You are on page 1of 7

Pediatr Nephrol (2012) 27:1213–1219

DOI 10.1007/s00467-011-1963-1

REVIEW

The influence of gender and sexual hormones on incidence


and outcome of chronic kidney disease
Sebastian Kummer & Gero von Gersdorff &
Markus J. Kemper & Jun Oh

Received: 28 April 2011 / Revised: 19 June 2011 / Accepted: 19 June 2011 / Published online: 16 July 2011
# IPNA 2011

Abstract It has long been known that the female sex is and the progression of kidney diseases. Experimental
associated with a better clinical outcome in chronic renal models that analyze the effects of sexual hormones on
diseases. Although many experimental, clinical, and epide- renal structure and function are discussed. It is hoped that
miological studies in adults have attempted to explain the this review will stimulate researchers to focus on pediatric
difference in disease progression between females and studies that will provide a deeper insight into the interaction
males, a definitive understanding of the underlying mech- of gender hormones and the kidney both before and during
anisms is still lacking. Hormone-modulating therapies are puberty.
being increasingly used for various indications (such as
post-menopausal hormone replacement, estrogen- or Keywords Sex . Disease progression . Risk factor . FSGS .
androgen-receptor antagonists for cancer therapy). There- Renal disease
fore, a deeper knowledge of the interaction between sexual
hormones and progression of kidney disease is important,
as hormone-modulating therapy for non-renal indication Epidemiology: gender-dependent incidence
may influence both kidney structure and function. In and progression of chronic renal diseases
addition, specific modulation of the sexual hormone
system, such as the use of selective estrogen receptor In patients with chronic renal diseases, the prevalence of
modulators, may represent a therapeutic option for patients end-stage renal disease (ESRD) is higher in males than in
with renal diseases. Although conclusive data on this topic females (Fig. 1). These gender differences show remarkable
in the pediatric population are still lacking, the aim of this variations with age: when the male-to-female ratio of ESRD
review is to familiarize pediatric nephrologists with gender- is plotted as a function of age (Fig. 2), there are two peaks.
specific differences in renal physiology, pathophysiology, The first peak appears during infancy and early childhood
and most likely represents renal failure due to congenital
urologic anomalies, which are predominantly seen in boys.
S. Kummer (*) A second peak appears between ages 40–50 years, when
Department of General Pediatrics, University Children’s Hospital,
the activity of gender hormones in women begins to
Moorenstr. 5,
40225 Duesseldorf, Germany decline. These gender-linked differences disappear after
e-mail: sebastian.kummer@med.uni-duesseldorf.de the beginning of the menopausal years.
Thus, two different mechanisms can be distinguished,
G. von Gersdorff
which may cause the gender differences in renal diseases:
Renal Division, Department of Medicine,
University Hospital Cologne,
Cologne, Germany a) Gender-specific incidence of congenital anomalies of
the kidney and urinary tract (CAKUT). These differ-
M. J. Kemper : J. Oh
ences are presumably due to genetic factors. Hormonal
Department of Pediatric Nephrology,
University Medical Center Hamburg-Eppendorf, influences are of minor importance since maternal sex
Hamburg, Germany hormones cross the placental barrier easily, thus
1214 Pediatr Nephrol (2012) 27:1213–1219

differences were detectable through the whole age spectrum


(21.5–67.1 years, median 38.5 years).
The largest meta-analysis investigating gender differ-
ences in chronic renal diseases compared data collected on
11,345 patients participating in a total of 68 studies [2].
Female patients with polycystic kidney diseases, membra-
nous glomerulonephritis (MGN), and nephropathies of
unknown etiology had a favorable renal outcome compared
to their male counterparts. The clinical outcomes were
ESRD requiring renal replacement therapy, age of initiation
of renal replacement therapy, and changes in measured
GFR. The influence of menopause was not addressed in
Fig. 1 Prevalence of end-stage renal disease in the USA depending this study due to the heterogeneous patient population.
on age and gender (data are taken from the U.S. Renal Data System Cattran and colleagues further showed a significant
2000 Annual Data Report. The National Institutes of Health, NIDDK,
benefit of female gender in 395 patients with MGN and
Bethesda, MD)
370 patients with focal-segmental glomerulosclerosis
(FSGS) [3]. They measured the decrease in creatinine-
overriding gender differences in fetal hormone produc- based clearance and the overall survival of renal function.
tion. Therefore, this group of patients is of minor In particular, the negative impact of proteinuria on the rate
relevance for the subject of this review. of disease progression was clearly lower in women, and this
b) Influences of gender hormones on renal structure and interaction was independent of blood pressure.
function. These may either directly affect renal struc- Renal involvement in several systemic diseases also
tures (e.g., via gender hormone receptors on renal cells) shows distinct gender-dependent features. For example, renal
or indirectly via secondary influence factors (e.g. involvement occurs more frequently and more severely in
cardiovascular system). males with systemic lupus erythematodes (SLE), although
A multitude of epidemiologic studies have analyzed the SLE incidence is considerably higher in females [4]. Data on
interplay of gender and chronic renal diseases. Berg et al. renal involvement in diabetes mellitus (DM) type 2 are
demonstrated that the physiological decline of renal conflicting, likely due to the lack of multivariate analyses of
function during aging is significantly slower in healthy blood pressure and pre-/postmenopausal age influencing
females than in males [loss of glomerular filtration rate gender-dependent factors. Therefore, consistent evidence is
(GFR) of 1.4 ml/min/1.73 m2 per decade in healthy female still missing on potential gender differences in the renal
kidney donors compared to 8.7 ml/min/1.73 m2 per decade involvement of DM type 2 patients.
in males, as measured by inulin clearance] [1]. These A large meta-analysis of 27,805 adult patients with type
I DM showed an association of male gender with an
increased risk for an early development of diabetic
nephropathy [5]. Renal involvement was further dependent
on pre- or postpubertal onset of diabetes in children.
Prepubertal diagnosis of diabetes leads to significantly
prolonged latency until the first manifestation of nephrop-
athy. However, gender influences were not studied in this
trial [6].
Epidemiologic studies published to date which have
examined gender differences in renal diseases did not
analyze different racial groups separately. In turn, studies
demonstrating an increased risk of renal disease for several
ethnic groups, such as African Americans, Hispanics,
Fig. 2 Male-to-female ratio of end-stage renal disease as a function of
Native Americans, or Asians, have not specifically
age. The first peak during early infancy is caused by a male addressed gender differences in these populations [7].
predominance in congenital anomalies of the kidney and urinary tract Therefore, it remains unclear whether gender differences
(CAKUT), representing the most frequent cause for end-stage disease in renal diseases are also dependent on racial background.
in infancy. The second peak is located between menarche and
menopause, when hormonal influences are the most pronounced (data
Interestingly, the female advantage largely disappears
taken from the U.S. Renal Data System 2000 Annual Data Report. after the initiation of dialysis therapy, with women having
The National Institutes of Health, NIDDK, Bethesda, MD) as poor survival as men despite the continuing worse risk
Pediatr Nephrol (2012) 27:1213–1219 1215

profile of men also during dialysis therapy. The reasons for higher in boys than in girls [15]. Furthermore, girls with
this observation have not as yet been fully elucidated, but steroid-sensitive nephrotic syndrome show a trend towards
may be due to—among others—a more severe impact of less frequent post-pubertal recurrences than males (43 vs.
several risk factors on the overall prognosis of women 83%) [16]. However, the general frequency of MCGN
despite a less frequent occurrence (see [8] for an overview relapses reported in this study was higher than that found in
of this topic). other studies in which relapses were reported in only 30%
of patients after the initiation of puberty [17].
Kyrieleis et al. demonstrated that therapy with cyclo-
Gender differences associated with renal phosphamide of steroid-dependent and frequently relapsing
transplantation MCGN in prepubertal girls achieved definite remission
more often compared with boys, although this trend did not
It has long been noted that the outcome and survival of reach statistical significance [18]. Examination of the same
renal grafts is much better in female than in male recipients patients again after puberty revealed that, interestingly, of
[9], suggesting that the influence of the female recipient’s the 29% patients who developed a relapse, nine were boys
‘environment’ is protective of the transplanted graft. and only three were girls [19]. These data suggest a similar
In contrast, renal grafts from female donors have been trend towards a clinical advantage of female gender in
reported to have an inferior survival rate compared to those pediatric glomerular diseases.
from male donors [10]. Interestingly, in this study, the effect A retrospective analysis of 4,166 children with chronic
was observed to be even more pronounced for younger kidney disease (CKD) of different etiologies identified an
donors (16–45 years) than for older donors (>45 years). As association between CKD progression (among others) and
an explanation for this finding, the rapid loss of high male gender [20]. However, this finding did not show
premenopausal estrogen levels of the female donor statistical significance in the multivariate analysis, possibly
organism on the removed kidney may compromise graft due to the heterogeneous spectrum of disease etiologies.
function and survival after transplantation into a male Surprisingly, to the best of our knowledge, there have not
recipient. Another explanation for the inferior survival of been any subsequent studies specifically focusing on
grafts from female donors is provided by the concept of gender differences in pediatric patients with chronic
‘nephron underdosing’: fewer and smaller nephrons may glomerular diseases.
compromise organ function and survival by overloading the Taken together, the available epidemiological data show
capacity of individual nephrons. In contrast to early a significant benefit of female gender for the prognosis of
anatomic studies suggesting larger kidney weight in men, renal diseases. In the following part of this review, different
more recent data do not show any significant differences in experimental models explaining these gender differences
kidney weight when corrected for body weight [11]. will be discussed.
Furthermore, the number of glomeruli in females seems to
be equal to that in males [12]. Thus, ‘nephron underdosing’
seems to be of minor importance in terms of gender Influences of hormones on experimental models of renal
differences in renal transplantation. This conclusion is also function and disease
supported by a study showing that other transplanted organs
(e.g. heart) show similar drawbacks when taken from Various animal models of renal disease (hypertensive, age-
female donors [10]. The same study demonstrated that the related, polycystic, and renal mass-reduction) have shown
frequency of rejection treatments 1 year after transplanta- that the progression of renal injury is faster in male animals
tion was higher when the kidneys were taken from female than in their female littermates: as early as 1975 Elma and
donors. Consistent with these findings, Vereerstraten and colleagues were able to show that male rats spontaneously
coworkers saw a higher incidence of acute rejection develop proteinuria and glomerulosclerosis during aging,
episodes in female kidneys [13]. Both findings point to an whereas female animals were remarkably resistant against
immunological effect, for example, increased immunoge- these changes [21]. In another experiment, disease progres-
nicity of female grafts by the higher expression of HLA sion in male rats could be slowed by estrogen substitution
antigens [14]. or orchiectomy [22], suggesting a protective influence of
estrogens and adverse effects of androgens. Similarly,
experimental castration of 5/6 nephrectomized rats pro-
Gender differences in pediatric renal disease tected male animals against proteinuria, tubulointerstitial
damage, and renal accumulation of fibronectin, whereas
In children, the prevalence of glomerular diseases, such as female animals did not show any protective effect after
minimal-change glomerulonephritis (MCGN), is slightly removal of estrogen influence by ovariectomy [23].
1216 Pediatr Nephrol (2012) 27:1213–1219

In several other studies reviewed by Neugarten et al., angiotensin II induced an increase of GFR in men, but not
estrogen application reduced proteinuria and glomerular in women [39]. This difference was explained by the
fibrosis after experimental renal damage in different animal authors in terms of an enhanced glomerular capillary
models [24]. In addition, the expression of glomerular pressure as a response to angiotensin II in men, which
damage markers, such as desmin, in spontaneously hyper- was reduced or even absent in women. One possible
tensive rats and rats after puromycin treatment could be molecular mechanism might be that estrogens and andro-
attenuated by estrogen treatment [25, 26]. gens both modulate the expression of angiotensinogen,
Thus, female sexual hormones generally exhibit protec- angiotensin II, angiotensin II receptor, renin, and angioten-
tive effects, while androgens often accelerate disease sinogen converting enzyme (ACE) in multiple different cell
progress in animal models of renal disease. types, as shown in cell cultures and animal models [40, 41].
Correspondingly, human studies show an impact of estro-
gens and oral contraceptives on the regulation of the renin–
Possible mechanisms explaining gender differences angiontensin–aldosterone system (RAAS) [42, 43]. In
in the progression of renal disease contrast, endogenous increases in estrogen levels (e.g.,
during pregnancy) are associated with a decrease in arterial
Lifestyle factors and nutritional profile blood pressure despite the activation of RAAS [44]. Taken
together, these data show gender-specific influences on both
Factors such as physical and psychological stress, smoking, the regulation of systemic blood pressure and intrarenal
and especially nutritional factors seem to have a significant hemodynamics.
impact on the progression of renal diseases [27]. In
particular, an excessive consumption of protein, sodium, Other influences of the hormonal milieu
and phosphate has been shown to be harmful. Dyslipidemia
with high low-density lipoprotein (LDL), low high-density Premenopausal application of oral contraceptives as well as
lipoprotein (HDL), and hypertriglyceridemia [28], as well postmenopausal hormone replacement therapy (HRT)
as obesity in general [29], further facilitate disease increases microalbuminuria, a marker of early glomerular
progression. Compared to men, women have a more damage [45]. In 5,845 postmenopausal women older than
favorable nutritional profile, with higher intakes of fruit, 65 years, HRT was also associated with a significantly faster
vegetables, and dietary fibers and lower intakes of fat and decline of renal function compared to women without any
protein [30], and a beneficial serum lipid profile [31, 32]. HRT [46]. In a subgroup analysis, this effect was limited to
This may provide significant benefit for the prognosis of the estrogen monotherapy (additional decline of 1.21 ml/
renal diseases. min/1.73 m2 compared to non-substituted individuals).
Uric acid has recently turned out to be another risk factor Progesterone alone or in combination with estrogens did
for accelerated kidney disease progression—probably me- not induce an accelerated decline of kidney function.
diated by increasing the incidence of arterial hypertension Moreover, the effect was detectable only for oral adminis-
[33]. As uric acid levels are known to be higher in males tration, not for vaginal application, and was independent of
than in females [34], this may further contribute to a faster any other co-medications (such as non-steroidal antiphlo-
progression of CKD in men gistics, ACE inhibitors, angiotensin receptor blockers,
glucocorticoids, cyclosporine, etc.). Unfortunately, it was
Blood pressure and intrarenal hemodynamics not documented whether the therapy was initiated before or
during menopause.
Arterial hypertension is significantly associated with a In contrast, other studies have detected a reduced risk
worse prognosis in CKD [35]. Various studies have shown for albuminuria in postmenopausal women on HRT
that women have a lower mean arterial blood pressure [36] without further differentiation for the subtype of medica-
and a lower incidence of arterial hypertension than age- and tion [47]. In postmenopausal women with DM, HRT also
weight-matched men [37]. Interestingly, these differences in did not show any effect on albuminuria compared to
blood pressure level between men and women disappear women without HRT [48], whereas in premenopausal
after the onset of the menopause [37]. Therefore, hormonal patients with DM, HRT was clearly associated with
influences on blood pressure level may substantially affect albuminuria [43]. Experimental data reveal similar dis-
the outcome of renal diseases [15]. crepancies as epidemiological studies: in animal models,
When adjusted for body weight and body surface, GFR only continuous application of estrogen throughout a 9-
is identical for both genders in humans and rats [38]. month period was able to reduce experimentally induced
However, intrarenal hemodynamics show gender-specific glomerulosclerosis, while intermittent application every 3
reactions on numerous variables. The administration of months had no effect [49].
Pediatr Nephrol (2012) 27:1213–1219 1217

One possible explanation for this inconsistency in pathways [57]. Doublier and colleagues demonstrated that
experimental and clinical data is the high degree of this mechanism also leads to a protection of podocytes
heterogeneity in the therapeutic approach to HRT, with a against transforming growth factor beta 1 (TGFβ1)- or
variety of different hormone subtypes, application forms, tumor necrosis factor alpha (TNFα)-induced apoptosis in
and timing of therapy initiation. vitro and in vivo, while testosterone induces podocyte
apoptosis by signaling via androgen receptors [58]. Un-
Cellular pathways of sexual hormones and selective published data from our group show a corresponding
estrogen receptor modulators (SERM) estrogen-induced protection of podocytes in the model of
puromycin-induced apoptosis. Studies on metastatic cancer
Numerous studies have shown that gender hormones, in have implicated a novel set of signaling molecules as
particular estrogens, have pleiotropic effects on different potential mediators of estrogens, namely, focal adhesion
cell types through specific cellular receptors. Extensive kinase (FAK) and paxillin. Both regulate cell adhesion and
investigations in this field have been performed, especially the interaction of cytoskeletal components with the extra-
for neuronal cells and skeletal muscle cells. For example, cellular matrix. In podocytes, these proteins have been
the transcription of genes encoding mitochondrial proteins shown to be critical for the maintenance of cellular
is modified by estrogen receptor (ER) activation, represent- structures: The interactions of podocytes with the glomer-
ing a strong link between ER signaling and intact ular basement membrane result in increased stability
mitochondrial function [50]. One specific example is the against proteinuria in FAK knockout mice [59]. Recent
estrogen-induced expression of nuclear respiratory factor-1 data also show a complex interaction of estrogen signaling
(NRF-1), a transcriptional factor that regulates the expres- with the activity of FAK, thereby modulating cell motility
sion of nuclear-encoded mitochondrial genes, such as via control of the focal adhesion complex turnover in
mitochondrial transcription factor A (TFAM) [51]. This endothelial cells, breast cancer cells, and endometrial cells.
pathway leads to a coordinated increase in the expression of Further protective influences of estrogens are an inhibited
the mitochondrial genome and thereby the stimulation of production of matrix proteins, such as collagen type I and
mitochondrial biogenesis and function. This cellular path- IV, and stimulation of matrix-degrading enzymes, thereby
way could explain why skeletal muscle cells are stabilized reducing glomeruloslerosis [60, 61]. In TGF-β-transgenic
against oxidative damage by estrogens [52] and why mice, which have an increased susceptibility to extensive
degenerative diseases of muscle cells are associated with a glomerulosclerosis, estrogen substitution significantly
decrease of estrogen levels [53]. The stabilization of reduces sclerosis [62]. Protective effects have also been
mitochondrial function has also been shown in ocular lens shown for SERM such as tamoxifen and raloxifene. These
epithelium, where estrogen was found to reduce destabili- substances bind to estrogen receptors with tissue-specific
zation of mitochondrial membrane potential by oxidative effects and different binding specificities for ERα or ERβ:
stress [54]. In addition to these genomic ER effects, ‘non- for example, raloxifene has a fourfold higher affinity for
classical’ actions via membrane-associated ER are also able ERα than for ERβ and does not induce endometrial
to activate cytoplasmic signaling pathways in a G-protein- hyperplasia. In vitro, SERM are able to reduce collagen
coupled manner. For example, activation of mitogen- synthesis, similar to estrogen [60]. In vivo, renal prognosis
activated protein kinase (MAPK) or phosphatidylinositol is improved by SERM: raloxifene has very recently been
3-OH kinase (PI3K) pathways appears within minutes after found to be significantly renoprotective in postmenopausal
estrogen administration [55, 56]. The MAPK pathway women that were treated with this medication for reduction
involves a group of intracellular signaling molecules of bone fractures. In this study, raloxifene-treated women
comprising three major families with different downstream developed a significantly slower annual increase in serum
effects: extracellular signal-regulated kinase (ERK1/2), p38 creatinine and a lower decrease in GFR [63]. Several
MAPK, and c-Jun N-terminal kinases. These pathways estrogenic effects can also be induced by metabolites of
involve a group of intracellular signaling molecules with estradiol, such as catecholestradioles (e.g. 2-hydroxyestradiol
impacts on cell proliferation, survival, apoptosis, differen- and 4-hydroxyestradiol) [25]. Interestingly, these effects are
tiation, motility, among others. One example of protective independent of ER activation, indicating that additional
estrogen action is the reduction of apoptotic cell death of independent mechanisms must be involved.
neurons following ischemia by an estrogen-induced activa-
tion of the Raf–MEK–ERK–p90RSK cascade [56]. Inter-
estingly, similar pathways have recently been shown to be Summary/conclusion
active in podocytes: podocytes isolated, immortalized, and
subsequently analyzed in vitro were observed to show In terms of the incidence and progression of chronic renal
estrogen-induced modulation of TGFβ- and MAPK- diseases, epidemiological data demonstrate that the female
1218 Pediatr Nephrol (2012) 27:1213–1219

gender endows patients with significant benefits compared duration, A1C, hypertension, dyslipidemia, diabetes onset, and
sex. Diabetes Care 30:2523–2528
to the male gender. 6. Svensson M, Nystrom L, Schon S, Dahlquist G (2006) Age at
In this review, we have presented several factors that onset of childhood-onset type 1 diabetes and the development of
have been evaluated extensively (lifestyle, blood pressure, end-stage renal disease: a nationwide population-based study.
and hemodynamics). More recent data also show direct Diabetes Care 29:538–542
7. Norris KC, Agodoa LY (2005) Unraveling the racial disparities
actions of sexual hormones in the kidney at a cellular level associated with kidney disease. Kidney Int 68:914–924
(e.g., regulation of apoptosis, cytoskeletal dynamics, mito- 8. Carrero JJ (2010) Gender differences in chronic kidney disease:
chondrial function, nitric oxide signaling). Taken together, a underpinnings and therapeutic implications. Kidney Blood Press
complex interaction of estrogen effects in combination with Res 33:383–392
9. Reed E, Cohen DJ, Barr ML, Ho E, Reemtsma K, Rose EA,
partly adverse androgen effects seems likely, with the Hardy M, Suciu-Foca N (1992) Effect of recipient gender and race
balance of both contributing decisively to the overall renal on heart and kidney allograft survival. Transplant Proc 24:2670–
prognosis. 2671
However, the clinically most relevant question still 10. Zeier M, Dohler B, Opelz G, Ritz E (2002) The effect of donor
gender on graft survival. J Am Soc Nephrol 13:2570–2576
remains to be answered: will this knowledge find any 11. Kasiske BL, Umen AJ (1986) The influence of age, sex, race, and
therapeutic application in the near future? A first attempt body habitus on kidney weight in humans. Arch Pathol Lab Med
was published in 1955 [64]. Three patients with severe 110:55–60
nephrotic syndrome had complete resolution of symptoms 12. Nyengaard JR, Bendtsen TF (1992) Glomerular number and size
in relation to age, kidney weight, and body surface in normal man.
when treated with an initial course of estrogen application, Anat Rec 232:194–201
but proteinuria recurred after the discontinuation of the 13. Vereerstraeten P, Wissing M, De Pauw L, Abramowicz D, Kinnaert
medication. Two women had another course of therapy and P (1999) Male recipients of kidneys from female donors are at
achieved remission again, while the male patient refused to increased risk of graft loss from both rejection and technical
failure. Clin Transplant 13:181–186
take these hormones again and died in nephrotic crisis. 14. Panajotopoulos N, Ianhez LE, Neumann J, Sabbaga E, Kalil J
Although the substitution of 17β-estradiol does not (1990) Immunological tolerance in human transplantation. The
provide a reasonable therapeutic option due to significant possible existence of a maternal effect. Transplantation 50:443–445
side-effects (such as possible tumor induction and femini- 15. Silbiger SR, Neugarten J (1995) The impact of gender on the
progression of chronic renal disease. Am J Kidney Dis 25:515–
zation), SERM or non-receptor mediated estrogen metabo- 533
lites may offer promising options for a targeted therapy of 16. Fakhouri F, Bocquet N, Taupin P, Presne C, Gagnadoux MF,
renal diseases. Whether such an approach might also be Landais P, Lesavre P, Chauveau D, Knebelmann B, Broyer M,
extended to the pediatric population with a reasonable Grunfeld JP, Niaudet P (2003) Steroid-sensitive nephrotic syn-
drome: from childhood to adulthood. Am J Kidney Dis 41:550–557
balanced risk profile regarding specific problems of 17. Ruth EM, Kemper MJ, Leumann EP, Laube GF, Neuhaus TJ
prepubertal/pubertal patients remains to be determined. (2005) Children with steroid-sensitive nephrotic syndrome come
However, increasing awareness of gender aspects may of age: long-term outcome. J Pediatr 147:202–207
encourage pediatric nephrologists to consider them as risk 18. Kyrieleis HA, Levtchenko EN, Wetzels JF (2007) Long-term
outcome after cyclophosphamide treatment in children with
factors and prognostic indicators for renal diseases in the steroid-dependent and frequently relapsing minimal change
pediatric population. nephrotic syndrome. Am J Kidney Dis 49:592–597
19. Kyrieleis HA, Lowik MM, Pronk I, Cruysberg HR, Kremer JA,
Oyen WJ, van den Heuvel BL, Wetzels JF, Levtchenko EN (2009)
References Long-term outcome of biopsy-proven, frequently relapsing
minimal-change nephrotic syndrome in children. Clin J Am Soc
Nephrol 4:1593–1600
1. Berg UB (2006) Differences in decline in GFR with age between 20. Staples AO, Greenbaum LA, Smith JM, Gipson DS, Filler G,
males and females. Reference data on clearances of inulin and Warady BA, Martz K, Wong CS (2010) Association between
PAH in potential kidney donors. Nephrol Dial Transplant clinical risk factors and progression of chronic kidney disease in
21:2577–2582 children. Clin J Am Soc Nephrol 5:2172–2179
2. Neugarten J, Acharya A, Silbiger SR (2000) Effect of gender on 21. Elema JD, Arends A (1975) Focal and segmental glomerular
the progression of nondiabetic renal disease: a meta-analysis. J hyalinosis and sclerosis in the rat. Lab Invest 33:554–561
Am Soc Nephrol 11:319–329 22. Hajdu A, Rona G (1971) The protective effect of estrogens against
3. Cattran DC, Reich HN, Beanlands HJ, Miller JA, Scholey JW, spontaneous pancratic islet and renal changes in aging male rats.
Troyanov S (2008) The impact of sex in primary glomerulone- Experientia 27:956–957
phritis. Nephrol Dial Transplant 23:2247–2253 23. Lemos CC, Mandarim-de-Lacerda CA, Dorigo D, Coimbra TM,
4. Molina JF, Drenkard C, Molina J, Cardiel MH, Uribe O, Anaya Bregman R (2005) Chronic renal failure in male and female rats. J
JM, Gomez LJ, Felipe O, Ramirez LA, Alarcon-Segovia D (1996) Nephrol 18:368–373
Systemic lupus erythematosus in males. A study of 107 Latin 24. Neugarten J (2002) Gender and the progression of renal disease. J
American patients. Medicine 75:124–130 Am Soc Nephrol 13:2807–2809
5. Raile K, Galler A, Hofer S, Herbst A, Dunstheimer D, Busch P, 25. Tofovic SP, Dubey R, Salah EM, Jackson EK (2002) 2-
Holl RW (2007) Diabetic nephropathy in 27,805 children, Hydroxyestradiol attenuates renal disease in chronic puromycin
adolescents, and adults with type 1 diabetes: effect of diabetes aminonucleoside nephropathy. J Am Soc Nephrol 13:2737–2747
Pediatr Nephrol (2012) 27:1213–1219 1219

26. Gross ML, Adamczak M, Rabe T, Harbi NA, Krtil J, Koch A, postmenopausal women is associated with loss of kidney function.
Hamar P, Amann K, Ritz E (2004) Beneficial effects of estrogens Kidney Int 74:370–376
on indices of renal damage in uninephrectomized SHRsp rats. J 47. Schopick EL, Fisher ND, Lin J, Forman JP, Curhan GC (2009)
Am Soc Nephrol 15:348–358 Post-menopausal hormone use and albuminuria. Nephrol Dial
27. Ritz E, Schwenger V (2005) Lifestyle modification and progres- Transplant 24:3739–3744
sive renal failure. Nephrology 10:387–392 48. Manning PJ, Sutherland WH, Allum AR, de Jong SA, Jones SD
28. Tsimihodimos V, Dounousi E, Siamopoulos KC (2008) Dyslipi- (2003) HRT does not improve urinary albumin excretion in
demia in chronic kidney disease: an approach to pathogenesis and postmenopausal diabetic women. Diabetes Res Clin Pract 60:33–39
treatment. Am J Nephrol 28:958–973 49. Karl M, Berho M, Pignac-Kobinger J, Striker GE, Elliot SJ (2006)
29. Nguyen S, Hsu CY (2007) Excess weight as a risk factor for Differential effects of continuous and intermittent 17beta-estradiol
kidney failure. Curr Opin Nephrol Hypertens 16:71–76 replacement and tamoxifen therapy on the prevention of glomer-
30. Westenhoefer J (2005) Age and gender dependent profile of food ulosclerosis: modulation of the mesangial cell phenotype in vivo.
choice. Forum Nutr:44–51 Am J Pathol 169:351–361
31. Graff-Iversen S, Thelle DS, Hammar N (2008) Serum lipids, 50. Klinge CM (2008) Estrogenic control of mitochondrial function
blood pressure and body weight around the age of the menopause. and biogenesis. J Cell Biochem 105:1342–1351
Eur J Cardiovasc Prev Rehabil 15:83–88 51. Mattingly KA, Ivanova MM, Riggs KA, Wickramasinghe NS,
32. Mudali S, Dobs AS, Ding J, Cauley JA, Szklo M, Golden SH Barch MJ, Klinge CM (2008) Estradiol stimulates transcription of
(2005) Endogenous postmenopausal hormones and serum lipids: nuclear respiratory factor-1 and increases mitochondrial biogene-
the atherosclerosis risk in communities study. J Clin Endocrinol sis. Mol Endocrinol 22:609–622
Metab 90:1202–1209 52. Persky AM, Green PS, Stubley L, Howell CO, Zaulyanov L,
33. Feig DI (2009) Uric acid: a novel mediator and marker of risk in Brazeau GA, Simpkins JW (2000) Protective effect of estrogens
chronic kidney disease? Curr Opin Nephrol Hypertens 18:526– against oxidative damage to heart and skeletal muscle in vivo and
530 in vitro. Proc Soc Exp Biol Med 223:59–66
34. Krishnan E, Pandya BJ, Chung L, Dabbous O (2011) Hyperuri- 53. Dionne IJ, Kinaman KA, Poehlman ET (2000) Sarcopenia and
cemia and the risk for subclinical coronary atherosclerosis—data muscle function during menopause and hormone-replacement
from a prospective observational cohort study. Arthritis Res Ther therapy. J Nutr Health Aging 4:156–161
13:R66 54. Flynn JM, Lannigan DA, Clark DE, Garner MH, Cammarata PR
35. Martinez-Maldonado M (2001) Role of hypertension in the (2008) RNA suppression of ERK2 leads to collapse of mitochon-
progression of chronic renal disease. Nephrol Dial Transplant 16 drial membrane potential with acute oxidative stress in human lens
[Suppl 1]:63–66 epithelial cells. Am J Physiol Endocrinol Metab 294:E589–E599
36. Khoury S, Yarows SA, O’Brien TK, Sowers JR (1992) Ambula- 55. Bjornstrom L, Sjoberg M (2005) Mechanisms of estrogen receptor
tory blood pressure monitoring in a nonacademic setting. Effects signaling: convergence of genomic and nongenomic actions on
of age and sex. Am J Hypertens 5:616–623 target genes. Mol Endocrinol 19:833–842
37. McBride SM, Flynn FW, Ren J (2005) Cardiovascular alteration 56. Koh PO (2007) Estradiol prevents the injury-induced decrease of
and treatment of hypertension: do men and women differ? 90 ribosomal S6 kinase (p90RSK) and Bad phosphorylation.
Endocrine 28:199–207 Neurosci Lett 412:68–72
38. Munger K, Baylis C (1988) Sex differences in renal hemodynam- 57. Catanuto P, Doublier S, Lupia E, Fornoni A, Berho M, Karl M,
ics in rats. Am J Physiol 254:F223–231 Striker GE, Xia X, Elliot S (2009) 17 beta-estradiol and tamoxifen
39. Miller JA, Anacta LA, Cattran DC (1999) Impact of gender on the upregulate estrogen receptor beta expression and control podocyte
renal response to angiotensin II. Kidney Int 55:278–285 signaling pathways in a model of type 2 diabetes. Kidney Int
40. Reckelhoff JF, Zhang H, Srivastava K, Granger JP (1999) Gender 75:1194–1201
differences in hypertension in spontaneously hypertensive rats: 58. Doublier S, Lupia E, Catanuto P, Periera-Simon S, Xia X, Korach
role of androgens and androgen receptor. Hypertension 34:920– K, Berho M, Elliot SJ, Karl M (2011) Testosterone and 17beta-
923 estradiol have opposite effects on podocyte apoptosis that
41. Oelkers WK (1996) Effects of estrogens and progestogens on the precedes glomerulosclerosis in female estrogen receptor knockout
renin-aldosterone system and blood pressure. Steroids 61:166–171 mice. Kidney Int 79:404–413
42. Chidambaram M, Duncan JA, Lai VS, Cattran DC, Floras JS, 59. Ma H, Togawa A, Soda K, Zhang J, Lee S, Ma M, Yu Z, Ardito T,
Scholey JW, Miller JA (2002) Variation in the renin angiotensin Czyzyk J, Diggs L, Joly D, Hatakeyama S, Kawahara E, Holzman
system throughout the normal menstrual cycle. J Am Soc Nephrol L, Guan JL, Ishibe S (2010) Inhibition of podocyte FAK protects
13:446–452 against proteinuria and foot process effacement. J Am Soc
43. Ahmed SB, Hovind P, Parving HH, Rossing P, Price DA, Laffel Nephrol 21:1145–1156
LM, Lansang MC, Stevanovic R, Fisher ND, Hollenberg NK 60. Neugarten J, Acharya A, Lei J, Silbiger S (2000) Selective
(2005) Oral contraceptives, angiotensin-dependent renal vasocon- estrogen receptor modulators suppress mesangial cell collagen
striction, and risk of diabetic nephropathy. Diabetes Care synthesis. Am J Physiol Renal Physiol 279:F309–318
28:1988–1994 61. Guccione M, Silbiger S, Lei J, Neugarten J (2002) Estradiol
44. Chapman AB, Abraham WT, Zamudio S, Coffin C, Merouani A, upregulates mesangial cell MMP-2 activity via the transcription
Young D, Johnson A, Osorio F, Goldberg C, Moore LG, Dahms T, factor AP-2. Am J Physiol Renal Physiol 282:F164–169
Schrier RW (1998) Temporal relationships between hormonal and 62. Blush J, Lei J, Ju W, Silbiger S, Pullman J, Neugarten J (2004)
hemodynamic changes in early human pregnancy. Kidney Int Estradiol reverses renal injury in Alb/TGF-beta1 transgenic mice.
54:2056–2063 Kidney Int 66:2148–2154
45. Monster TB, Janssen WM, de Jong PE, de Jong-van den Berg LT 63. Melamed ML, Blackwell T, Neugarten J, Arnsten JH, Ensrud KE,
(2001) Oral contraceptive use and hormone replacement therapy Ishani A, Cummings SR, Silbiger SR (2011) Raloxifene, a
are associated with microalbuminuria. Arch Intern Med selective estrogen receptor modulator, is renoprotective: a post-
161:2000–2005 hoc analysis. Kidney Int 79:241–249
46. Ahmed SB, Culleton BF, Tonelli M, Klarenbach SW, Macrae JM, 64. Nitsch W, Thein K (1955) Therapy of the nephrotic syndrome
Zhang J, Hemmelgarn BR (2008) Oral estrogen therapy in with estradiol. Ther Ggw 94:364–366

You might also like