Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Front Immunol ; 13: 940577, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36248873

RESUMO

Background: Aging is usually accompanied by functional declines of the immune system, especially in T-cell responses. However, little is known about ways to alleviate this. Methods: Here, 37 middle-aged healthy participants were recruited, among which 32 were intravenously administrated with expanded NK cells and 5 with normal saline. Then, we monitored changes of peripheral senescent and exhausted T cells within 4 weeks after infusion by flow cytometry, as well as serum levels of senescence-associated secretory phenotype (SASP)-related factors. In vitro co-culture assays were performed to study NK-mediated cytotoxic activity against senescent or exhausted T cells. Functional and phenotypic alteration of NK cells before and after expansion was finally characterized. Results: After NK cell infusion, senescent CD28-, CD57+, CD28-CD57+, and CD28-KLRG1+ CD4+ and CD8+ T-cell populations decreased significantly, so did PD-1+ and TIM-3+ T cells. These changes were continuously observed for 4 weeks. Nevertheless, no significant changes were observed in the normal saline group. Moreover, SASP-related factors including IL-6, IL-8, IL-1α, IL-17, MIP-1α, MIP-1ß, and MMP1 were significantly decreased after NK cell infusion. Further co-culture assays showed that expanded NK cells specifically and dramatically eliminated senescent CD4+ T cells other than CD28+CD4+ T cells. They also showed improved cytotoxic activity, with different expression patterns of activating and inhibitory receptors including NKG2C, NKG2A, KLRG1, LAG3, CD57, and TIM3. Conclusion: Our findings imply that T-cell senescence and exhaustion is a reversible process in healthy individuals, and autologous NK cell administration can be introduced to alleviate the aging. Clinical Trial Registration: ClinicalTrials.gov, ChiCTR-OOh-17011878.


Assuntos
Antígenos CD28 , Receptor Celular 2 do Vírus da Hepatite A , Antígenos CD28/metabolismo , Quimiocina CCL3/metabolismo , Quimiocina CCL4/metabolismo , Receptor Celular 2 do Vírus da Hepatite A/metabolismo , Interleucina-17/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Células Matadoras Naturais , Metaloproteinase 1 da Matriz/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Ensaios Clínicos Controlados Aleatórios como Assunto , Solução Salina/metabolismo
2.
Front Genet ; 12: 671682, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34194471

RESUMO

Ankylosing spondylitis (AS) is a common, highly heritable inflammatory arthritis affecting the mainly axial joints in both East Asia and Europe. To date, the pathogenesis of AS is still unknown, although we know that genetics play a vital role in it. The HLA-B27 allele is found in over 85% of AS patients. However, strong evidence suggests that other major histocompatibility complex (MHC) and non-MHC genes are also involved in the pathogenesis. In addition, current data showed that there were significant differences in both genomics and metagenomics among the different ethnic populations. The investigation of the key role of the microbiome in AS pathogenesis also highlighted the host-microbiome genetic interactions. Here, we systematically review current AS genetic research data and further compare genetic differences, especially between East Asian and European groups, which may highlight the challenge in future genetic studies.

3.
Cell Physiol Biochem ; 47(5): 2046-2055, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29969781

RESUMO

BACKGROUND/AIMS: Hepatocellular carcinoma (HCC) is one of the most common human malignant diseases in the world, and the mechanisms underlying HCC carcinogenesis and progression need further investigation. MicroRNAs play important roles in the development of cancer, and miR-500a is suggested to be deregulated in some types of cancer. However, the underlying molecular mechanisms of miR-500a in HCC remain unknown. METHODS: The expression of miR-500a in HCC was analyzed in The Cancer Genome Atlas (TCGA) database and examined in 33 pairs of HCC tissues and matched nontumor tissues. The correlation between miR-500a expression and prognosis of HCC patients was analyzed from the survival data in TCGA. The mechanism of miR-500a upregulation in HCC was detected using chromatin immunoprecipitation-quantitative real-time PCR. The roles of miR-500a in HCC development were examined using a cell counting kit-8 assay in vitro and growth of transplanted tumors in nude mice in vivo. Apoptosis of HCC was detected using Annexin V/propidium iodide staining. The expression of BH3-interacting death agonist (BID) protein was examined using western blot analysis. RESULTS: miR-500a expression was upregulated in HCC tissues, and high miR-500a expression was significantly correlated with the poor prognosis of HCC patients. Histone modifications in the promoter region of miR-500a may be responsible for its increased expression. Inhibition of miR-500a in HCC cell lines significantly promoted apoptosis, as well as inhibiting the proliferation of HCC cells and growth of transplanted tumors in nude mice. miR-500a directly targeted the 3' untranslated region of BID mRNA, and inhibition of miR-500a-promoted apoptosis was almost completely abolished by the administration of ABT-199 via the BID-mitochondria pathway. CONCLUSION: Our results suggest that histone modifications in the promoter region of miR-500a may be responsible for the increased expression of miR-500a in HCC, which promotes cancer progression by targeting BID, indicating that miR-500a may be a potential prognostic predictor and therapeutic target for HCC patients.


Assuntos
Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Carcinoma Hepatocelular/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/metabolismo , MicroRNAs/metabolismo , Proteínas de Neoplasias/metabolismo , RNA Neoplásico/metabolismo , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Células HEK293 , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , MicroRNAs/genética , Proteínas de Neoplasias/genética , RNA Neoplásico/genética
4.
Biochem Cell Biol ; 96(5): 636-645, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29671340

RESUMO

Indirubin and isatin have been used in the treatment of inflammatory diseases due to their anti-inflammatory properties. This study aimed to evaluate the combined effect of indirubin and isatin on dextran sulfate sodium (DSS)-induced ulcerative colitis (UC). UC was induced by the administration of 3% (w/v) DSS solution, and then the model mice were administered indirubin (10 mg/kg body mass) and (or) isatin (10 mg/kg body mass) by gavage once daily for 7 days. The results showed that indirubin and isatin, individually or combined, significantly inhibited weight loss, lowered disease activity index (DAI), ameliorated pathological changes, decreased the levels of pro-inflammatory mediators and myeloperoxidase (MPO) activity, increased the expression of anti-inflammatory cytokines and Foxp3, suppressed CD4+ T cell infiltration, and inhibited oxidative stress and epithelial cell apoptosis. Additionally, indirubin and isatin, both individually and combined, can also inhibit activation of the NF-κB and MAPK pathways induced by DSS. The protective effect of combination therapy against UC was superior to that of single-agent treatment. These results suggest that indirubin combined with isatin attenuates DSS-induced UC, and changes to the NF-κB and MAPK signaling pathways may mediate the protective effects of indirubin and isatin in UC.


Assuntos
Linfócitos T CD4-Positivos , Colite Ulcerativa , Sulfato de Dextrana/toxicidade , Isatina/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Colite Ulcerativa/induzido quimicamente , Colite Ulcerativa/tratamento farmacológico , Colite Ulcerativa/imunologia , Colite Ulcerativa/patologia , Relação Dose-Resposta a Droga , Células Epiteliais/imunologia , Células Epiteliais/patologia , Indóis/farmacologia , Sistema de Sinalização das MAP Quinases/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/imunologia
5.
Int J Clin Exp Pathol ; 7(2): 509-20, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24551271

RESUMO

Previously, we demonstrated that Tim-1-Fc prevents acute cardiac graft rejection by inhibiting Th1 response. In the present report, we tackled the impact of Tim-1-Fc on Th17 cells in a model of cardiac chronic rejection. Administration of Tim-1-Fc did not result in a detectable impact on innate immunity and regulatory T cells, while it provided protection for Bm12-derive cardiac grafts against chronic rejection in B6 recipients, as manifested by the reduction of inflammatory infiltration along with less severity of vasculopathy. Studies in T-bet(-/-) recipients by implanting Bm12-derived cardiac grafts further revealed that Tim-1-Fc significantly protected cardiac grafts from chronic rejection along with attenuated production of IL-17 producing T cells. Depletion of CD4 and CD8 T cells or blockade of IL-17 in T-bet(-/-) recipients demonstrated that Tim-1-Fc selectively suppresses Th17 differentiation along with attenuated IL-17 secretion. Together, our data suggest that Tim-1-Fc protects cardiac grafts from chronic rejection by suppressing CD4 Th17 development and functionality. Therefore, Tim-1-Fc might be a potential immunosuppressive agent in the setting of cardiac transplantation.


Assuntos
Rejeição de Enxerto/prevenção & controle , Transplante de Coração/efeitos adversos , Fragmentos Fc das Imunoglobulinas/farmacologia , Imunossupressores/farmacologia , Interleucina-17/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Miocárdio/metabolismo , Células Th17/efeitos dos fármacos , Aloenxertos , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Doença Crônica , Técnicas de Cocultura , Modelos Animais de Doenças , Regulação para Baixo , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/metabolismo , Rejeição de Enxerto/patologia , Sobrevivência de Enxerto/efeitos dos fármacos , Receptor Celular 1 do Vírus da Hepatite A , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/imunologia , Miocárdio/patologia , Proteínas com Domínio T/deficiência , Proteínas com Domínio T/genética , Células Th17/imunologia , Células Th17/metabolismo , Fatores de Tempo
6.
Int J Clin Exp Pathol ; 6(9): 1702-12, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24040435

RESUMO

Sinomenine (SIN) is a purified alkaloid from the Chinese herb Sinomenium acutum. Previous studies demonstrated that SIN possesses anti-inflammatory and anti-apoptotic properties. We thus in the present report conducted studies to examine its impact on ischemia reperfusion (IR) induced renal injury. Precondition of mice with 200 mg/kg of SIN provided significant protection for mice against IR-induced renal injury as manifested by the attenuated serum creatinine (Cre) and blood urea nitrogen (BUN) along with less severity for histological changes and tubular cell apoptosis. In line with these results, treatment of mice with SIN suppressed IR-induced inflammatory infiltration and the expression of chemokine CXCL-10, adhesion molecule ICAM-1, and cytokines TNF-а/IL-6. Mechanistic studies revealed that SIN inhibits NF-κB transcriptional activity to suppress IR-induced inflammatory response in the kidney, while it attenuates MAP kinase signaling to prevent tubular cells undergoing apoptosis after IR insult. Altogether, our data support that SIN could be a useful therapeutic agent for prevention and treatment of IR-induced renal injury in the clinical settings.


Assuntos
Anti-Inflamatórios/farmacologia , Apoptose/efeitos dos fármacos , Mediadores da Inflamação/metabolismo , Túbulos Renais/efeitos dos fármacos , Morfinanos/farmacologia , Nefrite/prevenção & controle , Traumatismo por Reperfusão/prevenção & controle , Animais , Biomarcadores/sangue , Nitrogênio da Ureia Sanguínea , Quimiocina CXCL10/metabolismo , Creatinina/sangue , Citoproteção , Modelos Animais de Doenças , Molécula 1 de Adesão Intercelular/metabolismo , Interleucina-6/metabolismo , Túbulos Renais/imunologia , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Nefrite/imunologia , Nefrite/metabolismo , Nefrite/patologia , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo
7.
J Neuroimmunol ; 260(1-2): 55-9, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23623189

RESUMO

Despite increasing evidence highlighting the role of NPY in the modulation of inflammatory reaction, surprisingly little is known about the direct effects of NPY on the release of proinflammatory mediators. In the present work, we have evaluated the effects of NPY on the release of TNF-α, IL-1ß, IL-6 and HMGB1 mediators in peritoneal macrophages. Our results demonstrate for the first time that NPY can directly induce active HMGB1 release and cytoplasmic translocation, while the production of TNF-α, IL-1ß and IL-6 is not affected. PKC and ERK pathway inhibitors can abolish the promotive effect of NPY on HMGB1 secretion. Thus, our results indicate that NPY might impact on the innate immune system by directly potentiating the HMGB1 release from the macrophage.


Assuntos
Proteína HMGB1/metabolismo , Sistema de Sinalização das MAP Quinases/imunologia , Macrófagos/metabolismo , Neuroimunomodulação/fisiologia , Neuropeptídeo Y/metabolismo , Proteína Quinase C/metabolismo , Animais , Arginina/análogos & derivados , Arginina/farmacologia , Butadienos/farmacologia , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Proteína HMGB1/imunologia , Macrófagos/citologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neuropeptídeo Y/antagonistas & inibidores , Neuropeptídeo Y/imunologia , Nitrilas/farmacologia , Proteína Quinase C/imunologia , Receptores de Neuropeptídeo Y/antagonistas & inibidores , Receptores de Neuropeptídeo Y/metabolismo , Estaurosporina/farmacologia
8.
Front Biosci (Elite Ed) ; 4(4): 1303-13, 2012 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-22201956

RESUMO

Tanshinone IIA (Tan IIA), an active component derived from Salvia miltiorrhiza root, has been used to treat various ischemic cardiovascular and cerebrovascular diseases. However, its impact on hepatic ischemia/reperfusion (I/R) injury remains unclear. Here, we addressed this issue by using a 90-minute partial liver ischemia model. Mice were administered Tan IIA intragastrically for 3 days before ischemia and were assessed for liver damage 6-h after reperfusion. Tan IIA pretreatment significantly inhibited serum aminotransferases and proinflammatory cytokine levels along with reduced inflammatory infiltration and liver damage. Mechanistic studies revealed that Tan IIA suppressed TLR4 expression in nonparenchymal cells (NPCs) and induced heme oxygenase-1 (HO-1) production in both parenchymal and NPCs. Moreover, the phosphorylation of AKT and ERK1/2 in the liver was enhanced, while the phosphorylation of JNK, p38 and p65 was suppressed. These results suggest Tan IIA can suppress TLR4 signaling which then enhances HO-1 expression along with reduced proinflammatory cytokine expressions in the liver, and Tan IIA could be a useful candidate drug in clinic for prevention and treatment of hepatic I/R injury.


Assuntos
Abietanos/uso terapêutico , Fígado/irrigação sanguínea , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Sequência de Bases , Western Blotting , Primers do DNA , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase em Tempo Real
9.
Chin Med J (Engl) ; 124(19): 3127-32, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22040567

RESUMO

BACKGROUND: The immunomodulatory effects of glucocorticoids (GCs) have been described as bimodal. High concentration of GCs exerts immunosuppressive effects and low levels of GCs are immunopermissive. While the immunosuppressive mechanisms of GCs have been investigated intensely, the immunopermissive effects of GCs remain unclear. A lot of studies showed GCs could exert rapid non-genomic actions. We herein studied the rapid immunopromoting effects of GCs. METHODS: We observed the rapid (within 30 minutes) effects of corticosterone on respiratory burst of mouse peritoneal macrophages and studied their mechanisms. The superoxide anions were measured by cytochrome C reduction assay. Protein kinase C phosphorylation was measured by Western blotting and membrane fluidity was evaluated by fluorescence polarization measurement. RESULTS: The 10(-8) mol/L and 10(-7) mol/L corticosterone rapidly increased the superoxide anions production by macrophages, which were insensitive to GC-receptor antagonist, mifepristone, and protein-synthesis inhibitor, cycloheximide. Corticosterone coupled to bovine serum albumin was able to mimic the effects of corticosterone. The effects were independent of protein kinase C pathway and the change in membrane fluidity. CONCLUSIONS: The results indicate that corticosterone rapidly promote the superoxide anions production by mouse peritoneal macrophages may through non-genomic mechanisms. This study may contribute to understanding the effects of GCs under stress condition and the physiological significance of nongenomic effects of GCs.


Assuntos
Corticosterona/farmacologia , Macrófagos Peritoneais/fisiologia , Explosão Respiratória/efeitos dos fármacos , Animais , Macrófagos Peritoneais/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Superóxidos/metabolismo
10.
PLoS One ; 6(7): e21697, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21750723

RESUMO

Engagement of T-cell immunoglobulin mucin (Tim)-1 on T cells with its ligand, Tim-4, on antigen presenting cells delivers positive costimulatory signals to T cells. However, the molecular mechanisms for Tim-1-mediated regulation of T-cell activation and differentiation are relatively poorly understood. Here we investigated the role of Tim-1 in T-cell responses and allograft rejection using recombinant human Tim-1 extracellular domain and IgG1-Fc fusion proteins (Tim-1-Fc). In vitro assays confirmed that Tim-1-Fc selectively binds to CD4(+) effector T cells, but not dendritic cells or natural regulatory T cells (nTregs). Tim-1-Fc was able to inhibit the responses of purified CD4(+) T cells that do not express Tim-4 to stimulation by anti-CD3/CD28 mAbs, and this inhibition was associated with reduced AKT and ERK1/2 phosphorylation, but it had no influence on nTregs. Moreover, Tim-1-Fc inhibited the proliferation of CD4(+) T cells stimulated by allogeneic dendritic cells. Treatment of recipient mice with Tim-1-Fc significantly prolonged cardiac allograft survival in a fully MHC-mismatched strain combination, which was associated with impaired Th1 response and preserved Th2 and nTregs function. Importantly, the frequency of Foxp3(+) cells in splenic CD4(+) T cells was increased, thus shifting the balance toward regulators, even though Tim-1-Fc did not induce Foxp3 expression in CD4(+)CD25(-) T cells directly. These results indicate that Tim-1-Fc can inhibit T-cell responses through an unknown Tim-1 binding partner on T cells, and it is a promising immunosuppressive agent for preventing allograft rejection.


Assuntos
Rejeição de Enxerto/imunologia , Fragmentos Fc das Imunoglobulinas/imunologia , Glicoproteínas de Membrana/imunologia , Receptores Virais/imunologia , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Citometria de Fluxo , Fatores de Transcrição Forkhead/imunologia , Fatores de Transcrição Forkhead/metabolismo , Rejeição de Enxerto/prevenção & controle , Transplante de Coração/imunologia , Receptor Celular 1 do Vírus da Hepatite A , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Proteína Quinase 1 Ativada por Mitógeno/imunologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/imunologia , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/imunologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Virais/genética , Receptores Virais/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Transplante Homólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA