You are on page 1of 6

J Appl Physiol 119: 1481–1486, 2015.

First published August 13, 2015; doi:10.1152/japplphysiol.00271.2015. Review

HIGHLIGHTED TOPIC Hypoxia 2015

New genetic and physiological factors for excessive erythrocytosis and


Chronic Mountain Sickness
Francisco C. Villafuerte
Laboratorio de Fisiología Comparada, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Perú
Submitted 7 April 2015; accepted in final form 7 August 2015

Villafuerte FC. New genetic and physiological factors for excessive erythrocy-
tosis and Chronic Mountain Sickness. J Appl Physiol 119: 1481–1486, 2015. First
published August 13, 2015; doi:10.1152/japplphysiol.00271.2015.—In the last few
years, genetic and functional studies have provided important insight on the
pathophysiology of excessive erythrocytosis (EE), the main sign of Chronic
Mountain Sickness (CMS). The recent finding of the association of the CMS
phenotype with a single-nucleotide polymorphism (SNP) in the Sentrin-specific
Protease 1 (SENP1) gene, and its differential expression pattern in Andean
highlanders with and without CMS, has triggered large interest in high-altitude
studies because of the potential role of its gene product in the control of erythro-
poiesis. The SENP1 gene encodes for a protease that regulates the function of
hypoxia-relevant transcription factors such as Hypoxia-Inducible Factor (HIF) and
GATA, and thus might have an erythropoietic regulatory role in CMS through the
modulation of the expression of erythropoietin (Epo) or Epo receptors. The
different physiological patterns in the Epo-EpoR system found among Andeans,
even among highlanders with CMS, together with their different degrees of
erythropoietic response, might indicate specific underlying genetic backgrounds,
which in turn might reflect different levels of adaptation to lifelong high-altitude
hypoxia. This minireview discusses recent genetic findings potentially underlying
EE and CMS, and their possible physiological mechanisms in Andean highlanders.
Chronic Mountain Sickness; excessive erythrocytosis; chronic hypoxia; high alti-
tude; Andes

IN THE LAST FEW YEARS, GENETIC and functional studies have Consensus scoring system (30). Pulmonary hypertension is a
provided important insight into the pathophysiology of exces- frequent complication of the condition and may lead to right-
sive erythrocytosis (EE) and Chronic Mountain Sickness heart failure in advanced stages of the disease (42, 43). CMS
(CMS). The use of genomic technologies has allowed the affects people who reside at altitudes ⱖ2,500 m above sea
identification of functionally relevant single-nucleotide poly- level, and it is estimated that 5-10% of the world’s population
morphism (SNPs) as markers of “maladaptation” to life at high living at high altitude may develop the condition (13, 21, 30,
altitude, as well as some potential functional correlates. This 48, 51). In the central Andes of Peru (⬎4,000 m), 15–20% of
minireview discusses recent genetic findings potentially under- the adult male population suffers from CMS, and its prevalence
lying EE and CMS, and their possible physiological mecha- increases with age, rising up to 30% by the age of 50 (29, 39).
nisms among residents of the Andean highlands. The pathophysiological mechanism of EE is still debatable;
however, it is well known that hypoxemia represents its un-
CHRONIC MOUNTAIN SICKNESS OR MONGE DISEASE derlying stimulus because relocation of patients with CMS to
sea level reverses the disorder completely. Also, it has been
CMS is a clinical syndrome characterized by EE, which is shown that phlebotomy, with or without isovolemic hemodi-
defined by hemoglobin (Hb) concentration values ⱖ21 g/dl for lution, alleviates CMS-related symptoms, suggesting that
men or ⱖ19 g/dl for women (30). CMS is diagnosed on the many of these are associated with EE (37, 46, 57, 58).
basis of the presence of EE accompanied by three or more of
the following symptoms: breathlessness, palpitations, sleep A POTENTIAL GENETIC MARKER FOR EXCESSIVE
disturbance, cyanosis, dilation of veins, paresthesia, headache, ERYTHROPOIESIS
or tinnitus (29, 33, 39) according to the Qinghai International
Although it has been known for some time that high Hb
concentration values among Andean highlanders show herita-
Address for reprint requests and other correspondence: F.C. Villafuerte,
Laboratorio de Fisiología Comparada, Facultad de Ciencias y Filosofía, Uni-
bility and familial character (4, 31), until recently no specific
versidad Peruana Cayetano Heredia, Av. Honorio Delgado 430, Lima 31, alleles had been found to be associated with the exaggerated
Lima, Peru (e-mail: francisco.villafuerte@upch.pe). red blood cell count in patients with CMS. Recent studies have
http://www.jappl.org 8750-7587/15 Copyright © 2015 the American Physiological Society 1481
Downloaded from journals.physiology.org/journal/jappl (200.105.212.020) on June 20, 2023.
Review
1482 Genetics and Physiology of Excessive Erythrocytosis and CMS • Villafuerte FC

revealed that specific SNPs in the EPAS1 and EGLN1 genes are PHYSIOLOGICAL CORRELATES TO THE CANDIDATE GENE
associated with low Hb concentration values in Tibetan high- SENP1
landers and suggest that these markers may be linked to a
SENP1 plays important roles in the regulation of multiple
physiological mechanism that confers protection to this popu-
lation against the development of EE and CMS (5, 49, 62). The cellular signaling pathways, including glucose and mitochon-
products of these genes, Hypoxia-Inducible Factor-2 (HIF-2) drial metabolism, and hormone receptor activity regulation (1,
and Prolyl Hydroxylase Domain 2 (PHD2), respectively, are 6, 9, 11, 34, 52, 60). Also, studies in animal models show that
major regulators of erythropoiesis given their central role in the SENP1 plays a key function in normal erythropoiesis. But is
control of the hypoxic response of erythropoietin (Epo) expres- there a potential link between SENP1 and the pathophysiolog-
sion at the transcriptional level. However, among Andeans, no ical erythropoietic response? SENP1 is a small ubiquitin-
such “protective” alleles have been identified, and no differ- related modifier (SUMO) protein-specific protease that regu-
ences in the frequencies of these SNPs between healthy high- lates the function of hypoxia-relevant transcription factors such
landers and those with CMS have been demonstrated (31). as HIF and GATA (8, 65). SENP1 deconjugates the SUMO
Differences are also absent in other candidate genes associated from HIF1␣ and rescues this subunit from ubiquitination and
with erythropoiesis, such as Epo and Epo receptor (EpoR). degradation (Fig. 1). Studies in mouse embryos have shown
The recent finding of an association of the CMS phenotype that SENP1 is required for hepatic Epo production and eryth-
among Andeans with an SNP in the sentrin-specific protease 1 ropoiesis, and that senp1⫺/⫺ mice die from anemia (8). Thus
(SENP1) gene has triggered large interest in the field of although this pathway has not been yet verified in humans, it
high-altitude studies because of the potential role of its gene seems likely that SENP1-dependent regulation of HIF2␣, the
product in the control of erythropoiesis (7, 47, 66). The main erythropoietic HIF␣ isoform in adults, shares similar
identified candidate gene was validated by hypoxic functional direct and indirect regulatory pathways with HIF1␣ (8, 25).
testing in cultured fibroblasts derived from skin biopsies ob- In addition, SENP1 regulates erythropoiesis via GATA-1
tained from the same subjects. Compared with normoxic con- deSUMOylation (65). Consistently, GATA1-null mouse em-
trols, hypoxia treatment (1.5% O2 for 24 h) induced a signif- bryos die from severe anemia (15). The critical role of GATA1
icant downregulation of SENP1 expression in non-CMS cells in erythropoiesis is attributed to its activity in driving expres-
and an upregulation in CMS cells (66). Therefore, it is likely sion of many erythropoietic genes including enzymes involved
that the lower arterial O2 saturation in patients with CMS in heme biosynthesis, hemoglobin, and EpoR (10, 17, 26, 67).
induces a higher expression of SENP1 and favors exaggerated Among these, EpoR expression has been well established to be
erythropoiesis. Thus in contrast with Tibetans and Ethiopians, critical for definitive erythropoiesis in murine fetal liver (28,
Andeans show a proerythropoietic SNP rather than protective 35, 59). Yu et al. (65) showed that a SUMOylated form of
SNPs against erythrocytosis. This supports the hypothesis of GATA1 accumulated in senp1⫺/⫺ mice and correlated with a
different degrees of high-altitude adaptation among popula- downregulation of GATA1-dependent gene expression includ-
tions. It would be interesting to explore whether the SENP1 ing EpoR and ␤-hemoglobin. The study also showed that
SNP found in Tibetans and Ethiopians correspond to the GATA1 can be directly deSUMOylated by SENP1, resulting in
“adapted variant” of the SENP1 allele found in healthy Andean the modulation of GATA1-dependent EpoR expression and
highlanders. erythropoiesis.

Fig. 1. Schematic model of sentrin-specific protease 1


(SENP1) on Hypoxia-Inducible Factor ␣ (HIF␣) and
GATA-1 regulation. A: under normoxia, HIF␣ is sub-
ject to oxygen-dependent prolyl hydroxylation by
Prolyl-Hydroxylase Domain-containing enzymes
(PHDs), which allows for substrate recognition and
ubiquitylation by Von Hippel-Lindau protein (pVHL)
and its associated ubiquitin-ligase complex. Polyubiq-
uitylated (Ubn) HIF␣ is then degraded by the protea-
some. B: under hypoxia, HIF␣ escapes prolyl hydroxy-
lation and undergoes SUMOylation. SENP1-dependent
deSUMOylation rescues HIF␣ from polyubiquitination
and degradation, and allows its association with nuclear
HIF␤. The heterodimer binds to a core consensus se-
quence at the promoters of HIF-responsive genes, and
upon binding to coactivators, initiates transcription.
SENP1-dependent GATA-1 deSUMOylation enhances
its transcriptional activity, and thus upregulation of
SENP1 expression might increase available deSUMOy-
lated GATA-1 and enhance gene transcription. OH,
hydroxyl group; Ub, ubiquitin moiety.

J Appl Physiol • doi:10.1152/japplphysiol.00271.2015 • www.jappl.org


Downloaded from journals.physiology.org/journal/jappl (200.105.212.020) on June 20, 2023.
Review
Genetics and Physiology of Excessive Erythrocytosis and CMS • Villafuerte FC 1483
It is interesting that these two SENP1 targets, HIF and IS IT POSSIBLE THEN TO EXPLAIN AN INCREASED
GATA, control the Epo-EpoR system, and thus it is likely that PRODUCTION OF RED BLOOD CELLS WITH NORMAL Epo
hypoxic SENP1 upregulation as shown in patients with CMS VALUES?
(66) alters the Epo-EpoR system and therefore erythropoiesis.
Epo action depends both on Epo concentration and on the
abundance and type of Epo receptors (24, 50). Thus modula-
ARE THERE ANY OTHER POTENTIAL FUNCTIONAL
tion of EpoR expression or differential regulation of expression
CORRELATES TO SENP1-INCREASED EXPRESSION IN CMS?
of EpoR splice variants represents a potential erythropoiesis
HIF-mediated hypoxic expression of genes such as VEGF regulatory mechanism. SENP1-mediated regulation of EpoR
might be enhanced by SENP1 due to deSUMOylation and expression is likely to occur through GATA-1. The 5=-flanking
increased HIF␣ subunit stability (8). Apenzeller and coworkers region of the EpoR gene has several potential GATA-1 binding
(2) showed that VEGF expression was increased in patients sites and EpoR expression is regulated by promoters that bind
with CMS, and it was positively correlated with CMS score to GATA-1 (64, 65, 67). Also, it has been demonstrated that
and negatively with arterial saturation. More recently, Xu et al. this regulation is specific for cells of the hematopoietic line
(61) have provided evidence that SENP1 plays an essential role (63).
as a positive regulator of hypoxia-driven VEGF production and In addition to EpoR gene expression modulation, the ex-
angiogenesis by showing that silencing of SENP1 expression pressed proportion of splice variants of the gene can also affect
decreases VEGF production and abrogates the angiogenic the sensitivity of the system to Epo. EpoR can be synthesized
functions of endothelial cells. Therefore, there is a possibility in several forms: full-length, truncated, and soluble. Exons 1 to
that an increased SENP1 expression in CMS relates to changes 5 encode the extracellular domain, exon 6 encodes the mem-
in the expression of VEGF. brane-spanning domain, and exons 7 and 8 encode the intra-
cellular domain (20). The soluble receptor form (sEpoR) with
THE Epo-EpoR SYSTEM AND ITS PHYSIOLOGICAL a molecular weight of 29 kDa corresponds with the extracel-
ASSOCIATION WITH EE lular domain of the complete membrane EpoR (mEpoR) (18,
41, 56), and its synthesis occurs by alternative splicing of EpoR
The role of SENP1 in erythropoiesis supposes HIF and
GATA stabilization with the consequent expression of target mRNA (14, 19, 56). Recent findings suggest that the sEpoR
genes. SENP-dependent (direct or indirect) stabilization of plays a role in modulating Epo action in patients with CMS
HIF2␣ would lead to increased Epo expression and increased (53).
erythropoiesis, similarly to what is observed in other condi- Although it is likely that genetic variation at the splicing
tions with augmented HIF2␣ levels (27, 44). However, ele- machinery level has a significant effect on the abundance of
vated blood Epo values are not common in patients with CMS. membrane-bound or soluble Epo receptors, SUMOylation of
Most highlanders with CMS exhibit EE but normal Epo values spliceosome factors might also play an important role. Pro-
for the altitude of residence, similar to those observed in their teomic analysis has revealed that SENP isoforms are key in the
healthy counterparts (16, 23, 32). Yet a subgroup of patients deSUMOylation and function of these factors (55). Thus in
with CMS show significantly higher Epo values (2 SD above addition to the role of SENP1 in EpoR expression via
the altitude-normal average) usually with slightly higher Hb GATA-1, it has also a potential role modifying the splicing
(12, 32, 53) (Table 1). This finding might help explain why machinery and therefore the production of EpoR isoforms.
most studies, but not all, report normal Epo average values in Functionally, several studies have shown that the binding
CMS (16, 23, 32, 45, 53), because these values would depend of Epo to sEpoR limits its ability to bind mEpoR (3, 38, 50),
on the proportion of participants with normal and high Epo and thus this soluble isoform acts as an Epo “buffer”
values. Moreover, given that increased SENP1 expression regulating available circulating Epo concentration. Our
supposes increased Epo expression, it would be important to group has recently shown that EE is strongly associated with
question whether the strength of the association between the low circulating sEpoR values and high Epo-to-sEpoR ratios
SENP1 polymorphism and the CMS phenotype remains the (Epo/sEpoR), leading to elevated plasma Epo availability,
same when the proportion of highlander participants with high and thereby a stronger stimulus for erythropoiesis (53) (Fig. 2).
Epo is changed. This is definitely an aspect that must be taken For this reason, when comparing healthy highlanders and those
into consideration in future research. The remarkable differ- with CMS with low and high Epo levels, Hb concentration
ence in Epo levels between the two subgroups of patients with shows an exponential rise with increasing Epo/sEpoR ratio
CMS suggests underlying genetic differences. values, reaching the highest level in the high Epo group.

Table 1. Characteristics of sea-level residents, healthy highlanders, and patients with CMS
Sea-Level Residents Healthy Highlanders Normal-Epo CMS High-Epo CMS

Hb, g/dl 14.5 ⫾ 0.2 17.7 ⫾ 0.2 b


22.2 ⫾ 0.2b,c
24.2 ⫾ 0.8b,c,d
CMS score 1.4 ⫾ 0.3 2.7 ⫾ 0.4a 7.2 ⫾ 0.9b,c 8.0 ⫾ 1.4b,c
Arterial O2 saturation, % 98.3 ⫾ 0.2 88.2 ⫾ 0.6b 84.4 ⫾ 0.7b,c 82.5 ⫾ 1.1b,c
Epo, pg/ml 23.6 ⫾ 1.0 37.8 ⫾ 1.9b 43.5 ⫾ 2.4b 87.1 ⫾ 8.4b,c,d
CMS, Chronic Mountain Sickness; Epo, erythropoietin. Sea-level residents (n ⫽ 25), healthy highlanders (n ⫽ 44), normal-Epo CMS (n ⫽ 32), and high-Epo
CMS (n ⫽ 10). Values are means ⫾ SE. Significance aP ⬍ 0.01 vs. sea-level residents, bP ⬍ 0.001 vs. sea-level residents, cP ⬍ 0.001 vs. healthy highlanders,
d
P ⬍ 0.001 vs. normal-Epo CMS. Data from (53).

J Appl Physiol • doi:10.1152/japplphysiol.00271.2015 • www.jappl.org


Downloaded from journals.physiology.org/journal/jappl (200.105.212.020) on June 20, 2023.
Review
1484 Genetics and Physiology of Excessive Erythrocytosis and CMS • Villafuerte FC

machinery level caused by the chronically lower arterial O2


saturation in patients with CMS or from genetic differences
has yet to be clarified. Also, it is unknown whether differ-
ential expression of the splice EpoR isoforms, sEpoR mE-
poR, contributes to increased Epo action in persons with
similar Epo values, and also whether factors such as SENP1
are involved in EpoR expression and the balance between
mEpoR and sEpoR. It is possible that reduced sEpoR levels
in CMS are accompanied by increased levels of its mem-
brane counterpart leading to increased sensitivity to Epo in
addition to increased Epo availability. Finally, EpoR expres-
sion and the relative abundance of its splice isoforms also
might be influenced by epigenetic factors. Several studies
have suggested that perinatal hypoxia, for example, predis-
poses highlanders to develop EE and CMS during adulthood
(22, 23, 36, 40). Whether perinatal hypoxia or other envi-
ronmental factors affect the Epo-EpoR system is one im-
portant aspect that must be considered in future research.
The different physiological patterns in the Epo-EpoR system
among Andean highlanders together with their different de-
grees of erythropoietic response might indicate specific under-
lying genetic backgrounds, which in turn, might reflect differ-
ent levels of adaptation to chronic high-altitude hypoxia. Pa-
tients with CMS and high Epo, with their exaggerated
erythrocytosis and elevated plasma Epo, could be regarded as
the least adapted, followed by highlanders with CMS and
normal Epo, with still excessive but lower Hb and normal
circulating Epo concentration, and finally healthy Andeans,
with a modestly elevated Hb and normal Epo values for the
altitude of residence, as the most adapted among groups within
the same population.
Therefore, functional and phenotypic differences between
healthy Andean highlanders and those with CMS, and even
among patients with CMS, suggest the involvement of
multilevel physiological control points, each with a poten-
tial specific combination of genetic variants. These variants
would ultimately determine the presence and severity of EE
Fig. 2. Plasma soluble Epo receptor (sEpoR) and the relationship between and the degree of maladaptation to chronic high-altitude
hemoglobin (Hb) and the Epo-to-sEpoR ratio among highlanders and patients
with Chronic Mountain Sickness (CMS). A: plasma sEpoR concentration in hypoxia.
healthy highlanders and individuals with CMS with normal and high plasma
Epo concentration (normal Epo CMS and high-Epo CMS, respectively) at high ACKNOWLEDGMENT
altitude. Bars are means ⫾ SE. *P ⬍ 0.05, ***P ⬍ 0.001. B: relationship
between Epo/sEpoR and Hb concentration. The figure shows nonlinear best-fit I thank Noemi Corante for helpful comments and manuscript
for mean Hb values as a function of mean Epo/sEpoR, including sea level
residents, healthy highlanders, and patients with normal Epo and high Epo
editing.
CMS. Values are means ⫾ SE.
GRANTS
Support for this study was provided by Wellcome Trust Public Health and
At present, information does not exist on the effect of Tropical Medicine Fellowship 097275/Z/11/Z.
lifelong chronic hypoxia exposure on sEpoR. However, DISCLOSURES
experiments in mice have shown that sEpoR expression can
be downregulated by prolonged exposure to hypoxia (33). No conflicts of interest, financial or otherwise, are declared by the author.
Changes in alternative splicing of several genes have been AUTHOR CONTRIBUTIONS
reported as a physiological response to hypoxia, whether by
upregulating or downregulating splice isoform expression F.C.V. conception and design of research; F.C.V. performed experiments;
F.C.V. analyzed data; F.C.V. interpreted results of experiments; F.C.V. drafted
(54). The fact that the accentuated hypoxemia in patients manuscript; F.C.V. edited and revised manuscript; F.C.V. approved final
with CMS and normal Epo compared with healthy highland- version of manuscript.
ers is not associated with higher plasma Epo but with
decreased sEpoR concentration suggests that chronic hyp- REFERENCES
oxia affects Epo and sEpoR in CMS in differential manners. 1. Abdel-Hafiz HA, Horwitz KB. Control of progesterone receptor tran-
Whether reduced blood sEpoR levels result from a chronic scriptional synergy by SUMOylation and deSUMOylation. BMC Mol Biol
downregulation of sEpoR production at the mRNA splicing 13: 10, 2012.

J Appl Physiol • doi:10.1152/japplphysiol.00271.2015 • www.jappl.org


Downloaded from journals.physiology.org/journal/jappl (200.105.212.020) on June 20, 2023.
Review
Genetics and Physiology of Excessive Erythrocytosis and CMS • Villafuerte FC 1485
2. Appenzeller O, Minko T, Qualls C, Pozharov V, Gamboa J, Gamboa monary vascular dysfunction. Am J Physiol Heart Circ Physiol 309:
A, Wang Y. Gene expression, autonomic function and chronic hypoxia: H565–H573, 2015.
lessons from the Andes. Clin Auton Res 16: 217–222, 2006. 23. Julian CG, Vargas E, Gonzales M, Davila RD, Ladenburger A,
3. Baynes RD, Reddy GK, Shih YJ, Skikne BS, Cook JD. Serum form of Reardon L, Schoo C, Powers RW, Lee-Chiong T, Moore LG. Sleep-
the erythropoietin receptor identified by a sequence-specific peptide anti- disordered breathing and oxidative stress in preclinical chronic mountain
body. Blood 82: 2088 –2095, 1993. sickness (excessive erythrocytosis). Respir Physiol Neurobiol 186: 188 –
4. Beall CM, Brittenham GM, Strohl KP, Blangero J, Williams- 196, 2013.
Blangero S, Goldstein MC, Decker MJ, Vargas E, Villena M, Soria R, 24. Khankin EV, Mutter WP, Tamez H, Yuan HT, Karumanchi SA,
Alarcon AM, Gonzales C. Hemoglobin concentration of high-altitude Thadhani R. Soluble erythropoietin receptor contributes to erythropoietin
Tibetans and Bolivian Aymara. Am J Phys Anthropol 106: 385–400, 1998. resistance in end-stage renal disease. PLoS One 5: e9246, 2010.
5. Beall CM, Cavalleri GL, Deng L, Elston RC, Gao Y, Knight J, Li C, 25. Koh MY, Nguyen V, Lemos R Jr, Darnay BG, Kiriakova G, Ab-
Li JC, Liang Y, McCormack M, Montgomery HE, Pan H, Robbins delmelek M, Ho TH, Karam J, Monzon FA, Jonasch E, Powis G.
PA, Shianna KV, Tam SC, Tsering N, Veeramah KR, Wang W, Hypoxia-induced SUMOylation of E3 ligase HAF determines specific
Wangdui P, Weale ME, Xu Y, Xu Z, Yang L, Zaman MJ, Zeng C, activation of HIF2 in clear-cell renal cell carcinoma. Cancer Res 75:
Zhang L, Zhang X, Zhaxi P, Zheng YT. Natural selection on EPAS1 316 –329, 2015.
(HIF2alpha) associated with low hemoglobin concentration in Tibetan 26. Lacombe C, Mayeux P. The molecular biology of erythropoietin. Neph-
highlanders. Proc Natl Acad Sci USA 107: 11459 –11464, 2010. rol Dial Transplant 14, Suppl 2: 22–28, 1999.
6. Cai R, Gu J, Sun H, Liu X, Mei W, Qi Y, Xue S, Ren S, Rabinowitz 27. Lee FS, Percy MJ. The HIF pathway and erythrocytosis. Annu Rev Pathol
JE, Wang Y, Yeh ET, Cheng J. Induction of SENP1 in myocardium 6: 165–192, 2011.
contributes to abnormities of mitochondria and cardiomyopathy. J Mol 28. Lee R, Kertesz N, Joseph SB, Jegalian A, Wu H. Erythropoietin (Epo)
Cell Cardiol 79: 115–122, 2015. and EpoR expression and 2 waves of erythropoiesis. Blood 98: 1408 –
7. Cole AM, Petousi N, Cavalleri GL, Robbins PA. Genetic variation in 1415, 2001.
SENP1 and ANP32D as predictors of Chronic Mountain Sickness. High 29. Leon-Velarde F, Arregui A. Desadaptacion a la vida en las grandes
Alt Med Biol 15: 497–499, 2014. alturas. Lima: Institut français d’études andines (IFEA), 1994.
8. Cheng J, Kang X, Zhang S, Yeh ET. SUMO-specific protease 1 is 30. Leon-Velarde F, Maggiorini M, Reeves JT, Aldashev A, Asmus I,
essential for stabilization of HIF1alpha during hypoxia. Cell 131: 584 – Bernardi L, Ri-Li G, Hackett P, Kobayashi T, Moore LG, Penaloza D,
595, 2007. Richalet JP, Roach R, Wu T, Vargas E, Zubieta-Castillo G, Zubieta-
9. Cheng J, Wang D, Wang Z, Yeh ET. SENP1 enhances androgen Calleja G. Consensus statement on chronic and subacute high altitude
receptor-dependent transcription through desumoylation of histone diseases. High Alt Med Biol 6: 147–157, 2005.
deacetylase 1. Mol Cell Biol 24: 6021–6028, 2004. 31. Leon-Velarde F, Mejia O. Gene expression in chronic high altitude
10. Chiba T, Nagata Y, Kishi A, Sakamaki K, Miyajima A, Yamamoto M, diseases. High Alt Med Biol 9: 130 –139, 2008.
Engel JD, Todokoro K. Induction of erythroid-specific gene expression 32. Leon-Velarde F, Monge CC, Vidal A, Carcagno M, Criscuolo M,
Bozzini CE. Serum immunoreactive erythropoietin in high altitude natives
in lymphoid cells. Proc Natl Acad Sci USA 90: 11593–11597, 1993.
with and without excessive erythrocytosis. Exp Hematol 19: 257–260,
11. Dai XQ, Spigelman AF, Khan S, Braun M, Manning Fox JE, Mac-
1991.
Donald PE. SUMO1 enhances cAMP-dependent exocytosis and glucagon
33. Leon-Velarde F, Rivera-Ch M, Huicho L, Villafuerte FC. Chronic
secretion from pancreatic alpha-cells. J Physiol 592: 3715–3726, 2014.
Mountain Sickness. In: Hypoxia: Human Adaption to High Altitude (2nd
12. Dainiak N, Spielvogel H, Sorba S, Cudkowicz L. Erythropoietin and the
ed.), edited by Swenson ER, Bartsch P. New York: Springer, 2014, p.
polycythemia of high-altitude dwellers. In: Molecular biology of erythro-
429 –448.
poiesis, edited by Ascensao J. New York: Plenum, 1989, p. 17–21.
34. Li S, Wang M, Ao X, Chang AK, Yang C, Zhao F, Bi H, Liu Y, Xiao
13. De Ferrari A, Miranda JJ, Gilman RH, Davila-Roman VG, Leon-
L, Wu H. CLOCK is a substrate of SUMO and sumoylation of CLOCK
Velarde F, Rivera-Ch M, Huicho L, Bernabe-Ortiz A, Wise RA,
upregulates the transcriptional activity of estrogen receptor-alpha. Onco-
Checkley W. Prevalence, clinical profile, iron status, and subject-specific gene 32: 4883–4891, 2013.
traits for excessive erythrocytosis in Andean adults living permanently at 35. Lin CS, Lim SK, D’Agati V, Costantini F. Differential effects of an
3,825 meters above sea level. Chest 146: 1327–1336, 2014. erythropoietin receptor gene disruption on primitive and definitive eryth-
14. Fujita M, Takahashi R, Kitada K, Watanabe R, Kitazawa S, Ashoori ropoiesis. Genes Dev 10: 154 –164, 1996.
F, Liang P, Saya H, Serikawa T, Maeda S. Alternative splicing of the 36. Lumbroso D, Joseph V. Impaired acclimatization to chronic hypoxia in
erythropoietin receptor gene correlates with erythroid differentiation in rat adult male and female rats following neonatal hypoxia. Am J Physiol
hematopoietic and leukemic cells. Cancer Lett 112: 47–55, 1997. Regul Integr Comp Physiol 297: R421–R427, 2009.
15. Fujiwara Y, Browne CP, Cunniff K, Goff SC, Orkin SH. Arrested 37. Manier G, Guenard H, Castaing Y, Varene N, Vargas E. Pulmonary
development of embryonic red cell precursors in mouse embryos lacking gas exchange in Andean natives with excessive polycythemia– effect of
transcription factor GATA-1. Proc Natl Acad Sci USA 93: 12355–12358, hemodilution. J Appl Physiol 65: 2107–2117, 1988.
1996. 38. Maruyama H, Higuchi M, Higuchi N, Kameda S, Saito M, Sugawa M,
16. Gonzales GF, Gasco M, Tapia V, Gonzales-Castaneda C. High serum Matsuzaki J, Neichi T, Yokoyama S, Miyazaki Y, Miyazaki J, Gejyo
testosterone levels are associated to excessive erythrocytosis of chronic F. Post-secretion neutralization of transgene-derived effect: soluble eryth-
mountain sickness in men. Am J Physiol Endocrinol Metab 296: E1319 – ropoietin receptor/IgG1Fc expressed in liver neutralizes erythropoietin
E1325, 2009. produced in muscle. J Gene Med 6: 228 –237, 2004.
17. Gregory T, Yu C, Ma A, Orkin SH, Blobel GA, Weiss MJ. GATA-1 39. Monge CC, Arregui A, Leon-Velarde F. Pathophysiology and epidemi-
and erythropoietin cooperate to promote erythroid cell survival by regu- ology of chronic mountain sickness. Int J Sports Med 13, Suppl 1:
lating bcl-xL expression. Blood 94: 87–96, 1999. S79 –S81, 1992.
18. Harris KW, Winkelmann JC. Enzyme-linked immunosorbent assay 40. Moore LG, Niermeyer S, Vargas E. Does chronic mountain sickness
detects a potential soluble form of the erythropoietin receptor in human (CMS) have perinatal origins? Respir Physiol Neurobiol 158: 180 –189,
plasma. Am J Hematol 52: 8 –13, 1996. 2007.
19. Heaney ML, Golde DW. Soluble cytokine receptors. Blood 87: 847–857, 41. Nagao M, Masuda S, Abe S, Ueda M, Sasaki R. Production and
1996. ligand-binding characteristics of the soluble form of murine erythropoietin
20. Jelkmann W, Metzen E. Erythropoietin in the control of red cell receptor. Biochem Biophys Res Commun 188: 888 –897, 1992.
production. Ann Anat 178: 391–403, 1996. 42. Penaloza D, Arias-Stella J. The heart and pulmonary circulation at high
21. Jiang C, Chen J, Liu F, Luo Y, Xu G, Shen HY, Gao Y, Gao W. altitudes: healthy highlanders and chronic mountain sickness. Circulation
Chronic mountain sickness in Chinese Han males who migrated to the 115: 1132–1146, 2007.
Qinghai-Tibetan plateau: application and evaluation of diagnostic criteria 43. Penaloza D, Sime F. Chronic cor pulmonale due to loss of altitude
for chronic mountain sickness. BMC Public Health 14: 701, 2014. acclimatization (chronic mountain sickness). Am J Med 50: 728 –743,
22. Julian CG, Gonzales M, Rodriguez A, Bellido D, Salinas Salmon C, 1971.
Ladenburger A, Reardon L, Vargas E, Moore LG. Perinatal hypoxia 44. Percy MJ, Chung YJ, Harrison C, Mercieca J, Hoffbrand AV, Di-
increases susceptibility to high-altitude polycythemia and attendant pul- nardo CL, Santos PC, Fonseca GH, Gualandro SF, Pereira AC,

J Appl Physiol • doi:10.1152/japplphysiol.00271.2015 • www.jappl.org


Downloaded from journals.physiology.org/journal/jappl (200.105.212.020) on June 20, 2023.
Review
1486 Genetics and Physiology of Excessive Erythrocytosis and CMS • Villafuerte FC

Lappin TR, McMullin MF, Lee FS. Two new mutations in the HIF2A 58. Winslow RM, Monge CC, Brown EG, Klein HG, Sarnquist F, Win-
gene associated with erythrocytosis. Am J Hematol 87: 439 –442, 2012. slow NJ, McKneally SS. Effects of hemodilution on O2 transport in
45. Richalet JP, Rivera M, Bouchet P, Chirinos E, Onnen I, Petitjean O, high-altitude polycythemia. J Appl Physiol 59: 1495–1502, 1985.
Bienvenu A, Lasne F, Moutereau S, Leon-Velarde F. Acetazolamide: a 59. Wu H, Liu X, Jaenisch R, Lodish HF. Generation of committed
treatment for chronic mountain sickness. Am J Respir Crit Care Med 172: erythroid BFU-E and CFU-E progenitors does not require erythropoietin
1427–1433, 2005. or the erythropoietin receptor. Cell 83: 59 –67, 1995.
46. Rivera-Ch M, Leon-Velarde F, Huicho L. Treatment of chronic moun- 60. Wu R, Cui Y, Yuan X, Yuan H, Wang Y, He J, Zhao J, Peng S.
tain sickness: critical reappraisal of an old problem. Respir Physiol SUMO-specific protease 1 modulates cadmium-augmented transcriptional
Neurobiol 158: 251–265, 2007. activity of androgen receptor (AR) by reversing AR SUMOylation. Toxi-
col Lett 229: 405–413, 2014.
47. Ronen R, Zhou D, Bafna V, Haddad GG. The genetic basis of chronic
61. Xu Y, Zuo Y, Zhang H, Kang X, Yue F, Yi Z, Liu M, Yeh ET, Chen
mountain sickness. Physiology (Bethesda) 29: 403–412, 2014.
G, Cheng J. Induction of SENP1 in endothelial cells contributes to
48. Sahota IS, Panwar NS. Prevalence of Chronic Mountain Sickness in high
hypoxia-driven VEGF expression and angiogenesis. J Biol Chem 285:
altitude districts of Himachal Pradesh. Indian J Occup Environ Med 17: 36682–36688, 2010.
94 –100, 2013. 62. Yi X, Liang Y, Huerta-Sanchez E, Jin X, Cuo ZX, Pool JE, Xu X,
49. Simonson TS, Yang Y, Huff CD, Yun H, Qin G, Witherspoon DJ, Bai Jiang H, Vinckenbosch N, Korneliussen TS, Zheng H, Liu T, He W, Li
Z, Lorenzo FR, Xing J, Jorde LB, Prchal JT, Ge R. Genetic evidence K, Luo R, Nie X, Wu H, Zhao M, Cao H, Zou J, Shan Y, Li S, Yang
for high-altitude adaptation in Tibet. Science 329: 72–75, 2010. Q, Asan Ni P, Tian G, Xu J, Liu X, Jiang T, Wu R, Zhou G, Tang M,
50. Soliz J, Gassmann M, Joseph V. Soluble erythropoietin receptor is Qin J, Wang T, Feng S, Li G, Huasang Luosang J, Wang W, Chen F,
present in the mouse brain and is required for the ventilatory acclimati- Wang Y, Zheng X, Li Z, Bianba Z, Yang G, Wang X, Tang S, Gao G,
zation to hypoxia. J Physiol 583: 329 –336, 2007. Chen Y, Luo Z, Gusang L, Cao Z, Zhang Q, Ouyang W, Ren X, Liang
51. Vargas E, Spielvogel H. Chronic mountain sickness, optimal hemoglo- H, Zheng H, Huang Y, Li J, Bolund L, Kristiansen K, Li Y, Zhang Y,
bin, and heart disease. High Alt Med Biol 7: 138 –149, 2006. Zhang X, Li R, Li S, Yang H, Nielsen R, Wang J, Wang J. Sequencing
52. Vergari E, Plummer G, Dai X, MacDonald PE. DeSUMOylation of 50 human exomes reveals adaptation to high altitude. Science 329:
controls insulin exocytosis in response to metabolic signals. Biomolecules 75–78, 2010.
2: 269 –281, 2012. 63. Yoon D, Kim B, Prchal JT. Cre recombinase expression controlled by
53. Villafuerte FC, Macarlupu JL, Anza-Ramirez C, Corrales-Melgar D, the hematopoietic regulatory domain of Gata-1 is erythroid-specific. Blood
Vizcardo-Galindo G, Corante N, Leon-Velarde F. Decreased plasma Cells Mol Dis 40: 381–387, 2008.
soluble erythropoietin receptor in high-altitude excessive erythrocytosis 64. Youssoufian H, Longmore G, Neumann D, Yoshimura A, Lodish HF.
and Chronic Mountain Sickness. J Appl Physiol 117: 1356 –1362, 2014. Structure, function, and activation of the erythropoietin receptor. Blood
81: 2223–2236, 1993.
54. Weigand JE, Boeckel JN, Gellert P, Dimmeler S. Hypoxia-induced
65. Yu L, Ji W, Zhang H, Renda MJ, He Y, Lin S, Cheng EC, Chen H,
alternative splicing in endothelial cells. PLoS One 7: e42697, 2012.
Krause DS, Min W. SENP1-mediated GATA1 deSUMOylation is critical
55. Wen D, Xu Z, Xia L, Liu X, Tu Y, Lei H, Wang W, Wang T, Song L, for definitive erythropoiesis. J Exp Med 207: 1183–1195, 2010.
Ma C, Xu H, Zhu W, Chen G, Wu Y. Important role of SUMOylation 66. Zhou D, Udpa N, Ronen R, Stobdan T, Liang J, Appenzeller O, Zhao
of Spliceosome factors in prostate cancer cells. J Proteome Res 13: HW, Yin Y, Du Y, Guo L, Cao R, Wang Y, Jin X, Huang C, Jia W,
3571–3582, 2014. Cao D, Guo G, Gamboa JL, Villafuerte F, Callacondo D, Xue J, Liu
56. Westphal G, Braun K, Debus J. Detection and quantification of the S, Frazer KA, Li Y, Bafna V, Haddad GG. Whole-genome sequencing
soluble form of the human erythropoietin receptor (sEpoR) in the growth uncovers the genetic basis of chronic mountain sickness in Andean
medium of tumor cell lines and in the plasma of blood samples. Clin Exp highlanders. Am J Hum Genet 93: 452–462, 2013.
Med 2: 45–52, 2002. 67. Zon LI, Youssoufian H, Mather C, Lodish HF, Orkin SH. Activation
57. Winslow R, Monge-CC. Hypoxia, Polycythemia, and Chronic Mountain of the erythropoietin receptor promoter by transcription factor GATA-1.
Sickness. Baltimore, MD: Johns Hopkins University Press, 1987. Proc Natl Acad Sci USA 88: 10638 –10641, 1991.

J Appl Physiol • doi:10.1152/japplphysiol.00271.2015 • www.jappl.org


Downloaded from journals.physiology.org/journal/jappl (200.105.212.020) on June 20, 2023.

You might also like