Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Nat Neurosci ; 27(6): 1116-1124, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38637622

RESUMO

Alzheimer's disease (AD) and dementia in general are age-related diseases with multiple contributing factors, including brain inflammation. Microglia, and specifically those expressing the AD risk gene TREM2, are considered important players in AD, but their exact contribution to pathology remains unclear. In this study, using high-throughput mass cytometry in the 5×FAD mouse model of amyloidosis, we identified senescent microglia that express high levels of TREM2 but also exhibit a distinct signature from TREM2-dependent disease-associated microglia (DAM). This senescent microglial protein signature was found in various mouse models that show cognitive decline, including aging, amyloidosis and tauopathy. TREM2-null mice had fewer microglia with a senescent signature. Treating 5×FAD mice with the senolytic BCL2 family inhibitor ABT-737 reduced senescent microglia, but not the DAM population, and this was accompanied by improved cognition and reduced brain inflammation. Our results suggest a dual and opposite involvement of TREM2 in microglial states, which must be considered when contemplating TREM2 as a therapeutic target in AD.


Assuntos
Envelhecimento , Doença de Alzheimer , Encéfalo , Modelos Animais de Doenças , Glicoproteínas de Membrana , Microglia , Receptores Imunológicos , Animais , Receptores Imunológicos/metabolismo , Receptores Imunológicos/genética , Microglia/metabolismo , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/genética , Camundongos , Envelhecimento/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Camundongos Transgênicos , Senescência Celular/fisiologia , Senescência Celular/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Knockout
2.
Cell Rep Med ; 4(11): 101278, 2023 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-37944529

RESUMO

The choroid plexus (CP) plays a key role in remotely controlling brain function in health, aging, and disease. Here, we report that CP epithelial cells express the brain-specific cholesterol 24-hydroxylase (CYP46A1) and that its levels are decreased under different mouse and human brain conditions, including amyloidosis, aging, and SARS-CoV-2 infection. Using primary mouse CP cell cultures, we demonstrate that the enzymatic product of CYP46A1, 24(S)-hydroxycholesterol, downregulates inflammatory transcriptomic signatures within the CP, found here to be elevated across multiple neurological conditions. In vitro, the pro-inflammatory cytokine tumor necrosis factor α (TNF-α) downregulates CYP46A1 expression, while overexpression of CYP46A1 or its pharmacological activation in mouse CP organ cultures increases resilience to TNF-α. In vivo, overexpression of CYP46A1 in the CP in transgenic mice with amyloidosis is associated with better cognitive performance and decreased brain inflammation. Our findings suggest that CYP46A1 expression in the CP impacts the role of this niche as a guardian of brain immune homeostasis.


Assuntos
Amiloidose , Plexo Corióideo , Humanos , Camundongos , Animais , Colesterol 24-Hidroxilase/metabolismo , Plexo Corióideo/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Encéfalo/patologia , Homeostase/fisiologia , Camundongos Transgênicos , Amiloidose/metabolismo , Amiloidose/patologia
3.
Nat Commun ; 14(1): 1293, 2023 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-36894557

RESUMO

Systemic immunity supports lifelong brain function. Obesity posits a chronic burden on systemic immunity. Independently, obesity was shown as a risk factor for Alzheimer's disease (AD). Here we show that high-fat obesogenic diet accelerated recognition-memory impairment in an AD mouse model (5xFAD). In obese 5xFAD mice, hippocampal cells displayed only minor diet-related transcriptional changes, whereas the splenic immune landscape exhibited aging-like CD4+ T-cell deregulation. Following plasma metabolite profiling, we identified free N-acetylneuraminic acid (NANA), the predominant sialic acid, as the metabolite linking recognition-memory impairment to increased splenic immune-suppressive cells in mice. Single-nucleus RNA-sequencing revealed mouse visceral adipose macrophages as a potential source of NANA. In vitro, NANA reduced CD4+ T-cell proliferation, tested in both mouse and human. In vivo, NANA administration to standard diet-fed mice recapitulated high-fat diet effects on CD4+ T cells and accelerated recognition-memory impairment in 5xFAD mice. We suggest that obesity accelerates disease manifestation in a mouse model of AD via systemic immune exhaustion.


Assuntos
Doença de Alzheimer , Camundongos , Humanos , Animais , Doença de Alzheimer/metabolismo , Ácido N-Acetilneuramínico , Camundongos Transgênicos , Transtornos da Memória/etiologia , Obesidade/complicações , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças
4.
Curr Opin Immunol ; 76: 102182, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35576769

RESUMO

For decades, neurodegenerative diseases were thought to be caused by the accumulation of toxic compounds, exacerbated by local inflammation, which together lead to neuronal loss and cognitive impairment . An additional factor that was long overlooked , is the role of the systemic immune system, which provides a defense mechanism against internal and external intruders in all bodily tissues. The evolving understanding of the life-long cross-talk between the CNS and the immune system led to an awareness of the function of systemic adaptive immunity in containing emerging destructive factors within the brain. . This includes harnessing of circulating myeloid cells to help the brain. However, as damage accumulates within the brain, the systemic immune system loses its protective capacity. Under such conditions, the dysregulated immune system becomes an escalating factor itself, thereby driving a vicious cycle that must be arrested.


Assuntos
Doenças Neurodegenerativas , Imunidade Adaptativa , Encéfalo , Humanos , Sistema Imunitário , Inflamação
5.
Nat Aging ; 2(1): 60-73, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-37118355

RESUMO

Microglia and monocyte-derived macrophages (MDM) are key players in dealing with Alzheimer's disease. In amyloidosis mouse models, activation of microglia was found to be TREM2 dependent. Here, using Trem2-/-5xFAD mice, we assessed whether MDM act via a TREM2-dependent pathway. We adopted a treatment protocol targeting the programmed cell death ligand-1 (PD-L1) immune checkpoint, previously shown to modify Alzheimer's disease via MDM involvement. Blockade of PD-L1 in Trem2-/-5xFAD mice resulted in cognitive improvement and reduced levels of water-soluble amyloid beta1-42 with no effect on amyloid plaque burden. Single-cell RNA sequencing revealed that MDM, derived from both Trem2-/- and Trem2+/+5xFAD mouse brains, express a unique set of genes encoding scavenger receptors (for example, Mrc1, Msr1). Blockade of monocyte trafficking using anti-CCR2 antibody completely abrogated the cognitive improvement induced by anti-PD-L1 treatment in Trem2-/-5xFAD mice and similarly, but to a lesser extent, in Trem2+/+5xFAD mice. These results highlight a TREM2-independent, disease-modifying activity of MDM in an amyloidosis mouse model.


Assuntos
Doença de Alzheimer , Amiloidose , Camundongos , Animais , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Camundongos Transgênicos , Macrófagos/metabolismo , Amiloidose/genética , Glicoproteínas de Membrana/genética , Receptores Imunológicos/genética
6.
Mol Neurodegener ; 16(1): 39, 2021 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-34172073

RESUMO

BACKGROUND: For decades, dementia has been characterized by accumulation of waste in the brain and low-grade inflammation. Over the years, emerging studies highlighted the involvement of the immune system in neurodegenerative disease emergence and severity. Numerous studies in animal models of amyloidosis demonstrated the beneficial role of monocyte-derived macrophages in mitigating the disease, though less is known regarding tauopathy. Boosting the immune system in animal models of both amyloidosis and tauopathy, resulted in improved cognitive performance and in a reduction of pathological manifestations. However, a full understanding of the chain of events that is involved, starting from the activation of the immune system, and leading to disease mitigation, remained elusive. Here, we hypothesized that the brain-immune communication pathway that is needed to be activated to combat tauopathy involves monocyte mobilization via the C-C chemokine receptor 2 (CCR2)/CCL2 axis, and additional immune cells, such as CD4+ T cells, including FOXP3+ regulatory CD4+ T cells. METHODS: We used DM-hTAU transgenic mice, a mouse model of tauopathy, and applied an approach that boosts the immune system, via blocking the inhibitory Programmed cell death protein-1 (PD-1)/PD-L1 pathway, a manipulation previously shown to alleviate disease symptoms and pathology. An anti-CCR2 monoclonal antibody (αCCR2), was used to block the CCR2 axis in a protocol that partially eliminates monocytes from the circulation at the time of anti-PD-L1 antibody (αPD-L1) injection, and for the critical period of their recruitment into the brain following treatment. RESULTS: Performance of DM-hTAU mice in short-term and working memory tasks, revealed that the beneficial effect of αPD-L1, assessed 1 month after a single injection, was abrogated following blockade of CCR2. This was accompanied by the loss of the beneficial effect on disease pathology, assessed by measurement of cortical aggregated human tau load using Homogeneous Time Resolved Fluorescence-based immunoassay, and by evaluation of hippocampal neuronal survival. Using both multiparametric flow cytometry, and Cytometry by Time Of Flight, we further demonstrated the accumulation of FOXP3+ regulatory CD4+ T cells in the brain, 12 days following the treatment, which was absent subsequent to CCR2 blockade. In addition, measurement of hippocampal levels of the T-cell chemoattractant, C-X-C motif chemokine ligand 12 (Cxcl12), and of inflammatory cytokines, revealed that αPD-L1 treatment reduced their expression, while blocking CCR2 reversed this effect. CONCLUSIONS: The CCR2/CCL2 axis is required to modify pathology using PD-L1 blockade in a mouse model of tauopathy. This modification involves, in addition to monocytes, the accumulation of FOXP3+ regulatory CD4+ T cells in the brain, and the T-cell chemoattractant, Cxcl12.


Assuntos
Quimiocina CCL2/metabolismo , Receptores CCR2/metabolismo , Tauopatias/imunologia , Tauopatias/metabolismo , Animais , Linfócitos T CD4-Positivos/imunologia , Quimiocina CCL2/imunologia , Modelos Animais de Doenças , Inibidores de Checkpoint Imunológico/farmacologia , Camundongos , Camundongos Transgênicos , Monócitos/imunologia , Receptores CCR2/imunologia , Tauopatias/patologia
7.
Nat Commun ; 10(1): 465, 2019 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-30692527

RESUMO

Alzheimer's disease (AD) is a heterogeneous disorder with multiple etiologies. Harnessing the immune system by blocking the programmed cell death receptor (PD)-1 pathway in an amyloid beta mouse model was shown to evoke a sequence of immune responses that lead to disease modification. Here, blocking PD-L1, a PD-1 ligand, was found to have similar efficacy to that of PD-1 blocking in disease modification, in both animal models of AD and of tauopathy. Targeting PD-L1 in a tau-driven disease model resulted in increased immunomodulatory monocyte-derived macrophages within the brain parenchyma. Single cell RNA-seq revealed that the homing macrophages expressed unique scavenger molecules including macrophage scavenger receptor 1 (MSR1), which was shown here to be required for the effect of PD-L1 blockade in disease modification. Overall, our results demonstrate that immune checkpoint blockade targeting the PD-1/PD-L1 pathway leads to modification of common factors that go awry in AD and dementia, and thus can potentially provide an immunotherapy to help combat these diseases.


Assuntos
Antígeno B7-H1/metabolismo , Disfunção Cognitiva/metabolismo , Macrófagos/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Tauopatias/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Anticorpos Bloqueadores/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/imunologia , Encéfalo/imunologia , Encéfalo/metabolismo , Disfunção Cognitiva/genética , Modelos Animais de Doenças , Humanos , Macrófagos/imunologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Transgênicos , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Tauopatias/genética
9.
Nat Commun ; 6: 7967, 2015 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-26284939

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disorder in which chronic neuroinflammation contributes to disease escalation. Nevertheless, while immunosuppressive drugs have repeatedly failed in treating this disease, recruitment of myeloid cells to the CNS was shown to play a reparative role in animal models. Here we show, using the 5XFAD AD mouse model, that transient depletion of Foxp3(+) regulatory T cells (Tregs), or pharmacological inhibition of their activity, is followed by amyloid-ß plaque clearance, mitigation of the neuroinflammatory response and reversal of cognitive decline. We further show that transient Treg depletion affects the brain's choroid plexus, a selective gateway for immune cell trafficking to the CNS, and is associated with subsequent recruitment of immunoregulatory cells, including monocyte-derived macrophages and Tregs, to cerebral sites of plaque pathology. Our findings suggest targeting Treg-mediated systemic immunosuppression for treating AD.


Assuntos
Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Linfócitos T Reguladores/metabolismo , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Giro Denteado/patologia , Giro Denteado/fisiologia , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica/fisiologia , Acetato de Glatiramer/farmacologia , Tolerância Imunológica , Imunomodulação , Camundongos , Camundongos Transgênicos , RNA/genética , RNA/metabolismo
10.
Proc Natl Acad Sci U S A ; 110(6): 2264-9, 2013 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-23335631

RESUMO

The adaptive arm of the immune system has been suggested as an important factor in brain function. However, given the fact that interactions of neurons or glial cells with T lymphocytes rarely occur within the healthy CNS parenchyma, the underlying mechanism is still a mystery. Here we found that at the interface between the brain and blood circulation, the epithelial layers of the choroid plexus (CP) are constitutively populated with CD4(+) effector memory cells with a T-cell receptor repertoire specific to CNS antigens. With age, whereas CNS specificity in this compartment was largely maintained, the cytokine balance shifted in favor of the T helper type 2 (Th2) response; the Th2-derived cytokine IL-4 was elevated in the CP of old mice, relative to IFN-γ, which decreased. We found this local cytokine shift to critically affect the CP epithelium, triggering it to produce the chemokine CCL11 shown to be associated with cognitive dysfunction. Partial restoration of cognitive ability in aged mice, by lymphopenia-induced homeostasis-driven proliferation of memory T cells, was correlated with restoration of the IL-4:IFN-γ ratio at the CP and modulated the expression of plasticity-related genes at the hippocampus. Our data indicate that the cytokine milieu at the CP epithelium is affected by peripheral immunosenescence, with detrimental consequences to the aged brain. Amenable to immunomodulation, this interface is a unique target for arresting age-related cognitive decline.


Assuntos
Envelhecimento/imunologia , Envelhecimento/patologia , Encéfalo/imunologia , Encéfalo/patologia , Plexo Corióideo/imunologia , Plexo Corióideo/patologia , Células Th2/imunologia , Células Th2/patologia , Imunidade Adaptativa , Animais , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/patologia , Proliferação de Células , Epitélio/imunologia , Epitélio/patologia , Hipocampo/imunologia , Hipocampo/patologia , Memória Imunológica , Linfopenia/imunologia , Linfopenia/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuroimunomodulação , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Receptor de Interferon gama
11.
J Cell Mol Med ; 14(10): 2470-82, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19650830

RESUMO

Amyotrophic lateral sclerosis (ALS) is a devastating disease, characterized by extremely rapid loss of motor neurons. Our studies over the last decade have established CD4(+) T cells as important players in central nervous system maintenance and repair. Those results, together with recent findings that CD4(+) T cells play a protective role in mouse models of ALS, led us to the current hypothesis that in ALS, a rapid T-cell malfunction may develop in parallel to the motor neuron dysfunction. Here, we tested this hypothesis by assessing thymic function, which serves as a measure of peripheral T-cell availability, in an animal model of ALS (mSOD1 [superoxide dismutase] mice; G93A) and in human patients. We found a significant reduction in thymic progenitor-cell content, and abnormal thymic histology in 3-4-month-old mSOD1 mice. In ALS patients, we found a decline in thymic output, manifested in the reduction in blood levels of T-cell receptor rearrangement excision circles, a non-invasive measure of thymic function, and demonstrated a restricted T-cell repertoire. The morbidity of the peripheral immune cells was also manifested in the increase of pro-apoptotic BAX/BCXL2 expression ratio in peripheral blood mononuclear cells (PBMCs) of these patients. In addition, gene expression screening in the same PBMCs, revealed in the ALS patients a reduction in key genes known to be associated with T-cell activity, including: CD80, CD86, IFNG and IL18. In light of the reported beneficial role of T cells in animal models of ALS, the present observation of thymic dysfunction, both in human patients and in an animal model, might be a co-pathological factor in ALS, regardless of the disease aetiology. These findings may lead to the development of novel therapeutic approaches directed at overcoming the thymic defect and T-cell deficiency.


Assuntos
Esclerose Lateral Amiotrófica/imunologia , Esclerose Lateral Amiotrófica/patologia , Perfilação da Expressão Gênica , Timo/fisiopatologia , Adulto , Animais , Linfócitos T CD4-Positivos/imunologia , Modelos Animais de Doenças , Feminino , Rearranjo Gênico do Linfócito T , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Timo/imunologia
12.
J Immunol ; 179(1): 41-4, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17579019

RESUMO

LPS, a molecule produced by Gram-negative bacteria, is known to activate both innate immune cells such as macrophages and adaptive immune B cells via TLR4 signaling. Although TLR4 is also expressed on T cells, LPS was observed not to affect T cell proliferation or cytokine secretion. We now report, however, that LPS can induce human T cells to adhere to fibronectin via TLR4 signaling. This response to LPS was confirmed in mouse T cells; functional TLR4 and MyD88 were required, but T cells from TLR2 knockout mice could respond to LPS. The human T cell response to LPS depended on protein kinase C signaling and involved the phosphorylation of the proline-rich tyrosine kinase (Pyk-2) and p38. LPS also up-regulated the T cell expression of suppressor of cytokine signaling 3, which led to inhibition of T cell chemotaxis toward the chemokine stromal cell-derived factor 1alpha (CXCL12). Thus, LPS, through TLR4 signaling, can affect T cell behavior in inflammation.


Assuntos
Imunidade Inata , Lipopolissacarídeos/farmacologia , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Receptor 4 Toll-Like/fisiologia , Animais , Adesão Celular/imunologia , Inibição de Migração Celular , Movimento Celular/imunologia , Quimiocina CXCL12 , Quimiocinas CXC/antagonistas & inibidores , Quimiocinas CXC/fisiologia , Quimiotaxia de Leucócito/imunologia , Feminino , Fibronectinas/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/fisiologia , Linfócitos T/citologia , Linfócitos T/metabolismo , Receptor 4 Toll-Like/antagonistas & inibidores
13.
Eur J Immunol ; 36(11): 2971-80, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17051618

RESUMO

A peptide (hCDR1) based on the complementarity determining region-1 of an anti-DNA antibody ameliorates systemic lupus erythematosus (SLE) in induced and spontaneous lupus models. Our objectives were to determine the effects of hCDR1 on TCR signaling and on its negative regulators, Foxj1 and Foxo3a. BALB/c mice were immunized with the SLE-inducing anti-DNA antibody, designated 16/6Id, and treated with hCDR1. hCDR1 treatment specifically inhibited IFN-gamma secretion by T cells in association with down-regulated T-bet expression and NF-kappaB activation; however, GATA-3 expression was not affected. Furthermore, TCR signaling (ZAP-70 phosphorylation) was inhibited, and the mRNA expression of the two modulators of Th1 activation, Foxj1 and Foxo3a, was significantly up-regulated. The latter were also elevated in SLE-afflicted (NZBxNZW)F1 mice that were treated with hCDR1. Addition of TGF-beta, which was elevated following treatment with hCDR1, to T cells from 16/6Id immunized mice, up-regulated Foxj1 and Foxo3a mRNA expression, similarly to hCDR1. In contrast, anti-TGF-beta antibodies added to hCDR1-treated T cells abrogated its effect. Thus, hCDR1 elevates TGF-beta, which contributes to the up-regulation of T cell Foxj1 and Foxo3a expression, leading to inhibition of NF-kappaB activation and IFN-gamma secretion, which is required for the maintenance of SLE.


Assuntos
Anticorpos Monoclonais/farmacologia , Fatores de Transcrição Forkhead/metabolismo , Interferon gama/antagonistas & inibidores , Lúpus Eritematoso Sistêmico/imunologia , Fragmentos de Peptídeos/farmacologia , Linfócitos T/efeitos dos fármacos , Animais , Feminino , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/genética , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/metabolismo , Peptídeos/farmacologia , Fosforilação/efeitos dos fármacos , RNA Mensageiro/metabolismo , Receptores de Antígenos de Linfócitos T/antagonistas & inibidores , Receptores de Antígenos de Linfócitos T/metabolismo , Proteínas com Domínio T/metabolismo , Linfócitos T/imunologia , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Regulação para Cima , Proteína-Tirosina Quinase ZAP-70/metabolismo
14.
J Clin Invest ; 116(7): 2022-32, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16767222

RESUMO

CD4+CD25+ Tregs regulate immunity, but little is known about their own regulation. We now report that the human 60-kDa heat shock protein (HSP60) acts as a costimulator of human Tregs, both CD4+CD25int and CD4+CD25hi. Treatment of Tregs with HSP60, or its peptide p277, before anti-CD3 activation significantly enhanced the ability of relatively low concentrations of the Tregs to downregulate CD4+CD25- or CD8+ target T cells, detected as inhibition of target T cell proliferation and IFN-gamma and TNF-alpha secretion. The enhancing effects of HSP60 costimulation on Tregs involved innate signaling via TLR2, led to activation of PKC, PI3K, and p38, and were further enhanced by inhibition of ERK. HSP60-treated Tregs suppressed target T cells both by cell-to-cell contact and by secretion of TGF-beta and IL-10. In addition, the expression of ERK, NF-kappaB, and T-bet by downregulated target T cells was inhibited. Thus, HSP60, a self-molecule, can downregulate adaptive immune responses by upregulating Tregs innately through TLR2 signaling.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Chaperonina 60/metabolismo , Receptores de Interleucina-2/metabolismo , Transdução de Sinais/fisiologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/metabolismo , Receptor 2 Toll-Like/metabolismo , Animais , Complexo CD3/metabolismo , Células Cultivadas , Chaperonina 60/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Quinase 2 de Adesão Focal/metabolismo , Humanos , Interferon gama/metabolismo , Interleucina-10/metabolismo , Camundongos , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas com Domínio T , Fatores de Transcrição/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
FASEB J ; 20(3): 547-9, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16396993

RESUMO

Chondroitin sulfate proteoglycan (CSPG), a matrix protein that occurs naturally in the central nervous system (CNS), is considered to be a major inhibitor of axonal regeneration and is known to participate in activation of the inflammatory response. The degradation of CSPG by a specific enzyme, chondroitinase ABC, promotes repair. We postulated that a disaccharidic degradation product of this glycoprotein (CSPG-DS), generated following such degradation, participates in the modulation of the inflammatory responses and can, therefore, promote recovery in immune-induced neuropathologies of the CNS, such as experimental autoimmune encephalomyelitis (EAE) and experimental autoimmune uveitis (EAU). In these pathologies, the dramatic increase in T cells infiltrating the CNS is far in excess of the numbers needed for regular maintenance. Here, we show that CSPG-DS markedly alleviated the clinical symptoms of EAE and protected against the neuronal loss in EAU. The last effect was associated with a reduction in the numbers of infiltrating T cells and marked microglia activation. This is further supported by our in vitro results indicating that CSPG-DS attenuated T cell motility and decreased secretion of the cytokines interferon-gamma and tumor necrosis factor-alpha. Mechanistically, these effects are associated with an increase in SOCS-3 levels and a decrease in NF-kappaB. Our results point to a potential therapeutic modality, in which a compound derived from an endogenous CNS-resident molecule, known for its destructive role in CNS recovery, might be helpful in overcoming inflammation-induced neurodegenerative conditions.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Doenças Autoimunes/tratamento farmacológico , Proteoglicanas de Sulfatos de Condroitina/química , Proteoglicanas de Sulfatos de Condroitina/uso terapêutico , Dissacarídeos/uso terapêutico , Encefalomielite Autoimune Experimental/tratamento farmacológico , Fatores Imunológicos/uso terapêutico , Degeneração Neural/prevenção & controle , Células Ganglionares da Retina/efeitos dos fármacos , Uveíte/tratamento farmacológico , Sequência de Aminoácidos , Animais , Anti-Inflamatórios não Esteroides/isolamento & purificação , Anti-Inflamatórios não Esteroides/farmacologia , Apoptose/efeitos dos fármacos , Doenças Autoimunes/complicações , Doenças Autoimunes/patologia , Adesão Celular , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/imunologia , Células Cultivadas/metabolismo , Quimiotaxia/efeitos dos fármacos , Proteoglicanas de Sulfatos de Condroitina/isolamento & purificação , Proteoglicanas de Sulfatos de Condroitina/farmacologia , Citocinas/metabolismo , Dissacarídeos/isolamento & purificação , Dissacarídeos/farmacologia , Avaliação Pré-Clínica de Medicamentos , Encefalomielite Autoimune Experimental/complicações , Encefalomielite Autoimune Experimental/patologia , Feminino , Humanos , Hipersensibilidade Tardia/tratamento farmacológico , Hipersensibilidade Tardia/prevenção & controle , Fatores Imunológicos/isolamento & purificação , Fatores Imunológicos/farmacologia , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/patologia , Dados de Sequência Molecular , NF-kappa B/metabolismo , Degeneração Neural/etiologia , Ratos , Ratos Endogâmicos Lew , Células Ganglionares da Retina/patologia , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/biossíntese , Proteínas Supressoras da Sinalização de Citocina/genética , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Uveíte/complicações , Uveíte/patologia
16.
J Immunol ; 175(11): 7255-63, 2005 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16301630

RESUMO

Systemic lupus erythematosus (SLE), which is characterized by the increased production of autoantibodies and defective T cell responses, can be induced in mice by immunization with a human anti-DNA mAb that expresses a major Id, designated 16/6Id. A peptide based on the sequence of the CDR1 of the 16/6Id (human CDR1 (hCDR1)) ameliorated the clinical manifestations of SLE and down-regulated, ex vivo, the 16/6Id-induced T cell proliferation. In this study, we examined the mechanism responsible for the hCDR1-induced modulation of T cell functions related to the pathogenesis of SLE. We found that injection of hCDR1 into BALB/c mice concomitant with their immunization with 16/6Id resulted in a marked elevation of TGF-beta secretion 10 days later. Addition of TGF-beta suppressed the 16/6Id-stimulated T cell proliferation similarly to hCDR1. In addition, we provide evidence that one possible mechanism underlying the hCDR1- and TGFbeta-induced inhibition of T cell proliferation is by down-regulating the expression, and therefore the functions, of a pair of key cell adhesion receptors, LFA-1 (alphaLbeta2) and CD44, which operate as accessory molecules in mediating APC-T cell interactions. Indeed, T cells of mice treated with hCDR1 showed a TGF-beta-induced suppression of adhesion to the LFA-1 and CD44 ligands, hyaluronic acid and ICAM-1, respectively, induced by stromal cell-derived factor-1alpha and PMA. The latter suppression is through the inhibition of ERK phosphorylation. Thus, the down-regulation of SLE-associated responses by hCDR1 treatment may be due to the effect of the up-regulated TGF-beta on the expression and function of T cell adhesion receptors and, consequently, on T cell stimulation, adhesion, and proliferation.


Assuntos
Anticorpos Antinucleares/imunologia , Autoimunidade/imunologia , Receptores de Hialuronatos/imunologia , Antígeno-1 Associado à Função Linfocitária/imunologia , Linfócitos T/imunologia , Fator de Crescimento Transformador beta/imunologia , Animais , Anticorpos Monoclonais/imunologia , Adesão Celular/imunologia , Comunicação Celular/imunologia , Proliferação de Células , Regiões Determinantes de Complementaridade/imunologia , Modelos Animais de Doenças , Citometria de Fluxo , Humanos , Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/metabolismo , Idiótipos de Imunoglobulinas , Molécula 1 de Adesão Intercelular/metabolismo , Lúpus Eritematoso Sistêmico/imunologia , Ativação Linfocitária/imunologia , Antígeno-1 Associado à Função Linfocitária/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Peptídeos/imunologia , Fator de Crescimento Transformador beta/metabolismo
17.
J Immunol ; 174(1): 302-9, 2005 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-15611253

RESUMO

Systemic lupus erythematosus (SLE) can be induced in mice by immunizing them with a monoclonal human anti-DNA Ab that expresses a major Id, designated 16/6Id. In addition, a peptide based on the sequence of the CDR 1 (hCDR1) of the 16/6Id ameliorated the clinical manifestations of SLE in experimental models. In this study we examined the effects of treating mice with human complementary-determining region 1 (hCDR1) on the subsequent chemotaxis of T cells derived from 16/6Id-primed mice. First we demonstrated elevated levels of stromal cell-derived factor-1alpha (SDF-1alpha) in the sera of SLE-afflicted mice and in the sera and lymphoid tissues of 16/6Id-immunized BALB/c mice shortly after the immunization. We then found that administration of hCDR1 to 16/6Id-immunized mice specifically down-regulated SDF1alpha-induced T cell chemotaxis through fibronectin and collagen type I. This was accompanied by diminished SDF1-alpha-induced T cell adhesion and ERK phosphorylation. Treatment with hCDR1 up-regulated TGF-beta secretion, which, in turn, inhibited the murine T cell adhesion to and chemotaxis through fibronectin as well as their ERK phosphorylation. Thus, the secretion of TGF-beta after treatment of 16/6Id-immunized mice with hCDR1 plays an important role in the down-regulation of SDF-1alpha-mediated T cell activation and the interactions with extracellular matrix moieties observed in the present study.


Assuntos
Anticorpos Antinucleares/imunologia , Quimiocinas CXC/imunologia , Quimiotaxia de Leucócito/imunologia , Regiões Determinantes de Complementaridade/imunologia , Linfócitos T/imunologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Western Blotting , Adesão Celular/imunologia , Quimiocina CXCL12 , Quimiocinas CXC/farmacologia , Quimiotaxia de Leucócito/efeitos dos fármacos , Modelos Animais de Doenças , Regulação para Baixo , Fibronectinas/metabolismo , Humanos , Lúpus Eritematoso Sistêmico/imunologia , Ativação Linfocitária/imunologia , Camundongos , Peptídeos/imunologia , Regulação para Cima
18.
Eur J Neurosci ; 20(8): 1973-83, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15450076

RESUMO

Chondroitin sulphate proteoglycan (CSPG) inhibits axonal regeneration in the central nervous system (CNS) and its local degradation promotes repair. We postulated that the enzymatic degradation of CSPG generates reparative products. Here we show that an enzymatic degradation product of CSPG, a specific disaccharide (CSPG-DS), promoted CNS recovery by modulating both neuronal and microglial behaviour. In neurons, acting via a mechanism that involves the PKCalpha and PYK2 intracellular signalling pathways, CSPG-DS induced neurite outgrowth and protected against neuronal toxicity and axonal collapse in vitro. In microglia, via a mechanism that involves ERK1/2 and PYK2, CSPG-DS evoked a response that allowed these cells to manifest a neuroprotective phenotype ex vivo. In vivo, systemically or locally injected CSPG-DS protected neurons in mice subjected to glutamate or aggregated beta-amyloid intoxication. Our results suggest that treatment with CSPG-DS might provide a way to promote post-traumatic recovery, via multiple cellular targets.


Assuntos
Sistema Nervoso Central/efeitos dos fármacos , Proteoglicanas de Sulfatos de Condroitina/farmacologia , Dissacarídeos/farmacologia , Regeneração Nervosa/efeitos dos fármacos , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Sistema Nervoso Central/citologia , Sistema Nervoso Central/fisiologia , Dissacarídeos/isolamento & purificação , Relação Dose-Resposta a Droga , Camundongos , Camundongos Endogâmicos C57BL , Regeneração Nervosa/fisiologia , Técnicas de Cultura de Órgãos , Células PC12 , Ratos , Ratos Endogâmicos Lew , Ratos Wistar
19.
Clin Dev Immunol ; 11(2): 143-9, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15330450

RESUMO

Traumeel S (Traumeel), a mixture of highly diluted (10(-1)-10(-9)) extracts from medicinal plants and minerals is widely used in humans to relieve trauma, inflammation and degenerative processes. However, little is known about its possible effects on the behavior of immune cells. The effects of Traumeel were examined in vitro on the ability of resting and PHA-, PMA- or TNF-alpha-activated human T cells, monocytes, and gut epithelial cells to secrete the prototypic pro-inflammatory mediators IL-1beta, TNF-alpha and IL-8 over a period of 24-72 h. Traumeel inhibited the secretion of all three agents in resting, as well as activated immune cells. IL-beta secretion was reduced by up to 70% in both resting and activated cells; TNF-alpha secretion was reduced by up to 65 and 54%, respectively, and IL-8 secretion was reduced by 50% in both resting and activated cells (P < 0.01 for all cells). Interestingly, the effect appeared to be inversely dose-related; maximal inhibition (usually 30-60% inhibition; P < 0.01) was seen with dilutions of 10(-3)-10(-6) of the Traumeel stock material. This finding suggests that Traumeel does not inhibit immune cells functions by exerting a toxic effect. Indeed, Traumeel did not affect T cell and monocyte proliferation. Although additional studies are needed to clarify the mode of action of Traumeel and to demonstrate causative relationship between the inhibition of cytokine/chemokine secretion in cell culture and the reported clinical effects of the preparation, our in vitro results offer a mechanism for the anti-inflammatory effects of Traumeel observed in clinical use.


Assuntos
Interleucina-1/metabolismo , Ativação Linfocitária , Minerais/farmacologia , Extratos Vegetais/farmacologia , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Linhagem Celular , Células Cultivadas , Técnicas de Cocultura , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Interleucina-8/metabolismo , Linfócitos T/metabolismo , Acetato de Tetradecanoilforbol/farmacologia
20.
J Immunol ; 172(9): 5185-93, 2004 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15100255

RESUMO

During their migration into inflammatory sites, immune cells, such as T cells, secrete extracellular matrix (ECM)-degrading enzymes, such as heparanase, which, under mildly acidic conditions, degrade heparan sulfate proteoglycans (HSPG). We have previously shown that at pH 7.2, human placental heparanase loses its enzymatic activity, while retaining its ability to bind HSPG and promote T cell adhesion to unfractionated ECM. We now demonstrate that the 65-kDa recombinant human heparanase, which is devoid of enzymatic activity, but can still bind HSPG, captures T cells under shear flow conditions and mediates their rolling and arrest, in the absence or presence of stromal cell-derived factor 1 alpha (SDF-1 alpha; CXCL12), in an alpha(4)beta(1)-VCAM-1-dependent manner. Furthermore, heparanase binds to and induces T cell adhesion to key ECM components, like fibronectin and hyaluronic acid, in beta(1) integrin- and CD44-specific manners, respectively, via the activation of the protein kinase C and phosphatidylinositol 3-kinase intracellular signaling machineries. Although the nature of the putative T cell heparanase-binding moiety is unknown, it appears that heparanase exerts its proadhesive activity by interacting with the T cells' surface HSPG, because pretreatment of the cells with heparinase abolished their subsequent response to heparanase. Also, heparanase augmented the SDF-1 alpha-triggered phosphorylation of Pyk-2 and extracellular signal-regulated kinase-2 implicated in integrin functioning. Moreover, heparanase, which had no chemotactic effect on T cells on its own, augmented the SDF-1 alpha-induced T cell chemotaxis across fibronectin. These findings add another dimension to the known versatility of heparanase as a key regulator of T cell activities during inflammation, both in the context of the vasculature and at extravascular sites.


Assuntos
Adjuvantes Imunológicos/fisiologia , Comunicação Celular/imunologia , Matriz Extracelular/enzimologia , Matriz Extracelular/imunologia , Glucuronidase/fisiologia , Proteínas Recombinantes/farmacologia , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/fisiologia , Molécula 1 de Adesão de Célula Vascular/fisiologia , Adjuvantes Imunológicos/metabolismo , Adesão Celular/imunologia , Comunicação Celular/fisiologia , Linhagem Celular , Células Cultivadas , Quimiocina CXCL12 , Quimiocinas CXC/farmacologia , Quimiotaxia de Leucócito/imunologia , Colágeno Tipo IV/fisiologia , Matriz Extracelular/fisiologia , Fibronectinas/metabolismo , Fibronectinas/fisiologia , Quinase 2 de Adesão Focal , Glucuronidase/metabolismo , Humanos , Ácido Hialurônico/fisiologia , Interfase/imunologia , Migração e Rolagem de Leucócitos/imunologia , Ativação Linfocitária , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Proteínas Tirosina Quinases/metabolismo , Proteínas Recombinantes/metabolismo , Transdução de Sinais/imunologia , Especificidade por Substrato/imunologia , Subpopulações de Linfócitos T/enzimologia , Molécula 1 de Adesão de Célula Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA