Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Viral Immunol ; 37(1): 24-35, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38301135

RESUMO

Interleukin-36 (IL-36) signaling plays an important role in promoting CD8+ T cell-mediated antitumor immune responses. The role of IL-36 signaling in CD8+ T cells that are involved in host immune responses during human immunodeficiency virus-1 (HIV-1) infection has not been characterized. Sixty-one patients living with chronic HIV-1 infection and 23 controls were enrolled in this study. The levels of IL-36 cytokine family members were measured by enzyme-linked immunosorbent assay. Purified CD8+ T cells were stimulated with recombinant IL-36gamma (1 or 10 ng/mL). The expression of inhibitory receptors, the secretion of cytotoxic molecules and interferon-gamma, and the mRNA levels of apoptosis-related ligands were assessed to evaluate the effect of IL-36gamma on CD8+ T cell function in vitro. There were no significant differences in IL-36alpha, IL-36beta, or IL-36 receptor antagonist levels between patients living with chronic HIV-1 infection and controls. Plasma IL-36gamma levels were reduced in patients living with chronic HIV-1 infection. Perforin, granzyme B, and granulysin secretion, as well as tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and Fas ligand (FasL) mRNA expression, but not programmed death-1 (PD-1) or cytotoxic T lymphocyte-associated protein-4 (CTLA-4) expression was downregulated in CD8+ T cells from patients living with chronic HIV-1 infection. The addition of both 1 and 10 ng/mL IL-36gamma enhanced perforin, granzyme B, granulysin, and interferon-gamma secretion by CD8+ T cells without affecting PD-1/CTLA-4 or TRAIL/FasL mRNA expression in CD8+ T cells from patients living with chronic HIV-1 infection. The addition of 1 ng/mL IL-36gamma also promoted perforin and granzyme B secretion by HIV-1-specific CD8+ T cells from patients living with chronic HIV-1 infection. The reduced IL-36gamma levels in patients living with chronic HIV-1 infection might be insufficient for the activation of CD8+ T cells, leading to CD8+ T cell exhaustion.


Assuntos
Linfócitos T CD8-Positivos , Infecções por HIV , Humanos , Antígeno CTLA-4 , Granzimas/farmacologia , HIV , Interferon gama , Interleucinas/farmacologia , Perforina/farmacologia , Receptor de Morte Celular Programada 1 , RNA Mensageiro
2.
J Cereb Blood Flow Metab ; 44(3): 367-383, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37974301

RESUMO

The crosstalk between reactive astrocytes and infiltrated immune cells plays a critical role in maintaining blood-brain barrier (BBB) integrity. However, how astrocytes interact with immune cells and the effect of their interaction on BBB integrity after hemorrhagic stroke are still unclear. By performing RNA sequencing in astrocytes that were activated by interleukin-1α (IL-1α), tumor necrosis factor α (TNFα), and complement component 1q (C1q) treatment, we found CCL5 was among the top upregulated genes. Immunostaining and western blot results demonstrated that CCL5 was increased in mice brain after hemorrhagic stroke. Flow cytometry showed that knockout of astrocytic CCL5 reduced the infiltration of CD8+ but not CD4+ T and myeloid cells into the brain (p < 0.05). In addition, knockout CCL5 in astrocytes increased tight junction-related proteins ZO-1 and Occludin expression; reduced Evans blue leakage, perforin and granzyme B expression; improved neurobehavioral outcomes in hemorrhagic stroke mice (p < 0.05), while transplantation of CD8+ T cells reversed these protective effects. Moreover, co-culture of CD8+ T cells with bEnd.3 cells induced the apoptosis of bEnd.3 cells, which was rescued by inhibiting perforin. In conclusion, our study suggests that CCL5 mediated crosstalk between astrocytes and CD8+ T cells represents an important therapeutic target for protecting BBB in stroke.


Assuntos
Barreira Hematoencefálica , Quimiocina CCL5 , Acidente Vascular Cerebral Hemorrágico , Animais , Camundongos , Astrócitos/metabolismo , Barreira Hematoencefálica/metabolismo , Linfócitos T CD8-Positivos , Comunicação Celular , Células Endoteliais/metabolismo , Acidente Vascular Cerebral Hemorrágico/metabolismo , Perforina/metabolismo , Perforina/farmacologia , Quimiocina CCL5/metabolismo
3.
Eur J Med Res ; 28(1): 460, 2023 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-37880804

RESUMO

BACKGROUND: Hypomethylation of the perforin gene promoter in CD4 + T cells, inflammation and oxidative stress, might be involved in alveolar septal cell apoptosis associated with emphysema in rats. This study aimed to investigate the effects of S-adenosylmethionine (SAM) on this kind of apoptosis in rats with autoimmune emphysema. METHODS: Twenty-four rats were randomly divided into three groups: a normal control group, a model group, and a SAM group. Pathological changes in lung tissues were observed, and the mean linear intercept (MLI) and mean alveolar number (MAN) were measured. The levels of anti-endothelial cell antibodies (AECA) in serum, alveolar septal cell apoptosis, perforin gene promotor methylation in CD4 + T cells in the spleen, and the levels of cytokines, malondialdehyde (MDA), and glutathione (GSH) and the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) in bronchoalveolar lavage fluid (BALF) were investigated. RESULTS: The MLI, apoptosis index (AI) of alveolar septal cells, levels of AECA in serum, and levels of tumour necrosis factor-α (TNF-α), matrix metalloproteinase-9 (MMP-9) and MDA in BALF were increased, while the MAN, methylation levels, and the activities of GSH, SOD and GSH-Px in BALF were decreased in the model group compared with those in the normal control group and the SAM group (all P < 0.05). The levels of interleukin-8 (IL-8) in BALF were greater in the model group than in the normal control group (P < 0.05). CONCLUSIONS: SAM protects against alveolar septal cell apoptosis, airway inflammation and oxidative stress in rats with autoimmune emphysema possibly by partly reversing the hypomethylation of the perforin gene promoter in CD4 + T cells.


Assuntos
Enfisema , Enfisema Pulmonar , Humanos , Ratos , Animais , S-Adenosilmetionina/farmacologia , Ratos Sprague-Dawley , Perforina/farmacologia , Enfisema Pulmonar/patologia , Pulmão/patologia , Enfisema/patologia , Apoptose , Glutationa/farmacologia , Inflamação/patologia , Superóxido Dismutase
4.
Hematology ; 28(1): 2166333, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36651499

RESUMO

OBJECTIVE: This experiment will explore the role of TIGIT/PVR signaling pathway in the pathogenesis of MDS immune tolerance through in vitro co-culture of NK cells and MDSC cells. METHODS: Flow cytometry was used to detect the expression percentage of MDSCs and CD155 on MDSCs in the bone marrow of MDS patients and controls. The expression of NK cell surface receptors (NKG2D, NKp30, NKp46), secreted cytokines (perforin, granzyme B, CD107a, IFN-γ) and NK cell apoptosis rate were detected by flow cytometry to evaluate the effect of MDSCs on NK cell function. RESULTS: The number of MDSCs in bone marrow of MDS patients was notably higher than that of the control group (8.39 ± 7.01 vs 2.31 ± 1.65, P = 0.0001). Compared with the control group, the expression of CD155 on MDSCs in MDS group was increased (31.81 ± 21.33 vs. 10.49 ± 6.53, P < 0.0001). After NK cells were co-cultured with MDSCs, NKG2D, NKp30, NKp46, CD107a, IFN-γ, perforin and granzyme B were decreased, and the NK function partially recovered after the addition of inhibitors. CONCLUSION: Compared with the normal control, MDSCs and CD155 on MDSCs were highly expressed in MDS patients. After co-culture with MDSCs, the expression of NK cells' surface receptors decreased, the secretion of cytokines decreased and the apoptosis rate increased. After blocking TIGIT/CD155 pathway, NK cell function was reversed, but NK cell apoptosis was not reduced.


Assuntos
Síndromes Mielodisplásicas , Células Supressoras Mieloides , Humanos , Células Supressoras Mieloides/metabolismo , Granzimas/metabolismo , Granzimas/farmacologia , Perforina/metabolismo , Perforina/farmacologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Células Matadoras Naturais , Síndromes Mielodisplásicas/metabolismo , Citocinas , Receptores Imunológicos/metabolismo
5.
Life Sci Alliance ; 5(11)2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36271507

RESUMO

CD3 bispecific antibodies (bsAbs) show great promise as anticancer therapeutics. Here, we show in-depth mechanistic studies of a CD3 bsAb in solid cancer, using DuoBody-CD3x5T4. Cross-linking T cells with tumor cells expressing the oncofetal antigen 5T4 was required to induce cytotoxicity. Naive and memory CD4+ and CD8+ T cells were equally effective at mediating cytotoxicity, and DuoBody-CD3x5T4 induced partial differentiation of naive T-cell subsets into memory-like cells. Tumor cell kill was associated with T-cell activation, proliferation, and production of cytokines, granzyme B, and perforin. Genetic knockout of FAS or IFNGR1 in 5T4+ tumor cells abrogated tumor cell kill. In the presence of 5T4+ tumor cells, bystander kill of 5T4- but not of 5T4-IFNGR1- tumor cells was observed. In humanized xenograft models, DuoBody-CD3x5T4 antitumor activity was associated with intratumoral and peripheral blood T-cell activation. Lastly, in dissociated patient-derived tumor samples, DuoBody-CD3x5T4 activated tumor-infiltrating lymphocytes and induced tumor-cell cytotoxicity, even when most tumor-infiltrating lymphocytes expressed PD-1. These data provide an in-depth view on the mechanism of action of a CD3 bsAb in preclinical models of solid cancer.


Assuntos
Anticorpos Biespecíficos , Neoplasias , Humanos , Anticorpos Biespecíficos/farmacologia , Linfócitos T CD8-Positivos , Granzimas/farmacologia , Complexo CD3/farmacologia , Citotoxicidade Imunológica , Perforina/farmacologia , Receptor de Morte Celular Programada 1 , Neoplasias/tratamento farmacológico , Citocinas
6.
Biotechnol Prog ; 38(5): e3279, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35661450

RESUMO

As a water-soluble macromolecule polysaccharide, xanthan gum (XG) has several biological activities, such as antitumor, antiviral, and immunomodulatory function. However, the effect of XG on the proliferation and cytotoxicity of cytokines induced killer (CIK) cells is rarely studied. In this study, the effect of XG on CIK cells derived from peripheral blood was investigated by analyzing the expansion fold of total cells, phenotype, cytotoxicity, degranulation, and apoptosis in serum-free medium. The results showed that the expansion fold of total cells with 100 µg/ml XG which molecule weight is 2.95 × 106 Da reached 4534.0 folds, significantly higher than that without XG (1299.0 folds, p < 0.05). The percentage of main effector cells-CD3+ CD56+ cells increased to 25.5% and the cytotoxic activity of CIK cells increased to 45.3%. The cell proportions of expression granzyme B and perforin that related to cytotoxicity in CIK cells reached 53.6% and 48.3%, respectively, significantly higher than 27.5% and 37.5% in the group without XG (p < 0.05). Collectively, XG could stimulate the ex vivo expansion of CIK cells and enhance the cytotoxicity of expanded CIK cells. The above results provide technical support for optimizing the expansion process of CIK cells ex vivo.


Assuntos
Células Matadoras Induzidas por Citocinas , Antivirais/farmacologia , Células Cultivadas , Células Matadoras Induzidas por Citocinas/metabolismo , Granzimas/metabolismo , Granzimas/farmacologia , Perforina/metabolismo , Perforina/farmacologia , Polissacarídeos Bacterianos , Fator de Necrose Tumoral alfa , Água/metabolismo
7.
Phytomedicine ; 104: 154206, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35724525

RESUMO

BACKGROUND: Immune activation, chronic inflammation, and renal interstitial fibrosis (RIF) are associated with chronic kidney disease (CKD). The herbal formula, Shenkang injection (SKI), has been reported to attenuate RIF. However, the mechanisms by which SKI alleviates renal fibrosis, especially the role of natural killer (NK) cells, are unknown and require exploration. PURPOSE: This study aimed to determine the mechanisms by which SKI alleviates RIF. METHODS: Differential gene expression between CKD mice and control groups was explored using bioinformatics analysis. To reveal how SKI reduces RIF in CKD, a CKD mouse model was established using folic acid for in vivo studies, and human kidney-2 cells were used for in vitro experiments. The effects of various SKI doses were then determined. Immunohistochemical staining, Enzyme-linked immunosorbent assay, western blotting, and quantitative real-time PCR were used for pathological and molecular expression detection. RESULTS: We first investigated the potential immune dysfunction in CKD using bioinformatics analysis. Some differentially expressed genes were enriched in immune-related functions. The expressions of perforin and interferon (IFN)-γ, which are mainly released by NK cells, were significantly higher in patients with CKD (p< 0.05). In vivo experiments showed that SKI alleviated renal fibrosis in a folic acid-induced renal fibrosis model. Serum creatinine and blood urea nitrogen levels were reduced in the high-dose SKI-treated group. Additionally, the mRNA and protein expression levels of type IV collagen and alpha-spinal muscular atrophy were reduced. Biochemical detection showed that SKI could also downregulate the activity of NK cells (by decreasing the expressions of perforin and IFN-γ). Increased levels of stimulator of interferon genes (STING)/TANK-binding kinase 1 (TBK1)/IFN regulatory factor 3 (IRF3), phosphorylation of TBK1, and IRF3 in FA-induced RIF mice were alleviated by SKI treatment, which was consistent with the results of in vitro experiments. CONCLUSION: These results demonstrated that SKI could decrease the activation of NK cells via the STING/TBK1/IRF3 signaling pathway, thereby alleviating RIF and protecting renal function in CKD. This may provide valuable evidence supporting the clinical use of SKI in the treatment of patients with CKD.


Assuntos
Fator Regulador 3 de Interferon , Insuficiência Renal Crônica , Animais , Medicamentos de Ervas Chinesas , Fibrose , Ácido Fólico , Humanos , Fator Regulador 3 de Interferon/metabolismo , Interferons/metabolismo , Interferons/farmacologia , Células Matadoras Naturais , Proteínas de Membrana/metabolismo , Camundongos , Perforina/metabolismo , Perforina/farmacologia , Proteínas Serina-Treonina Quinases , Insuficiência Renal Crônica/tratamento farmacológico , Transdução de Sinais
8.
Viruses ; 14(3)2022 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-35336897

RESUMO

Cytolysin-positive Enterococcus faecalis (E. faecalis) cause more severe alcohol-associated hepatitis, and phages might be used to specifically target these bacteria in a clinical trial. Using a humanized mouse model of ethanol-induced liver disease, the effect of cytolytic E. faecalis phage treatment on the intestinal and liver immune response was evaluated. The observed immune response was predominantly anti-inflammatory and tissue-restoring. Besides, live phages could be readily recovered from the serum, spleen, and liver following oral gavage in ethanol-fed mice. We also isolated 20 new phages from the sewage water; six of them exhibited a relatively broad host range. Taken together, the oral administration of cytolytic E. faecalis phages leads to the translocation of phages to the systemic circulation and appears to be safe, following chronic-binge ethanol administration. A cocktail of three phages covers the majority of tested cytolysin-positive E. faecalis strains and could be tested in a clinical trial.


Assuntos
Bacteriófagos , Hepatopatias , Animais , Bacteriófagos/fisiologia , Enterococcus faecalis , Etanol/farmacologia , Imunidade , Camundongos , Perforina/farmacologia
9.
Nat Nanotechnol ; 16(10): 1130-1140, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34385682

RESUMO

Despite the enormous therapeutic potential of immune checkpoint blockade (ICB), it benefits only a small subset of patients. Some chemotherapeutics can switch 'immune-cold' tumours to 'immune-hot' to synergize with ICB. However, safe and universal therapeutic platforms implementing such immune effects remain scarce. We demonstrate that sphingomyelin-derived camptothecin nanovesicles (camptothesomes) elicit potent granzyme-B- and perforin-mediated cytotoxic T lymphocyte (CTL) responses, potentiating PD-L1/PD-1 co-blockade to eradicate subcutaneous MC38 adenocarcinoma with developed memory immunity. In addition, camptothesomes improve the pharmacokinetics and lactone stability of camptothecin, avoid systemic toxicities, penetrate deeply into the tumour and outperform the antitumour efficacy of Onivyde. Camptothesome co-load the indoleamine 2,3-dioxygenase inhibitor indoximod into its interior using the lipid-bilayer-crossing capability of the immunogenic cell death inducer doxorubicin, eliminating clinically relevant advanced orthotopic CT26-Luc tumours and late-stage B16-F10-Luc2 melanoma, and achieving complete metastasis remission when combined with ICB and folate targeting. The sphingomyelin-derived nanotherapeutic platform and doxorubicin-enabled transmembrane transporting technology are generalizable to various therapeutics, paving the way for transformation of the cancer immunochemotherapy paradigm.


Assuntos
Camptotecina/farmacologia , Tratamento Farmacológico , Imunoterapia , Nanopartículas/química , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/imunologia , Adenocarcinoma/patologia , Animais , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/genética , Camptotecina/química , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/genética , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Granzimas/química , Granzimas/farmacologia , Humanos , Bicamadas Lipídicas/química , Bicamadas Lipídicas/farmacologia , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Perforina/química , Perforina/farmacologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/genética , Esfingomielinas/química , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia
10.
Thorac Cancer ; 11(11): 3223-3233, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32915511

RESUMO

BACKGROUND: Currently used biomarkers for immunotherapy are inadequate because they are only based on tumor properties. In view of microenvironment changes by tumors, host immunity should be considered, which may result in identifying more accurate and easily detectable biomarkers for daily clinical practice. Here, we assessed serum immune-modulating factor levels for the response to anti-PD-1 antibodies during the first cycle in non-small cell lung cancer (NSCLC) patients. METHODS: Serum was collected from patients with advanced NSCLC treated with nivolumab or pembrolizumab at several time points during the first cycle. We applied the enzyme-linked immunosorbent assays (ELISAs) and multiplex assays to measure the levels of immune modulators. RESULTS: A total of 40 patients treated with nivolumab and 26 patients treated with pembrolizumab were studied. By ELISA, serum perforin, but not granzyme B, was measured in all samples. By multiplex assay, 10 immune modulators, including granzyme B, were measured in some, but not all, samples. Serum baseline perforin levels were strongly associated with increased progression-free survival (PFS) and overall survival (OS) times. Sequential changes in perforin levels during the first cycle were weakly associated with the clinical outcome. CONCLUSIONS: Serum baseline perforin levels may be used to predict the prognosis of NSCLC patients treated with anti-PD-1 antibody therapy. KEY POINTS: To identify a useful predictive marker for anti-PD-1 antibody therapy, using blood samples might be helpful. Serum baseline perforin levels were closely associated with prognosis with anti-PD-1 antibody therapy in non-small cell lung cancer.


Assuntos
Biomarcadores Tumorais/sangue , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Citotoxinas/uso terapêutico , Imunoterapia/métodos , Neoplasias Pulmonares/tratamento farmacológico , Perforina/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma Pulmonar de Células não Pequenas/patologia , Citotoxinas/farmacologia , Feminino , Humanos , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Perforina/farmacologia
11.
PLoS One ; 15(4): e0231430, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32275689

RESUMO

Extracellular vesicles (EVs) in the tumor microenvironment facilitate intercellular communication. Cancer cell-derived EVs act as an immunosuppressor by transporting cargos and presenting transmembrane proteins. By contrast, CD8+ cytotoxic T-lymphocytes (CTLs) exert anti-cancer cytotoxicity via the pore-forming protein perforin. Here, we hypothesize that although EVs are destroyed by perforin, cancer cell-derived EVs might possess mechanisms that enable them to avoid this destruction. We used a breast cancer cell line, MDA-MB-231-luc-D3H2LN (D3H2LN), to generate EVs. Destruction of the EVs by perforin was demonstrated visually using atomic force microscopy. To investigate immunosuppressive metabolites within cancer cell-derived EVs, we performed metabolomic profiling of EVs from D3H2LN cells cultured for 48 h with or without IFN-γ, which induces metabolic changes in the cells. We found that both types of EV from IFN-γ treated D3H2LN cells and non-treated D3H2LN cells contained adenosine, which has immunosuppressive effects. When we exposed cancer cell-derived EVs to CTLs, perforin secretion by CTLs fell significantly. In addition, the decreases in perforin secretion were ameliorated by treatment with adenosine deaminase, which degrades extracellular adenosine. Taken together, these results suggest that after perforin secreted by CTLs disrupts the membrane of EVs, adenosine released from the EVs acts as an immunosuppressive metabolite by binding to the adenosine receptor on the CTL membrane. This mechanism provides a novel survival strategy using cancer cell-derived EVs.


Assuntos
Adenosina/metabolismo , Vesículas Extracelulares/metabolismo , Perforina/metabolismo , Linfócitos T Citotóxicos/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Vesículas Extracelulares/efeitos dos fármacos , Humanos , Interferon gama/farmacologia , Perforina/farmacologia , Linfócitos T Citotóxicos/efeitos dos fármacos
12.
Cell Death Dis ; 10(10): 695, 2019 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-31541080

RESUMO

Cytotoxic T lymphocytes (CTL) and natural killer cells (NK)-mediated elimination of tumor cells is mostly dependent on Granzyme B apoptotic pathway, which is regulated by the wild type (wt) p53 protein. Because TP53 inactivating mutations, frequently found in human tumors, could interfere with Granzyme B-mediated cell death, the use of small molecules developed to reactivate wtp53 function in p53-mutated tumor cells could optimize their lysis by CTL or NK cells. Here, we show that the pharmalogical reactivation of a wt-like p53 function in p53-mutated breast cancer cells using the small molecule CP-31398 increases their sensitivity to NK-mediated lysis. This potentiation is dependent on p53-mediated induction of autophagy via the sestrin-AMPK-mTOR pathway and the ULK axis. This CP31398-induced autophagy sequestrates in autophagosomes several anti-apoptotic proteins, including Bcl-XL and XIAP, facilitating Granzyme B-mediated mitochondrial outer membrane permeabilization, caspase-3 activation and Granzyme B- or NK cell-induced apoptosis. Together, our results define a new way to increase cytotoxic lymphocyte-mediated lysis of p53-mutated breast cancer cell, through a p53-dependent autophagy induction, with potential applications in combined immunotherapeutic approaches.


Assuntos
Granzimas/farmacologia , Células Matadoras Naturais/imunologia , Proteína Supressora de Tumor p53/imunologia , Animais , Autofagia/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/imunologia , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Feminino , Granzimas/metabolismo , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/metabolismo , Camundongos , Perforina/farmacologia , Pirimidinas/farmacologia , Transdução de Sinais , Transfecção , Proteína Supressora de Tumor p53/metabolismo
13.
Sci Rep ; 9(1): 7212, 2019 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-31076615

RESUMO

The small intestinal epithelium of Vibrio cholerae infected patients expresses the immunomodulatory microRNAs miR-146a and miR-155 at acute stage of disease. V. cholerae release outer membrane vesicles (OMVs) that serve as vehicles for translocation of virulence factors including V. cholerae cytolysin (VCC). The aim was to investigate whether OMVs, with and/or without VCC-cargo could be responsible for induction of microRNAs in intestinal epithelial cells and thereby contribute to immunomodulation. Polarized tight monolayers of T84 cells were challenged with OMVs of wildtype and a VCC deletion mutant of the non-O1/non-O139 (NOVC) V. cholerae strain V:5/04 and with soluble VCC. OMVs, with and without VCC-cargo, caused significantly increased levels of miR-146a. Increase was seen already after 2 hours challenge with OMVs and persisted after 12 hours. Challenge with soluble VCC caused significant increases in interleukin-8 (IL-8), tumour necrosis factor-α (TNF-α), CCL20, IL-1ß, and IRAK2 mRNA levels while challenge with OMVs did not cause increases in expression levels of any of these mRNAs. These results suggest that V. cholerae bacteria release OMVs that induce miR-146a in order to pave the way for colonization by reducing the strength of an epithelial innate immune defence reaction and also preventing inflammation in the mucosa that factors like VCC can evoke.


Assuntos
Proteínas de Bactérias/farmacologia , Imunomodulação/efeitos dos fármacos , MicroRNAs/metabolismo , Vesículas Secretórias/metabolismo , Regulação para Cima/efeitos dos fármacos , Vibrio cholerae/metabolismo , Proteínas de Bactérias/metabolismo , Quimiocina CCL20/genética , Quimiocina CCL20/metabolismo , Cólera/microbiologia , Cólera/patologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Quinases Associadas a Receptores de Interleucina-1/genética , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Intestinos/citologia , Nanopartículas/química , Perforina/metabolismo , Perforina/farmacologia
14.
JCI Insight ; 3(8)2018 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-29669928

RESUMO

Although immune checkpoint inhibitors have resulted in durable clinical benefits in a subset of patients with advanced cancer, some patients who did not respond to initial anti-PD-1 therapy have been found to benefit from the addition of salvage chemotherapy. However, the mechanism responsible for the successful chemoimmunotherapy is not completely understood. Here we show that a subset of circulating CD8+ T cells expressing the chemokine receptor CX3CR1 are able to withstand the toxicity of chemotherapy and are increased in patients with metastatic melanoma who responded to chemoimmunotherapy (paclitaxel and carboplatin plus PD-1 blockade). These CX3CR1+CD8+ T cells have effector memory phenotypes and the ability to efflux chemotherapy drugs via the ABCB1 transporter. In line with clinical observation, our preclinical models identified an optimal sequencing of chemoimmunotherapy that resulted in an increase of CX3CR1+CD8+ T cells. Taken together, we found a subset of PD-1 therapy-responsive CD8+ T cells that were capable of withstanding chemotherapy and executing tumor rejection with their unique abilities of drug efflux (ABCB1), cytolytic activity (granzyme B and perforin), and migration to and retention (CX3CR1 and CD11a) at tumor sites. Future strategies to monitor and increase the frequency of CX3CR1+CD8+ T cells may help to design effective chemoimmunotherapy to overcome cancer resistance to immune checkpoint blockade therapy.


Assuntos
Linfócitos T CD8-Positivos/efeitos dos fármacos , Receptor 1 de Quimiocina CX3C/efeitos dos fármacos , Imunoterapia/métodos , Melanoma/imunologia , Neoplasias/tratamento farmacológico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Animais , Antineoplásicos/uso terapêutico , Linfócitos T CD8-Positivos/imunologia , Receptor 1 de Quimiocina CX3C/imunologia , Carboplatina/uso terapêutico , Citotoxinas/farmacologia , Quimioterapia Combinada , Feminino , Granzimas/farmacologia , Humanos , Masculino , Melanoma/tratamento farmacológico , Melanoma/secundário , Camundongos , Neoplasias/imunologia , Paclitaxel/uso terapêutico , Perforina/farmacologia
15.
Methods ; 140-141: 198-211, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29175337

RESUMO

Dynamic heterogeneity (DH) at nanoscale due to lipid-lipid and/or lipid-protein interactions in cell membranes plays a crucial role in determining a broad range of important cell functions. In cell membranes, the dimensions of these nanodomains have been postulated to be in the order of 10's of nm and transient in nature. While the structural features of membranes have been studied in detail, little is known about their dynamical characteristics due to paucity of techniques which can probe nanoscale phenomena with simultaneous high temporal resolution. A combination of super-resolution stimulated emission depletion (STED) and fluorescence correlation spectroscopy (FCS) technique can overcome this limitation and provide information about the nanoscale dynamic heterogeneity in cell membranes. Using STED-FCS and FCS diffusion law, we provide an understanding of how nanoscale dynamically organizing lipid platforms can emerge in minimal system of model biomembranes. To illustrate the utility of the technique we have chosen cholesterol containing supported lipid bilayers and demonstrated the role of cholesterol concentration and/or added pore-forming protein, Listeriolysin O (LLO) in determining onset of lipid DH. In addition we have also looked at multi-component lipid bilayers with and without cholesterol to infer about the role of phospholipid and cholesterol composition on lipid dynamics. These results on simple biomimetic systems provide insights into fundamental pathways for the emergence of complex nanodomain substructures with implications for a wide variety of membrane mediated cellular events and depict the significant contribution that STED-FCS can make in resolving several outstanding issues in membrane biology.


Assuntos
Membrana Celular/metabolismo , Colesterol/metabolismo , Fosfolipídeos/metabolismo , Espectrometria de Fluorescência/métodos , Membrana Celular/efeitos dos fármacos , Difusão , Fluorescência , Corantes Fluorescentes/química , Bicamadas Lipídicas/metabolismo , Proteínas de Membrana/metabolismo , Microscopia Confocal/instrumentação , Microscopia Confocal/métodos , Nanotecnologia/instrumentação , Nanotecnologia/métodos , Perforina/farmacologia , Espectrometria de Fluorescência/instrumentação
16.
Protein Eng Des Sel ; 30(2): 105-111, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27980121

RESUMO

Perforin is a pore-forming, immune protein that functions to deliver an apoptotic cocktail of proteins into a target pathogen. Recent studies of the bacterial cholesterol-dependent cytolysins (CDCs) have provided a model for perforin's pore-forming mechanism. Both perforin and CDC family members share a conserved ß-sheet flanked by two clusters of α-helices. Within the CDCs, these helices refold into two transmembrane ß-hairpins, TMH1 and TMH2. Based upon structural conservation and electron microscopy imaging, the analogous helices within perforin are predicted to also be membrane inserting; however, these regions are approximately twice the length of the CDC TMHs. To test the membrane-insertion potential of one of these regions, chimeras were created using a well-characterized CDC, perfringolysin-O (PFO), as the backbone of these constructs. PFO's TMH2 region was replaced with perforin's corresponding helical region. Although hemolytic activity was observed, the chimera was poorly soluble. A second chimera contained the same region truncated to match the length of the PFO TMH2 region. The truncated chimera demonstrated improved solubility, significant hemolytic activity and the ability to form pores characteristic of those created by PFO. These results provide the first evidence that perforin's helices function as TMHs and more importantly narrows the residues responsible for membrane insertion.


Assuntos
Membrana Celular/metabolismo , Colesterol/metabolismo , Perforina/química , Perforina/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Sequência de Aminoácidos , Animais , Hemólise/efeitos dos fármacos , Humanos , Camundongos , Modelos Moleculares , Perforina/genética , Perforina/farmacologia , Porosidade , Estrutura Secundária de Proteína , Coelhos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia
17.
Methods Mol Biol ; 1535: 275-284, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27914086

RESUMO

Bacterial pathogens represent a constant threat to human health that was exacerbated in recent years by a dramatic increase of strains resistant to last resort antibiotics. The immune system of higher vertebrates generally evolved several efficient innate and adaptive mechanisms to fight ubiquitous bacterial pathogens. Among those mechanisms, immune proteases were recognized to contribute essentially to antibacterial immune defense. The effector serine proteases of the adaptive immune system, the granzymes, exert potent antimicrobial activity when they are delivered into the bacterial cytosol by prokaryotic membrane disrupting proteins, such as granulysin.In this chapter, we are detailing experimental protocols to study the synergistic cytotoxic effects of human granzymes and granulysin on extracellular as well as on intracellular bacterial pathogens in vitro. In addition, we provide a simple and fast-forward method to biochemically purify native cytotoxic effector molecules necessary to perform this kind of investigations.


Assuntos
Antibacterianos/farmacologia , Antígenos de Diferenciação de Linfócitos T/farmacologia , Bactérias/efeitos dos fármacos , Bactérias/imunologia , Citotoxicidade Imunológica , Granzimas/farmacologia , Perforina/farmacologia , Linhagem Celular , Relação Dose-Resposta a Droga , Espaço Extracelular/imunologia , Espaço Extracelular/microbiologia , Humanos , Espaço Intracelular/imunologia , Espaço Intracelular/microbiologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo
18.
Mol Neurobiol ; 54(2): 874-887, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-26780457

RESUMO

Studies on the mechanisms of neuronal amyloid-ß (Aß) internalisation are crucial for understanding the neuropathological progression of Alzheimer's disease (AD). We here investigated how extracellular Aß peptides are internalised and focused on three different pathways: (i) via endocytic mechanisms, (ii) via the receptor for advanced glycation end products (RAGE) and (iii) via the pore-forming protein perforin. Both Aß40 and Aß42 were internalised in retinoic acid differentiated neuroblastoma (RA-SH-SY5Y) cells. A higher concentration was required for Aß40 (250 nM) compared with Aß42 (100 nM). The internalised Aß40 showed a dot-like pattern of distribution whereas Aß42 accumulated in larger and distinct formations. By confocal microscopy, we showed that Aß40 and Aß42 co-localised with mitochondria, endoplasmic reticulum (ER) and lysosomes. Aß treatment of human primary cortical neurons (hPCN) confirmed our findings in RA-SH-SY5Y cells, but hPCN were less sensitive to Aß; therefore, a 20 (Aß40) and 50 (Aß42) times higher concentration was needed for inducing uptake. The blocking of endocytosis completely inhibited the internalisation of Aß peptides in RA-SH-SY5Y cells and hPCN, indicating that this is a major pathway by which Aß enters the cells. In addition, the internalisation of Aß42, but not Aß40, was reduced by 55 % by blocking RAGE. Finally, for the first time we showed that pore formation in cell membranes by perforin led to Aß internalisation in hPCN. Understanding how Aß is internalised sheds light on the pathological role of Aß and provides further ideas of inhibitory strategies for preventing Aß internalisation and the spreading of neurodegeneration in AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Endocitose/fisiologia , Neurônios/metabolismo , Fragmentos de Peptídeos/metabolismo , Perforina/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Endocitose/efeitos dos fármacos , Humanos , Neurônios/efeitos dos fármacos , Perforina/farmacologia
19.
Org Lett ; 18(23): 6188-6191, 2016 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-27934350

RESUMO

Cytolysin, a two-component lanthipeptide comprising cytolysin S (CylLS″) and cytolysin L (CylLL″), is the only family member to exhibit lytic activity against mammalian cells in addition to synergistic antimicrobial activity. A subset of the thioether cross-links of CylLS″ and CylLL″ have ll stereochemistry instead of the canonical dl stereochemistry in all previously characterized lanthipeptides. The synthesis of a CylLS″ variant with dl stereochemistry is reported. Its antimicrobial activity was found to be decreased, but not its lytic activity against red blood cells. Hence, the unusual ll stereochemistry is not responsible for the lytic activity.


Assuntos
Antibacterianos/farmacologia , Lactococcus lactis/efeitos dos fármacos , Perforina/farmacologia , Antibacterianos/síntese química , Antibacterianos/química , Relação Dose-Resposta a Droga , Lactococcus lactis/patogenicidade , Testes de Sensibilidade Microbiana , Conformação Molecular , Perforina/síntese química , Perforina/química , Estereoisomerismo , Relação Estrutura-Atividade , Virulência/efeitos dos fármacos
20.
Cell Death Dis ; 7: e2193, 2016 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-27077809

RESUMO

CD4(+) T lymphocytes are key players in the adaptive immune system and can differentiate into a variety of effector and regulatory T cells. Here, we provide evidence that a novel differentiation pathway of CD4(+) T cells shifts the balance from a destructive T-cell response to one that favors regulation in an immune-mediated liver injury model. Peripheral CD4(-)CD8(-)NK1.1(-) double-negative T cells (DNT) was increased following Concanavalin A administration in mice. Adoptive transfer of DNT led to significant protection from hepatocyte necrosis by direct inhibition on the activation of lymphocytes, a process that occurred primarily through the perforin-granzyme B route. These DNT converted from CD4(+) rather than CD8(+) T cells, a process primarily regulated by OX40. DNT migrated to the liver through the CXCR3-CXCL9/CXCL10 interaction. In conclusion, we elucidated a novel differentiation pathway from activated CD4(+) T cells to regulatory DNT cells for maintaining homeostasis of the immune system in vivo, and provided key evidence that utilizing this novel differentiation pathway has potential application in the prevention and treatment of autoimmune diseases.


Assuntos
Linfócitos T CD4-Positivos/citologia , Linfócitos T/citologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Quimiocina CXCL9/genética , Quimiocina CXCL9/metabolismo , Concanavalina A/farmacologia , Citocinas/sangue , Granzimas/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ligante OX40/deficiência , Ligante OX40/genética , Perforina/deficiência , Perforina/genética , Perforina/farmacologia , RNA Mensageiro/metabolismo , Receptores CXCR3/genética , Receptores CXCR3/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...