Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Front Endocrinol (Lausanne) ; 14: 1168186, 2023.
Article in English | MEDLINE | ID: mdl-37529610

ABSTRACT

The thymus gland is a central lymphoid organ in which developing T cell precursors, known as thymocytes, undergo differentiation into distinct type of mature T cells, ultimately migrating to the periphery where they exert specialized effector functions and orchestrate the immune responses against tumor cells, pathogens and self-antigens. The mechanisms supporting intrathymic T cell differentiation are pleiotropically regulated by thymic peptide hormones and cytokines produced by stromal cells in the thymic microenvironment and developing thymocytes. Interestingly, in the same way as T cells, thymic hormones (herein exemplified by thymosin, thymulin and thymopoietin), can circulate to impact immune cells and other cellular components in the periphery. Evidence on how thymic function influences tumor cell biology and response of patients with cancer to therapies remains unsatisfactory, although there has been some improvement in the knowledge provided by recent studies. Herein, we summarize research progression in the field of thymus-mediated immunoendocrine control of cancer, providing insights into how manipulation of the thymic microenvironment can influence treatment outcomes, including clinical responses and adverse effects of therapies. We review data obtained from clinical and preclinical cancer research to evidence the complexity of immunoendocrine interactions underpinning anti-tumor immunity.


Subject(s)
Neoplasms , Thymus Gland , Humans , T-Lymphocytes , Cytokines/metabolism , Neoplasms/metabolism , Peptides/metabolism , Tumor Microenvironment
2.
Article in English | MEDLINE | ID: mdl-30042731

ABSTRACT

Maintenance of thymus homeostasis is a delicate interplay involving hormones, neurotransmitters and local microenvironmental proteins, as well as saccharides, acting on both thymocytes and stromal cells. Disturbances in these interactions may lead to alterations on thymocyte development. We previously showed that galectin-3, a ß-galactoside-binding protein, is constitutively expressed in the thymus, interacting with extracellular matrix glycoproteins and acting as a de-adhesion molecule, thus modulating thymocyte-stromal cell interactions. In this work, we aimed to investigate the participation of galectin-3 in the maintenance of thymus homeostasis, including hormonal-mediated circuits. For that, we used genetically engineered galectin-3-deficient mice. We observed that the thymus of galectin-3-deficient mice was reduced in mass and cellularity compared to wild-type controls; however, the proportions of different thymocyte subpopulations defined by CD4/CD8 expression were not changed. Considering the CD4-CD8- double-negative (DN) subpopulation, an accumulation of the most immature (DN1) stage was observed. Additionally, the proliferative capacity of thymocytes was decreased in all thymocyte subsets, whereas the percentage of apoptosis was increased, especially in the CD4+CD8+ double-positive thymocytes. As glucocorticoid hormones are known to be involved in thymus homeostasis, we evaluated serum and intrathymic corticosterone levels by radioimmunoassay, and the expression of steroidogenic machinery using real-time PCR. We detected a significant increase in corticosterone levels in both serum and thymus samples of galectin-3-deficient mice, as compared to age-matched controls. This was paralleled by an increase of gene transcription of the steroidogenic enzymes, steroidogenic acute regulatory protein (Star) and Cyp11b1 in thymus, 11ß-Hydroxysteroid Dehydrogenase (Hsd11b1) in the adrenal, and Cyp11a1 in both glands. In conclusion, our findings show that the absence of galectin-3 subverts mouse thymus homeostasis by a mechanism likely associated to intrathymic and systemic stress-related endocrine circuitries, affecting thymocyte number, proliferation and apoptosis.

3.
Cell Adh Migr ; 12(2): 152-167, 2018 03 04.
Article in English | MEDLINE | ID: mdl-28494186

ABSTRACT

The thymus supports differentiation of T cell precursors. This process requires relocation of developing thymocytes throughout multiple microenvironments of the organ, mainly with thymic epithelial cells (TEC), which control intrathymic T cell differentiation influencing the formation and maintenance of the immunological synapse. In addition to the proteins of the major histocompatibility complex (MHC), this structure is supported by several adhesion molecules. During the process of thymopoiesis, we previously showed that laminin-mediated interactions are involved in the entrance of T-cell precursors into the thymus, as well as migration of differentiating thymocytes within the organ. Using small interference RNA strategy, we knocked-down the ITGA6 gene (which encodes the CD49f integrin α-chain) in cultured human TEC, generating a decrease in the expression of the corresponding CD49f subunit, in addition to modulation in several other genes related to cell adhesion and migration. Thymocyte adhesion to TEC was significantly impaired, comprising both immature and mature thymocyte subsets. Moreover, we found a modulation of the MHC, with a decrease in membrane expression of HLA-ABC, in contrast with increase in the expression of HLA-DR. Interestingly, the knockdown of the B2M gene (encoding the ß-2 microglobulin of the HLA-ABC complex) increased CD49f expression levels, thus unraveling the existence of a cross-talk event in the reciprocal control of CD49f and HLA-ABC. Our data suggest that the expression levels of CD49f may be relevant in the general control of MHC expression by TEC and consequently the corresponding synapse with developing thymocytes mediated by the T-cell receptor.


Subject(s)
Cell Adhesion Molecules/metabolism , Epithelial Cells/metabolism , Epithelium/metabolism , Immunological Synapses/metabolism , Integrin alpha6/genetics , Cell Adhesion/physiology , Cell Differentiation/physiology , Cell Movement/physiology , Extracellular Matrix/metabolism , Gene Targeting/methods , Humans , Integrins/metabolism
4.
Nat Rev Endocrinol ; 12(2): 77-89, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26437623

ABSTRACT

The physiology of the thymus, the primary lymphoid organ in which T cells are generated, is controlled by hormones. Data from animal models indicate that several peptide and nonpeptide hormones act pleiotropically within the thymus to modulate the proliferation, differentiation, migration and death by apoptosis of developing thymocytes. For example, growth hormone and prolactin can enhance thymocyte proliferation and migration, whereas glucocorticoids lead to the apoptosis of these developing cells. The thymus undergoes progressive age-dependent atrophy with a loss of cells being generated and exported, therefore, hormone-based therapies are being developed as an alternative strategy to rejuvenate the organ, as well as to augment thymocyte proliferation and the export of mature T cells to peripheral lymphoid organs. Some hormones (such as growth hormone and progonadoliberin-1) are also being used as therapeutic agents to treat immunodeficiency disorders associated with thymic atrophy, such as HIV infection. In this Review, we discuss the accumulating data that shows the thymus gland is under complex and multifaceted hormonal control that affects the process of T-cell development in health and disease.


Subject(s)
Cell Differentiation/immunology , Human Growth Hormone/immunology , Prolactin/immunology , T-Lymphocytes/immunology , Thymocytes/immunology , Thymus Gland/immunology , Animals , Cell Movement/immunology , Cell Proliferation , Gonadotropin-Releasing Hormone/therapeutic use , Growth Hormone/immunology , HIV Infections/drug therapy , Human Growth Hormone/therapeutic use , Humans , Immunologic Deficiency Syndromes/drug therapy , Lymphoid Tissue/immunology , Protein Precursors/therapeutic use
5.
Gerontology ; 61(6): 504-14, 2015.
Article in English | MEDLINE | ID: mdl-25765703

ABSTRACT

Thymic involution during aging is a major reason for the decreased production of naive T cells and reduced immunity. Alterations within the thymic microenvironment, characterized by the loss of function of thymic epithelial cells (TECs) and fibro-adipogenetic transformation, seem to underlie this process, mainly through declining communication between thymic stromal cells and developing thymocytes. Specifically, the signaling mediated by cytokines and hormones secreted by TECs declines during aging. Many therapies based on the manipulation of growth factors and hormones have succeeded in partially recovering the lymphoid compartment and promoting thymic function. However, considering that aging-induced thymic involution is multifactorial, the thymic reestablishment achieved with treatments that target isolated pathways is incomplete and transitory. Here, we discuss the development of three novel approaches for potentially sustained thymic recovery: the induction of sustained forkhead box N1 expression, the activation of endogenous thymic epithelial progenitor cells (TEPCs), and the generation of TEPCs from pluripotent stem cells. Combined approaches targeting both TECs and lymphoid cells will provide a potentially more effective strategy for sustained rejuvenation of the thymus.


Subject(s)
Aging/pathology , Epithelial Cells/physiology , Thymus Gland/pathology , Aging/physiology , Cellular Microenvironment , Forkhead Transcription Factors , Humans , Signal Transduction/physiology
6.
PLoS Negl Trop Dis ; 8(10): e3203, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25330249

ABSTRACT

The protozoan parasite Trypanosoma cruzi is able to target the thymus and induce alterations of the thymic microenvironmental and lymphoid compartments. Acute infection results in severe atrophy of the organ and early release of immature thymocytes into the periphery. To date, the pathophysiological effects of thymic changes promoted by parasite-inducing premature release of thymocytes to the periphery has remained elusive. Herein, we show that sphingosine-1-phosphate (S1P), a potent mediator of T cell chemotaxis, plays a role in the exit of immature double-negative thymocytes in experimental Chagas disease. In thymuses from T. cruzi-infected mice we detected reduced transcription of the S1P kinase 1 and 2 genes related to S1P biosynthesis, together with increased transcription of the SGPL1 sphingosine-1-lyase gene, whose product inactivates S1P. These changes were associated with reduced intrathymic levels of S1P kinase activity. Interestingly, double-negative thymocytes from infected animals expressed high levels of the S1P receptor during infection, and migrated to lower levels of S1P. Moreover, during T. cruzi infection, this thymocyte subset expresses high levels of IL-17 and TNF-α cytokines upon polyclonal stimulation. In vivo treatment with the S1P receptor antagonist FTY720 resulted in recovery the numbers of double-negative thymocytes in infected thymuses to physiological levels. Finally, we showed increased numbers of double-negative T cells in the peripheral blood in severe cardiac forms of human Chagas disease.


Subject(s)
Chagas Disease/physiopathology , Lysophospholipids/metabolism , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Thymocytes/metabolism , Trypanosoma cruzi , Animals , Case-Control Studies , Humans , Leukocytes, Mononuclear/physiology , Male , Mice , Mice, Inbred BALB C , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Lysosphingolipid/genetics , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors , T-Lymphocytes
7.
PLoS Negl Trop Dis ; 7(11): e2470, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24324845

ABSTRACT

We have previously shown that experimental infection caused by Trypanosoma cruzi is associated with changes in the hypothalamus-pituitary-adrenal axis. Increased glucocorticoid (GC) levels are believed to be protective against the effects of acute stress during infection but result in depletion of CD4(+)CD8(+) thymocytes by apoptosis, driving to thymic atrophy. However, very few data are available concerning prolactin (PRL), another stress-related hormone, which seems to be decreased during T. cruzi infection. Considering the immunomodulatory role of PRL upon the effects caused by GC, we investigated if intrathymic cross-talk between GC and PRL receptors (GR and PRLR, respectively) might influence T. cruzi-induced thymic atrophy. Using an acute experimental model, we observed changes in GR/PRLR cross-activation related with the survival of CD4(+)CD8(+) thymocytes during infection. These alterations were closely related with systemic changes, characterized by a stress hormone imbalance, with progressive GC augmentation simultaneously to PRL reduction. The intrathymic hormone circuitry exhibited an inverse modulation that seemed to counteract the GC-related systemic deleterious effects. During infection, adrenalectomy protected the thymus from the increase in apoptosis ratio without changing PRL levels, whereas an additional inhibition of circulating PRL accelerated the thymic atrophy and led to an increase in corticosterone systemic levels. These results demonstrate that the PRL impairment during infection is not caused by the increase of corticosterone levels, but the opposite seems to occur. Accordingly, metoclopramide (MET)-induced enhancement of PRL secretion protected thymic atrophy in acutely infected animals as well as the abnormal export of immature and potentially autoreactive CD4(+)CD8(+) thymocytes to the periphery. In conclusion, our findings clearly show that Trypanosoma cruzi subverts mouse thymus homeostasis by altering intrathymic and systemic stress-related endocrine circuitries with major consequences upon the normal process of intrathymic T cell development.


Subject(s)
Chagas Disease/immunology , Homeostasis , Hormones/metabolism , Host-Pathogen Interactions , Stress, Physiological , Thymus Gland/physiology , Trypanosoma cruzi/immunology , Animals , Disease Models, Animal , Male , Mice , Mice, Inbred BALB C , Thymocytes/physiology
8.
PLoS One ; 8(5): e63814, 2013.
Article in English | MEDLINE | ID: mdl-23717489

ABSTRACT

Adrenal steroidogenesis is under a complex regulation involving extrinsic and intrinsic adrenal factors. TNF-α is an inflammatory cytokine produced in response to tissue injury and several other stimuli. We have previously demonstrated that TNF-R1 knockout (TNF-R1(-/-)) mice have a dysregulated synthesis of glucocorticoids (GCs) during Trypanosoma cruzi acute infection. Since TNF-α may influence GCs production, not only through the hypothalamus-pituitary axis, but also at the adrenal level, we now investigated the role of this cytokine on the adrenal GCs production. Wild type (WT) and TNF-R1(-/-) mice undergoing acute infection (Tc-WT and Tc-TNF-R1(-/-) groups), displayed adrenal hyperplasia together with increased GCs levels. Notably, systemic ACTH remained unchanged in Tc-WT and Tc-TNF-R1(-/-) compared with uninfected mice, suggesting some degree of ACTH-independence of GCs synthesis. TNF-α expression was increased within the adrenal gland from both infected mouse groups, with Tc-WT mice showing an augmented TNF-R1 expression. Tc-WT mice showed increased levels of P-p38 and P-ERK compared to uninfected WT animals, whereas Tc-TNF-R1(-/-) mice had increased p38 and JNK phosphorylation respect to Tc-WT mice. Strikingly, adrenal NF-κB and AP-1 activation during infection was blunted in Tc-TNF-R1(-/-) mice. The accumulation of mRNAs for steroidogenic acute regulatory protein and cytochrome P450 were significantly increased in both Tc-WT and Tc-TNF-R1(-/-) mice; being much more augmented in the latter group, which also had remarkably increased GCs levels. TNF-α emerges as a potent modulator of steroidogenesis in adrenocortical cells during T. cruzi infection in which MAPK pathways, NF-κB and AP-1 seem to play a role in the adrenal synthesis of pro-inflammatory cytokines and enzymes regulating GCs synthesis. These results suggest the existence of an intrinsic immune-adrenal interaction involved in the dysregulated synthesis of GCs during murine Chagas disease.


Subject(s)
Adrenal Glands/metabolism , Chagas Disease/metabolism , Chagas Disease/parasitology , Glucocorticoids/biosynthesis , Glucocorticoids/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adrenocorticotropic Hormone/metabolism , Animals , Chagas Disease/blood , Chagas Disease/genetics , Corticosterone/genetics , Corticosterone/metabolism , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Gene Expression/genetics , Glucocorticoids/genetics , Inflammation/genetics , Inflammation/metabolism , Inflammation/parasitology , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation/genetics , RNA, Messenger/genetics , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Trypanosoma cruzi , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics
9.
Ann N Y Acad Sci ; 1262: 45-50, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22823434

ABSTRACT

Disorders in the hypothalamic-pituitary-adrenal axis are associated with the pathogenesis of Trypanosoma cruzi infection. During the acute phase of this disease, increased levels of circulating glucocorticoids (GCs) correlate with thymic atrophy. Recently, we demonstrated that this phenomenon is paralleled by a decrease of prolactin (PRL) secretion, another stress hormone that seems to counteract many immunosuppressive effects of GCs. Both GCs and PRL are intrathymically produced and exhibit mutual antagonism through the activation of their respective receptors, GR, and PRLR. Considering that GCs induce apoptosis and inhibit double-positive (DP) thymocyte proliferation and that PRL administration prevents these effects, it seems plausible that a local imbalance of GR-PRLR crosstalk underlies the thymic involution occurring in acute T. cruzi infection. In this respect, preserving PRLR signaling seems to be crucial for protecting DP from GC-induced apoptosis.


Subject(s)
Chagas Disease/immunology , Chagas Disease/pathology , Thymus Gland/pathology , Animals , Apoptosis/immunology , Atrophy , Glucocorticoids/immunology , Host-Pathogen Interactions/immunology , Humans , Mice , Neuroimmunomodulation , Neurosecretory Systems/immunology , Prolactin/immunology , Receptor Cross-Talk , Receptors, Glucocorticoid/immunology , Receptors, Prolactin/immunology , Signal Transduction , Stress, Physiological , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , Thymus Gland/immunology
10.
PLoS One ; 7(3): e34360, 2012.
Article in English | MEDLINE | ID: mdl-22461911

ABSTRACT

Previous studies revealed a significant production of inflammatory cytokines together with severe thymic atrophy and thymocyte migratory disturbances during experimental Chagas disease. Migratory activity of thymocytes and mature T cells seem to be finely tuned by cytokines, chemokines and extracellular matrix (ECM) components. Systemic TNF-α is enhanced during infection and appears to be crucial in the response against the parasite. However, it also seems to be involved in disease pathology, since it is implicated in the arrival of T cells to effector sites, including the myocardium. Herein, we analyzed the role of TNF-α in the migratory activity of thymocytes in Trypanosoma cruzi (T. cruzi) acutely-infected mice. We found increased expression and deposition of TNF-α in the thymus of infected animals compared to controls, accompanied by increased co-localization of fibronectin, a cell migration-related ECM molecule, whose contents in the thymus of infected mice is also augmented. In-vivo studies showed an enhanced export of thymocytes in T. cruzi-infected mice, as ascertained by intrathymic injection of FITC alone or in combination with TNF-α. The increase of immature CD4(+)CD8(+) T cells in secondary lymphoid organs was even more clear-cut when TNF-α was co-injected with FITC. Ex-vivo transmigration assays also revealed higher number of migrating cells when TNF-α was added onto fibronectin lattices, with higher input of all thymocyte subsets, including immature CD4(+)CD8(+). Infected animals also exhibit enhanced levels of expression of both mRNA TNF-α receptors in the CD4(+)CD8(+) subpopulation. Our findings suggest that in T. cruzi acute infection, when TNF-α is complexed with fibronectin, it favours the altered migration of thymocytes, promoting the release of mature and immature T cells to different compartments of the immune system. Conceptually, this work reinforces the notion that thymocyte migration is a multivectorial biological event in health and disease, and that TNF-α is a further player in the process.


Subject(s)
Cell Movement/immunology , Chagas Disease/immunology , Thymocytes/immunology , Trypanosoma cruzi/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Atrophy , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Movement/drug effects , Chagas Disease/metabolism , Chagas Disease/parasitology , Fibronectins/immunology , Fibronectins/metabolism , Flow Cytometry , Gene Expression , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thymocytes/cytology , Thymocytes/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism , Thymus Gland/pathology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
11.
J Renin Angiotensin Aldosterone Syst ; 8(4): 169-75, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18205095

ABSTRACT

OBJECTIVE: Considering the controversial data regarding the role of the brain renin-angiotensin system (RAS) on the thirst and sodium appetite in ovariectomised rats, we aimed to evaluate the role of the brain angiotensin II (Ang II) AT1-receptor on the nocturnal fluids intake. MATERIALS AND METHODS: Groups of Wistar female rats were ovariectomised and chronically given oestrogen or vehicle to evaluate its influence on effects induced by i.c.v. injection of losartan, Ang I and Ang II. RESULTS: The i.c.v. losartan decreased basal water intake in the ovariectomised group. Ang II but not Ang I-induced nocturnal dipsogenic and natriorexigenic responses in ovariectomised rats. In oestrogen-treated rats, both peptides increased fluids intake. Previously, i.c.v. losartan abolished these effects in all groups. Oestrogen replacement decreased the nocturnal fluids intake, attenuated the losartan and Ang II effects, and highlighted the Ang I response. CONCLUSIONS: The present study has shown for the first time the involvement of AT1-receptor in regulating nocturnal basal water and salt intake in ovariectomised rats. In addition, our data have revealed an unexpected increased brain Ang I-mediated fluid intake in oestrogen-treated ovariectomised compared to ovariectomised rats, which was blocked by previous i.c.v. losartan. Our data have therefore shown that oestrogen influences homeostatic behaviours dependent on brain RAS.


Subject(s)
Angiotensins/pharmacology , Appetite/drug effects , Brain/metabolism , Circadian Rhythm/physiology , Receptor, Angiotensin, Type 1/metabolism , Sodium/metabolism , Thirst/drug effects , Administration, Oral , Angiotensin I/pharmacology , Angiotensin II/pharmacology , Animals , Antihypertensive Agents/pharmacology , Brain/drug effects , Darkness , Drinking/drug effects , Estrogens/pharmacology , Female , Injections, Intraventricular , Losartan/administration & dosage , Losartan/pharmacology , Ovariectomy , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL