Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Transl Med ; 15(690): eabk1900, 2023 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-37018415

RESUMEN

Patients who receive chimeric antigen receptor (CAR)-T cells that are enriched in memory T cells exhibit better disease control as a result of increased expansion and persistence of the CAR-T cells. Human memory T cells include stem-like CD8+ memory T cell progenitors that can become either functional stem-like T (TSTEM) cells or dysfunctional T progenitor exhausted (TPEX) cells. To that end, we demonstrated that TSTEM cells were less abundant in infused CAR-T cell products in a phase 1 clinical trial testing Lewis Y-CAR-T cells (NCT03851146), and the infused CAR-T cells displayed poor persistence in patients. To address this issue, we developed a production protocol to generate TSTEM-like CAR-T cells enriched for expression of genes in cell replication pathways. Compared with conventional CAR-T cells, TSTEM-like CAR-T cells had enhanced proliferative capacity and increased cytokine secretion after CAR stimulation, including after chronic CAR stimulation in vitro. These responses were dependent on the presence of CD4+ T cells during TSTEM-like CAR-T cell production. Adoptive transfer of TSTEM-like CAR-T cells induced better control of established tumors and resistance to tumor rechallenge in preclinical models. These more favorable outcomes were associated with increased persistence of TSTEM-like CAR-T cells and an increased memory T cell pool. Last, TSTEM-like CAR-T cells and anti-programmed cell death protein 1 (PD-1) treatment eradicated established tumors, and this was associated with increased tumor-infiltrating CD8+CAR+ T cells producing interferon-γ. In conclusion, our CAR-T cell protocol generated TSTEM-like CAR-T cells with enhanced therapeutic efficacy, resulting in increased proliferative capacity and persistence in vivo.


Asunto(s)
Inmunoterapia Adoptiva , Neoplasias , Humanos , Inmunoterapia Adoptiva/métodos , Linfocitos T , Citocinas/metabolismo , Células Madre/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo
2.
Front Immunol ; 14: 1140541, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36949946

RESUMEN

Chemotherapy has long been a standard treatment for a wide range of malignancies, where patients typically undergo multiple rounds of chemotherapy regimens to control tumor growth. In the clinic, the chemotherapy drugs cyclophosphamide and fludarabine are commonly used prior to Chimeric Antigen Receptor T (CAR-T) cell therapy to lymphodeplete and improve CAR-T cell engraftment. In this review, we discuss the use of chemotherapy in combination with CAR-T cell therapy. We also show that chemotherapy can deplete immunosuppressive cells, promote a pro-inflammatory tumor microenvironment, disrupt tumor stroma, and improve CAR-T cell recruitment to the tumor. Although the combination of chemotherapy plus CAR-T cell therapy is promising, certain aspects of chemotherapy also pose a challenge. In addition, the combined therapeutic effect may be heavily dependent on the dose and the treatment schedule. Thus, we also discussed the obstacles to effective clinical outcomes of the combination therapy.


Asunto(s)
Neoplasias , Receptores Quiméricos de Antígenos , Humanos , Neoplasias/terapia , Inmunoterapia Adoptiva , Linfocitos T , Tratamiento Basado en Trasplante de Células y Tejidos , Microambiente Tumoral
3.
Haematologica ; 108(1): 83-97, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-35770527

RESUMEN

Patients with refractory relapsed multiple myeloma respond to combination treatment with elotuzumab and lenalidomide. The mechanisms underlying this observation are not fully understood. Furthermore, biomarkers predictive of response have not been identified to date. To address these issues, we used a humanized myeloma mouse model and adoptive transfer of human natural killer (NK) cells to show that elotuzumab and lenalidomide treatment controlled myeloma growth, and this was mediated through CD16 on NK cells. In co-culture studies, we showed that peripheral blood mononuclear cells from a subset of patients with refractory relapsed multiple myeloma were effective killers of OPM2 myeloma cells when treated with elotuzumab and lenalidomide, and this was associated with significantly increased expression of CD54 on OPM2 cells. Furthermore, elotuzumab- and lenalidomide-induced OPM2 cell killing and increased OPM2 CD54 expression were dependent on both monocytes and NK cells, and these effects were not mediated by soluble factors alone. At the transcript level, elotuzumab and lenalidomide treatment significantly increased OPM2 myeloma cell expression of genes for trafficking and adhesion molecules, NK cell activation ligands and antigen presentation molecules. In conclusion, our findings suggest that multiple myeloma patients require elotuzumab- and lenalidomide-mediated upregulation of CD54 on autologous myeloma cells, in combination with NK cells and monocytes to mediate an effective anti-tumor response. Furthermore, our data suggest that increased myeloma cell CD54 expression levels could be a powerful predictive biomarker for response to elotuzumab and lenalidomide treatment.


Asunto(s)
Mieloma Múltiple , Animales , Ratones , Humanos , Lenalidomida/farmacología , Lenalidomida/uso terapéutico , Lenalidomida/metabolismo , Mieloma Múltiple/metabolismo , Monocitos/metabolismo , Leucocitos Mononucleares/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Células Asesinas Naturales , Dexametasona/uso terapéutico
5.
Front Immunol ; 12: 632399, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33746969

RESUMEN

Immunomodulatory drugs (IMiDs) are effective treatments for patients with multiple myeloma. IMiDs have pleotropic effects including targeting the myeloma cells directly, and improving the anti-myeloma immune response. In the absence of myeloma cells, lenalidomide and pomalidomide induce CD4+ T cell secretion of IL-2 and indirect activation of Natural Killer (NK) cells. In the context of T cell receptor ligation, IMiDs enhance T cell proliferation, cytokine release and Th1 responses, both in vivo and in vitro. Furthermore, combination treatment of IMiDs and myeloma-targeting monoclonal antibodies eg. daratumumab (anti-CD38) and elotuzumab (anti-SLAMF7), checkpoint inhibitors, or bispecific T cell engagers showed synergistic effects, mainly via enhanced T and NK cell dependent cellular toxicity and T cell proliferation. Conversely, the corticosteroid dexamethasone can impair the immune modulatory effects of IMiDs, indicating that careful choice of myeloma drugs in combination with IMiDs is key for the best anti-myeloma therapeutic efficacy. This review presents an overview of the role for T cells in the overall anti-myeloma effects of immunomodulatory drugs.


Asunto(s)
Factores Inmunológicos/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Linfocitos T/inmunología , Resistencia a Antineoplásicos , Quimioterapia Combinada , Humanos , Factores Inmunológicos/farmacología , Inmunoterapia , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Mieloma Múltiple/inmunología , Células Plasmáticas/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos
6.
Methods Mol Biol ; 2265: 529-541, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33704738

RESUMEN

We describe here a protocol to measure gene expression, T cell receptor (TCR) sequence, and protein expression by single T cells extracted from melanoma, using 10× Chromium technology. This method includes freezing and thawing of the melanoma infiltrating lymphocytes, staining of cells with fluorescent and barcode-conjugated antibodies, sorting of T cells, and loading the cells on the 10× Chromium Controller. After sequencing, analysis includes quality control, genetic demultiplexing to resolve genetically different samples, and T cell clonality and clustering analysis. Single cell RNA sequencing paints the complete portrait of individual T cells, including their clonality and phenotype, and it reconstructs a complete picture of the T cell infiltrate in a tumor that is represented as cell clustering similar to a pointillism painting.


Asunto(s)
Linfocitos Infiltrantes de Tumor/inmunología , Melanoma , RNA-Seq , Receptores de Antígenos de Linfocitos T , Análisis de la Célula Individual , Humanos , Melanoma/genética , Melanoma/inmunología , Receptores de Antígenos de Linfocitos T/inmunología
7.
Methods Mol Biol ; 2265: 543-555, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33704739

RESUMEN

Here we describe the application of mass cytometry to analyze tumor-infiltrating lymphocytes in human melanoma. Mass cytometry is the coupling of flow cytometry and mass spectrometry, which allows for the simultaneous measurement of 40+ cell parameters on a per cell basis. Heavy metal-labeled antibodies can bind to proteins (CD markers, transcription factors, cytokines) on the cell surface and in the cytoplasm/nucleus. As labeled cells pass through the CyTOF, the instrument detects the heavy metals. Combining these signals allows description of melanoma tumor-infiltrating lymphocytes at a greater depth than alternative phenotyping strategies and enables detailed analyses of a variety of cellular parameters, including immune cell lineage, activation status, and functional polarization.


Asunto(s)
Citometría de Flujo , Linfocitos Infiltrantes de Tumor , Espectrometría de Masas , Melanoma , Análisis de la Célula Individual , Humanos , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Melanoma/metabolismo , Melanoma/patología
8.
Cancers (Basel) ; 13(3)2021 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-33499101

RESUMEN

Chimeric antigen receptors (CAR) are genetically engineered receptors that can recognise specific antigens and subsequently activate downstream signalling. Human T cells engineered to express a CAR, also known as CAR-T cells, can target a specific tumour antigen on the cell surface to mediate a cytotoxic response against the tumour. CAR-T cell therapy has achieved remarkable success in treating hematologic malignancies, but not in solid tumours. Currently, extensive research is being carried out to make CAR-T cells a therapy for solid tumours. To date, most of the research interest in the field has focused on cytotoxic T lymphocytes as the carrier of CAR products. However, in addition to T cells, the CAR design can be introduced in other immune cells, such as natural killer (NK)/NKT cells, γδ T cells, mucosal-associated invariant T (MAIT) cells, dendritic cells (DC), macrophages, regulatory T cells (Treg), B cells, etc. Some of the CAR-engineered immune cells, such as CAR- γδ T and CAR-NK/NK-T cells, are directly involved in the anti-tumour response, demonstrated in preclinical studies and/or clinical trials. CAR-Tregs showed promising therapeutic potential in treating autoimmune diseases. In particular, B cells engineered with chimeric receptors can be used as a platform for long-term delivery of therapeutic proteins, such as recombinant antibodies or protein replacement, in an antigen-specific manner. CAR technology is one of the most powerful engineering platforms in immunotherapy, especially for the treatment of cancers. In this review, we will discuss the recent application of the CAR design in non-CAR-T cells and future opportunities in immunotherapy.

9.
Haematologica ; 106(12): 3115-3124, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33147937

RESUMEN

This study explored the novel immune checkpoint poliovirus receptor-related immunoglobulin domain-containing (PVRIG) in acute myeloid leukemia (AML). We showed that AML patient blasts consistently expressed the PVRIG ligand (poliovirus receptor-related 2, PVRL2). Furthermore, PVRIG blockade significantly enhanced NK cell killing of PVRL2+, poliovirus receptor (PVR)lo AML cell lines, and significantly increased NK cell activation and degranulation in the context of patient primary AML blasts. However, in AML patient bone marrow, NK cell PVRIG expression levels were not increased. To understand how PVRIG blockade might potentially be exploited therapeutically, we investigated the biology of PVRIG and revealed that NK cell activation resulted in reduced PVRIG expression on the cell surface. This occurred whether NK cells were activated by tumour cell recognition, cytokines (IL-2 and IL-12) or activating receptor stimulation (CD16 and NKp46). PVRIG was present at higher levels in the cytoplasm than on the cell surface, particularly on CD56bright NK cells, which further increased cytoplasmic PVRIG levels following IL-2 and IL-12 activation. PVRIG was continually transported to the cell surface via the endoplasmic reticulum (ER) and Golgi in both unstimulated and activated NK cells. Taken together, our findings suggest that anti- PVRIG blocking antibody functions by binding to surface-bound PVRIG, which undergoes rapid turnover in both unstimulated and activated NK cells. We conclude that the PVRIGPVRL2 immune checkpoint axis can feasibly be targeted with PVRIG blocking antibody for NK-mediated immunotherapy of PVRL2+ AML.


Asunto(s)
Proteínas de Punto de Control Inmunitario , Células Asesinas Naturales , Leucemia Mieloide Aguda , Receptores de Superficie Celular , Humanos , Inmunoterapia , Activación de Linfocitos , Receptores de Células Asesinas Naturales
10.
Front Immunol ; 12: 813832, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35095911

RESUMEN

Radiotherapy (RT) is the standard-of-care treatment for more than half of cancer patients with localized tumors and is also used as palliative care to facilitate symptom relief in metastatic cancers. In addition, RT can alter the immunosuppressive tumor microenvironment (TME) of solid tumors to augment the anti-tumor immune response of immune checkpoint blockade (ICB). The rationale of this combination therapy can also be extended to other forms of immunotherapy, such as chimeric antigen receptor T cell (CAR-T) therapy. Similar to ICB, the efficacy of CAR-T therapy is also significantly impacted by the immunosuppressive TME, leading to compromised T cell function and/or insufficient T cell infiltration. In this review, we will discuss some of the key barriers to the activity of CAR-T cells in the immunosuppressive TME and focus on how RT can be used to eliminate or bypass these barriers. We will present the challenges to achieving success with this therapeutic partnership. Looking forward, we will also provide strategies currently being investigated to ensure the success of this combination strategy in the clinic.


Asunto(s)
Inmunoterapia Adoptiva , Neoplasias/terapia , Radioterapia , Terapia Combinada , Manejo de la Enfermedad , Humanos , Terapia de Inmunosupresión/métodos , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Neoplasias/diagnóstico , Neoplasias/etiología , Neoplasias/mortalidad , Pronóstico , Radioterapia/efectos adversos , Radioterapia/métodos , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Resultado del Tratamiento
11.
Sci Immunol ; 4(41)2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31732518

RESUMEN

Mucosal-associated invariant T (MAIT) cells are activated in a TCR-dependent manner by antigens derived from the riboflavin synthesis pathway, including 5-(2-oxopropylideneamino)-6-d-ribitylaminouracil (5-OP-RU), bound to MHC-related protein-1 (MR1). However, MAIT cell activation in vivo has not been studied in detail. Here, we have found and characterized additional molecular signals required for optimal activation and expansion of MAIT cells after pulmonary Legionella or Salmonella infection in mice. We show that either bone marrow-derived APCs or non-bone marrow-derived cells can activate MAIT cells in vivo, depending on the pathogen. Optimal MAIT cell activation in vivo requires signaling through the inducible T cell costimulator (ICOS), which is highly expressed on MAIT cells. Subsequent expansion and maintenance of MAIT-17/1-type responses are dependent on IL-23. Vaccination with IL-23 plus 5-OP-RU augments MAIT cell-mediated control of pulmonary Legionella infection. These findings reveal cellular and molecular targets for manipulating MAIT cell function under physiological conditions.


Asunto(s)
Antígenos Bacterianos/inmunología , Interleucina-23/inmunología , Legionella/inmunología , Enfermedad de los Legionarios/inmunología , Células T Invariantes Asociadas a Mucosa/inmunología , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vacunación
12.
Curr Protoc Immunol ; 127(1): e89, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31763782

RESUMEN

This unit describes the utility of various mouse models of infection and immunization for studying mucosal-associated invariant T (MAIT) cell immunity: MAIT cells can be isolated from the lungs (or from other tissues/organs) and then identified and characterized by flow cytometry using MR1 tetramers in combination with a range of antibodies. The response kinetics, cytokine profiles, and functional differentiation of lung MAIT cells are studied following infection with the bacterial pathogen Legionella longbeachae or Salmonella enterica Typhimurium or immunization with synthetic MAIT cell antigen plus Toll-like receptor agonist. MAIT cells enriched or expanded during the process can be used for further studies. A step-by-step protocol is provided for MAIT cell sorting and adoptive transfer. Mice can then be challenged and MAIT cells tracked and further examined. © 2019 by John Wiley & Sons, Inc.


Asunto(s)
Citometría de Flujo , Antígenos de Histocompatibilidad Menor/inmunología , Células T Invariantes Asociadas a Mucosa/citología , Células T Invariantes Asociadas a Mucosa/inmunología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL
13.
Nat Commun ; 9(1): 4706, 2018 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-30413689

RESUMEN

Mucosal associated invariant T (MAIT) cells are evolutionarily-conserved, innate-like lymphocytes which are abundant in human lungs and can contribute to protection against pulmonary bacterial infection. MAIT cells are also activated during human viral infections, yet it remains unknown whether MAIT cells play a significant protective or even detrimental role during viral infections in vivo. Using murine experimental challenge with two strains of influenza A virus, we show that MAIT cells accumulate and are activated early in infection, with upregulation of CD25, CD69 and Granzyme B, peaking at 5 days post-infection. Activation is modulated via cytokines independently of MR1. MAIT cell-deficient MR1-/- mice show enhanced weight loss and mortality to severe (H1N1) influenza. This is ameliorated by prior adoptive transfer of pulmonary MAIT cells in both immunocompetent and immunodeficient RAG2-/-γC-/- mice. Thus, MAIT cells contribute to protection during respiratory viral infections, and constitute a potential target for therapeutic manipulation.


Asunto(s)
Gripe Humana/patología , Gripe Humana/virología , Células T Invariantes Asociadas a Mucosa/virología , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Traslado Adoptivo , Animales , Citocinas/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Pulmón/patología , Ratones Endogámicos C57BL , Antígenos de Histocompatibilidad Menor/metabolismo
14.
Mol Immunol ; 103: 46-54, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30196233

RESUMEN

Mucosal-associated Invariant T (MAIT) cells represent a large proportion of T cells in human blood, and are also present throughout the body, being concentrated at mucosal sites. Their high level of conservation throughout mammalian evolution and recognition of conserved microbial antigens, derived from precursors of riboflavin (vitamin B2) biosynthesis, suggest an important role in protective immunity to pathogens. However, the picture that is emerging of MAIT cell immune function is increasingly complex, with numerous correlations of MAIT cell numbers with human diseases, and with recent studies demonstrating their pathogenic potential. The conditions that drive MAIT cell responses towards a protective versus pathogenic role are only beginning to be deciphered and, yet, must be understood for any attempt to harness MAIT cells therapeutically. In this review we summarise our current knowledge of immune protection and pathology driven by MAIT cells, models used to study their role in immunity and steps towards elucidating the immune signals driving these responses.


Asunto(s)
Presentación de Antígeno/inmunología , Inmunidad/inmunología , Células T Invariantes Asociadas a Mucosa/inmunología , Subgrupos de Linfocitos T/inmunología , Inmunidad Adaptativa/inmunología , Animales , Antígenos Bacterianos/inmunología , Helicobacter pylori/inmunología , Helicobacter pylori/fisiología , Humanos , Inmunidad Mucosa/inmunología , Subgrupos de Linfocitos T/microbiología
15.
Nat Commun ; 9(1): 3350, 2018 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-30135490

RESUMEN

Mucosal associated invariant T (MAIT) cells recognise conserved microbial metabolites from riboflavin synthesis. Striking evolutionary conservation and pulmonary abundance implicate them in antibacterial host defence, yet their functions in protection against clinically important pathogens are unknown. Here we show that mouse Legionella longbeachae infection induces MR1-dependent MAIT cell activation and rapid pulmonary accumulation of MAIT cells associated with immune protection detectable in immunocompetent host animals. MAIT cell protection is more evident in mice lacking CD4+ cells, and adoptive transfer of MAIT cells rescues immunodeficient Rag2-/-γC-/- mice from lethal Legionella infection. Protection is dependent on MR1, IFN-γ and GM-CSF, but not IL-17A, TNF or perforin, and enhanced protection is detected earlier after infection of mice antigen-primed to boost MAIT cell numbers before infection. Our findings define a function for MAIT cells in protection against a major human pathogen and indicate a potential role for vaccination to enhance MAIT cell immunity.


Asunto(s)
Legionella longbeachae/patogenicidad , Pulmón/microbiología , Células T Invariantes Asociadas a Mucosa/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Interleucina-17/metabolismo , Legionella longbeachae/inmunología , Legionelosis/inmunología , Legionelosis/microbiología , Pulmón/metabolismo , Masculino , Ratones , Células T Invariantes Asociadas a Mucosa/metabolismo , Perforina/metabolismo
16.
J Immunol ; 200(5): 1901-1916, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29378910

RESUMEN

Mucosal-associated invariant T (MAIT) cells produce inflammatory cytokines and cytotoxic granzymes in response to by-products of microbial riboflavin synthesis. Although MAIT cells are protective against some pathogens, we reasoned that they might contribute to pathology in chronic bacterial infection. We observed MAIT cells in proximity to Helicobacter pylori bacteria in human gastric tissue, and so, using MR1-tetramers, we examined whether MAIT cells contribute to chronic gastritis in a mouse H. pylori SS1 infection model. Following infection, MAIT cells accumulated to high numbers in the gastric mucosa of wild-type C57BL/6 mice, and this was even more pronounced in MAIT TCR transgenic mice or in C57BL/6 mice where MAIT cells were preprimed by Ag exposure or prior infection. Gastric MAIT cells possessed an effector memory Tc1/Tc17 phenotype, and were associated with accelerated gastritis characterized by augmented recruitment of neutrophils, macrophages, dendritic cells, eosinophils, and non-MAIT T cells and by marked gastric atrophy. Similarly treated MR1-/- mice, which lack MAIT cells, showed significantly less gastric pathology. Thus, we demonstrate the pathogenic potential of MAIT cells in Helicobacter-associated immunopathology, with implications for other chronic bacterial infections.


Asunto(s)
Gastritis/inmunología , Infecciones por Helicobacter/inmunología , Helicobacter pylori/inmunología , Células T Invariantes Asociadas a Mucosa/inmunología , Adulto , Animales , Línea Celular Tumoral , Femenino , Mucosa Gástrica/inmunología , Humanos , Memoria Inmunológica/inmunología , Células Jurkat , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Linfocitos T Citotóxicos/inmunología
17.
Nat Immunol ; 18(4): 402-411, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28166217

RESUMEN

The major-histocompatibility-complex-(MHC)-class-I-related molecule MR1 can present activating and non-activating vitamin-B-based ligands to mucosal-associated invariant T cells (MAIT cells). Whether MR1 binds other ligands is unknown. Here we identified a range of small organic molecules, drugs, drug metabolites and drug-like molecules, including salicylates and diclofenac, as MR1-binding ligands. Some of these ligands inhibited MAIT cells ex vivo and in vivo, while others, including diclofenac metabolites, were agonists. Crystal structures of a T cell antigen receptor (TCR) from a MAIT cell in complex with MR1 bound to the non-stimulatory and stimulatory compounds showed distinct ligand orientations and contacts within MR1, which highlighted the versatility of the MR1 binding pocket. The findings demonstrated that MR1 was able to capture chemically diverse structures, spanning mono- and bicyclic compounds, that either inhibited or activated MAIT cells. This indicated that drugs and drug-like molecules can modulate MAIT cell function in mammals.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Antígenos de Histocompatibilidad Menor/metabolismo , Células T Invariantes Asociadas a Mucosa/efectos de los fármacos , Células T Invariantes Asociadas a Mucosa/metabolismo , Sitios de Unión , Línea Celular , Cristalografía por Rayos X , Descubrimiento de Drogas , Antígenos de Histocompatibilidad Clase I/química , Humanos , Enlace de Hidrógeno , Ligandos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Antígenos de Histocompatibilidad Menor/química , Modelos Moleculares , Conformación Molecular , Estructura Molecular , Células T Invariantes Asociadas a Mucosa/inmunología , Unión Proteica , Receptores de Antígenos de Linfocitos T/química , Receptores de Antígenos de Linfocitos T/metabolismo , Relación Estructura-Actividad
18.
Mol Biol Rep ; 40(8): 4747-57, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23653003

RESUMEN

Bisphenol A (BPA) is an estrogenic compound commonly used in manufacture of various consumer products. Earlier studies from our group have demonstrated that neonatal exposure of male rats to BPA causes decrease in sperm count and motility, increase in post implantation loss, ultimately leading to subfertility during adulthood. One of the factors contributing for post implantation loss is altered methylation pattern of imprinted genes. The present study was undertaken to investigate the molecular effects of neonatal exposure of male rats to BPA (2.4 µg/pup) (F0) on the methylation of H19 imprinting control region (ICR) in resorbed embryo (F1) and compared with spermatozoa of their respective sires (F0). We observed a significant down regulation in the transcript expression of Igf2 and H19 genes in BPA resorbed embryo (F1) as compared to control viable embryo. A significant hypomethylation was observed at the H19 ICR in the spermatozoa as well as in resorbed embryo sired by rats exposed neonatally to BPA. These results indicated that the aberrant methylation at ICR in spermatozoa was inherited by embryo which causes perturbation in the expression of Igf2 and H19, ultimately leading to post implantation loss. This could be one of the possible mechanisms of BPA induced adverse epigenetic effects on male fertility.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Metilación de ADN/efectos de los fármacos , Pérdida del Embrión/inducido químicamente , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Factor II del Crecimiento Similar a la Insulina/metabolismo , Fenoles/toxicidad , ARN Largo no Codificante/metabolismo , Espermatozoides/efectos de los fármacos , Animales , Animales Recién Nacidos , Secuencia de Bases , Compuestos de Bencidrilo/administración & dosificación , Clonación Molecular , Cartilla de ADN/genética , ADN Complementario/biosíntesis , Inyecciones Subcutáneas , Masculino , Datos de Secuencia Molecular , Fenoles/administración & dosificación , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ADN
19.
J Biochem Mol Toxicol ; 26(9): 337-43, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22730197

RESUMEN

Bisphenol A (BPA) is used in the production of polycarbonate plastics and epoxy resins. Our previous studies have demonstrated that neonatal exposure of male rats to BPA causes decrease in sperm count and motility, increase in postimplantation loss (POL), ultimately leading to subfertility during adulthood. Epigenetic mechanisms such as DNA methylation play an important role in embryo development. DNA methyltransferases (Dnmts) are the key players involved in regulating DNA methylation marks. The objective of the present study was to determine the mechanism involved in resorption of embryo as a result of BPA exposure. The results of the present study demonstrate that neonatal exposure of male rats to BPA down regulates the gene expression of Dnmts and related transcription factors in resorbed embryos as compared with the viable embryo. Thereby, suggesting that BPA may have altered the sperm epigenome, which might have affected the embryo development and leading to an increase in the POL.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Metilación de ADN/efectos de los fármacos , Pérdida del Embrión/inducido químicamente , Desarrollo Embrionario/efectos de los fármacos , Disruptores Endocrinos/toxicidad , Fenoles/toxicidad , Animales , Animales Recién Nacidos , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Epigénesis Genética , Femenino , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Infertilidad Masculina/inducido químicamente , Masculino , Oligospermia/inducido químicamente , Embarazo , Ratas , Ratas Sprague-Dawley , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción Sp3/genética , Factor de Transcripción Sp3/metabolismo , Motilidad Espermática/efectos de los fármacos , Transcripción Genética , ADN Metiltransferasa 3B
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...