Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36.222
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Immunol ; 38: 541-566, 2020 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-32017635

RESUMEN

Naturally occurring CD4+ regulatory T cells (Tregs), which specifically express the transcription factor FoxP3 in the nucleus and CD25 and CTLA-4 on the cell surface, are a functionally distinct T cell subpopulation actively engaged in the maintenance of immunological self-tolerance and homeostasis. Recent studies have facilitated our understanding of the cellular and molecular basis of their generation, function, phenotypic and functional stability, and adaptability. It is under investigation in humans how functional or numerical Treg anomalies, whether genetically determined or environmentally induced, contribute to immunological diseases such as autoimmune diseases. Also being addressed is how Tregs can be targeted to control physiological and pathological immune responses, for example, by depleting them to enhance tumor immunity or by expanding them to treat immunological diseases. This review discusses our current understanding of Treg immunobiology in normal and disease states, with a perspective on the realization of Treg-targeting therapies in the clinic.


Asunto(s)
Susceptibilidad a Enfermedades , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/patología , Enfermedades Autoinmunes/terapia , Autoinmunidad , Biomarcadores , Manejo de la Enfermedad , Humanos , Activación de Linfocitos/inmunología , Terapia Molecular Dirigida , Autotolerancia/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
2.
Annu Rev Immunol ; 36: 755-781, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29677472

RESUMEN

Inflammatory bowel disease (IBD) defines a spectrum of complex disorders. Understanding how environmental risk factors, alterations of the intestinal microbiota, and polygenetic and epigenetic susceptibility impact on immune pathways is key for developing targeted therapies. Mechanistic understanding of polygenic IBD is complemented by Mendelian disorders that present with IBD, pharmacological interventions that cause colitis, autoimmunity, and multiple animal models. Collectively, this multifactorial pathogenesis supports a concept of immune checkpoints that control microbial-host interactions in the gut by modulating innate and adaptive immunity, as well as epithelial and mesenchymal cell responses. In addition to classical immunosuppressive strategies, we discuss how resetting the microbiota and restoring innate immune responses, in particular autophagy and epithelial barrier function, might be key for maintaining remission or preventing IBD. Targeting checkpoints in genetically stratified subgroups of patients with Mendelian disorder-associated IBD increasingly directs treatment strategies as part of personalized medicine.


Asunto(s)
Susceptibilidad a Enfermedades/inmunología , Enfermedades Inflamatorias del Intestino/etiología , Enfermedades Inflamatorias del Intestino/terapia , Animales , Biomarcadores , Enfermedad Crónica , Manejo de la Enfermedad , Modelos Animales de Enfermedad , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Disbiosis , Microbioma Gastrointestinal , Predisposición Genética a la Enfermedad , Humanos , Enfermedades Inflamatorias del Intestino/prevención & control , Terapia Molecular Dirigida , Investigación Biomédica Traslacional
3.
Annu Rev Immunol ; 35: 337-370, 2017 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-28142321

RESUMEN

Transcriptomics, the high-throughput characterization of RNAs, has been instrumental in defining pathogenic signatures in human autoimmunity and autoinflammation. It enabled the identification of new therapeutic targets in IFN-, IL-1- and IL-17-mediated diseases. Applied to immunomonitoring, transcriptomics is starting to unravel diagnostic and prognostic signatures that stratify patients, track molecular changes associated with disease activity, define personalized treatment strategies, and generally inform clinical practice. Herein, we review the use of transcriptomics to define mechanistic, diagnostic, and predictive signatures in human autoimmunity and autoinflammation. We discuss some of the analytical approaches applied to extract biological knowledge from high-dimensional data sets. Finally, we touch upon emerging applications of transcriptomics to study eQTLs, B and T cell repertoire diversity, and isoform usage.


Asunto(s)
Enfermedades Autoinmunes/diagnóstico , Inflamación/diagnóstico , Transcriptoma , Enfermedades Autoinmunes/inmunología , Conjuntos de Datos como Asunto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inflamación/inmunología , Almacenamiento y Recuperación de la Información , Terapia Molecular Dirigida , Monitorización Inmunológica , Pronóstico
4.
Cell ; 187(16): 4389-4407.e15, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-38917788

RESUMEN

Fewer than 200 proteins are targeted by cancer drugs approved by the Food and Drug Administration (FDA). We integrate Clinical Proteomic Tumor Analysis Consortium (CPTAC) proteogenomics data from 1,043 patients across 10 cancer types with additional public datasets to identify potential therapeutic targets. Pan-cancer analysis of 2,863 druggable proteins reveals a wide abundance range and identifies biological factors that affect mRNA-protein correlation. Integration of proteomic data from tumors and genetic screen data from cell lines identifies protein overexpression- or hyperactivation-driven druggable dependencies, enabling accurate predictions of effective drug targets. Proteogenomic identification of synthetic lethality provides a strategy to target tumor suppressor gene loss. Combining proteogenomic analysis and MHC binding prediction prioritizes mutant KRAS peptides as promising public neoantigens. Computational identification of shared tumor-associated antigens followed by experimental confirmation nominates peptides as immunotherapy targets. These analyses, summarized at https://targets.linkedomics.org, form a comprehensive landscape of protein and peptide targets for companion diagnostics, drug repurposing, and therapy development.


Asunto(s)
Neoplasias , Proteogenómica , Humanos , Proteogenómica/métodos , Neoplasias/genética , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Neoplasias/metabolismo , Terapia Molecular Dirigida , Inmunoterapia/métodos , Antígenos de Neoplasias/metabolismo , Antígenos de Neoplasias/genética , Línea Celular Tumoral , Antineoplásicos/uso terapéutico , Antineoplásicos/farmacología , Péptidos/metabolismo , Proteómica , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo
5.
Annu Rev Immunol ; 34: 31-64, 2016 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-27168239

RESUMEN

Inflammatory bowel disease (IBD), including Crohn disease and ulcerative colitis, is characterized by chronic intestinal inflammation due to a complex interaction of genetic determinants, disruption of mucosal barriers, aberrant inflammatory signals, loss of tolerance, and environmental triggers. Importantly, the incidence of pediatric IBD is rising, particularly in children younger than 10 years. In this review, we discuss the clinical presentation of these patients and highlight environmental exposures that may affect disease risk, particularly among people with a background genetic risk. With regard to both children and adults, we review advancements in understanding the intestinal epithelium, the mucosal immune system, and the resident microbiota, describing how dysfunction at any level can lead to diseases like IBD. We conclude with future directions for applying advances in IBD genetics to better understand pathogenesis and develop therapeutics targeting key pathogenic nodes.


Asunto(s)
Disbiosis/inmunología , Microbioma Gastrointestinal/inmunología , Inmunidad Mucosa , Inflamación/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Mucosa Intestinal/inmunología , Adulto , Animales , Niño , Preescolar , Exposición a Riesgos Ambientales/efectos adversos , Interacción Gen-Ambiente , Predisposición Genética a la Enfermedad , Humanos , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/terapia , Terapia Molecular Dirigida
6.
Annu Rev Immunol ; 34: 121-49, 2016 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-26735698

RESUMEN

Genomic DNA sequencing technologies have been one of the great advances of the 21st century, having decreased in cost by seven orders of magnitude and opening up new fields of investigation throughout research and clinical medicine. Genomics coupled with biochemical investigation has allowed the molecular definition of a growing number of new genetic diseases that reveal new concepts of immune regulation. Also, defining the genetic pathogenesis of these diseases has led to improved diagnosis, prognosis, genetic counseling, and, most importantly, new therapies. We highlight the investigational journey from patient phenotype to treatment using the newly defined XMEN disease, caused by the genetic loss of the MAGT1 magnesium transporter, as an example. This disease illustrates how genomics yields new fundamental immunoregulatory insights as well as how research genomics is integrated into clinical immunology. At the end, we discuss two other recently described diseases, CHAI/LATAIE (CTLA-4 deficiency) and PASLI (PI3K dysregulation), as additional examples of the journey from unknown immunological diseases to new precision medicine treatments using genomics.


Asunto(s)
Antígeno CTLA-4/genética , Proteínas de Transporte de Catión/genética , Genómica , Enfermedades del Sistema Inmune/genética , Mutación/genética , Fosfatidilinositol 3-Quinasas/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Animales , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Enfermedades del Sistema Inmune/terapia , Masculino , Terapia Molecular Dirigida , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia
7.
Annu Rev Immunol ; 34: 1-30, 2016 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-27168238

RESUMEN

I started research in high school, experimenting on immunological tolerance to transplantation antigens. This led to studies of the thymus as the site of maturation of T cells, which led to the discovery, isolation, and clinical transplantation of purified hematopoietic stem cells (HSCs). The induction of immune tolerance with HSCs has led to isolation of other tissue-specific stem cells for regenerative medicine. Our studies of circulating competing germline stem cells in colonial protochordates led us to document competing HSCs. In human acute myelogenous leukemia we showed that all preleukemic mutations occur in HSCs, and determined their order; the final mutations occur in a multipotent progenitor derived from the preleukemic HSC clone. With these, we discovered that CD47 is an upregulated gene in all human cancers and is a "don't eat me" signal; blocking it with antibodies leads to cancer cell phagocytosis. CD47 is the first known gene common to all cancers and is a target for cancer immunotherapy.


Asunto(s)
Antígeno CD47/metabolismo , Células Madre Hematopoyéticas/inmunología , Inmunoterapia/tendencias , Leucemia Mieloide Aguda/inmunología , Células Madre Multipotentes/fisiología , Linfocitos T/inmunología , Animales , Biomarcadores de Tumor/metabolismo , Antígeno CD47/genética , Humanos , Tolerancia Inmunológica , Leucemia Mieloide Aguda/terapia , Terapia Molecular Dirigida , Mutación/genética , Medicina Regenerativa , Inmunología del Trasplante
8.
Annu Rev Immunol ; 33: 291-353, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25861976

RESUMEN

Ion channels and transporters mediate the transport of charged ions across hydrophobic lipid membranes. In immune cells, divalent cations such as calcium, magnesium, and zinc have important roles as second messengers to regulate intracellular signaling pathways. By contrast, monovalent cations such as sodium and potassium mainly regulate the membrane potential, which indirectly controls the influx of calcium and immune cell signaling. Studies investigating human patients with mutations in ion channels and transporters, analysis of gene-targeted mice, or pharmacological experiments with ion channel inhibitors have revealed important roles of ionic signals in lymphocyte development and in innate and adaptive immune responses. We here review the mechanisms underlying the function of ion channels and transporters in lymphocytes and innate immune cells and discuss their roles in lymphocyte development, adaptive and innate immune responses, and autoimmunity, as well as recent efforts to develop pharmacological inhibitors of ion channels for immunomodulatory therapy.


Asunto(s)
Inmunidad Adaptativa/fisiología , Inmunidad Innata/fisiología , Canales Iónicos/metabolismo , Animales , Canales de Calcio/genética , Canales de Calcio/metabolismo , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Humanos , Hipersensibilidad/genética , Hipersensibilidad/inmunología , Hipersensibilidad/metabolismo , Síndromes de Inmunodeficiencia/tratamiento farmacológico , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Síndromes de Inmunodeficiencia/metabolismo , Inmunoterapia/métodos , Canales Iónicos/genética , Linfocitos/citología , Linfocitos/inmunología , Linfocitos/metabolismo , Mastocitos/inmunología , Mastocitos/metabolismo , Terapia Molecular Dirigida , Mutación , Transducción de Señal
9.
Annu Rev Immunol ; 32: 25-50, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24215318

RESUMEN

CD47 is a broadly expressed membrane protein that interacts with the myeloid inhibitory immunoreceptor SIRPα (also termed CD172a or SHPS-1). SIRPα is the prototypic member of the SIRP paired receptor family of closely related SIRP proteins. Engagement of SIRPα by CD47 provides a downregulatory signal that inhibits host cell phagocytosis, and CD47 therefore functions as a "don't-eat-me" signal. Here, we discuss recent structural analysis of CD47-SIRPα interactions and implications of this for the function and evolution of SIRPα and paired receptors in general. Furthermore, we review the proposed roles of CD47-SIRPα interactions in phagocytosis, (auto)immunity, and host defense, as well as its potential significance as a therapeutic target in cancer and inflammation and for improving graft survival in xenotransplantation.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Antígeno CD47/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Antígenos de Diferenciación/química , Antígenos de Diferenciación/genética , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/etiología , Antígeno CD47/química , Antígeno CD47/genética , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Enfermedades Hematológicas/tratamiento farmacológico , Enfermedades Hematológicas/etiología , Humanos , Sinapsis Inmunológicas/inmunología , Sinapsis Inmunológicas/metabolismo , Terapia Molecular Dirigida , Familia de Multigenes , Neoplasias/tratamiento farmacológico , Neoplasias/etiología , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología , Unión Proteica/efectos de los fármacos , Receptores Inmunológicos/química , Receptores Inmunológicos/genética , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo
10.
Cell ; 184(15): 3852-3872, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34297930

RESUMEN

Fibroblasts are diverse mesenchymal cells that participate in tissue homeostasis and disease by producing complex extracellular matrix and creating signaling niches through biophysical and biochemical cues. Transcriptionally and functionally heterogeneous across and within organs, fibroblasts encode regional positional information and maintain distinct cellular progeny. We summarize their development, lineages, functions, and contributions to fibrosis in four fibroblast-rich organs: skin, lung, skeletal muscle, and heart. We propose that fibroblasts are uniquely poised for tissue repair by easily reentering the cell cycle and exhibiting a reversible plasticity in phenotype and cell fate. These properties, when activated aberrantly, drive fibrotic disorders in humans.


Asunto(s)
Enfermedad , Fibroblastos/metabolismo , Salud , Animales , Linaje de la Célula , Humanos , Terapia Molecular Dirigida , Transducción de Señal
11.
Cell ; 184(2): 293, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33482093

RESUMEN

The ultra-rare, pediatric premature aging disorder Hutchinson-Gilford progeria syndrome (HGPS) is caused by mutation of LMNA, encoding the nuclear architectural protein lamin A. Patients develop atherosclerosis and typically die of heart failure in their teens. FDA-approved Zokinvy prevents farnesylation of lamin A, reduces vascular stiffness, and extends survival in HGPS patients. To view this Bench to Bedside, open or download the PDF.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Farnesiltransferasa/antagonistas & inhibidores , Progeria/tratamiento farmacológico , Progeria/enzimología , Inhibidores Enzimáticos/farmacología , Farnesiltransferasa/metabolismo , Humanos , Terapia Molecular Dirigida
12.
Cell ; 184(6): 1604-1620, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33740455

RESUMEN

Historically, emerging viruses appear constantly and have cost millions of human lives. Currently, climate change and intense globalization have created favorable conditions for viral transmission. Therefore, effective antivirals, especially those targeting the conserved protein in multiple unrelated viruses, such as the compounds targeting RNA-dependent RNA polymerase, are urgently needed to combat more emerging and re-emerging viruses in the future. Here we reviewed the development of antivirals with common targets, including those against the same protein across viruses, or the same viral function, to provide clues for development of antivirals for future epidemics.


Asunto(s)
Antivirales/uso terapéutico , Enfermedades Transmisibles Emergentes/tratamiento farmacológico , Enfermedades Transmisibles Emergentes/epidemiología , Terapia Molecular Dirigida/métodos , Pandemias , Virosis/tratamiento farmacológico , Virosis/epidemiología , Virus/enzimología , Animales , Antivirales/farmacología , Enfermedades Transmisibles Emergentes/virología , Humanos , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , Proteínas del Envoltorio Viral/antagonistas & inhibidores , Virosis/virología , Internalización del Virus/efectos de los fármacos
13.
Cell ; 184(12): 3143-3162.e32, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34004147

RESUMEN

Gene expression by RNA polymerase II (RNAPII) is tightly controlled by cyclin-dependent kinases (CDKs) at discrete checkpoints during the transcription cycle. The pausing checkpoint following transcription initiation is primarily controlled by CDK9. We discovered that CDK9-mediated, RNAPII-driven transcription is functionally opposed by a protein phosphatase 2A (PP2A) complex that is recruited to transcription sites by the Integrator complex subunit INTS6. PP2A dynamically antagonizes phosphorylation of key CDK9 substrates including DSIF and RNAPII-CTD. Loss of INTS6 results in resistance to tumor cell death mediated by CDK9 inhibition, decreased turnover of CDK9 phospho-substrates, and amplification of acute oncogenic transcriptional responses. Pharmacological PP2A activation synergizes with CDK9 inhibition to kill both leukemic and solid tumor cells, providing therapeutic benefit in vivo. These data demonstrate that fine control of gene expression relies on the balance between kinase and phosphatase activity throughout the transcription cycle, a process dysregulated in cancer that can be exploited therapeutically.


Asunto(s)
Quinasa 9 Dependiente de la Ciclina/metabolismo , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Proteína Fosfatasa 2/metabolismo , Proteínas de Unión al ARN/metabolismo , Transcripción Genética , Proteínas Supresoras de Tumor/metabolismo , Animales , Línea Celular Tumoral , Quinasa 9 Dependiente de la Ciclina/antagonistas & inhibidores , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones Endogámicos NOD , Fosforilación , Unión Proteica , ARN Polimerasa II/química , ARN Polimerasa II/metabolismo , Especificidad por Sustrato
14.
Cell ; 184(9): 2487-2502.e13, 2021 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-33857424

RESUMEN

Precision oncology has made significant advances, mainly by targeting actionable mutations in cancer driver genes. Aiming to expand treatment opportunities, recent studies have begun to explore the utility of tumor transcriptome to guide patient treatment. Here, we introduce SELECT (synthetic lethality and rescue-mediated precision oncology via the transcriptome), a precision oncology framework harnessing genetic interactions to predict patient response to cancer therapy from the tumor transcriptome. SELECT is tested on a broad collection of 35 published targeted and immunotherapy clinical trials from 10 different cancer types. It is predictive of patients' response in 80% of these clinical trials and in the recent multi-arm WINTHER trial. The predictive signatures and the code are made publicly available for academic use, laying a basis for future prospective clinical studies.


Asunto(s)
Biomarcadores de Tumor/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Medicina de Precisión , Mutaciones Letales Sintéticas , Transcriptoma/efectos de los fármacos , Anciano , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/inmunología , Ensayos Clínicos como Asunto , Femenino , Estudios de Seguimiento , Humanos , Inmunoterapia , Masculino , Neoplasias/genética , Neoplasias/patología , Pronóstico , Estudios Prospectivos , Estudios Retrospectivos , Tasa de Supervivencia
15.
Cell ; 184(19): 5031-5052.e26, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34534465

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with poor patient survival. Toward understanding the underlying molecular alterations that drive PDAC oncogenesis, we conducted comprehensive proteogenomic analysis of 140 pancreatic cancers, 67 normal adjacent tissues, and 9 normal pancreatic ductal tissues. Proteomic, phosphoproteomic, and glycoproteomic analyses were used to characterize proteins and their modifications. In addition, whole-genome sequencing, whole-exome sequencing, methylation, RNA sequencing (RNA-seq), and microRNA sequencing (miRNA-seq) were performed on the same tissues to facilitate an integrated proteogenomic analysis and determine the impact of genomic alterations on protein expression, signaling pathways, and post-translational modifications. To ensure robust downstream analyses, tumor neoplastic cellularity was assessed via multiple orthogonal strategies using molecular features and verified via pathological estimation of tumor cellularity based on histological review. This integrated proteogenomic characterization of PDAC will serve as a valuable resource for the community, paving the way for early detection and identification of novel therapeutic targets.


Asunto(s)
Adenocarcinoma/genética , Carcinoma Ductal Pancreático/genética , Neoplasias Pancreáticas/genética , Proteogenómica , Adenocarcinoma/diagnóstico , Adulto , Anciano , Anciano de 80 o más Años , Algoritmos , Carcinoma Ductal Pancreático/diagnóstico , Estudios de Cohortes , Células Endoteliales/metabolismo , Epigénesis Genética , Femenino , Dosificación de Gen , Genoma Humano , Glucólisis , Glicoproteínas/biosíntesis , Humanos , Masculino , Persona de Mediana Edad , Terapia Molecular Dirigida , Neoplasias Pancreáticas/diagnóstico , Fenotipo , Fosfoproteínas/metabolismo , Fosforilación , Pronóstico , Proteínas Quinasas/metabolismo , Proteoma/metabolismo , Especificidad por Sustrato , Transcriptoma/genética
16.
Cell ; 184(2): 384-403.e21, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33450205

RESUMEN

Many oncogenic insults deregulate RNA splicing, often leading to hypersensitivity of tumors to spliceosome-targeted therapies (STTs). However, the mechanisms by which STTs selectively kill cancers remain largely unknown. Herein, we discover that mis-spliced RNA itself is a molecular trigger for tumor killing through viral mimicry. In MYC-driven triple-negative breast cancer, STTs cause widespread cytoplasmic accumulation of mis-spliced mRNAs, many of which form double-stranded structures. Double-stranded RNA (dsRNA)-binding proteins recognize these endogenous dsRNAs, triggering antiviral signaling and extrinsic apoptosis. In immune-competent models of breast cancer, STTs cause tumor cell-intrinsic antiviral signaling, downstream adaptive immune signaling, and tumor cell death. Furthermore, RNA mis-splicing in human breast cancers correlates with innate and adaptive immune signatures, especially in MYC-amplified tumors that are typically immune cold. These findings indicate that dsRNA-sensing pathways respond to global aberrations of RNA splicing in cancer and provoke the hypothesis that STTs may provide unexplored strategies to activate anti-tumor immune pathways.


Asunto(s)
Antivirales/farmacología , Inmunidad/efectos de los fármacos , Empalmosomas/metabolismo , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/patología , Inmunidad Adaptativa/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Femenino , Amplificación de Genes/efectos de los fármacos , Humanos , Intrones/genética , Ratones , Terapia Molecular Dirigida , Proteínas Proto-Oncogénicas c-myc/metabolismo , Empalme del ARN/efectos de los fármacos , Empalme del ARN/genética , ARN Bicatenario/metabolismo , Transducción de Señal/efectos de los fármacos , Empalmosomas/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/genética
17.
Cell ; 184(18): 4753-4771.e27, 2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34388391

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is characterized by notorious resistance to current therapies attributed to inherent tumor heterogeneity and highly desmoplastic and immunosuppressive tumor microenvironment (TME). Unique proline isomerase Pin1 regulates multiple cancer pathways, but its role in the TME and cancer immunotherapy is unknown. Here, we find that Pin1 is overexpressed both in cancer cells and cancer-associated fibroblasts (CAFs) and correlates with poor survival in PDAC patients. Targeting Pin1 using clinically available drugs induces complete elimination or sustained remissions of aggressive PDAC by synergizing with anti-PD-1 and gemcitabine in diverse model systems. Mechanistically, Pin1 drives the desmoplastic and immunosuppressive TME by acting on CAFs and induces lysosomal degradation of the PD-1 ligand PD-L1 and the gemcitabine transporter ENT1 in cancer cells, besides activating multiple cancer pathways. Thus, Pin1 inhibition simultaneously blocks multiple cancer pathways, disrupts the desmoplastic and immunosuppressive TME, and upregulates PD-L1 and ENT1, rendering PDAC eradicable by immunochemotherapy.


Asunto(s)
Inmunoterapia , Terapia Molecular Dirigida , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/inmunología , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Aloinjertos/inmunología , Secuencias de Aminoácidos , Animales , Apoptosis/efectos de los fármacos , Antígeno B7-H1/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Sinergismo Farmacológico , Endocitosis/efectos de los fármacos , Tranportador Equilibrativo 1 de Nucleósido/metabolismo , Humanos , Terapia de Inmunosupresión , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Ratones , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/metabolismo , Oncogenes , Organoides/efectos de los fármacos , Organoides/patología , Transducción de Señal/efectos de los fármacos , Análisis de Supervivencia , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
18.
Annu Rev Biochem ; 89: 557-581, 2020 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-32208767

RESUMEN

The binding affinity and kinetics of target engagement are fundamental to establishing structure-activity relationships (SARs) for prospective therapeutic agents. Enhancing these binding parameters for operative targets, while minimizing binding to off-target sites, can translate to improved drug efficacy and a widened therapeutic window. Compound activity is typically assessed through modulation of an observed phenotype in cultured cells. Quantifying the corresponding binding properties under common cellular conditions can provide more meaningful interpretation of the cellular SAR analysis. Consequently, methods for assessing drug binding in living cells have advanced and are now integral to medicinal chemistry workflows. In this review, we survey key technological advancements that support quantitative assessments of target occupancy in cultured cells, emphasizing generalizable methodologies able to deliver analytical precision that heretofore required reductionist biochemical approaches.


Asunto(s)
Química Farmacéutica/métodos , Colorantes Fluorescentes/química , Ensayos Analíticos de Alto Rendimiento , Técnicas de Sonda Molecular , Terapia Molecular Dirigida/métodos , Transferencia de Energía por Resonancia de Bioluminiscencia , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Genes Reporteros , Humanos , Cinética , Imagen Óptica/métodos , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad
19.
Annu Rev Immunol ; 31: 705-42, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23330954

RESUMEN

A fascinating aspect of viral evolution relates to the ability of viruses to escape the adaptive immune response. The widely held view has been that the great variability of viral glycoproteins would be an absolute obstacle to the development of antibody-based therapies or vaccines that could confer broad and long-lasting protection. In the past five years, new approaches have been developed to interrogate human memory B cells and plasma cells with high efficiency and to isolate several broadly neutralizing antiviral antibodies against highly variable pathogens such as HIV-1 and influenza virus. These antibodies not only provide new tools for prophylaxis and therapy for viral diseases but also identify conserved epitopes that may be used to design new vaccines capable of conferring broader protection.


Asunto(s)
Anticuerpos Antivirales/fisiología , Pruebas de Neutralización/métodos , Animales , Anticuerpos Antivirales/uso terapéutico , Anticuerpos Anti-VIH/uso terapéutico , Infecciones por VIH/inmunología , Infecciones por VIH/prevención & control , Infecciones por VIH/virología , Humanos , Inmunización Pasiva/métodos , Gripe Humana/inmunología , Gripe Humana/prevención & control , Gripe Humana/virología , Terapia Molecular Dirigida/métodos
20.
Cell ; 180(1): 9-14, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31951522

RESUMEN

This commentary introduces a new clinical trial construct, the Master Observational Trial (MOT), which hybridizes the power of molecularly based master interventional protocols with the breadth of real-world data. The MOT provides a clinical venue to allow molecular medicine to rapidly advance, answers questions that traditional interventional trials generally do not address, and seamlessly integrates with interventional trials in both diagnostic and therapeutic arenas. The result is a more comprehensive data collection ecosystem in precision medicine.


Asunto(s)
Estudios Observacionales como Asunto/métodos , Medicina de Precisión/métodos , Proyectos de Investigación/normas , Macrodatos , Protocolos de Ensayos Clínicos como Asunto , Humanos , Terapia Molecular Dirigida/métodos , Terapia Molecular Dirigida/tendencias , Estudios Observacionales como Asunto/normas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA