World J Mens Health.  2014 Dec;32(3):123-132. 10.5534/wjmh.2014.32.3.123.

Understanding the Role of Heat Shock Protein Isoforms in Male Fertility, Aging and Apoptosis

Affiliations
  • 1Institute of Science, Nirma University, Gujarat, India. sriram.seshadri@nirmauni.ac.in

Abstract

Heat shock proteins (HSPs) play a role in the homeostasis, apoptosis regulation and the maintenance of the various other physiological processes. Aging is accompanied by a decrease in the resistance to environmental stress, while mitochondria are primary targets in the process of aging, their expression decreasing with age. Mitochondrion also plays a significant role in the process of spermatogenesis. HSPs have been shown to be involved in apoptosis with some of acting as apoptotic inhibitors and are involved in cytoprotection. In this review we discuss the roles of Hsp 27, 60, 70, and 90 in aging and male infertility and have concluded that these particular HSPs can be used as a molecular markers for mitochondrially- mediated apoptosis, aging and male infertility.

Keyword

Aging; Heat-shock proteins; Infertility, male; Mitochondria; Apoptosis

MeSH Terms

Aging*
Apoptosis*
Cytoprotection
Fertility*
Heat-Shock Proteins*
Homeostasis
Humans
Infertility, Male
Male
Mitochondria
Physiological Processes
Protein Isoforms*
Spermatogenesis
Heat-Shock Proteins
Protein Isoforms

Figure

  • Fig. 1 Showing the intrinsic and the extrinsic apoptotic pathways. Adapted from Favoloro, et al. Aging (Albany NY) 2012;4:735-42 [78].

  • Fig. 2 Diagram summarizing the common mechanism for the role of HSPs in aging and male fertility.


Reference

1. De Maio A. Heat shock proteins: facts, thoughts, and dreams. Shock. 1999; 11:1–12.
Article
2. Li Z, Srivastava P. Heat-shock proteins. Curr Protoc Immunol. 2004; 02. Appendix 1:Appendix 1T.
3. Simar D, Ruell P, Caillaud C. Hsp responses to exercising in a warm environment. [Internet]. Doha: ASPETAR;c2013. cited 2014 Jul 20. Available from: http://www.aspetar.com/ResearchEducationCentre/HSPResponses.aspx.
4. Li P, Nijhawan D, Budihardjo I, Srinivasula SM, Ahmad M, Alnemri ES, et al. Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell. 1997; 91:479–489.
Article
5. Gupta S, Knowlton AA. HSP60, Bax, apoptosis and the heart. J Cell Mol Med. 2005; 9:51–58.
Article
6. Chandra D, Choy G, Tang DG. Cytosolic accumulation of HSP60 during apoptosis with or without apparent mitochondrial release: evidence that its pro-apoptotic or pro-survival functions involve differential interactions with caspase-3. J Biol Chem. 2007; 282:31289–31301.
7. Zirkin BR, Chen H. Regulation of Leydig cell steroidogenic function during aging. Biol Reprod. 2000; 63:977–981.
8. Kimura M, Itoh N, Takagi S, Sasao T, Takahashi A, Masumori N, et al. Balance of apoptosis and proliferation of germ cells related to spermatogenesis in aged men. J Androl. 2003; 24:185–191.
Article
9. Meinhardt A, Wilhelm B, Seitz J. Expression of mitochondrial marker proteins during spermatogenesis. Hum Reprod Update. 1999; 5:108–119.
10. Adly MA, Assaf HA, Hussein MR. Heat shock protein 27 expression in the human testis showing normal and abnormal spermatogenesis. Cell Biol Int. 2008; 32:1247–1255.
Article
11. Werner A, Meinhardt A, Seitz J, Bergmann M. Distribution of heat-shock protein 60 immunoreactivity in testes of infertile men. Cell Tissue Res. 1997; 288:539–544.
Article
12. Boulanger J, Faulds D, Eddy EM, Lingwood CA. Members of the 70 kDa heat shock protein family specifically recognize sulfoglycolipids: role in gamete recognition and mycoplasma-related infertility. J Cell Physiol. 1995; 165:7–17.
Article
13. Miller D, Brough S, al-Harbi O. Characterization and cellular distribution of human spermatozoal heat shock proteins. Hum Reprod. 1992; 7:637–645.
Article
14. Kamaruddin M, Kroetsch T, Basrur PK, Hansen PJ, King WA. Immunolocalization of heat shock protein 70 in bovine spermatozoa. Andrologia. 2004; 36:327–334.
Article
15. Huang SY, Tam MF, Hsu YT, Lin JH, Chen HH, Chuang CK, et al. Developmental changes of heat-shock proteins in porcine testis by a proteomic analysis. Theriogenology. 2005; 64:1940–1955.
Article
16. Dix DJ, Allen JW, Collins BW, Mori C, Nakamura N, Poorman-Allen P, et al. Targeted gene disruption of Hsp70-2 results in failed meiosis, germ cell apoptosis, and male infertility. Proc Natl Acad Sci U S A. 1996; 93:3264–3268.
Article
17. Minami Y, Kawasaki H, Miyata Y, Suzuki K, Yahara I. Analysis of native forms and isoform compositions of the mouse 90-kDa heat shock protein, HSP90. J Biol Chem. 1991; 266:10099–10103.
Article
18. Lee SJ. Expression of HSP86 in male germ cells. Mol Cell Biol. 1990; 10:3239–3242.
Article
19. Erata GO, Koçak Toker N, Durlanik O, Kadioğlu A, Aktan G, Aykaç Toker G. The role of heat shock protein 70 (Hsp 70) in male infertility: is it a line of defense against sperm DNA fragmentation? Fertil Steril. 2008; 90:322–327.
Article
20. Feng HL, Sandlow JI, Sparks AE. Decreased expression of the heat shock protein hsp70-2 is associated with the pathogenesis of male infertility. Fertil Steril. 2001; 76:1136–1139.
Article
21. Hunt C, Morimoto RI. Conserved features of eukaryotic hsp70 genes revealed by comparison with the nucleotide sequence of human hsp70. Proc Natl Acad Sci U S A. 1985; 82:6455–6459.
Article
22. Sarge KD, Cullen KE. Regulation of hsp expression during rodent spermatogenesis. Cell Mol Life Sci. 1997; 53:191–197.
Article
23. Rong C, Han J, Du Z. Expression of heat shock protein 90β and its regulation in the reproductive system of male mice. Nan Fang Yi Ke Da Xue Xue Bao. 2013; 33:491–495.
24. Moore SK, Kozak C, Robinson EA, Ullrich SJ, Appella E. Murine 86- and 84-kDa heat shock proteins, cDNA sequences, chromosome assignments, and evolutionary origins. J Biol Chem. 1989; 264:5343–5351.
Article
25. Ohsako S, Bunick D, Hayashi Y. Immunocytochemical observation of the 90 KD heat shock protein (HSP90): high expression in primordial and pre-meiotic germ cells of male and female rat gonads. J Histochem Cytochem. 1995; 43:67–76.
26. Ecroyd H, Jones RC, Aitken RJ. Tyrosine phosphorylation of HSP-90 during mammalian sperm capacitation. Biol Reprod. 2003; 69:1801–1807.
27. Wu C. Heat shock transcription factors: structure and regulation. Annu Rev Cell Dev Biol. 1995; 11:441–469.
Article
28. Liu Z, Wang G, Pan Y, Zhu C. Expression of androgen receptor and heat shock protein 90alpha in the testicular biopsy specimens of infertile patients with spermatogenic arrest. Zhonghua Nan Ke Xue. 2004; 10:662–666.
29. Muller FL, Lustgarten MS, Jang Y, Richardson A, Van Remmen H. Trends in oxidative aging theories. Free Radic Biol Med. 2007; 43:477–503.
Article
30. Landis GN, Tower J. Superoxide dismutase evolution and life span regulation. Mech Ageing Dev. 2005; 126:365–379.
Article
31. Macario AJ, Conway de Macario E. Sick chaperones, cellular stress, and disease. N Engl J Med. 2005; 06. 353:1489–1501.
Article
32. Soti C, Csermely P. Chaperones come of age. Cell Stress Chaperones. 2002; 7:186–190.
33. Tatar M, Khazaeli AA, Curtsinger JW. Chaperoning extended life. Nature. 1997; 390:30.
Article
34. Tower J. Hsps and aging. Trends Endocrinol Metab. 2009; 20:216–222.
35. Jonak C, Klosner G, Trautinger F. Heat shock proteins in the skin. Int J Cosmet Sci. 2006; 28:233–241.
Article
36. Préville X, Salvemini F, Giraud S, Chaufour S, Paul C, Stepien G, et al. Mammalian small stress proteins protect against oxidative stress through their ability to increase glucose-6-phosphate dehydrogenase activity and by maintaining optimal cellular detoxifying machinery. Exp Cell Res. 1999; 247:61–78.
Article
37. Yan LJ, Christians ES, Liu L, Xiao X, Sohal RS, Benjamin IJ. Mouse heat shock transcription factor 1 deficiency alters cardiac redox homeostasis and increases mitochondrial oxidative damage. EMBO J. 2002; 21:5164–5172.
Article
38. Facchinetti F, Dawson VL, Dawson TM. Free radicals as mediators of neuronal injury. Cell Mol Neurobiol. 1998; 18:667–682.
Article
39. Merendino AM, Paul C, Vignola AM, Costa MA, Melis M, Chiappara G, et al. Heat shock protein-27 protects human bronchial epithelial cells against oxidative stress-mediated apoptosis: possible implication in asthma. Cell Stress Chaperones. 2002; 7:269–280.
Article
40. Bryantsev AL, Kurchashova SY, Golyshev SA, Polyakov VY, Wunderink HF, Kanon B, et al. Regulation of stress-induced intracellular sorting and chaperone function of Hsp27 (HspB1) in mammalian cells. Biochem J. 2007; 407:407–417.
Article
41. Renkawek K, Stege GJ, Bosman GJ. Dementia, gliosis and expression of the small heat shock proteins hsp27 and alpha B-crystallin in Parkinson's disease. Neuroreport. 1999; 10:2273–2276.
42. Cappello F, Conway de Macario E, Marino Gammazza A, Bonaventura G, Carini F, Czarnecka AM, et al. Hsp60 and human aging: Les liaisons dangereuses. Front Biosci (Landmark Ed). 2013; 18:626–637.
43. Spector NL, Mehlen P, Ryan C, Hardy L, Samson W, Levine H, et al. Regulation of the 28 kDa heat shock protein by retinoic acid during differentiation of human leukemic HL-60 cells. FEBS Lett. 1994; 337:184–188.
44. Proctor CJ, Soti C, Boys RJ, Gillespie CS, Shanley DP, Wilkinson DJ, et al. Modelling the actions of chaperones and their role in ageing. Mech Ageing Dev. 2005; 126:119–131.
Article
45. Buchner J. Hsp90 & Co. - a holding for folding. Trends Biochem Sci. 1999; 24:136–141.
46. Rao DV, Watson K, Jones GL. Age-related attenuation in the expression of the major heat shock proteins in human peripheral lymphocytes. Mech Ageing Dev. 1999; 107:105–118.
Article
47. Zhang HJ, Drake VJ, Morrison JP, Oberley LW, Kregel KC. Selected contribution: differential expression of stress-related genes with aging and hyperthermia. J Appl Physiol (1985). 2002; 92:1762–1769. discussion 1749.
Article
48. Stolzing A, Sethe S, Scutt AM. Stressed stem cells: Temperature response in aged mesenchymal stem cells. Stem Cells Dev. 2006; 15:478–487.
Article
49. Faassen AE, O'Leary JJ, Rodysill KJ, Bergh N, Hallgren HM. Diminished heat-shock protein synthesis following mitogen stimulation of lymphocytes from aged donors. Exp Cell Res. 1989; 183:326–334.
Article
50. Hunter T, Poon RY. Cdc37: a protein kinase chaperone? Trends Cell Biol. 1997; 7:157–161.
Article
51. Holt SE, Aisner DL, Baur J, Tesmer VM, Dy M, Ouellette M, et al. Functional requirement of p23 and Hsp90 in telomerase complexes. Genes Dev. 1999; 13:817–826.
Article
52. Martin JA, Buckwalter JA. Aging, articular cartilage chondrocyte senescence and osteoarthritis. Biogerontology. 2002; 3:257–264.
53. Ferlin A, Speltra E, Patassini C, Pati MA, Garolla A, Caretta N, et al. Heat shock protein and heat shock factor expression in sperm: relation to oligozoospermia and varicocele. J Urol. 2010; 183:1248–1252.
Article
54. Mazzoli S, Cai T, Addonisio P, Bechi A, Mondaini N, Bartoletti R. Chlamydia trachomatis infection is related to poor semen quality in young prostatitis patients. Eur Urol. 2010; 57:708–714.
Article
55. Venkataraman S, Wagner BA, Jiang X, Wang HP, Schafer FQ, Ritchie JM, et al. Overexpression of manganese superoxide dismutase promotes the survival of prostate cancer cells exposed to hyperthermia. Free Radic Res. 2004; 38:1119–1132.
Article
56. Davidson JF, Schiestl RH. Mitochondrial respiratory electron carriers are involved in oxidative stress during heat stress in Saccharomyces cerevisiae. Mol Cell Biol. 2001; 21:8483–8489.
57. Heise K, Puntarulo S, Pörtner HO, Abele D. Production of reactive oxygen species by isolated mitochondria of the Antarctic bivalve Laternula elliptica (King and Broderip) under heat stress. Comp Biochem Physiol C Toxicol Pharmacol. 2003; 134:79–90.
Article
58. Qian L, Song X, Ren H, Gong J, Cheng S. Mitochondrial mechanism of heat stress-induced injury in rat cardiomyocyte. Cell Stress Chaperones. 2004; 9:281–293.
Article
59. Skulachev VP, Longo VD. Aging as a mitochondria-mediated atavistic program: can aging be switched off? Ann N Y Acad Sci. 2005; 1057:145–164.
Article
60. Haak JL, Buettner GR, Spitz DR, Kregel KC. Aging augments mitochondrial susceptibility to heat stress. Am J Physiol Regul Integr Comp Physiol. 2009; 296:R812–R820.
Article
61. Landis GN, Abdueva D, Skvortsov D, Yang J, Rabin BE, Carrick J, et al. Similar gene expression patterns characterize aging and oxidative stress in Drosophila melanogaster. Proc Natl Acad Sci U S A. 2004; 101:7663–7668.
Article
62. Pletcher SD, Macdonald SJ, Marguerie R, Certa U, Stearns SC, Goldstein DB, et al. Genome-wide transcript profiles in aging and calorically restricted Drosophila melanogaster. Curr Biol. 2002; 12:712–723.
Article
63. Zahn JM, Sonu R, Vogel H, Crane E, Mazan-Mamczarz K, Rabkin R, et al. Transcriptional profiling of aging in human muscle reveals a common aging signature. PLoS Genet. 2006; 2:e115.
Article
64. Shigenaga MK, Hagen TM, Ames BN. Oxidative damage and mitochondrial decay in aging. Proc Natl Acad Sci U S A. 1994; 91:10771–10778.
Article
65. Ames BA, Shingenaga MK, Park EM. Davies KJA, editor. Oxidative damage and repair, chemical, biological and medical aspects. Elmstad, New York: Pergamon;1991. p. 181–187.
66. Laganiere S, Yu BP. Modulation of membrane phospholipid fatty acid composition by age and food restriction. Gerontology. 1993; 39:7–18.
Article
67. Deocaris CC, Kaul SC, Wadhwa R. On the brotherhood of the mitochondrial chaperones mortalin and heat shock protein 60. Cell Stress Chaperones. 2006; 11:116–128.
Article
68. Wadhwa R, Taira K, Kaul SC. An Hsp70 family chaperone, mortalin/mthsp70/PBP74/Grp75: what, when, and where? Cell Stress Chaperones. 2002; 7:309–316.
Article
69. Bulteau AL, Szweda LI, Friguet B. Mitochondrial protein oxidation and degradation in response to oxidative stress and aging. Exp Gerontol. 2006; 41:653–657.
Article
70. Rea IM, McNerlan S, Pockley AG. Serum heat shock protein and anti-heat shock protein antibody levels in aging. Exp Gerontol. 2001; 36:341–352.
Article
71. Imao M, Nagaki M, Moriwaki H. Dual effects of heat stress on tumor necrosis factor-alpha-induced hepatocyte apoptosis in mice. Lab Invest. 2006; 86:959–967.
72. Sachidhanandam SB, Lu J, Low KS, Moochhala SM. Herbimycin A attenuates apoptosis during heat stress in rats. Eur J Pharmacol. 2003; 474:121–128.
Article
73. Concannon CG, Gorman AM, Samali A. On the role of Hsp27 in regulating apoptosis. Apoptosis. 2003; 8:61–70.
74. Mehlen P, Schulze-Osthoff K, Arrigo AP. Small stress proteins as novel regulators of apoptosis. Heat shock protein 27 blocks Fas/APO-1- and staurosporine-induced cell death. J Biol Chem. 1996; 271:16510–16514.
75. Samali A, Robertson JD, Peterson E, Manero F, van Zeijl L, Paul C, et al. Hsp27 protects mitochondria of thermotolerant cells against apoptotic stimuli. Cell Stress Chaperones. 2001; 6:49–58.
Article
76. Moriyama-Gonda N, Igawa M, Shiina H, Urakami S, Wada Y, Terashima M. Heat-induced cellular damage and tolerance in combination with adriamycin for the PC-3 prostate cancer cell line: relationships with cytotoxicity, reactive oxygen species and heat shock protein 70 expression. Eur Urol. 2000; 38:235–240.
77. Pallepati P, Averill-Bates DA. Mild thermotolerance induced at 40℃ protects HeLa cells against activation of death receptor-mediated apoptosis by hydrogen peroxide. Free Radic Biol Med. 2011; 50:667–679.
78. Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer. 1972; 26:239–257.
Article
79. Steller H. Mechanisms and genes of cellular suicide. Science. 1995; 267:1445–1449.
Article
80. Favaloro B, Allocati N, Graziano V, Di Ilio C, De Laurenzi V. Role of apoptosis in disease. Aging (Albany NY). 2012; 4:330–349.
Article
81. Wickman G, Julian L, Olson MF. How apoptotic cells aid in the removal of their own cold dead bodies. Cell Death Differ. 2012; 19:735–742.
Article
82. Ai X, Butts B, Vora K, Li W, Tache-Talmadge C, Fridman A, et al. Generation and characterization of antibodies specific for caspase-cleaved neo-epitopes: a novel approach. Cell Death Dis. 2011; 2:e205.
Article
83. Lüthi AU, Martin SJ. The CASBAH: a searchable database of caspase substrates. Cell Death Differ. 2007; 14:641–650.
Article
84. Xanthoudakis S, Roy S, Rasper D, Hennessey T, Aubin Y, Cassady R, et al. Hsp60 accelerates the maturation of pro-caspase-3 by upstream activator proteases during apoptosis. EMBO J. 1999; 18:2049–2056.
Article
85. Morimoto RI. Proteotoxic stress and inducible chaperone networks in neurodegenerative disease and aging. Genes Dev. 2008; 22:1427–1438.
Article
86. Garrido C, Brunet M, Didelot C, Zermati Y, Schmitt E, Kroemer G. Heat shock proteins 27 and 70: anti-apoptotic proteins with tumorigenic properties. Cell Cycle. 2006; 5:2592–2601.
Article
87. Mosser DD, Caron AW, Bourget L, Meriin AB, Sherman MY, Morimoto RI, et al. The chaperone function of hsp70 is required for protection against stress-induced apoptosis. Mol Cell Biol. 2000; 20:7146–7159.
Article
88. Mosser DD, Morimoto RI. Molecular chaperones and the stress of oncogenesis. Oncogene. 2004; 23:2907–2918.
Article
89. Kamada M, So A, Muramaki M, Rocchi P, Beraldi E, Gleave M. Hsp27 knockdown using nucleotide-based therapies inhibit tumor growth and enhance chemotherapy in human bladder cancer cells. Mol Cancer Ther. 2007; 6:299–308.
Article
90. Aghdassi A, Phillips P, Dudeja V, Dhaulakhandi D, Sharif R, Dawra R, et al. Heat shock protein 70 increases tumorigenicity and inhibits apoptosis in pancreatic adenocarcinoma. Cancer Res. 2007; 67:616–625.
Article
91. Compton SA, Elmore LW, Haydu K, Jackson-Cook CK, Holt SE. Induction of nitric oxide synthase-dependent telomere shortening after functional inhibition of Hsp90 in human tumor cells. Mol Cell Biol. 2006; 26:1452–1462.
Article
92. Gurbuxani S, Bruey JM, Fromentin A, Larmonier N, Parcellier A, Jäättelä M, et al. Selective depletion of inducible HSP70 enhances immunogenicity of rat colon cancer cells. Oncogene. 2001; 20:7478–7485.
Article
93. Matsumori Y, Hong SM, Aoyama K, Fan Y, Kayama T, Sheldon RA, et al. Hsp70 overexpression sequesters AIF and reduces neonatal hypoxic/ischemic brain injury. J Cereb Blood Flow Metab. 2005; 25:899–910.
Article
94. Marin-Vinader L, Shin C, Onnekink C, Manley JL, Lubsen NH. Hsp27 enhances recovery of splicing as well as rephosphorylation of SRp38 after heat shock. Mol Biol Cell. 2006; 17:886–894.
Article
95. Liao W, Li X, Mancini M, Chan L. Proteasome inhibition induces differential heat shock protein response but not unfolded protein response in HepG2 cells. J Cell Biochem. 2006; 99:1085–1095.
Article
96. Gusarova V, Caplan AJ, Brodsky JL, Fisher EA. Apoprotein B degradation is promoted by the molecular chaperones hsp90 and hsp70. J Biol Chem. 2001; 276:24891–24900.
Article
97. Garrido C, Solary E. A role of HSPs in apoptosis through "protein triage"? Cell Death Differ. 2003; 10:619–620.
Article
98. Steel R, Doherty JP, Buzzard K, Clemons N, Hawkins CJ, Anderson RL. Hsp72 inhibits apoptosis upstream of the mitochondria and not through interactions with Apaf-1. J Biol Chem. 2004; 279:51490–51499.
Article
99. Pandey P, Saleh A, Nakazawa A, Kumar S, Srinivasula SM, Kumar V, et al. Negative regulation of cytochrome c-mediated oligomerization of Apaf-1 and activation of procaspase-9 by heat shock protein 90. EMBO J. 2000; 19:4310–4322.
Article
100. Kim HE, Jiang X, Du F, Wang X. PHAPI, CAS, and Hsp70 promote apoptosome formation by preventing Apaf-1 aggregation and enhancing nucleotide exchange on Apaf-1. Mol Cell. 2008; 30:239–247.
Article
Full Text Links
  • WJMH
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr