51
|
Modelling naive T-cell homeostasis: consequences of heritable cellular lifespan during ageing. Immunol Cell Biol 2009; 87:445-56. [PMID: 19290017 DOI: 10.1038/icb.2009.11] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Within an individual, the population of mature naive T cells is maintained throughout life by both input from the thymus and homeostatic proliferation in the periphery. Here, we develop a mathematical model of this process of naive T-cell homeostasis, and use it to explore questions of lifespan, inheritance and receptor repertoire during ageing. By assuming lifespan is largely determined by a heritable trait reset on mitosis, we show that homeostatic proliferation leads naturally to a longer lived population with age. A plausible candidate for the heritable trait influencing lifespan is T-cell receptor affinity for major histocompatibility molecules loaded with self-peptides. Concurrently with increasing lifespan, receptor diversity decreases with age, thus quantitatively linking these two phenomena. These results depend on the thymus involuting with age so that homeostatic proliferation becomes the dominant mode of replacement of the naive T-cell repertoire.
Collapse
|
52
|
Aw D, Silva AB, Palmer DB. Is thymocyte development functional in the aged? Aging (Albany NY) 2009; 1:146-53. [PMID: 20157506 PMCID: PMC2806005 DOI: 10.18632/aging.100027] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2009] [Accepted: 02/10/2009] [Indexed: 01/09/2023]
Abstract
T cells are an
integral part of a functional immune system with the majority being produced
in the thymus. Of all the changes related to immunosenescence, regression of the thymus is
considered one of the most universally recognised alterations. Despite the reduction of
thymic size, there is evidence to suggest that T cell output is still
present into old age, albeit much diminished; leading to the assumption
that thymocyte development is normal. However, current data suggests that
recent thymic emigrant from the aged thymus are functionally less
responsive, giving rise to the possibility that the generation of
naïve T cell may be intrinsically impaired in the elderly. In light of
these findings we discuss the evidence that suggest aged T cells may be
flawed even before exiting to the periphery and could contribute to the
age-associated decline in immune function.
Collapse
Affiliation(s)
- Danielle Aw
- Infection & Immunity and Genes & Development Group, Department of Veterinary Basic Sciences, Royal Veterinary College, UK
| | | | | |
Collapse
|
53
|
Mef2C is a lineage-restricted target of Scl/Tal1 and regulates megakaryopoiesis and B-cell homeostasis. Blood 2009; 113:3461-71. [PMID: 19211936 DOI: 10.1182/blood-2008-07-167577] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The basic helix-loop-helix transcription factor stem cell leukemia gene (Scl) is a master regulator for hematopoiesis essential for hematopoietic specification and proper differentiation of the erythroid and megakaryocyte lineages. However, the critical downstream targets of Scl remain undefined. Here, we identified a novel Scl target gene, transcription factor myocyte enhancer factor 2 C (Mef2C) from Scl(fl/fl) fetal liver progenitor cell lines. Analysis of Mef2C(-/-) embryos showed that Mef2C, in contrast to Scl, is not essential for specification into primitive or definitive hematopoietic lineages. However, adult VavCre(+)Mef2C(fl/fl) mice exhibited platelet defects similar to those observed in Scl-deficient mice. The platelet counts were reduced, whereas platelet size was increased and the platelet shape and granularity were altered. Furthermore, megakaryopoiesis was severely impaired in vitro. Chromatin immunoprecipitation microarray hybridization analysis revealed that Mef2C is directly regulated by Scl in megakaryocytic cells, but not in erythroid cells. In addition, an Scl-independent requirement for Mef2C in B-lymphoid homeostasis was observed in Mef2C-deficient mice, characterized as severe age-dependent reduction of specific B-cell progenitor populations reminiscent of premature aging. In summary, this work identifies Mef2C as an integral member of hematopoietic transcription factors with distinct upstream regulatory mechanisms and functional requirements in megakaryocyte and B-lymphoid lineages.
Collapse
|
54
|
Leaky severe combined immunodeficiency and aberrant DNA rearrangements due to a hypomorphic RAG1 mutation. Blood 2009; 113:2965-75. [PMID: 19126872 DOI: 10.1182/blood-2008-07-165167] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The RAG1/2 endonuclease initiates programmed DNA rearrangements in progenitor lymphocytes by generating double-strand breaks at specific recombination signal sequences. This process, known as V(D)J recombination, assembles the vastly diverse antigen receptor genes from numerous V, D, and J coding segments. In vitro biochemical and cellular transfection studies suggest that RAG1/2 may also play postcleavage roles by forming complexes with the recombining ends to facilitate DNA end processing and ligation. In the current study, we examine the in vivo consequences of a mutant form of RAG1, RAG1-S723C, that is proficient for DNA cleavage, yet exhibits defects in postcleavage complex formation and end joining in vitro. We generated a knockin mouse model harboring the RAG1-S723C hypomorphic mutation and examined the immune system in this fully in vivo setting. RAG1-S723C homozygous mice exhibit impaired lymphocyte development and decreased V(D)J rearrangements. Distinct from RAG nullizygosity, the RAG1-S723C hypomorph results in aberrant DNA double-strand breaks within rearranging loci. RAG1-S723C also predisposes to thymic lymphomas associated with chromosomal translocations in a p53 mutant background, and heterozygosity for the mutant allele accelerates age-associated immune system dysfunction. Thus, our study provides in vivo evidence that implicates aberrant RAG1/2 activity in lymphoid tumor development and premature immunosenescence.
Collapse
|
55
|
Larbi A, Franceschi C, Mazzatti D, Solana R, Wikby A, Pawelec G. Aging of the immune system as a prognostic factor for human longevity. Physiology (Bethesda) 2008; 23:64-74. [PMID: 18400689 DOI: 10.1152/physiol.00040.2007] [Citation(s) in RCA: 200] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Accumulating data are documenting an inverse relationship between immune status, response to vaccination, health, and longevity, suggesting that the immune system becomes less effective with advancing age and that this is clinically relevant. The mechanisms and consequences of age-associated immune alterations, designated immunosenescence, are briefly reviewed here.
Collapse
Affiliation(s)
- Anis Larbi
- University of Tübingen, Center for Medical Research,Tübingen, Germany.
| | | | | | | | | | | |
Collapse
|
56
|
Age-related differences in expression of signal differentiation factors for human thymic epithelial cells. Bull Exp Biol Med 2008; 144:235-7. [PMID: 18399289 DOI: 10.1007/s10517-007-0298-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Expression of transcription proteins PAX1, Hoxa3, and TLP regulating differentiation of thymic epithelial cells is detected in human thymus starting from gestation week 22 until the age of 95 years. Expression of transcription factors significantly decreased during aging. Apart from the decrease in the expression of signal differentiation factors in cultured thymic epithelial cells, proliferative activity of T lymphocytes cocultured with thymic epithelial cells also decreased in aging cultures, which demonstrated the important regulatory effect of transcription proteins on maturation and maintenance of T lymphocytes. Taking into account the important role of transcription proteins in the regulation of proliferation and function of T lymphocytes, whose number sharply decreases during aging, the maintenance of the level of expression of transcription factors during aging is a promising trend in modern biogerontology.
Collapse
|
57
|
Spyridopoulos I, Erben Y, Brummendorf TH, Haendeler J, Dietz K, Seeger F, Kissel CK, Martin H, Hoffmann J, Assmus B, Zeiher AM, Dimmeler S. Telomere Gap Between Granulocytes and Lymphocytes Is a Determinant for Hematopoetic Progenitor Cell Impairment in Patients With Previous Myocardial Infarction. Arterioscler Thromb Vasc Biol 2008; 28:968-74. [DOI: 10.1161/atvbaha.107.160846] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Objective—
We have previously demonstrated that ischemic cardiomyopathy is associated with selective impairment of progenitor cell function in the bone marrow and in the peripheral blood, which may contribute to an unfavorable left ventricular remodeling process.
Methods and Results—
With this study, we intended to identify the influence of telomere length on bone marrow functionality in 50 patients with coronary artery disease (CAD) and previous myocardial infarction. Mean telomere length (mTL) was measured simultaneously in peripheral blood leukocytes and mononuclear bone marrow cells (BMC), using the flow-FISH method. Telomere erosion already occurred at the bone marrow level, whereby age (39 bp/yr,
P
=0.025) and the number of affected vessels (434 bp/vessel,
P
=0.029) were the only independent predictors. Lymphocytes demonstrated significant TL shortening between BMCs and peripheral blood in CAD patients (−1011±897 bp) as opposed to an increase in a young control group (+235±459 bp,
P
<0.001). SDF- and VEGF-specific migration of BMCs correlated with mTL of lymphocytes (
r
=0.42,
P
<0.001) and was significantly reduced in CAD patients. Finally, the telomere length difference between granulocytes and lymphocytes was the most determinant for telomere-associated bone marrow impairment (
P
<0.001).
Conclusion—
In patients with CAD, telomere shortening of BMCs is dependent on both age and the extent of CAD and correlates with bone marrow cell functionality.
Collapse
Affiliation(s)
- Ioakim Spyridopoulos
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Young Erben
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Tim H. Brummendorf
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Judith Haendeler
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Klaus Dietz
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Florian Seeger
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Christine K. Kissel
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Hans Martin
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Jedrzej Hoffmann
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Birgit Assmus
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Andreas M. Zeiher
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Stefanie Dimmeler
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| |
Collapse
|
58
|
Marinova TT, Kuerten S, Petrov DB, Angelov DN. Thymic epithelial cells of human patients affected by myasthenia gravis overexpress IGF-I immunoreactivity. APMIS 2008; 116:50-8. [PMID: 18254780 DOI: 10.1111/j.1600-0463.2008.00666.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Accumulating evidence shows that several kinds of thymic cells express insulin-like growth factor-I (IGF-I), which is known to play an important role in T cell ontogeny under both physiological and pathological conditions. Still, little is known about the mechanisms of IGF-I involvement in the pathological transformation of the thymocyte microenvironment. The present study focuses on a comparative analysis of the IGF-I immunoreactivity of thymic epithelial cells (EC) from human patients with hyperplasia-associated myasthenia gravis (MG) versus physiological thymic tissue from healthy controls using immunohistochemistry and immunoelectron microscopy. We show that myasthenic EC overexpress IGF-I in comparison to EC from control subjects. The IGF-I immunoreactivity in the medullary and cortical EC from MG patients was stronger than in the normal gland. The increased expression of IGF-I and more frequent distribution of IGF-I and IGF-I-receptor (IGF-IR) immunopositive EC correlated with modulation in the immunoreactivity of double (IGF-I/IGF-IR) positive EC. Our data provide new immunocytochemial evidence for alterations of IGF-I and IGF-IR immunoreactivity in EC from pathological thymi. The persisting expression of IGF-I and IGF-IR most likely indicates that the myasthenic thymus is still capable of governing IGF-I signaling pathways, which are involved in the local regulation of T cell development and plasticity.
Collapse
|
59
|
Laconi E, Doratiotto S, Vineis P. The microenvironments of multistage carcinogenesis. Semin Cancer Biol 2008; 18:322-9. [PMID: 18456510 DOI: 10.1016/j.semcancer.2008.03.019] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2008] [Accepted: 03/25/2008] [Indexed: 12/19/2022]
Abstract
Overt neoplasia is often the result of a chronic disease process encompassing an extended segment of the lifespan of any species. A common pathway in the natural history of the disease is the appearance of focal proliferative lesions that are known to act as precursors for cancer development. It is becoming increasingly apparent that the emergence of such lesions is not a cell-autonomous phenomenon, but is heavily dependent on microenvironmental cues derived from the surrounding tissue. Specific alterations in the tissue microenvironment that can foster the selective growth of focal lesions are discussed herein. Furthermore, we argue that a fundamental property of focal lesions as it relates to their precancerous nature lies in their altered growth pattern as compared to the tissue where they reside. The resulting altered tissue architecture translates into the emergence of a unique tumor microenvironment inside these lesions, associated with altered blood vessels and/or blood supply which in turn can trigger biochemical and metabolic changes fueling tumor progression. A deeper understanding of the role(s) of tissue and tumor microenvironments in the pathogenesis of cancer is essential to design more effective strategies for the management of this disease.
Collapse
Affiliation(s)
- Ezio Laconi
- Dipartimento di Scienze e Tecnologie Biomediche, Sezione di Patologia Sperimentale, Università di Cagliari, 09125 Cagliari, Italy.
| | | | | |
Collapse
|
60
|
Signer RAJ, Montecino-Rodriguez E, Witte ON, McLaughlin J, Dorshkind K. Age-related defects in B lymphopoiesis underlie the myeloid dominance of adult leukemia. Blood 2007; 110:1831-9. [PMID: 17554060 PMCID: PMC1976345 DOI: 10.1182/blood-2007-01-069401] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2007] [Accepted: 06/02/2007] [Indexed: 01/12/2023] Open
Abstract
Reduced lymphopoiesis during aging contributes to declines in immunity, but little consideration has been given to its effect on the development of hematologic disease. This report demonstrates that age-related defects in lymphopoiesis underlie the myeloid dominance of adult leukemia. Using a murine model of chronic myeloid leukemia, an adult-onset malignancy that arises from transformation of hematopoietic stem cells by the BCR-ABL(P210) oncogene, we demonstrate that young bone marrow (BM) cells that were transformed with BCR-ABL(P210) initiated both a myeloproliferative disorder (MPD) and B-lymphoid leukemia, whereas BCR-ABL(P210)-transformed old BM cells recapitulated the human disease by inducing an MPD with rare lymphoid involvement. In addition, the lesser severity of MPDs initiated from old BCR-ABL(P210)-transduced BM cells revealed unappreciated defects in aged myeloid progenitors. These data demonstrate that aging affects patterns of leukemogenesis and indicate that the effects of senescence on hematopoiesis are more extensive than previously appreciated.
Collapse
Affiliation(s)
- Robert A J Signer
- Department of Pathology and Laboratory Medicine and Hematopoietic Malignancies Program, Jonsson Comprehensive Cancer Center, Los Angels, CA 90095, USA
| | | | | | | | | |
Collapse
|
61
|
Ju Z, Jiang H, Jaworski M, Rathinam C, Gompf A, Klein C, Trumpp A, Rudolph KL. Telomere dysfunction induces environmental alterations limiting hematopoietic stem cell function and engraftment. Nat Med 2007; 13:742-7. [PMID: 17486088 DOI: 10.1038/nm1578] [Citation(s) in RCA: 217] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2006] [Accepted: 03/20/2007] [Indexed: 12/15/2022]
Abstract
Cell-intrinsic checkpoints limit the proliferative capacity of primary cells in response to telomere dysfunction. It is not known, however, whether telomere dysfunction contributes to cell-extrinsic alterations that impair stem cell function and organ homeostasis. Here we show that telomere dysfunction provokes defects of the hematopoietic environment that impair B lymphopoiesis but increase myeloid proliferation in aging telomerase knockout (Terc(-/-)) mice. Moreover, the dysfunctional environment limited the engraftment of transplanted wild-type hematopoietic stem cells (HSCs). Dysfunction of the hematopoietic environment was age dependent and correlated with progressive telomere shortening in bone marrow stromal cells. Telomere dysfunction impaired mesenchymal progenitor cell function, reduced the capacity of bone marrow stromal cells to maintain functional HSCs, and increased the expression of various cytokines, including granulocyte colony-stimulating factor (G-CSF), in the plasma of aging mice. Administration of G-CSF to wild-type mice mimicked some of the defects seen in aging Terc(-/-) mice, including impairment of B lymphopoiesis and HSC engraftment. Conversely, inhibition of G-CSF improved HSC engraftment in aged Terc(-/-) mice. Taken together, these results show that telomere dysfunction induces alterations of the environment that can have implications for organismal aging and cell transplantation therapies.
Collapse
Affiliation(s)
- Zhenyu Ju
- Department of Gastroenterology, Hepatology and Endocrinology, Medical School Hannover, 30625 Hannover, Germany
| | | | | | | | | | | | | | | |
Collapse
|
62
|
Abstract
OBJECTIVE The aims of this study were to measure the normal concentration of nerve growth factor (NGF) in healthy human saliva and to investigate the effects of age and gender differences on saliva NGF level. MATERIALS AND METHODS Resting whole, stimulated parotid, and stimulated submandibular/sublingual saliva were collected from 127 healthy volunteers with ages ranging from 20 to 81 years. The saliva NGF concentration was measured by enzyme immunoassay. RESULTS AND CONCLUSIONS The mean concentrations of NGF were 901.4 +/- 75.6 pg ml(-1) in resting whole saliva, 885.9 +/- 79.9 pg ml(-1) in stimulated parotid saliva, and 1066.1 +/- 88.1 pg ml(-1) in stimulated submandibular/sublingual saliva. The stimulated submandibular saliva showed lower NGF concentrations with increasing age (rho = -0.296, P = 0.001). The NGF concentrations of resting whole saliva (P = 0.025) and stimulated parotid saliva (P = 0.005) were significantly higher in women than men. The NGF concentration of stimulated submandibular saliva was significantly higher than stimulated parotid saliva (P = 0.005) and significantly correlated with stimulated parotid saliva NGF level (rho = -0.244, P = 0.008). We found measurable concentrations of NGF in all three sources of saliva; the concentration was affected by the source for the stimulated parotid and submandibular saliva, age for stimulated submandibular saliva, and gender difference for resting whole saliva and stimulated parotid saliva.
Collapse
Affiliation(s)
- J-W Nam
- Department of Oral Medicine and Oral Diagnosis, School of Dentistry and Dental Research Institute, Seoul National University, Yunkeun-Dong 28, Chongro-Ku, Seoul 110-749, Korea
| | | | | | | | | |
Collapse
|
63
|
Zhao L, Sun L, Wang H, Ma H, Liu G, Zhao Y. Changes of CD4+CD25+Foxp3+ regulatory T cells in aged Balb/c mice. J Leukoc Biol 2007; 81:1386-94. [PMID: 17369496 DOI: 10.1189/jlb.0506364] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
A progressive decline in the integrity of the immune system is one of the physiologic changes during aging. The frequency of autoimmune diseases or immune disorders increases in the aging population, but the state of regulatory T (Treg) cells in aged individuals has not been well determined. In the present study, we investigated the levels, phenotypes, and function of CD4(+)CD25(+) Treg cells in Balb/c mice, which were older than 20 months. Significantly enhanced percentages of CD4(+)CD25(+) Treg cells in the periphery (blood, spleen, and lymph nodes) of the aged mice were observed. These Treg cells showed modified Vbeta family distribution, reduced levels of CD45 receptor B and CD62 ligand molecules, as well as normal levels of forkhead box p3. However, when the inhibiting function of Treg cells was assayed in the in vitro assays and in a delayed-type hypersensitivity (DTH) model, CD4(+)CD25(+) Treg cells of aged mice displayed significantly lower inhibiting ability on alloantigen-induced DTH reaction or cytokine productions (IL-2 and IFN-gamma) but not cell proliferation of effector T cells, as compared with CD4(+)CD25(+) Treg cells of young mice. In addition, the percentages of CD4(+)CD8(-)CD25(+) Treg cells in the thymi of aged mice increased significantly, but their total cell numbers decreased markedly in these mice. Our present studies indicated collectively that the percentages, phenotypes, the size of TCR repertoire, and function of CD4(+)CD25(+) Treg cells were altered significantly with aging in mice. The functional defects of CD4(+)CD25(+) Treg cells may shed light on the role of CD4(+)CD25(+) Treg cells in the increased sensitivity to autoimmune diseases of aged populations.
Collapse
Affiliation(s)
- Liang Zhao
- Transplantation Biology Research Division, State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beisihuan Xi Road 25, Beijing, China 100080
| | | | | | | | | | | |
Collapse
|
64
|
Douat-Casassus C, Marchand-Geneste N, Diez E, Gervois N, Jotereau F, Quideau S. Synthetic anticancer vaccine candidates: rational design of antigenic peptide mimetics that activate tumor-specific T-cells. J Med Chem 2007; 50:1598-609. [PMID: 17328535 DOI: 10.1021/jm0613368] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A rational design approach was followed to develop peptidomimetic analogues of a cytotoxic T-cell epitope capable of stimulating T-cell responses as strong as or stronger (heteroclytic) than those of parental antigenic peptides. The work described herein focused on structural alterations of the central amino acids of the melanoma tumor-associated antigenic peptide Melan-A/MART-1(26-35) using nonpeptidic units. A screening was first realized in silico to select altered peptides potentially capable of fitting at the interface between the major histocompatibilty complex (MHC) class-I HLA-A2 molecule and T-cell receptors (TCRs). Two compounds appeared to be high-affinity ligands to the HLA-A2 molecule and stimulated several Melan-A/MART-1 specific T-cell clones. Most remarkably, one of them even managed to amplify the response of one clone. Together, these results indicate that central TCR-contact residues of antigenic peptides can be replaced by nonpeptidic motifs without loss of binding affinity to MHC class-I molecules and T-cell triggering capacity.
Collapse
Affiliation(s)
- Céline Douat-Casassus
- Institut Européen de Chimie et Biologie, 2 rue Robert Escarpit, 33607 Pessac Cedex, France
| | | | | | | | | | | |
Collapse
|
65
|
Abstract
It is now becoming apparent that the immune system undergoes age-associated alterations, which accumulate to produce a progressive deterioration in the ability to respond to infections and to develop immunity after vaccination, both of which are associated with a higher mortality rate in the elderly. Immunosenescence, defined as the changes in the immune system associated with age, has been gathering interest in the scientific and health-care sectors alike. The rise in its recognition is both pertinent and timely given the increasing average age and the corresponding failure to increase healthy life expectancy. This review attempts to highlight the age-dependent defects in the innate and adaptive immune systems. While discussing the mechanisms that contribute to immunosenescence, with emphasis on the extrinsic factors, particular attention will be focused on thymic involution. Finally, we illuminate potential therapies that could be employed to help us live a longer, fuller and healthier life.
Collapse
Affiliation(s)
- Danielle Aw
- Royal Veterinary College, Host Response and Genes and Development Group, Department of Veterinary Basic Sciences, Royal College Street, London, United Kingdom
| | | | | |
Collapse
|
66
|
Signer RAJ, Montecino-Rodriguez E, Dorshkind K. Aging, B lymphopoiesis, and patterns of leukemogenesis. Exp Gerontol 2006; 42:391-5. [PMID: 17184948 PMCID: PMC1924911 DOI: 10.1016/j.exger.2006.11.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2006] [Revised: 11/07/2006] [Accepted: 11/07/2006] [Indexed: 11/30/2022]
Abstract
The production of B lymphocytes begins to decline steadily early in adult life and is severely compromised in the elderly. This occurrence has been attributed to intrinsic defects in early hematopoietic progenitors and B cell precursors as well as to microenvironmental changes in aged bone marrow. The aim of this review is to present an overview of B lymphocyte senescence and its underlying causes and to discuss its impact on immune function and leukemogenesis in aged individuals.
Collapse
Affiliation(s)
- Robert A J Signer
- Department of Pathology and Laboratory Medicine and Hematopoietic Malignancies Program, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|
67
|
Choudhury AR, Ju Z, Djojosubroto MW, Schienke A, Lechel A, Schaetzlein S, Jiang H, Stepczynska A, Wang C, Buer J, Lee HW, von Zglinicki T, Ganser A, Schirmacher P, Nakauchi H, Rudolph KL. Cdkn1a deletion improves stem cell function and lifespan of mice with dysfunctional telomeres without accelerating cancer formation. Nat Genet 2006; 39:99-105. [PMID: 17143283 DOI: 10.1038/ng1937] [Citation(s) in RCA: 340] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2006] [Accepted: 11/03/2006] [Indexed: 12/27/2022]
Abstract
Telomere shortening limits the proliferative lifespan of human cells by activation of DNA damage pathways, including upregulation of the cell cycle inhibitor p21 (encoded by Cdkn1a, also known as Cip1 and Waf1)) (refs. 1-5). Telomere shortening in response to mutation of the gene encoding telomerase is associated with impaired organ maintenance and shortened lifespan in humans and in mice. The in vivo function of p21 in the context of telomere dysfunction is unknown. Here we show that deletion of p21 prolongs the lifespan of telomerase-deficient mice with dysfunctional telomeres. p21 deletion improved hematolymphopoiesis and the maintenance of intestinal epithelia without rescuing telomere function. Moreover, deletion of p21 rescued proliferation of intestinal progenitor cells and improved the repopulation capacity and self-renewal of hematopoietic stem cells from mice with dysfunctional telomeres. In these mice, apoptotic responses remained intact, and p21 deletion did not accelerate chromosomal instability or cancer formation. This study provides experimental evidence that telomere dysfunction induces p21-dependent checkpoints in vivo that can limit longevity at the organismal level.
Collapse
Affiliation(s)
- Aaheli Roy Choudhury
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Sadighi Akha AA, Berger SB, Miller RA. Enhancement of CD8 T-cell function through modifying surface glycoproteins in young and old mice. Immunology 2006; 119:187-94. [PMID: 16805788 PMCID: PMC1782347 DOI: 10.1111/j.1365-2567.2006.02420.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Previous work from our laboratory has shown that modifying cell surface glycosylation with either a Clostridium perfringens-derived sialidase (CP-Siase), or an O-linked glycoprotein endopeptidase (OSGE) can enhance the function of CD4 T cells from both young and old mice at multiple levels. Here we have re-assessed the effect of age on CD8 T-cell function, and examined the outcome of enzymatic treatment with CP-Siase and OSGE on its different aspects. Pre-treatment of CD8 T cells with either CP-Siase or OSGE led to a significant increase in anti-CD3-mediated Ca2+ response in both young and old mice. Pre-treated CD8 T cells from both age groups also displayed a significant increase in activation-induced CD69 and CD25 expression, and produced significantly higher amounts of interleukin-2 and interferon-gamma in comparison to their untreated counterparts. Furthermore, pretreatment with either enzyme enhanced granzyme B expression in CD8 T cells, and increased their cytolytic activity in vitro. These data support the notion that glycosylated surface proteins hinder CD8 T-cell activation and function in both young and old mice, and raise the possibility of significantly improving CD8 T cell function in older individuals through enzymatic alteration of surface glycoproteins.
Collapse
Affiliation(s)
- Amir A Sadighi Akha
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA.
| | | | | |
Collapse
|
69
|
Montecino-Rodriguez E, Dorshkind K. Evolving patterns of lymphopoiesis from embryogenesis through senescence. Immunity 2006; 24:659-662. [PMID: 16782019 DOI: 10.1016/j.immuni.2006.06.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Lymphopoiesis occurs throughout adult life, but the number of lymphocytes produced is substantially reduced in old individuals. This commentary will highlight observations suggesting that common mechanisms underlie declines in both B and T cell production and that reductions in lymphopoiesis are initiated much earlier than has generally been appreciated.
Collapse
Affiliation(s)
- Encarnacion Montecino-Rodriguez
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue, Los Angeles, California 90095
| | - Kenneth Dorshkind
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue, Los Angeles, California 90095.
| |
Collapse
|
70
|
Barten DM, Meredith JE, Zaczek R, Houston JG, Albright CF. Gamma-secretase inhibitors for Alzheimer's disease: balancing efficacy and toxicity. Drugs R D 2006; 7:87-97. [PMID: 16542055 DOI: 10.2165/00126839-200607020-00003] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The amyloid hypothesis, which states that beta-amyloid (Abeta) aggregates cause the onset and progression of Alzheimer's disease (AD), is a leading proposal to explain AD aetiology. Based on this hypothesis, compounds that inhibit gamma-secretase, one of the enzymes responsible for forming Abeta, are potential therapeutics for AD. Preclinical studies clearly establish that gamma-secretase inhibitors can reduce brain Abeta in rodent models. The initial investigation of the effects of a gamma-secretase inhibitor on Abeta-induced cognitive deficits in transgenic mice showed that modest Abeta reductions (15-30%) are sufficient to reverse Abeta-induced cognitive deficits in Tg2576 mice. Extending these studies to other gamma-secretase inhibitors and other models with Abeta-induced cognitive deficits will be important. Unfortunately, gamma-secretase inhibitors also cause abnormalities in the gastrointestinal tract, thymus and spleen in rodents. These changes likely result from inhibition of Notch cleavage, a transmembrane receptor involved in regulating cell-fate decisions. Two recent studies in rodents suggest that Abeta reduction using gamma-secretase inhibitors can be partially separated from Notch inhibition. Given the uncertain Abeta reduction target and the potential for mechanism-based toxicity, biomarkers for efficacy and toxicity would be helpful in clinical trials. The first report of gamma-secretase inhibitors in clinical trials was recently published. In this study, LY-450139 reduced plasma Abeta, but not cerebrospinal fluid Abeta. Taken together, the results of studies to date suggest that gamma-secretase inhibitors have the potential to address a large unmet medical need if the technical challenges can be overcome.
Collapse
Affiliation(s)
- Donna M Barten
- Bristol-Myers Squibb, Pharmaceutical Research Institute, Neuroscience Drug Discovery, Wallingford, Connecticut 06492, USA.
| | | | | | | | | |
Collapse
|
71
|
Sehl M, Sawhney R, Naeim A. Physiologic aspects of aging: impact on cancer management and decision making, part II. Cancer J 2006; 11:461-73. [PMID: 16393480 DOI: 10.1097/00130404-200511000-00005] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In this second article of our two-part review, we focus on age-associated physiologic changes involving the nervous, endocrine, hematologic, immune, and musculoskeletal systems, with close attention to the interconnected nature of these systems. There is a well-known connection between the neuroendocrine and immune systems via the hypothalamic-pituitary-adrenal axis and via interaction by means of cytokines, hormones, and neurotransmitters. These changes may lead to a loss of integration and resiliency with age, thus decreasing the ability of the elderly patient with cancer to adapt to stressful circumstances. Prominent changes include decline in memory and cognition, and increased susceptibility to peripheral neuropathy. Hematologic and immune changes like reduced bone marrow reserve and increased susceptibility to infections have far reaching implications for cancer care in the elderly. Gradual decline in hormone levels, and changes in muscle and body composition, can lead to functional decline and frailty. Use of the clinical interventions suggested in this article, along with an appreciation of the interplay of these age-related physiologic changes and their consequences, allows oncology professionals to customize therapy and minimize side effects in the geriatric oncology patient.
Collapse
Affiliation(s)
- Mary Sehl
- Division of Hematology-Oncology and Geriatrics, David Geffen School of Medicine, University of California, Los Angeles, California 90095-1687, USA
| | | | | |
Collapse
|