51
|
Ongrádi J, Stercz B, Kövesdi V, Vértes L. Immunosenescence and vaccination of the elderly II. New strategies to restore age-related immune impairment. Acta Microbiol Immunol Hung 2009; 56:301-12. [PMID: 20038483 DOI: 10.1556/amicr.56.2009.4.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
One of the greatest health-care challenges in the elderly is to ensure that vaccination against infections are optimally effective, but vaccination can only be effective if cells that are capable of responding are still present in the repertoire. The reversing of immunosenescence could be achieved by improving immune responses or altering vaccine formulation. Recent vaccination strategies in the elderly exert low effectiveness. Nutritional interventions and moderate exercise delay T cell senescence. Telomerase activity and expression of toll-like receptors can be improved by chemotherapy. Reversion of thymic atrophy could be achieved by thymus transplantation, depletion of accumulated dysfunctional naive T cells and herpesvirus-specific exhausted memory cells. Administration of immunostimulatory and anti-inflammatory cytokines show the best practical approach. Reduced dendritic cell activity and co-receptor expression might be increased by interleukin (IL)-2 administration. IL-7 protects both B and T lymphocytes, but IL-2, IL-10, keratinocyte growth factor, thymic stromal lymphopoietin, as well as leptin and growth hormone also have a stimulatory effect on thymopoiesis. In animals, several strategies have been explored to produce more efficacious vaccines including high dose vaccines, DNA vaccines with immunostimulatory patch, virosomal vaccines and vaccines containing new adjuvants. Hopefully, one of these approaches will be translated into human therapy in a short time.
Collapse
Affiliation(s)
- J Ongrádi
- Institute of Public Health, Semmelweis University, Budapest, Hungary.
| | | | | | | |
Collapse
|
52
|
Chen J, Li J, Lim FC, Wu Q, Douek DC, Scott DK, Ravussin E, Hsu HC, Jazwinski SM, Mountz JD. Maintenance of naïve CD8 T cells in nonagenarians by leptin, IGFBP3 and T3. Mech Ageing Dev 2009; 131:29-37. [PMID: 19941883 DOI: 10.1016/j.mad.2009.11.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2009] [Revised: 11/03/2009] [Accepted: 11/19/2009] [Indexed: 10/20/2022]
Abstract
Research into the age-associated decline in the immune system has focused on the factors that contribute to the accumulation of senescent CD8 T cells. Less attention has been paid to the non-immune factors that may maintain the pool of naïve CD8 T cells. Here, we analyzed the status of the naïve CD8 T-cell population in healthy nonagenarians (>or=90-year-old), old (60-79-year-old), and young (20-34-year-old) subjects. Naïve CD8 T cells were defined as CD28(+)CD95(-) as this phenotype showed a strong co-expression of the CD45RA(+), CD45RO(-), and CD127(+) phenotypes. Although there was an age-associated decline in the percentage of CD28(+)CD95(-) CD8 T cells, the healthy nonagenarians maintained a pool of naïve CD28(+)CD95(-) cells that contained T-cell receptor excision circles (TREC)(+) cells. The percentages of naïve CD28(+)CD95(-) CD8 T cells in the nonagenarians correlated with the sera levels of insulin-like growth factor binding protein 3 (IGFBP3) and leptin. Higher levels of triiodothyronine (T3) negatively correlated with the accumulation of TREC(-)CD28(-)CD95(+) CD8 T cells from nonagenarians. These results suggest a model in which IGFBP3, leptin and T3 act as non-immune factors to maintain a larger pool of naïve CD8 T cells in healthy nonagenarians.
Collapse
Affiliation(s)
- Jian Chen
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
53
|
Gruver AL, Ventevogel MS, Sempowski GD. Leptin receptor is expressed in thymus medulla and leptin protects against thymic remodeling during endotoxemia-induced thymus involution. J Endocrinol 2009; 203:75-85. [PMID: 19587263 PMCID: PMC3747557 DOI: 10.1677/joe-09-0179] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Leptin deficiency in mice results in chronic thymic atrophy, suppressed cell-mediated immunity, and decreased numbers of total lymphocytes, suggesting a key role for the metabolic hormone leptin in regulating thymopoiesis and overall immune homeostasis. Unfortunately, the thymus is highly susceptible to stress-induced acute involution. Prolonged thymus atrophy in stress situations can contribute to peripheral T cell deficiency or inhibit immune reconstitution. Little is known, however, about specific roles for leptin signaling in the thymus or the underlying mechanisms driving thymic involution or thymic recovery after acute stress. We report here that leptin receptor expression is restricted in thymus to medullary epithelial cells. Using a model of endotoxemia-induced acute thymic involution and recovery, we have demonstrated a role for supraphysiologic leptin in protection of thymic epithelial cells (TECs). We also present data in support of our hypothesis that leptin treatment decreases in vivo endotoxemia-induced apoptosis of double positive thymocytes and promotes proliferation of double negative thymocytes through a leptin receptor isoform b-specific mechanism. Furthermore, our studies have revealed that leptin treatment increases thymic expression of interleukin-7, an important soluble thymocyte growth factor produced by medullary TECs. Taken together, these studies support an intrathymic role for the metabolic hormone leptin in maintaining healthy thymic epithelium and promoting thymopoiesis, which is revealed when thymus homeostasis is perturbed by endotoxemia.
Collapse
Affiliation(s)
- Amanda L Gruver
- Department of Medicine, Duke University Medical Center, 102 Research Drive, Global Health Research Building (Room 1033), Durham, North Carolina 27710, USA
| | | | | |
Collapse
|
54
|
Leptin contributes to the adaptive responses of mice to high-fat diet intake through suppressing the lipogenic pathway. PLoS One 2009; 4:e6884. [PMID: 19727392 PMCID: PMC2731220 DOI: 10.1371/journal.pone.0006884] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Accepted: 08/05/2009] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Leptin is an adipocyte-derived hormone that plays a critical role in energy homeostasis and lipid metabolism. Overnutrition-associated obesity is known to be accompanied by hyperleptinemia. However, the physiological actions of leptin in the metabolic responses to high-fat diet (HFD) intake remain to be completely elucidated. Here we characterized the metabolic features of mice fed high-fat diets and investigated the impact of leptin upon the lipogenic program which was found to be suppressed by HFD feeding through a proteomics approach. RESULTS When maintained on two types of high-fat diets for up to 16 weeks, mice with a higher fat intake exhibited increased body fat accumulation at a greater pace, developing more severely impaired glucose tolerance. Notably, HFD feeding at 4 weeks elicited the onset of marked hyperleptinemia, prior to the occurrence of apparent insulin resistance and hyperinsulinemia. Proteomic analysis revealed dramatically decreased expression of lipogenic enzymes in the white adipose tissue (WAT) from HFD-fed mice, including ATP-citrate lyase (ACL) and fatty acid synthase (FAS). The expression of ACL and FAS in the liver was similarly suppressed in response to HFD feeding. By contrast, HFD-induced downregulation of hepatic ACL and FAS was significantly attenuated in leptin receptor-deficient db/db mice. Furthermore, in the liver and WAT of wild type animals, intraperitoneal leptin administration was able to directly suppress the expression of these two lipogenic enzymes, accompanied by reduced triglyceride levels both in the liver and serum. CONCLUSIONS These results suggest that leptin contributes to the metabolic responses in adaptation to overnutrition through suppressing the expression of lipogenic enzymes, and that the lipogenic pathway represents a key targeted peripheral component in exerting leptin's liporegulatory actions.
Collapse
|
55
|
Ongrádi J, Stercz B, Kövesdi V, Vértes L. Immunosenescence and vaccination of the elderly, I. Age-related immune impairment. Acta Microbiol Immunol Hung 2009; 56:199-210. [PMID: 19789136 DOI: 10.1556/amicr.56.2009.3.1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The sharp increase of life expectancy and the increasing ratio of ageing population pose new challenges for the public health system. The elderly suffer from more frequent and severe infections than young people. Theoretically, vaccination could protect the elderly against several infectious diseases, but due to their age-related immune impairment, vaccination might fail in many cases. Instead of ineffective vaccination campaigns, exploration and restoration of age-dependent dysregulation of their immune functions have to be placed into the focus of recent research. Frequent comorbidities in these people augment immune defects. Immunosenescence affects both the innate and adaptive immunity. Disturbances in macrophage-derived cytokine release and reduction of the natural killer cell mediated cytotoxicity lead to increased frequency of respiratory, gastrointestinal and skin infections. Although the humoral immunity retains most of its original activity through life span, ageing dampens the ability of B cells to produce antibodies against novel antigens. Age-related declination of the cellular immunity is the consequence of thymic atrophy, reduced output of new T lymphocytes, accumulation of anergic memory cells, deficiencies in the cytokine production and uncertain antigen presentation. Persistent infection by different herpesviruses and other parasites contribute to the loss of immunosurveillance and premature exhaustion of T cells.
Collapse
Affiliation(s)
- J Ongrádi
- Institute of Public Health, Semmelweis University, Budapest Hungary.
| | | | | | | |
Collapse
|
56
|
Diet-induced obesity increases NF-kappaB signaling in reporter mice. GENES AND NUTRITION 2009; 4:215-22. [PMID: 19707810 DOI: 10.1007/s12263-009-0133-6] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Accepted: 03/10/2009] [Indexed: 01/04/2023]
Abstract
The nuclear factor (NF)-kappaB is a primary regulator of inflammatory responses and may be linked to pathology associated with obesity. We investigated the progression of NF-kappaB activity during a 12-week feeding period on a high-fat diet (HFD) or a low-fat diet (LFD) using NF-kappaB luciferase reporter mice. In vivo imaging of luciferase activity showed that NF-kappaB activity was higher in the HFD mice compared with LFD-fed mice. Thorax region of HFD females displayed fourfold higher activity compared with LFD females, while no such increase was evident in males. In male HFD mice, abdominal NF-kappaB activity was increased twofold compared with the LFD males, while females had unchanged NF-kappaB activity in the abdomen by HFD. HFD males, but not females, exhibited evident glucose intolerance during the study. In conclusion, HFD increased NF-kappaB activity in both female and male mice. However, HFD differentially increased activity in males and females. The moderate increase in abdomen of male mice may be linked to glucose intolerance.
Collapse
|
57
|
Borghetti P, Saleri R, Mocchegiani E, Corradi A, Martelli P. Infection, immunity and the neuroendocrine response. Vet Immunol Immunopathol 2009; 130:141-62. [PMID: 19261335 PMCID: PMC7112574 DOI: 10.1016/j.vetimm.2009.01.013] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2008] [Revised: 01/17/2009] [Accepted: 01/27/2009] [Indexed: 12/11/2022]
Abstract
The Central Nervous (CNS) and Immune Systems (IS) are the two major adaptive systems which respond rapidly to numerous challenges that are able to compromise health. The defensive response strictly linking innate to acquired immunity, works continuously to limit pathogen invasion and damage. The efficiency of the innate response is crucial for survival and for an optimum priming of acquired immunity. During infection, the immune response is modulated by an integrated neuro-immune network which potentiates innate immunity, controls potential harmful effects and also addresses metabolic and nutritional modifications supporting immune function. In the last decade much knowledge has been gained on the molecular signals that orchestrate this integrated adaptive response, with focus on the systemic mediators which have a crucial role in driving and controlling an efficient protective response. These mediators are also able to signal alterations and control pathway dysfunctions which may be involved in the persistence and/or overexpression of inflammation that may lead to tissue damage and to a negative metabolic impact, causing retarded growth. This review aims to describe some important signalling pathways which drive bidirectional communication between the Immune and Nervous Systems during infection. Particular emphasis is placed on pro-inflammatory cytokines, immunomodulator hormones such as Glucocorticoids (GCs), Growth hormone (GH), Insulin-like Growth Factor-1 (IGF-1), and Leptin, as well as nutritional factors such as Zinc (Zn). Finally, the review includes up-to-date information on this neuroimmune cross-talk in domestic animals. Data in domestic animal species are still limited, but there are several exciting areas of research, like the potential interaction pathways between mediators (i.e. cytokine-HPA regulation, IL-6-GCS-Zn, cytokines-GH/IGF-1, IL-6-GH-Leptin and thymus activity) that are or could be promising topics of future research in veterinary medicine.
Collapse
|
58
|
Castellheim A, Brekke OL, Espevik T, Harboe M, Mollnes TE. Innate immune responses to danger signals in systemic inflammatory response syndrome and sepsis. Scand J Immunol 2009; 69:479-91. [PMID: 19439008 DOI: 10.1111/j.1365-3083.2009.02255.x] [Citation(s) in RCA: 167] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The systemic immune response induced by non-infectious agents is called systemic inflammatory response syndrome (SIRS) and infection-induced systemic immune response is called sepsis. The host inflammatory response in SIRS and sepsis is similar and may lead to multiple organ dysfunction syndrome (MODS) and ultimately death. The mortality and morbidity in SIRS and sepsis (i.e. critical illness) remain high despite advances in diagnostic and organ supporting possibilities in intensive care units. In critical illness, the acute immune response is organized and executed by innate immunity influenced by the neuroendocrine system. This response starts with sensing of danger by pattern-recognition receptors on the immune competent cells and endothelium. The sensed danger signals, through specific signalling pathways, activate nuclear transcription factor kappaB and other transcription factors and gene regulatory systems which up-regulate the expression of pro-inflammatory mediators. The plasma cascades are also activated which together with the produced pro-inflammatory mediators stimulate further the production of inflammatory biomarkers. The acute inflammatory response underlies the pathophysiological mechanisms involved in the development of MODS. The inflammatory mediators directly affect organ function and cause a decline in remote organ function by mediating the production of nitric oxide leading to mitochondrial anergy and cytopathic hypoxia, a condition of cellular inability to use oxygen. Understanding the mechanisms of acute immune responses in critical illness is necessary for the development of urgently needed therapeutics. The aim of this review is to provide a description of the key components and mechanisms involved in the immune response in SIRS and sepsis.
Collapse
Affiliation(s)
- A Castellheim
- Institute of Immunology, Rikshospitalet University Hospital and University of Oslo, Oslo, Norway.
| | | | | | | | | |
Collapse
|
59
|
Patel K, Taub DD. Role of neuropeptides, hormones, and growth factors in regulating thymopoiesis in middle to old age. F1000 BIOLOGY REPORTS 2009; 1:42. [PMID: 20948643 PMCID: PMC2924688 DOI: 10.3410/b1-42] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The deterioration in adaptive immunity and T-lymphocyte output and the narrowing of the T cell receptor repertoire with age are largely attributable to thymic involution. The loss of thymic function with age may be due to diminished numbers of early thymic progenitors and epithelial cells, and the loss of critical tropic factors within the thymic microenvironment. Here we review some of the recent literature demonstrating a role for neuropeptides, hormones, and growth factors that can influence thymopoiesis associated with stress and aging.
Collapse
Affiliation(s)
- Kalpesh Patel
- Laboratory of Immunology, National Institute of Aging, Intramural Research Program, National Institutes of Health, Biomedical Research Center 251 Bayview Boulevard, Room 8C222, Baltimore, MD 21224 USA
| | | |
Collapse
|
60
|
Three questions about leptin and immunity. Brain Behav Immun 2009; 23:405-10. [PMID: 18996468 PMCID: PMC2699448 DOI: 10.1016/j.bbi.2008.10.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2008] [Revised: 10/14/2008] [Accepted: 10/16/2008] [Indexed: 12/11/2022] Open
Abstract
Leptin is a protein produced by adipocytes (and other cell types) that acts in the brain to regulate appetite and energy expenditure according to the amount of energy stored in adipose tissue. Leptin also exerts a variety of other functions, including important roles as a regulator of immune and inflammatory reactions. The present article is not meant to be a comprehensive review on leptin and immunity, but rather highlights a few controversial issues about leptin's place in the complex network of mediators regulating immune and inflammatory responses. Three issues are discussed: (1) Where am I going, or What is the cellular target of leptin for modulation of immune responses?; (2) Where am I coming from, or Is the cellular source important in determining leptin's effects on immune responses? and (3) What am I doing, or What are leptin's effects on immune and inflammatory responses?
Collapse
|
61
|
Goldberg AC, Goldberg-Eliaschewitz F, Sogayar MC, Genre J, Rizzo LV. Leptin and the Immune Response. Ann N Y Acad Sci 2009; 1153:184-92. [DOI: 10.1111/j.1749-6632.2008.03971.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
62
|
|
63
|
Keratinocyte growth factor enhances DNA plasmid tumor vaccine responses after murine allogeneic bone marrow transplantation. Blood 2008; 113:1574-80. [PMID: 19011222 DOI: 10.1182/blood-2008-05-155697] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Keratinocyte growth factor (KGF), which is given exogenously to allogeneic bone marrow transplantation (allo-BMT) recipients, supports thymic epithelial cells and increases thymic output of naive T cells. Here, we demonstrate that this improved T-cell reconstitution leads to enhanced responses to DNA plasmid tumor vaccination. Tumor-bearing mice treated with KGF and DNA vaccination have improved long-term survival and decreased tumor burden after allo-BMT. When assayed before vaccination, KGF-treated allo-BMT recipients have increased numbers of peripheral T cells, including CD8(+) T cells with vaccine-recognition potential. In response to vaccination, KGF-treated allo-BMT recipients, compared with control subjects, generate increased numbers of tumor-specific CD8(+) cells, as well as increased numbers of CD8(+) cells producing interferon-gamma (IFN-gamma) and tumor necrosis factor-alpha (TNF-alpha). We also found unanticipated benefits to antitumor immunity with the administration of KGF. KGF-treated allo-BMT recipients have an improved ratio of T effector cells to regulatory T cells, a larger fraction of effector cells that display a central memory phenotype, and effector cells that are derived from a broader T-cell-receptor repertoire. In conclusion, our data suggest that KGF can function as a potent vaccine adjuvant after allo-BMT through its effects on posttransplantation T-cell reconstitution.
Collapse
|
64
|
Trotter-Mayo RN, Roberts MR. Leptin acts in the periphery to protect thymocytes from glucocorticoid-mediated apoptosis in the absence of weight loss. Endocrinology 2008; 149:5209-18. [PMID: 18583419 PMCID: PMC2582910 DOI: 10.1210/en.2008-0476] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Leptin is a member of the IL-6 cytokine family and is primarily produced by adipose tissue. At high enough concentration, leptin engages leptin receptors expressed in the hypothalamus that regulate a variety of functions, including induction of weight loss. Mice deficient in leptin (ob/ob) or leptin receptor (db/db) function exhibit thymic atrophy associated with a reduction in double-positive (DP) thymocytes. However, the mediator of such thymic atrophy remains to be identified, and the extent to which leptin acts in the periphery vs. the hypothalamus to promote thymocyte cellularity is unknown. In the present study, we first demonstrate that thymic cellularity and composition is fully restored in ob/ob mice subjected to adrenalectomy. Second, we observe that ob/ob mice treated with low-dose leptin peripherally but not centrally exhibit increased thymocyte cellularity in the absence of any weight loss or significant reduction in systemic corticosterone levels. Third, we demonstrate that reconstitution of db/db mice with wild-type bone marrow augments thymocyte cellularity and restores DP cell frequency despite elevated corticosterone levels. These and additional data support a mode of action whereby leptin acts in the periphery to reduce the sensitivity of DP thymocytes to glucocorticoid-mediated apoptosis in vivo. Strikingly, our data reveal that leptin's actions on thymic cellularity in the periphery can be uncoupled from its anorectic actions in the hypothalamus.
Collapse
Affiliation(s)
- Robert N Trotter-Mayo
- Department of Microbiology, University of Virginia, P.O. Box 801394, Charlottesville, Virginia 22908, USA
| | | |
Collapse
|
65
|
Dixit VD. Adipose-immune interactions during obesity and caloric restriction: reciprocal mechanisms regulating immunity and health span. J Leukoc Biol 2008; 84:882-92. [PMID: 18579754 DOI: 10.1189/jlb.0108028] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Increasing evidence suggests a tight coupling of metabolic and immune systems. This cross-talk mediated by neuroendocrine peptides as well as numerous cytokines and chemokines is believed to be responsible for integrating energy balance to immune function. These neuroendocrine-immune interactions are heightened during the state of chronic positive energy balance, as seen during obesity, and negative energy balance caused by caloric restriction (CR). Emerging evidence suggests that obesity may be associated with an immunodeficient state and chronic inflammation, which contribute to an increased risk of premature death. The direct interactions between expanded leukocyte populations within the adipose tissue during obesity and an increased number of adipocytes within an aging lymphoid microenvironment may constitute an important adaptive or pathological response as a result of change in energy balance. In stark contrast to obesity, CR causes negative energy balance and robustly prolongs a healthy lifespan in all of the species studied to date. Therefore, the endogenous neuroendocrine-metabolic sensors elevated or suppressed as a result of changes in energy balance may offer an important mechanism in understanding the antiaging and potential immune-enhancing nature of CR. Ghrelin, one such sensor of negative energy balance, is reduced during obesity and increased by CR. Ghrelin also regulates immune function by reducing proinflammatory cytokines and promotes thymopoiesis during aging and thus, may be a new CR mimetic target. The identification of immune effects and molecular pathways used by such orexigenic metabolic factors could offer potentially novel approaches to enhance immunity and increase healthy lifespan.
Collapse
Affiliation(s)
- Vishwa Deep Dixit
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA.
| |
Collapse
|
66
|
Gruver AL, Sempowski GD. Cytokines, leptin, and stress-induced thymic atrophy. J Leukoc Biol 2008; 84:915-23. [PMID: 18495786 DOI: 10.1189/jlb.0108025] [Citation(s) in RCA: 167] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Thymopoiesis is essential for development and maintenance of a robust and healthy immune system. Acute thymic atrophy is a complication of many infections, environmental stressors, clinical preparative regimens, and cancer treatments used today. This undesirable sequela can decrease host ability to reconstitute the peripheral T cell repertoire and respond to new antigens. Currently, there are no treatments available to protect against acute thymic atrophy or accelerate recovery, thus leaving the immune system compromised during acute stress events. Several useful murine models are available for mechanistic studies of acute thymic atrophy, including a sepsis model of endotoxin-induced thymic involution. We have identified the IL-6 cytokine gene family members (i.e., leukemia inhibitory factor, IL-6, and oncostatin M) as thymosuppressive agents by the observation that they can acutely involute the thymus when injected into a young, healthy mouse. We have gone on to explore the role of thymosuppressive cytokines and specifically defined a corticosteroid-dependent mechanism of action for the leukemia inhibitory factor in acute thymic atrophy. We also have identified leptin as a novel, thymostimulatory agent that can protect against endotoxin-induced acute thymic atrophy. This review will highlight mechanisms of stress-induced thymic involution and focus on thymosuppressive agents involved in atrophy induction and thymostimulatory agents that may be exploited for therapeutic use.
Collapse
Affiliation(s)
- Amanda L Gruver
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | | |
Collapse
|
67
|
Meadows SK, Dressman HK, Muramoto GG, Himburg H, Salter A, Wei Z, Ginsburg G, Chao NJ, Nevins JR, Chute JP. Gene expression signatures of radiation response are specific, durable and accurate in mice and humans. PLoS One 2008; 3:e1912. [PMID: 18382685 PMCID: PMC2271127 DOI: 10.1371/journal.pone.0001912] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Accepted: 02/28/2008] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Previous work has demonstrated the potential for peripheral blood (PB) gene expression profiling for the detection of disease or environmental exposures. METHODS AND FINDINGS We have sought to determine the impact of several variables on the PB gene expression profile of an environmental exposure, ionizing radiation, and to determine the specificity of the PB signature of radiation versus other genotoxic stresses. Neither genotype differences nor the time of PB sampling caused any lessening of the accuracy of PB signatures to predict radiation exposure, but sex difference did influence the accuracy of the prediction of radiation exposure at the lowest level (50 cGy). A PB signature of sepsis was also generated and both the PB signature of radiation and the PB signature of sepsis were found to be 100% specific at distinguishing irradiated from septic animals. We also identified human PB signatures of radiation exposure and chemotherapy treatment which distinguished irradiated patients and chemotherapy-treated individuals within a heterogeneous population with accuracies of 90% and 81%, respectively. CONCLUSIONS We conclude that PB gene expression profiles can be identified in mice and humans that are accurate in predicting medical conditions, are specific to each condition and remain highly accurate over time.
Collapse
Affiliation(s)
- Sarah K. Meadows
- Division of Cellular Therapy, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Holly K. Dressman
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
- Institute for Genome Sciences and Policy, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Garrett G. Muramoto
- Division of Cellular Therapy, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Heather Himburg
- Division of Cellular Therapy, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Alice Salter
- Division of Cellular Therapy, Duke University Medical Center, Durham, North Carolina, United States of America
| | - ZhengZheng Wei
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Geoff Ginsburg
- Institute for Genome Sciences and Policy, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Nelson J. Chao
- Division of Cellular Therapy, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Joseph R. Nevins
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
- Institute for Genome Sciences and Policy, Duke University Medical Center, Durham, North Carolina, United States of America
| | - John P. Chute
- Division of Cellular Therapy, Duke University Medical Center, Durham, North Carolina, United States of America
- Institute for Genome Sciences and Policy, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
68
|
Fernández-Riejos P, Goberna R, Sánchez-Margalet V. Leptin promotes cell survival and activates Jurkat T lymphocytes by stimulation of mitogen-activated protein kinase. Clin Exp Immunol 2008; 151:505-18. [PMID: 18234059 DOI: 10.1111/j.1365-2249.2007.03563.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Leptin (Ob) is a non-glycosylated peptide hormone that regulates energy homeostasis centrally, but also has systemic effects including the regulation of the immune function. We have reported previously that leptin activates human peripheral blood lymphocytes co-stimulated with phytohaemagglutinin (PHA) (4 microg/ml), which prevented the employment of pharmacological inhibitors of signalling pathways. In the present study, we used Jurkat T cells that responded to leptin with minimal PHA co-stimulation (0.25 microg/ml). The long isoform of leptin receptor is expressed on Jurkat T cells and upon leptin stimulation, the expression of early activation marker CD69 increases in a dose-dependent manner (0.1-10 nM). We have also found that leptin activates receptor-associated kinases of the Janus family-signal transucers and activators of transcription (JAK-STAT), mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3 kinase (PI3K) signalling pathways. Moreover, we sought to study the possible effect of leptin on cell survival and apoptosis of Jurkat T cells by culture in serum-free conditions. We have assayed the early phases of apoptosis by flow cytometric detection of fluorescein isothiocyanate (FITC)-labelled annexin V simultaneously with dye exclusion of propidium iodide (PI). As well, we have assayed the activation level of caspase-3 by inmunoblot with a specific antibody that recognizes active caspase-3. We have found that leptin inhibits the apoptotic process dose-dependently. By using pharmacological inhibitors, we have found that the stimulatory and anti-apoptotic effects of leptin in Jurkat T cells are dependent on MAPK activation, rather than the PI3K pathway, providing new data regarding the mechanism of action of leptin in T cells, which may be useful to understand more clearly the association between nutritional status and the immune function.
Collapse
Affiliation(s)
- P Fernández-Riejos
- Department of Clinical Biochemistry, Virgen Macarena University Hospital, Medical School University of Seville, Seville, Spain
| | | | | |
Collapse
|
69
|
Abstract
Malnutrition, secondary to deficiency in intake of proteins, minerals or vitamins, consistently results in changes in the thymus. This organ undergoes a severe atrophy due to apoptosis-induced thymocyte depletion, particularly affecting the immature CD4+CD8+ cells, as well as a decrease in cell proliferation. This feature is apparently linked to a hormonal imbalance, involving a decrease in leptin and consequent increase in glucocorticoid hormone levels in the serum. The thymic microenvironment is also affected in malnutrition: morphological changes in thymic epithelial cells have been found, together with a decrease of thymic hormone production by these cells. Additionally, intrathymic contents of extracellular proteins, such as fibronectin, laminin and collagens, are increased in thymuses from malnourished children. Taken together, these data clearly point to the notion that the thymus is significantly affected in malnutrition. Similar patterns of thymic changes occur in acute infectious diseases, including a severe atrophy of the organ, mainly due to the apoptosis-related depletion of immature CD4+CD8+ thymocytes. Additionally, thymocyte proliferation is compromised in acutely-infected subjects. The microenvironmental compartment of the thymus is also affected in acute infections, with an increased density of the epithelial network and an increase in the deposition of extracellular matrix. In conclusion, it seems clear that the thymus is targeted in malnutrition as well as in acute infections. These changes are related to the impaired peripheral immune response seen in malnourished and infected individuals. Thus, strategies inducing thymus replenishment should be considered in therapeutic approaches, in both malnutrition and acute infectious diseases.
Collapse
|
70
|
Abstract
Although leptin is known for its regulation of food intake, it has many emerging roles in immune function. To better define the role of leptin in hematopoietic processes, a leptin-deficient obese mouse (ob/ob) and C57BL/6 lean wild-type controls were compared. Despite their large size and consumption of substantial amounts of nutrients, the ob/ob mice had only 60% as many nucleated cells in their marrow as controls. The greatest impact of leptin deficiency was on the B cell compartment that had 70% fewer cells, reducing the absolute number of pre-B and immature B cells to 21% and 12% of normal, respectively, and indicating a significant reduction in lymphopoiesis in ob/ob mice. Whereas the proportion of myeloids remained nearly normal in the obese mice, they also exhibited a reduction of 40% and 25%, respectively, in absolute numbers of granulocytes and monocytes. Seven days of provision of recombinant leptin promoted substantial lymphopoiesis, increasing the numbers of B cells in the marrow of the obese mice twofold, while doubling and tripling, respectively, the numbers of pre-B and immature B cells. Twelve days of supplementation brought these subpopulations to near-normal proportions. Leptin treatment also facilitated myelopoiesis such that the marrow of the obese mice contained normal numbers of monocytes and granulocytes after 7 days. Taken together, the data support an important role for leptin in sustaining lymphopoiesis and myelopoiesis.
Collapse
|
71
|
Dixit VD, Yang H, Sun Y, Weeraratna AT, Youm YH, Smith RG, Taub DD. Ghrelin promotes thymopoiesis during aging. J Clin Invest 2007; 117:2778-90. [PMID: 17823656 PMCID: PMC1964507 DOI: 10.1172/jci30248] [Citation(s) in RCA: 155] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2006] [Accepted: 06/26/2007] [Indexed: 11/17/2022] Open
Abstract
The decline in adaptive immunity, T lymphocyte output, and the contraction of the TCR repertoire with age is largely attributable to thymic involution. The loss of thymic function with age may be due to diminished numbers of progenitors and the loss of critical cytokines and hormones from the thymic microenvironment. We have previously demonstrated that the orexigenic hormone ghrelin is expressed by immune cells and regulates T cell activation and inflammation. Here we report that ghrelin and ghrelin receptor expression within the thymus diminished with progressive aging. Infusion of ghrelin into 14-month-old mice significantly improved the age-associated changes in thymic architecture and thymocyte numbers, increasing recent thymic emigrants and improving TCR diversity of peripheral T cell subsets. Ghrelin-induced thymopoiesis during aging was associated with enhanced early thymocyte progenitors and bone marrow-derived Lin(-)Sca1(+)cKit(+) cells, while ghrelin- and growth hormone secretagogue receptor-deficient (GHS-R-deficient) mice displayed enhanced age-associated thymic involution. Leptin also enhanced thymopoiesis in aged but not young mice. Our findings demonstrate what we believe to be a novel role for ghrelin and its receptor in thymic biology and suggest a possible therapeutic benefit of harnessing this pathway in the reconstitution of thymic function in immunocompromised subjects.
Collapse
Affiliation(s)
- Vishwa Deep Dixit
- Clinical Immunology Section, Laboratory of Immunology, Intramural Research Program, National Institute on Aging, NIH, Baltimore, Maryland, USA.
Laboratory of Neuroendocrine Immunology, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA.
Roy M. and Phyllis Gough Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA
| | - Hyunwon Yang
- Clinical Immunology Section, Laboratory of Immunology, Intramural Research Program, National Institute on Aging, NIH, Baltimore, Maryland, USA.
Laboratory of Neuroendocrine Immunology, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA.
Roy M. and Phyllis Gough Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA
| | - Yuxiang Sun
- Clinical Immunology Section, Laboratory of Immunology, Intramural Research Program, National Institute on Aging, NIH, Baltimore, Maryland, USA.
Laboratory of Neuroendocrine Immunology, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA.
Roy M. and Phyllis Gough Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA
| | - Ashani T. Weeraratna
- Clinical Immunology Section, Laboratory of Immunology, Intramural Research Program, National Institute on Aging, NIH, Baltimore, Maryland, USA.
Laboratory of Neuroendocrine Immunology, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA.
Roy M. and Phyllis Gough Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA
| | - Yun-Hee Youm
- Clinical Immunology Section, Laboratory of Immunology, Intramural Research Program, National Institute on Aging, NIH, Baltimore, Maryland, USA.
Laboratory of Neuroendocrine Immunology, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA.
Roy M. and Phyllis Gough Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA
| | - Roy G. Smith
- Clinical Immunology Section, Laboratory of Immunology, Intramural Research Program, National Institute on Aging, NIH, Baltimore, Maryland, USA.
Laboratory of Neuroendocrine Immunology, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA.
Roy M. and Phyllis Gough Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA
| | - Dennis D. Taub
- Clinical Immunology Section, Laboratory of Immunology, Intramural Research Program, National Institute on Aging, NIH, Baltimore, Maryland, USA.
Laboratory of Neuroendocrine Immunology, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA.
Roy M. and Phyllis Gough Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
72
|
Abstract
Ageing is a complex process that negatively impacts the development of the immune system and its ability to function. The mechanisms that underlie these age-related defects are broad and range from defects in the haematopoietic bone marrow to defects in peripheral lymphocyte migration, maturation and function. The thymus is a central lymphoid organ responsible for production of naïve T cells, which play a vital role in mediating both cellular and humoral immunity. Chronic involution of the thymus gland is thought to be one of the major contributing factors to loss of immune function with increasing age. It has recently been demonstrated that thymic atrophy is mediated by a shift from a stimulatory to a suppressive cytokine microenvironment. In this review we present an overview of the morphological, cellular and biochemical changes that have been implicated in the decline of thymic and peripheral immune function with ageing. We conclude with the clinical implications of age-associated immunosenescence to vaccine development for tumours and infectious disease. A fundamental understanding of the complex mechanisms by which ageing attenuates immune function will enable translational research teams to develop new therapies and vaccines specifically aimed at overcoming these defects in immunological function in the aged.
Collapse
Affiliation(s)
- AL Gruver
- Duke University Human Vaccine Institute and Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - LL Hudson
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| | - GD Sempowski
- Duke University Human Vaccine Institute and Department of Pathology, Duke University Medical Center, Durham, NC, USA
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
- Correspondence to: GD Sempowski, DUMC Box 3258, Research Park Building I, Room 113, Research Drive, Durham, NC 27710, USA. E-mail:
| |
Collapse
|