Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Blood ; 142(5): 421-433, 2023 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-37146250

RESUMEN

Although BCL2 mutations are reported as later occurring events leading to venetoclax resistance, many other mechanisms of progression have been reported though remain poorly understood. Here, we analyze longitudinal tumor samples from 11 patients with disease progression while receiving venetoclax to characterize the clonal evolution of resistance. All patients tested showed increased in vitro resistance to venetoclax at the posttreatment time point. We found the previously described acquired BCL2-G101V mutation in only 4 of 11 patients, with 2 patients showing a very low variant allele fraction (0.03%-4.68%). Whole-exome sequencing revealed acquired loss(8p) in 4 of 11 patients, of which 2 patients also had gain (1q21.2-21.3) in the same cells affecting the MCL1 gene. In vitro experiments showed that CLL cells from the 4 patients with loss(8p) were more resistant to venetoclax than cells from those without it, with the cells from 2 patients also carrying gain (1q21.2-21.3) showing increased sensitivity to MCL1 inhibition. Progression samples with gain (1q21.2-21.3) were more susceptible to the combination of MCL1 inhibitor and venetoclax. Differential gene expression analysis comparing bulk RNA sequencing data from pretreatment and progression time points of all patients showed upregulation of proliferation, B-cell receptor (BCR), and NF-κB gene sets including MAPK genes. Cells from progression time points demonstrated upregulation of surface immunoglobulin M and higher pERK levels compared with those from the preprogression time point, suggesting an upregulation of BCR signaling that activates the MAPK pathway. Overall, our data suggest several mechanisms of acquired resistance to venetoclax in CLL that could pave the way for rationally designed combination treatments for patients with venetoclax-resistant CLL.


Asunto(s)
Antineoplásicos , Leucemia Linfocítica Crónica de Células B , Humanos , Antineoplásicos/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Resistencia a Antineoplásicos/genética , Secuenciación del Exoma , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/patología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteínas Proto-Oncogénicas c-bcl-2
3.
Blood Cancer J ; 13(1): 22, 2023 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-36732326

RESUMEN

PI3Kδ inhibitors are approved for the therapy of B cell malignancies, but their clinical use has been limited by unpredictable autoimmune toxicity, despite promising efficacy and evidence that toxicity is associated with improved clinical outcomes. Prior phenotypic evaluation by CyTOF has identified increases in activated CD8 T cells with activation of Th17 T cells, as well as decreases in Tregs, particularly in patients with toxicity. Here we sought to further understand the effects of idelalisib and duvelisib in vitro, and demonstrate that both idelalisib and duvelisib can inhibit T cell proliferation as well as Th1 and Treg differentiation in vitro, while promoting Th2 and Th17 differentiation. We further demonstrate directly using intracellular flow cytometry that autoimmune toxicity in patients is associated with higher absolute numbers of CD4 and CD8 T cells with Th17 differentiation in peripheral blood prior to therapy, and that gastrointestinal tissues from patients with active autoimmune complications of PI3Kδ inhibitors show infiltration with Th17+ T cells. These same tissues show depletion of Tregs as compared to CLL patients without toxicity, suggesting that loss of Tregs may be permissive for Th17 activation to lead to autoimmune toxicity. Clinical trials to restore this balance are warranted.


Asunto(s)
Linfocitos T Reguladores , Células Th17 , Humanos , Linfocitos T CD8-positivos , Diferenciación Celular , Citometría de Flujo
4.
Nat Commun ; 13(1): 2810, 2022 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-35589724

RESUMEN

Cellular therapies offer a promising therapeutic strategy for the highly malignant brain tumor, glioblastoma (GBM). However, their clinical translation is limited by the lack of effective target identification and stringent testing in pre-clinical models that replicate standard treatment in GBM patients. In this study, we show the detection of cell surface death receptor (DR) target on CD146-enriched circulating tumor cells (CTC) captured from the blood of mice bearing GBM and patients diagnosed with GBM. Next, we developed allogeneic "off-the-shelf" clinical-grade bifunctional mesenchymal stem cells (MSCBif) expressing DR-targeted ligand and a safety kill switch. We show that biodegradable hydrogel encapsulated MSCBif (EnMSCBif) has a profound therapeutic efficacy in mice bearing patient-derived invasive, primary and recurrent GBM tumors following surgical resection. Activation of the kill switch enhances the efficacy of MSCBif and results in their elimination post-tumor treatment which can be tracked by positron emission tomography (PET) imaging. This study establishes a foundation towards a clinical trial of EnMSCBif in primary and recurrent GBM patients.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Trasplante de Células Madre Hematopoyéticas , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/terapia , Línea Celular Tumoral , Glioblastoma/tratamiento farmacológico , Glioblastoma/terapia , Humanos , Ratones , Recurrencia Local de Neoplasia/terapia
5.
Blood ; 138(1): 44-56, 2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-33684943

RESUMEN

Inhibitors of Bruton tyrosine kinase (BTK) and phosphatidylinositol 3-kinase δ (PI3Kδ) that target the B-cell receptor (BCR) signaling pathway have revolutionized the treatment of chronic lymphocytic leukemia (CLL). Mutations associated with resistance to BTK inhibitors have been identified, but limited data are available on mechanisms of resistance to PI3Kδ inhibitors. Here we present findings from longitudinal whole-exome sequencing of cells from patients with multiply relapsed CLL (N = 28) enrolled in trials of PI3K inhibitors. The nonresponder subgroup was characterized by baseline activating mutations in MAP2K1, BRAF, and KRAS genes in 60% of patients. PI3Kδ inhibition failed to inhibit ERK phosphorylation (pERK) in nonresponder CLL cells with and without mutations, whereas treatment with a MEK inhibitor rescued ERK inhibition. Overexpression of MAP2K1 mutants in vitro led to increased basal and inducible pERK and resistance to idelalisib. These data demonstrate that MAPK/ERK activation plays a key role in resistance to PI3Kδ inhibitors in CLL and provide a rationale for therapy with a combination of PI3Kδ and ERK inhibitors.


Asunto(s)
Resistencia a Antineoplásicos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/enzimología , Sistema de Señalización de MAP Quinasas , Inhibidores de las Quinasa Fosfoinosítidos-3/uso terapéutico , Adulto , Anciano , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Genoma Humano , Humanos , Leucemia Linfocítica Crónica de Células B/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Persona de Mediana Edad , Mutación/genética , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Purinas/farmacología , Purinas/uso terapéutico , Quinazolinonas/farmacología , Quinazolinonas/uso terapéutico , Resultado del Tratamiento , Regulación hacia Arriba/genética
6.
Cells ; 10(3)2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-33668856

RESUMEN

Immunotherapy for brain tumors remains elusive, unlike many other cancer types for which it is one of the most promising therapeutic options. Recent studies have comprehensively profiled the immune-landscape of the highly malignant brain tumor, glioblastoma (GBM) in patients and identified novel immune-modulatory targets. However, given that pre-clinical exploration of potential novel therapeutics is primarily performed in immune-competent mice, it is vital to compare the immune-profiling data obtained from syngeneic mouse GBM models with GBM patient samples. This will pave the way for utilizing appropriate clinically relevant mouse GBM models for evaluating novel immune-therapies in pre-clinical settings. Recent brain tumor immune-profiling studies using state-of-the-art time of flight cytometry (CyTOF) analysis compared different human and mouse GBM types and reported immunological distinctions amongst these mouse models. These studies also contrast the immune phenotype of brain tumor patients with commonly used pre-clinical immune-competent mouse models. In this perspective, we provide the outcomes of very recent brain tumor immune-profiling studies and their implications on designing and translating unique, tumor-subtype specific therapeutics.


Asunto(s)
Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/terapia , Glioblastoma/inmunología , Glioblastoma/terapia , Inmunoterapia , Investigación Biomédica Traslacional , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Microambiente Tumoral/inmunología
7.
Stem Cells ; 39(6): 707-722, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33586320

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic has grown to be a global public health crisis with no safe and effective treatments available yet. Recent findings suggest that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the coronavirus pathogen that causes COVID-19, could elicit a cytokine storm that drives edema, dysfunction of the airway exchange, and acute respiratory distress syndrome in the lung, followed by acute cardiac injury and thromboembolic events leading to multiorgan failure and death. Mesenchymal stem cells (MSCs), owing to their powerful immunomodulatory abilities, have the potential to attenuate the cytokine storm and have therefore been proposed as a potential therapeutic approach for which several clinical trials are underway. Given that intravenous infusion of MSCs results in a significant trapping in the lung, MSC therapy could directly mitigate inflammation, protect alveolar epithelial cells, and reverse lung dysfunction by normalizing the pulmonary microenvironment and preventing pulmonary fibrosis. In this review, we present an overview and perspectives of the SARS-CoV-2 induced inflammatory dysfunction and the potential of MSC immunomodulation for the prevention and treatment of COVID-19 related pulmonary disease.


Asunto(s)
COVID-19/inmunología , Síndrome de Liberación de Citoquinas/inmunología , Células Madre Mesenquimatosas/inmunología , SARS-CoV-2/inmunología , COVID-19/terapia , COVID-19/virología , Síndrome de Liberación de Citoquinas/terapia , Síndrome de Liberación de Citoquinas/virología , Humanos , Inmunomodulación , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/virología , Pandemias , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/terapia , Fibrosis Pulmonar/virología , Síndrome de Dificultad Respiratoria/inmunología , Síndrome de Dificultad Respiratoria/terapia , Síndrome de Dificultad Respiratoria/virología , SARS-CoV-2/genética
8.
Nat Commun ; 11(1): 3912, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32764562

RESUMEN

Immunotherapy has emerged as a promising approach to treat cancer, however, its efficacy in highly malignant brain-tumors, glioblastomas (GBM), is limited. Here, we generate distinct imageable syngeneic mouse GBM-tumor models and utilize RNA-sequencing, CyTOF and correlative immunohistochemistry to assess immune-profiles in these models. We identify immunologically-inert and -active syngeneic-tumor types and show that inert tumors have an immune-suppressive phenotype with numerous exhausted CD8 T cells and resident macrophages; fewer eosinophils and SiglecF+ macrophages. To mimic the clinical-settings of first line of GBM-treatment, we show that tumor-resection invigorates an anti-tumor response via increasing T cells, activated microglia and SiglecF+ macrophages and decreasing resident macrophages. A comparative CyTOF analysis of resected-tumor samples from GBM-patients and mouse GBM-tumors show stark similarities in one of the mouse GBM-tumors tested. These findings guide informed choices for use of GBM models for immunotherapeutic interventions and offer a potential to facilitate immune-therapies in GBM patients.


Asunto(s)
Neoplasias Encefálicas/inmunología , Glioblastoma/inmunología , Animales , Encéfalo/inmunología , Encéfalo/patología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Línea Celular Tumoral , Glioblastoma/patología , Glioblastoma/terapia , Humanos , Tolerancia Inmunológica , Inmunofenotipificación , Inmunoterapia , Isoinjertos , Linfocitos Infiltrantes de Tumor/clasificación , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/patología , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Microambiente Tumoral/inmunología
9.
Mol Cell ; 78(6): 1207-1223.e8, 2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32504554

RESUMEN

Tumor interferon (IFN) signaling promotes PD-L1 expression to suppress T cell-mediated immunosurveillance. We identify the IFN-stimulated non-coding RNA 1 (INCR1) as a long noncoding RNA (lncRNA) transcribed from the PD-L1 locus and show that INCR1 controls IFNγ signaling in multiple tumor types. Silencing INCR1 decreases the expression of PD-L1, JAK2, and several other IFNγ-stimulated genes. INCR1 knockdown sensitizes tumor cells to cytotoxic T cell-mediated killing, improving CAR T cell therapy. We discover that PD-L1 and JAK2 transcripts are negatively regulated by binding to HNRNPH1, a nuclear ribonucleoprotein. The primary transcript of INCR1 binds HNRNPH1 to block its inhibitory effects on the neighboring genes PD-L1 and JAK2, enabling their expression. These findings introduce a mechanism of tumor IFNγ signaling regulation mediated by the lncRNA INCR1 and suggest a therapeutic target for cancer immunotherapy.


Asunto(s)
Antígeno B7-H1/genética , Interferón gamma/metabolismo , ARN Largo no Codificante/genética , Anciano , Animales , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inmunoterapia , Inmunoterapia Adoptiva/métodos , Interferón gamma/genética , Interferones/genética , Interferones/metabolismo , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Masculino , Ratones , Ratones Endogámicos NOD , Persona de Mediana Edad , Proteína 2 Ligando de Muerte Celular Programada 1/genética , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/efectos de los fármacos , Linfocitos T Citotóxicos
10.
Immunology ; 160(3): 295-309, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32187647

RESUMEN

Activated T-cells make both interleukin-2 (IL2) and its high-affinity receptor component CD25. Regulatory CD4 T-cells (Treg cells) do not make IL2, and the IL2-CD25 circuit is considered a paracrine circuit crucial in their generation and maintenance. Yet, all T-cells are capable of making IL2 at some stage during differentiation, making a cell-intrinsic autocrine circuit additionally possible. When we re-visited experiments with mixed bone marrow chimeras using a wide range of ratios of wild-type (WT) and IL2-/- genotype progenitors, we found that, as expected, thymic Treg cells were almost equivalent between WT and IL2-/- genotypes at ratios with WT prominence. However, at WT-limiting ratios, the IL2-/- genotype showed lower thymic Treg frequencies, indicating a role for cell-intrinsic autocrine IL2 in thymic Treg generation under IL2-limiting conditions. Further, peripheral IL2-/- naive CD4 T-cells showed poor conversion to inducible Tregs (pTregs) both in vivo and in vitro, again indicating a significant role for cell-intrinsic autocrine IL2 in their generation. Peripherally, the IL2-/- genotype was less prominent at all WT:IL2-/- ratios among both thymic Tregs (tTregs) and pTregs, adoptively transferred IL2-/- Tregs showed poorer survival than WT Tregs did, and RNA-seq analysis of WT and IL2-/- Tregs showed interesting differences in the T-cell receptor and transforming growth factor-beta-bone morphogenetic protein-JNK pathways between them, suggesting a non-titrating role for cell-intrinsic autocrine IL2 in Treg programming. These data indicate that cell-intrinsic autocrine IL2 plays significant roles in Treg generation and maintenance.


Asunto(s)
Interleucina-2/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Comunicación Autocrina , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Homeostasis , Interleucina-2/genética , Activación de Linfocitos , MAP Quinasa Quinasa 4/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Quimera por Trasplante
11.
Immunology ; 158(2): 104-120, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31318442

RESUMEN

Activation of B and T lymphocytes leads to major remodelling of the metabolic landscape of the cells enabling their post-activation functions. However, naive B and T lymphocytes also show metabolic differences, and the genesis, nature and functional significance of these differences are not yet well understood. Here we show that resting B-cells appeared to have lower energy demands than resting T-cells as they consumed lower levels of glucose and fatty acids and produced less ATP. Resting B-cells are more dependent on OXPHOS, while T-cells show more dependence on aerobic glycolysis. However, despite an apparently higher energy demand, T lineage cells showed lower rates of protein synthesis than equivalent B lineage stages. These metabolic differences between the two lineages were established early during lineage differentiation, and were functionally significant. Higher levels of protein synthesis in B-cells were associated with increased synthesis of MHC class II molecules and other proteins associated with antigen internalization, transport and presentation. The combination of higher energy demand and lower protein synthesis in T-cells was consistent with their higher ATP-dependent motility. Our data provide an integrated perspective of the metabolic differences and their functional implications between the B and T lymphocyte lineages.


Asunto(s)
Linfocitos B/metabolismo , Glucólisis/inmunología , Fosforilación Oxidativa , Linfocitos T/metabolismo , Adenosina Trifosfato/biosíntesis , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Diferenciación Celular/inmunología , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Ácidos Grasos/metabolismo , Expresión Génica , Glucosa/metabolismo , Glucólisis/genética , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Inmunofenotipificación , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Especificidad de Órganos , Cultivo Primario de Células , Biosíntesis de Proteínas/inmunología , Linfocitos T/citología , Linfocitos T/inmunología
12.
Clin Cancer Res ; 23(22): 7047-7058, 2017 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-28912136

RESUMEN

Purpose: Despite tumor resection being the first-line clinical care for glioblastoma (GBM) patients, nearly all preclinical immune therapy models intend to treat established GBM. Characterizing cytoreductive surgery-induced immune response combined with the administration of immune cytokines has the potential of offering a new treatment paradigm of immune therapy for GBMs.Experimental Design: We developed syngeneic orthotopic mouse GBM models of tumor resection and characterized the immune response of intact and resected tumors. We also created a highly secretable variant of immune cytokine IFNß to enhance its release from engineered mouse mesenchymal stem cells (MSC-IFNß) and assessed whether surgical resection of intracranial GBM tumor significantly enhanced the antitumor efficacy of targeted on-site delivery of encapsulated MSC-IFNß.Results: We show that tumor debulking results in substantial reduction of myeloid-derived suppressor cells (MDSC) and simultaneous recruitment of CD4/CD8 T cells. This immune response significantly enhanced the antitumor efficacy of locally delivered encapsulated MSC-IFNß via enhanced selective postsurgical infiltration of CD8 T cells and directly induced cell-cycle arrest in tumor cells, resulting in increased survival of mice. Utilizing encapsulated human MSC-IFNß in resected orthotopic tumor xenografts of patient-derived GBM, we further show that IFNß induces cell-cycle arrest followed by apoptosis, resulting in increased survival in immunocompromised mice despite their absence of an intact immune system.Conclusions: This study demonstrates the importance of syngeneic tumor resection models in developing cancer immunotherapies and emphasizes the translational potential of local delivery of immunotherapeutic agents in treating cancer. Clin Cancer Res; 23(22); 7047-58. ©2017 AACR.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/inmunología , Interferón beta/genética , Células Madre/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Quimiotaxis de Leucocito/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Glioblastoma/patología , Glioblastoma/terapia , Humanos , Interferón beta/metabolismo , Ratones , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase S del Ciclo Celular/genética , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
13.
J Immunol ; 198(5): 1823-1837, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28100678

RESUMEN

T cell response magnitudes increase with increasing antigenic dosage. However, it is unclear whether ligand density only modulates the proportions of responding ligand-specific T cells or also alters responses at the single cell level. Using brief (3 h) exposure of TCR-transgenic mouse CD8 T cells in vitro to varying densities of cognate peptide-MHC ligand followed by ligand-free culture in IL-2, we found that ligand density determined the frequencies of responding cells but not the expression levels of the early activation marker molecule, CD69. Cells with low glucose uptake capacity and low protein synthesis rates were less ligand-sensitive, implicating metabolic competence in the response heterogeneity of CD8 T cell populations. Although most responding cells proliferated, ligand density was associated with time of entry into proliferation and with the extent of cell surface TCR downmodulation. TCR internalization was associated, regardless of the ligand density, with rapidity of c-myc induction, loss of the cell cycle inhibitor p27kip1, metabolic reprogramming, and cell cycle entry. A low affinity peptide ligand behaved, regardless of ligand density, like a low density, high affinity ligand in all these parameters. Inhibition of signaling after ligand exposure selectively delayed proliferation in cells with internalized TCRs. Finally, internalized TCRs continued to signal and genetic modification of TCR internalization and trafficking altered the duration of signaling in a T cell hybridoma. Together, our findings indicate that heterogeneity among responding CD8 T cell populations in their ability to respond to TCR-mediated stimulation and internalize TCRs mediates detection of ligand density or affinity, contributing to graded response magnitudes.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos T/genética , Antígenos de Diferenciación de Linfocitos T/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Línea Celular , Células Dendríticas/inmunología , Glucosa/metabolismo , Interleucina-2/farmacología , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Ligandos , Activación de Linfocitos/inmunología , Ratones , Péptidos/metabolismo , Péptidos/farmacología , Receptores de Antígenos de Linfocitos T/inmunología
14.
Mol Cell Biol ; 35(17): 2932-46, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26100020

RESUMEN

Posttranslational modifications (PTMs) can alter many fundamental properties of a protein. One or combinations of them have been known to regulate the dynamics of many cellular pathways and consequently regulate all vital processes. Understandably, pathogens have evolved sophisticated strategies to subvert these mechanisms to achieve instantaneous control over host functions. Here, we present the first report of modulation by intestinal pathogen Salmonella enterica serovar Typhimurium (S. Typhimurium) of host SUMOylation, a PTM pathway central to all fundamental cellular processes. Both in cell culture and in a mouse model, we observed that S. Typhimurium infection led to a dynamic SUMO-conjugated proteome alteration. The intracellular survival of S. Typhimurium was dependent on SUMO status as revealed by reduced infection and Salmonella-induced filaments (SIFs) in SUMO-upregulated cells. S. Typhimurium-dependent SUMO modulation was seen as a result of depletion of crucial SUMO pathway enzymes Ubc-9 and PIAS1, at both the protein and the transcript levels. Mechanistically, depletion of Ubc-9 relied on upregulation of small noncoding RNAs miR30c and miR30e during S. Typhimurium infection. This was necessary and sufficient for both down-modulation of Ubc-9 and a successful infection. Thus, we demonstrate a novel strategy of pathogen-mediated perturbation of host SUMOylation, an integral mechanism underlying S. Typhimurium infection and intracellular survival.


Asunto(s)
MicroARNs/genética , Proteínas Inhibidoras de STAT Activados/metabolismo , Salmonella typhimurium/patogenicidad , Sumoilación/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo , Animales , Línea Celular Tumoral , Femenino , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Ratones , Ratones Endogámicos C57BL , Infecciones por Salmonella/patología
15.
J Biol Chem ; 290(7): 4131-48, 2015 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-25512377

RESUMEN

Amyloids are highly organized protein aggregates that arise from inappropriately folded versions of proteins or polypeptides under both physiological as well as simulated ambiences. Once thought to be irreversible assemblies, amyloids have begun to expose their more dynamic and reversible attributes depending upon the intrinsic properties of the precursor protein/peptide and experimental conditions such as temperature, pressure, structural modifications in proteins, or presence of chemicals in the reaction mixture. It has been repeatedly proposed that amyloids undergo transformation to the bioactive peptide/protein forms under specific conditions. In the present study, amyloids assembled from the model protein ovalbumin (OVA) were found to release the precursor protein in a slow and steady manner over an extended time period. Interestingly, the released OVA from amyloid depot was found to exhibit biophysical characteristics of native protein and reacted with native-OVA specific monoclonal as well as polyclonal antibodies. Moreover, antibodies generated upon immunization of OVA amyloidal aggregates or fibrils were found to recognize the native form of OVA. The study suggests that amyloids may act as depots for the native form of the protein and therefore can be exploited as vaccine candidates, where slow antigen release over extended time periods is a pre-requisite for the development of desired immune response.


Asunto(s)
Amiloide/inmunología , Anticuerpos Monoclonales/inmunología , Ovalbúmina/inmunología , Péptidos/inmunología , Linfocitos T/inmunología , Amiloide/química , Amiloide/metabolismo , Animales , Anticuerpos Monoclonales/sangre , Dicroismo Circular , Citocinas/metabolismo , Femenino , Inmunización , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Óxido Nítrico/metabolismo , Ovalbúmina/química , Ovalbúmina/metabolismo , Péptidos/química , Péptidos/metabolismo , Conformación Proteica , Multimerización de Proteína
16.
Indian J Med Res ; 138(5): 577-90, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24434313

RESUMEN

Multiple checkpoints regulating finely balanced death-versus-survival decisions characterize both thymic development and peripheral homeostasis of T lymphocytes. While exploring the mechanisms of T cell death involved at various stages during the life of a T cell, we have observed and reported a variety of non-redundant roles for apoptosis inducing factor (Aif), a mitochondrial flavoprotein. Aif is ubiquitously expressed in all cell lineages and functions as an NADH oxidase in its mitochondrial location. It is released following the mitochondrial death signals, whereupon it translocates to the nucleus, binds to DNA and causes large-scale DNA fragmentation. During T cell development, Aif is important for developing thymocytes to navigate the double negative (DN)3 to DN4 transition (beta-selection), via its oxidoreductase property which protects the rapidly proliferating cells from death due to reactive oxygen species (ROS). In peripheral mature T cells, Aif deficiency leads to an increased susceptibility of T cell blasts to activation induced cell death (AICD), possibly mediated by its antioxidant function, and decreased sensitivity to neglect-induced death (NID). Thus, Aif seems to have pro-apoptotic and anti-apoptotic roles in the same lineage in different contexts and at different stages. Surprisingly, in the closely related B lymphocyte lineage, Aif deficiency does not result in any abnormality. These findings generate the possibility of specific T cell dysfunction in human disease caused by Aif deficiency, as well as in mitochondriopathies due to other causes. Also, these data raise questions regarding the basis of lineage-specific consequences of the dysfunction/deficiency of apparently ubiquitous molecules.


Asunto(s)
Factor Inductor de la Apoptosis/metabolismo , Diferenciación Celular/genética , Linfocitos T/metabolismo , Timo/crecimiento & desarrollo , Antioxidantes/metabolismo , Apoptosis/genética , Factor Inductor de la Apoptosis/genética , Linaje de la Célula/inmunología , Humanos , Mitocondrias/genética , Mitocondrias/metabolismo , Complejos Multienzimáticos/metabolismo , NADH NADPH Oxidorreductasas/metabolismo , Linfocitos T/inmunología , Timo/metabolismo
17.
J Exp Med ; 209(9): 1641-53, 2012 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-22869892

RESUMEN

Apoptosis-inducing factor (Aif) is a mitochondrial flavoprotein that regulates cell metabolism and survival in many tissues. We report that aif-hypomorphic harlequin (Hq) mice show thymic hypocellularity and a cell-autonomous thymocyte developmental block associated with apoptosis at the ß-selection stage, independent of T cell receptor ß recombination. No abnormalities are observed in the B cell lineage. Transgenes encoding wild-type or DNA-binding-deficient mutant Aif rectify the thymic defect, but a transgene encoding oxidoreductase activity-deficient mutant Aif does not. The Hq thymic block is reversed in vivo by antioxidant treatment, and Hq T but not B lineage cells show enhanced oxidative stress. Thus, Aif, a ubiquitous protein, serves a lineage-specific nonredundant antiapoptotic role in the T cell lineage by regulating reactive oxygen species during thymic ß-selection.


Asunto(s)
Factor Inductor de la Apoptosis/fisiología , Linfocitos T/fisiología , Animales , Apoptosis , Muerte Celular , Linaje de la Célula , ADN/metabolismo , Femenino , Genes Codificadores de la Cadena beta de los Receptores de Linfocito T , Masculino , Ratones , Ratones Mutantes , Ratones Transgénicos , Especies Reactivas de Oxígeno/metabolismo , Recombinación Genética , Timocitos/metabolismo , Timocitos/patología , Timo/crecimiento & desarrollo , Timo/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA