Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros











Intervalo de año de publicación
1.
bioRxiv ; 2023 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-37292765

RESUMEN

Overexpression of repetitive elements is an emerging hallmark of human cancers 1 . Diverse repeats can mimic viruses by replicating within the cancer genome through retrotransposition, or presenting pathogen-associated molecular patterns (PAMPs) to the pattern recognition receptors (PRRs) of the innate immune system 2-5 . Yet, how specific repeats affect tumor evolution and shape the tumor immune microenvironment (TME) in a pro- or anti-tumorigenic manner remains poorly defined. Here, we integrate whole genome and total transcriptome data from a unique autopsy cohort of multiregional samples collected in pancreatic ductal adenocarcinoma (PDAC) patients, into a comprehensive evolutionary analysis. We find that more recently evolved S hort I nterspersed N uclear E lements (SINE), a family of retrotransposable repeats, are more likely to form immunostimulatory double-strand RNAs (dsRNAs). Consequently, younger SINEs are strongly co-regulated with RIG-I like receptor associated type-I interferon genes but anti-correlated with pro-tumorigenic macrophage infiltration. We discover that immunostimulatory SINE expression in tumors is regulated by either L ong I nterspersed N uclear E lements 1 (LINE1/L1) mobility or ADAR1 activity in a TP53 mutation dependent manner. Moreover, L1 retrotransposition activity tracks with tumor evolution and is associated with TP53 mutation status. Altogether, our results suggest pancreatic tumors actively evolve to modulate immunogenic SINE stress and induce pro-tumorigenic inflammation. Our integrative, evolutionary analysis therefore illustrates, for the first time, how dark matter genomic repeats enable tumors to co-evolve with the TME by actively regulating viral mimicry to their selective advantage.

2.
iScience ; 25(7): 104599, 2022 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-35789859

RESUMEN

Pattern recognition receptors (PRRs) protect against microbial invasion by detecting specific molecular patterns found in pathogens and initiating an immune response. Although microbial-derived PRR ligands have been extensively characterized, the contribution and relevance of endogenous ligands to PRR activation remains overlooked. Here, we characterize the landscape of endogenous ligands that engage RIG-I-like receptors (RLRs) upon infection by different RNA viruses. In each infection, several RNAs transcribed by RNA polymerase III (Pol3) specifically engaged RLRs, particularly the family of Y RNAs. Sensing of Y RNAs was dependent on their mimicking of viral secondary structure and their 5'-triphosphate extremity. Further, we found that HIV-1 triggered a VPR-dependent downregulation of RNA triphosphatase DUSP11 in vitro and in vivo, inducing a transcriptome-wide change of cellular RNA 5'-triphosphorylation that licenses Y RNA immunogenicity. Overall, our work uncovers the contribution of endogenous RNAs to antiviral immunity and demonstrates the importance of this pathway in HIV-1 infection.

3.
J Clin Invest ; 132(16)2022 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-35708912

RESUMEN

Aberrant expression of viral-like repeat elements is a common feature of epithelial cancers, and the substantial diversity of repeat species provides a distinct view of the cancer transcriptome. Repeatome profiling across ovarian, pancreatic, and colorectal cell lines identifies distinct clustering independent of tissue origin that is seen with coding gene analysis. Deeper analysis of ovarian cancer cell lines demonstrated that human satellite II (HSATII) satellite repeat expression was highly associated with epithelial-mesenchymal transition (EMT) and anticorrelated with IFN-response genes indicative of a more aggressive phenotype. SATII expression - and its correlation with EMT and anticorrelation with IFN-response genes - was also found in ovarian cancer RNA-Seq data and was associated with significantly shorter survival in a second independent cohort of patients with ovarian cancer. Repeat RNAs were enriched in tumor-derived extracellular vesicles capable of stimulating monocyte-derived macrophages, demonstrating a mechanism that alters the tumor microenvironment with these viral-like sequences. Targeting of HSATII with antisense locked nucleic acids stimulated IFN response and induced MHC I expression in ovarian cancer cell lines, highlighting a potential strategy of modulating the repeatome to reestablish antitumor cell immune surveillance.


Asunto(s)
Neoplasias Ováricas , Satélite de ARN , Carcinoma Epitelial de Ovario/genética , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Ováricas/genética , Fenotipo , ARN , Microambiente Tumoral/genética
4.
Cancer Cell ; 40(7): 738-753.e5, 2022 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-35679859

RESUMEN

How immune dysregulation affects recovery from COVID-19 infection in patients with cancer remains unclear. We analyzed cellular and humoral immune responses in 103 patients with prior COVID-19 infection, more than 20% of whom had delayed viral clearance. Delayed clearance was associated with loss of antibodies to nucleocapsid and spike proteins with a compensatory increase in functional T cell responses. High-dimensional analysis of peripheral blood samples demonstrated increased CD8+ effector T cell differentiation and a broad but poorly converged COVID-specific T cell receptor (TCR) repertoire in patients with prolonged disease. Conversely, patients with a CD4+ dominant immunophenotype had a lower incidence of prolonged disease and exhibited a deep and highly select COVID-associated TCR repertoire, consistent with effective viral clearance and development of T cell memory. These results highlight the importance of B cells and CD4+ T cells in promoting durable SARS-CoV-2 clearance and the significance of coordinated cellular and humoral immunity for long-term disease control.


Asunto(s)
COVID-19 , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Humanos , Inmunidad Celular , Inmunidad Humoral , Memoria Inmunológica , Receptores de Antígenos de Linfocitos T , SARS-CoV-2
5.
Cancer Discov ; 12(6): 1462-1481, 2022 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-35320348

RESUMEN

Altered RNA expression of repetitive sequences and retrotransposition are frequently seen in colorectal cancer, implicating a functional importance of repeat activity in cancer progression. We show the nucleoside reverse transcriptase inhibitor 3TC targets activities of these repeat elements in colorectal cancer preclinical models with a preferential effect in p53-mutant cell lines linked with direct binding of p53 to repeat elements. We translate these findings to a human phase II trial of single-agent 3TC treatment in metastatic colorectal cancer with demonstration of clinical benefit in 9 of 32 patients. Analysis of 3TC effects on colorectal cancer tumorspheres demonstrates accumulation of immunogenic RNA:DNA hybrids linked with induction of interferon response genes and DNA damage response. Epigenetic and DNA-damaging agents induce repeat RNAs and have enhanced cytotoxicity with 3TC. These findings identify a vulnerability in colorectal cancer by targeting the viral mimicry of repeat elements. SIGNIFICANCE: Colorectal cancers express abundant repeat elements that have a viral-like life cycle that can be therapeutically targeted with nucleoside reverse transcriptase inhibitors (NRTI) commonly used for viral diseases. NRTIs induce DNA damage and interferon response that provide a new anticancer therapeutic strategy. This article is highlighted in the In This Issue feature, p. 1397.


Asunto(s)
Neoplasias Colorrectales , ADN Polimerasa Dirigida por ARN , Animales , Antivirales , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , ADN , Humanos , Interferones/metabolismo , Lamivudine , Estadios del Ciclo de Vida , ARN , ADN Polimerasa Dirigida por ARN/metabolismo , Proteína p53 Supresora de Tumor/genética
6.
Clin Infect Dis ; 75(1): e774-e782, 2022 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34644393

RESUMEN

BACKGROUND: Vaccine-induced clinical protection against severe acute respiratory syndrome coronavirus 2 (SARS CoV-2) variants is an evolving target. There are limited genomic level data on SARS CoV-2 breakthrough infections and vaccine effectiveness (VE) since the global spread of the B.1.617.2 (Delta) variant. METHODS: In a retrospective study from 1 November 2020 to 31 August 2021, divided as pre-Delta and Delta-dominant periods, laboratory-confirmed SARS CoV-2 infections among healthcare personnel (HCP) at a large tertiary cancer center in New York City were examined to compare the weekly infection rate-ratio in vaccinated, partially vaccinated, and unvaccinated HCP. We describe the clinical and genomic epidemiologic features of post-vaccine infections to assess for selection of variants of concern (VOC)/variants of interest (VOI) in the early post-vaccine period and impact of B.1.617.2 (Delta) variant domination on VE. RESULTS: Among 13658 HCP in our cohort, 12379 received at least 1 dose of a messenger RNA (mRNA) vaccine. In the pre-Delta period overall VE was 94.5%. Whole genome sequencing (WGS) of 369 isolates in the pre-Delta period did not reveal a clade bias for VOC/VOI specific to post-vaccine infections. VE in the Delta dominant phase was 75.6%. No hospitalizations occurred among vaccinated HCP in the entire study period, compared to 17 hospitalizations and 1 death among unvaccinated HCP. CONCLUSIONS: Findings show high VE among HCP in New York City in the pre-Delta phase, with moderate decline in VE post-Delta emergence. SARS CoV-2 clades were similarly distributed among vaccinated and unvaccinated infected HCP without apparent clustering during the pre-Delta period of diverse clade circulation. Strong vaccine protection against hospitalization was maintained through the entire study period.


Asunto(s)
COVID-19 , SARS-CoV-2 , COVID-19/epidemiología , COVID-19/prevención & control , Atención a la Salud , Genómica , Humanos , Ciudad de Nueva York/epidemiología , ARN Mensajero , Estudios Retrospectivos , SARS-CoV-2/genética
7.
Cancer Discov ; 12(1): 62-73, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34753749

RESUMEN

Coronavirus disease 2019 (COVID-19) infection results in both acute mortality and persistent and/or recurrent disease in patients with hematologic malignancies, but the drivers of persistent infection in this population are unknown. We found that B-cell lymphomas were at particularly high risk for persistent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) positivity. Further analysis of these patients identified discrete risk factors for initial disease severity compared with disease chronicity. Active therapy and diminished T-cell counts were drivers of acute mortality in COVID-19-infected patients with lymphoma. Conversely, B cell-depleting therapy was the primary driver of rehospitalization for COVID-19. In patients with persistent SARS-CoV-2 positivity, we observed high levels of viral entropy consistent with intrahost viral evolution, particularly in patients with impaired CD8+ T-cell immunity. These results suggest that persistent COVID-19 infection is likely to remain a risk in patients with impaired adaptive immunity and that additional therapeutic strategies are needed to enable viral clearance in this high-risk population. SIGNIFICANCE: We describe the largest cohort of persistent symptomatic COVID-19 infection in patients with lymphoid malignancies and identify B-cell depletion as the key immunologic driver of persistent infection. Furthermore, we demonstrate ongoing intrahost viral evolution in patients with persistent COVID-19 infection, particularly in patients with impaired CD8+ T-cell immunity.This article is highlighted in the In This Issue feature, p. 1.


Asunto(s)
COVID-19/inmunología , COVID-19/virología , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/virología , Infección Persistente/inmunología , Infección Persistente/virología , Adulto , Anciano , Anciano de 80 o más Años , Linfocitos B/inmunología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Factores de Riesgo , SARS-CoV-2/inmunología , Linfocitos T/inmunología
8.
Nat Commun ; 11(1): 6319, 2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-33298930

RESUMEN

The relationship of SARS-CoV-2 pulmonary infection and severity of disease is not fully understood. Here we show analysis of autopsy specimens from 24 patients who succumbed to SARS-CoV-2 infection using a combination of different RNA and protein analytical platforms to characterize inter-patient and intra-patient heterogeneity of pulmonary virus infection. There is a spectrum of high and low virus cases associated with duration of disease. High viral cases have high activation of interferon pathway genes and a predominant M1-like macrophage infiltrate. Low viral cases are more heterogeneous likely reflecting inherent patient differences in the evolution of host response, but there is consistent indication of pulmonary epithelial cell recovery based on napsin A immunohistochemistry and RNA expression of surfactant and mucin genes. Using a digital spatial profiling platform, we find the virus corresponds to distinct spatial expression of interferon response genes demonstrating the intra-pulmonary heterogeneity of SARS-CoV-2 infection.


Asunto(s)
COVID-19 , Interacciones Microbiota-Huesped , Interferones/metabolismo , Pulmón , Adulto , Anciano , Anciano de 80 o más Años , Ácido Aspártico Endopeptidasas/metabolismo , Autopsia , COVID-19/inmunología , COVID-19/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Células Epiteliales/virología , Femenino , Humanos , Inmunidad , Inmunohistoquímica , Hibridación in Situ , Interferones/genética , Pulmón/patología , Pulmón/virología , Macrófagos/inmunología , Masculino , Persona de Mediana Edad , Mucinas/genética , Mucinas/metabolismo , Tensoactivos/metabolismo , Transcriptoma , Carga Viral
9.
Melanoma Res ; 30(6): 594-598, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32976222

RESUMEN

Mounting evidence supports a role for dysregulated long non-coding RNAs (lncRNA) in the development of many cancers. A recently discovered function of lncRNAs is to act as microRNA (miR) decoys or competing endogenous RNAs, which sequester specific miRs and relieve negative regulation of mRNA expression by miRs. Although a large number of non-coding RNAs are thought to function as competing endogenous RNAs, miR-sequestering lncRNAs involved in nevus to melanoma transformation remain largely unknown. In this study, we applied a bioinformatics approach to a unique dataset of benign melanocytic nevi and primary melanomas of the skin in order to fill this research gap. We modified a previously published miR target prediction algorithm, RNAhybrid, and improved its search efficiency. We reported the presence of many lncRNAs and miRs deregulated when transitioning from a senescence-like state of nevi to melanoma. We provided evidence of a relatively new and understudied mechanism of gene regulation during this process and identified for the first time lncRNAs (n = 122) that may potentially function as miR decoys as well as their target miRs during nevus to melanoma transformation. The knowledge presented here can be employed for developing biomarkers for diagnostic and risk stratification purposes.


Asunto(s)
Melanoma/genética , MicroARNs/metabolismo , Nevo/genética , ARN Largo no Codificante/metabolismo , Neoplasias Cutáneas/genética , Humanos
10.
medRxiv ; 2020 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-32766600

RESUMEN

The relationship of SARS-CoV-2 lung infection and severity of pulmonary disease is not fully understood. We analyzed autopsy specimens from 24 patients who succumbed to SARS-CoV-2 infection using a combination of different RNA and protein analytical platforms to characterize inter- and intra- patient heterogeneity of pulmonary virus infection. There was a spectrum of high and low virus cases that was associated with duration of disease and activation of interferon pathway genes. Using a digital spatial profiling platform, the virus corresponded to distinct spatial expression of interferon response genes and immune checkpoint genes demonstrating the intra-pulmonary heterogeneity of SARS-CoV-2 infection.

12.
Clin Cancer Res ; 26(2): 450-464, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31857430

RESUMEN

PURPOSE: Somatic mutations in cancer cells can give rise to novel protein sequences that can be presented by antigen-presenting cells as neoantigens to the host immune system. Tumor neoantigens represent excellent targets for immunotherapy, due to their specific expression in cancer tissue. Despite the widespread use of immunomodulatory drugs and immunotherapies that recharge T and NK cells, there has been no direct evidence that neoantigen-specific T-cell responses are elicited in multiple myeloma. EXPERIMENTAL DESIGN: Using next-generation sequencing data we describe the landscape of neo-antigens in 184 patients with multiple myeloma and successfully validate neoantigen-specific T cells in patients with multiple myeloma and support the feasibility of neoantigen-based therapeutic vaccines for use in cancers with intermediate mutational loads such as multiple myeloma. RESULTS: In this study, we demonstrate an increase in neoantigen load in relapsed patients with multiple myeloma as compared with newly diagnosed patients with multiple myeloma. Moreover, we identify shared neoantigens across multiple patients in three multiple myeloma oncogenic driver genes (KRAS, NRAS, and IRF4). Next, we validate neoantigen T-cell response and clonal expansion in correlation with clinical response in relapsed patients with multiple myeloma. This is the first study to experimentally validate the immunogenicity of predicted neoantigens from next-generation sequencing in relapsed patients with multiple myeloma. CONCLUSIONS: Our findings demonstrate that somatic mutations in multiple myeloma can be immunogenic and induce neoantigen-specific T-cell activation that is associated with antitumor activity in vitro and clinical response in vivo. Our results provide the foundation for using neoantigen targeting strategies such as peptide vaccines in future trials for patients with multiple myeloma.


Asunto(s)
Antígenos de Neoplasias/genética , Vacunas contra el Cáncer/inmunología , Mieloma Múltiple/genética , Mieloma Múltiple/inmunología , Mutación , Péptidos/inmunología , Linfocitos T/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Vacunas contra el Cáncer/uso terapéutico , Supervivencia sin Enfermedad , Resistencia a Antineoplásicos , Femenino , Humanos , Inmunoterapia/métodos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/patología , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/patología , Tasa de Supervivencia
13.
Nat Commun ; 10(1): 90, 2019 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-30626867

RESUMEN

Tandem satellite repeats account for 3% of the human genome. One of them, Human Satellite II (HSATII), is highly expressed in several epithelial cancers and cancer cell lines. Here we report an acute induction of HSATII RNA in human cells infected with two herpes viruses. We show that human cytomegalovirus (HCMV) IE1 and IE2 proteins cooperate to induce HSATII RNA affecting several aspects of the HCMV replication cycle, viral titers and infected-cell processes. HSATII RNA expression in tissue from two chronic HCMV colitis patients correlates with the strength of CMV antigen staining. Thus, endogenous HSATII RNA synthesis after herpesvirus infections appears to have functionally important consequences for viral replication and may provide a novel insight into viral pathogenesis. The HSATII induction seen in both infected and cancer cells suggests possible convergence upon common HSATII-based regulatory mechanisms in these seemingly disparate diseases.


Asunto(s)
Secuencias Repetitivas Esparcidas/fisiología , Línea Celular , Movimiento Celular , Fibroblastos/metabolismo , Fibroblastos/virología , Regulación de la Expresión Génica , Herpesviridae , Genética Humana , Humanos , Hibridación in Situ , ARN/genética , ARN/metabolismo , Regulación hacia Arriba
14.
Pigment Cell Melanoma Res ; 31(5): 636-640, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29665239

RESUMEN

FBXW7 is well characterized as a tumor suppressor in many human cancers including melanoma; however, the mechanisms of tumor-suppressive function have not been fully elucidated. We leveraged two distinct RNA sequencing datasets: human melanoma cell lines (n = 10) with control versus silenced FBXW7 and a cohort of human melanoma tumor samples (n = 51) to define the transcriptomic fingerprint regulated by FBXW7. Here, we report that loss of FBXW7 enhances a mitochondrial gene transcriptional program that is dependent on MITF in human melanoma and confers poor patient outcomes. MITF is a lineage-specific master regulator of melanocytes and together with PGC-1alpha is a marker for melanoma subtypes with dependence for mitochondrial oxidative metabolism. We found that inactivation of FBXW7 elevates MITF protein levels in melanoma cells. In vitro studies examining loss of FBXW7 and MITF alone or in combination showed that FBXW7 is an upstream regulator for the MITF/PGC-1 signaling.


Asunto(s)
Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Regulación Neoplásica de la Expresión Génica , Melanoma/genética , Melanoma/patología , Factor de Transcripción Asociado a Microftalmía/metabolismo , Mitocondrias/genética , Células Cultivadas , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Humanos , Melanocitos/metabolismo , Melanocitos/patología , Melanoma/metabolismo , Factor de Transcripción Asociado a Microftalmía/genética , Mitocondrias/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Pronóstico , Transducción de Señal , Tasa de Supervivencia , Transcripción Genética
15.
Cell Rep ; 23(2): 512-521, 2018 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-29642008

RESUMEN

It has been posited that anti-tumoral innate activation is driven by derepression of endogenous repeats. We compared RNA sequencing protocols to assess repeat transcriptomes in The Cancer Genome Atlas (TCGA). Although poly(A) selection efficiently detects coding genes, most non-coding genes, and limited subsets of repeats, it fails to capture overall repeat expression and co-expression. Alternatively, total RNA expression reveals distinct repeat co-expression subgroups and delivers greater dynamic changes, implying they may serve as better biomarkers of clinical outcomes. We show that endogenous retrovirus expression predicts immunotherapy response better than conventional immune signatures in one cohort yet is not predictive in another. Moreover, we find that global repeat derepression, including the HSATII satellite repeat, correlates with an immunosuppressive phenotype in colorectal and pancreatic tumors and validate in situ. In conclusion, we stress the importance of analyzing the full spectrum of repeat transcription to decode their role in tumor immunity.


Asunto(s)
Neoplasias/genética , Secuencias Repetitivas de Ácidos Nucleicos/genética , Subgrupos de Linfocitos T/metabolismo , Transcriptoma , Anticuerpos Monoclonales/uso terapéutico , Análisis por Conglomerados , Retrovirus Endógenos/metabolismo , Humanos , Inmunoterapia , Estimación de Kaplan-Meier , Elementos de Nucleótido Esparcido Largo/genética , Neoplasias/mortalidad , Neoplasias/terapia , Análisis de Secuencia de ARN , Subgrupos de Linfocitos T/citología
16.
Nature ; 551(7681): 517-520, 2017 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-29132144

RESUMEN

Checkpoint blockade immunotherapies enable the host immune system to recognize and destroy tumour cells. Their clinical activity has been correlated with activated T-cell recognition of neoantigens, which are tumour-specific, mutated peptides presented on the surface of cancer cells. Here we present a fitness model for tumours based on immune interactions of neoantigens that predicts response to immunotherapy. Two main factors determine neoantigen fitness: the likelihood of neoantigen presentation by the major histocompatibility complex (MHC) and subsequent recognition by T cells. We estimate these components using the relative MHC binding affinity of each neoantigen to its wild type and a nonlinear dependence on sequence similarity of neoantigens to known antigens. To describe the evolution of a heterogeneous tumour, we evaluate its fitness as a weighted effect of dominant neoantigens in the subclones of the tumour. Our model predicts survival in anti-CTLA-4-treated patients with melanoma and anti-PD-1-treated patients with lung cancer. Importantly, low-fitness neoantigens identified by our method may be leveraged for developing novel immunotherapies. By using an immune fitness model to study immunotherapy, we reveal broad similarities between the evolution of tumours and rapidly evolving pathogens.


Asunto(s)
Antígenos de Neoplasias/inmunología , Inmunoterapia , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia , Melanoma/inmunología , Melanoma/terapia , Modelos Inmunológicos , Presentación de Antígeno , Antígenos de Neoplasias/genética , Antígeno CTLA-4/antagonistas & inhibidores , Antígeno CTLA-4/genética , Antígeno CTLA-4/inmunología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Puntos de Control del Ciclo Celular/genética , Puntos de Control del Ciclo Celular/inmunología , Estudios de Cohortes , Evolución Molecular , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Activación de Linfocitos , Melanoma/genética , Melanoma/patología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Análisis de Supervivencia , Linfocitos T/inmunología
17.
JCI Insight ; 2(9)2017 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-28469092

RESUMEN

BACKGROUND: Melanoma is a heterogeneous malignancy. We set out to identify the molecular underpinnings of high-risk melanomas, those that are likely to progress rapidly, metastasize, and result in poor outcomes. METHODS: We examined transcriptome changes from benign states to early-, intermediate-, and late-stage tumors using a set of 78 treatment-naive melanocytic tumors consisting of primary melanomas of the skin and benign melanocytic lesions. We utilized a next-generation sequencing platform that enabled a comprehensive analysis of protein-coding and -noncoding RNA transcripts. RESULTS: Gene expression changes unequivocally discriminated between benign and malignant states, and a dual epigenetic and immune signature emerged defining this transition. To our knowledge, we discovered previously unrecognized melanoma subtypes. A high-risk primary melanoma subset was distinguished by a 122-epigenetic gene signature ("epigenetic" cluster) and TP53 family gene deregulation (TP53, TP63, and TP73). This subtype associated with poor overall survival and showed enrichment of cell cycle genes. Noncoding repetitive element transcripts (LINEs, SINEs, and ERVs) that can result in immunostimulatory signals recapitulating a state of "viral mimicry" were significantly repressed. The high-risk subtype and its poor predictive characteristics were validated in several independent cohorts. Additionally, primary melanomas distinguished by specific immune signatures ("immune" clusters) were identified. CONCLUSION: The TP53 family of genes and genes regulating the epigenetic machinery demonstrate strong prognostic and biological relevance during progression of early disease. Gene expression profiling of protein-coding and -noncoding RNA transcripts may be a better predictor for disease course in melanoma. This study outlines the transcriptional interplay of the cancer cell's epigenome with the immune milieu with potential for future therapeutic targeting. FUNDING: National Institutes of Health (CA154683, CA158557, CA177940, CA087497-13), Tisch Cancer Institute, Melanoma Research Foundation, the Dow Family Charitable Foundation, and the Icahn School of Medicine at Mount Sinai.

18.
Cell ; 169(4): 750-765.e17, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-28475900

RESUMEN

To guide the design of immunotherapy strategies for patients with early stage lung tumors, we developed a multiscale immune profiling strategy to map the immune landscape of early lung adenocarcinoma lesions to search for tumor-driven immune changes. Utilizing a barcoding method that allows a simultaneous single-cell analysis of the tumor, non-involved lung, and blood cells, we provide a detailed immune cell atlas of early lung tumors. We show that stage I lung adenocarcinoma lesions already harbor significantly altered T cell and NK cell compartments. Moreover, we identified changes in tumor-infiltrating myeloid cell (TIM) subsets that likely compromise anti-tumor T cell immunity. Paired single-cell analyses thus offer valuable knowledge of tumor-driven immune changes, providing a powerful tool for the rational design of immune therapies. VIDEO ABSTRACT.


Asunto(s)
Adenocarcinoma/inmunología , Adenocarcinoma/patología , Inmunidad Innata , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Análisis de la Célula Individual/métodos , Adenocarcinoma del Pulmón , Células Dendríticas/patología , Humanos , Células Asesinas Naturales/patología , Macrófagos/patología , Linfocitos T/patología , Microambiente Tumoral
19.
Nat Microbiol ; 2: 17037, 2017 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-28346446

RESUMEN

During the last few decades, the global incidence of dengue virus (DENV) has increased dramatically, and it is now endemic in more than 100 countries. To establish a productive infection in humans, DENV uses different strategies to inhibit or avoid the host innate immune system. Several DENV proteins have been shown to strategically target crucial components of the type I interferon system. Here, we report that the DENV NS2B protease cofactor targets the DNA sensor cyclic GMP-AMP synthase (cGAS) for lysosomal degradation to avoid the detection of mitochondrial DNA during infection. Such degradation subsequently results in the inhibition of type I interferon production in the infected cell. Our data demonstrate a mechanism by which cGAS senses cellular damage upon DENV infection.


Asunto(s)
ADN Mitocondrial/fisiología , Virus del Dengue/genética , Interacciones Huésped-Patógeno , Nucleotidiltransferasas/metabolismo , Proteínas no Estructurales Virales/metabolismo , ADN Mitocondrial/genética , Células Dendríticas/virología , Dengue/inmunología , Dengue/virología , Virus del Dengue/química , Virus del Dengue/enzimología , Virus del Dengue/inmunología , Células HEK293 , Humanos , Inmunidad Innata , Interferón Tipo I/inmunología , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/deficiencia , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/inmunología , Transducción de Señal , Proteínas no Estructurales Virales/genética
20.
Cell Stem Cell ; 20(3): 315-328.e7, 2017 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-28215825

RESUMEN

Myeloid malignancy is increasingly viewed as a disease spectrum, comprising hematopoietic disorders that extend across a phenotypic continuum ranging from clonal hematopoiesis to myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). In this study, we derived a collection of induced pluripotent stem cell (iPSC) lines capturing a range of disease stages encompassing preleukemia, low-risk MDS, high-risk MDS, and secondary AML. Upon their differentiation, we found hematopoietic phenotypes of graded severity and/or stage specificity that together delineate a phenotypic roadmap of disease progression culminating in serially transplantable leukemia. We also show that disease stage transitions, both reversal and progression, can be modeled in this system using genetic correction or introduction of mutations via CRISPR/Cas9 and that this iPSC-based approach can be used to uncover disease-stage-specific responses to drugs. Our study therefore provides insight into the cellular events demarcating the initiation and progression of myeloid transformation and a new platform for testing genetic and pharmacological interventions.


Asunto(s)
Transformación Celular Neoplásica/patología , Progresión de la Enfermedad , Células Madre Pluripotentes Inducidas/citología , Leucemia Mieloide Aguda/patología , Animales , Antineoplásicos/farmacología , Transformación Celular Neoplásica/efectos de los fármacos , Reprogramación Celular/efectos de los fármacos , Reprogramación Celular/genética , Análisis Mutacional de ADN , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Leucemia Mieloide Aguda/genética , Ratones , Modelos Biológicos , Síndromes Mielodisplásicos/patología , Trasplante de Neoplasias , Fenotipo , Transcriptoma/efectos de los fármacos , Transcriptoma/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA