Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Neurochem Res ; 49(7): 1762-1781, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38551797

RESUMO

Lactate has received attention as a potential therapeutic intervention for brain diseases, particularly those including energy deficit, exacerbated inflammation, and disrupted redox status, such as cerebral ischemia. However, lactate roles in metabolic or signaling pathways in neural cells remain elusive in the hypoxic and ischemic contexts. Here, we tested the effects of lactate on the survival of a microglial (BV-2) and a neuronal (SH-SY5Y) cell lines during oxygen and glucose deprivation (OGD) or OGD followed by reoxygenation (OGD/R). Lactate signaling was studied by using 3,5-DHBA, an exogenous agonist of lactate receptor GPR81. Inhibition of lactate dehydrogenase (LDH) or monocarboxylate transporters (MCT), using oxamate or 4-CIN, respectively, was performed to evaluate the impact of lactate metabolization and transport on cell viability. The OGD lasted 6 h and the reoxygenation lasted 24 h following OGD (OGD/R). Cell viability, extracellular lactate concentrations, microglial intracellular pH and TNF-ɑ release, and neurite elongation were evaluated. Lactate or 3,5-DHBA treatment during OGD increased microglial survival during reoxygenation. Inhibition of lactate metabolism and transport impaired microglial and neuronal viability. OGD led to intracellular acidification in BV-2 cells, and reoxygenation increased the release of TNF-ɑ, which was reverted by lactate and 3,5-DHBA treatment. Our results suggest that lactate plays a dual role in OGD, acting as a metabolic and a signaling molecule in BV-2 and SH-SY5Y cells. Lactate metabolism and transport are vital for cell survival during OGD. Moreover, lactate treatment and GPR81 activation during OGD promote long-term adaptations that potentially protect cells against secondary cell death during reoxygenation.


Assuntos
Sobrevivência Celular , Glucose , Ácido Láctico , Microglia , Neurônios , Oxigênio , Microglia/metabolismo , Microglia/efeitos dos fármacos , Glucose/metabolismo , Glucose/deficiência , Humanos , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Oxigênio/metabolismo , Ácido Láctico/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Animais , Camundongos , Fármacos Neuroprotetores/farmacologia , Hipóxia Celular/fisiologia , Hipóxia Celular/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Linhagem Celular Tumoral , Linhagem Celular , Transportadores de Ácidos Monocarboxílicos/metabolismo
2.
Neuroimmunomodulation ; 31(1): 51-61, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38272012

RESUMO

BACKGROUND: T-cell acute lymphoblastic leukemia (T-ALL) is a malignant hematologic disease caused by the transformation and uncontrolled proliferation of T-cell precursors. T-ALL is generally thought to originate in the thymus since lymphoblasts express phenotypic markers comparable to those described in thymocytes in distinct stages of development. Although around 50% of T-ALL patients present a thymic mass, T-ALL is characterized by peripheral blood and bone marrow involvement, and central nervous system (CNS) infiltration is one of the most severe complications of the disease. SUMMARY: The CNS invasion is related to the expression of specific adhesion molecules and receptors commonly expressed in developing T cells, such as L-selectin, CD44, integrins, and chemokine receptors. Furthermore, T-ALL blasts also express neurotransmitters, neuropeptides, and cognate receptors that are usually present in the CNS and can affect both the brain and thymus, participating in the crosstalk between the organs. KEY MESSAGES: This review discusses how the thymus-brain connections, mediated by innervation and common molecules and receptors, can impact the development and migration of T-ALL blasts, including CNS infiltration.


Assuntos
Encéfalo , Leucemia-Linfoma Linfoblástico de Células T Precursoras , Timo , Humanos , Timo/patologia , Encéfalo/patologia , Encéfalo/metabolismo , Encéfalo/imunologia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Animais
3.
Neuroimmunomodulation ; 25(5-6): 280-284, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30121669

RESUMO

OBJECTIVE: Considering the potential role of lymphocytes in the pathophysiology of autism spectrum disorder (ASD), we aimed to evaluate possible alterations of T cell pools in the lymphoid organs of an animal model of autism induced by valproic acid (VPA). Pregnant Swiss mice received a single intraperitoneal injection of 600 mg/kg of VPA (VPA group) or saline (control group) on day 11 of gestation. Male offspring were euthanized on postnatal day 60 for removal of thy-muses, spleens, and a pool of inguinal, axillary and brachial lymph nodes. Cellularity was evaluated, and flow cytometry analysis was performed on cell suspensions incubated with the mouse antibodies anti-CD3-FITC, anti-CD4-PE, and anti-CD8-PE-Cy7. We observed that the prenatal exposure to VPA induced a reduction in the numbers of CD3+CD4+ T cells in their lymph nodes when compared to the control animals. This was specific since it was not seen in the thymus or spleen. The consistent decrease in the number of CD4+ T cells in subcutaneous lymph nodes of mice from the animal model of autism may be related to the allergic symptoms frequently observed in ASD. Further research is necessary to characterize the immunological patterns in ASD and the connection with the pathophysiology of this disorder.


Assuntos
Transtorno Autístico/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfonodos/imunologia , Animais , Transtorno Autístico/induzido quimicamente , Modelos Animais de Doenças , Inibidores Enzimáticos/toxicidade , Feminino , Masculino , Camundongos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Ácido Valproico/toxicidade
4.
FASEB J ; 26(11): 4390-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22815386

RESUMO

Cell migration is a crucial event for normal T-cell development, and various ligand/receptor pairs have been implicated. Most of them, including chemokines and extracellular matrix proteins, have attractant properties on thymocytes. We discuss herein two further groups of ligand/receptor pairs, semaphorins/neuropilins and ephs/ephrins, which are constitutively expressed by thymocytes and thymic microenvironmental cells. Evidence shows that the corresponding interactions are relevant for developing T-cell migration, including the entry of bone marrow progenitor cells, migration of CD4/CD8-defined thymocyte subpopulations triggered by chemokines and/or extracellular matrix proteins, and thymocyte export. Conceptually, the data summarized here show that thymocyte migration results from a complex network of molecular interactions, which generate not only attraction, but also repulsion of migrating T-cell precursors.


Assuntos
Movimento Celular/fisiologia , Efrinas/metabolismo , Semaforinas/metabolismo , Timócitos/fisiologia , Animais , Efrinas/genética , Regulação da Expressão Gênica , Semaforinas/genética
5.
Front Immunol ; 14: 1108630, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37426675

RESUMO

Growth hormone (GH) is a classic pituitary-derived hormone crucial to body growth and metabolism. In the pituitary gland, GH production is stimulated by GH-releasing hormone and inhibited by somatostatin. GH secretion can also be induced by other peptides, such as ghrelin, which interacts with receptors present in somatotropic cells. It is well established that GH acts directly on target cells or indirectly by stimulating the production of insulin-like growth factors (IGFs), particularly IGF-1. Notably, such somatotropic circuitry is also involved in the development and function of immune cells and organs, including the thymus. Interestingly, GH, IGF-1, ghrelin, and somatostatin are expressed in the thymus in the lymphoid and microenvironmental compartments, where they stimulate the secretion of soluble factors and extracellular matrix molecules involved in the general process of intrathymic T-cell development. Clinical trials in which GH was used to treat immunocompromised patients successfully recovered thymic function. Additionally, there is evidence that the reduction in the function of the somatotropic axis is associated with age-related thymus atrophy. Treatment with GH, IGF-1 or ghrelin can restore thymopoiesis of old animals, thus in keeping with a clinical study showing that treatment with GH, associated with metformin and dehydroepiandrosterone, could induce thymus regeneration in healthy aged individuals. In conclusion, the molecules of the somatotrophic axis can be envisioned as potential therapeutic targets for thymus regeneration in age-related or pathological thymus involution.


Assuntos
Grelina , Fator de Crescimento Insulin-Like I , Animais , Fator de Crescimento Insulin-Like I/metabolismo , Hormônio do Crescimento , Timo , Somatostatina
6.
Front Immunol ; 14: 1202834, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37920473

RESUMO

Growing evidence demonstrates a continuous interaction between the immune system, the nerve and the muscle in neuromuscular disorders of different pathogenetic origins, such as Duchenne Muscular Dystrophy (DMD) and Amyotrophic Lateral Sclerosis (ALS), the focus of this review. Herein we highlight the complexity of the cellular and molecular interactions involving the immune system in neuromuscular disorders, as exemplified by DMD and ALS. We describe the distinct types of cell-mediated interactions, such as cytokine/chemokine production as well as cell-matrix and cell-cell interactions between T lymphocytes and other immune cells, which target cells of the muscular or nervous tissues. Most of these interactions occur independently of exogenous pathogens, through ligand-receptor binding and subsequent signal transduction cascades, at distinct levels of specificity. Although this issue reveals the complexity of the system, it can also be envisioned as a window of opportunity to design therapeutic strategies (including synthetic moieties, cell and gene therapy, as well as immunotherapy) by acting upon one or more targets. In this respect, we discuss ongoing clinical trials using VLA-4 inhibition in DMD, and in ALS, with a focus on regulatory T cells, both revealing promising results.


Assuntos
Esclerose Lateral Amiotrófica , Distrofia Muscular de Duchenne , Doenças Neuromusculares , Humanos , Distrofia Muscular de Duchenne/terapia , Esclerose Lateral Amiotrófica/terapia , Esclerose Lateral Amiotrófica/genética , Músculos , Terapia Genética/métodos
7.
Exp Physiol ; 97(11): 1146-50, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22327328

RESUMO

Cell migration is a key event for proper intrathymic T-cell differentiation, and several ligand-receptor interactions contribute to the well-co ordinated movement of developing thymocytes within the thymic lobules. Herein we summarize recent data that place semaphorin 3A (Sema3A) and its receptor neuropilin 1 (NRP1) as further players in the physiological process of cell migration in the human thymus. These molecules, as well as class A plexins (necessary for the intracellular signalling transduction triggered by Sema3A-NRP1 ligation), are constitutively expressed by both developing thymocytes and components of the thymic microenvironment, including epithelial and dendritic cells. Functionally, Sema3A decreases the adhesion of human thymocytes on thymic epithelial cell monolayers and exerts per se a dose-dependent chemorepulsive effect on human thymocytes. Moreover, Sema3A inhibits chemoattractant migratory responses induced by other ligands, including fibronectin, laminin and CXCL12 (chemokine CXC motif ligand 12). These data should be placed in the context of the concept that migration of developing T cells is a multivectorial system, in which the resulting migration vector derives from a balance of several simultaneous and/or sequential ligand-receptor pair interactions. Accordingly, semaphorins and neuropilins can be considered as further players in the system.


Assuntos
Movimento Celular/fisiologia , Neuropilinas/fisiologia , Sistemas Neurossecretores/fisiologia , Semaforinas/fisiologia , Linfócitos T/fisiologia , Timócitos/fisiologia , Humanos , Sistemas Neurossecretores/metabolismo , Timócitos/metabolismo , Timo/metabolismo , Timo/fisiologia
8.
Front Nutr ; 9: 948488, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36225882

RESUMO

Undernutrition remains a major issue in global health. Low protein-energy consumption, results in stunting, wasting and/or underweight, three deleterious forms of malnutrition that affect roughly 200 million children under the age of five years. Undernutrition compromises the immune system with the generation of various degrees of immunodeficiency, which in turn, renders undernourished individuals more sensitive to acute infections. The severity of various infectious diseases including visceral leishmaniasis (VL), influenza, and tuberculosis is associated with undernutrition. Immunosuppression resulting from protein-energy undernutrition severely impacts primary and secondary lymphoid organs involved in the response to related pathogens. The thymus-a primary lymphoid organ responsible for the generation of T lymphocytes-is particularly compromised by both undernutrition and infectious diseases. In this respect, we will discuss herein various intrathymic cellular and molecular interactions seen in undernutrition alone or in combination with acute infections. Many examples illustrated in studies on humans and experimental animals clearly revealed that protein-related undernutrition causes thymic atrophy, with cortical thymocyte depletion. Moreover, the non-lymphoid microenvironmental compartment of the organ undergoes important changes in thymic epithelial cells, including their secretory products such as hormones and extracellular matrix proteins. Of note, deficiencies in vitamins and trace elements also induce thymic atrophy. Interestingly, among the molecular interactions involved in the control of undernutrition-induced thymic atrophy is a hormonal imbalance with a rise in glucocorticoids and a decrease in leptin serum levels. Undernutrition also yields a negative impact of acute infections upon the thymus, frequently with the intrathymic detection of pathogens or their antigens. For instance, undernourished mice infected with Leishmania infantum (that causes VL) undergo drastic thymic atrophy, with significant reduction in thymocyte numbers, and decreased levels of intrathymic chemokines and cytokines, indicating that both lymphoid and microenvironmental compartments of the organ are affected. Lastly, recent data revealed that some probiotic bacteria or probiotic fermented milks improve the thymus status in a model of malnutrition, thus raising a new field for investigation, namely the thymus-gut connection, indicating that probiotics can be envisioned as a further adjuvant therapy in the control of thymic changes in undernutrition accompanied or not by infection.

9.
Front Immunol ; 13: 874064, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35757754

RESUMO

Autism spectrum disorder (ASD) is a heterogeneous neurodevelopmental disorder characterised by stereotyped behaviours, specific interests, and impaired communication skills. Elevated levels of pro-inflammatory cytokines, such as interleukin-17A (IL-17A or IL-17), have been implicated as part of immune alterations that may contribute to this outcome. In this context, rodent models have helped elucidate the role of T-cell activation and IL-17 secretion in the pathogenesis of ASD. Regarding the preclinical findings, the data available is contradictory in offspring but not in the pregnant dams, pointing to IL-17 as one of the main drivers of altered behaviour in some models ASD, whilst there are no alterations described in IL-17 levels in others. To address this gap in the literature, a systematic review of altered IL-17 levels in rodent models of ASD was conducted. In total, 28 studies that explored IL-17 levels were included and observed that this cytokine was generally increased among the different models of ASD. The data compiled in this review can help the choice of animal models to study the role of cytokines in the development of ASD, seeking a parallel with immune alterations observed in individuals with this condition. Systematic Review Registration: PROSPERO, identifier CRD42022306558.


Assuntos
Transtorno do Espectro Autista , Interleucina-17 , Animais , Citocinas , Feminino , Gravidez , Roedores
10.
PLoS Negl Trop Dis ; 16(2): e0010166, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35171909

RESUMO

The tropism of Zika virus (ZIKV) has been described in the nervous system, blood, placenta, thymus, and skeletal muscle. We investigated the mechanisms of skeletal muscle susceptibility to ZIKV using an in vitro model of human skeletal muscle myogenesis, in which myoblasts differentiate into myotubes. Myoblasts were permissive to ZIKV infection, generating productive viral particles, while myotubes controlled ZIKV replication. To investigate the underlying mechanisms, we used gene expression profiling. First, we assessed gene changes in myotubes compared with myoblasts in the model without infection. As expected, we observed an increase in genes and pathways related to the contractile muscle system in the myotubes, a reduction in processes linked to proliferation, migration and cytokine production, among others, confirming the myogenic capacity of our system in vitro. A comparison between non-infected and infected myoblasts revealed more than 500 differentially expressed genes (DEGs). In contrast, infected myotubes showed almost 2,000 DEGs, among which we detected genes and pathways highly or exclusively expressed in myotubes, including those related to antiviral and innate immune responses. Such gene modulation could explain our findings showing that ZIKV also invades myotubes but does not replicate in these differentiated cells. In conclusion, we showed that ZIKV largely (but differentially) disrupts gene expression in human myoblasts and myotubes. Identifying genes involved in myotube resistance can shed light on potential antiviral mechanisms against ZIKV infection.


Assuntos
Infecção por Zika virus , Zika virus , Antivirais/metabolismo , Feminino , Expressão Gênica , Humanos , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/metabolismo , Gravidez , Zika virus/fisiologia , Infecção por Zika virus/genética
11.
Front Cell Dev Biol ; 9: 668067, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33928093

RESUMO

The existence of a crosstalk between the nervous and immune systems is well established. Neurotransmitters can be produced by immune cells, whereas cytokines can be secreted by cells of nervous tissues. Additionally, cells of both systems express the corresponding receptors. Herein, we discuss the thymus as a paradigm for studies on the neuroimmune network. The thymus is a primary lymphoid organ responsible for the maturation of T lymphocytes. Intrathymic T-cell development is mostly controlled by the thymic microenvironment, formed by thymic epithelial cells (TEC), dendritic cells, macrophages, and fibroblasts. Developing thymocytes and microenvironmental cells can be influenced by exogenous and endogenous stimuli; neurotransmitters are among the endogenous molecules. Norepinephrine is secreted at nerve endings in the thymus, but are also produced by thymic cells, being involved in controlling thymocyte death. Thymocytes and TEC express acetylcholine receptors, but the cognate neurotransmitter seems to be produced and released by lymphoid and microenvironmental cells, not by nerve endings. Evidence indicates that, among others, TECs also produce serotonin and dopamine, as well as somatostatin, substance P, vasoactive intestinal peptide (VIP) and the typical pituitary neurohormones, oxytocin and arg-vasopressin. Although functional data of these molecules in the thymus are scarce, they are likely involved in intrathymic T cell development, as exemplified by somatostatin, which inhibits thymocyte proliferation, differentiation, migration and cytokine production. Overall, intrathymic neuroimmune interactions include various neurotransmitters, most of them of non-neuronal origin, and that should be placed as further physiological players in the general process of T-cell development.

12.
BMC Genomics ; 11 Suppl 5: S2, 2010 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-21210968

RESUMO

BACKGROUND: The thymus is a central lymphoid organ, in which bone marrow-derived T cell precursors undergo a complex process of maturation. Developing thymocytes interact with thymic microenvironment in a defined spatial order. A component of thymic microenvironment, the thymic epithelial cells, is crucial for the maturation of T-lymphocytes through cell-cell contact, cell matrix interactions and secretory of cytokines/chemokines. There is evidence that extracellular matrix molecules play a fundamental role in guiding differentiating thymocytes in both cortical and medullary regions of the thymic lobules. The interaction between the integrin α5ß1 (CD49e/CD29; VLA-5) and fibronectin is relevant for thymocyte adhesion and migration within the thymic tissue. Our previous results have shown that adhesion of thymocytes to cultured TEC line is enhanced in the presence of fibronectin, and can be blocked with anti-VLA-5 antibody. RESULTS: Herein, we studied the role of CD49e expressed by the human thymic epithelium. For this purpose we knocked down the CD49e by means of RNA interference. This procedure resulted in the modulation of more than 100 genes, some of them coding for other proteins also involved in adhesion of thymocytes; others related to signaling pathways triggered after integrin activation, or even involved in the control of F-actin stress fiber formation. Functionally, we demonstrated that disruption of VLA-5 in human TEC by CD49e-siRNA-induced gene knockdown decreased the ability of TEC to promote thymocyte adhesion. Such a decrease comprised all CD4/CD8-defined thymocyte subsets. CONCLUSION: Conceptually, our findings unravel the complexity of gene regulation, as regards key genes involved in the heterocellular cell adhesion between developing thymocytes and the major component of the thymic microenvironment, an interaction that is a mandatory event for proper intrathymic T cell differentiation.


Assuntos
Adesão Celular/fisiologia , Células Epiteliais/metabolismo , Regulação da Expressão Gênica/genética , Integrina alfa5/genética , Linfócitos T/fisiologia , Timo/citologia , Adesão Celular/genética , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Fibronectinas/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Integrina alfa5/metabolismo , Interferência de RNA
13.
Brain Behav Immun ; 24(3): 451-61, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19948213

RESUMO

Previous evidence indicated that growth hormone (GH) modulates cell migration in the thymus, and that extracellular matrix and chemokines are involved. Herein, we studied migration of peripheral lymphocytes derived from spleen and lymph nodes of GH-transgenic (GH-Tg) mice. We initially found that the relative cell numbers (normalized per gram of body weight) in lymph nodes and spleens from GH-Tg were higher at all ages tested (2-3, 7 and 12 months), as compared to wild type age-matched controls. Functionally, we found that lymphocyte migration triggered by laminin or fibronectin was enhanced in cells from GH-Tg versus control mice, independent of the organ from which the cells were derived (as ascertained in young adult animals). However, such an enhancement in migration was statistically significant only for CD4+ and CD8+ T cells from mesenteric lymph nodes. Migration of lymphocytes from mesenteric lymph nodes of GH-Tg mice, triggered by the chemokine CXCL12, in conjunction with laminin or fibronectin, was enhanced compared to lymphocytes from control mice. Rather surprisingly, the membrane levels of the corresponding extracellular matrix or chemokine receptors in peripheral lymphoid organs of GH-Tg mice did not necessarily correlate with the changes seen in migratory responses. In conclusion, our data show for the first time that GH alters lymphocyte migration in the periphery of the immune system. Considering that GH is used as an adjuvant therapeutic agent in immunodeficiencies, including AIDS, the concepts defined herein provide relevant background knowledge for future GH-related immune interventions.


Assuntos
Quimiocinas/metabolismo , Matriz Extracelular/fisiologia , Hormônio do Crescimento/genética , Hormônio do Crescimento/fisiologia , Linfócitos/fisiologia , Animais , Subpopulações de Linfócitos B/fisiologia , Movimento Celular , Quimiotaxia de Leucócito , Feminino , Fibronectinas/metabolismo , Citometria por Imagem , Imuno-Histoquímica , Linfonodos/patologia , Masculino , Camundongos , Camundongos Transgênicos , Baço/patologia , Subpopulações de Linfócitos T/fisiologia
14.
Cytokine Growth Factor Rev ; 18(1-2): 107-24, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17339126

RESUMO

Pathophysiology of Chagas' disease is not completely defined, although innate and adaptative immune responses are crucial. In acute infection some parasite antigens can activate macrophages, and this may result in pro-inflammatory cytokine production, nitric oxide synthesis, and consequent control of parasitemia and mortality. Cell-mediated immunity in Trypanosoma cruzi infection is also modulated by cytokines, but in addition to parasite-specific responses, autoimmunity can be also triggered. Importantly, cytokines may also play a role in the cell-mediated immunity of infected subjects. Finally, leukocyte influx towards target tissues is regulated by cytokines, chemokines, and extracellular matrix components which may represent potential therapeutic targets in infected patients. Here we will discuss recent findings on the role of cytokines, chemokines and extracellular matrix components in the regulation of innate and adaptive immunity during T. cruzi infection.


Assuntos
Moléculas de Adesão Celular/imunologia , Doença de Chagas/imunologia , Quimiocinas/imunologia , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Trypanosoma cruzi/imunologia , Animais , Doença de Chagas/fisiopatologia , Matriz Extracelular/imunologia , Humanos , Imunidade Celular , Imunidade Inata , Macrófagos/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Parasitemia/microbiologia , Parasitemia/parasitologia
15.
Sci Rep ; 9(1): 12962, 2019 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-31506501

RESUMO

In canine visceral leishmaniasis (CVL), splenic white pulp (SWP) disorganization has been associated with disease progression, reduced cytokine and chemokine expression and failure to control the parasite load. This profile is compatible with the cellular exhaustion previously shown in human visceral leishmaniasis. The present study aimed to evaluate the in situ expression of cellular exhaustion markers and their relation to clinical signs, SWP disorganization and parasite load. Forty dogs naturally infected by Leishmania infantum were grouped according to levels of SWP organization and parasite load. SWP disorganization was associated with reductions in the periarteriolar lymphatic sheath and lymphoid follicles/mm2 and worsening of the disease. Apoptotic cells expressing CTLA-4+ increased in dogs with disorganized SWP and a high parasite load. In the same group, PD-L1 and LAG-3 gene expression were reduced. A higher number of CD21+TIM-3+ B cells was detected in disorganized spleens than in organized spleens. Apoptosis is involved in periarteriolar lymphatic sheath reduction and lymphoid follicle atrophy and is associated with CTLA-4+ cell reductions in the splenic tissue of dogs with visceral leishmaniasis (VL). Failure to control the parasite load was observed, suggesting that cell exhaustion followed by T and B cell apoptosis plays a role in the immunosuppression observed in CVL.


Assuntos
Biomarcadores/análise , Doenças do Cão/imunologia , Leishmania infantum/imunologia , Leishmaniose Visceral/veterinária , Carga Parasitária , Baço/imunologia , Baço/parasitologia , Animais , Citocinas/metabolismo , Doenças do Cão/parasitologia , Doenças do Cão/patologia , Cães , Leishmaniose Visceral/imunologia , Leishmaniose Visceral/patologia , Baço/patologia
16.
Artigo em Inglês | MEDLINE | ID: mdl-31355153

RESUMO

Detrimental effects of malnutrition on immune responses to pathogens have long been recognized and it is considered a main risk factor for various infectious diseases, including visceral leishmaniasis (VL). Thymus is a target of both malnutrition and infection, but its role in the immune response to Leishmania infantum in malnourished individuals is barely studied. Because we previously observed thymic atrophy and significant reduction in cellularity and chemokine levels in malnourished mice infected with L. infantum, we postulated that the thymic microenvironment is severely compromised in those animals. To test this, we analyzed the microarchitecture of the organ and measured the protein abundance in its interstitial space in malnourished BALB/c mice infected or not with L. infantum. Malnourished-infected animals exhibited a significant reduction of the thymic cortex:medulla ratio and altered abundance of proteins secreted in the thymic interstitial fluid. Eighty-one percent of identified proteins are secreted by exosomes and malnourished-infected mice showed significant decrease in exosomal proteins, suggesting that exosomal carrier system, and therefore intrathymic communication, is dysregulated in those animals. Malnourished-infected mice also exhibited a significant increase in the abundance of proteins involved in lipid metabolism and tricarboxylic acid cycle, suggestive of a non-proliferative microenvironment. Accordingly, flow cytometry analysis revealed decreased proliferation of single positive and double positive T cells in those animals. Together, the reduced cortical area, decreased proliferation, and altered protein abundance suggest a dysfunctional thymic microenvironment where T cell migration, proliferation, and maturation are compromised, contributing for the thymic atrophy observed in malnourished animals. All these alterations could affect the control of the local and systemic infection, resulting in an impaired response to L. infantum infection.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Leishmania infantum/imunologia , Leishmaniose Visceral/imunologia , Desnutrição/imunologia , Linfócitos T/imunologia , Timo/imunologia , Animais , Transporte Biológico , Movimento Celular , Proliferação de Células , Ciclo do Ácido Cítrico/genética , Ciclo do Ácido Cítrico/imunologia , Exossomos/imunologia , Exossomos/metabolismo , Exossomos/parasitologia , Líquido Extracelular/imunologia , Líquido Extracelular/metabolismo , Líquido Extracelular/parasitologia , Galectina 1/genética , Galectina 1/imunologia , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Imunidade Inata , Leishmania infantum/crescimento & desenvolvimento , Leishmaniose Visceral/genética , Leishmaniose Visceral/metabolismo , Leishmaniose Visceral/parasitologia , Metabolismo dos Lipídeos , Masculino , Desnutrição/genética , Desnutrição/metabolismo , Desnutrição/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Plasminogênio/genética , Plasminogênio/imunologia , Proteoma/genética , Proteoma/imunologia , Linfócitos T/parasitologia , Timo/metabolismo , Timo/parasitologia
17.
Cell Adh Migr ; 12(2): 152-167, 2018 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-28494186

RESUMO

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.


Assuntos
Moléculas de Adesão Celular/metabolismo , Células Epiteliais/metabolismo , Epitélio/metabolismo , Sinapses Imunológicas/metabolismo , Integrina alfa6/genética , Adesão Celular/fisiologia , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Matriz Extracelular/metabolismo , Marcação de Genes/métodos , Humanos , Integrinas/metabolismo
19.
Sci Rep ; 7: 45991, 2017 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-28397794

RESUMO

Protein malnutrition, the most deleterious cause of malnutrition in developing countries, has been considered a primary risk factor for the development of clinical visceral leishmaniasis (VL). Protein malnutrition and infection with Leishmania infantum leads to lymphoid tissue disorganization, including changes in cellularity and lymphocyte subpopulations in the thymus and spleen. Here we report that protein malnutrition modifies thymic chemotactic factors by diminishing the CCL5, CXCL12, IGF1, CXCL9 and CXCL10 protein levels in infected animals. Nevertheless, T cells preserve their migratory capability, as they were able to migrate ex vivo in response to chemotactic stimuli, indicating that malnutrition may compromise the thymic microenvironment and alter in vivo thymocyte migration. Decrease in chemotactic factors protein levels was accompanied by an early increase in the parasite load of the spleen. These results suggest that the precondition of malnutrition is affecting the cell-mediated immune response to L. infantum by altering T cell migration and interfering with the capacity of protein-deprived animals to control parasite spreading and proliferation. Our data provide evidence for a disturbance of T lymphocyte migration involving both central and peripheral T-cells, which likely contribute to the pathophysiology of VL that occurs in malnourished individuals.


Assuntos
Movimento Celular , Leishmania infantum/patogenicidade , Leishmaniose Visceral/complicações , Leishmaniose Visceral/imunologia , Desnutrição/complicações , Desnutrição/imunologia , Linfócitos T/patologia , Timo/patologia , Animais , Apoptose , Atrofia , Peso Corporal , Quimiotaxia , Citocinas/sangue , Fator de Crescimento Insulin-Like I/metabolismo , Leishmaniose Visceral/sangue , Leishmaniose Visceral/parasitologia , Leptina/sangue , Ligantes , Macrófagos/metabolismo , Macrófagos/patologia , Desnutrição/sangue , Desnutrição/parasitologia , Camundongos Endogâmicos BALB C , Carga Parasitária , Parasitos/patogenicidade , Receptores CXCR3/metabolismo , Baço/parasitologia , Timócitos/patologia
20.
Microbes Infect ; 8(1): 221-31, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16239119

RESUMO

It is currently accepted that experimental acute infection by Trypanosoma cruzi promotes changes in secondary lymphoid organs, with general T and B lymphocyte polyclonal activation. Here we show that mesenteric lymph nodes (MLN) of acutely infected mice show severe atrophy due to extensive lymphocyte apoptosis. Accordingly, clusters of apoptotic cells are detected in the initial phase of infection in MLN but not in subcutaneous nodes. Moreover, such atrophy is independent of the infection route, parasite load or the mouse strain used. Studies in Fas-L deficient (BALB gld/gld+/+) and in TNF type 1 receptor (p55-/-) knockout mice indicate that both molecules are involved in MLN atrophy: Fas-L participates in cell death of CD4+ as well as B lymphocytes, whereas the TNF type 1 receptor is important for the apoptosis of CD4+ and CD8+ T lymphocytes. In contrast, perforin does not play a role, as lymph nodes from perforin-deficient mice do not behave differently from the corresponding wild types. Our data support the concept that, even in a systemic infection, differential (even opposing) responses can be found in different lymph node chains.


Assuntos
Doença de Chagas/metabolismo , Linfonodos/metabolismo , Linfonodos/patologia , Glicoproteínas de Membrana/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Fatores de Necrose Tumoral/metabolismo , Animais , Apoptose , Atrofia/metabolismo , Linfócitos B/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Doença de Chagas/patologia , Proteína Ligante Fas , Deleção de Genes , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas Citotóxicas Formadoras de Poros , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptor fas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA