You are on page 1of 33

Endocrine Reviews, 2024, 00, 1–33

https://doi.org/10.1210/endrev/bnae011
Advance access publication 15 March 2024
Review

Relative Energy Deficiency in Sport (REDs): Endocrine


Manifestations, Pathophysiology and Treatments
Angeliki M. Angelidi,1,2,* Konstantinos Stefanakis,1,2,3,4,* Sharon H. Chou,5

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


Laura Valenzuela-Vallejo,1,2 Konstantina Dipla,6 Chrysoula Boutari,7 Konstantinos Ntoskas,4
Panagiotis Tokmakidis,3,4 Alexander Kokkinos,3 Dimitrios G. Goulis,8 Helen A. Papadaki,9
and Christos S. Mantzoros1,2,5
1
Department of Medicine, Boston VA Healthcare System, Boston, MA 02115, USA
2
Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
3
First Propaedeutic Department of Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens,
Athens 11527, Greece
4
Department of Internal Medicine, 251 Air Force General Hospital, Athens 11525, Greece
5
Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital (BWH), Harvard Medical School, Boston, MA 02115,
USA
6
Exercise Physiology and Biochemistry Laboratory, Department of Sports Science at Serres, Aristotle University of Thessaloniki, Serres 62100,
Greece
7
Second Propaedeutic Department of Internal Medicine, Hippokration Hospital, School of Medicine, Aristotle University of Thessaloniki,
Thessaloniki 54642, Greece
8
Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki,
Thessaloniki 54124, Greece
9
Department of Hematology, University Hospital of Heraklion, School of Medicine, University of Crete, Heraklion 71500, Greece
Correspondence: Christos S. Mantzoros, MD, DSc, Beth Israel Deaconess Medical Center, 330 Brookline Ave, East Campus, ASN-249, Boston, MA 02215, USA.
Email: cmantzor@bidmc.harvard.edu.
*These authors have contributed equally to this work and share co-first authorship.

Abstract
Research on lean, energy-deficient athletic and military cohorts has broadened the concept of the Female Athlete Triad into the Relative Energy
Deficiency in Sport (REDs) syndrome. REDs represents a spectrum of abnormalities induced by low energy availability (LEA), which serves as the
underlying cause of all symptoms described within the REDs concept, affecting exercising populations of either biological sex. Both short- and
long-term LEA, in conjunction with other moderating factors, may produce a multitude of maladaptive changes that impair various physiological
systems and adversely affect health, well-being, and sport performance. Consequently, the comprehensive definition of REDs encompasses a
broad spectrum of physiological sequelae and adverse clinical outcomes related to LEA, such as neuroendocrine, bone, immune, and
hematological effects, ultimately resulting in compromised health and performance. In this review, we discuss the pathophysiology of REDs
and associated disorders. We briefly examine current treatment recommendations for REDs, primarily focusing on nonpharmacological,
behavioral, and lifestyle modifications that target its underlying cause-energy deficit. We also discuss treatment approaches aimed at
managing symptoms, such as menstrual dysfunction and bone stress injuries, and explore potential novel treatments that target the
underlying physiology, emphasizing the roles of leptin and the activin-follistatin-inhibin axis, the roles of which remain to be fully elucidated, in
the pathophysiology and management of REDs.
In the near future, novel therapies leveraging our emerging understanding of molecules and physiological axes underlying energy availability or
lack thereof may restore LEA-related abnormalities, thus preventing and/or treating REDs-related health complications, such as stress fractures,
and improving performance.

Received: 17 November 2023. Editorial Decision: 12 March 2024. Corrected and Typeset: 16 April 2024
Published by Oxford University Press on behalf of the Endocrine Society 2024.
This work is written by (a) US Government employee(s) and is in the public domain in the US.
2 Endocrine Reviews, 2024, Vol. 00, No. 0

Graphical Abstract

Graphical Abstract
Underlying etiologies
Elite athletic and military cohorts
Strict diet regimens

Demanding Persistently
training regimens increased energy Peer-related and trainer pressure
expenditure

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


Energy imbalance Persistently low energy intake

Physiological mediators Glucose, Insulin,


BCAA surge

Sustained shift from carbohydrate


Glycogen utilization depletion
to fatty acid metabolism
Low energy availability
Adipose tissue depletion
Leptin levels
Impaired energy homeostasis
Bodily adaptations both
as a result of and in response to
Endocrine manifestations low energy availability

Activin A GH pulse LH, FSH BMD Immunological


Follistatin GH sensitivity Sex steroids and hematological
F F IGFs, IGFBPs consequences
F A F
A F Functional HA Osteoporosis
A
A F Male infertility Stress fractures

Thyroid Catecholamines (?) Gastrointestinal Increased cardiovascular Performance


hormones ACTH Cortisol disturbances risk (limited evidence) Endurance
(long-term) Strength
Negative mental
health and sleep
consequences
Neuroendocrine axes Organ systems Performance
© 2024 Endocrine Society

Key Words: relative energy deficiency in sport, female athlete triad, low energy availability, pathophysiology, endocrinological disorders, therapeutic
approaches
Abbreviations: AFI, activin-follistatin-inhibin; AT, adipose tissue; BAT, brown adipose tissue; BM, bone marrow; BMAT, bone marrow adipose tissue; BMC,
bone mineral content; BMD, bone mineral density; BMI, body mass index; CLD, congenital leptin deficiency; DE, disordered eating; EA, energy availability;
EB, energy balance; EEE, exercise energy expenditure; EI, energy intake; FFM, fat-free mass; FHA, functional hypothalamic amenorrhea; FSTL3, follistatin
and follistatin-like 3; HA, hypothalamic amenorrhea; HPA, hypothalamic-pituitary-adrenal; HPG, hypothalamic-pituitary-gonadal; HRV, heart rate variability;
IOC, International Olympic Committee; JAK, Janus kinase; LEA, low energy availability; OPG, osteoprotegerin; P1NP, pro-peptide of type 1 collagen;
r-metHuLeptin, recombinant methionyl human leptin; RANKL, receptor-activator of nuclear factor κΒ ligand; REDs, Relative Energy Deficiency in Sport;
RMR, resting metabolic rate; SF, stress fracture; STAT, signal transducer and activator of transcription; TEE, total energy expenditure; WAT, white adipose
tissue.

• Basic, translational, and clinical research during en­


ESSENTIAL POINTS
ergy deprivation indicates the adipokine leptin as a
• Relative Energy Deficiency in Sport (REDs) refers to promising regulator of neuroendocrine function
a frequently overlooked constellation of disorders downregulated during LEA, which might be associ­
stemming from low energy availability (LEA) in di­ ated with several REDs-related disorders.
verse exercising populations of either biological • Currently, the front-line treatment approach targeting
sex. the underlying causes of LEA entails the incorporation
• REDs encompasses neuroendocrine abnormalities in of nutritional, behavioral, and lifestyle alterations.
central hypothalamic-pituitary axes, mainly the re­ • Leptin replacement therapy is currently not recom­
productive, thyroid, adrenal, and somatotropic axes mended by consensus guidelines for individuals with
as well as bone, hematological, immunological, hypothalamic amenorrhea or during low energy avail­
gastrointestinal, cardiovascular, neuromuscular, ability because its safety and effectiveness, including
and psychological abnormalities. potential benefits like improved neuroendocrine
Endocrine Reviews, 2024, Vol. 00, No. 0 3

[SF]) observed mainly in high-performance female athletes


function, menstrual and reproductive health, growth
with or without a disordered eating (DE) behavior (9). Low
factors, and bone health, require further investigation
energy availability has been acknowledged as the cornerstone
for confirmation.
for these unfavorable health outcomes recognized as the
• The ACTH-cortisol and activin-follistatin-inhibin axes
Female Athlete Triad (10-13). Since its inception in 1992
have also been shown to be implicated as potential reg­
(10) and reevaluation by the American College of Sports
ulators of a variety of biological processes, including
Medicine in 1997 (12), the term “Female Athlete Triad” in­
development, reproduction, body composition, and en­
volves any 1 of the following 3 interrelated components: (1)
ergy metabolism, independently of leptin; however, the
LEA with or without DE, (2) menstrual dysfunction, and (3)
full spectrum of diagnostic and therapeutic relevance of
low BMD (9, 12, 14).
the activin-follistatin-inhibin signaling pathways in

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


Furthermore, over the past several decades, animal experi­
REDs is under investigation.
ments and clinical research (mainly observational studies
• Because more studies describing the diagnosis, clinic­
and survey data) have revealed that some male athletes and
al relevance, and potential treatments of REDs are
soldiers may also experience similar LEA-induced physio­
being published, compounds in development, target­
logical abnormalities and health disruptions indicative of
ing the physiological pathways involved, could help
those identified in the Female Athlete Triad (15-21). Thus, a
improve therapeutic outcomes in energy-deficient
similar model was defined as the “Male Athlete Triad,” and
populations suffering from REDs.
it consists of 3 interrelated conditions, namely the LEA (as
the key etiologic factor) and perturbation of bone and repro­
ductive health (alterations of the hypothalamic-pituitary-
Over the past 3 decades, the fundamental role of energy avail­ gonadal [HPG] axis) (22, 23). However, he available evidence
ability (EA) in athletes’ health and overall well-being has at­ in males is still more limited, chiefly focused in military popu­
tracted increasing attention from the medical community, lations and based mainly on epidemiological information, but
international sports committees and federations, as well as contradictory data still exist (17, 21, 24, 25). Therefore, fur­
sports professionals and scientists in general (1). ther research efforts focusing on the underlying physiological
In sports science and nutrition, EA is defined as the differ­ mechanisms and long-term effects of the Male Athlete Triad
ence between total daily energy intake (EI) and exercise energy on health and performance are required.
expenditure (EEE) divided by the fat-free mass (FFM) (ie, EA In 2014, the International Olympic Committee (IOC) ex­
= (EI – EEE)/FFM, expressed in kcal/kg of FFM per day) (2-6). pert working group introduced a broader term, coined
EA represents the total energy at the body’s disposal to sup­ Relative Energy Deficiency in Sport (REDs) syndrome (26), re­
port fundamental physiological functions (such as thermo­ ferring but not limited to the LEA-related abnormalities (in­
regulation, growth, maintenance of cellular homeostasis, cluding also endocrine, hematological, immunological,
immune system functions, and reproduction) that underpin cardiovascular, cardiovascular, and psychological health dis­
optimal performance, adaptation to training, and ultimately orders) affecting athletic performance, physiological function,
health (3, 4, 7). It should be emphasized that EA indicates and health status in physically active men and women (21, 26-
the total available energy after deducting the energy expended 31). As awareness of REDs increases, regulatory organiza­
during exercise, thereby highlighting the possibility that tions in the field of sports have begun outlining the clinical
excessive energy expenditure during exercise can divert energy evaluation, diagnostic workup, and basic treatment of the
away from other critical physiological functions (8). syndrome (26, 30). There has also been extensive research
Moreover, energy balance (EB) is the difference between EI on DE as well as debate on the definition of REDs as a distinct
and total energy expenditure (TEE) (ie, EB = EI – TEE). In a clinical entity, juxtaposed against overlapping features of the
broader sense, EB is the amount of energy remaining from Overtraining Syndrome (32) and, importantly, the Female
EI after subtracting total energy used by the body, including Athlete Triad (33). Thus, a “pathway for progress” has been
the energy consumed for the function of all physiological sys­ suggested to facilitate cooperation between the Female
tems (and is thus either positive or negative) (4). Therefore, it Athlete Triad and REDs investigators, calling for quality stud­
is essential to acknowledge that EA and EB are not inter­ ies with clinically relevant outcomes and aiming to establish
changeable but reflect 2 different parameters and should be causality and address previous research gaps (34). In recent
clearly differentiated to identify athletes at risk of or athletes years, significant scientific progress has advanced our under­
in a pathophysiologically abnormal state. standing of the underlying physiology and psychology related
Both research evidence and clinical experience show that to REDs, culminating in the publication of the IOC consensus
athletes may adapt to situations of energy deficiency, particu­ statement on REDs in 2023 (6). Therefore, according to the
larly during prolonged periods of severely low energy avail­ revised 2023 definition, REDs is identified as a syndrome re­
ability (LEA). This adaptation occurs by suppressing sulting from prolonged and/or severe LEA (problematic),
energetically costly physiological processes (such as neuroen­ which affects athletes of both sexes, leading to impaired
docrine function, and growth), reducing the athlete’s overall physiological and/or psychological functioning, including det­
energy requirements, and altering physiological functions rimental health issues, compromised well-being, performance
such as reproductive and bone health or insulin resistance sta­ challenges, and elevated injury risks (6).
tus and lipolysis. These changes aim to promote survival in en­ The management of REDs should be multidisciplinary and
vironments of limited energy resources (4, 7, 9). carried out by a team of sports and exercise professionals.
Considerable research, commencing several decades ago, Current practice guidelines and consensus statements (14,
has been performed to elucidate the underlying causes of men­ 19, 35) highlight nutritional, behavioral, and lifestyle changes
strual dysfunction and low bone mineral density (BMD) lead­ as the recommended primary treatment strategies for both the
ing to poor bone health (eg, osteoporosis and stress fractures Female and Male Athlete Triad, as well as for REDs. To date,
4 Endocrine Reviews, 2024, Vol. 00, No. 0

there is a lack of effective pharmacological remedies to coun­ between males and females and interpersonal variability, diet­
ter the negative consequences of REDs. The adipokine leptin, ary macronutrient composition, the lack of previous models
the circulating levels of which reflect primarily the size of en­ to consider jointly acute energy changes and chronic energy
ergy stores in adipose tissue and secondarily acute energy de­ availability and the fact that the type of methods for EA assess­
privation, may be considered a potential contributory factor ment vary across studies and are not universally standardized
to the pathophysiology of the syndrome because of its involve­ (eg, self-reported questionnaires) (2, 4, 5, 55).
ment in regulating neuroendocrine function and energy In more detail, based on findings from early randomized
homeostasis. Leptin orchestrates an intricate response system clinical trials investigating gonadotropin pulsatility in regu­
to conserve energy by suppressing nonessential functions vs larly menstruating, habitually sedentary, young females of
deficits, aiming to ensure the survival of the organism through normal body composition, LH pulse frequency and ampli­

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


limiting procreation and preserving energy during LEA (36- tude were considered not disrupted when EA levels were
41) by influencing several neurohormonal axes affected in set at 45 kcal/kg FFM/day (56). Moreover, it was found
REDs. Administration of recombinant methionyl human lep­ that LH pulsatility did not follow a linearly proportional to
tin (r-metHuLeptin) to lean individuals with hypoleptinemia EA, whereas females with shorter luteal phases were more
in replacement doses to normalize the falling levels in response susceptible to disruption of LH pulsatility by restricted EA
to energy deprivation, confers neuroendocrine benefits (42- (56). Therefore, an EA of at least 45 kcal/kg FFM/day is typ­
44), including reversing the clinical manifestations of hypo­ ically recommended as the optimal EA to ensure sufficient
thalamic amenorrhea (HA) (45-47). Furthermore, leptin energy for healthy physiological function (4, 30). Likewise,
may help protect against LEA-induced low BMD and SF in EA <30 kcal/kg of FFM/day, even for a short period, has
the long term (48-52). However, leptin replacement therapy been associated with substantial perturbation of various
is not currently recommended pending further research on body systems (mainly including alterations in circulating thy­
its effectiveness and potential risks in addressing the syn­ roid hormones, disruptions in LH pulse frequency, and
drome’s pathophysiology. The activin-follistatin-inhibin increases in bone resorption) (56-58). Furthermore, our
(AFI) axis is an additional hormonal system known for regu­ comprehension of the LEA threshold or the range within
lating reproductive function and has secondary roles in main­ which males manifest REDs-related symptoms remains rela­
taining muscle mass, altering bone mass, and influencing tively limited. Nonetheless, current evidence suggests that
insulin sensitivity and thus lipolysis and glycemia (ie, provi­ this threshold is likely lower in males compared with females
sion of energy) (43, 53, 54). The role of the AFI axis in (59). Furthermore, it is crucial to recognize that the manifes­
REDs is starting to emerge and may also pinpoint to novel tations of REDs in males and the thresholds for LEA are less
therapeutic strategies, as is the stress hormone axis of understood and appear to differ from those in females, indi­
ACTH-cortisol, which is affected by both the leptin and the cating a need for gender-specific research in this area.
AFI axis. Consequently, our recent and evolving understand­ Therefore, an EA threshold lower than 30 kcal/kg FFM per
ing of the underlying and mediating pathophysiological mech­ day has been considered a potential LEA threshold, mainly
anisms is anticipated to change the way we approach, based on short-term controlled clinical trials in females.
diagnose, and treat this significant condition. However, because of limited epidemiological information on
Military training may negatively affect well-being and free-living individuals, paucity of long-term studies, and diver­
sports performance, especially among personnel who experi­ gent findings among research studies, fail to establish a stand­
ence energy deficits during training and field exercises, which ardized and robust threshold for LEA, which continues to be a
can be linked to the Triad and REDs because of the high en­ topic of active discussion (2, 5, 17, 24, 25, 30, 60, 61).
ergy expenditures and reduced energy intake (21). Our previous research highlights the importance of not only
Therefore, this review summarizes available research on the considering the acute daily energy intake but also longer term
definition and recognition of LEA/REDs in athletic and mili­ caloric reserves, specifically the amount of energy stored in
tary cohorts. It further explores the characteristics of REDs adipose tissue. We have found that the hormone leptin acts
and discusses the pathophysiology of endocrine conduits be­ as a “thermostat” by reflecting both acute changes in energy
hind REDs, including leptin and the AFI axis. Additionally, intake (ie, leptin levels drop by approximately 50% of base­
current and potential treatments for REDs, the Female line in response to acute and absolute caloric deprivation
Athlete Triad, and related disorders are discussed. and by only ∼10% of baseline after 3 days of acute and abso­
lute caloric deprivation even before any changes in total fat
mass and body weight occur) (62). The decreased leptin levels
LEA and the Manifestations of REDs
inform the brain of these changes and activate molecular path­
Definition of LEA ways that lead to the physiological changes depicted in the
As mentioned, EA is calculated by subtracting the EEE from EI graphical abstract. Nevertheless, it is important to note that
and dividing it by the FFM (6). Severe and/or extended LEA reductions in circulating leptin in relation to LEA have been
has been associated with several detrimental effects, with im­ reported in earlier studies (63).
paired reproductive function and bone health being the most No consensus has been developed to date concerning a
widely described. However, a consensus threshold for the targeted LEA threshold between optimal and impaired
minimum EA required to sustain all the physiological func­ physiological mechanisms, highlighting the complexity of ac­
tions necessary for optimal health and sports performance curately identifying individuals at risk, and this needs to be a
has not been achieved. Therefore, a precise threshold of priority for research in the future. Therefore, the currently
LEA remains undetermined, with ongoing debates in the sci­ used EA cutoff points (ie, 30 kcal/kg FFM/day) serve as a pre­
entific community. This uncertainty in defining a clear-cut liminary guide and should not be used diagnostically because
threshold could also be attributed to the length of LEA expos­ of the complex interplay of factors affecting LEA and REDs.
ure, type of exercise, body size and composition, differences However, EA thresholds might be helpful to direct systematic
Endocrine Reviews, 2024, Vol. 00, No. 0 5

research on the mechanistic underpinnings of LEA and pro­ However, additional research is warranted in this area.
vide a gross tool to identify athletes at risk of LEA and subse­ More rigorous study designs are essential to improve the over­
quent performance decline and REDs development. all quality of evidence regarding the LEA definition and the
In conclusion, because of individual variability, including Female Athlete Triad and REDs-related outcomes (75).
gender differences as previously described, and the outlined Therefore, more prospective long-term studies considering
limitations, a precise threshold of LEA remains undetermined. the impact of potential moderating and confounding factors
Thus, it is imperative to acknowledge the ongoing debates and are needed to elucidate further the causality and timing of
contradictions, which highlight the complexities and inherent the effects of energy deficiency on the Female Athlete Triad
challenges in the evidence-based classification and precise def­ and REDs development. It is also crucial to consider all of
inition of both LEA and REDs. This underscores the need for the aforementioned factors jointly rather than independently

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


further research to refine our understanding and methods, of each other.
which is essential for advancing both scientific knowledge
and clinical applications related to this condition. Key Abnormalities of REDs
The physiological consequences of REDs stem from problem­
LEA: Causative Factor of REDs atic LEA (graphical abstract) and encompass most neuroendo­
crine systems in the human body (Table 1). However, several
It is crucial to acknowledge that LEA is the primary etiological
short- and long-term outcomes and related signs and symp­
key factor of the REDs, which expands the concept of the
toms are not REDs-specific, and these consequences can be
Female and Male Athlete Triad to encompass a wide range of
observed in the absence of LEA under the prism of different
potentially LEA-related severe health consequences beyond
health disorders. Therefore, the differential diagnosis should
those included in the aforementioned Triad models (26, 56,
always be considered and carefully conducted based on a com­
57, 64-66) and negatively affect the sport performance and well-
prehensive assessment and thorough evaluation of the patient’s
being of individuals at risk (67, 68). The underlying conditions
clinical presentation, imaging findings, and laboratory exami­
underpinning the development of LEA are multifactorial and
nations, as appropriate.
complex, including, but not limited to, an intentional or unin­
tentional reduction in caloric intake (eg, cultural eating habits,
restriction, binging, fasting, anorexia nervosa, other eating dis­ Substrate Metabolism and Food Intake
order/DE disorders) (69), an increase in daily exercise energy ex­ The biochemical fingerprint of short-term LEA can be ob­
penditure (eg, prolonged acute exercise, intense and extended served in the context of acute nutrient deprivation, which is
duration training demands), and a poor awareness or miscon­ well known to trigger a classical physiological cascade.
ceptions about the syndrome resulting in an inappropriate Fasting- and energy-deficiency induced hypoglycemia (172,
matching of intake energy to the exercise energy demands (9, 173) can be partially prevented, owing to reduced insulin pro­
26, 30, 70-72). Interestingly, psychological impairments can ei­ duction and elevated glucagon, epinephrine, and cortisol se­
ther contribute to LEA and precede REDs (as an exposure vari­ cretion secondary to starvation (174, 175). However,
able) or result from REDs (as an outcome) (26, 67). carbohydrate and glycogen stores are soon depleted, and glu­
However, it should be noted that LEA may not always dis­ coneogenesis takes over using amino acids and glycerol for 24
rupt physiological processes and result in the clinical conse­ to 48 hours, followed by ketogenesis (174). After a few days, a
quences described in the REDs, a fact that is more apparent, branched-chain amino acid surge triggers the shift to lipid me­
especially in males (30, 55, 72). Consequently, LEA may be tabolism and ketogenesis, as attested by elevated cortisol, free
considered adaptable, particularly in the short term and in fatty acids, β-hydroxybutyrate, and ketone body concentra­
the context of transient and reversible conditions, in which tions (173, 176, 177). Once adipose tissue (AT) reserves are
LEA might be associated with a necessary and valuable activity depleted, the lipolytic metabolic shift is followed by protein
in sport (eg, body composition management and intensified catabolism. Caloric deprivation importantly produces
training) (6). On the contrary, LEA could be considered prob­ changes in circulating adipokines, the prime sensors of energy
lematic when it persists over an extended duration or exhibits deficiency and appetite and energy intake controlling hor­
specific characteristics indicative of a maladaptive response mones and, thus, calibrators of adipose tissue energy reserves
(6). LEA exposure, in conjunction with other moderating and function. Acute energy deficits have a major effect on
factors, may lead to impaired physiological processes. body composition, energy balance, and hormone concentra­
Consequently, normal physiology can transition into patho­ tions, including leptin (42, 85, 178) but exerts little effect on
physiology by disrupting physiological systems, ultimately re­ adiponectin (179). A sustained energy deficit leading to dimin­
sulting in poor performance and health issues from ished AT reserves decreases leptin production and alters rest­
inadequate energy to support optimal functions (8). The char­ ing metabolic rate (RMR) and neuroendocrine profile to
acteristics of problematic LEA and the potential moderating prolong survival (180). In the long term, LEA can dysregulate
factors may differ among individuals and body systems (eg, the secretion of orexigenic peptides (ie, ghrelin and PYY), with
sex, genetic characteristics, preexisting medical conditions, studies reporting both increases and decreases in circulating
and most importantly, energy reserves such as overall fat levels (181, 182).
mass at baseline, type of sport, exercise training duration and
characteristics). Moreover, emerging evidence suggests that be­ Neuroendocrine Effects
sides the amount of caloric consumption, the dietary patterns
followed (eg, low carbohydrate consumption) may exert a Hypothalamic-pituitary-gonadal axis
negative impact on athletes’ health, but this remains to be stud­ Suppression of the HPG axis is a major adaptive mechanism in
ied further and in conjunction with underlying neuroendocrine states of energy deprivation. This adaptation occurs because
pathways activated or inactivated (73, 74). procreation may not be prioritized when the survival of the
6 Endocrine Reviews, 2024, Vol. 00, No. 0

Table 1. Most commonly reported clinical and biochemical manifestations reported in milieus of REDs

Physiological axis Manifestation Most common study type

Endocrine Hypothalamic-pituitary-gonadal Functional HA (12, 36, 61, 65, 68, 76-79) Systematic review
Society guidelines
Consensus statements
Prospective cohort
Cross-sectional
Diminished gonadotropin (LH, FSH) Open-label clinical trial
concentrations and pulses (56, 80-84) Prospective cohort
Decreased testosterone concentrations (81-83, Prospective cohort

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


85-91) Cross-sectional
Decreased motile sperm count (86, 92) Cross-sectional
Abnormal sperm morphology (93)
Hypothalamic-pituitary-thyroid Reduced thyroid hormone concentrations (42, 57, Case-control
85, 94-97) Prospective cohort
Cross-sectional
Blunted TSH pulse and response to TRH (42, 96) Cross-sectional
Prospective (short-term)
Hypothalamic-pituitary-somatotropic Accentuated GH pulse peaks (short-term) (98, 99) Prospective cohort
Decreased IGF-1 levels (56, 85, 99-103) Prospective cohort
Increased IGFBP-1 levels (104) Clinical trial
Decreased IGFBP-4, IGFBP-6, GHBP levels (104) Clinical trial
Hypothalamic-pituitary-adrenal Elevated cortisol concentrations (105-109) Review
Prospective cohort
Blunted diurnal salivary cortisol pulse (42, 110, 111) Prospective cohort
Leptin, insulin, and ghrelin Reduced insulin levels (112) Prospective (short-term)
Elevated ghrelin levels (113) Cross-sectional
Reduced leptin levels (42, 45, 46, 104, 112, Clinical trial
114-116)
Bone Loss of BMD and premenopausal osteoporosis (17, 24, 25, 58, 70, 117, 118) Randomized controlled trials
Reviews, Society guidelines,
consensus statements
↑ NTX, CTX, bone resorption markers (58, 70, 119) Randomized and nonrandomized
trials
Cross-sectional
Stress fractures (120-136) Systematic review
Epidemiological
Prospective cohort
Case-control
Immunological Diminished lymphocyte and increased neutrophil counts (137-139) Review
Randomized controlled trial
Prospective cohort
Increased white blood cell counts (73) Prospective cohort
Increased IL-6 levels (140-142) Prospective cohort
Decreased monocytes and T-cell subpopulation (143) Prospective cohort
T-cell hyporesponsiveness (144) Clinical trial
Neutrophil hyporesponsiveness and decreased phagocytic activity (145, 146) Prospective cohort
Decreased IgA levels (64, 147) Prospective cohort
Cross-sectional
Increased likelihood of infections and illnesses (64, 147-151) Cross-sectional
Muscle and Decreased muscle mass and strength, increased risk of injuries (152-154) Randomized controlled trial
performance Cross-sectional
Prospective
Decreased explosive power (155) Prospective
Dysregulation of myokines (156, 157) Review
Reduced muscle glycogen content (158) Crossover randomized controlled trial
Decreased muscle protein synthesis (159) Clinical trial
Diminished activin and increased follistatin concentrations (43, 53, 111, 160, 161) Clinical trial
Prospective
Impaired sports performance (162) Review
Hematological Iron deficiency anemia (163, 164) Cross-sectional
Abnormal levels of ferritin and transferrin, elevated levels of hepcidin (73, 142, 165-167) Cross-sectional
Prospective
Case-control
Cardiovascular Endothelial dysfunction concurrent with HA—association with increased cardiovascular risk Cross-sectional
(168)
Gastrointestinal Nausea, regurgitation, upper abdominal bloating, diarrhoea, delayed gastric emptying and Systematic review
constipation (169, 170)
Increased intestinal permeability (170, 171) Systematic review

Abbreviations: BMD, bone mineral density; CTX, collagen type 1 C-telopeptide; GHBP, GH-binding protein; HA, hypothalamic amenorrhea; IGFBP, IGF binding
protein; NTX, N-terminal telopeptide.
Endocrine Reviews, 2024, Vol. 00, No. 0 7

organism is at risk because of LEA, which may not provide on T3 levels has been observed, emphasizing the role of base­
sufficient energy to support the creation and development of line energy stores and sex (as indicated by varying leptin levels
a new organism. LEA is associated with suppression of in males and females) (112, 189).
GnRH signaling and pulsatility, reduced pituitary gonadotro­ LEA association with thyroid hormone alterations is likely
pins, primarily LH levels (56, 80), and gonadal steroid hor­ to be subject to several parameters, including participant’s
mones, including decreased estrone-1-glucuronide (principal characteristics (within and between participant variability,
metabolite of estradiol) and pregnanediol glucuronide (pri­ sex, menstrual status), the underlying causes of LEA (dietary
mary metabolite of progesterone), resulting in menstrual dis­ restriction, increased exercise energy expenditure), and the
turbances (60) and functional hypothalamic amenorrhea type, duration, and volume of athletic training. Therefore,
(FHA). Past studies have indicated that up to 50% of exercis­ the full consequences of LEA on thyroid-binding proteins

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


ing women experience menstrual irregularities (76), whereas and other components of the axis remain to be fully investi­
FHA in itself constitutes 1 of the most common manifestations gated. This research should be conducted in the context of
of REDs (36, 77, 78). The presence of FHA depends on the the effects of those factors on both acute and chronic energy
duration and severity of LEA (30) and was recently reviewed availability (ie, acute changes in energy balance in relation
(79). The highest prevalence for primary amenorrhea was re­ to the baseline energy stored in adipose tissue).
ported in rhythmic gymnastics at 25%, for secondary amenor­
rhea in cycling at 56%, and for oligomenorrhea in boxing at Hypothalamic-pituitary-somatotropic axis
55% (79). However, study and group heterogeneity, as well
In the anterior pituitary gland, GH is released by somatotroph
as menstruation reporting, should be taken under careful con­
cells and binds to GH receptors in multiple tissues, including
sideration (77, 183).
the liver, which is the main organ responsible for IGF-1 secre­
There are conflicting results from studies investigating the
tion. GH may stimulate the production of IGF-1 via binding to
occurrence of hypogonadism in male athletes, with some inves­
GH receptor, activating Janus kinase 2 (JAK2) and subse­
tigations underlining differences from female manifestations
quently the phosphorylation of signal transducer and activa­
and varying forms of gonadal disruption in duration, quality,
tor of transcription 5 (STAT5), which plays an essential role
and reversibility, encompassing a vast array of hypogonadism
in IGF-1 gene expression (190, 191).
forms that occur in elite male athletes (184). Likewise, albeit
LEA accelerates GH pulse frequency and accentuates GH
studied to a lesser extent, elite male endurance athletes may
peaks (98, 99), whereas it reduces IGF-1 production, poten­
display notable downregulation of gonadotropin concentra­
tially reflecting peripheral resistance or reduced sensitivity to
tions, blunted gonadotropin pulses, suppressed testosterone
growth signals through diminished liver production of IGFs
levels, and impaired spermatogenesis (30, 81-83, 86). In mili­
and downregulated hepatic and peripheral somatogenic re­
tary milieus, a series of surveys on male Norwegian cadets
ceptor expression (56, 99-102).
under a 5-day arduous training course has demonstrated
LEA in the form of energy deprivation culminating in hypo­
downregulated gonadotropin and sex steroid hormone levels
thalamic amenorrhea, leads to increased levels of IGF binding
(87-89). Numerous other studies in energy-deprived male sol­
protein 1 (IGFBP-1) and diminished levels of IGFBP-4,
diers have proven similar changes in testosterone and sex ster­
IGFBP-6, and growth hormone-binding globulin compared
oid hormones (185-188), even reaching “castrate levels”
with healthy individuals (104), potentially indicating a con­
following prolonged ranger training courses (85).
sistent downregulation of hepatic GH receptors and reduced
bioactivity of growth factors, hampering overall circulating
Hypothalamic-pituitary-thyroid axis GH activity. Moreover, resistance exercise, a potent anabolic
stimulus, increased GH response and decreased IGF-1 levels in
The hypothalamic-pituitary-thyroid axis also adapts to energy
female and male participants during a short-term, calorie-
deprivation (31) by implementing mechanisms that promote
restricted LEA state while on a postexercise protein or carbo­
and prolong survival through energy conservation (78).
hydrate supplementation (103). Significant and consistent ef­
Short-term energy deprivation exercise studies, including pre­
fects have been observed in male soldiers, wherein increased
viously untrained regularly menstruating females, showed
energy expenditure leads to an upregulation of IGFBP-1 and
that LEA induces a nonlinear reduction of T3 and free T3 lev­
an intermediate downregulation of IGFBP-3 (186, 192,
els, which is associated with decreased metabolism and energy
193). These results suggest that a catabolic state may also limit
expenditure in states of reduced energy availability, and an in­
the effectiveness and/or the potential benefits of resistance ex­
crease of reverse T3 levels (ie, the inactive form of thyroid hor­
ercise during the calorie-restricted state.
mone) (57, 94). Downregulation of T3 has also been
Finally, GH may exert effects on carbohydrate and lipid
consistently observed in female athletes with LEA and FHA
metabolism and help to maintain euglycemia via the mobil­
compared with eumenorrheic individuals (95-97).
ization of fat stores in an IGF-1-independent way (194,
More complex findings regarding T4 and free T4 changes
195). Therefore, GH resistance could be considered an
have been observed. An increase of T4 has been described
adaptive response to LEA. The decreased IGF-1 levels may
(94), suggesting a potential mechanism related to decreased
help to conserve energy, and through a decreased negative
conversion of T4 to T3, which is the active thyroid hormone.
feedback at the pituitary, the subsequent increased circulat­
Other studies either identified a decrease in T4 levels (96) or
ing GH levels may help to maintain euglycemia under LEA
did not detect a difference (57). Similar findings have been
conditions (190).
noted in soldiers, wherein marked downregulations of T3,
T4, and TSH have been noted through longitudinal evalua­
tions of training male cadets and conscripts (85, 90, 186, Hypothalamic-pituitary-adrenal axis
188). Supporting literature on male athletes is limited, and Increased serum and cerebrospinal fluid cortisol concentra­
in contrast to females, either a decrease or a neutral effect tion (105-107), reduced cortisol response to intense exercise
8 Endocrine Reviews, 2024, Vol. 00, No. 0

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


Figure 1. Acute and chronic exercise-induced stress responses of neuroendocrine axes. Acute and chronic exercise, particularly in excess, constitute
important stress effectors. Acutely, this response activates the locus coeruleus-norepinephrine system and induces the secretion of catecholamines
(sympathetic response), while also boosting glucocorticoid production, which are known to acutely produce a spike in GH. Chronic stress, on the other
hand, also produces sustained increases in glucocorticoids through activation of the hypothalamic-pituitary-adrenal axis. The parent hormone, CRH, is
known to suppress GnRH through β-endorphins, as well as TRH, and increase somatostatin, which in turn inhibits GH and TRH. Meanwhile, increased
glucocorticoid production suppresses the concentrations of thyroid hormones, GH, and GnRH. These effects, when compounded and persistently
accentuated in chronic stress, which entails excessive exercise, may lead to lower muscle mass and BMD, as well as to increased visceral adiposity risk.
Abbreviations: BMD, bone mineral density; CRH, corticotropin-releasing hormone; GnRH, gonadotropin releasing hormone; IGFBP, IGF binding protein;
LC/NE, locus coeruleus-norepinephrine; SNS, sympathetic nervous system; TRH, thyroid releasing hormone.

(106), as well as accentuated cortisol pulse amplitude com­ Immunological Effects


pared with eumenorrheic exercisers (108) have all been estab­ As attested by experiments in mice, mainly because of falling
lished in women with HA. The diurnal pulsatility of salivary leptin levels in response to energy deprivation, an abnormal
cortisol appears to be abolished in elite gymnasts, which Th1/Th2 balance, upregulated production of proinflamma­
may indicate an adaptation to prolonged periods of LEA tory cytokines (ie, IL-6 and TNF-α), and cortisol is observed
(196). In the long term, however, cortisol levels may be in­ with LEA and may increase the risk of specific infections
creased, reflecting an increasingly stressful situation from pro­ (201). In the 2018 and 2023 consensus statements (30,
longed energy deprivation. Malnutrition and anorexia, on the 202), the IOC noted that LEA might impair the immune sys­
extreme spectrum of energy deficiency, as well as chronic ex­ tem, highlighting the increased rates of upper respiratory
cessive exercise, directly related to REDs, have been well- and gastrointestinal tract infections based on observational
established as effectuators of acute and chronic stress and data from amenorrheic elite female distance runners (64)
activate the hypothalamic-pituitary-adrenal (HPA) axis and Olympic athletes assessed as being at risk for LEA (using
(109, 197). In turn, this effect entails alterations in almost the low energy availability in females questionnaire) (148,
all hypothalamic-pituitary axes and may lead to reductions 149). Findings of various studies have suggested adverse ef­
in muscle mass and BMD (Fig. 1). This increase in cortisol lev­ fects of energy restriction and weight loss on immunological
els is a mechanistically multifactorial process, which depends parameters, such as impaired humoral and cell-mediated im­
not only on decreasing leptin levels, as all the previously de­ mune function and subsequent increased risk of potential in­
scribed neuroendocrine changes, but also on alterations in fections in athletes. In particular, 2 observational studies
the AFI axis and other proinflammatory molecules in the per­ exploring the effects of intensive training and rapid weight
iphery, as well as changes in neuropeptide levels centrally loss on immunological markers revealed suppressed mucosal
(Figs. 1-3). The elucidation of energy deficiency-induced immunity indicated by the reduced levels of salivary Ig A
changes in the HPA axis is further evident in studies of both and an increased upper respiratory tract infection incidence
male and female soldiers, consistently reporting an upregula­ in male and female elite taekwondo athletes (147, 150).
tion of cortisol (187, 188, 198, 199), which has been shown to Decreased salivary Ig A levels and increased upper respiratory
be more pronounced in females (200). tract infection symptoms were also observed in amenorrheic
Endocrine Reviews, 2024, Vol. 00, No. 0 9

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


Figure 2. Endocrine effects underlying REDs-associated states across tissues and systems, highlighting the independent functions of leptin and the AFI
axis. Arrows with plus signs indicate positive relationships or feedback in states of hypoleptinemia; arrows with minus signs indicate negative feedback;
up and down arrows represent changes (increase or decrease, respectively) occurring under energy deprivation. Leptin is a central regulator of the HPG
axis and can simultaneously influence bone health both directly and indirectly through modulations in the reproductive system. In states of LEA, leptin
levels are decreased, thus leading to downregulation of the reproductive system and its hormones, bringing about long-term directly or indirectly bone
loss; these changes can be reversed through leptin supplementation. However, the AFI axis functions independently of leptin. LEA upregulates folli­
statins and decreases activins. Follistatins thus render the reproductive system inactive, help preserve precious muscle tissue, which is achieved through
follistatins blunting the catabolic functions of activins and myostatin. In the liver, follistatin concentrations are inversely correlated with insulin sensitivity
and improved glucose metabolism, thus leading to de novo lipogenesis and glycemia. Emerging evidence demonstrates that lower activins/follistatin
ratios are also linked to osteopenia and osteoporosis. Abbreviations: ACTRII, activin receptor type 2; AMH, anti-müllerian hormone; BMD, bone mineral
density; fT3, free triiodothyronine; FSTL3, follistatin-like 3; HPA, hypothalamic-pituitary-adrenal; HPG, hypothalamic-pituitary-gonadal; HPT, hypothal­
amic-pituitary-thyroid; IGF-1, insulin-like growth factor; IGFBP, IGF binding protein; TG, triglyceride.

elite female distance runners compared with the eumenorrheic also outlined in the hematological effects section that follows
runners (203). Furthermore, weight loss (reduced energy in­ and can display downregulated in vitro T-lymphocyte re­
take) was associated with impaired cell-mediated immune sponses and increases in infection rates (151, 206). Further re­
function (ie, decreased counts of specific monocytes and search is needed to more precisely identify the relationship
T-cell subpopulations) and high susceptibility to upper re­ between LEA, immunological parameters, immune functions,
spiratory tract infection symptoms in an observational study and susceptibility to infectious diseases. However, these
of judo athletes (143). In addition, weight reduction resulted changes should be seen in the broader context of energy de­
in decreased cytokine production (ie, interferon-g), prolifer­ privation leading to immune dysfunction in a direct but also
ation of T cells (144), and neutrophil phagocytic activity in a potentially indirect way through alterations in leptin,
(145, 146) in case-control studies in amateur wrestlers and AFI, and cortisol levels. Pathophysiologically, similar but
judo athletes. However, in a randomized control trial, calorie more pronounced changes occur in other states of more severe
restriction during high-intensity exercise was associated with energy deprivation, such as in starving populations in devel­
an increase in IL-6 and TNF-a levels and a significant variation oping countries.
in lymphocyte, leukocyte, and neutrophil counts (204). Of
note, a large cross-sectional evaluation of 2247 elite athletes
has shown markedly downregulated white blood cell and neu­ Hematological Effects
trophil counts in athletes trained for aerobic vs skill-based Lower hemoglobin concentrations and iron deficiency have
sports, indicating a negative immune adaptive response which been observed in both male and female professional athletes
is distinct for individuals at risk for LEA (205). On the other (163, 164, 207). LEA has been associated with iron deficiency
hand, male soldiers also display reportedly increased circulat­ and impaired ferritin levels, which may result in iron defi­
ing levels of IL-6 and TNF-a (140-142) as well as hepcidin, ciency anemia commonly seen in young female athletes
10 Endocrine Reviews, 2024, Vol. 00, No. 0

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


Figure 3. Neuroendocrine regulation of reproduction, bone health, growth, thyroid, and adrenal function by leptin. Leptin concentrations are directly
proportional to AT energy reserves, yet obese patients demonstrate significant resistance to its hormonal effects. In the brain, leptin can act directly on
dopaminergic neurons or bind to its receptor in the hypothalamus, leading to upregulation of proopiomelanocortin and the downregulation of the ore­
xigenic NPY and agouti-related protein (AgRP) neurons, thus exerting a holistic control over appetite and food intake. Leptin directly influences the SNS in
animals but not in humans. The hypothalamic effects of leptin influence the pituitary downstream, affecting several vital endocrine axes in both animal
models and human cohorts. Through kisspeptin-1 and other neurons, GnRH secretion triggers LH and FSH production in a pulsatile manner directly
influenced by leptin’s circadian variations. Thyroid and corticoid hormone adaptations, driven by perturbations in TRH, TSH, and CRH, respectively, are
also influenced by leptin, especially in animal models but also in humans in lesser degrees. Leptin also exerts control over the growth factor IGF axis, with
administration in humans effectuating increases in IGF-1 and IGFBP-1. Last, leptin constitutes a key conduit of bone health, either directly or indirectly by
influencing estrogen and other trophic hormone concentrations. Abbreviations: AgRP, agouti-related protein; CNS, central nervous system; CRH, cor­
ticotropin-releasing hormone; E2, estradiol; IGF, insulin-like growth factor; Kiss1, kisspeptin-1; NPY, neuropeptide-Y; POMC, proopiomelanocortin; SNS,
sympathetic nervous system; TRH, thyrotropin-releasing hormone.

(208, 209). Iron deficiency has been suggested as a hemato­ increase hepcidin levels, which may induce potential de­
logical concern of interest in the context of REDs, according clines in iron status (142, 166). However, the duration and
to the updated 2018 IOC consensus statement, and a potential type of exercise might also have different effects on iron con­
driver for other REDs-related health complications (30). The centrations (167). In addition, an indirect effect of LEA on
prevalence of iron deficiency has also been frequently reported iron levels may be secondary to undernutrition or decreased
in both male and female soldiers, both throughout and follow­ macronutrient (ie, low carbohydrate availability) and
ing training (210-214), alongside elevated hepcidin and fer­ micronutrient intake combined with increased iron de­
ritin concentrations (141, 142). mands and a substantial reduction in bioavailable iron in
LEA may impact the iron balance in both direct and athletes (215).
indirect ways. Innately, an impaired reproductive axis in fe­ On the other hand, iron deficiency may also interact with
males results in menstrual dysfunction and amenorrhea. multiple physiological systems, resulting in exacerbated en­
Therefore, LEA may improve iron status by reducing iron ergy deficiency, impaired reproductive functions, and poor
losses from menstrual bleeding. However, elevated acute bone health (the 3 pillars of the known Female Athlete
phase reactants and proinflammatory molecules, including Triad), which are REDs-related health consequences (30,
hepcidin concentrations (a known iron-regulating protein 216). In more detail, iron deficiency may be associated with re­
associated with reduced dietary iron absorption and im­ productive endocrine dysfunction, hyperprolactinemia, infertil­
paired iron metabolism) and abnormal ferritin levels, and ity issues, reduced metabolic efficiency, and worsened energy
low-grade inflammation-induced iron deficiency have been status via impaired thyroid function (209). In addition, iron de­
observed in athletes with a high-intensity workload (165). ficiency may induce neuroendocrine abnormalities by impair­
Moreover, LEA accompanied by conditions with high en­ ing the levels and function of dopamine, serotonin, and
ergy expenditure may also trigger an immune response and norepinephrine neurotransmitters (217). These neuropeptide
Endocrine Reviews, 2024, Vol. 00, No. 0 11

alterations might contribute to traits associated with (229). Athletes classified in the “fatigue state” (score exceeded
anxiety-related disorder symptoms and disturbed appetitive 20 negative items of 54) experienced a lower and more vari­
and eating behaviors (218, 219). In addition, iron deficiency able HRV than their counterparts not classified in the fatigue
per se may induce negative feelings of anxiety, depression, state (229). However, direct evidence regarding cardiovascu­
and low perceived quality of life (220). Therefore, it has lar health and HRV in REDs in athletic populations and the
been proposed that iron deficiency may also affect the risk military is still limited; more research is warranted (33).
for DE by increasing anxiety-related behaviors and disrupt­
ing eating behaviors via neuroendocrine aberrations (209).
Gastrointestinal Effects
Iron deficiency may also compromise bone health through
imbalances in growth factors (eg, GH and IGF-1 suppres­ The redistribution of blood and nutrients toward metabolical­
ly active tissues during exercise can lead to gastrointestinal

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


sion), hypoxia-induced bone resorption, and thyroid dysre­
gulation (209). disturbances, alterations in the gut microbiome, and changes
The available research is limited. Future investigation is re­ in markers of gut health, damage, and permeability in healthy
quired to elucidate further the relationships between iron de­ exercising athletes (171, 230, 231). REDs has been associated
ficiency and LEA and its components and explore the with gastrointestinal symptoms in athletes, such as gut dam­
potential effects of dietary interventions, supplementation, age, increased intestinal permeability, delayed gastric empty­
neuroendocrine regulation, and restoration of menstrual func­ ing, constipation, and worsened sphincter control (30, 171).
tion on iron metabolism and REDs outcomes. Several gastrointestinal disturbances (increased intestinal per­
Bone marrow fat actively contributes to various metabolic meability, diarrhea, irritable bowel, chronic pain, and consti­
processes such as energy storage, endocrine function, and pation) (140, 232, 233) and even relevant clinical entities (eg,
bone metabolism. The most abundant type of cells in the Gulf War syndrome) (234) have been reported in soldiers, yet
bone marrow cavity and hematopoietic microenvironment these have not been adequately delineated in the context of
are bone marrow adipocytes. These cells play a crucial role REDs.
in maintaining the balance between the proliferation and dif­
ferentiation of hematopoietic stem cells, thereby supporting Muscle and Performance Effects
normal blood cell formation (221). Bone marrow (BM) adi­ The beneficial effects of exercise include reductions in body fat
pose tissue (BMAT) is regulated in a distinct manner that re­ and inflammation and improvements in strength and endur­
flects visceral fat stores in obesity and does not shrink under ance (235, 236). Moreover, adequate carbohydrate and pro­
caloric deprivation (221). BMAT possesses both metabolic-, tein intake are essential to maintain blood glucose levels,
bone-, and immune-related properties and is considered the replenish muscle glycogen, repair and synthesize muscle
third-largest fat storage after subcutaneous and visceral fat, tissue, promote muscle protein synthesis (237) and muscle
accounting for up to 70% of BM volume and ∼10% of total adaptation to training (238, 239), and maximize training out­
adipose mass. It derives from marrow adipogenic lineage pre­ comes. In addition, it has been suggested that resistance exer­
cursors, which are cells that possess adipocyte markers but do cise combined with increased postexercise protein availability
not contain lipid droplets and function by preserving marrow may promote muscle protein synthesis in a dose-dependent
vasculature and preventing the differentiation of mesenchy­ way during a short-term energy deficiency state and ultimately
mal progenitor cells into osteoblasts (222). preserve lean (muscle) mass in the long term (159).
BMAT, a metabolically active, insulin-sensitive tissue, cor­ Therefore, energy deficits, often accompanied by inad­
relates with the amount of visceral fat in obesity, whereas equate carbohydrate or protein intake (240), may lead to
obesity and aging induce ectopic adipocyte accumulation in lower muscle glycogen stores (56, 158), which may result
the BM (223, 224). BMAT accumulates in individuals with in unfavorable health outcomes (240) and sports perform­
chronic caloric restriction, such as in anorexia nervosa and ance (74). In addition, incomplete recovery and increased fa­
possibly REDs, or during skeletal growth periods, localized tigue further impede athletic or military goals (21, 30).
principally within the tibia or femur (225). Studies in mice Importantly, as shown by a recent investigation in elite
have shown how BMAT fails to mobilize its lipid reserve dur­ male endurance athletes, reduced performance and explosive
ing moderate starvation (226) and is resistant to β-adrenergic power as a result of reduced energy availability may precede
lipolysis stimulation. hormonal changes (ie, reductions in IGF-1 levels) (155),
whereas previous studies on Norwegian soldiers have dem­
onstrated a progressive deterioration of muscle damage,
Cardiovascular Effects
soreness, and blunted performance (241), and a marked
In young endurance athletes, a link between amenorrhea and and persistent drop in performance even 1 week after hormo­
endothelial dysfunction has been suggested (168). Low estro­ nal imbalances were normalized following refeeding (185).
gen levels have also been associated with unfavorable lipid
profiles and increased cardiovascular risk as well as compro­
mised orthostatic and cardiac sympathetic responses (168, Bone Health and Stress Fractures
227). Along with decreased muscle mass and reduced performance,
Heart rate variability (HRV), reflecting autonomic nervous LEA may result in compromised bone health linked to in­
system function, is a metric of fitness and performance in creased risk of SF in athletes and military personnel as well
sports and the military (228). Elite endurance athletes who as long-term consequences (ie, osteoporosis) (21, 118, 242).
participated in a 4-year longitudinal study were classified as Moreover, a negative dose-response association between EA
experiencing fatigue or not based on their scoring on a specific and bone turnover was found in a randomized control trial
questionnaire designed and validated by the consensus group in which the energy availability (ie, 10, 20, or 30 kcal/kg
on overtraining of the French Society of Sports Medicine FFM/day) was controlled in 29 young, regularly menstruating
12 Endocrine Reviews, 2024, Vol. 00, No. 0

exercising women (58). Potential sex differences have been de­ Adipose Tissue
scribed, with females displaying increased bone resorption White AT (WAT) stores energy, brown AT (BAT) is consid­
and decreased bone formation (70). In male athletes, loss of ered a key mediator of thermogenesis. A third type of AT
BMD is often characterized as a direct or indirect effect of termed “beige” or “brite” (brown-in-white) fat comprises re­
LEA, particularly in weight-restrictive, lean endurance sports cruitable thermogenic brown adipocytes within WAT depots,
(eg, cycling, rowing, distance running), leading to prolonged as demonstrated through in vitro and preclinical experiments
periods of LEA (71). (246). BAT was first investigated as a key thermogenetic tissue
Both short-term diet-induced LEA and exercise-induced in small mammals (247). In humans, less than half of BAT de­
LEA have been associated with reduced bone formation in posits are activated under cold exposure, and BAT appears
eumenorrheic women. Further evidence suggests that the less activated in obese individuals compared with lean coun­

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


type of diet may also play a role in bone formation. In par­ terparts (248). Exercise can induce “browning” of WAT,
ticular, a low-carbohydrate diet has been found to be associ­ causing AT to switch from storing white adipocytes to beige
ated with a more profound reduced bone formation, whereas adipocytes, and has been hypothesized to be an adaptive pro­
sufficient energy, including adequate carbohydrate support, cess (249). Interestingly, BAT thermogenic activity may be
may mitigate the exercise-induced unfavorable bone turn­ lower in male endurance athletes and chronically exercising
over responses (74). Inadequate nutrition, regarding both women compared with nonexercising controls (250, 251).
macronutrients (eg, carbohydrates, protein) and vitamins BAT activity is negatively correlated with amenorrhea dur­
and micronutrients (eg, vitamin D and calcium, iron), as ation (251), potentially indicating compromised metabolic ac­
well as hormonal disturbances (eg, sex steroid deficiency, tivity of BAT in REDs to preserve energy, but these data
GH resistance, hypercortisolism, mainly from hypoleptine­ remain to be confirmed, especially given that BAT is extremely
mia and disturbances in other adipokines and the AFI axis) low to start with in normal adults. BMAT has also been impli­
may all directly and indirectly contribute to increased bone cated in energy regulation, bone health, and hematopoiesis,
resorption and low BMD. Additional SF risk factors among and its role as a mediator in REDs-related disorders is
athletes and military personnel include pronounced mechan­ emerging.
ical stress without sufficient recovery (120).
Early case reports in athletes indicate that the most common
injuries are localized in the tibia (49.1%), tarsals (25.3%), and Leptin
metatarsals (8.8%), as well as the femur, fibula, and pelvis
(243). Furthermore, low weight and BMD have been associ­ Leptin physiology
ated with a higher risk of bone stress injuries in trabecular-rich Leptin is a hormone secreted by adipose tissue. It is an adipo­
compared with cortical-rich locations (121). Military person­ kine that plays a crucial role in regulating energy metabolism
nel is also heavily burdened by SF reaching a cumulative and adipose tissue. Leptin helps maintain the overall physio­
incidence of 5.69 per 1000 person-years in large-scale epi­ logical balance of the body and regulates important functions
demiological studies (122). These numbers differ between related to metabolism (252). The circulating levels of leptin
men and women (reportedly 2.05 vs 7.47 per 1000 person- display sexual dimorphism, being higher in women than in
years in male and female soldiers, respectively, representing men, and reflect mainly acute changes in energy intake (with
>5.2 million person-years of risk (123); other reports delin­ a precipitous drop in acute energy deprivation) as well as long-
eate an incidence of 19.3 vs 79.9/1000 recruits respectively term energy stores (with decreasing levels reflecting LEA).
in general SF (124) and a prevalence of 2.76 vs 5.78 per Leptin binds and activates its receptors not only in the brain
1000 person-years respectively for ankle-foot complex SFs) but also in peripheral organs, thus mediating the effects of dys­
(125). Apart from epidemiology, soldiers are known to dis­ regulated energy homeostasis in most REDs-related hormonal
play elevated levels of bone turnover markers and parathor­ subsystems (Fig. 3) (1, 27, 37, 253). Therefore, leptin is a re­
mone, which are, however, higher in male soldiers (119) liable indicator of energy availability and, by extension, en­
who similarly display a lower total bone mineral content ergy deprivation in animals and humans (47, 62, 253).
(BMC) compared with female counterparts (126). SFs impose In humans, the leptin gene is expressed primarily in WAT and
an estimated cost of $100 million annually to the US military, in much lower concentrations in other tissues such as the stom­
not including lost duty (244). ach, placenta, mammary gland, and the immune system (254).
More prolonged (58) studies with larger sample sizes are Leptin displays a pulsatile pattern of secretion, which is notably
needed to refine the dose-response relationships between accentuated in women and produces elevated basal leptin con­
chronic restrictions of EA and bone turnover and the under­ centrations in the female sex (255). On a molecular level, leptin
lying mechanisms and determine the optimal nutritional inter­ can cross the blood-brain barrier through a facilitated system
ventions to restore bone health (117). and directly (256) binds in the brain but also in other organs
to its receptor LEPR, which belongs to the JAK-STAT family
of transcription factors, primarily in the hypothalamus and
Pathophysiology and Mechanistic Pathways
more specifically in areas such as the arcuate nucleus of the
Underlying REDs-associated Disorders hypothalamus (256-258). In the context of neuroendocrine
In states of LEA, the body strives to preserve the function of sys­ regulation, falling leptin levels in the context of energy depriv­
tems necessary for immediate survival, diverting energy from ation activate orexigenic NPY/agouti-related protein neurons
nonessential systems (eg, reproduction). From an endocrine (258) and inhibit anorexigenic proopiomelanocortin activity
point of view, exercise and inflammation both induce and are in the brain, thus constituting the afferent signal of an
influenced by various hormonal changes, particularly in adipo­ AT-mediated negative feedback loop regulating energy avail­
kines, which constitute the main cellular and metabolic signals ability by influencing food intake and altering physiology (37,
of AT and are prime regulators of energy balance (245). 259). Leptin regulates the hypothalamic-pituitary-peripheral
Endocrine Reviews, 2024, Vol. 00, No. 0 13

hormonal axes, as mentioned previously, and has secondary ac­ Disruptions in circulating leptin and reproductive hormone
tions in peripheral tissues, including the immune system, bones, profiles are evident in LEA and have been observed in healthy
muscle, and adipose tissue, all aiming at maintaining energy individuals after short-term starvation (88, 275). In 8 healthy
homeostasis under normal physiological conditions but leading males, 72 hours of fasting altered LH pulsatility and markedly
to pathophysiological abnormalities and diseases such as SF decreased testosterone. Concomitant r-metHuLeptin admin­
when the physiologically adaptive processes last longer than istration maintained normal LH pulsatility and testosterone
they should. levels (42). In a pilot study of 14 adult females with hypo­
Mice with deleted leptin genes, now identified as the ob/ob thalamic amenorrhea resulting from vigorous exercise or
phenotype, as well as humans with similar genetic defects, are low weight, the administration of recombinant leptin
innately obese and display impaired thermogenesis, dimin­ (r-metHuLeptin) appeared to improve reproductive func­

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


ished energy expenditure, and endocrine and metabolic ab­ tion. In particular, women who received r-metHuLeptin
normalities (246, 260). Leptin replacement in these mice demonstrated increases in LH pulsatility, estradiol concen­
and humans reduces fat mass without affecting lean body trations, ovarian activity, and the number and size of follicles
mass and improves neuroendocrine function and metabolism (46). Another randomized placebo-controlled trial in women
(261). In addition, females tend to have increased leptin pro­ with HA, 11 of whom were assigned to r-metHuLeptin and 9
duction and pulse amplitude, potentially indicating an innate to placebo, reported elevated estradiol and progesterone lev­
relative leptin resistance (255, 262). However, in LEA, short- els in the leptin-treated women, in addition to improvements
term fasting induces a profound and rapid decrease in serum in other neuroendocrine abnormalities after 36 weeks (45).
leptin concentrations, potentially through increased leptin Treatment with r-metHuLeptin resulted in the resumption
clearance (263), which precede and are disproportionate to of menses in a significant proportion of adhering participants
changes in fat mass (42, 44, 264). Moreover, the weight loss (114). During the entire study period, the r-metHuLeptin
attained during fasting with concomitant leptin administra­ dose was reduced in 5 of 11 metreleptin-treated participants
tion in physiologic, supraphysiologic, or pharmacologic doses who lost more than 5% of their baseline body weight, where­
is independent of dose or circulating leptin levels achieved as 2 of the participants withdrew from the study because of
(263). Long-term energy deprivation through caloric restric­ weight loss when circulating leptin levels exceeded the
tion likewise results in markedly downregulated leptin con­ physiologically normal range. Besides local injection site re­
centrations (265). Women with DE and amenorrhea are also actions developed by 1 participant, no other clinically signifi­
known to have reduced circulating leptin levels and blunted cant drug-related adverse events were documented (114).
pulses (107, 266, 267). In this energy-deficient milieu of low Thus, leptin administration at replacement doses, sufficient
basal leptin, human studies show notable neuroendocrine to maintain circulating leptin levels within the normal range,
benefits in response to both physiological and supraphysiolog­ normalizes neuroendocrine axes impairments from energy
ical doses of r-metHuLeptin (42, 45-47, 114) (Fig. 2). deprivation. However, when leptin is administered in supra­
physiological doses, the side effect of further suppression of
fat mass and weight loss is observed (apparently as part of a
Neuroendocrine effects of leptin feedback loop to limit the endogenous contributions to circu­
The pivotal role of leptin as key regulator of neuroendocrine lating leptin when the exogenously administered leptin is too
axes, whose dysfunctions constitute components of REDs, is high).
physiologically distinct between animals and humans.

Hypothalamic-pituitary-thyroid axis
Hypothalamic-pituitary-gonadal axis Leptin stimulates thyrotropin-releasing hormone production
Leptin stimulates GnRH release from incubated neuronal cells in hypothalamic cells of fasted rodents (276) and prevents
and accentuates GnRH pulsatility, but not amplitude, in the fasting-induced downregulation of TSH pulsatility and
hypothalamic neurons (268). Low leptin concentrations, com­ T4 secretion, albeit partially (271). In humans, LEPR muta­
mon in LEA, downregulate GnRH secretion from the hypo­ tions result in low T4 levels but normal basal TSH levels
thalamus, through either direct action or indirect changes in and sustained TSH responses to thyrotropin-releasing hor­
neuropeptide systems, such as neurokinin or kisspeptin- mone (277). In leptin-deficient children, leptin replacement in­
mediated pathways (269). creased free T3 and free T4 concentrations (273).
Ob/ob mice, which are sterile, regain reproductive capacity In lean healthy men, r-metHuLeptin administration in re­
following leptin administration (270). In calorically deprived placement doses prevented the energy derivation-induced sup­
mice with low leptin levels, leptin administration has also been pression of TSH pulsatility and increased the concentrations of
shown to reverse the LEA-induced reductions in LH and tes­ free T4 but did not affect T3 and reverse T3 (42). However, in
tosterone (271). Likewise, r-metHuLeptin replacement stimu­ 7 women following a similar protocol, r-metHuLeptin did not
lated gonadotropin pulsatility, culminating in normal LH and prevent fasting-induced fluctuations in thyroid hormones (44).
FSH secretion in a seminal study on a child with congenital Women with HA receiving high-dose r-metHuLeptin for 3
leptin deficiency (CLD) (272) and another case report of leptin months had increased TSH pulse frequency and amplitude
replacement in a female child with CLD for 24 months ob­ and transiently increased free T3 and free T4 levels (46),
served restoration of LH pulsatility and pubertal develop­ whereas treatment over 36 weeks resulted in increased free
ment, including menarche (273). In a case series of 3 adults T3 but not free T4 or TSH levels, compared with placebo
with CLD, leptin replacement increased LH and testosterone (45). Thus, although the precise effects of leptin on the thyroid
levels, associated with the development of secondary sex char­ axis remain to be fully elucidated, findings to date are consist­
acteristics, in a male patient and induced ovulation in 2 female ent with the hypothesis that fasting-induced thyroid hormone
patients (274). changes are mediated through falling leptin levels due to
14 Endocrine Reviews, 2024, Vol. 00, No. 0

energy deprivation. It is however established that among over­ body temperature, resting heart rate, blood pressure, or urin­
weight or obese individuals, who are leptin resistant, long-term ary catecholamine levels (285).
leptin replacement therapy to push circulating leptin levels to
even higher supraphysiologic levels does not produce any
changes in circulating TSH or any of the active thyroid hor­ Immune and hemopoietic systems
mones, both total and free, indicating a neuroendocrine milieu LEPR is expressed in almost all immune cells (286, 287), and
of leptin resistance (278). its long form, which is considered to be fully capable of JAK/
STAT signaling, is expressed on natural killer cells and acti­
Hypothalamic-pituitary-somatotropic axis vated T-lymphocytes (287). Incubation with leptin has been
shown to activate B cells and induce the expression of cyto­
Incubation of human pituitary cells in leptin increases GH secre­

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


kines and signaling molecules in macrophages and mono­
tion (279). and in mice, leptin prevents the fasting-induced sup­
nuclear cells (288). When administered in fasted mice, leptin
pression of GH and IGF-1 and partially corrects GHRH mRNA
reversed the starvation-induced suppression of T-lymphocyte
expression (280). In adults with CLD, r-metHuLeptin, in
responses by increasing Th1 cells and downregulating Th2
physiological replacement doses, elevated IGFBP-1 and 2, but
cytokine production (289). In humans, leptin administration
not IGF-1 (274).
in children with congenital leptin deficiency boosted T-cell re­
When administered in healthy men (42) or women (44)
sponsiveness and increased CD4+ T-lymphocytes (273), and in
undergoing a 72-hour complete energy deprivation, leptin
adults it boosted signals that stimulate chemotaxis and in­
in replacement doses does not markedly influence GH pulsa­
crease CD4+ cell counts (290, 291).
tility or IGFBP levels but blunts the starvation-induced de­
In a study of 14 women with HA (6 treated with placebo and
crease (104) of total, but not free, IGF-1 secretion. In
8 with replacement-dose r-metHuLeptin for 36 weeks) and 13
contrast, in a 3-month pilot study of women with HA treated
healthy controls, individuals with HA had lower basal leptin
with r-metHuLeptin, IGF-1 increased in the first month and
levels and T-cell counts compared with controls. Treatment
returned to baseline levels, whereas IGFBP-3 was markedly in­
with r-MetHuLeptin improved CD4+ cell counts and their
creased during months 2 and 3 (46). A longer study of female
proliferative responses following in vitro stimulation. Leptin
athletes with HA also found that r-metHuLeptin treatment for
additionally boosted the expression of cell survival and hormo­
36 weeks significantly increased total IGF-1 and tended to ele­
nal response genes and blunted apoptosis-related genes.
vate free IGF-1 and IGFBP-3 levels compared with controls
Finally, leptin activated cell proliferation and growth path­
(104). These changes in women with HA are evident with
ways within CD4+ T-lymphocytes, such as AMPK, STAT3,
up to 2 years of r-metHuLeptin treatment (114). Overall, lep­
and mTOR (291).
tin administration seems to have beneficial effects on the
Leptin is also known to promote the proliferation, differen­
GH-IGF1 axis in states of long-term energy deficiency rather
tiation, and activation of marrow hemopoietic cells, induce
than short-term starvation.
bone marrow colonies, and interact with granulocyte precur­
sors (292-294). More recently, the presence of LEPR on hem­
Hypothalamic-pituitary-adrenal axis
atopoietic stem cells was identified as an indicator of
LEA increases ACTH and corticosterone in mice, and leptin hematopoietic stem cell engrafting quality and capacity (295).
administration inhibits corticotropin-releasing hormone
from hypothalamic cells and reverses the LEA-induced activa­
tion of the HPA axis (281). However, humans with LEPR mu­ Bone Health
tations do not present with HPA abnormalities (277), Loss of BMD constitutes 1 of the prime characteristics of the
indicating a key interspecies difference. Female Athlete Triad and is a pivotal feature of REDs. The sta­
Short-term pharmacological leptin administration in tus of bone tissue is a direct reflection of human nutritional
healthy fasted lean men (42) and women (44) does not affect status, including but not limited to calcium and vitamin D in­
fasting-induced changes in plasma or urine cortisol concentra­ take, and is regulated by PTH and other hormonal conduits,
tions, cortisol pulsatility, or the HPA axis overall. Three including leptin, particularly in energy-insufficiency states
months of r-metHuLeptin treatment in women with HA failed (296) (Fig. 4).
to induce changes in cortisol or ACTH levels (46); however, When elucidating the inner workings of bone regulation, it
long-term administration of replacement-dose leptin of 36 is important to mention the roles of receptor-activator of nu­
weeks to 2 years in women with HA results in marked declines clear factor κΒ ligand (RANKL) pathway signaling, osteopro­
in cortisol levels (42, 45, 46, 114). Overall, leptin administra­ tegerin (OPG), and sclerostin, among others, in regulating the
tion seems to have beneficial effects on the ACTH-cortisol axis osteoblast-osteoclast equilibrium. Osteoclasts, responsible
in states of long-term energy deficiency rather than short-term for bone resorption, are activated through RANKL ligand
starvation. signaling, which is naturally inhibited by OPG, itself secreted
by osteoblasts (297). On the other hand, sclerostin, which is
Sympathetic nervous system also produced by osteocytes, binds to low-density lipoprotein
Leptin also differentially affects components of the sympa­ receptor-related protein receptors, which are involved in
thetic nervous system (heart rate, blood pressure, catechol­ WNT signaling and the regulation of bone mass (298).
amines) in animals and humans. In mice, leptin increases Normally, WNT activation induces osteoblast activation,
BAT sympathetic tone in the dorsomedial hypothalamus whereas sclerostin inhibits lipoprotein receptor-related pro­
and activates sympathetic nerves (282-284). However, leptin tein and WNT signaling, thus reducing bone formation
administration in short-term studies in healthy lean men and (298). Sex steroid hormones and estrogen directly regulate
women and long-term studies in women with HA do not dem­ OPG and RANKL production (299) and may influence
onstrate any significant changes in resting metabolic rate, RANKL-induced osteoclast differentiation (300), whereas
Endocrine Reviews, 2024, Vol. 00, No. 0 15

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


Figure 4. Functions and pathways of leptin in bone health. Arrows with plus signs indicate positive relationships or feedback; arrows with minus signs
indicate negative feedback. Physiologically, RANKL is critical for the differentiation of osteocytes into osteoclasts and is inhibited by osteoprotegerin
(OPG). Meanwhile, sclerostin binds to LRP receptors and antagonizes WNT signaling, which is key for osteoblast activation. Leptin’s role in bone ph­
ysiology can be discerned into indirect and direct. Leptin indirectly influences bone status by producing changes in hormone levels such as estrogens and
growth factors, especially IGF-1. Studies in animals and humans receiving leptin have demonstrated positive effects on bone mass through downre­
gulation of bone resorption and increased osteoblast formation and activity. Leptin may also act through CART to deactivate osteoblasts while it add­
itionally increases osteoprotegerin expression, which binds RANKL and thus deactivates osteoclasts in a manner similar to estrogen. This is evident in
human studies showcasing a downregulation of CTX, a marker of bone catabolism. On the other hand, leptin upregulates a series of important bone
formation factors, including TGF-B, collagen 1a, bone-specific alkaline phosphatase, and osteocalcin. Leptin may additionally influence PTH secretion,
while it also regulates vitamin D metabolism and activates FGF-23 in animal models, which is central to bone and mineral metabolism, yet does not impact
sclerostin, and only tends to increase osteoprotegerin (OPG) and decrease RANKL levels in humans. Intracellularly, leptin drives osteoblast proliferation
through cyclin D1, activated through the same molecular clock the hormone oversees. Overall, leptin demonstrates a tendency to improve cortical bone
integrity and thus may pinpoint a strategy to reduce SF risk in athletes and military personnel. Abbreviations: AP-1, activator protein 1; BAP, bone-specific
alkaline phosphatase; CART, cocaine and amphetamine regulated transcript; Col1A, collagen type I alpha 1; CTX, C-terminal telopeptide of collagen; CY,
cytochrome; E2, estradiol; FGF23, fibroblast growth factor-23; LPR, low-density lipoprotein receptor-related protein; OPG, osteoprotegerin; RANKL, r­
eceptor activator of nuclear factor kappa-Β ligand; VDR, vitamin D receptor.

in animals, estrogen targets and suppresses RANKL expres­ whereas conflicting results are present in long-term exercising
sion in bone lining cells (301). cohorts, even displaying decreasing trends (306). On the other
High cortisol may additionally directly lead to increased hand, vitamin D deficiency is a frequent occurrence in athletes
bone resorption by means of reducing OPG and increasing and often correlated with increased fracture risk (307), pre­
RANKL expression, promoting osteoclast survival and de­ cipitating therapeutic recommendations for supplementation
creased bone formation via inducing osteoblast apoptosis, in amenorrhea with osteoporosis.
and by preferentially driving BM stromal cell differentiation Estrogen promotes osteoblast proliferation and blocks
to adipocytes over osteocytes (253, 302-304). Moreover, cor­ osteoclast differentiation (308). Thus, hypoestrogenism re­
tisol concentrations have been positively correlated with colla­ sults in increased bone resorption with a relative deficit in
gen type 1 C-telopeptide (CTX), a bone resorption marker, bone formation and is associated with low BMD (309-312).
and inversely associated with LH pulsatility and procollagen Furthermore, androgens play important antiresorptive and
type 1 N-terminal propeptide (P1NP), a marker of bone colla­ anabolic roles in bone for men and women by decreasing os­
gen deposition (110). teoclastogenesis and stimulating osteoclast apoptosis while in­
PTH aims to maintain and mobilize circulating calcium hibiting osteoblast apoptosis (313).
from the bones, kidneys, and gut, while reducing the renal re­ Low IGF-1 levels and activity are also culprits of compro­
absorption of phosphate. Vitamin D, on the other hand, stim­ mised BMD in LEA. IGF-1 has been positively correlated
ulates both calcium and phosphate metabolism, providing with bone mass (314) and shown to stimulate osteoblast pro­
adequate minerals for bone formation (305). In exercising hu­ liferation and activity (315), is considered essential for the dif­
mans, short-term exercise often increases circulating PTH, ferentiation of mesenchymal osteoprogenitors (316), has been
16 Endocrine Reviews, 2024, Vol. 00, No. 0

associated with bone resorption, formation, and healing, and trabecular volume and BMD in axial bones, but diminished
linked, alongside GH administration, to rapid clinical im­ length and mineralization, along with increased marrow
provements in tibial fractures (317). IGFBPs also possess an adiposity, in the peripheral skeleton (322, 336).
important anabolic role within the skeleton by directing the Intracerebroventricular leptin administration in ob/ob mice
actions of IGFs and particularly IGF-1 within bone tissue inhibits bone formation and stimulates bone resorption
(314). (284) to reduce vertebral trabecular bone volume but increase
The HPA axis, which responds to stress and LEA by excess femur length and bone volume, normalizing the bone pheno­
cortisol production, influences bone health as well. Cortisol type (322). In several animal models, leptin administration
can inhibit osteoblast and osteoclast growth, hinder the ana­ exerts positive effects on bone mass except at markedly ele­
bolic effects of growth hormone, and suppress IGF-1 synthesis vated doses, which may decrease body weight (49, 322, 337).

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


in skeletal cells (99, 318-320). Excess cortisol is also known to A retrospective analysis of 1193 Japanese postmenopausal
suppress the HPG axis, which is pivotal for bone regulation women found that low leptin concentrations were associated
(12, 26, 36, 321). with a higher risk of long-bone osteoporotic fractures (hazard
ratio 0.70) (48), confirming the results of an older report in
men and women (51). Leptin replacement in leptin-deficient
Role of leptin in bone health individuals may improve bone health directly and indirectly
Leptin serves as a link between AT and bone both directly by via restoration of the HPG axis, increased IGF-1 activity,
stimulating osteoblasts, chondrocytes, and BM stromal cells and decreased cortisol concentrations (46). In a randomized
and indirectly through alterations in neuroendocrine activity controlled trial of 19 women with HA, 36 weeks of
(49, 322). At the molecular level, leptin regulates osteoblasts r-metHuLeptin treatment upregulated osteocalcin levels, a
by upregulating Ap-1 gene expression, which activates osteo­ marker of bone formation, and prevented an increase in urin­
blast proliferation through cyclin D1 (323). Leptin has also ary N-terminal telopeptide-to-creatinine ratio, a marker of
been shown to promote osteoblast differentiation by upregu­ bone resorption, compared with placebo (45). Lumbar BMC
lating TGF-β, IGF-1, collagen-1α, alkaline phosphatase, and and BMD improved with r-metHuLeptin treatment (114).
osteocalcin mRNA in human iliac crest osteoblasts (324). The extension of treatment in 6 subjects for 1 additional
Osteocalcin, in particular, is known to be affected by leptin year increased BMC by 1.4% to 6.5% and BMD by 2.2%
via the hypothalamus and constitutes a notable orchestrator to 10.8% from baseline at the lumbar spine and maintained
of bone health, insulin sensitivity, and energy balance (325, low levels of CTX, another marker of bone resorption
326). Moreover, leptin may also promote osteoblastogenesis (114). These r-metHuLeptin-treated women with HA also
by upregulating CYP27B1, CYP27A1, and VDR, which are had increases in estradiol concentrations and resumed menses,
mediators in vitamin D metabolism (327). In rodent models, which would also contribute to decreased bone resorption
leptin also increases FGF-23 expression in bone, leading to (114). In another randomized placebo-controlled trial, leptin
suppression of renal 1α-hydroxylase expression (328). It is administration in women with hypothalamic amenorrhea
now accepted that leptin represses osteoclast differentiation for 36 weeks did not influence circulating sclerostin or
through cocaine- and amphetamine-regulated transcript and FGF23 levels but markedly downregulated intact PTH and
acts in a similar way to estrogen, increasing OPG levels, which tended to downregulate serum RANKL and increase OPG,
binds to RANKL (329) and thus blunts osteoclast activity. while also significantly decreasing the RANKL-to-OPG ratio
Finally, leptin also exerts beneficial effects on bone through (330), indicating reduced bone resorption and osteoclastic ac­
an overall amelioration of metabolism and neuroendocrine tivity, which are beneficial to bone integrity.
health (49). In lean hypoleptinemic women, r-metHuLeptin
administration for 36 weeks markedly downregulated intact
PTH and RANKL to OPG ratio did not alter sclerostin, The AFI Axis
dickkopf-1, and FGF-23 levels, but it nevertheless tended to Leptin significantly influences the energy-consuming process of
increase serum OPG and decreased serum RANKL (330). reproduction, as attested by the downregulation of the HPG
This indicates differences in mechanisms of action between axis in energy-deprived hypoleptinemic states, leading to HA
humans and animal models. and infertility (43). However, only 60% of patients with HA
Low leptin levels have also been observed among female improve their reproductive function with leptin replacement,
athletes with elevated bone turnover as indicated by levels of suggesting the presence of additional regulators. We have pro­
CTX (331). Even though early animal models displayed in­ posed that the AFI axis may have a role in reproduction, as well
verse relationships with leptin concentrations and BMD, re­ as muscle mass and metabolic processes, in energy-deficiency
porting predominantly antiosteogenic pathways through the states, that is independent of leptin (43, 338) (Fig. 2).
central hypothalamus as a result of intracerebroventricular Activins and inhibins are members of the TGF-β superfamily,
leptin infusions (284, 332), such pathways are not activated chiefly functioning through SMAD signaling. They are impli­
in humans (42, 114). Although leptin levels have not been cated in reproduction, muscle tissue growth and differentiation,
linked to estrogens in healthy individuals with sufficient leptin and energy metabolism (339). Under normal circumstances, ac­
levels (333), leptin has an intricate relationship with the regu­ tivins and inhibins bind to their receptors on pituitary, ovarian,
lation of sex steroid hormone production through its para­ and testicular cells. Principally, inhibins suppress whereas acti­
mount influence on the HPG and other axes, particularly vins upregulate FSH production from the anterior pituitary,
when circulating leptin levels are low as in cases characterized thus maintaining an equilibrium (339-341). Activins also in­
by acute or chronic energy deprivation (334, 335). duce muscle atrophy and limit muscle mass alongside myosta­
In vitro, leptin directly activates osteoblasts, chondrocytes, tin (53, 342).
and bone marrow stromal cells and promotes osteoblast dif­ Follistatin and follistatin-like 3 (FSTL3), which increase
ferentiation (49). Leptin-deficient mice present with increased with LEA, suppress the bioactivity of activins by irreversibly
Endocrine Reviews, 2024, Vol. 00, No. 0 17

binding to them (339, 340). By neutralizing activins’ role in re­ energy expenditure to improve EA for the prevention and
production, follistatins shunt energy away from the repro­ treatment of the clinical consequences of LEA and therefore
ductive system (43, 54, 340). Follistatins also block the of the Female and Male Athlete Triad and REDs.
catabolic function of activins and myostatin to preserve The IOC panel of experts also highlights the need for edu­
muscle mass (339, 341). cational programs on nutritional options as well as the risks
Follistatins also mediate liver metabolic processes, lipid and consequences in health from inadequate dietary patterns
homeostasis, and glucose metabolism (54). FSTL3 knockout (26). Besides, they describe the REDs clinical treatment as
mice have reduced visceral fat, decreased insulin resistance, in­ based on realistic health-promoting goals for weight and
creased pancreatic islet number and size, and improved glu­ body composition (26). Patients are usually advised to in­
cose tolerance (343). In humans, follistatin concentrations crease their daily calorie intake by 300 to 600 kcal (26) or

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


are inversely correlated with insulin sensitivity and positively to set energy intake at a minimum of 2000 kcal/day, depend­
associated with body mass index, fat mass, lipid profiles, and ing on exercise energy expenditure (14), but there is no estab­
blood pressure (43, 53, 54, 111). lished protocol for gradually increasing calorie consumption
Activin A and B concentrations are elevated in physically (26). In addition, it is suggested to restore body weight at lev­
active individuals (without known LEA), and exercise tends els that are correlated with the resumption of regular men­
to increase the levels of activins, but also follistatin, which strual cycles (14). It has been observed that resumption of
acts as a “binding protein” for activins and FSTL3 (53). In menses may occur after achieving approximately 90% of the
contrast, acute energy deprivation by fasting in healthy males ideal body weight for age and height (348). However, the
results in decreased activin A and increased follistatin concen­ timeframe for recovery typically spans several months, al­
trations (37). In the context of chronic energy deficiency, though it can vary significantly and may even extend to years
women with HA also have higher follistatin levels and lower in some cases (348, 349).
circulating levels of activins but lower FSTL3 levels compared Moreover, it is crucial to emphasize that the relationship be­
with healthy women (43). These changes would lead to sup­ tween body weight gain and the restoration of menstrual func­
pression of reproduction, preservation of muscle mass, and in­ tion is not a linear process, and additional factors may play a
sulin resistance, mainly at the level of the liver, which would role (350). Recovery can be complex, particularly in the con­
lead to higher availability of circulating levels of glucose and text of REDs, as intense physical activity may lead to the per­
lipids (ie, energy sources). Importantly, 72-hour crossover ex­ sistence of amenorrhea (6, 30). Therefore, it is essential to
periments in lean men and women, as well as long-term acknowledge both the complexity and individual variability
placebo-controlled leptin replacement in women with hypo­ in the resumption of the menstrual cycle while emphasizing
thalamic amenorrhea attaining resumption of menses and re­ the importance of personalized goals and targets in clinical
ductions in body fat, have shown that LEA-induced practice to facilitate menstrual function recovery. This
alterations of the AFI axis are not influenced by leptin admin­ highlights that achieving a specific weight is not a universal so­
istration and thus mostly function independently of leptin (53, lution, and a “1-size-fits-all” approach may not be appropri­
54, 111, 338, 344) (Fig. 2). ate and adjustments may be required over time.
Besides observational data (351), recently, the first random­
ized controlled trial (ie, REFUEL study) showed that increased
Management of REDs energy intake 20% to 40% above baseline energy require­
The management of REDs should be multidisciplinary and ments was associated with an improved menstrual function
undertaken by a team of sports and exercise professionals, in­ in exercising women with oligomenorrhoea or amenorrhoea
cluding nutritionists, physicians, psychologists, psychiatrists, (352). It is also important to focus on diet quality, ensuring
physiotherapists, and physiologists. However, REDs is often a proper balance of macronutrients and appropriate
misdiagnosed by health professionals, coaches, and athletes, consumption of micronutrients (such as iron, calcium, and
with studies demonstrating insufficient education, poor man­ vitamin D) (14). In particular, vitamin D and calcium supple­
agement, and prioritization of performance over health (345- mentation, as needed, may promote bone health and prevent
347). Initial recommendations start with specialized dietary SF with a potential benefit during the recovery phase (127,
plans, nutritional education, and supplementation, followed 296, 353-356), whereas an optimal daily intake of 1000 to
by mitigation of exercise, before proceeding with specialized 1300 mg of calcium and vitamin D concentrations within
hormonal or pharmaceutical treatments. the range of 32 to 50 ng/mL are recommended (14).
Additional factors such as the diversity of food choices, indi­
vidual taste, and the practicality of food availability (practical
Nonpharmacological Strategies aspects of food availability) should be considered while de­
Nonpharmacological therapy is the preferred mainstay strat­ signing a meal plan (14). As a result, potential challenges of
egy, which frequently results in the successful resolution of correcting the underlying LEA by altering energy intake and/
most cases. The treatment of REDs should involve a multifa­ or exercise energy expenditure in individual athletes or sol­
ceted approach, taking into account the complexity and indi­ diers (from difficulties in changing the antecedents that under­
vidual variability in each athlete’s nutritional needs and pin LEA or noncompliance) should also be acknowledged.
recovery processes. Therefore, it is crucial to recognize the sig­ Finally, the European College of Sport Science and the
nificance of addressing the underlying cause of REDs rather American College of Sports Medicine consensus also highlight
than solely focusing on treating its symptoms. Current con­ resting, adequate sleeping, adjusting dietary patterns, and de­
sensus statements and practice guidelines (6, 14, 19, 35) em­ creasing training as important treatment factors (357).
phasize nutritional, behavioral, and lifestyle changes, Regarding eating disorders, treatments for medically stable
including an appropriate exercise regimen and the importance and unstable patients differ but also require multidisciplinary
of increasing dietary energy intake and/or reducing exercise treatment. Although cognitive behavioral therapy is a first-line
18 Endocrine Reviews, 2024, Vol. 00, No. 0

treatment option, each patient evaluation should consider the 12 months of transdermal estradiol treatment, compared
need for an inpatient hospitalization, outpatient care, or day with oral estradiol or no treatment (368). Unlike transder­
programs based on their stability, associated complications, mal estrogen, oral formulations of estrogen can decrease
and comorbidities (358). Specifically, for anorexia nervosa, IGF-1 activity (369), reduce free testosterone concentrations
it can be attempted to be treated with cognitive behavioral (by increasing SHBG levels), and as a result, the potential
therapy, which has been expanded to females with HA and anabolic testosterone-induced effects on bone (370, 371).
shown to normalize leptin, TSH, and cortisol concentrations The Endocrine Society suggests against using oral contra­
(probably because body weight and fat mass increased) ceptives to improve BMD if the underlying energy deficit is
(353, 359). More precisely, its first-line management should not addressed (78). However, the 2014 Female Athlete
include weight restoration with nutritional rehabilitation. It Triad Coalition recommended considering transdermal es­

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


should include restoring a structured meal routine, supervised tradiol if BMD Z-scores are ≤−2.0, there are ongoing risk
meals, and psychotherapy to address dysfunctional thoughts factors, and an adequate response has not been achieved
and behaviors (358, 360). Of note, nutritional repletion might after 1 year of lifestyle modification (ie, BMD loss or new
restore several related endocrine abnormalities but is not com­ fracture) (14).
monly sufficient by itself (361, 362). PTH analogs are used for the treatment of osteoporosis and
Several ongoing (Table 2) and recently completed trials are extremely effective at reducing vertebral fractures.
have investigated risk assessment for REDs, as well as Teriparatide, an injectable synthetic recombinant human
pharmacological and nonpharmacological lifestyle modifica­ parathyroid hormone analog, has been shown to markedly in­
tion strategies for REDs and related disorders. One study en­ crease BMD in women with anorexia nervosa after 6 months
rolled 481 female college athletes who were assigned to a of treatment (372) and is stipulated by the Endocrine Society’s
comprehensive lifestyle modification termed the Female guidelines for HA-associated delayed fracture healing and low
Athlete Body Project, a behavioral eating disorder risk factor BMD (78). Two ongoing clinical trials (Table 2) are currently
reduction program, or a control arm, and found benefit in investigating its use in military populations to prevent SF.
body perception-related outcomes through the Female Romosozumab, an anti-sclerostin antibody, has proven
Athlete Body Project (363). Under the same principles, the fracture reduction benefits in postmenopausal women with
Male Athlete Body Project has also been investigated up to 1 osteoporosis and improve bone profiles in men, albeit with
month follow-up (364); yet, these studies chiefly pertain to some potential cardiovascular side effects, chiefly myocardial
DE and do not specifically address outcomes on endocrine infarction and stroke (373). Currently, there is only 1 small
REDs markers. Another more recent study implementing a trial examining BMD changes in premenopausal women
food and nutrition learning program intervention of online with idiopathic osteoporosis (NCT04800367). Denosumab
lectures applied to a multinational cohort alongside a control has also been studied for 12 and 24 months in 32 women
intervention, demonstrated improvements in REDs symp­ with premenopausal idiopathic osteoporosis following a
toms, particularly menstrual function and, to a lesser extent, treatment course of 24-month teriparatide, attaining marked
gastrointestinal issues, in athletes who underwent the FUEL increases in lumbar, hip, and femoral BMD (374). These med­
intervention, with these benefits persisting up to 12 months ications have yet to be studied in energy-deficient cohorts of
after intervention (365). Another study in healthy females athletes or soldiers. Moreover, it should be noted that the
under low energy availability led to decreased P1NP levels in­ Endocrine Society does not recommend bisphosphonates, de­
dicative of bone resorption, which were prevented through the nosumab, testosterone, or leptin for bone health based on lim­
incorporation of high-impact jumping, indicated by stable ited evidence, and these therapies are only reserved for
β-CTx levels, suggesting a protective effect on bone health individuals who have failed or have contraindications to es­
(366). On the other hand, in male cyclists, 4 weeks of intensive trogen replacement. Recombinant parathyroid hormone is
endurance interval training was shown to improve aerobic suggested in cases of delayed fracture healing and markedly
performance and testosterone but produced reductions in low BMD (78).
RMR, T3, and increases in cortisol, all of which constituted Given the reduced GH activity associated with LEA, GH
risk markers of REDs (367). and IGF-1 therapies have been tried in women with anorexia
nervosa. Although treatment with GH does not seem effective
because of GH resistance, recombinant IGF-1 treatment with
Pharmacological Strategies oral contraceptives has been shown to modestly increase spine
Targeting underlying causes of LEA incorporating nutritional, BMD in women with anorexia (195, 375). Growth hormone
behavioral, and lifestyle alterations is the front-line treatment secretagogues are currently being investigated, but not in co­
approach. However, pharmacological management could be horts with REDs. LUM201, also known as ibutamoren or
considered, especially in specific cases regarding the treatment MK-677, is a GH secretagogue currently being investigated
of problematic LEA-related symptoms. The nonpharmacolog­ for pediatric GH deficiency (NCT04614337). MK-677 has
ical approach should continue along with the initiation of been shown to increase serum levels of GH, IGF-1, and
pharmacological treatment. IGFBP-3, as well as lean body mass in a 2-month study in
Despite recommendations for treatment of FHA with estro­ obese patients (376). MK-677 additionally increased both
gen with or without calcium and vitamin D, a recent meta- markers of bone formation and resorption, elevating CTX
analysis on estradiol treatment in premenopausal women by 23% and procollagen III by 28% in early stages of treat­
with FHA and low BMD found no significant improvements ment and osteocalcin at later stages (377). A randomized con­
in lumbar BMD overall, though there may be a benefit with trolled trial in 165 older adults with hip fractures found
transdermal estradiol (309). A recent randomized controlled marked increases in IGF-1 levels but no improvements in func­
study in young, amenorrheic female athletes demonstrated ro­ tional or performance metrics (378). In another trial of elderly
bust improvements in spine, femoral neck, and hip BMD with patients with hip fracture, MK-677 was found to improve gait
Endocrine Reviews, 2024, Vol. 00, No. 0 19

Table 2. Ongoing studies, as of November 2023, investigating the pathophysiology and treatment of LEA-, REDs-, and the Female Athlete Triad-associated
conditions in athletic, training, healthy, and energy-deficient populations

Registered trials investigating prevalence, risk or mechanisms, and characteristics of LEA, REDs, and the Female Athlete Triad

Identifier Design Participants Arms Outcomes Time frame

NCT05649267 Prospective/ 10 Judoka athletes, 1. Year 1—Energy deficit, 1. Body weight and composition including 3y
cross-sectional aged 18-30 y, dietary risk BMD
training ≥4 times/ 2. Years 2 and 3—effect of 2. Food intake
week for at least 2 h carbohydrates and protein

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


on function and
performance
NCT04821076 Prospective Females, aged 18-30 Trained population: 1. Changes in muscle protein synthesis 10 d
RCT y, BMI 18.5-30, 1. Low-calorie diet + exercise rate
normally 2. Energy-balanced diet + 2. Muscle and adipose tissue biopsies,
menstruating: exercise anthropometrics, ergometric
1. 30 trained Recreationally active assessment, hormones, inflammatory
2. 30 recreationally population: markers, cardiovascular indices
active 1. Low-calorie diet
2. Energy-balanced diet
NCT02858336 Crossover 150 females ages 1. Neutral energy availability 1. LH pulse frequency 5d
18-28 y, BMI (45 kcal/kg lean body 2. Levels of hormones, adipokines,
18.5-27, normal mass/d) assessment of glucose homeostasis
menstruation 2. Deficient energy
availability (20 kcal/kg
lean body mass/d)
NCT04910724 Prospective Individuals, ages Energy deficit resulting from a 1. Whole body protein balance (leucine 2d
18-35 y, BMI < 30, combination of exercise and isotope)
stable weight for 2 diet: 2. Muscle protein synthesis (phenylalanine
months 1. Energy deficit (20% of isotope)
daily requirements) 3. Carbohydrate oxidation (glucose
2. Energy deficit (40% of isotope)
daily requirements)
3. Energy deficit (60% of
daily requirements)
NCT04900701 Prospective 36 individuals, aged 1. Hypocaloric energy status 1. Muscle protein fractional synthetic rate 6d
35-65 y 2. Energy balance 2. Muscle morphology
3. Hypercaloric energy status
NCT05259969 Prospective 25 competitive male Observational study 1. Energy intake, expenditure and fat-free 1 y, (visits on
cohort athletes aged 18-40 examining a series of mass baseline, 6
y markers across the sport 2. Resting metabolic rate, body mass and mo, 12 mo)
seasons to assess the risk of composition, iron status markers,
LEA in male adult athletes. thyroid markers, testosterone, cortisol,
1. Preparation phase (peak LEAM-Q, eating attitude and disorder
exercise workload) questionnaires, inflammation markers,
2. Competition phase ghrelin, GH, gut microbiota,
3. transition phase (lowest metabolites and performance markers,
exercise workload) lipid profiles, leptin concentrations,
IGF-1
NCT03593382 Cross-sectional Female endurance Energy availability 1. Resting energy metabolism 6 wk to 6 mo
athlete exercising at 2. Secondary outcomes include: work after
least 5 times/week, efficiency, substrate utilization, bone inclusion
18-39 y mineral content and density, hormonal
measurements, lipid profile, vital signs,
dietary and activity questionnaires
NCT04254900 Prospective Athletes from a Energy availability evaluation 1. Energy intake, exercise energy 7 consecutive
national team in expenditure and fat-free mass and days during
wheelchair sports, using the following formula: energy preseason
18-60 y availability = (energy intake – exercise
energy expenditure)/fat-free mass)
NCT05587270 Prospective Swedish climbers on Web-based surveys and DXA 1. Eating disorders based on the Eating 3y
elite or subelite Disorders Examination-Questionnaire
levels of 2. Basic symptoms indicative of REDs,
competition, >13 y questionnaires on meals, menstruation,
injuries
(continued)
20 Endocrine Reviews, 2024, Vol. 00, No. 0

Table 2. Continued

Registered trials investigating prevalence, risk or mechanisms, and characteristics of LEA, REDs, and the Female Athlete Triad

Identifier Design Participants Arms Outcomes Time frame


3. Sleep quality; depression, anxiety, and
stress assessment; body image;
compulsive exercise and perfectionism
behaviors; overuse injuries

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


Registered interventional trials investigating treatments for energy-deficiency related disorders in healthy and unhealthy leanness, athletes, and soldiers

Identifier Design Phase Participants Arms Outcomes Time frame

Behavioral modifications and dietary supplementation


NCT04748250 Prospective RCT N/A 60 female adolescent 1. Acupuncture Bone mineral density—DXA 3 mo
gymnasts with 2. Soy phytoestrogens
diagnosed Female 3. Acupuncture + soy
Athlete Triad, phytoestrogens
particularly
osteoporosis
NCT05390346 Cross-sectional, N/A 100 male or female college 1. Evaluation of subjects’ 1. Energy availability Up to 21 mo
prospective athletes energy availability and 2. REDs-CAT risk scores,
risk stratification improvements in energy
2. Intervention through availability after nutrition
nutrition education education
NCT04766203 Prospective N/A 2000 athletes or 1. REDs screening 1. REDs CAT, menstrual 6 mo
nonrandomized para-athletes >15 y 2. REDs screening function, sex hormone Treatment arm:
(blood draw) + levels, biochemical blood 3-6 mo
follow-up tests, RMR and
3. REDs screening ergometric capacities,
(DXA scan, RMR IGF-1, BMD,
and exercise testing) + cardiovascular parameters
follow-up 2. Anthropometrics,
4. Treatment of REDs complete blood counts,
using a holistic glucose homeostasis,
treatment arm through hormones, vitamin D,
individualized questionnaires
nutrition plans and
counselling
NCT04823156 Prospective RCT N/A 45 females, ages 18-36 y, 1. Daily calcium + 1. Urine calcium isotopes 2 wk
BMI 18-30, stable vitamin D 2. At rest and during load
weight over 2 mo supplementation carriage: calcium isotopes,
2. LEA (45 kcal/kg lean P1NP, calcium
body mass/d for 7 metabolism, reproductive
days proceeding to and metabolic hormones,
15 kcal/kg lean body muscle strength,
mass/d thereafter) + anthropometrics, BMD
vitamin D
3. LEA + calcium +
vitamin D
NCT03963128 Prospective N/A 4450 male soldiers, ages 1. Vitamin D3 1. SF risk reduction 32 wk
RCT 16-32 y supplementation 2. Skin, soft tissue, and
(800 IU/d) respiratory infection,
2. Placebo vitamin D and hormone
levels, bone turnover
markers
NCT05341700 Cross-sectional N/A 15 females, ages 18-30 y, 1. Impact loading 1. Changes in osteocalcin, 5d
with regular menstrual exercises sclerostin, CTX, PTH,
cycles, exercising at 2. No impact load estrogen, IGF-1,
least 5 days per week exercises hepcidin, insulin, thyroid
hormones, 24-h glucose
patterns
2. Running economy—
oxygen consumption,
change in body weight
NCT05709678 Randomized N/A Physically active (training, 1. General nutrition and 1. Platform to Evaluate 3 wk
sport nutrition with Athlete Knowledge of
(continued)
Endocrine Reviews, 2024, Vol. 00, No. 0 21

Table 2. Continued

Registered interventional trials investigating treatments for energy-deficiency related disorders in healthy and unhealthy leanness, athletes, and soldiers

Identifier Design Phase Participants Arms Outcomes Time frame


competition-level) athlete testimonials on Sports Nutrition
adults aged 18-40 y impacts of REDs on Questionnaire (Peak-Nq)
health 2. Carbohydrate intake
2. Control group 3. Food frequency
questionnaire

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


NCT05589077 Interventional N/A Females aged 18-25 y, 1. Behavioral: Education 1. Change in Knowledge, Cross-sectional
that participate in on Female Athlete Confidence, and Impact
organized sport Nutritional Concerns Scores
NCT05709639 Prospective N/A 8 tier 4 and 5 elite Individualized counselling 1) Nutrition knowledge 12 wk
Olympic and in nutrition 2) Change in carbohydrate
paralympic athletes, intake
aged ≥ 18 years old
NCT06116097 Interventional N/A 100 competitive female 1. Experimental: 1. Energy availability 6 mo
endurance athletes aged Nutrition education 2. LEAF-Q
14-18 y training (behavioral) 3. Sports nutrition
minimum 6 h weekly intervention group knowledge questionnaire
(not taking a break from 2. No intervention: (SNKQ), Eating Attitude
sports for more than 3 Control group Test, Dietary intake,
mo) Energy Expenditure,
BMR, body composition
NCT06220240 Interventional N/A Professional female 1. Eccentric muscle 1. Energy intake and 2 wk
basketball players aged contraction modified availability, RED-S
18-35 y with a BMI training program CAT, biomechanics,
18.5-25, energy 2. Routine training running speed, activity
deficient. program 2. Hormones (gonadal,
thyroid, adrenal)
Pharmacological options for BMD loss in soldiers and athletes
NCT04196855 Prospective RCT 3 136 individuals 1. Teriparatide 20 μg/d 1. Radiological healing of SF 8, 10, 12, 14,
undergoing military 2. No intervention, 2. Time to healing, 16, 20, 24 wk
training, aged 18-40 y, standard SF care rehabilitation, pain
with lower limb SF quantification, quality of
life, adverse effects, P1NP,
CTX
NCT04589819 Prospective RCT 4 183 soldiers, diagnosed 1. Teriparatide 20μg/d 1. Time to return to full 3y
with tibial SF 2. Placebo activity
2. Long-term side effects,
injury recurrence, effects
on bone stress injuries
NCT05382026 Prospective N/A 114 adolescent athletes 1. Chocolate milk 1. Changes in lean mass 6 mo
RCT aged 12-17 y, having supplement (DXA)
reached menarche and 2. Pea-based beverage 2. Changes in fat mass,
performing resistance supplement lumbar BMD, hip BMD,
training 3x/wk 3. Placebo supplement bench press strength,
squat strength

Abbreviations: BMD, bone mineral density; BMI, body mass index; BMR, basal metabolic rate; DXA, dual-energy X-ray absorptiometry; LEA, low energy availability;
LEAF-Q, low energy availability in females questionnaire; N/A, not applicable; P1NP, pro-peptide of type 1 collagen; RCT, randomized controlled trial; REDs CAT,
relative energy deficiency in sport clinical assessment tool; RMR, resting metabolic rate; SF, stress fracture; β-CTX, β-carboxyl-terminal cross-linked telopeptide of type 1
collagen.

speed but not other performance measures and did not signifi­ (7.5 kg), waist circumference, and hemoglobin A1c (382).
cantly reduce falls (379). However, it is important to clarify Whether bimagrumab may have a role in maintaining muscle
that GH, IGF-1, and GH secretagogues are considered forms mass in lean, energy-deficient cohorts remains to be seen.
of doping and therefore are prohibited for use by competitive On the basis of improving performance, testosterone ad­
athletes. ministration has been recently assessed in a controlled study
Bimagrumab, an antibody blocking the activin type II recep­ of 48 young female athletes. Therein, daily application of tes­
tor, has been shown to increase lean body mass in older men tosterone cream led to marked enhancements in aerobic cap­
and women with sarcopenia and in older patients with recent acity and total lean mass, but not anaerobic performance or
hip fractures (380, 381). However, no improvements were muscle strength. Although this trial assessed circulating testos­
seen in physical performance. Another randomized controlled terone and performance-specific characteristics, it did not spe­
trial in adults with type 2 diabetes and obesity found that bima­ cifically address potential improvements in REDs-related
grumab over 48 weeks decreased fat mass by a mean of 20.5% markers, warranting further research (383).
22 Endocrine Reviews, 2024, Vol. 00, No. 0

According to findings from studies on college athletes, leptin analogues currently in development (388, 389). Large,
weight gain was identified as the most important indicator randomized phase 3 clinical trials investigating the effects of lep­
of normal menstrual function restoration (351, 384). The se­ tin in women and men with energy-deficiency-related disorders
verity of energy deficiency and the duration of the menstrual are warranted. Thus far, r-metHuLeptin has only been approved
disturbances affect the timeline of the menstrual cycle resump­ for patients with CLD or congenital or acquired generalized lip­
tion (385). Although not REDs-specific, according to the 2017 odystrophy (390, 391).
Endocrine Society Clinical Practice Guidelines, the adminis­ In contrast to its effects to normalize immune and neuroendo­
tration of GnRH analogs is considered the first line of treat­ crine function when leptin administration leads to normalization
ment for patients with FHA who wish to conceive, followed of the low leptin levels seen in energy deprivation states, leptin
by gonadotropin therapy and ovulation induction with clomi­ might exert anorexigenic properties and therefore suppress appe­

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


phene citrate (78). Short-term use of transdermal estradiol tite when administered in supraphysiological doses and only
with cyclic oral progesterone is suggested for those who when circulating levels exceed the upper physiological range
have not resumed menses after 6 to 12 months of lifestyle (392-394). Moreover, according to studies conducted on
modifications, but all these have a limited effect in normaliz­ women with FHA, recombinant leptin administration has been
ing only a small part of the big picture (78). associated with significant weight loss and fat mass reduction
when doses exceeded the upper limit of normal (45, 46, 114).
Therefore, as previously highlighted, leptin is not currently rec­
Leptin as a Potential Approach for REDs ommended for treating REDs-related health consequences be­
Current pharmacological options for the Female Athlete Triad cause randomized clinical trials on leptin administration with
and REDs fail to address the complete physiological picture. doses within the normal range (ie, beyond the proof of concept
GnRH analogs, estradiol, bone active agents, and GH analogs, studies discussed previously) have not been performed. The po­
as outlined previously, only contend with the endocrine tential role of leptin in specific athletic and military populations
changes downstream of leptin. Based on available data regard­ experiencing problematic LEA as a pharmacological approach
ing the efficacy of leptin in reversing FHA and remedying should be explored and confirmed in larger clinical trials
LEA-induced neuroendocrine perturbations in experimental involving both females and males with problematic LEA and
and small clinical settings, leptin could hypothetically consti­ REDs-related health consequences.
tute an effective candidate treatment for the broader family
of REDs-related disorders, especially in individuals with clinic­
ally low concentrations of leptin. Thus, from a pharmacologic­ Conclusions
al perspective, administering leptin, which is suppressed under REDs is a frequently overlooked constellation of disorders
LEA conditions (113, 115, 116, 386, 387), could potentially stemming from LEA in diverse exercising populations of ei­
serve as a complementary treatment strategy that could ad­ ther biological sex, resulting in various physiological conse­
dress problematic LEA-related pathophysiological alterations quences and impaired health and overall performance.
(eg, FHA, compromised bone health). However, leptin re­ Addressing the underlying causes of LEA by incorporating nu­
placement therapy is currently not recommended by consensus tritional, behavioral, and lifestyle modifications is currently
guidelines for individuals with hypothalamic amenorrhea or the primary treatment approach.
during low energy availability because its safety and effective­ As the principal arbiter of energy homeostasis, leptin may con­
ness require further investigation for confirmation. stitute a key neuroendocrine conduit for several REDs-related
Leptin administration in energy-deficient individuals over­ disturbances. The AFI system also seems to contribute to the
rides the energy conservation milieu that hinders the HPG axis pathophysiology of REDs. Decades of investigation have offered
in REDs (36, 45, 46), reversing gonadotropin pulsatility in males valuable insight into the potential therapeutic implications of
(42) and inducing ovulation, restoring menstruation, and thus leptin and the AFI axis, which exert a wide array of
resolving HA in lean energy-deficient females as discussed pro-metabolic effects (37, 254), including direct and indirect os­
previously (36, 45-47). Moreover, leptin replacement in teoprotective mechanisms (49).
energy-deprived amenorrheic women increased osteocalcin Furthermore, the practicality and feasibility of routinely
levels, decreased CTX levels, and increased lumbar BMC measuring leptin levels or AFI in clinical practice for REDs re­
and BMD (45, 114) while also decreasing intact PTH levels mains a significant consideration. However, it is important to
and tending to diminish the RANKL/OPG ratio indicative of recognize that REDs is a multifaceted disorder involving vari­
osteoclast differentiation and activity (330). Notwithstanding ous physiological and psychological factors, underscoring the
favorable results, because no large clinical trials beyond our need for a more comprehensive understanding of REDs,
proof-of-concept studies have been performed, the safety and ef­ which includes exploring other contributing factors. This
ficacy of leptin have not been adequately evaluated and thus lep­ has paved the way for new observational studies, prospective
tin cannot be recommended at this time as a definitive treatment cohorts, and randomized controlled trials in athletes and mili­
for HA. A major physiological caveat to the use of leptin in hy­ tary personnel, which should aim to encompass a broader
poleptinemic individuals is weight loss from overtreatment, range of factors impacting REDs for a more holistic approach
which can lead to supraphysiological circulating leptin levels to treatment and understanding.
(62, 388). Although leptin treatment reduces caloric intake
when administered in supraphysiological levels in lean individu­
als, it does not affect resting metabolic rate or physical activity, Additional Information
nor does it prevent the metabolomic shift from carbohydrate to Review criteria: The authors consulted the relevant position
fatty acid metabolism (285). At this time, it is unknown if anti­ statements of the American College of Sports Medicine and
bodies with any neutralizing effects may develop in subjects the IOC. Each author queried PubMed, Clinicaltrials.gov, tri­
with REDs in response to therapy with anti-r-metHuLeptin or al registries accepted by the International Committee of
Endocrine Reviews, 2024, Vol. 00, No. 0 23

Medical Journal Editors, and Google Scholar using the terms 3. Loucks AB. Energy balance and body composition in sports and
“energy deficiency,” “RED-S,” “REDs,” “female athlete tri­ exercise. J Sports Sci. 2004;22(1):1-14.
ad,” “anorexia,” “low energy availability,” “hypothalamic 4. Loucks AB, Kiens B, Wright HH. Energy availability in athletes. J
amenorrhea,” “bone,” “stress fractures,” “leptin,” “activin,” Sports Sci. 2011;29(Suppl 1):S7-S15.
5. Heikura IA, Stellingwerff T, Areta JL. Low energy availability in
“follistatin,” “adipose,” “soldier,” and “military.” The au­
female athletes: from the lab to the field. Eur J Sport Sci.
thors also reviewed relevant references cited within retrieved 2022;22(5):709-719.
articles. Cumulative results were presented as summaries in 6. Mountjoy M, Ackerman KE, Bailey DM, et al. 2023 International
tabular format, to be screened, assessed, and selected for in­ Olympic Committee’s (IOC) consensus statement on Relative
clusion in the current manuscript based on topic relevance, Energy Deficiency in Sport (REDs). Br J Sports Med.
conciseness, and study quality by the authors. Of note, for 2023;57(17):1073-1097.

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


the completeness of this review, the authors searched the lit­ 7. Wade GN, Jones JE. Neuroendocrinology of nutritional infertil­
erature for both Triad and REDs syndromes. The use of ity. Am J Physiol Regul Integr Comp Physiol. 2004;287(6):
each term inside the manuscript depends on the findings of R1277-R1296.
the original paper. 8. Burke LM, Ackerman KE, Heikura IA, Hackney AC, Stellingwerff
T. Mapping the complexities of Relative Energy Deficiency in
Sport (REDs): development of a physiological model by a sub­
group of the International Olympic Committee (IOC)
Author Contributions Consensus on REDs. Br J Sports Med. 2023;57(17):1098-1108.
C.S.M. conceptualized the review. K.S. and C.S.M. performed 9. Nattiv A, Loucks AB, Manore MM, Sanborn CF, Sundgot-Borgen
the initial search and wrote the first draft. A.M.A. performed J, Warren MP. American College of Sports Medicine position
extensive review of the first draft and A.M.A. and K.S. wrote stand. The female athlete triad. Med Sci Sports Exerc.
the manuscript. All coauthors (A.M.A., K.S., S.H.C., L.V.V., 2007;39(10):1867-1882.
D.G.G., K.D., K.N., C.B., P.T., A.K., H.A.P., and C.S.M.) 10. Yeager KK, Agostini R, Nattiv A, Drinkwater B. The female ath­
lete triad: disordered eating, amenorrhea, osteoporosis. Med Sci
performed literature searches, wrote sections of the text, con­
Sports Exerc. 1993;25(7):775-777.
tributed to the discussion of the content, edited, corrected, ex­ 11. Nattiv A, Agostini R, Drinkwater B, Yeager KK. The female ath­
tended, and reappraised all versions of the manuscript. All lete triad. The inter-relatedness of disordered eating, amenorrhea,
authors have approved the final version of the submitted and osteoporosis. Clin Sports Med. 1994;13(2):405-418.
manuscript. 12. Otis CL, Drinkwater B, Johnson M, Loucks A, Wilmore J.
American College of Sports Medicine position stand. The female
athlete triad. Med Sci Sports Exerc. 1997;29(5):i-ix.
Disclosures 13. Ikegami N, Samukawa M, Sakamaki-Sunaga M, et al. The influ­
ence of low energy availability on bone mineral density and tra­
No funding sources or conflicts of interest pertaining to the
becular bone microarchitecture of pubescent female athletes: a
present work were reported from the authors. This review is preliminary study. Int J Environ Res Public Health. 2022;19(9):
not funded, and none of the authors have received any funding 5580.
or nonfinancial support relevant to its completion. Other un­ 14. De Souza MJ, Nattiv A, Joy E, et al. 2014 Female athlete triad co­
related disclosures are as follows: A.K. reports grants and ad­ alition consensus statement on treatment and return to play of the
visory services for Novo Nordisk, advisory services for Eli female athlete triad: 1st International Conference held in
Lilly, Bausch Health, Sanofi-Aventis, MSD, AstraZeneca, San Francisco, California, May 2012 and 2nd International
Elpen Pharmaceuticals, Boehringer-Ingelheim, Galenica, Conference held in Indianapolis, Indiana, May 2013. Br J Sports
Ethicon, and Epsilon Health. None is related to the work pre­ Med. 2014;48(4):289.
sented herein. C.S.M. reports grants through his institution 15. Hattori S, Aikawa Y, Omi N. Female athlete triad and male athlete
triad syndrome induced by low energy availability: an animal
from Merck, Massachusetts Life Sciences Center, and
model. Calcif Tissue Int. 2022;111(2):116-123.
Boehringer Ingelheim, has been a shareholder of and has re­
16. Hattori S, Park J-H, Agata U, et al. Food restriction causes low
ceived grants through his institution and personal consulting bone strength and microarchitectural deterioration in exercised
fees from Coherus Inc. and AltrixBio; he reports personal con­ growing male rats. J Nutr Sci Vitaminol (Tokyo). 2014;60(1):
sulting fees and support with research reagents from Ansh 35-42.
Inc., collaborative research support from LabCorp Inc., re­ 17. Stewart AD, Hannan J. Total and regional bone density in male
ports personal consulting fees from Genfit, Lumos, Amgen, runners, cyclists, and controls. Med Sci Sports Exerc. 2000;32(8):
Corcept, Aligos, Intercept, 89 Bio, Madrigal, and 1373-1377.
Regeneron, reports travel support and fees from TMIOA, 18. Kraus E, Tenforde AS, Nattiv A, et al. Bone stress injuries in male
Elsevier, and the Cardio Metabolic Health Conference. distance runners: higher modified Female Athlete Triad
None is related to the work presented herein. The other au­ Cumulative Risk Assessment scores predict increased rates of in­
jury. Br J Sports Med. 2019;53(4):237-242.
thors have nothing to disclose.
19. Viner RT, Harris M, Berning JR, Meyer NL. Energy availability
and dietary patterns of adult male and female competitive cyclists
with lower than expected bone mineral density. Int J Sport Nutr
References Exerc Metab. 2015;25(6):594-602.
1. Areta JL, Taylor HL, Koehler K. Low energy availability: history, 20. Hackney AC, Moore AW, Brownlee KK. Testosterone and endur­
definition and evidence of its endocrine, metabolic and physio­ ance exercise: development of the “exercise-hypogonadal male
logical effects in prospective studies in females and males. Eur J condition”. Acta Physiol Hung. 2005;92(2):121-137.
Appl Physiol. 2021;121(1):1-21. 21. O’Leary TJ, Wardle SL, Greeves JP. Energy deficiency in soldiers:
2. Burke LM, Lundy B, Fahrenholtz IL, Melin AK. Pitfalls of con­ the risk of the athlete triad and relative energy deficiency in sport
ducting and interpreting estimates of energy availability in free- syndromes in the military. Front Nutr. 2020;7:142.
living athletes. Int J Sport Nutr Exerc Metab. 2018;28(4): 22. Nattiv A, De Souza MJ, Koltun KJ, et al. The male athlete triad-a
350-363. consensus statement from the female and male athlete triad
24 Endocrine Reviews, 2024, Vol. 00, No. 0

coalition part 1: definition and scientific basis. Clin J Sport Med. 43. Perakakis N, Upadhyay J, Ghaly W, et al. Regulation of the
2021;31(4):345-348. activins-follistatins-inhibins axis by energy status: impact on re­
23. Fredericson M, Kussman A, Misra M, et al. The male athlete productive function. Metabolism. 2018;85:240-249.
triad-a consensus statement from the female and male athlete triad 44. Chan JL, Matarese G, Shetty GK, et al. Differential regulation of
coalition part II: diagnosis, treatment, and return-to-play. Clin J metabolic, neuroendocrine, and immune function by leptin in hu­
Sport Med. 2021;31(4):349-366. mans. Proc Natl Acad Sci U S A. 2006;103(22):8481-8486.
24. MacDougall JD, Webber CE, Martin J, et al. Relationship among 45. Chou SH, Chamberland JP, Liu X, et al. Leptin is an effective
running mileage, bone density, and serum testosterone in male treatment for hypothalamic amenorrhea. Proc Natl Acad Sci U
runners. J Appl Physiol (1985). 1992;73(3):1165-1170. S A. 2011;108(16):6585-6590.
25. Hattori S, Uchizawa A, Omi N. Comparison of bone mineral 46. Welt CK, Chan JL, Bullen J, et al. Recombinant human leptin in
density between male long distance runners and non-athletes. J women with hypothalamic amenorrhea. N Engl J Med.

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


Run Sci. 2020;31(1):9-16. 2004;351(10):987-997.
26. Mountjoy M, Sundgot-Borgen J, Burke L, et al. The IOC consen­ 47. Chan JL, Mantzoros CS. Role of leptin in energy-deprivation
sus statement: beyond the Female Athlete Triad–Relative Energy states: normal human physiology and clinical implications for
Deficiency in Sport (RED-S). Br J Sports Med. 2014;48(7): hypothalamic amenorrhoea and anorexia nervosa. Lancet.
491-497. 2005;366(9479):74-85.
27. Dipla K, Kraemer RR, Constantini NW, Hackney AC. Relative 48. Nakamura Y, Nakano M, Suzuki T, et al. Two adipocytokines,
energy deficiency in sports (RED-S): elucidation of endocrine leptin and adiponectin, independently predict osteoporotic frac­
changes affecting the health of males and females. Hormones ture risk at different bone sites in postmenopausal women.
(Athens). 2021;20(1):35-47. Bone. 2020;137:115404.
28. Rogers MA, Appaneal RN, Hughes D, et al. Prevalence of im­ 49. Upadhyay J, Farr OM, Mantzoros CS. The role of leptin in regu­
paired physiological function consistent with Relative Energy lating bone metabolism. Metabolism. 2015;64(1):105-113.
Deficiency in Sport (RED-S): an Australian elite and pre-elite co­ 50. Takeda S, Elefteriou F, Levasseur R, et al. Leptin regulates bone for­
hort. Br J Sports Med. 2021;55(1):38-45. mation via the sympathetic nervous system. Cell. 2002;111(3):
29. Melin AK, Heikura IA, Tenforde A, Mountjoy M. Energy avail­ 305-317.
ability in athletics: health, performance, and physique. Int J 51. Schett G, Kiechl S, Bonora E, et al. Serum leptin level and the risk
Sport Nutr Exerc Metab. 2019;29(2):152-164. of nontraumatic fracture. Am J Med. 2004;117(12):952-956.
30. Mountjoy M, Sundgot-Borgen JK, Burke LM, et al. IOC consen­ 52. Papageorgiou M, Dolan E, Elliott-Sale KJ, Sale C. Reduced energy
sus statement on relative energy deficiency in sport (RED-S): availability: implications for bone health in physically active pop­
2018 update. Br J Sports Med. 2018;52(11):687-697. ulations. Eur J Nutr. 2018;57(3):847-859.
31. Elliott-Sale KJ, Tenforde AS, Parziale AL, Holtzman B, Ackerman 53. Perakakis N, Mougios V, Fatouros I, et al. Physiology of activins/
KE. Endocrine effects of relative energy deficiency in sport. Int J follistatins: associations with metabolic and anthropometric vari­
Sport Nutr Exerc Metab. 2018;28(4):335-349. ables and response to exercise. J Clin Endocrinol Metab.
32. Stellingwerff T, Heikura IA, Meeusen R, et al. Overtraining syn­ 2018;103(10):3890-3899.
drome (OTS) and relative energy deficiency in sport (RED-S): 54. Perakakis N, Kokkinos A, Peradze N, et al. Follistatins in glucose
shared pathways, symptoms and complexities. Sports Med. regulation in healthy and obese individuals. Diabetes Obes Metab.
2021;51(11):2251-2280. 2019;21(3):683-690.
33. Williams NI, Koltun KJ, Strock NCA, De Souza MJ. Female ath­ 55. Logue D, Madigan SM, Delahunt E, Heinen M, Mc Donnell SJ,
lete triad and relative energy deficiency in sport: a focus on scien­ Corish CA. Low energy availability in athletes: a review of preva­
tific rigor. Exerc Sport Sci Rev. 2019;47(4):197-205. lence, dietary patterns, physiological health, and sports perform­
34. De Souza MJ, Strock NCA, Ricker EA, et al. The path towards ance. Sports Med. 2018;48(1):73-96.
progress: a critical review to advance the science of the female 56. Loucks AB, Thuma JR. Luteinizing hormone pulsatility is dis­
and male athlete triad and relative energy deficiency in sport. rupted at a threshold of energy availability in regularly menstruat­
Sports Med. 2022;52(1):13-23. ing women. J Clin Endocrinol Metab. 2003;88(1):297-311.
35. Torstveit MK, Fahrenholtz IL, Lichtenstein MB, Stenqvist TB, 57. Loucks AB, Heath EM. Induction of low-T3 syndrome in exercis­
Melin AK. Exercise dependence, eating disorder symptoms and bi­ ing women occurs at a threshold of energy availability. Am J
omarkers of Relative Energy Deficiency in Sports (RED-S) among Physiol. 1994;266(3 Pt 2):R817-R823.
male endurance athletes. BMJ Open Sport Exerc Med. 2019;5(1): 58. Ihle R, Loucks AB. Dose-response relationships between energy
e000439. availability and bone turnover in young exercising women. J
36. Boutari C, Pappas PD, Mintziori G, et al. The effect of underweight Bone Miner Res. 2004;19(8):1231-1240.
on female and male reproduction. Metabolism. 2020;107:154229. 59. Lane AR, Hackney AC, Smith-Ryan A, Kucera K, Registar-Mihalik
37. Friedman JM. Leptin and the endocrine control of energy balance. J, Ondrak K. Prevalence of low energy availability in competitively
Nat Metab. 2019;1(8):754-764. trained male endurance athletes. Medicina (Kaunas). 2019;55(10):
38. Zhang Y, Chua S Jr. Leptin function and regulation. Compr 665.
Physiol. 2017;8(1):351-369. 60. Lieberman JL, De Souza MJ, Wagstaff DA, Williams NI.
39. Dardeno TA, Chou SH, Moon H-S, Chamberland JP, Fiorenza Menstrual disruption with exercise is not linked to an energy
CG, Mantzoros CS. Leptin in human physiology and therapeutics. availability threshold. Med Sci Sports Exerc. 2018;50(3):551-561.
Front Neuroendocrinol. 2010;31(3):377-393. 61. Williams NI, Leidy HJ, Hill BR, Lieberman JL, Legro RS, De Souza
40. Mantzoros CS. The role of leptin in human obesity and disease: a MJ. Magnitude of daily energy deficit predicts frequency but not se­
review of current evidence. Ann Intern Med. 1999;130(8): verity of menstrual disturbances associated with exercise and calor­
671-680. ic restriction. Am J Physiol Endocrinol Metab. 2015;308(1):
41. Mantzoros CS, Moschos SJ. Leptin: in search of role(s) in human E29-E39.
physiology and pathophysiology. Clin Endocrinol (Oxf). 62. Kelesidis T, Kelesidis I, Chou S, Mantzoros CS. Narrative review:
1998;49(5):551-567. the role of leptin in human physiology: emerging clinical applica­
42. Chan JL, Heist K, DePaoli AM, Veldhuis JD, Mantzoros CS. The tions. Ann Intern Med. 2010;152(2):93-100.
role of falling leptin levels in the neuroendocrine and metabolic 63. Hilton LK, Loucks AB. Low energy availability, not exercise
adaptation to short-term starvation in healthy men. J Clin stress, suppresses the diurnal rhythm of leptin in healthy young
Invest. 2003;111(9):1409-1421. women. Am J Physiol Endocrinol Metab. 2000;278(1):E43-E49.
Endocrine Reviews, 2024, Vol. 00, No. 0 25

64. Shimizu K, Suzuki N, Nakamura M, et al. Mucosal immune func­ 85. Friedl KE, Moore RJ, Hoyt RW, Marchitelli LJ, Martinez-Lopez
tion comparison between amenorrheic and eumenorrheic distance LE, Askew EW. Endocrine markers of semistarvation in healthy
runners. J Strength Cond Res. 2012;26(5):1402-1406. lean men in a multistressor environment. J Appl Physiol (1985).
65. Lieberman JL, Hill BR, De Souza MJ, Williams NI. Luteal phase 2000;88(5):1820-1830.
defects induced by exercise and diet are associated with low energy 86. Roberts AC, McClure RD, Weiner RI, Brooks GA. Overtraining
availability. Med Sci Sports Exerc. 2013;(S5):521-521. affects male reproductive status. Fertil Steril. 1993;60(4):
66. Barrack MT, Ackerman KE, Gibbs JC. Update on the female ath­ 686-692.
lete triad. Curr Rev Musculoskelet Med. 2013;6(2):195-204. 87. Aakvaag A, Bentdal Ø, Quigstad K, Walstad P, Rønningen H,
67. Woods AL, Garvican-Lewis LA, Lundy B, Rice AJ, Thompson Fonnum F. Testosterone and Testosterone Binding Globulin
KG. New approaches to determine fatigue in elite athletes during (TeBG) in young men during prolonged stress. Int J Androl.
intensified training: resting metabolic rate and pacing profile. 1978;1(1-6):22-31.

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


PLoS One. 2017;12(3):e0173807. 88. Aakvaag A, Sand T, Opstad PK, Fonnum F. Hormonal changes in
68. Vanheest JL, Rodgers CD, Mahoney CE, De Souza MJ. Ovarian serum in young men during prolonged physical strain. Eur J Appl
suppression impairs sport performance in junior elite female Physiol Occup Physiol. 1978;39(4):283-291.
swimmers. Med Sci Sports Exerc. 2014;46(1):156-166. 89. Opstad PK, Aakvaag A. Decreased serum levels of oestradiol, tes­
69. Nazem TG, Ackerman KE. The female athlete triad. Sports tosterone and prolactin during prolonged physical strain and sleep
Health. 2012;4(4):302-311. deprivation, and the influence of a high calorie diet. Eur J Appl
70. Papageorgiou M, Elliott-Sale KJ, Parsons A, et al. Effects of re­ Physiol Occup Physiol. 1982;49(3):343-348.
duced energy availability on bone metabolism in women and 90. Opstad PK, Falch D, Oktedalen O, Fonnum F, Wergeland R. The
men. Bone. 2017;105:191-199. thyroid function in young men during prolonged exercise and the
71. Burke LM, Close GL, Lundy B, Mooses M, Morton JP, Tenforde effect of energy and sleep deprivation. Clin Endocrinol (Oxf).
AS. Relative energy deficiency in sport in male athletes: a commen­ 1984;20(6):657-669.
tary on its presentation among selected groups of male athletes. 91. Opstad PK, Aakvaag A. The effect of a high calory diet on hormo­
Int J Sport Nutr Exerc Metab. 2018;28(4):364-374. nal changes in young men during prolonged physical strain and
72. Hackney AC, Lane AR, Register-Mihalik J, O’Leary CB. sleep deprivation. Eur J Appl Physiol Occup Physiol.
Endurance exercise training and male sexual libido. Med Sci
1981;46(1):31-39.
Sports Exerc. 2017;49(7):1383-1388.
92. De Souza MJ, Arce JC, Pescatello LS, Scherzer HS, Luciano AA.
73. McKay AK, Peeling P, Pyne DB, et al. Six days of low carbohy­
Gonadal hormones and semen quality in male runners. A volume
drate, not energy availability, alters the iron and immune response
threshold effect of endurance training. Int J Sports Med.
to exercise in elite athletes. Med Sci Sports Exerc. 2022;54(3):
1994;15(7):383-391.
377-387.
93. Gebreegziabher Y, Marcos E, McKinon W, Rogers G. Sperm char­
74. Fensham NC, Heikura IA, McKay AKA, Tee N, Ackerman KE,
acteristics of endurance trained cyclists. Int J Sports Med.
Burke LM. Short-term carbohydrate restriction impairs bone for­
2004;25(4):247-251.
mation at rest and during prolonged exercise to a greater degree
94. Loucks AB, Callister R. Induction and prevention of low-T3 syn­
than low energy availability. J Bone Miner Res. 2022;37(10):
drome in exercising women. Am J Physiol. 1993;264(5 Pt 2):
1915-1925.
R924-R930.
75. Guyatt GH, Oxman AD, Vist GE, et al. GRADE: an emerging con­
95. Tornberg ÅB, Melin A, Koivula FM, et al. Reduced neuromuscu­
sensus on rating quality of evidence and strength of recommenda­
lar performance in amenorrheic elite endurance athletes. Med Sci
tions. BMJ. 2008;336(7650):924-926.
Sports Exerc. 2017;49(12):2478-2485.
76. De Souza MJ, Toombs RJ, Scheid JL, O’Donnell E, West SL,
96. Loucks AB, Laughlin GA, Mortola JF, Girton L, Nelson JC, Yen
Williams NI. High prevalence of subtle and severe menstrual dis­
turbances in exercising women: confirmation using daily hormone SS. Hypothalamic-pituitary-thyroidal function in eumenorrheic
measures. Hum Reprod. 2010;25(2):491-503. and amenorrheic athletes. J Clin Endocrinol Metab. 1992;75(2):
77. Melin A, Tornberg ÅB, Skouby S, et al. Energy availability and the 514-518.
female athlete triad in elite endurance athletes. Scand J Med Sci 97. Harber VJ, Petersen SR, Chilibeck PD. Thyroid hormone concen­
Sports. 2015;25(5):610-622. trations and muscle metabolism in amenorrheic and eumenorrheic
78. Gordon CM, Ackerman KE, Berga SL, et al. Functional hypothal­ athletes. Can J Appl Physiol. 1998;23(3):293-306.
amic amenorrhea: an endocrine society clinical practice guideline. 98. Hartman ML, Veldhuis JD, Johnson ML, et al. Augmented
J Clin Endocrinol Metab. 2017;102(5):1413-1439. growth hormone (GH) secretory burst frequency and amplitude
79. Gimunová M, Paulínyová A, Bernaciková M, Paludo AC. The mediate enhanced GH secretion during a two-day fast in normal
prevalence of menstrual cycle disorders in female athletes from men. J Clin Endocrinol Metab. 1992;74(4):757-765.
different sports disciplines: a rapid review. Int J Environ Res 99. Misra M, Miller KK, Bjornson J, et al. Alterations in growth hor­
Public Health. 2022;19(21):14243. mone secretory dynamics in adolescent girls with anorexia ner­
80. Loucks AB, Verdun M, Heath EM. Low energy availability, not vosa and effects on bone metabolism. J Clin Endocrinol Metab.
stress of exercise, alters LH pulsatility in exercising women. J 2003;88(12):5615-5623.
Appl Physiol (1985). 1998;84(1):37-46. 100. Laughlin GA, Dominguez CE, Yen SS. Nutritional and endocrine-
81. MacConnie SE, Barkan A, Lampman RM, Schork MA, Beitins IZ. metabolic aberrations in women with functional hypothalamic
Decreased hypothalamic gonadotropin-releasing hormone secretion amenorrhea. J Clin Endocrinol Metab. 1998;83(1):25-32.
in male marathon runners. N Engl J Med. 1986;315(7):411-417. 101. Ipsa E, Cruzat VF, Kagize JN, Yovich JL, Keane KN. Growth hor­
82. Wheeler GD, Wall SR, Belcastro AN, Cumming DC. Reduced se­ mone and insulin-like growth factor action in reproductive tissues.
rum testosterone and prolactin levels in male distance runners. Front Endocrinol (Lausanne). 2019;10:777.
JAMA. 1984;252(4):514-516. 102. Thissen JP, Ketelslegers JM, Underwood LE. Nutritional regula­
83. Jin J-M, Yang W-X. Molecular regulation of hypothalamus- tion of the insulin-like growth factors. Endocr Rev. 1994;15(1):
pituitary-gonads axis in males. Gene. 2014;551(1):15-25. 80-101.
84. Ruffing KM, Koltun KJ, De Souza MJ, Williams NI. Moderate 103. Murphy C, Koehler K. Caloric restriction induces anabolic resist­
weight loss is associated with reductions in LH pulse frequency ance to resistance exercise. Eur J Appl Physiol. 2020;120(5):
and increases in 24-hour cortisol with no change in perceived 1155-1164.
stress in young ovulatory women. Physiol Behav. 2022;254: 104. Chan JL, Williams CJ, Raciti P, et al. Leptin does not mediate
113885. short-term fasting-induced changes in growth hormone pulsatility
26 Endocrine Reviews, 2024, Vol. 00, No. 0

but increases IGF-I in leptin deficiency states. J Clin Endocrinol 122. Waterman BR, Gun B, Bader JO, Orr JD, Belmont PJ Jr.
Metab. 2008;93(7):2819-2827. Epidemiology of lower extremity stress fractures in the United
105. Brundu B, Loucks TL, Adler LJ, Cameron JL, Berga SL. Increased States military. Mil Med. 2016;181(10):1308-1313.
cortisol in the cerebrospinal fluid of women with functional hypo­ 123. Bulathsinhala L, Hughes JM, McKinnon CJ, et al. Risk of stress
thalamic amenorrhea. J Clin Endocrinol Metab. 2006;91(4): fracture varies by race/ethnic origin in a cohort study of 1.3 mil­
1561-1565. lion US army soldiers. J Bone Miner Res. 2017;32(7):1546-1553.
106. De Souza MJ, Maguire MS, Maresh CM, Kraemer WJ, Rubin KR, 124. Knapik J, Montain SJ, McGraw S, Grier T, Ely M, Jones BH.
Loucks AB. Adrenal activation and the prolactin response to exer­ Stress fracture risk factors in basic combat training. Int J Sports
cise in eumenorrheic and amenorrheic runners. J Appl Physiol Med. 2012;33(11):940-946.
(1985). 1991;70(6):2378-2387. 125. MacGregor AJ, Fogleman SA, Dougherty AL, Ryans CP, Janney
107. Laughlin GA, Yen SSC. Hypoleptinemia in women athletes: ab­ CF, Fraser JJ. Sex differences in the incidence and risk of ankle–

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


sence of a diurnal rhythm with amenorrhea. J Clin Endocrinol foot complex stress fractures among U.S. military personnel. J
Metab. 1997;82(1):318-321. Women’s Health. 2022;31(4):586-592.
108. Berga SL, Daniels TL, Giles DE. Women with functional hypothal­ 126. Armstrong DW 3rd, Rue JP, Wilckens JH, Frassica FJ. Stress frac­
amic amenorrhea but not other forms of anovulation display amp­ ture injury in young military men and women. Bone. 2004; 35(3):
lified cortisol concentrations. Fertil Steril. 1997;67(6):1024-1030. 806-816.
109. Chrousos GP, Gold PW. The concepts of stress and stress system 127. Millward D, Root AD, Dubois J, et al. Association of serum vita­
disorders. Overview of physical and behavioral homeostasis. min D levels and stress fractures in collegiate athletes. Orthop J
JAMA. 1992;267(9):1244-1252. Sports Med. 2020;8(12):2325967120966967.
110. Ackerman KE, Patel KT, Guereca G, Pierce L, Herzog DB, Misra 128. Jones BH, Thacker SB, Gilchrist J, Kimsey CD Jr, Sosin DM.
M. Cortisol secretory parameters in young exercisers in relation to Prevention of lower extremity stress fractures in athletes and sol­
LH secretion and bone parameters. Clin Endocrinol (Oxf). diers: a systematic review. Epidemiol Rev. 2002;24(2):228-247.
2013;78(1):114-119. 129. Milner CE, Foch E, Gonzales JM, Petersen D. Biomechanics asso­
111. Vamvini MT, Aronis KN, Chamberland JP, Mantzoros CS. ciated with tibial stress fracture in runners: a systematic review
Energy deprivation alters in a leptin- and cortisol-independent and meta-analysis. J Sport Health Sci. 2023;12(3):333-342.
manner circulating levels of activin A and follistatin but not myo­ 130. Wright AA, Taylor JB, Ford KR, Siska L, Smoliga JM. Risk factors
statin in healthy males. J Clin Endocrinol Metab. 2011;96(11): associated with lower extremity stress fractures in runners: a sys­
3416-3423. tematic review with meta-analysis. Br J Sports Med. 2015;49(23):
112. Koehler K, Hoerner NR, Gibbs JC, et al. Low energy availability 1517-1523.
in exercising men is associated with reduced leptin and insulin but 131. Saxena A, Fullem B. Navicular stress fractures: a prospective study
not with changes in other metabolic hormones. J Sports Sci. on athletes. Foot Ankle Int. 2006;27(11):917-921.
2016;34(20):1921-1929. 132. Bennell KL, Malcolm SA, Thomas SA, et al. Risk factors for stress
113. Christo K, Cord J, Mendes N, et al. Acylated ghrelin and leptin in fractures in track and field athletes. A twelve-month prospective
adolescent athletes with amenorrhea, eumenorrheic athletes and study. Am J Sports Med. 1996;24(6):810-818.
controls: a cross-sectional study. Clin Endocrinol (Oxf). 133. Bennell KL, Malcolm SA, Thomas SA, Wark JD, Brukner PD. The
2008;69(4):628-633. incidence and distribution of stress fractures in competitive track
114. Sienkiewicz E, Magkos F, Aronis KN, et al. Long-term metreleptin and field athletes. A twelve-month prospective study. Am J
treatment increases bone mineral density and content at the lumbar Sports Med. 1996;24(2):211-217.
spine of lean hypoleptinemic women. Metabolism. 2011;60(9): 134. Takkar P, Prabhakar R. Stress fractures in military recruits: a pro­
1211-1221. spective study for evaluation of incidence, patterns of injury and
115. Donoso MA, Muñoz-Calvo MT, Barrios V, Garrido G, Hawkins invalidments out of service. Med J Armed Forces India.
F, Argente J. Increased circulating adiponectin levels and de­ 2019;75(3):330-334.
creased leptin/soluble leptin receptor ratio throughout puberty 135. Cosman F, Ruffing J, Zion M, et al. Determinants of stress fracture
in female ballet dancers: association with body composition and risk in United States Military Academy cadets. Bone. 2013;55(2):
the delay in puberty. Eur J Endocrinol. 2010;162(5):905-911. 359-366.
116. Zaccaria M, Ermolao A, Brugin E, Bergamin M. Plasma leptin and 136. Wentz L, Liu P-Y, Haymes E, Ilich JZ. Females have a greater in­
energy expenditure during prolonged, moderate intensity, tread­ cidence of stress fractures than males in both military and athletic
mill exercise. J Endocrinol Invest. 2013;36(6):396-401. populations: a systemic review. Mil Med. 2011;176(4):420-430.
117. De Souza MJ, Ricker EA, Mallinson RJ, et al. Bone mineral dens­ 137. Sarin HV, Gudelj I, Honkanen J, et al. Molecular pathways medi­
ity in response to increased energy intake in exercising women ating immunosuppression in response to prolonged intensive
with oligomenorrhea/amenorrhea: the REFUEL randomized con­ physical training, low-energy availability, and intensive weight
trolled trial. Am J Clin Nutr. 2022;115(6):1457-1472. loss. Front Immunol. 2019;10:907.
118. Popp KL, Cooke LM, Bouxsein ML, Hughes JM. Impact of low 138. Murakami S, Kurihara S, Titchenal CA, Ohtani M. Suppression of
energy availability on skeletal health in physically active adults. exercise-induced neutrophilia and lymphopenia in athletes by
Calcif Tissue Int. 2022;110(5):605-614. cystine/theanine intake: a randomized, double-blind, placebo-
119. Evans RK, Antczak AJ, Lester M, Yanovich R, Israeli E, Moran controlled trial. J Int Soc Sports Nutr. 2010;7(1):23.
DS. Effects of a 4-month recruit training program on markers of 139. Pedersen BK, Toft AD. Effects of exercise on lymphocytes and cy­
bone metabolism. Med Sci Sports Exerc. 2008;40(11 Suppl): tokines. Br J Sports Med. 2000;34(4):246-251.
S660-S670. 140. Li X, Kan EM, Lu J, et al. Combat-training increases intestinal
120. Abbott A, Wang C, Stamm M, Mulcahey MK. Part II: risk factors permeability, immune activation and gastrointestinal symptoms
for stress fractures in female military recruits. Mil Med. in soldiers. Aliment Pharmacol Ther. 2013;37(8):799-809.
2023;188(1-2):93-99. 141. McClung JP, Martini S, Murphy NE, et al. Effects of a 7-day mili­
121. Tenforde AS, Katz NB, Sainani KL, Carlson JL, Golden NH, tary training exercise on inflammatory biomarkers, serum hepci­
Fredericson M. Female athlete triad risk factors are more strongly din, and iron status. Nutr J. 2013;12(1):141.
associated with trabecular-rich versus cortical-rich bone stress in­ 142. Pasiakos SM, Margolis LM, Murphy NE, et al. Effects of exercise
juries in collegiate athletes. Orthop J Sports Med. 2022;10(9): mode, energy, and macronutrient interventions on inflammation
23259671221123588. during military training. Physiol Rep. 2016;4(11):e12820.
Endocrine Reviews, 2024, Vol. 00, No. 0 27

143. Shimizu K, Aizawa K, Suzuki N, et al. Influences of weight loss on on sports performance. Scand J Med Sci Sports. 2024;34(1):
monocytes and T-cell subpopulations in male judo athletes. J e14327.
Strength Cond Res. 2011;25(7):1943-1950. 163. Eliakim A, Nemet D, Constantini N. Screening blood tests in
144. Imai T, Seki S, Dobashi H, Ohkawa T, Habu Y, Hiraide H. Effect members of the Israeli National Olympic team. J Sports Med
of weight loss on T-cell receptor-mediated T-cell function in elite Phys Fitness. 2002;42(2):250-255.
athletes. Med Sci Sports Exerc. 2002;34(2):245-250. 164. Malczewska J, Szczepaǹ ska B, Stupnicki R, Sendecki W. The as­
145. Suzuki M, Nakaji S, Umeda T, et al. Effects of weight reduction on sessment of frequency of iron deficiency in athletes from the trans­
neutrophil phagocytic activity and oxidative burst activity in fe­ ferrin receptor-ferritin index. Int J Sport Nutr Exerc Metab.
male judoists. Luminescence. 2003;18(4):214-217. 2001;11(1):42-52.
146. Yaegaki M, Umeda T, Takahashi I, et al. Change in the capability 165. Ziemann E, Kasprowicz K, Kasperska A, Zembroǹ -Lacny A,
of reactive oxygen species production by neutrophils following Antosiewicz J, Laskowski R. Do high blood hepcidin concentra­

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


weight reduction in female judoists. Br J Sports Med. tions contribute to low ferritin levels in young tennis players at
2007;41(5):322-327. the end of tournament season? J Sports Sci Med. 2013;12(2):
147. Tsai M-L, Chou K-M, Chang C-K, Fang S-H. Changes of mucosal 249-258.
immunity and antioxidation activity in elite male Taiwanese taek­ 166. Badenhorst CE, Black KE, O’Brien WJ. Hepcidin as a prospective
wondo athletes associated with intensive training and rapid individualized biomarker for individuals at risk of low energy
weight loss. Br J Sports Med. 2011;45(9):729-734. availability. Int J Sport Nutr Exerc Metab. 2019;29(6):671-681.
148. Drew M, Vlahovich N, Hughes D, et al. Prevalence of illness, 167. Sim M, Dawson B, Landers GJ, et al. A seven day running training
poor mental health and sleep quality and low energy availability period increases basal urinary hepcidin levels as compared to cyc­
prior to the 2016 summer Olympic games. Br J Sports Med. ling. J Int Soc Sports Nutr. 2014;11(1):14.
2018;52(1):47-53. 168. Rickenlund A, Eriksson MJ, Schenck-Gustafsson K, Hirschberg
149. Drew MK, Vlahovich N, Hughes D, et al. A multifactorial evalu­ AL. Amenorrhea in female athletes is associated with endothelial
ation of illness risk factors in athletes preparing for the Summer dysfunction and unfavorable lipid profile. J Clin Endocrinol
Olympic Games. J Sci Med Sport. 2017;20(8):745-750. Metab. 2005;90(3):1354-1359.
150. Tsai M-L, Ko M-H, Chang C-K, Chou K-M, Fang S-H. Impact of 169. Costa RJS, Snipe RMJ, Kitic CM, Gibson PR. Systematic review:
intense training and rapid weight changes on salivary parameters exercise-induced gastrointestinal syndrome-implications for health
in elite female taekwondo athletes. Scand J Med Sci Sports. and intestinal disease. Aliment Pharmacol Ther. 2017;46(3):
2011;21(6):758-764. 246-265.
151. Martinez-Lopez LE, Friedl KE, Moore RJ, Kramer TR. A longitu­ 170. Norris ML, Harrison ME, Isserlin L, Robinson A, Feder S,
dinal study of infections and injuries of Ranger students. Mil Med. Sampson M. Gastrointestinal complications associated with ano­
1993;158(7):433-437. rexia nervosa: a systematic review. Int J Eat Disord. 2016;49(3):
152. Andreoli A, Monteleone M, Van Loan M, Promenzio L, Tarantino 216-237.
U, De Lorenzo A. Effects of different sports on bone density and 171. Chantler S, Griffiths A, Matu J, Davison G, Jones B, Deighton K.
muscle mass in highly trained athletes. Med Sci Sports Exerc. The effects of exercise on indirect markers of gut damage and per­
2001;33(4):507-511. meability: a systematic review and meta-analysis. Sports Med.
153. Rauh MJ, Nichols JF, Barrack MT. Relationships among injury 2021;51(1):113-124.
and disordered eating, menstrual dysfunction, and low bone min­ 172. O’Connor KL, Scisco JL, Smith TJ, et al. Altered appetite-
eral density in high school athletes: a prospective study. J Athl mediating hormone concentrations precede compensatory over­
Train. 2010;45(3):243-252. eating after severe, short-term energy deprivation in healthy
154. Miralles-Amorós L, Asencio-Mas N, Martínez-Olcina M, et al. adults. J Nutr. 2016;146(2):209-217.
Study the effect of relative energy deficiency on physiological 173. Karl JP, Smith TJ, Wilson MA, et al. Altered metabolic homeosta­
and physical variables in professional women athletes: a random­ sis is associated with appetite regulation during and following
ized controlled trial. Metabolites. 2023;13(2):168. 48-h of severe energy deprivation in adults. Metabolism.
155. Jurov I, Keay N, Spudic̀ D, Rauter S. Inducing low energy avail­ 2016;65(4):416-427.
ability in trained endurance male athletes results in poorer explo­ 174. Cahill GF Jr. Starvation in man. N Engl J Med. 1970;282(12):
sive power. Eur J Appl Physiol. 2022;122(2):503-513. 668-675.
156. Leal LG, Lopes MA, Batista ML Jr. Physical exercise-induced my­ 175. Boyle PJ, Shah SD, Cryer PE. Insulin, glucagon, and catechol­
okines and muscle-adipose tissue crosstalk: a review of current amines in prevention of hypoglycemia during fasting. Am J
knowledge and the implications for health and metabolic diseases. Physiol. 1989;256(5 Pt 1):E651-E661.
Front Physiol. 2018;9:1307. 176. Steinhauser ML, Olenchock BA, O’Keefe J, et al. The circulating
157. Chow LS, Gerszten RE, Taylor JM, et al. Exerkines in health, re­ metabolome of human starvation. JCI Insight. 2018;3(16):
silience and disease. Nat Rev Endocrinol. 2022;18(5):273-289. e121434.
158. Kojima C, Ishibashi A, Tanabe Y, et al. Muscle glycogen content 177. Cahill GF Jr. Fuel metabolism in starvation. Annu Rev Nutr.
during endurance training under low energy availability. Med 2006;26(1):1-22.
Sci Sports Exerc. 2020;52(1):187-195. 178. Nykänen T, Ojanen T, Heikkinen R, Fogelholm M, Kyröläinen H.
159. Areta JL, Burke LM, Camera DM, et al. Reduced resting skeletal Changes in body composition, energy metabolites and electrolytes
muscle protein synthesis is rescued by resistance exercise and pro­ during winter survival training in male soldiers. Front Physiol.
tein ingestion following short-term energy deficit. Am J Physiol 2022;13:797268.
Endocrinol Metab. 2014;306(8):E989-E997. 179. Gavrila A, Chan JL, Yiannakouris N, et al. Serum adiponectin lev­
160. Bouzoni E, Perakakis N, Mantzoros CS. Circulating profile of els are inversely associated with overall and central fat distribution
activin-follistatin-inhibin axis in women with hypothalamic but are not directly regulated by acute fasting or leptin administra­
amenorrhea in response to leptin treatment. Metab Clin Exp. tion in humans: cross-sectional and interventional studies. J Clin
2020;113:154392. Endocrinol Metab. 2003;88(10):4823-4831.
161. Polyzos SA, Perakakis N, Boutari C, et al. Targeted analysis of 180. Pasiakos SM, Caruso CM, Kellogg MD, Kramer FM, Lieberman
three hormonal systems identifies molecules associated with the HR. Appetite and endocrine regulators of energy balance after 2
presence and severity of NAFLD. J Clin Endocrinol Metab. days of energy restriction: insulin, leptin, ghrelin, and DHEA-S.
2020;105(3):e390-e400. Obesity (Silver Spring). 2011;19(6):1124-1130.
162. Melin AK, Areta JL, Heikura IA, Stellingwerff T, Torstveit MK, 181. De Souza MJ, Leidy HJ, O’Donnell E, Lasley B, Williams NI.
Hackney AC. Direct and indirect impact of low energy availability Fasting ghrelin levels in physically active women: relationship
28 Endocrine Reviews, 2024, Vol. 00, No. 0

with menstrual disturbances and metabolic hormones. J Clin 202. Torstveit MK, Ackerman KE, Constantini N, et al. Primary, sec­
Endocrinol Metab. 2004;89(7):3536-3542. ondary and tertiary prevention of Relative Energy Deficiency in
182. Sumithran P, Prendergast LA, Delbridge E, et al. Long-term per­ Sport (REDs): a narrative review by a subgroup of the IOC con­
sistence of hormonal adaptations to weight loss. N Engl J Med. sensus on REDs. Br J Sports Med. 2023;57(17):1119-1126.
2011;365(17):1597-1604. 203. Shimizu K, Suzuki N, Nakamura M, et al. Mucosal immune func­
183. Sygo J, Coates AM, Sesbreno E, Mountjoy ML, Burr JF. tion comparison between amenorrheic and eumenorrheic distance
Prevalence of indicators of low energy availability in elite female runners. J Strength Condition Res. 2012;26(5):1402-1406.
sprinters. Int J Sport Nutr Exerc Metab. 2018;28(5):490-496. 204. Abedelmalek S, Chtourou H, Souissi N, Tabka Z. Caloric restric­
184. Hackney AC. Hypogonadism in exercising males: dysfunction or tion effect on proinflammatory cytokines, growth hormone, and
adaptive-regulatory adjustment? Front Endocrinol (Lausanne). steroid hormone concentrations during exercise in judokas.
2020;11:11. Oxid Med Cell Longev. 2015;2015:809492.

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


185. Hamarsland H, Paulsen G, Solberg PA, Slaathaug OG, Raastad T. 205. Horn PL, Pyne DB, Hopkins WG, Barnes CJ. Lower white blood
Depressed physical performance outlasts hormonal disturbances cell counts in elite athletes training for highly aerobic sports. Eur J
after military training. Med Sci Sports Exerc. 2018;50(10): Appl Physiol. 2010;110(5):925-932.
2076-2084. 206. Kramer TR, Moore RJ, Shippee RL, et al. Effects of food restric­
186. Nindl BC, Alemany JA, Kellogg MD, et al. Utility of circulating tion in military training on T-lymphocyte responses. Int J Sports
IGF-I as a biomarker for assessing body composition changes in Med. 1997;18(Suppl 1):S84-S90.
men during periods of high physical activity superimposed upon en­ 207. Dubnov G, Constantini NW. Prevalence of iron depletion and an­
ergy and sleep restriction. J Appl Physiol (1985). 2007;103(1): emia in top-level basketball players. Int J Sport Nutr Exerc Metab.
340-346. 2004;14(1):30-37.
187. Kyröläinen H, Karinkanta J, Santtila M, Koski H, Mäntysaari M, 208. Ackerman KE, Holtzman B, Cooper KM, et al. Low energy avail­
Pullinen T. Hormonal responses during a prolonged military field ability surrogates correlate with health and performance conse­
exercise with variable exercise intensity. Eur J Appl Physiol. quences of Relative Energy Deficiency in Sport. Br J Sports Med.
2008;102(5):539-546. 2019;53(10):628-633.
188. Salonen M, Huovinen J, Kyröläinen H, Piirainen JM, Vaara JP. 209. Petkus DL, Murray-Kolb LE, De Souza MJ. The unexplored cross­
Neuromuscular performance and hormonal profile during mili­ roads of the female athlete triad and iron deficiency: a narrative re­
view. Sports Med. 2017;47(9):1721-1737.
tary training and subsequent recovery period. Mil Med.
210. Dubnov G, Foldes AJ, Mann G, Magazanik A, Siderer M,
2019;184(3-4):e113-e119.
Constantini N. High prevalence of iron deficiency and anemia in
189. Heikura IA, Uusitalo ALT, Stellingwerff T, Bergland D, Mero AA,
female military recruits. Mil Med. 2006;171(9):866-869.
Burke LM. Low energy availability is difficult to assess but out­
211. Epstein D, Borohovitz A, Merdler I, et al. Prevalence of iron defi­
comes have large impact on bone injury rates in elite distance ath­
ciency and iron deficiency anemia in strenuously training male
letes. Int J Sport Nutr Exerc Metab. 2018;28(4):403-411.
army recruits. Acta Haematol. 2018;139(3):141-147.
190. Fazeli PK, Klibanski A. Determinants of GH resistance in malnu­
212. Yanovich R, Merkel D, Israeli E, Evans RK, Erlich T, Moran DS.
trition. J Endocrinol. 2014;220(3):R57-R65.
Anemia, iron deficiency, and stress fractures in female combat­
191. Davey HW, Xie T, McLachlan MJ, Wilkins RJ, Waxman DJ,
ants during 16 months. J Strength Condition Res. 2011;25(12):
Grattan DR. STAT5b is required for GH-induced liver IGF-I
3412-3421.
gene expression. Endocrinology. 2001;142(9):3836-3841.
213. Yanovich R, Karl JP, Yanovich E, et al. Effects of basic combat
192. Nindl BC, Castellani JW, Young AJ, et al. Differential responses of
training on iron status in male and female soldiers: a comparative
IGF-I molecular complexes to military operational field training. J
study. US Army Med Dep J. 2015;Apr-Jun:67-73.
Appl Physiol (1985). 2003;95(3):1083-1089.
214. McClung JP, Marchitelli LJ, Friedl KE, Young AJ. Prevalence of
193. Gomez-Merino D, Chennaoui M, Drogou C, Guezennec CY.
iron deficiency and iron deficiency anemia among three popula­
Influence of energy deficiency on the insulin-like growth factor I tions of female military personnel in the US army. J Am Coll
axis in a military training program. Horm Metab Res. 2004;36(7): Nutr. 2006;25(1):64-69.
506-511. 215. McKay AKA, Pyne DB, Burke LM, Peeling P. Iron metabolism: in­
194. Misra M, Klibanski A. Endocrine consequences of anorexia ner­ teractions with energy and carbohydrate availability. Nutrients.
vosa. Lancet Diabetes Endocrinol. 2014;2(7):581-592. 2020;12(12):3692.
195. Fazeli PK, Lawson EA, Prabhakaran R, et al. Effects of recombin­ 216. Dave SC, Fisher M. Relative energy deficiency in sport (RED - S).
ant human growth hormone in anorexia nervosa: a randomized, Curr Probl Pediatr Adolesc Health Care. 2022;52(8):101242.
placebo-controlled study. J Clin Endocrinol Metab. 2010;95(11): 217. Berthou C, Iliou JP, Barba D. Iron, neuro-bioavailability and de­
4889-4897. pression. eJHaem. 2022;3(1):263-275.
196. Georgopoulos NA, Rottstein L, Tsekouras A, et al. Abolished cir­ 218. Barbarich NC, Kaye WH, Jimerson D. Neurotransmitter and im­
cadian rhythm of salivary cortisol in elite artistic gymnasts. aging studies in anorexia nervosa: new targets for treatment. Curr
Steroids. 2011;76(4):353-357. Drug Targets CNS Neurol Disord. 2003;2(1):61-72.
197. Charmandari E, Tsigos C, Chrousos G. Endocrinology of the 219. Kaye W. Neurobiology of anorexia and bulimia nervosa. Physiol
stress response. Annu Rev Physiol. 2005;67(1):259-284. Behav. 2008;94(1):121-135.
198. Cho GJ, Han SW, Shin J-H, Kim T. Effects of intensive training on 220. Peuranpää P, Heliövaara-Peippo S, Fraser I, Paavonen J,
menstrual function and certain serum hormones and peptides re­ Hurskainen R. Effects of anemia and iron deficiency on quality
lated to the female reproductive system. Medicine (Baltimore). of life in women with heavy menstrual bleeding. Acta Obstet
2017;96(21):e6876. Gynecol Scand. 2014;93(7):654-660.
199. Gifford RM, O’Leary TJ, Double RL, et al. Positive adaptation of 221. Wang H, Leng Y, Gong Y. Bone marrow fat and hematopoiesis.
HPA axis function in women during 44 weeks of infantry-based Front Endocrinol (Lausanne). 2018;9:694.
military training. Psychoneuroendocrinology. 2019;110:104432. 222. Zhong L, Yao L, Tower RJ, et al. Single cell transcriptomics iden­
200. Vikmoen O, Teien HK, Raustøl M, et al. Sex differences in the tifies a unique adipose lineage cell population that regulates bone
physiological response to a demanding military field exercise. marrow environment. Elife. 2020;9:e54695.
Scand J Med Sci Sports. 2020;30(8):1348-1359. 223. Deb A. How stem cells turn into bone and fat. N Engl J Med.
201. Koski KG, Su Z, Scott ME. Energy deficits suppress both systemic 2019;380(23):2268-2270.
and gut immunity during infection. Biochem Biophys Res 224. Devlin MJ. Why does starvation make bones fat? Am J Hum Biol.
Commun. 1999;264(3):796-801. 2011;23(5):577-585.
Endocrine Reviews, 2024, Vol. 00, No. 0 29

225. Devlin MJ, Cloutier AM, Thomas NA, et al. Caloric restriction redistribution and fat mass loss: a review. Nutrients.
leads to high marrow adiposity and low bone mass in growing 2020;12(6):1899.
mice. J Bone Miner Res. 2010;25(9):2078-2088. 246. Samuelson I, Vidal-Puig A. Studying brown adipose tissue in a hu­
226. Tavassoli M. Differential response of bone marrow and extrame­ man in vitro context. Front Endocrinol (Lausanne). 2020;11:629.
dullary adipose cells to starvation. Experientia. 1974;30(4): 247. Foster DO, Frydman ML. Tissue distribution of cold-induced
424-425. thermogenesis in conscious warm- or cold-acclimated rats reeval­
227. O’Donnell E, Goodman JM, Mak S, et al. Discordant orthostatic uated from changes in tissue blood flow: the dominant role of
reflex renin-angiotensin and sympathoneural responses in preme­ brown adipose tissue in the replacement of shivering by nonshiv­
nopausal exercising-hypoestrogenic women. Hypertension. ering thermogenesis. Can J Physiol Pharmacol. 1979;57(3):
2015;65(5):1089-1095. 257-270.
228. Tomes C, Schram B, Orr R. Relationships between heart rate vari­ 248. Leitner BP, Huang S, Brychta RJ, et al. Mapping of human brown

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


ability, occupational performance, and fitness for tactical person­ adipose tissue in lean and obese young men. Proc Natl Acad Sci U
nel: a systematic review. Front Public Health. 2020;8:583336. S A. 2017;114(32):8649-8654.
229. Schmitt L, Regnard J, Desmarets M, et al. Fatigue shifts and scat­ 249. Aldiss P, Betts J, Sale C, Pope M, Budge H, Symonds ME.
ters heart rate variability in elite endurance athletes. PLoS One. Exercise-induced ‘browning’ of adipose tissues. Metabolism.
2013;8(8):e71588. 2018;81:63-70.
230. Costa RJS, Snipe RMJ, Kitic CM, Gibson PR. Systematic review: 250. Vosselman MJ, Hoeks J, Brans B, et al. Low brown adipose tissue
exercise-induced gastrointestinal syndrome—implications for health activity in endurance-trained compared with lean sedentary men.
and intestinal disease. Aliment Pharmacol Ther. 2017;46(3): Int J Obesity. 2015;39(12):1696-1702.
246-265. 251. Singhal V, Maffazioli GD, Ackerman KE, et al. Effect of chronic
231. Pugh JN, Lydon KM, O’Donovan CM, O’Sullivan O, Madigan athletic activity on brown fat in young women. PLoS One.
SM. More than a gut feeling: what is the role of the gastrointestinal 2016;11(5):e0156353.
tract in female athlete health? Eur J Sport Sci. 2022;22(5): 252. Picó C, Palou M, Pomar CA, Rodríguez AM, Palou A. Leptin as a
755-764. key regulator of the adipose organ. Rev Endocr Metab Disord.
232. Zhang B, Verne ML, Fields JZ, Verne GN, Zhou Q. Intestinal hy­ 2022;23(1):13-30.
perpermeability in gulf war veterans with chronic gastrointestinal 253. Perakakis N, Farr OM, Mantzoros CS. Leptin in leanness and
symptoms. J Clin Gastroenterol. 2019;53(7):e298-e302. obesity: JACC state-of-the-art review. J Am Coll Cardiol.
233. Putnam SD, Sanders JW, Frenck RW, et al. Self-reported descrip­ 2021;77(6):745-760.
tion of diarrhea among military populations in operations Iraqi 254. Mantzoros CS. The role of leptin in human obesity and disease: a
freedom and enduring freedom. J Travel Med. 2006;13(2):92-99. review of current evidence. Ann Intern Med. 1999;130(8):
234. Kimono DA. Gastrointestinal problems, mechanisms and possible 671-680.
therapeutic directions in Gulf war illness: a mini review. Mil Med 255. Licinio J, Negrão AB, Mantzoros C, et al. Sex differences in circu­
Res. 2021;8(1):50. lating human leptin pulse amplitude: clinical implications. J Clin
235. Farina EK, Taylor JC, Means GE, et al. Effects of combat deploy­ Endocrinol Metab. 1998;83(11):4140-4147.
ment on anthropometrics and physiological status of U.S. army 256. Faouzi M, Leshan R, Björnholm M, Hennessey T, Jones J,
special operations forces soldiers. Mil Med. 2017;182(3): Münzberg H. Differential accessibility of circulating leptin to indi­
e1659-e1668. vidual hypothalamic sites. Endocrinology. 2007;148(11):
236. Sedliak M, Sedliak P, Vaara JP. Effects of 6-month military de­ 5414-5423.
ployment on physical fitness, body composition, and selected 257. Vaisse C, Halaas JL, Horvath CM, Darnell JE Jr, Stoffel M,
health-related biomarkers. J Strength Cond Res. 2021;35(4): Friedman JM. Leptin activation of Stat3 in the hypothalamus of
1074-1081. wild-type and ob/ob mice but not db/db mice. Nat Genet.
237. Burd NA, West DW, Moore DR, et al. Enhanced amino acid sen­ 1996;14(1):95-97.
sitivity of myofibrillar protein synthesis persists for up to 24 h 258. Baver SB, Hope K, Guyot S, Bjørbaek C, Kaczorowski C,
after resistance exercise in young men. J Nutr. 2011;141(4): O’Connell KM. Leptin modulates the intrinsic excitability of
568-573. AgRP/NPY neurons in the arcuate nucleus of the hypothalamus.
238. Philp A, Hargreaves M, Baar K. More than a store: regulatory J Neurosci. 2014;34(16):5486-5496.
roles for glycogen in skeletal muscle adaptation to exercise. Am 259. Farr OM, Li CR, Mantzoros CS. Central nervous system regula­
J Physiol Endocrinol Metab. 2012;302(11):E1343-E1351. tion of eating: insights from human brain imaging. Metabolism.
239. Phillips SM. Dietary protein requirements and adaptive advan­ 2016;65(5):699-713.
tages in athletes. Br J Nutr. 2012;108(Suppl 2):S158-S167. 260. Cohen P, Zhao C, Cai X, et al. Selective deletion of leptin receptor
240. Reed JL, De Souza MJ, Kindler JM, Williams NI. Nutritional in neurons leads to obesity. J Clin Invest. 2001;108(8):1113-1121.
practices associated with low energy availability in division I fe­ 261. Halaas JL, Gajiwala KS, Maffei M, et al. Weight-reducing effects
male soccer players. J Sports Sci. 2014;32(16):1499-1509. of the plasma protein encoded by the obese gene. Science.
241. Margolis LM, Murphy NE, Martini S, et al. Effects of winter mili­ 1995;269(5223):543-546.
tary training on energy balance, whole-body protein balance, 262. Wong SL, DePaoli AM, Lee JH, Mantzoros CS. Leptin hormonal
muscle damage, soreness, and physical performance. Appl kinetics in the fed state: effects of adiposity, age, and gender on en­
Physiol Nutr Metab. 2014;39(12):1395-1401. dogenous leptin production and clearance rates. J Clin Endocrinol
242. Henning PC, Park BS, Kim JS. Physiological decrements during Metab. 2004;89(6):2672-2677.
sustained military operational stress. Mil Med. 2011;176(9): 263. Chan JL, Wong SL, Mantzoros CS. Pharmacokinetics of subcuta­
991-997. neous recombinant methionyl human leptin administration in
243. Matheson GO, Clement DB, Mckenzie DC, Taunton JE, healthy subjects in the fed and fasting states: regulation by gender
Lloyd-Smith DR, Macintyre JG. Stress fractures in athletes: a and adiposity. Clin Pharmacokinet. 2008;47(11):753-764.
study of 320 cases. Am J Sports Med. 1987;15(1):46-58. 264. Boden G, Chen X, Mozzoli M, Ryan I. Effect of fasting on serum
244. Abbott A, Wang C, Stamm M, Mulcahey MK. Part I: background leptin in normal human subjects. J Clin Endocrinol Metab.
and clinical considerations for stress fractures in female military 1996;81(9):3419-3423.
recruits. Mil Med. 2023;188(1-2):86-92. 265. Wolfe BE, Jimerson DC, Orlova C, Mantzoros CS. Effect of diet­
245. Gonzalez-Gil AM, Elizondo-Montemayor L. The role of exercise ing on plasma leptin, soluble leptin receptor, adiponectin and re­
in the interplay between myokines, hepatokines, osteokines, adi­ sistin levels in healthy volunteers. Clin Endocrinol (Oxf).
pokines, and modulation of inflammation for energy substrate 2004;61(3):332-338.
30 Endocrine Reviews, 2024, Vol. 00, No. 0

266. Jimerson DC, Mantzoros C, Wolfe BE, Metzger ED. Decreased se­ 285. Chrysafi P, Perakakis N, Farr OM, et al. Leptin alters energy in­
rum leptin in bulimia nervosa. J Clin Endocrinol Metab. take and fat mass but not energy expenditure in lean subjects.
2000;85(12):4511-4514. Nat Commun. 2020;11(1):5145.
267. Audi L, Mantzoros CS, Vidal-Puig A, Vargas D, Gussinye M, 286. Kiernan K, MacIver NJ. The role of the adipokine leptin in im­
Carrascosa A. Leptin in relation to resumption of menses in wom­ mune cell function in health and disease. Front Immunol.
en with anorexia nervosa. Mol Psychiatry. 1998;3(6):544-547. 2021;11:622468.
268. Lebrethon MC, Vandersmissen E, Gérard A, Parent AS, Junien JL, 287. Matarese G, Moschos S, Mantzoros CS. Leptin in immunology. J
Bourguignon JP. In vitro stimulation of the prepubertal rat Immunol. 2005;174(6):3137-3142.
gonadotropin-releasing hormone pulse generator by leptin and 288. Agrawal S, Gollapudi S, Su H, Gupta S. Leptin activates human B
neuropeptide Y through distinct mechanisms. Endocrinology. cells to secrete TNF-α, IL-6, and IL-10 via JAK2/STAT3 and
2000;141(4):1464-1469. p38MAPK/ERK1/2 signaling pathway. J Clin Immunol.

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


269. Lehman MN, Coolen LM, Goodman RL. Minireview: kisspeptin/ 2011;31(3):472-478.
neurokinin B/dynorphin (KNDy) cells of the arcuate nucleus: a 289. Lord GM, Matarese G, Howard JK, Baker RJ, Bloom SR, Lechler
central node in the control of gonadotropin-releasing hormone se­ RI. Leptin modulates the T-cell immune response and reverses
cretion. Endocrinology. 2010;151(8):3479-3489. starvation-induced immunosuppression. Nature. 1998;394(6696):
270. Chehab FF, Lim ME, Lu R. Correction of the sterility defect in 897-901.
homozygous obese female mice by treatment with the human re­ 290. Chan JL, Moschos SJ, Bullen J, et al. Recombinant methionyl hu­
combinant leptin. Nat Genet. 1996;12(3):318-320. man leptin administration activates signal transducer and activa­
271. Ahima RS, Prabakaran D, Mantzoros C, et al. Role of leptin in the tor of transcription 3 signaling in peripheral blood mononuclear
neuroendocrine response to fasting. Nature. 1996;382(6588): cells in vivo and regulates soluble tumor necrosis factor-alpha re­
250-252. ceptor levels in humans with relative leptin deficiency. J Clin
272. Farooqi IS, Jebb SA, Langmack G, et al. Effects of recombinant Endocrinol Metab. 2005;90(3):1625-1631.
leptin therapy in a child with congenital leptin deficiency. N 291. Matarese G, La Rocca C, Moon HS, et al. Selective capacity of me­
Engl J Med. 1999;341(12):879-884. treleptin administration to reconstitute CD4+ T-cell number in fe­
273. Farooqi IS, Matarese G, Lord GM, et al. Beneficial effects of leptin males with acquired hypoleptinemia. Proc Natl Acad Sci U S A.
on obesity, T cell hyporesponsiveness, and neuroendocrine/meta­ 2013;110(9):E818-E827.
bolic dysfunction of human congenital leptin deficiency. J Clin 292. Gainsford T, Willson TA, Metcalf D, et al. Leptin can induce pro­
Invest. 2002;110(8):1093-1103. liferation, differentiation, and functional activation of hemopoi­
274. Licinio J, Caglayan S, Ozata M, et al. Phenotypic effects of leptin etic cells. Proc Natl Acad Sci U S A. 1996;93(25):14564-14568.
replacement on morbid obesity, diabetes mellitus, hypogonadism, 293. Bennett BD, Solar GP, Yuan JQ, Mathias J, Thomas GR,
and behavior in leptin-deficient adults. Proc Natl Acad Sci U S A. Matthews W. A role for leptin and its cognate receptor in hemato­
2004;101(13):4531-4536. poiesis. Curr Biol. 1996;6(9):1170-1180.
275. Nindl BC, Friedl KE, Frykman PN, Marchitelli LJ, Shippee RL, 294. Chung B, Matak P, McKie AT, Sharp P. Leptin increases the ex­
Patton JF. Physical performance and metabolic recovery among pression of the iron regulatory hormone hepcidin in HuH7 human
lean, healthy men following a prolonged energy deficit. Int J hepatoma cells. J Nutr. 2007;137(11):2366-2370.
Sports Med. 1997;18(5):317-324. 295. Trinh T, Ropa J, Aljoufi A, et al. Leptin receptor, a surface marker
276. Kim MS, Small CJ, Stanley SA, et al. The central melanocortin sys­ for a subset of highly engrafting long-term functional hematopoi­
tem affects the hypothalamo-pituitary thyroid axis and may medi­ etic stem cells. Leukemia. 2021;35(7):2064-2075.
ate the effect of leptin. J Clin Invest. 2000;105(7):1005-1011. 296. Rizzoli R, Biver E, Brennan-Speranza TC. Nutritional intake and
277. Clement K, Vaisse C, Lahlou N, et al. A mutation in the human bone health. Lancet Diabetes Endocrinol. 2021;9(9):606-621.
leptin receptor gene causes obesity and pituitary dysfunction. 297. Boyce BF, Xing L. Biology of RANK, RANKL, and osteoproteger­
Nature. 1998;392(6674):398-401. in. Arthritis Res Ther. 2007;9(Suppl 1):S1.
278. Shetty GK, Matarese G, Magkos F, et al. Leptin administration to 298. Li X, Zhang Y, Kang H, et al. Sclerostin binds to LRP5/6 and an­
overweight and obese subjects for 6 months increases free leptin tagonizes canonical Wnt signaling. J Biol Chem. 2005;280(20):
concentrations but does not alter circulating hormones of the thy­ 19883-19887.
roid and IGF axes during weight loss induced by a mild hypocalor­ 299. Bord S, Ireland DC, Beavan SR, Compston JE. The effects of estro­
ic diet. Eur J Endocrinol. 2011;165(2):249-254. gen on osteoprotegerin, RANKL, and estrogen receptor expres­
279. Giusti M, Bocca L, Florio T, et al. In vitro effect of human recom­ sion in human osteoblasts. Bone. 2003;32(2):136-141.
binant leptin and expression of leptin receptors on growth 300. Shevde NK, Bendixen AC, Dienger KM, Pike JW. Estrogens sup­
hormone-secreting human pituitary adenomas. Clin Endocrinol press RANK ligand-induced osteoclast differentiation via a stro­
(Oxf). 2002;57(4):449-455. mal cell independent mechanism involving c-Jun repression.
280. LaPaglia N, Steiner J, Kirsteins L, Emanuele M, Emanuele N. Proc Natl Acad Sci U S A. 2000;97(14):7829-7834.
Leptin alters the response of the growth hormone releasing factor- 301. Streicher C, Heyny A, Andrukhova O, et al. Estrogen regulates
growth hormone–insulin-like growth factor-I axis to fasting. J bone turnover by targeting RANKL expression in bone lining
Endocrinol. 1998;159(1):79-83. cells. Sci Rep. 2017;7(1):6460.
281. Heiman ML, Ahima RS, Craft LS, Schoner B, Stephens TW, Flier 302. Komori T. Glucocorticoid signaling and bone biology. Horm
JS. Leptin inhibition of the hypothalamic-pituitary-adrenal axis in Metab Res. 2016;48(11):755-763.
response to stress. Endocrinology. 1997;138(9):3859-3863. 303. Pralong FP, Roduit R, Waeber G, et al. Leptin inhibits directly
282. Haynes WG, Morgan DA, Walsh SA, Mark AL, Sivitz WI. glucocorticoid secretion by normal human and rat adrenal gland.
Receptor-mediated regional sympathetic nerve activation by lep­ Endocrinology. 1998;139(10):4264-4268.
tin. J Clin Invest. 1997;100(2):270-278. 304. Korner J, Conroy R, Febres G, et al. Randomized double-blind
283. Enriori PJ, Sinnayah P, Simonds SE, Garcia Rudaz C, Cowley MA. placebo-controlled study of leptin administration after gastric by­
Leptin action in the dorsomedial hypothalamus increases sympa­ pass. Obesity (Silver Spring). 2013;21(5):951-956.
thetic tone to brown adipose tissue in spite of systemic leptin re­ 305. Khundmiri SJ, Murray RD, Lederer E. PTH and vitamin D.
sistance. J Neurosci. 2011;31(34):12189-12197. Compr Physiol. 2016;6(2):561-601.
284. Ducy P, Amling M, Takeda S, et al. Leptin inhibits bone formation 306. Lombardi G, Ziemann E, Banfi G, Corbetta S. Physical activity-
through a hypothalamic relay: a central control of bone mass. dependent regulation of parathyroid hormone and calcium-
Cell. 2000;100(2):197-207. phosphorous metabolism. Int J Mol Sci. 2020;21(15):5388.
Endocrine Reviews, 2024, Vol. 00, No. 0 31

307. Ribbans WJ, Aujla R, Dalton S, Nunley JA. Vitamin D and the 329. Elefteriou F, Ahn JD, Takeda S, et al. Leptin regulation of bone re­
athlete–patient: state of the art. J ISAKOS. 2021;6(1):46-60. sorption by the sympathetic nervous system and CART. Nature.
308. Noirrit-Esclassan E, Valera M-C, Tremollieres F, et al. Critical role 2005;434(7032):514-520.
of estrogens on bone homeostasis in both male and female: from 330. Foo JP, Polyzos SA, Anastasilakis AD, Chou S, Mantzoros CS.
physiology to medical implications. Int J Mol Sci. 2021;22(4):1568. The effect of leptin replacement on parathyroid hormone,
309. Aalberg K, Stavem K, Norheim F, Russell MB, Chaibi A. Effect of RANKL-osteoprotegerin axis, and Wnt inhibitors in young wom­
oral and transdermal oestrogen therapy on bone mineral density in en with hypothalamic amenorrhea. J Clin Endocrinol Metab.
functional hypothalamic amenorrhoea: a systematic review and 2014;99(11):E2252-E2258.
meta-analysis. BMJ Open Sport Exerc Med. 2021;7(3):e001112. 331. Barrack MT, Van Loan MD, Rauh MJ, Nichols JF. Physiologic
310. Southmayd EA, Mallinson RJ, Williams NI, Mallinson DJ, De and behavioral indicators of energy deficiency in female adoles­
Souza MJ. Unique effects of energy versus estrogen deficiency on cent runners with elevated bone turnover. Am J Clin Nutr.

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


multiple components of bone strength in exercising women. 2010;92(3):652-659.
Osteoporos Int. 2017;28(4):1365-1376. 332. Pogoda P, Egermann M, Schnell JC, et al. Leptin inhibits bone for­
311. Meczekalski B, Podfigurna-Stopa A, Genazzani AR. mation not only in rodents, but also in sheep. J Bone Miner Res.
Hypoestrogenism in young women and its influence on bone 2006;21(10):1591-1599.
mass density. Gynecol Endocrinol. 2010;26(9):652-657. 333. Cento RM, Proto C, Spada RS, et al. Leptin levels in menopause:
312. De Souza MJ, West SL, Jamal SA, Hawker GA, Gundberg CM, effect of estrogen replacement therapy. Horm Res. 1999;52(6):
Williams NI. The presence of both an energy deficiency and estro­ 269-273.
gen deficiency exacerbate alterations of bone metabolism in exer­ 334. Yu WH, Kimura M, Walczewska A, Karanth S, McCann SM.
cising women. Bone. 2008;43(1):140-148. Role of leptin in hypothalamic-pituitary function. Proc Natl
313. Clarke BL, Khosla S. Androgens and bone. Steroids. 2009;74(3): Acad Sci U S A. 1997;94(3):1023-1028.
296-305. 335. Blüher S, Mantzoros CS. Leptin in reproduction. Curr Opin
314. Niu T, Rosen CJ. The insulin-like growth factor-I gene and osteo­ Endocrinol Diabetes Obes. 2007;14(6):458-464.
porosis: a critical appraisal. Gene. 2005;361:38-56. 336. Hamrick MW, Pennington C, Newton D, Xie D, Isales C. Leptin
315. Giustina A, Mazziotti G, Canalis E. Growth hormone, insulin-like deficiency produces contrasting phenotypes in bones of the limb
growth factors, and the skeleton. Endocr Rev. 2008;29(5): and spine. Bone. 2004;34(3):376-383.
535-559. 337. Philbrick KA, Wong CP, Branscum AJ, Turner RT, Iwaniec UT.
316. Crane JL, Zhao L, Frye JS, Xian L, Qiu T, Cao X. IGF-1 signaling
Leptin stimulates bone formation in ob/ob mice at doses having
is essential for differentiation of mesenchymal stem cells for peak
minimal impact on energy metabolism. J Endocrinol.
bone mass. Bone Res. 2013;1(2):186-194.
2017;232(3):461-474.
317. Locatelli V, Bianchi VE. Effect of GH/IGF-1 on bone metabolism
338. Brinkoetter M, Magkos F, Vamvini M, Mantzoros CS. Leptin
and osteoporsosis. Int J Endocrinol. 2014;2014:235060.
treatment reduces body fat but does not affect lean body mass
318. Tsunashima Y, Kondo A, Matsuda T, Togari A. Hydrocortisone
or the myostatin-follistatin-activin axis in lean hypoleptinemic
inhibits cellular proliferation by downregulating hepatocyte
women. Am J Physiol Endocrinol Metab. 2011;301(1):E99-E104.
growth factor synthesis in human osteoblasts. Biol Pharm Bull.
339. Namwanje M, Brown CW. Activins and inhibins: roles in develop­
2011;34(5):700-703.
ment, physiology, and disease. Cold Spring Harb Perspect Biol.
319. Misra M, Miller KK, Almazan C, et al. Alterations in cortisol se­
2016;8(7):a021881.
cretory dynamics in adolescent girls with anorexia nervosa and ef­
340. Wijayarathna R, de Kretser DM. Activins in reproductive biology
fects on bone metabolism. J Clin Endocrinol Metab. 2004;89(10):
and beyond. Hum Reprod Update. 2016;22(3):342-357.
4972-4980.
341. Makanji Y, Zhu J, Mishra R, et al. Inhibin at 90: from discovery to
320. McCarthy TL, Centrella M, Canalis E. Cortisol inhibits the syn­
clinical application, a historical review. Endocr Rev. 2014;35(5):
thesis of insulin-like growth factor-I in skeletal cells.
Endocrinology. 1990;126(3):1569-1575. 747-794.
321. McCosh RB, Breen KM, Kauffman AS. Neural and endocrine 342. Lee S-J, Lee Y-S, Zimmers TA, et al. Regulation of muscle mass by
mechanisms underlying stress-induced suppression of pulsatile follistatin and activins. Mol Endocrinol. 2010;24(10):1998-2008.
LH secretion. Mol Cell Endocrinol. 2019;498:110579. 343. Mukherjee A, Sidis Y, Mahan A, et al. FSTL3 deletion reveals roles
322. Reid IR, Baldock PA, Cornish J. Effects of leptin on the Skeleton. for TGF-beta family ligands in glucose and fat homeostasis in
Endocr Rev. 2018;39(6):938-959. adults. Proc Natl Acad Sci U S A. 2007;104(4):1348-1353.
323. Fu L, Patel MS, Bradley A, Wagner EF, Karsenty G. The molecular 344. Moragianni VA, Aronis KN, Chamberland JP, Mantzoros CS.
clock mediates leptin-regulated bone formation. Cell. Short-term energy deprivation alters activin a and follistatin but
2005;122(5):803-815. not inhibin B levels of lean healthy women in a leptin-independent
324. Gordeladze JO, Drevon CA, Syversen U, Reseland JE. Leptin stim­ manner. J Clin Endocrinol Metab. 2011;96(12):3750-3758.
ulates human osteoblastic cell proliferation, de novo collagen syn­ 345. Gillbanks L, Mountjoy M, Filbay SR. Insufficient knowledge and
thesis, and mineralization: impact on differentiation markers, inapproriate physiotherapy management of Relative Energy
apoptosis, and osteoclastic signaling. J Cell Biochem. 2002;85(4): Deficiency in Sport (RED-S) in lightweight rowers. Phys Ther
825-836. Sport. 2022;54:8-15.
325. Ferron M, Lacombe J. Regulation of energy metabolism by the 346. Charlton BT, Forsyth S, Clarke DC. Low energy availability and
skeleton: osteocalcin and beyond. Arch Biochem Biophys. relative energy deficiency in sport: what coaches should know.
2014;561:137-146. Int J Sports Sci Coach. 2022;17(2):445-460.
326. Kalra SP, Dube MG, Iwaniec UT. Leptin increases osteoblast- 347. Lodge MT, Ackerman KE, Garay J. Knowledge of the female ath­
specific osteocalcin release through a hypothalamic relay. lete triad and relative energy deficiency in sport among female cross-
Peptides. 2009;30(5):967-973. country athletes and support staff. J Athl Train. 2022;57(4):
327. Li J, Gao Y, Yu T, et al. Obesity and leptin influence vitamin D me­ 385-392.
tabolism and action in human marrow stromal cells. J Steroid 348. Golden NH, Jacobson MS, Schebendach J, Solanto MV, Hertz
Biochem Mol Biol. 2020;198:105564. SM, Shenker IR. Resumption of menses in anorexia nervosa.
328. Tsuji K, Maeda T, Kawane T, Matsunuma A, Horiuchi N. Leptin Arch Pediatr Adolesc Med. 1997;151(1):16-21.
stimulates fibroblast growth factor 23 expression in bone and sup­ 349. Herpertz-Dahlmann BM, Wewetzer C, Schulz E, Remschmidt H.
presses renal 1alpha,25-dihydroxyvitamin D3 synthesis in leptin- Course and outcome in adolescent anorexia nervosa. Int J Eat
deficient mice. J Bone Miner Res. 2010;25(8):1711-1723. Disord. 1996;19(4):335-345.
32 Endocrine Reviews, 2024, Vol. 00, No. 0

350. Cacciatore C, Cangiano B, Carbone E, et al. Body weight variation 368. Ackerman KE, Singhal V, Baskaran C, et al. Oestrogen replacement
is not an independent factor in the determination of functional improves bone mineral density in oligo-amenorrhoeic athletes: a
hypothalamic amenorrhea in anorexia nervosa. J Endocrinol randomised clinical trial. Br J Sports Med. 2019;53(4):229-236.
Invest. 2024;47(4):903-911. 369. Southmayd EA, De Souza MJ. A summary of the influence of ex­
351. Mallinson RJ, Williams NI, Olmsted MP, Scheid JL, Riddle ES, De ogenous estrogen administration across the lifespan on the GH/
Souza MJ. A case report of recovery of menstrual function follow­ IGF-1 axis and implications for bone health. Growth Horm IGF
ing a nutritional intervention in two exercising women with amen­ Res. 2017;32:2-13.
orrhea of varying duration. J Int Soc Sports Nutr. 2013;10(1):34. 370. Lee JS, LaCroix AZ, Wu L, et al. Associations of serum sex
352. De Souza MJ, Mallinson RJ, Strock NCA, et al. Randomised con­ hormone-binding globulin and sex hormone concentrations with
trolled trial of the effects of increased energy intake on menstrual hip fracture risk in postmenopausal women. J Clin Endocrinol
recovery in exercising women with menstrual disturbances: the Metab. 2008;93(5):1796-1803.

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


‘REFUEL’ study. Hum Reprod. 2021;36(8):2285-2297. 371. Singhal V, Ackerman KE, Bose A, Flores LPT, Lee H, Misra M.
353. Kuikman MA, Mountjoy M, Stellingwerff T, Burr JF. A review of Impact of route of estrogen administration on bone turnover
nonpharmacological strategies in the treatment of relative energy de­ markers in oligoamenorrheic athletes and its mediators. J Clin
ficiency in sport. Int J Sport Nutr Exerc Metab. 2021;31(3):268-275. Endocrinol Metab. 2019;104(5):1449-1458.
354. Holtzman B, Ackerman KE. Recommendations and nutritional 372. Fazeli PK, Wang IS, Miller KK, et al. Teriparatide increases bone
considerations for female athletes: health and performance. formation and bone mineral density in adult women with anorexia
Sports Med. 2021;51(Suppl 1):43-57. nervosa. J Clin Endocrinol Metab. 2014;99(4):1322-1329.
355. Moreira CA, Bilezikian JP. Stress fractures: concepts and thera­ 373. Paik J, Scott LJ. Romosozumab: a review in postmenopausal
peutics. J Clin Endocrinol Metab. 2017;102(2):525-534. osteoporosis. Drugs Aging. 2020;37(11):845-855.
356. Richards T, Wright C. British army recruits with low serum vita­ 374. Shane E, Shiau S, Recker RR, et al. Denosumab after teriparatide
min D take longer to recover from stress fractures. BMJ Mil in premenopausal women with idiopathic osteoporosis. J Clin
Health. 2020;166(4):240-242. Endocrinol Metab. 2022;107(4):e1528-e1540.
357. Meeusen R, Duclos M, Foster C, et al. Prevention, diagnosis, and 375. Grinspoon S, Thomas L, Miller K, Herzog D, Klibanski A. Effects
treatment of the overtraining syndrome: joint consensus statement of recombinant human IGF-I and oral contraceptive administra­
of the European College of Sport Science and the American College tion on bone density in anorexia nervosa. J Clin Endocrinol
of Sports Medicine. Med Sci Sports Exerc. 2013;45(1):186-205. Metab. 2002;87(6):2883-2891.
358. American Psychiatric Association. Treatment of patients with eat­ 376. Svensson J, Lö nn L, Jansson J-O, et al. Two-month treatment of
ing disorders, third edition. American Psychiatric Association. Am obese subjects with the oral growth hormone (GH) secretagogue
J Psychiatry. 2006;163(7 Suppl):4-54. MK-677 increases GH secretion, fat-free mass, and energy ex­
359. Michopoulos V, Mancini F, Loucks TL, Berga SL. Neuroendocrine penditure. The J Clin Endocrinol Metab. 1998;83(2):362-369.
recovery initiated by cognitive behavioral therapy in women with 377. Svensson J, Ohlsson C, Jansson JO, et al. Treatment with the oral
functional hypothalamic amenorrhea: a randomized, controlled tri­ growth hormone secretagogue MK-677 increases markers of bone
al. Fertil Steril. 2013;99(7):2084-2091.e1. formation and bone resorption in obese young males. J Bone
360. National Collaborating Centre for Mental Health (UK). National Miner Res. 1998;13(7):1158-1166.
Institute for Health and Clinical Excellence: Guidance. Eating 378. Bach MA, Rockwood K, Zetterberg C, et al. The effects of
Disorders: Core Interventions in the Treatment and Management MK-0677, an oral growth hormone secretagogue, in patients
of Anorexia Nervosa, Bulimia Nervosa and Related Eating with hip fracture. J Am Geriatr Soc. 2004;52(4):516-523.
Disorders. British Psychological Society (UK); 2004. 379. Adunsky A, Chandler J, Heyden N, et al. MK-0677 (ibutamoren
361. Pitts S, Blood E, Divasta A, Gordon CM. Percentage body fat by mesylate) for the treatment of patients recovering from hip frac­
dual-energy X-ray absorptiometry is associated with menstrual re­ ture: a multicenter, randomized, placebo-controlled phase IIb
covery in adolescents with anorexia nervosa. J Adolesc Health. study. Arch Gerontol Geriatrics. 2011;53(2):183-189.
2014;54(6):739-741. 380. Rooks D, Swan T, Goswami B, et al. Bimagrumab vs optimized
362. Jacoangeli F, Masala S, Staar Mezzasalma F, et al. Amenorrhea standard of care for treatment of sarcopenia in community-dwelling
after weight recover in anorexia nervosa: role of body composition older adults: a randomized clinical trial. JAMA Netw Open.
and endocrine abnormalities. Eat Weight Disord. 2006;11(1): 2020;3(10):e2020836.
e20-e26. 381. Hofbauer LC, Witvrouw R, Varga Z, et al. Bimagrumab to im­
363. Stewart TM, Pollard T, Hildebrandt T, Wesley NY, Kilpela LS, prove recovery after hip fracture in older adults: a multicentre,
Becker CB. The Female Athlete Body project study: 18-month out­ double-blind, randomised, parallel-group, placebo-controlled,
comes in eating disorder symptoms and risk factors. Int J Eat phase 2a/b trial. Lancet Healthy Longevity. 2021;2(5):e263-e274.
Disord. 2019;52(11):1291-1300. 382. Heymsfield SB, Coleman LA, Miller R, et al. Effect of bimagrumab
364. Perelman H, Schwartz N, Yeoward-Dodson J, et al. Reducing eat­ vs placebo on body fat mass among adults with type 2 diabetes and
ing disorder risk among male athletes: a randomized controlled obesity: a phase 2 randomized clinical trial. JAMA Netw Open.
trial investigating the male athlete body project. Int J Eat 2021;4(1):e2033457-e2033457.
Disord. 2022;55(2):193-206. 383. Hirschberg AL, Elings Knutsson J, Helge T, et al. Effects of mod­
365. Fahrenholtz IL, Melin AK, Garthe I, et al. Short-term effects and erately increased testosterone concentration on physical perform­
long-term changes of FUEL-a digital sports nutrition intervention ance in young women: a double blind, randomised, placebo
on REDs related symptoms in female athletes. Front Sports Act controlled study. Br J Sports Med. 2020;54(10):599-604.
Living. 2023;5:1254210. 384. Fredericson M, Kent K. Normalization of bone density in a previ­
366. Hutson MJ, O’Donnell E, Brooke-Wavell K, et al. High-impact ously amenorrheic runner with osteoporosis. Med Sci Sports
jumping mitigates the short-term effects of low energy availability Exerc. 2005;37(9):1481-1486.
on bone resorption but not formation in regularly menstruating fe­ 385. Arends JC, Cheung MY, Barrack MT, Nattiv A. Restoration of
males: a randomized control trial. Scand J Med Sci Sports. menses with nonpharmacologic therapy in college athletes with
2023;33(9):1690-1702. menstrual disturbances: a 5-year retrospective study. Int J Sport
367. Stenqvist TB, Torstveit MK, Faber J, Melin AK. Impact of a Nutr Exerc Metab. 2012;22(2):98-108.
4-week intensified endurance training intervention on markers 386. Inoue DS, Panissa VL, Antunes BM, et al. Reduced leptin level is
of Relative Energy Deficiency in Sport (RED-S) and performance independent of fat mass changes and hunger scores from high-
among well-trained male cyclists. Front Endocrinol (Lausanne). intensity intermittent plus strength training. J Sports Med Phys
2020;11:512365. Fitness. 2018;58(7-8):1045-1051.
Endocrine Reviews, 2024, Vol. 00, No. 0 33

387. Ackerman KE, Slusarz K, Guereca G, et al. Higher ghrelin and 391. Triantafyllou GA, Paschou SA, Mantzoros CS. Leptin and hor­
lower leptin secretion are associated with lower LH secretion in mones: energy homeostasis. Endocrinol Metab Clin North Am.
young amenorrheic athletes compared with eumenorrheic athletes 2016;45(3):633-645.
and controls. Am J Physiol Endocrinol Metab. 2012;302(7): 392. Shan X, Yeo GSH. Central leptin and ghrelin signalling: compar­
E800-E806. ing and contrasting their mechanisms of action in the brain. Rev
388. Paz-Filho G, Mastronardi CA, Licinio J. Leptin treatment: facts Endocr Metab Disord. 2011;12(3):197-209.
and expectations. Metabolism. 2015;64(1):146-156. 393. Mantzoros CS, Magkos F, Brinkoetter M, et al. Leptin in human
389. Chan JL, Koda J, Heilig JS, et al. Immunogenicity associated with physiology and pathophysiology. Am J Physiol Endocrinol
metreleptin treatment in patients with obesity or lipodystrophy. Metab. 2011;301(4):E567-E584.
Clin Endocrinol (Oxf). 2016;85(1):137-149. 394. Klok MD, Jakobsdottir S, Drent ML. The role of leptin and ghre­
390. Angelidi AM, Filippaios A, Mantzoros CS. Severe insulin resist­ lin in the regulation of food intake and body weight in humans: a

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnae011/7629683 by guest on 19 April 2024


ance syndromes. J Clin Invest. 2021;131(4):e142245. review. Obes Rev. 2007;8(1):21-34.

You might also like