Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
iScience ; 27(2): 108992, 2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38333703

RESUMEN

Human iPSC-derived cardiomyocytes (hiPSC-CMs) exhibit functional immaturity, potentially impacting their suitability for assessing drug proarrhythmic potential. We previously devised a traveling wave (TW) system to promote maturation in 3D cardiac tissue. To align with current drug assessment paradigms (CiPA and JiCSA), necessitating a 2D monolayer cardiac tissue, we integrated the TW system with a multi-electrode array. This gave rise to a hiPSC-derived closed-loop cardiac tissue (iCT), enabling spontaneous TW initiation and swift pacing of cardiomyocytes from various cell lines. The TW-paced cardiomyocytes demonstrated heightened sarcomeric and functional maturation, exhibiting enhanced response to isoproterenol. Moreover, these cells showcased diminished sensitivity to verapamil and maintained low arrhythmia rates with ranolazine-two drugs associated with a low risk of torsades de pointes (TdP). Notably, the TW group displayed increased arrhythmia rates with high and intermediate risk TdP drugs (quinidine and pimozide), underscoring the potential utility of this system in drug assessment applications.

2.
J Immunol ; 212(5): 917-927, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38214607

RESUMEN

The adaptive transfer of T cells redirected to cancer cells via chimeric Ag receptors (CARs) has produced clinical benefits for the treatment of hematologic diseases. To extend this approach to solid cancer, we screened CARs targeting surface Ags on human lung cancer cells using (to our knowledge) novel expression cloning based on the Ag receptor-induced transcriptional activation of IL-2. Isolated CARs were directed against fragile X mental retardation 1 neighbor (FMR1NB), a cancer-testis Ag that is expressed by malignant cells and adult testicular germ cells. Anti-FMR1NB CAR human T cells demonstrated target-specific cytotoxicity and successfully controlled tumor growth in mouse xenograft models of lung cancer. Furthermore, to protect CAR T cells from immune-inhibitory molecules, which are present in the tumor microenvironment, we introduced anti-FMR1NB CARs into 2-deoxy-glucose (2DG)-treated human T cells. These cells exhibited reduced binding affinity to immune-inhibitory molecules, and the suppressive effects of these molecules were resisted through blockade of the N-glycosylation of their receptors. Anti-FMR1NB CARs in 2DG-treated human T cells augmented target-specific cytotoxicity in vitro and in vivo. Thus, our findings demonstrated the feasibility of eradicating lung cancer cells using 2DG-treated human T cells, which are able to direct tumor-specific FMR1NB via CARs and survive in the suppressive tumor microenvironment.


Asunto(s)
Discapacidad Intelectual , Neoplasias Pulmonares , Receptores Quiméricos de Antígenos , Humanos , Ratones , Animales , Linfocitos T , Receptores Quiméricos de Antígenos/metabolismo , Receptores de Antígenos de Linfocitos T , Neoplasias Pulmonares/terapia , Glicosilación , Inmunoterapia Adoptiva , Ensayos Antitumor por Modelo de Xenoinjerto , Línea Celular Tumoral , Microambiente Tumoral
3.
Cell Transplant ; 32: 9636897221148457, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36624995

RESUMEN

Although mesenchymal stem cell transplantation has been successful in the treatment of ischemic cardiomyopathy, the underlying mechanisms remain unclear. Herein, we investigated whether mitochondrial transfer could explain the success of cell therapy in ischemic cardiomyopathy. Mitochondrial transfer in co-cultures of human adipose-derived mesenchymal stem cells and rat cardiomyocytes maintained under hypoxic conditions was examined. Functional recovery was monitored in a rat model of myocardial infarction following human adipose-derived mesenchymal stem cell transplantation. We observed mitochondrial transfer in vitro, which required the formation of cell-to-cell contacts and synergistically enhanced energy metabolism. Rat cardiomyocytes exhibited mitochondrial transfer 3 days following human adipose-derived mesenchymal stem cell transplantation to the ischemic heart surface post-myocardial infarction. We detected donor mitochondrial DNA in the recipient myocardium concomitant with a significant improvement in cardiac function. Mitochondrial transfer is vital for successful cell transplantation therapies and improves treatment outcomes in ischemic cardiomyopathy.


Asunto(s)
Cardiomiopatías , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Infarto del Miocardio , Ratas , Humanos , Animales , Miocardio/metabolismo , Infarto del Miocardio/terapia , Infarto del Miocardio/genética , Miocitos Cardíacos/metabolismo , Cardiomiopatías/terapia , Trasplante de Células Madre
4.
J Am Heart Assoc ; 9(16): e015841, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32783519

RESUMEN

Background Extracellular matrix, especially laminin-221, may play crucial roles in viability and survival of human-induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs) after in vivo transplant. Then, we hypothesized laminin-221 may have an adjuvant effect on therapeutic efficacy by enhancing cell viability and survival after transplantation of 3-dimensional engineered cardiac tissue (ECT) to a rat model of myocardial infarction. Methods and Results In vitro study indicates the impacts of laminin-221 on hiPS-CMs were analyzed on the basis of mechanical function, mitochondrial function, and tolerance to hypoxia. We constructed 3-dimensional ECT containing hiPS-CMs and fibrin gel conjugated with laminin-221. Heart function and in vivo behavior were assessed after engraftment of 3-dimensional ECT (laminin-conjugated ECT, n=10; ECT, n=10; control, n=10) in a rat model of myocardial infarction. In vitro assessment indicated that laminin-221 improves systolic velocity, diastolic velocity, and maximum capacity of oxidative metabolism of hiPS-CMs. Cell viability and lactate dehydrogenase production revealed that laminin-221 improved tolerance to hypoxia. Furthermore, analysis of mRNA expression revealed that antiapoptotic genes were upregulated in the laminin group under hypoxic conditions. Left ventricular ejection fraction of the laminin-conjugated ECT group was significantly better than that of other groups 4 weeks after transplantation. Laminin-conjugated ECT transplantation was associated with significant improvements in expression levels of rat vascular endothelial growth factor. In early assessments, cell survival was also improved in laminin-conjugated ECTs compared with ECT transplantation without laminin-221. Conclusions In vitro laminin-221 enhanced mechanical and metabolic function of hiPS-CMs and improved the therapeutic impact of 3-dimensional ECT in a rat ischemic cardiomyopathy model. These findings suggest that adjuvant laminin-221 may provide a clinical benefit to hiPS-CM constructs.


Asunto(s)
Supervivencia Celular , Células Madre Pluripotentes Inducidas/citología , Laminina/farmacología , Infarto del Miocardio/terapia , Miocitos Cardíacos/efectos de los fármacos , Ingeniería de Tejidos , Animales , Apoptosis/genética , Hipoxia de la Célula/efectos de los fármacos , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Trasplante de Corazón , Humanos , Células Madre Pluripotentes Inducidas/fisiología , L-Lactato Deshidrogenasa/biosíntesis , Masculino , Contracción Miocárdica/fisiología , Miocitos Cardíacos/fisiología , Miocitos Cardíacos/trasplante , Neovascularización Fisiológica , ARN Mensajero/metabolismo , Ratas , Ratas Desnudas , Proteínas Recombinantes/farmacología , Volumen Sistólico , Ingeniería de Tejidos/métodos , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo , Remodelación Ventricular
6.
Sci Rep ; 10(1): 4593, 2020 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-32165680

RESUMEN

Avoiding immune rejection after allogeneic induced pluripotent stem cell-derived cardiomyocyte (iPSC-CM) transplantation is a concern. However, mesenchymal stem cells (MSCs) can suppress immune rejection. To determine whether MSC co-transplantation can reduce immune rejection after allogeneic iPSC-CM transplantation, the latter cell type, harbouring a luciferase transgene, was subcutaneously transplanted alone or together with syngeneic MSCs into BALB/c mice. Bioluminescence imaging revealed that MSC co-transplantation significantly improved graft survival (day 7: iPSC-CMs alone 34 ± 5%; iPSC-CMs with MSCs, 61 ± 7%; P = 0.008). MSC co-transplantation increased CD4 + CD25 + FOXP3 + regulatory T cell numbers, apoptotic CD8-positive T cells, and IL-10 and TGF-beta expression at the implantation site. Analysis using a regulatory T cell depletion model indicated that enhanced regulatory T cell populations in the iPSC-CM with MSC group partially contributed to the extended iPSC-CM survival. Further, MSCs affected activated lymphocytes directly through cell-cell contact, which reduced the CD8/CD4 ratio, the proportion of Th1-positive cells among CD4-positive cells, and the secretion of several inflammation-related cytokines. Syngeneic MSC co-transplantation might thus control allogeneic iPSC-CM rejection by mediating immune tolerance via regulatory T cells and cell-cell contact with activated lymphocytes; this approach has promise for cardiomyogenesis-based therapy using allogeneic iPSC-CMs for severe heart failure.


Asunto(s)
Tolerancia Inmunológica , Células Madre Pluripotentes Inducidas/citología , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Miocitos Cardíacos/trasplante , Animales , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Células Cultivadas , Supervivencia de Injerto , Células Madre Pluripotentes Inducidas/metabolismo , Luciferasas/genética , Luciferasas/metabolismo , Mediciones Luminiscentes , Células Madre Mesenquimatosas/inmunología , Ratones , Ratones Endogámicos BALB C , Miocitos Cardíacos/citología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/metabolismo , Linfocitos T Reguladores/metabolismo , Trasplante Homólogo , Trasplante Isogénico
7.
J Immunol ; 204(5): 1373-1385, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31969386

RESUMEN

Adoptive cellular therapy and its derivative, chimeric AgR T cell therapy, have achieved significant progress against cancer. Major barriers persist, however, including insufficient induction of cytotoxic T cells and exhaustion of tumor-infiltrating lymphocytes. In this study, we discovered a new role for 2-deoxy-d-glucose (2DG) in enhancing the antitumor activity of human T cells against NKG2D ligand-expressing tumor cells. Human T cells treated with 2DG upregulated the NK-specific transcription factors TOX2 and EOMES, thereby acquiring NK cell properties, including high levels of perforin/granzyme and increased sensitivity to IL-2. Notably, rather than inhibiting glycolysis, 2DG modified N-glycosylation, which augmented antitumor activity and cell surface retention of IL-2R of T cells. Moreover, 2DG treatment prevented T cells from binding to galectin-3, a potent tumor Ag associated with T cell anergy. Our results, therefore, suggest that modifying N-glycosylation of T cells with 2DG could improve the efficacy of T cell-based immunotherapies against cancer.


Asunto(s)
Desoxiglucosa/farmacología , Inmunidad Celular/efectos de los fármacos , Neoplasias/inmunología , Linfocitos T/inmunología , Glicosilación/efectos de los fármacos , Proteínas HMGB/inmunología , Humanos , Inmunoterapia , Interleucina-2/inmunología , Células K562 , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias/patología , Neoplasias/terapia , Proteínas de Dominio T Box/inmunología , Linfocitos T/patología
8.
EMBO J ; 39(2): e100875, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31821596

RESUMEN

Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are the most common cause of familial Parkinson's disease (PD). Impaired mitochondrial function is suspected to play a major role in PD. Nonetheless, the underlying mechanism by which impaired LRRK2 activity contributes to PD pathology remains unclear. Here, we identified the role of LRRK2 in endoplasmic reticulum (ER)-mitochondrial tethering, which is essential for mitochondrial bioenergetics. LRRK2 regulated the activities of E3 ubiquitin ligases MARCH5, MULAN, and Parkin via kinase-dependent protein-protein interactions. Kinase-active LRRK2(G2019S) dissociated from these ligases, leading to their PERK-mediated phosphorylation and activation, thereby increasing ubiquitin-mediated degradation of ER-mitochondrial tethering proteins. By contrast, kinase-dead LRRK2(D1994A)-bound ligases blocked PERK-mediated phosphorylation and activation of E3 ligases, thereby increasing the levels of ER-mitochondrial tethering proteins. Thus, the role of LRRK2 in the ER-mitochondrial interaction represents an important control point for cell fate and pathogenesis in PD.


Asunto(s)
Retículo Endoplásmico/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/metabolismo , eIF-2 Quinasa/metabolismo , Animales , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Ratones , Proteínas Mitocondriales/genética , Mutación , Fosforilación , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , eIF-2 Quinasa/genética
10.
Sci Rep ; 9(1): 10840, 2019 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-31346220

RESUMEN

Transplantation of cardiomyocytes derived from induced pluripotent stem cell (iPSC-CMs) is a promising approach for increasing functional CMs during end-stage heart failure. Although major histocompatibility complex (MHC) class I matching between donor cells and recipient could reduce acquired immune rejection, innate immune responses may have negative effects on transplanted iPSC-CMs. Here, we demonstrated that natural killer cells (NKCs) infiltrated in iPSC-CM transplants even in a syngeneic mouse model. The depletion of NKCs using an anti-NKC antibody rescued transplanted iPSC-CMs, suggesting that iPSC-CMs activated NKC-mediated innate immunity. Surprisingly, iPSC-CMs lost inhibitory MHCs but not activating ligands for NKCs. Re-expression of MHC class I induced by IFN-γ as well as suppression of activating ligands by an antibody rescued the transplanted iPSC-CMs. Thus, NKCs impede the engraftment of transplanted iPSC-CMs because of lost MHC class I, and our results provide a basis for an approach to improve iPSC-CM engraftment.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Células Asesinas Naturales/citología , Miocitos Cardíacos/trasplante , Trasplante de Células Madre , Animales , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Supervivencia Celular/fisiología , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/terapia , Inmunidad Innata , Ratones
11.
Mol Ther ; 26(11): 2681-2695, 2018 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-30217728

RESUMEN

In this study, we proposed that the functionality or phenotype of differentiated cardiomyocytes derived from human induced pluripotent stem cells (iPSC-CMs) might be modified by co-culture with mesenchymal stem cells (MSCs), resulting in an improved therapeutic potential for failing myocardial tissues. Structural, motility, electrophysiological, and metabolic analyses revealed that iPSC-CMs co-cultured with MSCs displayed aligned myofibrils with A-, H-, and I-bands that could contract and relax quickly, indicating the promotion of differentiation and the establishment of the iPSC-CM structural framework, and showed clear gap junctions and an electric pacing of >2 Hz, indicating enhanced cell-cell interactions. In addition, soluble factors excreted by MSCs, including several cytokines and exosomes, enhanced cardiomyocyte-specific marker production, produced more energy under normal and stressed conditions, and reduced reactive oxygen species production by iPSC-CMs under stressed condition. Notably, gene ontology and pathway analysis revealed that microRNAs and proteins in the exosomes impacted the functionality and maturation of iPSC-CMs. Furthermore, cell sheets consisting of a mixture of iPSC-CMs and MSCs showed longer survival and enhanced therapeutic effects compared with those consisting of iPSC-CMs alone. This may lead to a new type of iPSC-based cardiomyogenesis therapy for patients with heart failure.


Asunto(s)
Diferenciación Celular/genética , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Mesenquimatosas/citología , Miocitos Cardíacos/metabolismo , Comunicación Celular/genética , Células Cultivadas , Técnicas de Cocultivo , Exosomas/genética , Exosomas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Mesenquimatosas/metabolismo , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/citología
12.
Nat Immunol ; 19(6): 561-570, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29777213

RESUMEN

Polarization of macrophages into pro-inflammatory or anti-inflammatory states has distinct metabolic requirements, with mechanistic target of rapamycin (mTOR) kinase signaling playing a critical role. However, it remains unclear how mTOR regulates metabolic status to promote polarization of these cells. Here we show that an mTOR-Semaphorin 6D (Sema6D)-Peroxisome proliferator receptor γ (PPARγ) axis plays critical roles in macrophage polarization. Inhibition of mTOR or loss of Sema6D blocked anti-inflammatory macrophage polarization, concomitant with severe impairments in PPARγ expression, uptake of fatty acids, and lipid metabolic reprogramming. Macrophage expression of the receptor Plexin-A4 is responsible for Sema6D-mediated anti-inflammatory polarization. We found that a tyrosine kinase, c-Abl, which associates with the cytoplasmic region of Sema6D, is required for PPARγ expression. Furthermore, Sema6D is important for generation of intestinal resident CX3CR1hi macrophages and prevents development of colitis. Collectively, these findings highlight crucial roles for Sema6D reverse signaling in macrophage polarization, coupling immunity, and metabolism via PPARγ.


Asunto(s)
Inflamación/metabolismo , Metabolismo de los Lípidos/inmunología , Macrófagos/metabolismo , PPAR gamma/metabolismo , Semaforinas/metabolismo , Animales , Diferenciación Celular/inmunología , Colitis/inmunología , Inflamación/inmunología , Macrófagos/citología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , PPAR gamma/inmunología , Semaforinas/inmunología , Transducción de Señal/inmunología , Serina-Treonina Quinasas TOR/inmunología , Serina-Treonina Quinasas TOR/metabolismo
14.
Sci Rep ; 6: 25738, 2016 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-27166870

RESUMEN

B-cell receptor (BCR) signaling plays a critical role in B-cell activation and humoral immunity. In this study, we discovered a critical function of leucine-rich repeat kinase 1 (LRRK1) in BCR-mediated immune responses. Lrrk1(-/-) mice exhibited altered B1a-cell development and basal immunoglobulin production. In addition, these mice failed to produce IgG3 antibody in response to T cell-independent type 2 antigen due to defects in IgG3 class-switch recombination. Concomitantly, B cells lacking LRRK1 exhibited a profound defect in proliferation and survival upon BCR stimulation, which correlated with impaired BCR-mediated NF-κB activation and reduced expression of NF-κB target genes including Bcl-xL, cyclin D2, and NFATc1/αA. Furthermore, LRRK1 physically interacted and potently synergized with CARMA1 to enhance NF-κB activation. Our results reveal a critical role of LRRK1 in NF-κB signaling in B cells and the humoral immune response.


Asunto(s)
Linfocitos B/metabolismo , Proteínas Adaptadoras de Señalización CARD/metabolismo , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Formación de Anticuerpos , Linfocitos B/citología , Proliferación Celular , Supervivencia Celular , Endocitosis , Células HEK293 , Humanos , Cambio de Clase de Inmunoglobulina , Inmunoglobulina G/metabolismo , Activación de Linfocitos , Cavidad Peritoneal/citología , Proteínas Serina-Treonina Quinasas/deficiencia , Receptores de Antígenos de Linfocitos B/metabolismo , Recombinación Genética/genética , Bazo/citología , Linfocitos T/metabolismo
15.
Mol Cell Biol ; 35(17): 3044-58, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26100023

RESUMEN

Autophagy is a conserved process that enables catabolic and degradative pathways. Rab family proteins, which are active in the GTP-bound form, regulate the transport and fusion of autophagosomes. However, it remains unclear how each cycle of Rab activation and inactivation is precisely regulated. Here, we show that leucine-rich repeat kinase 1 (LRRK1) regulates autophagic flux by controlling Rab7 activity in autolysosome formation. Upon induction of autophagy, LRRK1 was recruited via an association with VAMP7 to the autolysosome, where it activated the Rab7 GTPase-activating protein (GAP) TBC1D2, thereby switching off Rab7 signaling. Consistent with this model, LRRK1 deletion caused mice to be vulnerable to starvation and disrupted autolysosome formation, as evidenced by the accumulation of enlarged autolysosomes with undegraded LC3-II and persistently high levels of Rab7-GTP. This defect in autophagic flux was partially rescued by a mutant form of TBC1D2 with elevated Rab7-GAP activity. Thus, the spatiotemporal regulation of Rab7 activity during tunicamycin-induced autophagy is regulated by LRRK1.


Asunto(s)
Autofagia/genética , Proteínas Activadoras de GTPasa/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas de Unión al GTP rab/metabolismo , Animales , Autofagia/efectos de los fármacos , Células Cultivadas , Activación Enzimática , Proteínas Activadoras de GTPasa/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Lisosomas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptidos/análisis , Neuropéptidos/metabolismo , Fagosomas/fisiología , Proteínas R-SNARE/genética , Proteínas R-SNARE/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Tunicamicina , Proteínas de Unión al GTP rab/análisis , Proteínas de Unión a GTP rab7 , Proteína de Unión al GTP rac1/análisis , Proteína de Unión al GTP rac1/metabolismo
16.
J Immunol ; 195(3): 934-43, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26116513

RESUMEN

Mammalian target of rapamycin (mTOR) plays crucial roles in activation and differentiation of diverse types of immune cells. Although several lines of evidence have demonstrated the importance of mTOR-mediated signals in CD4(+) T cell responses, the involvement of mTOR in CD8(+) T cell responses is not fully understood. In this study, we show that a class IV semaphorin, SEMA4A, regulates CD8(+) T cell activation and differentiation through activation of mTOR complex (mTORC) 1. SEMA4A(-/-) CD8(+) T cells exhibited impairments in production of IFN-γ and TNF-α and induction of the effector molecules granzyme B, perforin, and FAS-L. Upon infection with OVA-expressing Listeria monocytogenes, pathogen-specific effector CD8(+) T cell responses were significantly impaired in SEMA4A(-/-) mice. Furthermore, SEMA4A(-/-) CD8(+) T cells exhibited reduced mTORC1 activity and elevated mTORC2 activity, suggesting that SEMA4A is required for optimal activation of mTORC1 in CD8(+) T cells. IFN-γ production and mTORC1 activity in SEMA4A(-/-) CD8(+) T cells were restored by administration of recombinant Sema4A protein. In addition, we show that plexin B2 is a functional receptor of SEMA4A in CD8(+) T cells. Collectively, these results not only demonstrate the role of SEMA4A in CD8(+) T cells, but also reveal a novel link between a semaphorin and mTOR signaling.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Activación de Linfocitos/inmunología , Complejos Multiproteicos/inmunología , Proteínas del Tejido Nervioso/fisiología , Semaforinas/fisiología , Serina-Treonina Quinasas TOR/inmunología , Animales , Linfocitos T CD8-positivos/citología , Diferenciación Celular/inmunología , Proliferación Celular , Proteína Ligando Fas/biosíntesis , Granzimas/biosíntesis , Interferón gamma/biosíntesis , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejos Multiproteicos/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas Citotóxicas Formadoras de Poros/biosíntesis , Unión Proteica/inmunología , Interferencia de ARN , ARN Interferente Pequeño , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Semaforinas/genética , Transducción de Señal/inmunología , Serina-Treonina Quinasas TOR/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis
17.
Autoimmune Dis ; 2013: 183487, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24222845

RESUMEN

Objective. To evaluate what types of DNA damages are detected in rheumatoid arthritis (RA). Methods. The DNA adducts such as 8-oxo-hydroxy-7,8-dihydro-2'-deoxyguanosine (8-oxo-dG), 1,N(6)-etheno-2'-deoxyadenosine ( ε dA), and heptanone-etheno-2'-deoxycytidine (H ε dC) in genomic DNAs, derived from whole blood cells from 46 RA patients and 31 healthy controls, were analyzed by high-performance liquid chromatography tandem mass spectrometry, and their levels in RA patients and controls were compared. In addition, correlation between DNA adducts and clinical parameters of RA was analyzed. Results. Compared with controls, the levels of H ε dC in RA were significantly higher (P < 0.0001) and age dependent (r = 0.43, P < 0.01), while there was no significant difference in 8-oxo-dG and ε dA accumulation between RA patients and controls. H ε dC levels correlated well with the number of swollen joints (r = 0.57, P < 0.0001) and weakly with the number of tender joints (r = 0.26, P = 0.08) of RA patients, while they did not show a significant association with serological markers such as C-reactive protein and matrix metalloproteinase 3. Conclusion. These findings indicate that H ε dC may have some influence on the development of RA and/or its complications.

18.
Nat Commun ; 4: 1406, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23360997

RESUMEN

Semaphorin 4A (Sema4A) has an essential role in photoreceptor survival. In humans, mutations in Sema4A are thought to contribute to retinal degenerative diseases. Here we generate a series of knock-in mouse lines with corresponding mutations (D345H, F350C or R713Q) in the Sema4A gene and find that Sema4A(F350C) causes retinal degeneration phenotypes. The F350C mutation results in abnormal localization of the Sema4A protein, leading to impaired endosomal sorting of molecules indispensable for photoreceptor survival. Additionally, protein structural modelling reveals that the side chain of the 350th amino acid is critical to retain the proper protein conformation. Furthermore, Sema4A gene transfer successfully prevents photoreceptor degeneration in Sema4A(F350C/F350C) and Sema4A(-/-) mice. Thus, our findings not only indicate the importance of the Sema4A protein conformation in human and mouse retina homeostasis but also identify a novel therapeutic target for retinal degenerative diseases.


Asunto(s)
Endosomas/metabolismo , Mutación Puntual/genética , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Semaforinas/genética , Semaforinas/metabolismo , Animales , Células COS , Chlorocebus aethiops , Endosomas/efectos de la radiación , Técnicas de Sustitución del Gen , Técnicas de Transferencia de Gen , Genes Dominantes , Sitios Genéticos , Humanos , Luz , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Modelos Moleculares , Células Fotorreceptoras de Vertebrados/patología , Células Fotorreceptoras de Vertebrados/efectos de la radiación , Transporte de Proteínas/efectos de la radiación , Degeneración Retiniana/prevención & control , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Epitelio Pigmentado de la Retina/efectos de la radiación , Semaforinas/química , Coloración y Etiquetado
19.
J Immunol ; 188(10): 4858-65, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22491253

RESUMEN

Multiple sclerosis (MS) is a demyelinating autoimmune disease of the CNS and a leading cause of lasting neurologic disabilities in young adults. Although the precise mechanism remains incompletely understood, Ag presentation and subsequent myelin-reactive CD4(+) T cell activation/differentiation are essential for the pathogenesis of MS. Although semaphorins were initially identified as axon guidance cues during neural development, several semaphorins are crucially involved in various phases of immune responses. Sema4A is one of the membrane-type class IV semaphorins, which we originally identified from the cDNA library of dendritic cell (DC). Sema4A plays critical roles in T cell activation and Th1 differentiation during the course of experimental autoimmune encephalomyelitis, an animal model of MS; however, its pathological involvement in human MS has not been determined. In this study, we report that Sema4A is increased in the sera of patients with MS. The expression of Sema4A is increased on DCs in MS patients and shed from these cells in a metalloproteinase-dependent manner. DC-derived Sema4A is not only critical for Th1 but also for Th17 cell differentiation, and MS patients with high Sema4A levels exhibit Th17 skewing. Furthermore, patients with high Sema4A levels have more severe disabilities and are unresponsive to IFN-ß treatment. Taken together, our results suggest that Sema4A is involved in the pathogenesis of MS by promoting Th17 skewing.


Asunto(s)
Diferenciación Celular/inmunología , Interferón beta/uso terapéutico , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/terapia , Semaforinas/biosíntesis , Células Th17/inmunología , Regulación hacia Arriba/inmunología , Secuencia de Aminoácidos , Animales , Diferenciación Celular/genética , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/patología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Esclerosis Múltiple/patología , Ratas , Semaforinas/sangre , Semaforinas/deficiencia , Semaforinas/metabolismo , Células Th17/metabolismo , Células Th17/patología , Regulación hacia Arriba/genética
20.
Genes Dev ; 26(8): 816-29, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22465952

RESUMEN

Photoreceptor cell death is the hallmark of a group of human inherited retinal degeneration. Although the causative genetic mutations are often known, the mechanisms leading to photoreceptor degeneration remain poorly defined. Here, we show that Semaphorin 4A (Sema4A), a member of axonal guidance molecule semaphorin, plays a role in Rab11/FIP2-mediated endosomal sorting in retinal pigment epithelial cells to support photoreceptor function. In response to oxidative stress, Sema4A switches the endosomal sorting of the lysosomal precursor protein prosaposin from the lysosome to the exosomal release, which prevents light-induced photoreceptor apoptosis. In the absence of oxidative stress, Sema4A sorts retinoid-binding proteins with retinoids between the cell surface and endoplasmic reticulum, by which 11-cis-retinal, a chromophore for phototransduction, is regenerated and transported back to photoreceptors. Owing to defects in these processes, Sema4A-deficient mice exhibit marked photoreceptor degeneration. Our findings therefore indicate that Sema4A regulates two distinct endosomal-sorting pathways that are critical for photoreceptor survival and phototransduction during the transition between daylight and darkness.


Asunto(s)
Endosomas/metabolismo , Células Fotorreceptoras de Vertebrados/citología , Epitelio Pigmentado de la Retina/metabolismo , Semaforinas/metabolismo , Animales , Proteínas de Ciclo Celular , Supervivencia Celular , Proteínas del Ojo/metabolismo , Luz , Proteínas de Transporte de Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Células Fotorreceptoras de Vertebrados/metabolismo , Saposinas/metabolismo , Proteínas de Unión al GTP rab/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA