Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38617240

RESUMEN

Regulatory T cells (Tregs) are promising cellular therapies to induce immune tolerance in organ transplantation and autoimmune disease. The success of chimeric antigen receptor (CAR) T-cell therapy for cancer has sparked interest in using CARs to generate antigen-specific Tregs. Here, we compared CAR with endogenous T cell receptor (TCR)/CD28 activation in human Tregs. Strikingly, CAR Tregs displayed increased cytotoxicity and diminished suppression of antigen-presenting cells and effector T (Teff) cells compared with TCR/CD28 activated Tregs. RNA sequencing revealed that CAR Tregs activate Teff cell gene programs. Indeed, CAR Tregs secreted high levels of inflammatory cytokines, with a subset of FOXP3+ CAR Tregs uniquely acquiring CD40L surface expression and producing IFNγ. Interestingly, decreasing CAR antigen affinity reduced Teff cell gene expression and inflammatory cytokine production by CAR Tregs. Our findings showcase the impact of engineered receptor activation on Treg biology and support tailoring CAR constructs to Tregs for maximal therapeutic efficacy.

2.
J Reprod Immunol ; 163: 104244, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38555747

RESUMEN

Implantation and maintenance of pregnancy involve intricate immunological processes that enable the developing fetus to coexist with the maternal immune system. Progesterone, a critical hormone during pregnancy, is known to promote immune tolerance and prevent preterm labor. However, the mechanism by which progesterone mediates these effects remains unclear. In this study, we investigated the role of the non-classical progesterone receptor membrane component 1 (PGRMC1) in progesterone signaling at the maternal-fetal interface. Using JEG3 cells, a trophoblast model cell line, we observed that progesterone stimulation increased the expression of human leukocyte antigen-C (HLA-C) and HLA-G, key molecules involved in immune tolerance. We also found that progesterone upregulated the expression of the transcription factor ELF3, which is known to regulate trophoblast-specific HLA-C expression. Interestingly, JEG3 cells lacked expression of classical progesterone receptors (PRs) but exhibited high expression of PGRMC1, a finding we confirmed in primary trophoblasts by mining sc-RNA seq data from human placenta. To investigate the role of PGRMC1 in progesterone signaling, we used CRISPR/Cas9 technology to knockout PGRMC1 in JEG3 cells. PGRMC1-deficient cells showed a diminished response to progesterone stimulation. Furthermore, we found that the progesterone antagonist RU486 inhibited ELF3 expression in a PGRMC1-dependent manner, suggesting that RU486 acts as a progesterone antagonist by competing for receptor binding. Additionally, we found that RU486 inhibited cell invasion, an important process for successful pregnancy, and this inhibitory effect was dependent on PGRMC1. Our findings highlight the crucial role of PGRMC1 in mediating the immunoregulatory effects of progesterone at the maternal-fetal interface.


Asunto(s)
Proteínas de la Membrana , Progesterona , Receptores de Progesterona , Trofoblastos , Humanos , Receptores de Progesterona/metabolismo , Femenino , Embarazo , Progesterona/metabolismo , Progesterona/farmacología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Trofoblastos/metabolismo , Trofoblastos/inmunología , Placenta/inmunología , Placenta/metabolismo , Transducción de Señal/inmunología , Intercambio Materno-Fetal/inmunología , Implantación del Embrión/inmunología
3.
Methods Mol Biol ; 2748: 243-265, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38070118

RESUMEN

Chimeric antigen receptor (CAR) T cell therapy has proven to be a successful treatment option for leukemias and lymphomas. These encouraging outcomes underscore the potential of adoptive cell therapy for other oncology applications, namely, solid tumors. However, CAR T cells are yet to succeed in treating solid tumors. Unlike liquid tumors, solid tumors create a hostile tumor microenvironment (TME). CAR T cells must traffic to the TME, survive, and retain their function to eradicate the tumor. Nevertheless, there is no universal preclinical model to systematically test candidate CARs and CAR targets for their capacity to infiltrate and eliminate human solid tumors in vivo. Here, we provide a detailed protocol to evaluate human CAR CD4+ helper T cells and CD8+ cytotoxic T cells in immunodeficient (NSG) mice bearing antigen-expressing human solid tumors.


Asunto(s)
Neoplasias , Receptores Quiméricos de Antígenos , Humanos , Animales , Ratones , Inmunoterapia Adoptiva/métodos , Receptores Quiméricos de Antígenos/genética , Neoplasias/patología , Linfocitos T , Microambiente Tumoral
4.
Methods Mol Biol ; 2748: 201-241, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38070117

RESUMEN

The adaptive immune system exhibits exquisite specificity and memory and is involved in virtually every process in the human body. Redirecting adaptive immune cells, in particular T cells, to desired targets has the potential to lead to the creation of powerful cell-based therapies for a wide range of maladies. While conventional effector T cells (Teff) would be targeted towards cells to be eliminated, such as cancer cells, immunosuppressive regulatory T cells (Treg) would be directed towards tissues to be protected, such as transplanted organs. Chimeric antigen receptors (CARs) are designer molecules comprising an extracellular recognition domain and an intracellular signaling domain that drives full T cell activation directly downstream of target binding. Here, we describe procedures to generate and evaluate human CAR CD4+ helper T cells, CD8+ cytotoxic T cells, and CD4+FOXP3+ regulatory T cells.


Asunto(s)
Receptores Quiméricos de Antígenos , Humanos , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T Reguladores , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo
5.
Mol Ther ; 31(9): 2591-2599, 2023 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-37481703

RESUMEN

Lymphodepleting pre-conditioning is a nearly universal component of T cell adoptive transfer protocols. The side effects of pre-conditioning regimens used in adoptive cell therapy are clinically significant and include pan-cytopenia, immune suppression, and reactive myelopoiesis. We conducted studies to test the hypothesis that the mechanisms underlying effective engraftment are cell autonomous and not dependent on a lymphodepleted host immune status. These studies leveraged mouse models to examine the role of Stat5 signaling during T cell adoptive transfer. We observed that, by transiently expressing a constitutively active mutamer of Stat5b during the process of adoptive transfer, we could completely obviate the need for lymphodepletion prior to adoptive transfer. Using several functional assays, we benchmark the function of the engrafted T cells against T cells transferred after conventional lymphodepletion. These studies identify a cell-autonomous mechanism driven by transient Stat5b signaling with lasting effects on T cell phenotype and function. Furthermore, the results presented suggest that adoptive T cell therapy could be improved by removing lymphodepletion protocols entirely and replacing them with RNA transfection of T cells with transcripts encoding active Stat5.


Asunto(s)
Transducción de Señal , Linfocitos T , Ratones , Animales , Traslado Adoptivo , Inmunoterapia Adoptiva/métodos
6.
Clin Immunol ; 246: 109201, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36470337

RESUMEN

Novel biologics are currently being tested in clinical trials for the treatment of autoimmune diseases and the prevention of transplant allograft rejection. Their premise is to deliver highly efficient immunosuppression while minimizing side-effects, as they specifically target inflammatory mediators involved in the dysregulation of the immune system. However, the pleiotropism of soluble mediators and cell-to-cell interactions with potential to exert both proinflammatory and regulatory influences on the outcome of the immune response can lead to unpredictable results. Predicting responses to biologic drugs requires mechanistic understanding of the cell type-specific effect of immune mediators. Elucidation of the central role of regulatory T cells (Treg), a small subset of T cells dedicated to immune homeostasis, in preventing the development of auto- and allo-immunity has provided a deeper understanding of the signaling pathways that govern immune tolerance. This review focuses on the requisite signals that promote Treg homeostasis and discusses the anticipated outcomes of biologics targeting these signals. Our goal is to inform and facilitate the design of cell-specific biologics that thwart T effector cells (Teff) while promoting Treg function for the treatment of autoimmune diseases and the prevention of transplant rejection.


Asunto(s)
Enfermedades Autoinmunes , Productos Biológicos , Humanos , Linfocitos T Reguladores , Productos Biológicos/farmacología , Productos Biológicos/uso terapéutico , Enfermedades Autoinmunes/tratamiento farmacológico , Tolerancia Inmunológica , Homeostasis
7.
Front Immunol ; 13: 1042622, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36466853

RESUMEN

TNFa blocking agents were the first-in-class biologic drugs used for the treatment of autoimmune disease. Paradoxically, however, exacerbation of autoimmunity was observed in some patients. TNFa is a pleiotropic cytokine that has both proinflammatory and regulatory effects on CD4+ T cells and can influence the adaptive immune response against autoantigens. Here, we critically appraise the literature and discuss the intricacies of TNFa signaling that may explain the controversial findings of previous studies. The pleiotropism of TNFa is based in part on the existence of two biologically active forms of TNFa, soluble and membrane-bound, with different affinities for two distinct TNF receptors, TNFR1 and TNFR2, leading to activation of diverse downstream molecular pathways involved in cell fate decisions and immune function. Distinct membrane expression patterns of TNF receptors by CD4+ T cell subsets and their preferential binding of distinct forms of TNFα produced by a diverse pool of cellular sources during different stages of an immune response are important determinants of the differential outcomes of TNFa-TNF receptor signaling. Targeted manipulation of TNFa-TNF receptor signaling on select CD4+ T cell subsets may offer specific therapeutic interventions to dampen inflammation while fortifying immune regulation for the treatment of autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes , Factor de Necrosis Tumoral alfa , Humanos , Linfocitos T , Recuento de Linfocitos , Transducción de Señal , Linfocitos T CD4-Positivos
8.
Mol Metab ; 56: 101417, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34902607

RESUMEN

BACKGROUND: Type 1 diabetes (T1D) is an autoimmune disease characterized by impaired immune tolerance to ß-cell antigens and progressive destruction of insulin-producing ß-cells. Animal models have provided valuable insights for understanding the etiology and pathogenesis of this disease, but they fall short of reflecting the extensive heterogeneity of the disease in humans, which is contributed by various combinations of risk gene alleles and unique environmental factors. Collectively, these factors have been used to define subgroups of patients, termed endotypes, with distinct predominating disease characteristics. SCOPE OF REVIEW: Here, we review the gaps filled by these models in understanding the intricate involvement and regulation of the immune system in human T1D pathogenesis. We describe the various models developed so far and the scientific questions that have been addressed using them. Finally, we discuss the limitations of these models, primarily ascribed to hosting a human immune system (HIS) in a xenogeneic recipient, and what remains to be done to improve their physiological relevance. MAJOR CONCLUSIONS: To understand the role of genetic and environmental factors or evaluate immune-modifying therapies in humans, it is critical to develop and apply models in which human cells can be manipulated and their functions studied under conditions that recapitulate as closely as possible the physiological conditions of the human body. While microphysiological systems and living tissue slices provide some of these conditions, HIS mice enable more extensive analyses using in vivo systems.


Asunto(s)
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Animales , Diabetes Mellitus Tipo 1/genética , Humanos , Sistema Inmunológico/patología , Células Secretoras de Insulina/patología , Ratones
9.
Front Immunol ; 13: 1075813, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36591309

RESUMEN

Autoimmune disease, caused by unwanted immune responses to self-antigens, affects millions of people each year and poses a great social and economic burden to individuals and communities. In the course of autoimmune disorders, including rheumatoid arthritis, systemic lupus erythematosus, type 1 diabetes mellitus, and multiple sclerosis, disturbances in the balance between the immune response against harmful agents and tolerance towards self-antigens lead to an immune response against self-tissues. In recent years, various regulatory immune cells have been identified. Disruptions in the quality, quantity, and function of these cells have been implicated in autoimmune disease development. Therefore, targeting or engineering these cells is a promising therapeutic for different autoimmune diseases. Regulatory T cells, regulatory B cells, regulatory dendritic cells, myeloid suppressor cells, and some subsets of innate lymphoid cells are arising as important players among this class of cells. Here, we review the roles of each suppressive cell type in the immune system during homeostasis and in the development of autoimmunity. Moreover, we discuss the current and future therapeutic potential of each one of these cell types for autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes , Inmunidad Innata , Humanos , Linfocitos , Enfermedades Autoinmunes/terapia , Autoinmunidad , Autoantígenos
11.
Front Immunol ; 12: 686439, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34616392

RESUMEN

Infusion of regulatory T cells (Tregs) engineered with a chimeric antigen receptor (CAR) targeting donor-derived human leukocyte antigen (HLA) is a promising strategy to promote transplant tolerance. Here, we describe an anti-HLA-A2 CAR (A2-CAR) generated by grafting the complementarity-determining regions (CDRs) of a human monoclonal anti-HLA-A2 antibody into the framework regions of the Herceptin 4D5 single-chain variable fragment and fusing it with a CD28-ζ signaling domain. The CDR-grafted A2-CAR maintained the specificity of the original antibody. We then generated HLA-A2 mono-specific human CAR Tregs either by deleting the endogenous T-cell receptor (TCR) via CRISPR/Cas9 and introducing the A2-CAR using lentiviral transduction or by directly integrating the CAR construct into the TCR alpha constant locus using homology-directed repair. These A2-CAR+TCRdeficient human Tregs maintained both Treg phenotype and function in vitro. Moreover, they selectively accumulated in HLA-A2-expressing islets transplanted from either HLA-A2 transgenic mice or deceased human donors. A2-CAR+TCRdeficient Tregs did not impair the function of these HLA-A2+ islets, whereas similarly engineered A2-CAR+TCRdeficientCD4+ conventional T cells rejected the islets in less than 2 weeks. A2-CAR+TCRdeficient Tregs delayed graft-versus-host disease only in the presence of HLA-A2, expressed either by co-transferred peripheral blood mononuclear cells or by the recipient mice. Altogether, we demonstrate that genome-engineered mono-antigen-specific A2-CAR Tregs localize to HLA-A2-expressing grafts and exhibit antigen-dependent in vivo suppression, independent of TCR expression. These approaches may be applied towards developing precision Treg cell therapies for transplant tolerance.


Asunto(s)
Anticuerpos/metabolismo , Antígeno HLA-A2/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T Reguladores/trasplante , Tolerancia al Trasplante , Animales , Ingeniería Celular , Femenino , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/metabolismo , Humanos , Inmunoterapia Adoptiva , Masculino , Ratones , Ratones Endogámicos NOD , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Quiméricos de Antígenos/genética , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
12.
Front Immunol ; 12: 639818, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33833759

RESUMEN

Anti-CD19 chimeric antigen receptor (CD19-CAR)-engineered T cells are approved therapeutics for malignancies. The impact of the hinge domain (HD) and the transmembrane domain (TMD) between the extracellular antigen-targeting CARs and the intracellular signaling modalities of CARs has not been systemically studied. In this study, a series of 19-CARs differing only by their HD (CD8, CD28, or IgG4) and TMD (CD8 or CD28) was generated. CARs containing a CD28-TMD, but not a CD8-TMD, formed heterodimers with the endogenous CD28 in human T cells, as shown by co-immunoprecipitation and CAR-dependent proliferation of anti-CD28 stimulation. This dimerization was dependent on polar amino acids in the CD28-TMD and was more efficient with CARs containing CD28 or CD8 HD than IgG4-HD. The CD28-CAR heterodimers did not respond to CD80 and CD86 stimulation but had a significantly reduced CD28 cell-surface expression. These data unveiled a fundamental difference between CD28-TMD and CD8-TMD and indicated that CD28-TMD can modulate CAR T-cell activities by engaging endogenous partners.


Asunto(s)
Antígenos CD28/inmunología , Dominios Proteicos/inmunología , Receptores Quiméricos de Antígenos/inmunología , Antígenos CD19/inmunología , Dimerización , Humanos , Activación de Linfocitos/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología
13.
Front Immunol ; 12: 783282, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35003100

RESUMEN

Treg therapies are being tested in clinical trials in transplantation and autoimmune diseases, however, the impact of inflammation on Tregs remains controversial. We challenged human Tregs ex-vivo with pro-inflammatory cytokines IL-6 and TNFα and observed greatly enhanced proliferation stimulated by anti-CD3 and anti-CD28 (aCD3/28) beads or CD28 superagonist (CD28SA). The cytokine-exposed Tregs maintained high expression of FOXP3 and HELIOS, demethylated FOXP3 enhancer, and low IFNγ, IL-4, and IL-17 secretion. Blocking TNF receptor using etanercept or deletion of TNF receptor 2 using CRISPR/Cas9 blunted Treg proliferation and attenuated FOXP3 and HELIOS expression. These results prompted us to consider using CD28SA together with IL-6 and TNFα without aCD3/28 beads (beadless) as an alternative protocol for therapeutic Treg manufacturing. Metabolomics profiling revealed more active glycolysis and oxidative phosphorylation, increased energy production, and higher antioxidant potential during beadless Treg expansion. Finally, beadless expanded Tregs maintained suppressive functions in vitro and in vivo. These results demonstrate that human Tregs positively respond to proinflammatory cytokines with enhanced proliferation without compromising their lineage identity or function. This property can be harnessed for therapeutic Treg manufacturing.


Asunto(s)
Enfermedad Injerto contra Huésped/terapia , Inmunoterapia Adoptiva/métodos , Interleucina-6/metabolismo , Linfocitos T Reguladores/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Adulto , Anciano , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Etanercept/farmacología , Femenino , Factores de Transcripción Forkhead/análisis , Factores de Transcripción Forkhead/metabolismo , Enfermedad Injerto contra Huésped/inmunología , Voluntarios Sanos , Humanos , Factor de Transcripción Ikaros/análisis , Factor de Transcripción Ikaros/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Cultivo Primario de Células , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/trasplante , Trasplante Heterólogo/efectos adversos , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Adulto Joven
14.
Int Immunol ; 32(12): 771-783, 2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-32808986

RESUMEN

Diet is an environmental factor in autoimmune disorders, where the immune system erroneously destroys one's own tissues. Yet, interactions between diet and autoimmunity remain largely unexplored, particularly the impact of immunogenetics, one's human leukocyte antigen (HLA) allele make-up, in this interplay. Here, we interrogated animals and plants for the presence of epitopes implicated in human autoimmune diseases. We mapped autoimmune epitope distribution across organisms and determined their tissue expression pattern. Interestingly, diet-derived epitopes implicated in a disease were more likely to bind to HLA alleles associated with that disease than to protective alleles, with visible differences between organisms with similar autoimmune epitope content. We then analyzed an individual's HLA haplotype, generating a personalized heatmap of potential dietary autoimmune triggers. Our work uncovered differences in autoimmunogenic potential across food sources and revealed differential binding of diet-derived epitopes to autoimmune disease-associated HLA alleles, shedding light on the impact of diet on autoimmunity.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Autoinmunidad/inmunología , Dieta , Complejo Mayor de Histocompatibilidad/inmunología , Alelos , Epítopos/inmunología , Humanos , Complejo Mayor de Histocompatibilidad/genética
15.
NPJ Vaccines ; 5: 58, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32655899

RESUMEN

Convalescent plasma therapy holds promise as a transient treatment for COVID-19. Yet, blood products are important sources of HIV infection in low- and middle-income nations. Great care must be taken to prevent plasma therapy from fueling HIV epidemics in the developing world.

16.
Biochim Biophys Acta Mol Basis Dis ; 1866(10): 165887, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32599141

RESUMEN

Intermediary metabolism is traditionally viewed as the large, highly integrated network of reactions that provides cells with metabolic energy, reducing power and biosynthetic intermediates. The elucidation of its major pathways and molecular mechanisms of energy transduction occupied some of the brightest scientific minds for almost two centuries. When these goals were achieved, a sense that intermediary metabolism was mostly a solved problem pervaded the broader biochemical community, and the field lost its vitality. However, intermediary metabolism has recently been re-energized by several paradigm-shifting discoveries that challenged its perception as a self-contained system and re-positioned it at the crossroads of all aspects of cell function, from cell growth, proliferation and death to epigenetics and immunity. Emphasis is now increasingly placed on the involvement of metabolic dysfunction in human disease. In this review, we will navigate from the dawn of intermediary metabolism research to present day work on this ever-expanding field.


Asunto(s)
Metabolismo Energético/fisiología , Animales , Apoptosis/fisiología , Proliferación Celular , Epigénesis Genética , Humanos , Inmunidad/fisiología , Modelos Animales , Transducción de Señal
17.
Cell ; 179(6): 1276-1288.e14, 2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31778654

RESUMEN

Although human genetic studies have implicated many susceptible genes associated with plasma lipid levels, their physiological and molecular functions are not fully characterized. Here we demonstrate that orphan G protein-coupled receptor 146 (GPR146) promotes activity of hepatic sterol regulatory element binding protein 2 (SREBP2) through activation of the extracellular signal-regulated kinase (ERK) signaling pathway, thereby regulating hepatic very low-density lipoprotein (VLDL) secretion, and subsequently circulating low-density lipoprotein cholesterol (LDL-C) and triglycerides (TG) levels. Remarkably, GPR146 deficiency reduces plasma cholesterol levels substantially in both wild-type and LDL receptor (LDLR)-deficient mice. Finally, aortic atherosclerotic lesions are reduced by 90% and 70%, respectively, in male and female LDLR-deficient mice upon GPR146 depletion. Taken together, these findings outline a regulatory role for the GPR146/ERK axis in systemic cholesterol metabolism and suggest that GPR146 inhibition could be an effective strategy to reduce plasma cholesterol levels and atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , Hipercolesterolemia/metabolismo , Receptores Acoplados a Proteínas G/deficiencia , Animales , Aterosclerosis/sangre , Secuencia de Bases , Colesterol/sangre , Dependovirus/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ayuno , Femenino , Hepatocitos/metabolismo , Humanos , Hipercolesterolemia/sangre , Lipoproteínas VLDL/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , ARN Interferente Pequeño/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de LDL/metabolismo , Transducción de Señal , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Triglicéridos/sangre , Regulación hacia Arriba
18.
Int J Mol Sci ; 20(19)2019 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-31623305

RESUMEN

Worldwide, several million workers are employed in the various chromium (Cr) industries. These workers may suffer from a variety of adverse health effects produced by dusts, mists and fumes containing Cr in the hexavalent oxidation state, Cr(VI). Of major importance, occupational exposure to Cr(VI) compounds has been firmly associated with the development of lung cancer. Counterintuitively, Cr(VI) is mostly unreactive towards most biomolecules, including nucleic acids. However, its intracellular reduction produces several species that react extensively with biomolecules. The diversity and chemical versatility of these species add great complexity to the study of the molecular mechanisms underlying Cr(VI) toxicity and carcinogenicity. As a consequence, these mechanisms are still poorly understood, in spite of intensive research efforts. Here, we discuss the impact of Cr(VI) on the stress response-an intricate cellular system against proteotoxic stress which is increasingly viewed as playing a critical role in carcinogenesis. This discussion is preceded by information regarding applications, chemical properties and adverse health effects of Cr(VI). A summary of our current understanding of cancer initiation, promotion and progression is also provided, followed by a brief description of the stress response and its links to cancer and by an overview of potential molecular mechanisms of Cr(VI) carcinogenicity.


Asunto(s)
Carcinógenos Ambientales/farmacología , Cromo/farmacología , Estrés Fisiológico/efectos de los fármacos , Carcinógenos Ambientales/toxicidad , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Cromo/toxicidad , Daño del ADN , Humanos , Neoplasias/etiología , Neoplasias/metabolismo , Neoplasias/patología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética
19.
Nat Rev Drug Discov ; 18(10): 749-769, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31541224

RESUMEN

Regulatory T cells (Treg cells) are a small subset of immune cells that are dedicated to curbing excessive immune activation and maintaining immune homeostasis. Accordingly, deficiencies in Treg cell development or function result in uncontrolled immune responses and tissue destruction and can lead to inflammatory disorders such as graft-versus-host disease, transplant rejection and autoimmune diseases. As Treg cells deploy more than a dozen molecular mechanisms to suppress immune responses, they have potential as multifaceted adaptable smart therapeutics for treating inflammatory disorders. Indeed, early-phase clinical trials of Treg cell therapy have shown feasibility, tolerability and potential efficacy in these disease settings. In the meantime, progress in the development of chimeric antigen receptors and in genome editing (including the application of CRISPR-Cas9) over the past two decades has facilitated the genetic optimization of primary T cell therapy for cancer. These technologies are now being used to enhance the specificity and functionality of Treg cells. In this Review, we describe the key advances and prospects in designing and implementing Treg cell-based therapy in autoimmunity and transplantation.


Asunto(s)
Enfermedades Autoinmunes/terapia , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Rechazo de Injerto/terapia , Neoplasias/terapia , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/trasplante , Animales , Enfermedades Autoinmunes/inmunología , Rechazo de Injerto/inmunología , Humanos , Neoplasias/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...