Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 69(6): 923-937.e8, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29547721

RESUMEN

Dietary supplements such as vitamins and minerals are widely used in the hope of improving health but may have unidentified risks and side effects. In particular, a pathogenic link between dietary supplements and specific oncogenes remains unknown. Here we report that chondroitin-4-sulfate (CHSA), a natural glycosaminoglycan approved as a dietary supplement used for osteoarthritis, selectively promotes the tumor growth potential of BRAF V600E-expressing human melanoma cells in patient- and cell line-derived xenograft mice and confers resistance to BRAF inhibitors. Mechanistically, chondroitin sulfate glucuronyltransferase (CSGlcA-T) signals through its product CHSA to enhance casein kinase 2 (CK2)-PTEN binding and consequent phosphorylation and inhibition of PTEN, which requires CHSA chains and is essential to sustain AKT activation in BRAF V600E-expressing melanoma cells. However, this CHSA-dependent PTEN inhibition is dispensable in cancer cells expressing mutant NRAS or PI3KCA, which directly activate the PI3K-AKT pathway. These results suggest that dietary supplements may exhibit oncogene-dependent pro-tumor effects.


Asunto(s)
Carcinógenos/toxicidad , Transformación Celular Neoplásica/genética , Sulfatos de Condroitina/toxicidad , Suplementos Dietéticos/toxicidad , Melanoma/inducido químicamente , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias Cutáneas/inducido químicamente , Animales , Antinematodos/farmacología , Quinasa de la Caseína II/metabolismo , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Femenino , GTP Fosfohidrolasas/genética , Células HEK293 , Células HT29 , Humanos , Melanoma/tratamiento farmacológico , Melanoma/enzimología , Melanoma/genética , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones Transgénicos , Células 3T3 NIH , Proteínas Nucleares/genética , Fosfohidrolasa PTEN/antagonistas & inhibidores , Fosfohidrolasa PTEN/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/genética , Factores de Transcripción/genética , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Nature ; 549(7672): 389-393, 2017 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-28854169

RESUMEN

The pathophysiology of neurodegenerative diseases is poorly understood and there are few therapeutic options. Neurodegenerative diseases are characterized by progressive neuronal dysfunction and loss, and chronic glial activation. Whether microglial activation, which is generally viewed as a secondary process, is harmful or protective in neurodegeneration remains unclear. Late-onset neurodegenerative disease observed in patients with histiocytoses, which are clonal myeloid diseases associated with somatic mutations in the RAS-MEK-ERK pathway such as BRAF(V600E), suggests a possible role of somatic mutations in myeloid cells in neurodegeneration. Yet the expression of BRAF(V600E) in the haematopoietic stem cell lineage causes leukaemic and tumoural diseases but not neurodegenerative disease. Microglia belong to a lineage of adult tissue-resident myeloid cells that develop during organogenesis from yolk-sac erythro-myeloid progenitors (EMPs) distinct from haematopoietic stem cells. We therefore hypothesized that a somatic BRAF(V600E) mutation in the EMP lineage may cause neurodegeneration. Here we show that mosaic expression of BRAF(V600E) in mouse EMPs results in clonal expansion of tissue-resident macrophages and a severe late-onset neurodegenerative disorder. This is associated with accumulation of ERK-activated amoeboid microglia in mice, and is also observed in human patients with histiocytoses. In the mouse model, neurobehavioural signs, astrogliosis, deposition of amyloid precursor protein, synaptic loss and neuronal death were driven by ERK-activated microglia and were preventable by BRAF inhibition. These results identify the fetal precursors of tissue-resident macrophages as a potential cell-of-origin for histiocytoses and demonstrate that a somatic mutation in the EMP lineage in mice can drive late-onset neurodegeneration. Moreover, these data identify activation of the MAP kinase pathway in microglia as a cause of neurodegeneration and this offers opportunities for therapeutic intervention aimed at the prevention of neuronal death in neurodegenerative diseases.


Asunto(s)
Células Precursoras Eritroides/patología , Sistema de Señalización de MAP Quinasas , Mutación , Células Progenitoras Mieloides/patología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Proteínas Proto-Oncogénicas B-raf/genética , Animales , Células Clonales/enzimología , Células Clonales/metabolismo , Células Clonales/patología , Modelos Animales de Enfermedad , Células Precursoras Eritroides/enzimología , Células Precursoras Eritroides/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Histiocitosis/enzimología , Histiocitosis/genética , Histiocitosis/metabolismo , Histiocitosis/patología , Humanos , Macrófagos/enzimología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Microglía/enzimología , Microglía/metabolismo , Microglía/patología , Mosaicismo , Células Progenitoras Mieloides/enzimología , Células Progenitoras Mieloides/metabolismo , Enfermedades Neurodegenerativas/enzimología , Enfermedades Neurodegenerativas/metabolismo , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/metabolismo
3.
PLoS Pathog ; 15(2): e1007583, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30726291

RESUMEN

CD8 T cells are necessary for the elimination of intracellular pathogens, but during chronic viral infections, CD8 T cells become exhausted and unable to control the persistent infection. Programmed cell death-1 (PD-1) blockade therapies have been shown to improve CD8 T cell responses during chronic viral infections. These therapies have been licensed to treat cancers in humans, but they have not yet been licensed to treat chronic viral infections because limited benefit is seen in pre-clinical animal models of chronic infection. In the present study, we investigated whether TLR4 triggering could improve PD-1 therapy during a chronic viral infection. Using the model of chronic lymphocytic choriomeningitis virus (LCMV) infection in mice, we show that TLR4 triggering with sublethal doses of lipopolysaccharide (LPS) followed by PD-1 blockade results in superior improvement in circulating virus-specific CD8 T cell responses, relative to PD-1 blockade alone. Moreover, we show that the synergy between LPS and PD-1 blockade is dependent on B7 costimulation and mediated by a dendritic cell (DC) intrinsic mechanism. Systemic LPS administration may have safety concerns, motivating us to devise a safer regimen. We show that ex vivo activation of DCs with LPS, followed by adoptive DC transfer, results in a similar potentiation of PD-1 therapy without inducing wasting disease. In summary, our data demonstrate a previously unidentified role for LPS/TLR4 signaling in modulating the host response to PD-1 therapy. These findings may be important for developing novel checkpoint therapies against chronic viral infection.


Asunto(s)
Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor Toll-Like 4/metabolismo , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/fisiología , Enfermedad Crónica , Células Dendríticas , Femenino , Lipopolisacáridos/farmacología , Activación de Linfocitos , Coriomeningitis Linfocítica , Virus de la Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1/inmunología , Transducción de Señal , Receptor Toll-Like 4/inmunología , Virosis/inmunología
4.
Blood ; 130(14): 1644-1648, 2017 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-28801450

RESUMEN

Classical hairy cell leukemia (cHCL) is characterized by a near 100% frequency of the BRAFV600E mutation, whereas ∼30% of variant HCLs (vHCLs) have MAP2K1 mutations. However, recurrent genetic alterations cooperating with BRAFV600E or MAP2K1 mutations in HCL, as well as those in MAP2K1 wild-type vHCL, are not well defined. We therefore performed deep targeted mutational and copy number analysis of cHCL (n = 53) and vHCL (n = 8). The most common genetic alteration in cHCL apart from BRAFV600E was heterozygous loss of chromosome 7q, the minimally deleted region of which targeted wild-type BRAF, subdividing cHCL into those hemizygous versus heterozygous for the BRAFV600E mutation. In addition to CDKN1B mutations in cHCL, recurrent inactivating mutations in KMT2C (MLL3) were identified in 15% and 25% of cHCLs and vHCLs, respectively. Moreover, 13% of vHCLs harbored predicted activating mutations in CCND3 A change-of-function mutation in the splicing factor U2AF1 was also present in 13% of vHCLs. Genomic analysis of de novo vemurafenib-resistant cHCL identified a novel gain-of-function mutation in IRS1 and losses of NF1 and NF2, each of which contributed to resistance. These data provide further insight into the genetic bases of cHCL and vHCL and mechanisms of RAF inhibitor resistance encountered clinically.


Asunto(s)
Leucemia de Células Pilosas/genética , Mutación , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Ciclina D3/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Proteínas de Unión al ADN/genética , Resistencia a Antineoplásicos , Genómica , Humanos , Indoles/farmacología , Indoles/uso terapéutico , Leucemia de Células Pilosas/tratamiento farmacológico , MAP Quinasa Quinasa 1/genética , Proteínas Proto-Oncogénicas B-raf/genética , Factor de Empalme U2AF/genética , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Vemurafenib
5.
Blood ; 130(4): 397-407, 2017 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-28576879

RESUMEN

Chronic myelomonocytic leukemia (CMML) and juvenile myelomonocytic leukemia (JMML) are myelodysplastic syndrome (MDS)/myeloproliferative neoplasm (MPN) overlap disorders characterized by monocytosis, myelodysplasia, and a characteristic hypersensitivity to granulocyte-macrophage colony-stimulating factor (GM-CSF). Currently, there are no available disease-modifying therapies for CMML, nor are there preclinical models that fully recapitulate the unique features of CMML. Through use of immunocompromised mice with transgenic expression of human GM-CSF, interleukin-3, and stem cell factor in a NOD/SCID-IL2Rγnull background (NSGS mice), we demonstrate remarkable engraftment of CMML and JMML providing the first examples of serially transplantable and genetically accurate models of CMML. Xenotransplantation of CD34+ cells (n = 8 patients) or unfractionated bone marrow (BM) or peripheral blood mononuclear cells (n = 10) resulted in robust engraftment of CMML in BM, spleen, liver, and lung of recipients (n = 82 total mice). Engrafted cells were myeloid-restricted and matched the immunophenotype, morphology, and genetic mutations of the corresponding patient. Similar levels of engraftment were seen upon serial transplantation of human CD34+ cells in secondary NSGS recipients (2/5 patients, 6/11 mice), demonstrating the durability of CMML grafts and functionally validating CD34+ cells as harboring the disease-initiating compartment in vivo. Successful engraftments of JMML primary samples were also achieved in all NSGS recipients (n = 4 patients, n = 12 mice). Engraftment of CMML and JMML resulted in overt phenotypic abnormalities and lethality in recipients, which facilitated evaluation of the JAK2/FLT3 inhibitor pacritinib in vivo. These data reveal that NSGS mice support the development of CMML and JMML disease-initiating and mature leukemic cells in vivo, allowing creation of genetically accurate preclinical models of these disorders.


Asunto(s)
Hidrocarburos Aromáticos con Puentes/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielomonocítica Juvenil/tratamiento farmacológico , Síndromes Mielodisplásicos/tratamiento farmacológico , Pirimidinas/farmacología , Animales , Femenino , Xenoinjertos , Humanos , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Leucemia Mielomonocítica Juvenil/genética , Leucemia Mielomonocítica Juvenil/metabolismo , Leucemia Mielomonocítica Juvenil/patología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/patología , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Trasplante de Neoplasias , Ensayos Antitumor por Modelo de Xenoinjerto , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/metabolismo
6.
Proc Natl Acad Sci U S A ; 111(50): E5401-10, 2014 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-25516983

RESUMEN

Patients with myeloproliferative neoplasms (MPNs) are at significant, cumulative risk of leukemic transformation to acute myeloid leukemia (AML), which is associated with adverse clinical outcome and resistance to standard AML therapies. We performed genomic profiling of post-MPN AML samples; these studies demonstrate somatic tumor protein 53 (TP53) mutations are common in JAK2V617F-mutant, post-MPN AML but not in chronic-phase MPN and lead to clonal dominance of JAK2V617F/TP53-mutant leukemic cells. Consistent with these data, expression of JAK2V617F combined with Tp53 loss led to fully penetrant AML in vivo. JAK2V617F-mutant, Tp53-deficient AML was characterized by an expanded megakaryocyte erythroid progenitor population that was able to propagate the disease in secondary recipients. In vitro studies revealed that post-MPN AML cells were sensitive to decitabine, the JAK1/2 inhibitor ruxolitinib, or the heat shock protein 90 inhibitor 8-(6-iodobenzo[d][1.3]dioxol-5-ylthio)-9-(3-(isopropylamino)propyl)-9H-purine-6-amine (PU-H71). Treatment with ruxolitinib or PU-H71 improved survival of mice engrafted with JAK2V617F-mutant, Tp53-deficient AML, demonstrating therapeutic efficacy for these targeted therapies and providing a rationale for testing these therapies in post-MPN AML.


Asunto(s)
Neoplasias Hematológicas/complicaciones , Janus Quinasa 2/genética , Leucemia Mieloide Aguda/genética , Trastornos Mieloproliferativos/complicaciones , Trastornos Mieloproliferativos/genética , Proteína p53 Supresora de Tumor/genética , Animales , Azacitidina/análogos & derivados , Azacitidina/farmacología , Benzodioxoles/farmacología , Western Blotting , Ensayo de Unidades Formadoras de Colonias , Decitabina , Exoma/genética , Citometría de Flujo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/etiología , Ratones , Mutación Missense/genética , Nitrilos , Purinas/farmacología , Pirazoles/farmacología , Pirimidinas
7.
N Engl J Med ; 367(24): 2316-21, 2012 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-23134356

RESUMEN

Vemurafenib, a selective RAF inhibitor, extends survival among patients with BRAF V600E-mutant melanoma. Vemurafenib inhibits ERK signaling in BRAF V600E-mutant cells but activates ERK signaling in BRAF wild-type cells. This paradoxical activation of ERK signaling is the mechanistic basis for the development of RAS-mutant squamous-cell skin cancers in patients treated with RAF inhibitors. We report the accelerated growth of a previously unsuspected RAS-mutant leukemia in a patient with melanoma who was receiving vemurafenib. Exposure to vemurafenib induced hyperactivation of ERK signaling and proliferation of the leukemic cell population, an effect that was reversed on drug withdrawal.


Asunto(s)
Genes ras , Indoles/efectos adversos , Leucemia Mielomonocítica Crónica/genética , Melanoma/genética , Inhibidores de Proteínas Quinasas/efectos adversos , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Sulfonamidas/efectos adversos , Anciano , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Humanos , Indoles/uso terapéutico , Leucemia Mielomonocítica Crónica/inducido químicamente , Recuento de Leucocitos , Masculino , Melanoma/tratamiento farmacológico , Mutación , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/genética , Sulfonamidas/uso terapéutico , Vemurafenib
8.
J Clin Invest ; 134(15)2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38861331

RESUMEN

Viral vectors are being used for the treatment of cancer. Yet, their efficacy varies among tumors and their use poses challenges in immunosuppressed patients, underscoring the need for alternatives. We report striking antitumoral effects by a nonlytic viral vector based on attenuated lymphocytic choriomeningitis virus (r3LCMV). We show in multiple tumor models that injection of tumor-bearing mice with this vector results in improved tumor control and survival. Importantly, r3LCMV improved tumor control in immunodeficient Rag1-/- mice and MyD88-/- mice, suggesting that multiple pathways contributed to the antitumoral effects. The antitumoral effects of r3LCMV were also observed when this vector was administered several weeks before tumor challenges, suggesting the induction of trained immunity. Single-cell RNA sequencing analyses, antibody blockade experiments, and knockout models revealed a critical role for host-intrinsic IFN-I in the antitumoral efficacy of r3LCMV vectors. Collectively, these data demonstrate potent antitumoral effects by r3LCMV vectors and unveil multiple mechanisms underlying their antitumoral efficacy.


Asunto(s)
Vectores Genéticos , Interferón Tipo I , Virus de la Coriomeningitis Linfocítica , Ratones Noqueados , Animales , Virus de la Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/genética , Ratones , Interferón Tipo I/inmunología , Interferón Tipo I/genética , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Humanos , Línea Celular Tumoral , Ratones Endogámicos C57BL , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/genética , Neoplasias Experimentales/terapia , Neoplasias Experimentales/patología , Proteínas de Homeodominio
9.
bioRxiv ; 2023 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-38106001

RESUMEN

Viral vectors are being used for the treatment of cancer. Yet their efficacy varies among tumors and their use poses challenges in immunosuppressed patients, underscoring the need for alternatives. We report striking antitumoral effects by a nonlytic viral vector based on attenuated lymphocytic choriomeningitis virus (r3LCMV). We show in multiple tumor models that injection of tumor-bearing mice with this novel vector results in improved tumor control and survival. Importantly, r3LCMV also improved tumor control in immunodeficient Rag1-/- mice. Single cell RNA-Seq analyses, antibody blockade experiments, and KO models revealed a critical role for host IFN-I in the antitumoral efficacy of r3LCMV vectors. Collectively, these data demonstrate potent antitumoral effects by a replication-attenuated LCMV vector and unveil mechanisms underlying its antitumoral efficacy.

10.
Blood Adv ; 7(23): 7319-7328, 2023 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-37874915

RESUMEN

Langerhans cell histiocytosis (LCH) is an inflammatory myeloid neoplasm characterized by the accumulation of clonal mononuclear phagocyte system cells expressing CD1a and CD207. In the past decade, molecular profiling of LCH as well as other histiocytic neoplasms demonstrated that these diseases are driven by MAPK activating alterations, with somatic BRAFV600E mutations in >50% of patients with LCH, and clinical inhibition of MAPK signaling has demonstrated remarkable clinical efficacy. At the same time, activating alterations in kinase-encoding genes, such as PIK3CA, ALK, RET, and CSF1R, which can activate mitogenic pathways independent from the MAPK pathway, have been reported in a subset of histiocytic neoplasms with anecdotal evidence of successful targeted treatment of histiocytoses harboring driver alterations in RET, ALK, and CSF1R. However, evidence supporting the biological consequences of expression of PIK3CA mutations in hematopoietic cells has been lacking, and whether targeted inhibition of PI3K is clinically efficacious in histiocytic neoplasms is unknown. Here, we provide evidence that activating mutations in PIK3CA can drive histiocytic neoplasms in vivo using a conditional knockin mouse expressing mutant PIK3CAH1047R in monocyte/dendritic cell progenitors. In parallel, we demonstrate successful treatment of PIK3CA-mutated, multisystemic LCH using alpelisib, an inhibitor of the alpha catalytic subunit of PI3K. Alpelisib demonstrated a tolerable safety profile at a dose of 750 mg per week and clinical and metabolic complete remission in a patient with PIK3CA-mutated LCH. These data demonstrate PIK3CA as a targetable noncanonical driver of LCH and underscore the importance of mutational analysis-based personalized treatment in histiocytic neoplasms.


Asunto(s)
Neoplasias Hematológicas , Histiocitosis de Células de Langerhans , Humanos , Animales , Ratones , Proteínas Proto-Oncogénicas B-raf/genética , Histiocitosis de Células de Langerhans/tratamiento farmacológico , Proteínas Tirosina Quinasas Receptoras , Fosfatidilinositol 3-Quinasas/uso terapéutico , Fosfatidilinositol 3-Quinasa Clase I/genética
11.
Front Immunol ; 13: 908707, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35958615

RESUMEN

T cell-based therapies have been widely explored for the treatment of cancer and chronic infection, but B cell-based therapies have remained largely unexplored. To study the effect of B cell therapy, we adoptively transferred virus-specific B cells into mice that were chronically infected with lymphocytic choriomeningitis virus (LCMV). Adoptive transfer of virus-specific B cells resulted in increase in antibody titers and reduction of viral loads. Importantly, the efficacy of B cell therapy was partly dependent on antibody effector functions, and was improved by co-transferring virus-specific CD4 T cells. These findings provide a proof-of-concept that adoptive B cell therapy can be effective for the treatment of chronic infections, but provision of virus-specific CD4 T cells may be critical for optimal virus neutralization.


Asunto(s)
Coriomeningitis Linfocítica , Animales , Tratamiento Basado en Trasplante de Células y Tejidos , Inmunoterapia Adoptiva , Coriomeningitis Linfocítica/terapia , Virus de la Coriomeningitis Linfocítica , Ratones , Ratones Endogámicos C57BL
12.
J Clin Invest ; 132(23)2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36219482

RESUMEN

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein is the main antigen in all approved COVID-19 vaccines and is also the only target for monoclonal antibody (mAb) therapies. Immune responses to other viral antigens are generated after SARS-CoV-2 infection, but their contribution to the antiviral response remains unclear. Here, we interrogated whether nucleocapsid-specific antibodies can improve protection against SARS-CoV-2. We first immunized mice with a nucleocapsid-based vaccine and then transferred sera from these mice into naive mice, followed by challenge with SARS-CoV-2. We show that mice that received nucleocapsid-specific sera or a nucleocapsid-specific mAb exhibited enhanced control of SARS-CoV-2. Nucleocapsid-specific antibodies elicited NK-mediated, antibody-dependent cellular cytotoxicity (ADCC) against infected cells. To our knowledge, these findings provide the first demonstration in the coronavirus literature that antibody responses specific to the nucleocapsid protein can improve viral clearance, providing a rationale for the clinical evaluation of nucleocapsid-based mAb therapies to treat COVID-19.


Asunto(s)
Anticuerpos Monoclonales , COVID-19 , Nucleocápside , Animales , Ratones , Anticuerpos Monoclonales/farmacología , Anticuerpos Antivirales , COVID-19/terapia , Vacunas contra la COVID-19 , Nucleocápside/inmunología , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/inmunología
13.
Curr Protoc Immunol ; 130(1): e99, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32940427

RESUMEN

In this invited article, we explain technical aspects of the lymphocytic choriomeningitis virus (LCMV) system, providing an update of a prior contribution by Matthias von Herrath and J. Lindsay Whitton. We provide an explanation of the LCMV infection models, highlighting the importance of selecting an appropriate route and viral strain. We also describe how to quantify virus-specific immune responses, followed by an explanation of useful transgenic systems. Specifically, our article will focus on the following protocols. © 2020 Wiley Periodicals LLC. Basic Protocol 1: LCMV infection routes in mice Support Protocol 1: Preparation of LCMV stocks ASSAYS TO MEASURE LCMV TITERS Support Protocol 2: Plaque assay Support Protocol 3: Immunofluorescence focus assay (IFA) to measure LCMV titer MEASUREMENT OF T CELL AND B CELL RESPONSES TO LCMV INFECTION Basic Protocol 2: Triple tetramer staining for detection of LCMV-specific CD8 T cells Basic Protocol 3: Intracellular cytokine staining (ICS) for detection of LCMV-specific T cells Basic Protocol 4: Enumeration of direct ex vivo LCMV-specific antibody-secreting cells (ASC) Basic Protocol 5: Limiting dilution assay (LDA) for detection of LCMV-specific memory B cells Basic Protocol 6: ELISA for quantification of LCMV-specific IgG antibody Support Protocol 4: Preparation of splenic lymphocytes Support Protocol 5: Making BHK21-LCMV lysate Basic Protocol 7: Challenge models TRANSGENIC MODELS Basic Protocol 8: Transfer of P14 cells to interrogate the role of IFN-I on CD8 T cell responses Basic Protocol 9: Comparing the expansion of naïve versus memory CD4 T cells following chronic viral challenge.


Asunto(s)
Inmunidad Adaptativa , Interacciones Huésped-Patógeno/inmunología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Animales , Anticuerpos Antivirales/inmunología , Especificidad de Anticuerpos/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Técnicas de Cultivo de Célula , Línea Celular , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática/métodos , Técnica del Anticuerpo Fluorescente/métodos , Memoria Inmunológica , Depleción Linfocítica , Coriomeningitis Linfocítica/transmisión , Ratones , Especificidad del Receptor de Antígeno de Linfocitos T , Linfocitos T/inmunología , Linfocitos T/metabolismo , Carga Viral/métodos , Ensayo de Placa Viral/métodos
14.
J Exp Med ; 217(12)2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-32820330

RESUMEN

Type I interferons (IFN-I) are a major antiviral defense and are critical for the activation of the adaptive immune system. However, early viral clearance by IFN-I could limit antigen availability, which could in turn impinge upon the priming of the adaptive immune system. In this study, we hypothesized that transient IFN-I blockade could increase antigen presentation after acute viral infection. To test this hypothesis, we infected mice with viruses coadministered with a single dose of IFN-I receptor-blocking antibody to induce a short-term blockade of the IFN-I pathway. This resulted in a transient "spike" in antigen levels, followed by rapid antigen clearance. Interestingly, short-term IFN-I blockade after coronavirus, flavivirus, rhabdovirus, or arenavirus infection induced a long-lasting enhancement of immunological memory that conferred improved protection upon subsequent reinfections. Short-term IFN-I blockade also improved the efficacy of viral vaccines. These findings demonstrate a novel mechanism by which IFN-I regulate immunological memory and provide insights for rational vaccine design.


Asunto(s)
Inmunogenicidad Vacunal/inmunología , Interferón Tipo I/antagonistas & inhibidores , Interferón-alfa/inmunología , Receptor de Interferón alfa y beta/inmunología , Vacunas Virales/inmunología , Infección por el Virus Zika/inmunología , Virus Zika/inmunología , Animales , Anticuerpos Bloqueadores/inmunología , Anticuerpos Bloqueadores/farmacología , Anticuerpos Antivirales/inmunología , Presentación de Antígeno/inmunología , Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Expresión Génica/inmunología , Células HEK293 , Humanos , Memoria Inmunológica , Interferón-alfa/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Interferón alfa y beta/genética , Transfección , Infección por el Virus Zika/virología
15.
Sci Transl Med ; 11(511)2019 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-31554738

RESUMEN

Adult stem and progenitor cells are uniquely capable of self-renewal, and targeting this process represents a potential therapeutic opportunity. The early erythroid progenitor, burst-forming unit erythroid (BFU-E), has substantial self-renewal potential and serves as a key cell type for the treatment of anemias. However, our understanding of mechanisms underlying BFU-E self-renewal is extremely limited. Here, we found that the muscarinic acetylcholine receptor, cholinergic receptor, muscarinic 4 (CHRM4), pathway regulates BFU-E self-renewal and that pharmacological inhibition of CHRM4 corrects anemias of myelodysplastic syndrome (MDS), aging, and hemolysis. Genetic down-regulation of CHRM4 or pharmacologic inhibition of CHRM4 using the selective antagonist PD102807 promoted BFU-E self-renewal, whereas deletion of Chrm4 increased erythroid cell production under stress conditions in vivo. Moreover, muscarinic acetylcholine receptor antagonists corrected anemias in mouse models of MDS, aging, and hemolysis in vivo, extending the survival of mice with MDS relative to that of controls. The effects of muscarinic receptor antagonism on promoting expansion of BFU-Es were mediated by cyclic AMP induction of the transcription factor CREB, whose targets up-regulated key regulators of BFU-E self-renewal. On the basis of these data, we propose a model of hematopoietic progenitor self-renewal through a cholinergic-mediated "hematopoietic reflex" and identify muscarinic acetylcholine receptor antagonists as potential therapies for anemias associated with MDS, aging, and hemolysis.


Asunto(s)
Autorrenovación de las Células , Células Eritroides/citología , Células Eritroides/metabolismo , Receptores Muscarínicos/metabolismo , Células Madre/citología , Anemia/tratamiento farmacológico , Animales , Autorrenovación de las Células/efectos de los fármacos , Autorrenovación de las Células/genética , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Células Eritroides/efectos de los fármacos , Células Precursoras Eritroides , Eritropoyesis/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Antagonistas Muscarínicos/farmacología , Antagonistas Muscarínicos/uso terapéutico , Células Madre/efectos de los fármacos , Células Madre/metabolismo
16.
Oncogene ; 37(34): 4692-4710, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29755131

RESUMEN

Estrogen receptor alpha (ERα) is a ligand-activated nuclear receptor that directs proliferation and differentiation in selected cancer cell types including mammary-derived carcinomas. These master-regulatory functions of ERα require trans-acting elements such as the pioneer factor FOXA1 to establish a genomic landscape conducive to ERα control. Here, we identify the H3K4 methyltransferase KMT2C as necessary for hormone-driven ERα activity and breast cancer proliferation. KMT2C knockdown suppresses estrogen-dependent gene expression and causes H3K4me1 and H3K27ac loss selectively at ERα enhancers. Correspondingly, KMT2C loss impairs estrogen-driven breast cancer proliferation but has no effect on ER- breast cells. Whereas KMT2C loss disrupts estrogen-driven proliferation, it conversely promotes tumor outgrowth under hormone-depleted conditions. In accordance, KMT2C is one of the most frequently mutated genes in ER-positive breast cancer with KMT2C deletion correlating with significantly shorter progression-free survival on anti-estrogen therapy. From a therapeutic standpoint, KMT2C-depleted cells that develop hormone-independence retain their dependence on ERα, displaying ongoing sensitivity to ERα antagonists. We conclude that KMT2C is a key regulator of ERα activity whose loss uncouples breast cancer proliferation from hormone abundance.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas de Unión al ADN/metabolismo , Receptor alfa de Estrógeno/metabolismo , Estrógenos/metabolismo , Proteínas de Neoplasias/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Proliferación Celular/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Células HEK293 , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Supervivencia sin Progresión , Transducción de Señal/fisiología
17.
J Clin Invest ; 128(9): 3819-3825, 2018 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-29920189

RESUMEN

Rearrangements involving the neurotrophic receptor kinase genes (NTRK1, NTRK2, and NTRK3; hereafter referred to as TRK) produce oncogenic fusions in a wide variety of cancers in adults and children. Although TRK fusions occur in fewer than 1% of all solid tumors, inhibition of TRK results in profound therapeutic responses, resulting in Breakthrough Therapy FDA approval of the TRK inhibitor larotrectinib for adult and pediatric patients with solid tumors, regardless of histology. In contrast to solid tumors, the frequency of TRK fusions and the clinical effects of targeting TRK in hematologic malignancies are unknown. Here, through an evaluation for TRK fusions across more than 7,000 patients with hematologic malignancies, we identified TRK fusions in acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), histiocytosis, multiple myeloma, and dendritic cell neoplasms. Although TRK fusions occurred in only 0.1% of patients (8 of 7,311 patients), they conferred responsiveness to TRK inhibition in vitro and in vivo in a patient-derived xenograft and a corresponding AML patient with ETV6-NTRK2 fusion. These data identify that despite their individual rarity, collectively, TRK fusions are present in a wide variety of hematologic malignancies and predict clinically significant therapeutic responses to TRK inhibition.


Asunto(s)
Neoplasias Hematológicas/genética , Fusión de Oncogenes , Proteínas Tirosina Quinasas Receptoras/genética , Adulto , Anciano , Animales , Niño , Femenino , Neoplasias Hematológicas/tratamiento farmacológico , Neoplasias Hematológicas/enzimología , Humanos , Lactante , Masculino , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/genética , Ratones , Persona de Mediana Edad , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteínas de Fusión Oncogénica/genética , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-ets/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-ets/genética , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Receptor trkA/genética , Receptor trkB/antagonistas & inhibidores , Receptor trkB/genética , Receptor trkC/genética , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto Joven , Proteína ETS de Variante de Translocación 6
18.
Cancer Cell ; 34(2): 225-241.e8, 2018 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-30107174

RESUMEN

Mutations affecting RNA splicing factors are the most common genetic alterations in myelodysplastic syndrome (MDS) patients and occur in a mutually exclusive manner. The basis for the mutual exclusivity of these mutations and how they contribute to MDS is not well understood. Here we report that although different spliceosome gene mutations impart distinct effects on splicing, they are negatively selected for when co-expressed due to aberrant splicing and downregulation of regulators of hematopoietic stem cell survival and quiescence. In addition to this synthetic lethal interaction, mutations in the splicing factors SF3B1 and SRSF2 share convergent effects on aberrant splicing of mRNAs that promote nuclear factor κB signaling. These data identify shared consequences of splicing-factor mutations and the basis for their mutual exclusivity.


Asunto(s)
Mutación , Neoplasias/genética , Empalmosomas , Animales , Caspasa 8/genética , Femenino , Hematopoyesis , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/fisiología , Fosfoproteínas/genética , Factores de Empalme de ARN/genética , Factores de Empalme Serina-Arginina/genética
19.
Nat Commun ; 8: 15429, 2017 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-28516957

RESUMEN

Additional sex combs-like (ASXL) proteins are mammalian homologues of additional sex combs (Asx), a regulator of trithorax and polycomb function in Drosophila. While there has been great interest in ASXL1 due to its frequent mutation in leukemia, little is known about its paralog ASXL2, which is frequently mutated in acute myeloid leukemia patients bearing the RUNX1-RUNX1T1 (AML1-ETO) fusion. Here we report that ASXL2 is required for normal haematopoiesis with distinct, non-overlapping effects from ASXL1 and acts as a haploinsufficient tumour suppressor. While Asxl2 was required for normal haematopoietic stem cell self-renewal, Asxl2 loss promoted AML1-ETO leukemogenesis. Moreover, ASXL2 target genes strongly overlapped with those of RUNX1 and AML1-ETO and ASXL2 loss was associated with increased chromatin accessibility at putative enhancers of key leukemogenic loci. These data reveal that Asxl2 is a critical regulator of haematopoiesis and mediates transcriptional effects that promote leukemogenesis driven by AML1-ETO.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Regulación Leucémica de la Expresión Génica , Leucemia Mieloide Aguda/genética , Proteínas de Fusión Oncogénica/genética , Proteínas Represoras/genética , Animales , Trasplante de Médula Ósea , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Modelos Animales de Enfermedad , Haploinsuficiencia , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Integrasas/genética , Integrasas/metabolismo , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Ratones Noqueados , Proteínas de Resistencia a Mixovirus/genética , Proteínas de Resistencia a Mixovirus/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Represoras/deficiencia , Transducción de Señal , Análisis de Supervivencia
20.
Cell Metab ; 25(2): 358-373, 2017 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-28089569

RESUMEN

Lifestyle factors, including diet, play an important role in the survival of cancer patients. However, the molecular mechanisms underlying pathogenic links between diet and particular oncogenic mutations in human cancers remain unclear. We recently reported that the ketone body acetoacetate selectively enhances BRAF V600E mutant-dependent MEK1 activation in human cancers. Here we show that a high-fat ketogenic diet increased serum levels of acetoacetate, leading to enhanced tumor growth potential of BRAF V600E-expressing human melanoma cells in xenograft mice. Treatment with hypolipidemic agents to lower circulating acetoacetate levels or an inhibitory homolog of acetoacetate, dehydroacetic acid, to antagonize acetoacetate-BRAF V600E binding attenuated BRAF V600E tumor growth. These findings reveal a signaling basis underlying a pathogenic role of dietary fat in BRAF V600E-expressing melanoma, providing insights into the design of conceptualized "precision diets" that may prevent or delay tumor progression based on an individual's specific oncogenic mutation profile.


Asunto(s)
Grasas de la Dieta/efectos adversos , Cuerpos Cetónicos/metabolismo , Melanoma/patología , Mutación/genética , Proteínas Proto-Oncogénicas B-raf/genética , Ácido 3-Hidroxibutírico/farmacología , Acetoacetatos/administración & dosificación , Acetoacetatos/sangre , Acetoacetatos/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Hipolipemiantes/farmacología , Inyecciones Intraperitoneales , Melanoma/sangre , Ratones , Ratones Desnudos , Pironas/química , Pironas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA