Blood Disorders in the Elderly - part 3 pdf

50 328 0
Blood Disorders in the Elderly - part 3 pdf

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

88 Rita B. Effros of dying from infectious causes compared to those individuals with the longest telomeres [57]. It should be emphasized that such correlative studies do not in any way suggest that it is telomere shortening per se that is the cause of mortality. Rather, it is more likely that the reduced telomere length is a biomarker of other physiological changes [58]. For example, in the case of shorter telomeres being associated with increased death from infec- tious causes, one possible mechanism that might be operating is that the T cells were working over- time (and eventually failing) to control a particular infection, and in the process undergoing extensive cell division and concomitant telomere shortening. Studies on HIV-infected persons are consistent with this notion, since over many years the chronic acti- vation and proliferation of CD8 T cells does eventu- ally lead to high proportions of CD8 T cells that lack CD28 expression and have shortened telomeres. An alternative possibility to explain the short-telomere/ infection association relates to the observation that telomere length is a heritable trait [59–61], and may be linked to other genetic factors that are the true cause of the increased death risk from infections. Given that infections are a major cause of mor- bidity and mortality in the elderly, vaccination is an important prophylactic strategy. Infl uenza, in particular, has been shown to be the fourth leading cause of death in elderly persons, so that this age group is a priority target population for infl uenza vaccination. Thus, it is highly relevant that two stud- ies have shown a signifi cant correlation between poor response to infl uenza vaccination and high proportions of senescent CD8 T cells. The under- lying mechanism for this association has not been identifi ed, but in other contexts, CD8 T cells that lack CD28 expression have been shown to have sup- pressor cell functions, leading to downregulation of antigen presentation as well as other T-cell activi- ties [62]. CD8ϩCD28– T cells also accumulate and mediate liver damage in hepatitis C infection [63]. Suppressor functions have also been attributed to CD8 T cells that are CD57-positive, a phenotype associated with loss of CD28 expression. These putatively senescent CD8 T cells exert suppressive infl uences on effector functions of HIV-specifi c CTL [64]. Another interesting correlation that has emerged from clinical studies is the association between high proportions of senescent CD8 T cells and oste- oporotic fractures in a group of elderly women [65]. Although this was a small-scale study, increasing evidence suggests that chronic immune activation is, in fact, associated with bone loss [66]. Moreover, the profi le of cytokines produced by senescent T cells (e.g., increased IL-6 and reduced IFN-γ) would be predicted to favor maturation and activation of osteoclasts, the bone-resorbing cells. Further research in the relatively new fi eld of osteoimmunol- ogy will undoubtedly uncover new and important mechanisms that link the immune system of the eld- erly with some of the well-documented age-related skeletal changes. Senescent T cells and cancer One of the fundamental questions spanning the fi elds of both cancer biology and immunology is whether immune surveillance plays a role in tumor initiation and progression. Although for cancers in general this issue has not been resolved, there is accumulating evidence suggesting that in certain virally related cancers, exhaustion of immune con- trol over the virus may play a role in tumor initiation [67]. Immune defi ciency is, in fact, closely correlated with several types of tumors that have viral etiologies. For example, in immunosuppressed individuals, vir- tually all lymphomas are EBV in origin, presumably resulting from the ultimate failure of T cells to effec- tively control EBV infection [68,69]. Another latent herpesvirus-associated tumor, Kaposi’s sarcoma, is increased in HIV-infected persons, and cervical cancer, which also increases during immune sup- pression, is associated with certain strains of human papillomavirus. Viruses that are able to establish latency develop a complex relationship with the host’s immune sys- tem. Evasion of immune recognition as well as spe- cifi c physiological effects on the T cells themselves Replicative senescence, aging, and cancer 89 are probably involved [70]. It is clear that the initial primary infection with these viruses does elicit an immune response. During the acute phase of infec- tious mononucleosis (EBV infection), for example, high levels of telomerase activity and activation markers can be detected on the antigen-specifi c CD8 T cells. Nevertheless, one year after infection, when presumably the virus has become latent, these same T cells show evidence of having experienced chronic antigenic stimulation, as indicated by telomere shortening of the tetramer-binding CD8 T cells [17]. These data suggest that, at least in the case of EBV, latency is associated with prolonged antigen-specifi c proliferation in vivo. Since EBV is involved not only in lymphomas, but also in invasive breast cancer and in some tumors of the prostate and of the liver [68], it is possible that immune exhaustion caused by replicative senescence of virus-specifi c CD8 T cells plays a role in the development of a broad spectrum of tumor types. Persons with virally associated tumors do, in fact, have increased proportions of CD8 T cells with char- acteristics reminiscent of T cells that reach replicative senescence in cell culture, suggesting an association between loss of control over the virus and transfor- mation of the latently infected cells [4]. Indeed, it has been shown that antigen-specifi c CD8 T cells in sev- eral chronic viral infections, such as HIV, CMV, and EBV, eventually lose their antiviral cytolytic function once the infection becomes chronic [71]. Interesting, in patients with certain EBV-associated nasopha- ryngeal tumors, such fundamental CD8-T-cell pro- tective functions as secretion of IFN-γ and perforin expression by CD8 T cells are also impaired [72]. Moreover, in many of these cancer patients, reduced EBV-specifi c CTL precursor frequency has also been documented and, importantly, the defi cit correlated with plasma viral burden [73]. Since the limiting dilution assay used to detect precursor frequency is critically dependent on proliferation, the above observation is consistent with a role for proliferative exhaustion. In addition, EBV-associated lymphomas are correlated with high tumor necrosis factor α lev- els, reminiscent of senescent T-cell cultures [74]. In sum, there is increasing evidence lending support to the hypothesis that chronic exposure to antigens of latent viruses (e.g., EBV, HPV) may facilitate tumor progression and metastasis by driving the relevant antigen-specifi c T cells to senescence. The potential to generate senescent antigen- reactive T cells may not be restricted to situations involving latent infections. Certain non-viral tumor- associated antigens may also be a source of chronic immune stimulation. For example, prostate-specifi c antigen (PSA), the blood levels of which increase in persons with prostate cancer, is also present in nor- mal prostate tissue, and is thus an antigen to which T cells have had prolonged exposure [75]. CD8 T cells from patients with prostate cancer do, in fact, show reactivity to PSA peptides immediately ex vivo [76], consistent with the notion that they were previously primed in vivo to this antigen. Similarly, melanoma- specifi c antigens, which cause chronic activation of T cells, have been suggested to play a role in the loss of CD28 expression in some melanoma patients [77]. Thus, like antigens of viruses that establish latency, tumor-associated antigens also have the potential to cause chronic T-cell activation, possibly driving some antigen-specifi c cells to senescence. As noted above, loss of CD28 expression is the signature change of CD8 T-cell senescence in cell culture. It is thus relevant to note that altered expres- sion of CD28, and by implication replicative senes- cence, has also been associated with the clinical outcome of certain non-viral cancers. In advanced renal carcinoma, for example, the proportion of CD8 T cells that are CD57ϩ (a marker present on a majority of CD28– T cells) has predictive value with respect to patient survival [78]. Further, in patients with head and neck tumors, it has been shown that tumor resection is associated with a reduction in the CD8ϩCD28– T-cell subset, which had undergone expansion during the period of tumor growth [37]. Thus, replicative senescence of CD8 T cells, already implicated in defective immunity to chronic viral infections [44], may also play a role in the failed immune surveillance that may facilitate the devel- opment or metastasis of certain types of cancer. In addition to possibly facilitating the develop- ment of some tumors, the process of CD8-T-cell 90 Rita B. Effros replicative senescence also has an impact on adop- tive immunotherapy for cancer, since sustained control over the tumor requires extensive T-cell pro- liferation and maintenance of functional integrity. The impediment of replicative senescence has, in fact, been documented in the case of EBV, where in- vitro expansion of EBV-specifi c CD8 T cells for the purpose of cancer immunotherapy is associated with loss of cytolytic function [79, 80]. This change is con- sistent with observations from cell culture studies on replicative senescence [81]. Thus, prevention or retardation of the process of replicative senescence will lead to improvement in immunotherapy directed at cancer, one of the major diseases of old age. Solutions to the problem of T-cell replicative senescence Given the spectrum of deleterious effects associated with senescent T cells, investigators are actively pursuing strategies to reverse, prevent or retard the process of replicative senescence. Based on the cen- tral role of telomere shortening in signaling the cell- cycle arrest, one of the major approaches has been manipulation of the enzyme telomerase, either by genetic or by pharmacologic methods. Gene trans- duction with the catalytic component of human telomerase (hTERT) has been extensively analyzed in human fi broblasts, epithelial cells, and keratino- cytes. These studies have documented that the transduced cells show unlimited proliferation, tel- omere length stabilization, normalization of func- tion, and, importantly, no evidence of altered growth or tumorogenesis in immunodefi cient (SCID) mice. In CD8 T cells, gene transduction with hTERT is able to reverse some, but not all, of the components of the replicative senescence program. CD8 T cells that are specifi c for tumors and for HIV have both been shown to acquire unlimited proliferative capac- ity following transduction with hTERT. Nevertheless, the ultimate loss of CD28 expression is not prevented by this strategy [81,82]. The importance of retaining CD28 expression has been documented in several studies of cancer immunotherapy and anti-tumor vaccines, in which incorporation of the CD28 ligand, B7, enhanced treatment effi cacy [34,83–85]. Genetic modulation of telomerase activity also fails to pre- vent the ultimate collapse of antigen-specifi c cyto- lytic function in virus-specifi c cultures [86]. Ongoing research is addressing whether combinations of hTERT and CD28 gene therapies will result in more comprehensive correction of the features of CD8- T-cell replicative senescence. Because of the complexity and impractical aspects of gene-therapy approaches, efforts are also directed at identifying pharmacologic agents that might accomplish the same goals. It has been known for some time that cells of the immune sys- tem contain estrogen receptors; the original radio- active estrogen binding studies suggested that CD8 T cells, in particular, bind estrogen with high affi nity [87]. Although little is known about the spectrum of T-cell genes that are modulated by estrogen, an estrogen-responsive element has been documented in the promoter region of IFN-γ [88], a cytokine that is often monitored in evaluating immune responses to viruses and cancer [89]. Interestingly, IFN-γ has also been recently shown to upregulate the enzyme telomerase in T cells [90]. Estrogen can also directly modulate telomerase activity; there is an estrogen-responsive element in the promoter of the hTERT gene in a variety of reproductive tissues [91]. Estrogen also affects cal- cium mobilization in T cells. Thus, evidence from a variety of systems suggests that estrogen has the potential to modulate several T-cell functions that are altered in senescent cells, and may therefore constitute a novel type of non-genetic strategy to modulate senescence. Clearly, application of these hormone-based approaches to cancer immuno- therapy or to modulation of antiviral immunity will require identifying designer estrogens that specifi - cally affect T cells, but not estrogen-sensitive tumor cells. Finally, research on non-hormonal modulators of T-cell telomerase activity may provide additional approaches to modulating replicative senescence, thereby expanding the effi cacy of cancer immuno- therapy and effective control over viral infections in the elderly [92]. Replicative senescence, aging, and cancer 91 ACKNOWLEDGEMENTS The research described in this chapter has been supported in part by the NIH and the UCLA Center on Aging. Dr. Effros holds the Thomas and Elizabeth Plott Endowed Chair in Gerontology. REFERENCES 1. Hayfl ick L, Moorhead PS. The serial cultivation of human diploid cell strains. Exp Cell Res 1961; 25: 585–621. 2. Effros RB, Dagarag M, Spaulding CC, Man J. The role of CD8ϩ T-cell replicative senescence in human aging. Immunol Rev 2005; 205: 147–57. 3. Akbar AN, Soares MV, Plunkett FJ, Salmon M. Differential regulation of CD8ϩ T cell senescence in mice and men. Mech Ageing Dev 2000; 121: 69–76. 4. Effros RB, Cai Z, Linton PJ. CD8 T cells and aging. Crit Rev Immunol 2003; 23: 45–64. 5. Janeway CA, Travers P, Walpert MSM. The Immune System in Health and Disease, 5th edn (New York, NY: Garland, 2001). 6. Effros RB, Pawelec G. Replicative senescence of T lymphocytes: does the Hayfl ick limit lead to immune exhaustion? Immunol Today 1997; 18: 450–4. 7. Perillo NL, Walford RL, Newman MA, Effros RB. Human T lymphocytes possess a limited in vitro lifespan. Exp Gerontol 1989; 24: 177–87. 8. Pawelec G, Rehbein A, Haehnel K, Meri A, Adibzadeh M. Human T cell clones in long-term culture as a model of immunosenescence. Immunol Rev 1997; 160: 31–42. 9. Levine BL, Cotte J, Small CC, et al. Large-scale produc- tion of CD4ϩ T cells from HIV-1-infected donors after CD3/CD28 costimulation. J Hematother 1998; 7: 437–48. 10. Harley C, Futcher AB, Greider C. Telomeres shorten during ageing of human fi broblasts. Nature 1990; 345: 458–60. 11. Bodnar AG, Kim NW, Effros RB, Chiu CP. Mechanism of telomerase induction during T cell activation. Exp Cell Res 1996; 228: 58–64. 12. Weng NP, Levine BL, June CH, Hodes RJ. Human naive and memory T lymphocytes differ in telomeric length and replicative potential. Proc Natl Acad Sci USA 1995; 92: 11091–4. 13. Smogorzewska A, de Lange T. Regulation of telomer- ase by telomeric proteins. Annu Rev Biochem 2004; 73: 177–208. 14. Blackburn EH. Telomeres and telomerase: their mech- anisms of action and the effects of altering their func- tions. FEBS Lett 2005; 579: 859–62. 15. Vaziri H, Schachter F, Uchida I, et al. Loss of telomeric DNA during aging of normal and trisomy 21 human lymphocytes. Am J Hum Genet 1993; 52: 661–7. 16. Weng NP, Palmer LD, Levine BL, Lane HC, June CH, Hodes RJ. Tales of tails: regulation of telomere length and telomerase activity during lymphocyte develop- ment, differentiation, activation, and aging. Immunol Rev 1997; 160: 43–54. 17. Maini MK, Soares MV, Zilch CF, Akbar AN, Beverley PC. Virus-induced CD8ϩ T cell clonal expansion is asso- ciated with telomerase up-regulation and telomere length preservation: a mechanism for rescue from rep- licative senescence. J Immunol 1999; 162: 4521–6. 18. Valenzuela HF, Effros RB. Loss of telomerase inducibil- ity in memory T cells with repeated antigenic stimula- tion. FASEB J 2000; 14: A991. 19. Spaulding CS, Guo W, Effros RB. Resistance to apop- tosis in human CD8ϩ T cells that reach replicative senescence after multiple rounds of antigen-specifi c proliferation. Exp Gerontol 1999; 34: 633–44. 20. Wang E, Lee MJ, Pandey S. Control of fi broblast senes- cence and activation of programmed cell death. J Cell Biochem 1994; 54: 432–9. 21. Effros RB, Zhu X, Walford RL. Stress response of senes- cent T lymphocytes: reduced hsp70 is independent of the proliferative block. J Gerontol 1994; 49: B65–B70. 22. Krichevsky S, Pawelec G, Gural A, et al. Age related microsatellite instability in T cells from healthy indi- viduals. Exp Gerontol 2004; 39: 507–15. 23. Effros RB, Boucher N, Porter V, et al. Decline in CD28ϩ T cells in centenarians and in long-term T cell cultures: a possible cause for both in vivo and in vitro immu- nosenescence. Exp Gerontol 1994; 29: 601–9. 24. Boucher N, Defeu-Duchesne T, Vicaut E, Farge D, Effros RB, Schachter F. CD28 expression in T cell aging and human longevity. Exp Gerontol 1998; 33: 267–82. 25. Azuma M, Phillips JH, Lanier LL. CD28– T lymphocytes: antigenic and functional properties. J Immunol 1993; 150: 1147–59. 26. Nilsson BO, Ernerudh J, Johansson B, et al. Morbidity does not infl uence the T-cell immune risk phenotype in the elderly: fi ndings in the Swedish NONA Immune Study using sample selection protocols. Mech Ageing Dev 2003; 124: 469–76. 27. Effros RB, Allsopp R, Chiu CP, et al. Shortened telomeres in the expanded CD28–CD8ϩ subset in HIV disease 92 Rita B. Effros implicate replicative senescence in HIV pathogenesis. AIDS/Fast Track 1996; 10: F17–F22. 28. Borthwick NJ, Bofi ll M, Gombert WM. Lymphocyte activation in HIV-1 infection II: functional defects of CD28– T cells. AIDS 1994; 8: 431–41. 29. Jennings C, Rich K, Siegel JN, Landay A. A phenotypic study of CD8ϩ lymphocyte subsets in infants using three-color fl ow cytometry. Clin Immunol Immunopath 1994; 71: 8–13. 30. Brinchmann JE, Dobloug JH, Heger BH, Haaheim LL, Sannes M, Egeland T. Expression of costimulatory mol- ecule CD28 on T cells in human immunodefi ciency virus type 1 infection: functional and clinical correla- tions. J Infect Dis 1994; 169: 730–8. 31. Pantaleo G, Koenig S, Baseler M, Lane HC, Fauci AS. Defective clonogenic potential of CD8ϩ T lymphocytes in patients with AIDS: expansion in vivo of a nonclo- nogenic CD3ϩCD8ϩDRϩCD25– T cell population. J Immunol 1990; 144: 1696–704. 32. Lewis DE, Tang DSN, Adu-Oppong A, Schober W, Rodgers JR. Anergy and apoptosis in CD8ϩ T cells from HIV-infected persons. J Immunol 1994; 153: 412–20. 33. Schirmer M, Goldberger C, Wurzner R, et al. Circulating cytotoxic CD8(ϩ) CD28(–) T cells in ankylosing spond- ylitis. Arthritis Res 2002; 4: 71–6. 34. Foss FM. Immunologic mechanisms of antitumor activity. Semin Oncol 2002; 29 (3 Suppl 7): 5–11. 35. Jonasson L, Tompa A, Wikby A. Expansion of peripheral CD8ϩ T cells in patients with coronary artery disease: relation to cytomegalovirus infection. J Intern Med 2003; 254: 472–8. 36. Pilch H, Hoehn H, Schmidt M, et al. CD8ϩ CD45RAϩCD27–CD28-T-cell subset in PBL of cervical cancer patients representing CD8ϩT-cells being able to recognize cervical cancer associated antigens provided by HPV 16 E7. Zentralbl Gynakol 2002; 124: 406–12. 37. Tsukishiro T, Donnenberg AD, Whiteside TL. Rapid turn- over of the CD8(ϩ)CD28(–) T-cell subset of effector cells in the circulation of patients with head and neck cancer. Cancer Immunol Immunother 2003; 52: 599–607. 38. Monteiro J, Batliwalla F, Ostrer H, Gregersen PK. Shortened telomeres in clonally expanded CD28–CD8ϩ T cells imply a replicative history that is distinct from their CD28ϩCD8ϩ counterparts. J Immunol 1996; 156: 3587–90. 39. Posnett DN, Edinger JW, Manavalan JS, Irwin C, Marodon G. Differentiation of human CD8 T cells: impli- cations for in vivo persistence of CD8ϩ CD28– cytotoxic effector clones. Int Immunol 1999; 11: 229–41. 40. Pawelec G, Akbar A, Caruso C, Effros RB, Grubeck- Loebenstein B, Wikby A. Is immunosenescence infec- tious? Trends Immunol 2004; 25: 406–10. 41. Wikby A, Johansson B, Olsson J, Lofgren S, Nilsson BO, Ferguson F. Expansions of peripheral blood CD8 T-lymphocyte subpopulations and an association with cytomegalovirus seropositivity in the elderly: the Swedish NONA immune study. Exp Gerontol 2002; 37: 445–53. 42. Dagarag MD, Evazyan T, Rao N, Effros RB. Genetic manipulation of telomerase in HIV-specifi c CD8ϩ T cells: enhanced anti-viral functions accompany the increased proliferative potential and telomere length stabilization. J Immunol 2004; 173: 6303–11. 43. Lieberman J, Shankar P, Manjunath N, Andersson J. Dressed to kill? A review of why antiviral CD8 T lym- phocytes fail to prevent progressive immunodefi ciency in HIV-1 infection. Blood 2001; 98: 1667–77. 44. Appay V, Rowland-Jones S. Premature ageing of the immune system: the cause of AIDS? Trends Immunol 2002; 23: 580–5. 45. Migueles SA, Laborico AC, Shupert WL, et al. HIV- specifi c CD8(ϩ) T cell proliferation is coupled to per- forin expression and is maintained in nonprogressors. Nat Immunol 2002; 3: 1061–8. 46. Riviaere Y, McChesney MB, Porrot F, et al. Gag-specifi c cytotoxic responses to HIV type 1 are associated with a decreased risk of progression to AIDS-related complex or AIDS. AIDS Res Hum Retroviruses 1995; 11: 903–7. 47. Rinaldo CJ, Beltz LA, Huang XL, Gupta P, Fan Z, Torpey DJ. Anti-HIV type 1 cytotoxic T lymphocyte effector activity and disease progression in the fi rst 8 years of HIV type 1 infection of homosexual men. AIDS Res Hum Retroviruses 1995; 11: 481–9. 48. Posnett DN, Sinha R, Kabak S, Russo C. Clonal popu- lations of T cells in normal elderly humans: the T cell equivalent to “benign monoclonal gammopathy”. J Exp Med 1994; 179 : 609–18. 49. Morley JK, Batliwalla FM, Hingorani R, Gregersen PK. Oligoclonal CD8ϩ T cells are preferentially expanded in the CD57ϩ subset. J Immunol 1995; 154: 6182–90. 50. Weekes MP, Wills MR, Mynard K, Hicks R, Sissons JG, Carmichael AJ. Large clonal expansions of human virus- specifi c memory cytotoxic T lymphocytes within the CD57ϩ CD28– CD8ϩ T-cell population. Immunology 1999; 98: 443–9. 51. Manfras BJ, Weidenbach H, Beckh KH, et al. Oligoclonal CD8ϩ T-cell expansion in patients with chronic hepa- titis C is associated with liver pathology and poor Replicative senescence, aging, and cancer 93 response to interferon-alpha therapy. J Clin Immunol 2004; 24: 258–71. 52. Scheuring UJ, Sabzevari H, Theofi lopoulos AN. Proliferative arrest and cell cycle regulation in CD8(ϩ)CD28(–) versus CD8(ϩ)CD28(ϩ) T cells. Hum Immunol 2002; 63: 1000–9. 53. McFarland EJ, Harding PA, Striebich CC, MaWhinney S, Kuritzkes DR, Kotzin BL. Clonal CD8ϩ T cell expan- sions in peripheral blood from human immunodefi - ciency virus type 1-infected children. J Infect Dis 2002; 186: 477–85. 54. Khan N, Shariff N, Cobbold M, et al. Cytomegalovirus seropositivity drives the CD8 T cell repertoire toward greater clonality in healthy elderly individuals. J Immunol 2002; 169: 1984–92. 55. Ouyang Q, Wagner WM, Wikby A, et al. Large numbers of dysfunctional CD8ϩ T lymphocytes bearing recep- tors for a single dominant CMV epitope in the very old. J Clin Immunol 2003; 23: 247–57. 56. Ferguson FG, Wikby A, Maxson P, Olsson J, Johansson B. Immune parameters in a longitudinal study of a very old population of Swedish people: a comparison between survivors and nonsurvivors. J Gerontol A Biol Sci Med Sci 1995; 50: B378–B382. 57. Cawthon RM, Smith KR, O’Brien E, Sivatchenko A, Kerber RA. Association between telomere length in blood and mortality in people aged 60 years or older. Lancet 2003; 361: 393–5. 58. Aviv A. Telomeres and human aging: facts and fi bs. Sci Aging Knowledge Environ 2004; 2004 (51): pe43. 59. Aviv A, Shay J, Christensen K, Wright W. The longevity gender gap: are telomeres the explanation? Sci Aging Knowledge Environ 2005; 2005 (23): pe16. 60. Unryn BM, Cook LS, Riabowol KT. Paternal age is posi- tively linked to telomere length of children. Aging Cell 2005; 4: 97–101. 61. Slagboom PE, Droog S, Boomsma DI. Genetic determi- nation of telomere size in humans: a twin study of three age groups. Am J Hum Genet 1994; 55: 876–82. 62. Suciu-Foca N, Manavalan JS, Scotto L, et al. Molecular characterization of allospecifi c T suppressor and toler- ogenic dendritic cells: review. Int Immunopharmacol 2005; 5: 7–11. 63. Kurokohchi K, Masaki T, Arima K, et al. CD28-negative CD8-positive cytotoxic T lymphocytes mediate hepa- tocellular damage in hepatitis C virus infection. J Clin Immunol 2003; 23: 518–27. 64. Sadat-Sowti B, Parrot A, Quint L, Mayaud C, Debre P, Autran B. Alveolar CD8ϩCD57ϩ lymphocytes in human immunodefi ciency virus infection produce an inhibi- tor of cytotoxic functions. Am J Respir Crit Care Med 1994; 149: 972–80. 65. Pietschmann P, Grisar J, Thien R, et al. Immune pheno- type and intracellular cytokine production of periph- eral blood mononuclear cells from postmenopausal patients with osteoporotic fractures. Exp Gerontol 2001; 36: 1749–59. 66. Arron JR, Choi Y. Bone versus immune system. Nature 2000; 408: 535–6. 67. Lanier LL. A renaissance for the tumor immunosurveil- lance hypothesis. Nat Med 2001; 7: 1178–80. 68. Israel BF, Kenney SC. Virally targeted therapies for EBV- associated malignancies. Oncogene 2003; 22: 5122–30. 69. Pardoll D. T cells and tumours. Nature 2001; 411: 1010–12. 70. Petersen JL, Morris CR, Solheim JC. Virus evasion of MHC class I molecule presentation. J Immunol 2003; 171: 4473–8. 71. Zhang D, Shankar P, Xu Z, et al. Most antiviral CD8 T cells during chronic viral infection do not express high levels of perforin and are not directly cytotoxic. Blood 2003; 101: 226–35. 72. Zanussi S, Vaccher E, Caffau C, et al. Interferon-gamma secretion and perforin expression are impaired in CD8ϩ T lymphocytes from patients with undiffer- entiated carcinoma of nasopharyngeal type. Cancer Immunol Immunother 2003; 52: 28–32. 73. Chua D, Huang J, Zheng B, et al. Adoptive transfer of autologous Epstein–Barr virus-specifi c cytotoxic T cells for nasopharyngeal carcinoma. Int J Cancer 2001; 94: 73–80. 74. Mori A, Takao S, Pradutkanchana J, Kietthubthew S, Mitarnun W, Ishida T. High tumor necrosis factor-alpha levels in the patients with Epstein–Barr virus-associated peripheral T-cell proliferative disease/lymphoma. Leuk Res 2003; 27: 493–8. 75. Kennedy-Smith AG, McKenzie JL, Owen MC, Davidson PJ, Vuckovic S, Hart DN. Prostate specifi c antigen inhibits immune responses in vitro: a potential role in prostate cancer. J Urol 2002; 168: 741–7. 76. Chakraborty NG, Stevens RL, Mehrotra S, et al. Recognition of PSA-derived peptide antigens by T cells from prostate cancer patients without any prior stimula- tion. Cancer Immunol Immunother 2003; 52: 497–505. 77. Hakansson A, Hakansson L, Gustafsson B, et al. Biochemotherapy of metastatic malignant melanoma: on down-regulation of CD28. Cancer Immunol Immunother 2002; 51: 499–504. 94 Rita B. Effros 78. Characiejus D, Pasukoniene V, Kazlauskaite N, et al. Predictive value of CD8highCD57ϩ lymphocyte subset in interferon therapy of patients with renal cell carci- noma. Anticancer Res 2002; 22: 3679–83. 79. Carlens S, Liu D, Ringden O, et al. Cytolytic T cell reactivity to Epstein–Barr virus is lost during in vitro T cell expansion. J Hematother Stem Cell Res 2002; 11: 669–74. 80. Straathof KC, Bollard CM, Rooney CM, Heslop HE. Immunotherapy for Epstein–Barr virus-associated cancers in children. Oncologist 2003; 8: 83–98. 81. Dagarag MD, Ng H, Lubong R, Effros RB, Yang OO. Differential impairment of lytic and cytokine functions in senescent HIV-1-specifi c cytotoxic T lymphocytes. J Virol 2003; 77: 3077–83. 82. Hooijberg E, Ruizendaal JJ, Snijders PJ, Kueter EW, Walboomers JM, Spits H. Immortalization of human CD8ϩ T cell clones by ectopic expression of telomerase reverse transcriptase. J Immunol 2000; 165: 4239–45. 83. Mitchell MS. Cancer vaccines, a critical review: part I. Curr Opin Investig Drugs 2002; 3: 140–9. 84. Gitlitz BJ, Belldegrun AS, Figlin RA. Vaccine and gene therapy of renal cell carcinoma. Semin Urol Oncol 2001; 19: 141–7. 85. Townsend SE, Allison JP. Tumor rejection after direct costimulation of CD28ϩ T cells by B7-transfected melanoma cells. Science 1993; 259: 368–70. 86. Dagarag MD, Effros RB. Ectopic telomerase expression prevents functional and phenotypic alterations associ- ated with replicative senescence in virus-specifi c CD8 T cells. Clin Immunol (FOCIS meeting abstract book). 2003. 87. Cutolo M, Sulli A, Seriolo B, Accardo S, Masi AT. Estrogens, the immune response, and autoimmunity. Clin Exp Rheumatol 1995; 13: 217–26. 88. Fox HS, Bond BL, Parslow TG. Estrogen regulates the IFN-gamma promoter. J Immunol 1991; 146: 4362–7. 89. Pittet MJ, Zippelius A, Speiser DE, et al. Ex vivo IFN- gamma secretion by circulating CD8 T lymphocytes: implications of a novel approach for T cell monitoring in infectious and malignant diseases. J Immunol 2001; 166: 7634–40. 90. Xu D, Erickson S, Szeps M, et al. Interferon alpha down-regulates telomerase reverse transcriptase and telomerase activity in human malignant and nonma- lignant hematopoietic cells. Blood 2000; 96: 4313–18. 91. Kyo S, Takakura M, Kanaya T, et al. Estrogen activates telomerase. Cancer Res 1999; 59: 5917–21. 92. Fauce S, Jamieson BD, Chin A, et al. Telomerase activa- tors increase HIV-specifi c CD8ϩ T cell function: a novel approach to prevent or delay immune exhaustion and progression to AIDS. In Cold Spring Harbor Symposium on Telomeres and Telomerase 2005, 197. Introduction This chapter explores the association of aging and qualitative abnormalities of hematopoiesis. While incidence and prevalence of benign and malignant hematologic conditions increase with age, it is not clear whether quantitative or qualitative abnormali- ties of hematopoiesis underlie these changes. A defi - nition of the mechanisms by which older individuals are more vulnerable to hematologic diseases is nec- essary for their prevention and treatment. A common example of a hematologic abnormality in the elderly is unexplained anemia [1–8]. Contro- versy lingers over whether hematopoietic exhaustion, erythropoietic abnormalities due to genomic dam- age, increased vulnerability to environmental stress, or a combination of these factors may lead to ane- mia. Likewise, changes in lymphocytic phenotype, a decline in immune function (immunosenescence), and reduced chemotaxis and bactericidal capacity of neutrophils have been reported in older individuals [9–15]. Despite these changes, hematopoiesis appears adequate to maintain the homeostasis of the periph- eral blood elements both in healthy elderly persons and in aging experimental animals, in the absence of hematopoietic stress [11–19]. Aging may be considered a condition of enhanced vulnerability to stress due to loss in functional reserve of multiple organ systems and simultaneous decline in personal and social resources [20–23]. Both envi- ronmental and genomic changes may conspire to restrict the functional reserve of the aged. Of the envi- ronmental changes the best defi ned is a condition of 8 Qualitative changes of hematopoiesis France Laurencet progressive infl ammation, associated with increased concentration of pro-infl ammatory cytokines in the circulation, that may hamper immune response, alter the ratio of various subpopulations of B and T cells in the circulation, promote apoptosis of hematopoi- etic progenitors, and reduce their responsiveness to growth factors [9,24,25]. At the same time, genomic alteration of these progenitors may prevent their dif- ferentiation and also reduce their responsiveness to growth factors [23,26]. Environmental and genomic alterations appear to converge in the pathogenesis of myelodysplastic syndromes (MDS), a group of common hematologic malignancies after age 60, characterized by clonal hematopoiesis and dysmorphic changes in the bone marrow (Fig. 8.1) and peripheral blood (Fig. 8.2), due to increased proliferation, increased apoptosis, and reduced maturation (Fig. 8.3) [27]. In the following discussion we will explore the mechanisms of MDS as the best-established manifestation of qualita- tively defi cient hematopoiesis in the elderly. Incidence of qualitative hematopoietic abnormalities Most industrialized countries have experienced an increase in the elderly population due to more pro- longed life expectancy and reduced birth rates [28]. In 2000, more than 20% of the people were older than 60 years, and this percentage is projected to increase in the foreseeable future [29,30]. Accumulation of oxidative damage to various organs and tissues is 95 8 Qualitative changes of hematopoiesis France Laurencet Blood Disorders in the Elderly, ed. Lodovico Balducci, William Ershler, Giovanni de Gaetano. Published by Cambridge University Press. © Cambridge University Press 2008. Figure 8.1 Aspirate smear showing hyperplastic and dysplastic features. See color plate section. Figure 8.2 Blood smear from a patient with myelodysplastic syndrome, showing an abnormal monocyte. See color plate section. Figure 8.3 (a and b) Aspirate smear: myelodysplastic dyserythropoiesis and mitosis. See color plate section. (a) (b) 96 Qualitative changes of hematopoiesis 97 partly responsible for age-related molecular changes [31,32]. In the hematopoietic system this damage may be manifested in minor dysplastic changes frequently observed in the bone marrow (BM) of elderly patients, the signifi cance of which is still not understood. In particular it is not clear whether these changes signal the development of MDS or are benign and self-lim- iting [33–35]. The incidence of hematopoietic neo- plasia [36–40] increases with age, but the incidence of MDS is diffi cult to defi ne precisely, because of the heterogeneity of the disease. The more benign sub- types might be under-diagnosed and under-reported [39–41]. MDSs are rarely seen before the age of 50, and the median age is about 70 years [42,43]. The overall disease incidence is about 3–4 per 100 000, but this may rise to 20–30 per 100 000 in the over-70s and up to 89 per 100 000 in the over-80s [44,45]. In Germany the incidence of MDS appears to have increased in recent decades, though this fi nding is con- troversial [46–49]. In a French analysis of 100 patients in a geriatric hospital with a median age of 86 years, the prevalence of macrocytosis was 21% [50,51]. Some of these cases may certainly be ascribed to B 12 and folate defi ciency, or to drugs interfering with nuclear metab- olisms. It is not farfetched to assume that at least in part these cases of macrocytosis represent early MDS or another form of qualitative hematopoietic defect. In conclusion, the incidence and prevalence of MDS, one example of a qualitative defect of hemato poiesis, increases with age. This fi nding suggests that the gen- eral prevalence of qualitative defects of hematopoiesis also increases with age. What is not yet clear is whether all forms of qualitative defects, and particularly mac- rocytosis, may end up as MDS. Risk factors The association of age and hematopoietic neopla- sias may be accounted for by both constitutional and environmental factors [36,37,52]. The occurrence of qualitative abnormalities of hematopoiesis may represent a likely step toward neoplasia [27,39,40]. Risk factors for MDS are shown in Table 8.1. Genetic factors Many karyotypic abnormalities, inherited mutations, polymorphism of several genes, and chromosomal instabilities are associated with disturbance of nor- mal hematopoiesis. Both gender and ethnic origin are important risk factors [53,54]. Of special interest in understanding the mechanism of hematopoietic abnormalities are the reports of familial MDS [55–61]. Inherited abnormalities as well as chromosomal instability may provide the initial genetic hit that predisposes to other hematopoietic abnormalities, including neoplasia [62,63]. Aging itself is associated Table 8.1. Risk factors for MDS. Age Environmental factors Drugs INH, anti-tuberculosis drugs, chloramphenicol, Cycloserine, penicillamine, immunosuppressors Toxins, drinking water Ethanol, zinc, arsenic, cadmium, chloroform, halomethanes Nutritional (malnutrition) Copper and pyridoxine defi ciency; phenols and hydroquinone Occupational Asbestos, paint products, benzene and organic solvents, ammonia, diesel fuel, or other petrochemicals Chronic exposure Pesticides, tobacco, agricultural chemicals, free radicals Infections Viruses Genetic factors Inherited mutations, karyotypic abnormalities, gene mutations, chromosomal instabilities, gender Immunological factors Lymphocytes and cytokines dysregulation Others Depression, obesity and endocrine status [...]... may be involved in the progression of MDS The 5q-minus syndrome consists of refractory anemia (RA), thrombocytosis, and abnormal megakaryocyte morphology with a relatively good prognosis [ 234 – 237 ] The deleted region of chromosome 5 contains the genes for several hematopoietic growth factors and is of particular interest regarding the maturation defect in MDS IL -3 , IL-4, IL-5, IL-9, GMCSF, the M-CSF receptor,... reduces the stress-coping ability of aged individuals In this chapter we explore the in uence of aging on response to hematopoietic stress and the potential mechanisms by which this response may be impaired The peripheral blood counts do not appear significantly reduced in the aged [1 3] , at least up to age 90, indicating that homeostasis is preserved even in the oldest old, in the absence of stress In the. .. (CFU-E) were less responsive to erythropoietin (EPO) It was then established in the mouse model that basal hematopoiesis was not altered in aging, but that the reserve capacity was compromised when mice were submitted to stress [10,11,91] During the last two decades there has been much progress in understanding the role of the immune system, pro -in ammatory cytokines [ 13] , gene interactions [92], point... progenitors in the circulation by stimulating their proliferation Chatta et al [39 ] studied the granulocyte reserve of younger ( 30 ) and older (у 70) individuals following hydrocortisone, epinephrine, and filgrastim, and found that the increment of neutrophils following epinephrine and filgrastim was unaffected by age, while the neutrophilic response to hydrocortisone was blunted in the elderly They concluded... Heijmans-Antonissen C, et al Reduced hematopoietic reserves in DNA interstrand 233 234 235 236 237 238 239 240 241 242 2 43 244 245 crosslink repair-deficient Ercc 1-/ - mice EMBO J 2005; 24: 861–71 Ueda M, Ota J, Yamashita Y, et al DNA microarray analysis of stage progression mechanism in myelodysplastic syndrome Br J Haematol 20 03; 1 23: 288–96 Van den Berghe H, Cassiman JJ, David G, Fryns JP Distinct haematological... unable to maintain a self-renewal capacity TGF-β activity, which inhibits proliferation, declines [181] Dendritic cells (DC) are the most powerful antigen-presenting cells able to activate T cells and partially regulate the adaptive and innate immune system Age does not seem to affect the function of DC in healthy older individuals [ 130 ], but it is compromised in the frail elderly [182] Another cause... and G-CSF [258] Thus, growth-promoting cytokines such as EPO, IL -3 , IL-6, and TPO, which are usually increased in MDS patients, may try to counterbalance the pro-apototic stimuli The role of the immune system One mechanism underlying the marrow failure in MDS is immunologic attack on the HSCs, which may also be found in aplastic anemia and paroxysmal nocturnal hemoglobinuria (PNH) T lymphocytes may inhibit... Following bleeding for blood donation the reticulocyte response was lower in older than in younger individuals, suggesting that the reconstitution of the red blood cell mass may take longer in the elderly [47,48] The mechanism for this reduced erythropoietic response to bleeding is disputed Some authors found that the production of erythropoietin may be blunted with aging [47,48,62], while others found... disease Clin Lab Haematol 2001; 23: 297 30 0 34 Malaguarnera M, Di Fazio I, Vinci E, et al Haematologic pattern in healthy subjects Panminerva Med 1999; 41: 227 31 35 Fernandez-Ferrero S, Ramos F Dyshaemopoietic bone marrow features in healthy subjects are related to age Leuk Res 2001; 25: 187–9 36 Nagura E, Minami S, Nagara K, et al Acute myeloid leukemia in the elderly: 159 Nagoya case studies 37 38 39 40... Am J Med Sci 2000; 31 9: 34 3–52 Mahmoud MY, Lugon M, Anderson CC Unexplained macrocytosis in elderly patients Age Ageing 1996; 25: 31 0–12 Gilleece MH, Dexter TM The biological ageing in bone marrow Rev Clin Gerontol 19 93; 3: 31 7–25 Sharp A, Zipori D, Toledo J, Tal S, Resnitzky P , Globerson A Age-related changes in hemopoietic capacity of bone marrow Mech Ageing Dev 1989; 48: 91–9 Pinto A, De Filippi . [10,11,91]. During the last two dec- ades there has been much progress in understanding the role of the immune system, pro -in ammatory cytokines [ 13] , gene interactions [92], point muta- tions,. [227– 231 ]. Spontaneous chro- mosomal instability and interstrand crosslink dam- age may contribute to the functional decline of the hematopoietic system associated with aging [ 232 , 233 ]. There. in older individuals [9–15]. Despite these changes, hematopoiesis appears adequate to maintain the homeostasis of the periph- eral blood elements both in healthy elderly persons and in aging

Ngày đăng: 10/08/2014, 16:22

Từ khóa liên quan

Tài liệu cùng người dùng

Tài liệu liên quan