Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Ther ; 31(8): 2309-2325, 2023 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-37312454

RESUMEN

Multiple clinical studies have treated mesothelin (MSLN)-positive solid tumors by administering MSLN-directed chimeric antigen receptor (CAR) T cells. Although these products are generally safe, efficacy is limited. Therefore, we generated and characterized a potent, fully human anti-MSLN CAR. In a phase 1 dose-escalation study of patients with solid tumors, we observed two cases of severe pulmonary toxicity following intravenous infusion of this product in the high-dose cohort (1-3 × 108 T cells per m2). Both patients demonstrated progressive hypoxemia within 48 h of infusion with clinical and laboratory findings consistent with cytokine release syndrome. One patient ultimately progressed to grade 5 respiratory failure. An autopsy revealed acute lung injury, extensive T cell infiltration, and accumulation of CAR T cells in the lungs. RNA and protein detection techniques confirmed low levels of MSLN expression by benign pulmonary epithelial cells in affected lung and lung samples obtained from other inflammatory or fibrotic conditions, indicating that pulmonary pneumocyte and not pleural expression of mesothelin may lead to dose-limiting toxicity. We suggest patient enrollment criteria and dosing regimens of MSLN-directed therapies consider the possibility of dynamic expression of mesothelin in benign lung with a special concern for patients with underlying inflammatory or fibrotic conditions.


Asunto(s)
Mesotelina , Neoplasias , Humanos , Proteínas Ligadas a GPI/genética , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Neoplasias/terapia , Linfocitos T
2.
Protein Cell ; 8(7): 514-526, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28523432

RESUMEN

The generation of T cells with maximal anti-tumor activities will significantly impact the field of T-cell-based adoptive immunotherapy. In this report, we found that OKT3/IL-2-stimulated T cells were phenotypically more heterogeneous, with enhanced anti-tumor activity in vitro and when locally administered in a solid tumor mouse model. To further improve the OKT3/IL-2-based T cell manufacturing procedure, we developed a novel T cell stimulation and expansion method in which peripheral blood mononuclear cells were electroporated with mRNA encoding a chimeric membrane protein consisting of a single-chain variable fragment against CD3 and the intracellular domains of CD28 and 4-1BB (OKT3-28BB). The expanded T cells were phenotypically and functionally similar to T cells expanded by OKT3/IL-2. Moreover, co-electroporation of CD86 and 4-1BBL could further change the phenotype and enhance the in vivo anti-tumor activity. Although T cells expanded by the co-electroporation of OKT3-28BB with CD86 and 4-1BBL showed an increased central memory phenotype, the T cells still maintained tumor lytic activities as potent as those of OKT3/IL-2 or OKT3-28BB-stimulated T cells. In different tumor mouse models, T cells expanded by OKT3-28BB RNA electroporation showed anti-tumor activities superior to those of OKT3/IL-2 T cells. Hence, T cells with both a less differentiated phenotype and potent tumor killing ability can be generated by RNA electroporation, and this T cell manufacturing procedure can be further optimized by simply co-delivering other splices of RNA, thus providing a simple and cost-effective method for generating high-quality T cells for adoptive immunotherapy.


Asunto(s)
Antígenos CD28 , Electroporación , Inmunidad Celular , Neoplasias Experimentales/inmunología , ARN Mensajero , Linfocitos T/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral , Animales , Antígenos CD28/genética , Antígenos CD28/inmunología , Humanos , Interleucina-2/inmunología , Células K562 , Ratones , Muromonab-CD3/inmunología , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , ARN Mensajero/genética , ARN Mensajero/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
3.
Oncotarget ; 8(10): 17002-17011, 2017 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-28199983

RESUMEN

The therapeutic potential of CRISPR system has already been demonstrated in many instances and begun to overlap with the rapidly expanding field of cancer immunotherapy, especially on the production of genetically modified T cell receptor or chimeric antigen receptor (CAR) T cells. Efficient genomic disruption of multiple gene loci to generate universal donor cells, as well as potent effector T cells resistant to multiple inhibitory pathways such as PD-1 and CTLA4 is an attractive strategy for cell therapy. In this study, we accomplished rapid and efficient multiplex genomic editing, and re-directing T cells with antigen specific CAR via a one-shot CRISPR protocol by incorporation of multiple gRNAs in a CAR lentiviral vector. High efficient double knockout of endogenous TCR and HLA class I could be easily achieved to generate allogeneic universal CAR T cells. We also generated Fas-resistant universal CAR T cells by triple gene disruption. Simultaneous gene editing of four gene loci using the one-shot CRISPR protocol to generate allogeneic universal T cells deficient of both PD1 and CTLA-4 was also attempted.


Asunto(s)
Edición Génica/métodos , Receptores de Antígenos de Linfocitos T/genética , Proteínas Recombinantes de Fusión/genética , Linfocitos T/metabolismo , Animales , Apoptosis/genética , Sistemas CRISPR-Cas , Antígeno CTLA-4/genética , Línea Celular Tumoral , Células Cultivadas , Genes MHC Clase I/genética , Humanos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Células K562 , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Receptor de Muerte Celular Programada 1/genética , Reproducibilidad de los Resultados , Trasplante Heterólogo
4.
Clin Cancer Res ; 23(9): 2255-2266, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-27815355

RESUMEN

Purpose: Using gene-disrupted allogeneic T cells as universal effector cells provides an alternative and potentially improves current chimeric antigen receptor (CAR) T-cell therapy against cancers and infectious diseases.Experimental Design: The CRISPR/Cas9 system has recently emerged as a simple and efficient way for multiplex genome engineering. By combining lentiviral delivery of CAR and electro-transfer of Cas9 mRNA and gRNAs targeting endogenous TCR, ß-2 microglobulin (B2M) and PD1 simultaneously, to generate gene-disrupted allogeneic CAR T cells deficient of TCR, HLA class I molecule and PD1.Results: The CRISPR gene-edited CAR T cells showed potent antitumor activities, both in vitro and in animal models and were as potent as non-gene-edited CAR T cells. In addition, the TCR and HLA class I double deficient T cells had reduced alloreactivity and did not cause graft-versus-host disease. Finally, simultaneous triple genome editing by adding the disruption of PD1 led to enhanced in vivo antitumor activity of the gene-disrupted CAR T cells.Conclusions: Gene-disrupted allogeneic CAR and TCR T cells could provide an alternative as a universal donor to autologous T cells, which carry difficulties and high production costs. Gene-disrupted CAR and TCR T cells with disabled checkpoint molecules may be potent effector cells against cancers and infectious diseases. Clin Cancer Res; 23(9); 2255-66. ©2016 AACR.


Asunto(s)
Edición Génica , Neoplasias/terapia , Receptor de Muerte Celular Programada 1/genética , Linfocitos T/inmunología , Sistemas CRISPR-Cas , Enfermedad Injerto contra Huésped , Humanos , Inmunoterapia Adoptiva , Activación de Linfocitos/inmunología , Neoplasias/genética , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/deficiencia , Receptores de Antígenos de Linfocitos T/deficiencia , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/inmunología
5.
Cancer Res ; 76(6): 1578-90, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26979791

RESUMEN

Chimeric antigen receptor (CAR)-modified adoptive T-cell therapy has been successfully applied to the treatment of hematologic malignancies, but faces many challenges in solid tumors. One major obstacle is the immune-suppressive effects induced in both naturally occurring and genetically modified tumor-infiltrating lymphocytes (TIL) by inhibitory receptors (IR), namely PD1. We hypothesized that interfering with PD1 signaling would augment CAR T-cell activity against solid tumors. To address this possibility, we introduced a genetically engineered switch receptor construct, comprising the truncated extracellular domain of PD1 and the transmembrane and cytoplasmic signaling domains of CD28, into CAR T cells. We tested the effect of this supplement, "PD1CD28," on human CAR T cells targeting aggressive models of human solid tumors expressing relevant tumor antigens. Treatment of mice bearing large, established solid tumors with PD1CD28 CAR T cells led to significant regression in tumor volume due to enhanced CAR TIL infiltrate, decreased susceptibility to tumor-induced hypofunction, and attenuation of IR expression compared with treatments with CAR T cells alone or PD1 antibodies. Taken together, our findings suggest that the application of PD1CD28 to boost CAR T-cell activity is efficacious against solid tumors via a variety of mechanisms, prompting clinical investigation of this potentially promising treatment modality.


Asunto(s)
Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T/inmunología , Animales , Antígenos de Neoplasias/inmunología , Antígenos CD28/inmunología , Línea Celular Tumoral , Femenino , Humanos , Inmunoterapia Adoptiva/métodos , Células K562 , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal/inmunología , Carga Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
6.
Cancer Res ; 75(17): 3596-607, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26330166

RESUMEN

Target-mediated toxicity is a major limitation in the development of chimeric antigen T-cell receptors (CAR) for adoptive cell therapy of solid tumors. In this study, we developed a strategy to adjust the affinities of the scFv component of CAR to discriminate tumors overexpressing the target from normal tissues that express it at physiologic levels. A CAR-expressing T-cell panel was generated with target antigen affinities varying over three orders of magnitude. High-affinity cells recognized target expressed at any level, including at levels in normal cells that were undetectable by flow cytometry. Affinity-tuned cells exhibited robust antitumor efficacy similar to high-affinity cells, but spared normal cells expressing physiologic target levels. The use of affinity-tuned scFvs offers a strategy to empower wider use of CAR T cells against validated targets widely overexpressed on solid tumors, including those considered undruggable by this approach.


Asunto(s)
Receptores ErbB/inmunología , Neoplasias/inmunología , Receptor ErbB-2/inmunología , Receptores de Antígenos/inmunología , Animales , Línea Celular Tumoral , Receptores ErbB/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Inmunoterapia Adoptiva , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Receptor ErbB-2/antagonistas & inhibidores , Receptores de Antígenos/antagonistas & inhibidores , Anticuerpos de Cadena Única/administración & dosificación , Anticuerpos de Cadena Única/inmunología , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Immunol ; 195(8): 4020-7, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26378078

RESUMEN

Mitochondria provide energy for cells via oxidative phosphorylation. Reactive oxygen species, a byproduct of this mitochondrial respiration, can damage mitochondrial DNA (mtDNA), and somatic mtDNA mutations have been found in all colorectal, ovarian, breast, urinary bladder, kidney, lung, and pancreatic tumors studied. The resulting altered mitochondrial proteins or tumor-associated mitochondrial Ags (TAMAs) are potentially immunogenic, suggesting that they may be targetable Ags for cancer immunotherapy. In this article, we show that the RENCA tumor cell line harbors TAMAs that can drive an antitumor immune response. We generated a cellular tumor vaccine by pulsing dendritic cells with enriched mitochondrial proteins from RENCA cells. Our dendritic cell-based RENCA mitochondrial lysate vaccine elicited a cytotoxic T cell response in vivo and conferred durable protection against challenge with RENCA cells when used in a prophylactic or therapeutic setting. By sequencing mtDNA from RENCA cells, we identified two mutated molecules: COX1 and ND5. Peptide vaccines generated from mitochondrial-encoded COX1 but not from ND5 had therapeutic properties similar to RENCA mitochondrial protein preparation. Thus, TAMAs can elicit effective antitumor immune responses, potentially providing a new immunotherapeutic strategy to treat cancer.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Carcinoma de Células Renales/prevención & control , Ciclooxigenasa 1/inmunología , Neoplasias Renales/prevención & control , Proteínas de la Membrana/inmunología , Proteínas Mitocondriales/inmunología , NADH Deshidrogenasa/inmunología , Neoplasias Experimentales/prevención & control , Animales , Antígenos de Neoplasias/farmacología , Vacunas contra el Cáncer/farmacología , Carcinoma de Células Renales/inmunología , Ciclooxigenasa 1/farmacología , Neoplasias Renales/inmunología , Proteínas de la Membrana/farmacología , Ratones , Ratones Endogámicos BALB C , Proteínas Mitocondriales/farmacología , NADH Deshidrogenasa/farmacología , Neoplasias Experimentales/inmunología
8.
Hum Gene Ther ; 24(3): 295-305, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23297870

RESUMEN

NKG2D ligands (NKG2DLs) are widely expressed on ovarian cancers to various degrees, making them attractive targets for immunotherapy. Here, we applied a chimeric antigen receptor (CAR) approach for the targeting of NKG2DLs expressed on human ovarian cancer cells and evaluated the impact of pharmacological upregulation of NKG2DLs on immune recognition. Various NKG2DLs, including MICA/B and ULBP-1, -2, -3, and -4, were expressed at various levels on the surface of all established ovarian cancer cell lines and primary ovarian cancer samples tested. To redirect human T cells against NKG2DLs, an NKG2DL-specific CAR was generated by fusing the extracellular domain of the NKG2D receptor to the 4-1BB costimulatory and CD3-ζ chain signaling domains. In vitro expansion of chimeric NKG2D CAR T cells was delayed compared with untransduced T cells and control CAR T cells; the likely result of fratricide among activated T cells expressing NKG2DLs. However, NKG2D CAR T cells did expand and were selectively enriched during prolonged culture. In coculture, CD4(+) and CD8(+) NKG2D CAR T cells specifically recognized and killed NKG2DL-expressing ovarian cancer cell lines but not NKG2DL-negative cells. Notably, pretreatment of ovarian cancer cells expressing moderate to low levels of NKG2DLs with the histone deacetylase inhibitor sodium valproate (VPA) upregulated NKG2DL cell surface expression and consequently enhanced their immune recognition by chimeric NKG2D CAR T cells. Our results demonstrate that VPA-induced upregulation of NKG2DL expression enhances the immune recognition of ovarian cancer cells by engineered NKG2D CAR T cells, and rationalizes the use of VPA in combination with NKG2DL-targeted immunotherapy in ovarian cancer.


Asunto(s)
Histona Desacetilasas/metabolismo , Subfamilia K de Receptores Similares a Lectina de Células NK/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo , Línea Celular , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Orden Génico , Vectores Genéticos/genética , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Inmunofenotipificación , Lentivirus/genética , Ligandos , Activación de Linfocitos , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Neoplasias Ováricas/metabolismo , Fenotipo , Receptores de Antígenos de Linfocitos T/metabolismo , Ácido Valproico/farmacología
9.
Blood ; 118(4): 1008-14, 2011 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-21652682

RESUMEN

Recent studies have indicated a role of complement in regulating T-cell immunity but the mechanism of action of complement in this process remains to be clarified. Here we studied mice deficient in decay-accelerating factor (DAF), a key membrane complement regulator whose deficiency led to increased complement-dependent T-cell immune responses in vivo. By crossing OT-II and OT-I T-cell receptor transgenic mice with DAF-knockout mice, we found that lack of DAF on T cells did not affect their responses to antigen stimulation. Similarly, lack of DAF on antigen-presenting cells (APCs) of naive mice did not alter their T-cell stimulating activity. In contrast, APCs from DAF-knockout mice treated with inflammatory stimuli were found to be more potent T-cell stimulators than cells from similarly treated wild-type mice. Acquisition of higher T-cell stimulating activity by APCs in challenged DAF-knockout mice required C3 and C5aR and was correlated with decreased surface PD-L1 and/or increased CD40 expression. These findings implied that DAF suppressed T-cell immunity as a complement regulator in the context of inflammation but did not play an intrinsic role on T cells or APCs. Collectively, our data suggest a systemic and indirect role of complement in T-cell immunity.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Antígenos CD55/inmunología , Activación de Complemento/inmunología , Activación de Linfocitos/inmunología , Linfocitos T/inmunología , Animales , Células Presentadoras de Antígenos/metabolismo , Western Blotting , Antígenos CD55/metabolismo , Separación Celular , Citometría de Flujo , Inflamación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Linfocitos T/metabolismo
10.
Blood ; 114(5): 1005-15, 2009 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-19491392

RESUMEN

Toll-like receptors (TLRs) and complement are 2 major components of innate immunity that provide a first-line host defense and shape the adaptive immune responses. We show here that coincidental activation of complement and several TLRs in mice led to the synergistic production of serum factors that promoted T-helper cell 17 (Th17) differentiation from anti-CD3/CD28 or antigen-stimulated T cells. Although multiple TLR-triggered cytokines were regulated by complement, Th17 cell-promoting activity in the serum was correlated with interleukin (IL)-6 induction, and antibody neutralization of IL-6 abrogated the complement effect. By using both in vitro and in vivo approaches, we examined in more detail the mechanism and physiologic implication of complement/TLR4 interaction on Th17-cell differentiation. We found that the complement effect required C5a receptor, was evident at physiologically relevant levels of C5a, and could be demonstrated in cultured peritoneal macrophages as well as in the setting of antigen immunization. Importantly, despite an inhibitory effect of complement on IL-23 production, complement-promoted Th17 cells were functionally competent in causing autoimmunity in an adoptive transfer model of experimental autoimmune encephalomyelitis. Collectively, these data establish a link between complement/TLR interaction and Th17-cell differentiation and provide new insight into the mechanism of action of complement in autoimmunity.


Asunto(s)
Activación de Complemento , Inflamación/inmunología , Interleucina-6/fisiología , Linfocitos T Colaboradores-Inductores/citología , Receptores Toll-Like/fisiología , Traslado Adoptivo , Animales , Antígenos CD55/genética , Células Cultivadas/citología , Células Cultivadas/metabolismo , Complemento C5a/fisiología , Complemento C5a/uso terapéutico , Citocinas/sangre , Citocinas/fisiología , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/inmunología , Inmunidad Innata , Inmunización , Inflamación/inducido químicamente , Interleucina-6/biosíntesis , Interleucina-6/sangre , Interleucina-6/deficiencia , Interleucina-6/genética , Linfopoyesis , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Anafilatoxina C5a/deficiencia , Receptor de Anafilatoxina C5a/fisiología , Proteínas Recombinantes/uso terapéutico , Organismos Libres de Patógenos Específicos , Linfocitos T Colaboradores-Inductores/clasificación , Linfocitos T Colaboradores-Inductores/inmunología , Receptor Toll-Like 4/fisiología
11.
J Immunol ; 179(5): 3178-86, 2007 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-17709533

RESUMEN

Decay-accelerating factor (DAF, CD55) is a GPI-anchored membrane protein that regulates complement activation on autologous cells. In addition to protecting host tissues from complement attack, DAF has been shown to inhibit CD4+ T cell immunity in the setting of model Ag immunization. However, whether DAF regulates natural T cell immune response during pathogenic infection is not known. We describe in this study a striking regulatory effect of DAF on the CD8+ T cell response to lymphocytic choriomeningitis virus (LCMV) infection. Compared with wild-type mice, DAF knockout (Daf-1(-/-)) mice had markedly increased expansion in the spleen of total and viral Ag-specific CD8+ T cells after acute or chronic LCMV infection. Splenocytes from LCMV-infected Daf-1(-/-) mice also displayed significantly higher killing activity than cells from wild-type mice toward viral Ag-loaded target cells, and Daf-1(-/-) mice cleared LCMV more efficiently. Importantly, deletion of the complement protein C3 or the receptor for the anaphylatoxin C5a (C5aR) from Daf-1(-/-) mice reversed the enhanced CD8+ T cell immunity phenotype. These results demonstrate that DAF is an important regulator of CD8+ T cell immunity in viral infection and that it fulfills this role by acting as a complement inhibitor to prevent virus-triggered complement activation and C5aR signaling. This mode of action of DAF contrasts with that of CD59 in viral infection and suggests that GPI-anchored membrane complement inhibitors can regulate T cell immunity to viral infection via either a complement-dependent or -independent mechanism.


Asunto(s)
Antígenos CD55/fisiología , Complemento C3/fisiología , Complemento C5a/fisiología , Coriomeningitis Linfocítica , Virus de la Coriomeningitis Linfocítica/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Antígenos Virales/inmunología , Antígenos CD55/genética , Antígenos CD8/análisis , Complemento C3/genética , Complemento C5a/genética , Memoria Inmunológica , Ratones , Ratones Mutantes , Mutación
12.
Blood ; 110(1): 228-36, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17363730

RESUMEN

Toll-like receptors (TLRs) and complement are 2 components of innate immunity that are critical for first-line host defense and elicitation of adaptive immune responses. Many pathogen-associated molecular patterns activate both TLR and complement, but whether and how these 2 systems, when coactivated in vivo, interact with each other has not been well studied. We demonstrate here a widespread regulation of TLR signaling by complement in vivo. The TLR ligands lipopolysacharride (TLR4), zymosan (TLR2/6), and CpG oligonucleotide (TLR9) caused, in a complement-dependent manner, strikingly elevated plasma interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-alpha), and IL-1beta, and/or decreased plasma IL-12 levels in mice deficient in the membrane complement inhibitor decay-accelerating factor (DAF). A similar outcome was observed in wild-type mice cotreated with the TLR ligands and cobra venom factor, a potent complement activator. The regulatory effect of complement on TLR-induced cytokine production in vivo was mediated by the anaphylatoxin receptors C5aR and C3aR. Additionally, changes in lipopolysaccharide (LPS)-induced cytokine production in DAF-deficient mice correlated with increased mitogen-activated protein kinase and nuclear factor-kappaB activation in the spleen. These results reveal a strong interaction between complement and TLR signaling in vivo and suggest a novel mechanism by which complement promotes inflammation and modulates adaptive immunity.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Inflamación/inmunología , Receptores Toll-Like/inmunología , Animales , Antígenos CD55/análisis , Citocinas/biosíntesis , Interleucina-12/sangre , Ligandos , Ratones , Ratones Noqueados , Transducción de Señal
13.
Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi ; 20(11): 1130-3, 2006 Nov.
Artículo en Chino | MEDLINE | ID: mdl-17191583

RESUMEN

OBJECTIVE: To establish a eukaryotic cell line that can express soluble human leucocyte antigen G1 (sHLA-G1) stably. METHODS: The recombinant plasmid pcDNA3-sHLA-G1 is transfected by a novel nonviral, electroporation-based gene transfer method termed nucleofection into the host cell lymphoblastoid cell line (LCL) 721.221 which does not express any HLA-classical I molecules. After selection by G418, the cell line stably expressing sHLA-G1 is identified by RT-PCR and Dot-ELISA with HLA-G1 specific monoclonal antibody MEM-G/9. RESULTS: The efficiency of transfection for LCL721. 221 is about 14% by nucleofection. The specific band for sHLA-G1 was found by RT-PCR assay from the transfections and the protein of sHLA-G1 in the supernatant of the transfections was detected by Dot-ELISA assay. Both confirmed that the eukaryotic cell line expressing sHLA-G1 has been established successfully at genic and proteinic levels. CONCLUSION: In this study, the eukaryotic cell line expressing sHLA-G1 have been established successfully by nucleofection.


Asunto(s)
Citometría de Flujo/métodos , Antígenos HLA/genética , Antígenos de Histocompatibilidad Clase I/genética , Línea Celular , ADN Complementario , Antígenos HLA-G , Humanos , Plásmidos , Transfección
14.
World J Gastroenterol ; 9(7): 1584-8, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12854169

RESUMEN

AIM: To investigate the expression of local renin and angiotensinogen mRNA in cirrhotic portal hypertensive patients. METHODS: The expression of local renin and angiotensinogen mRNA in the liver, splenic artery and vein of PH patients was detected by RT-PCR analysis. RESULTS: Expression of local renin mRNA in the liver of control group was (0.19+/-0.11), significantly lower than that in splenic artery (0.45+/-0.17) or splenic vein(0.39+/-0.12) respectively, (P<0.05). Expression of local angiotensinogen mRNA in the liver was (0.64+/-0.21), significantly higher than that in splenic artery(0.41+/-0.15) or in splenic vein (0.35+/-0.18) respectively, (P<0.05). Expression of local renin mRNA in the liver, splenic artery and vein of PH group was (0.78+/-0.28), (0.86+/-0.35) and (0.81+/-0.22) respectively, significantly higher than that in the control group, (P<0.05). Expression of local angiotensinogen mRNA in the liver, splenic artery and vein of PH group was (0.96+/-0.25), (0.83+/-0.18) and (0.79+/-0.23) respectively, significantly higher than that in the control group, (P<0.05). There was no significant difference between the liver, splenic artery and vein in the expression of local renin or local angiotensinogen mRNA in PH group, (P<0.05). CONCLUSION: In normal subjects the expression of local renin and angiotensinogen mRNA was organ specific, but with increase of the expression of LRAS, the organ-specificity became lost in cirrhotic patients. LRAS may contribute to increased resistance of portal vein with liver and formation of splanchnic vasculopathy.


Asunto(s)
Angiotensinógeno/genética , Hipertensión Portal/fisiopatología , Cirrosis Hepática/fisiopatología , Renina/genética , Adulto , Anciano , Endotelio Vascular/patología , Endotelio Vascular/ultraestructura , Femenino , Expresión Génica , Humanos , Hipertensión Portal/patología , Inmunohistoquímica , Cirrosis Hepática/patología , Masculino , Microscopía Electrónica de Rastreo , Persona de Mediana Edad , Especificidad de Órganos , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Vena Esplénica/patología
15.
Zhonghua Yi Xue Za Zhi ; 83(7): 584-7, 2003 Apr 10.
Artículo en Chino | MEDLINE | ID: mdl-12887750

RESUMEN

OBJECTIVE: To investigate the immune tolerance inducing effects of soluble human leucocyte antigen G1 (sHLA-G1) on natural killer (NK) cells and T cells. METHODS: A recombinant plasmid expressing sHLA-G1 was constructed and transfected into human lymphoblastoid cells LCL721.221. sHLA-G1 in the supernatant was purified by immuno-affinity chromatography and then added into the culture of NK cells obtained from the peripheral blood mononuclear cells of 3 unrelated individuals. Target cells, K562 cells, were added too. The killing rate of NK was calculated. Peripheral blood lymphocytes (PBLs) were obtained and stimulated by Ebstein-Barr-virus-transformed B lymphoblastoid cell line (EBV-LCL). The proliferation of the T cells in the mixed lymphocyte culture was examined by enzyme linked immunosorbent assay. The antigen-specific T cells in the peripheral blood was activated. sHLA-G1 was added into the culture. Then the T cells were suspended in the solution of fluorescence isothiocyanate (FITC)-annexin-V. Flow cytometry was used to detect the fluorescent intensity of FITC so as to examine the apoptosis of T cells. RESULTS: sHLAG-1 inhibited the cytotoxicity of NK cells dose-dependently. sHLAG-1 inhibited the proliferation of activated T cells, and induced the apoptosis of T cells dose-dependently, with a dose-saturation character and without antigen-specificity. CONCLUSION: sHLAG-1 is a kind of immune tolerance inducing molecule.


Asunto(s)
Antígenos HLA/fisiología , Antígenos de Histocompatibilidad Clase I/fisiología , Tolerancia Inmunológica , Células Asesinas Naturales/inmunología , Linfocitos T/inmunología , Apoptosis , Citotoxicidad Inmunológica , Antígenos HLA-G , Humanos , Prueba de Cultivo Mixto de Linfocitos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...