Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Cancer Res Commun ; 3(1): 130-139, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36968223

RESUMO

Purpose: The treatment of glioblastoma (GBM) poses challenges. The use of immune checkpoint inhibition (ICI) has been disappointing as GBM is characterized by low mutational burden and low T-cell infiltration. The combination of ICI with other treatment modalities may improve efficacy. Patient and Methods: Patients with recurrent GBM were treated with avelumab, a human IgG1 antibody directed against PD-L1 (part A), or avelumab within a week after laser interstitial thermal therapy (LITT) and continuation of avelumab (part B). Bevacizumab was allowed to be combined with ICI to spare steroid use. The primary objective was to characterize the tolerability and safety of the regimens. The secondary objectives included overall survival, progression-free survival (PFS), signatures of plasma analytes, and immune cells. Results: A total of 12 patients (median age 64; range, 37-73) enrolled, five in part A and seven in part B. Two serious adverse events occurred in the same patient, LITT treated, not leading to death. The median survival from enrollment was 13 months [95% confidence interval (CI), 4-16 months] with no differences for part A or B. The median PFS was 3 months (95% CI, 1.5-4.5 months). The decrease in MICA/MICB, γδT cells, and CD4+ T cell EMRA correlated with prolonged survival. Conclusions: Avelumab was generally well tolerated. Adding bevacizumab to ICI may be beneficial by lowering cytokine and immune cell expression. The development of this combinatorial treatment warrants further investigation. Exploring the modulation of adaptive and innate immune cells and plasma analytes as biomarker signatures may instruct future studies in this dismal refractory disease. Significance: Our phase I of PD-L1 inhibition combined with LITT and using bevacizumab to spare steroids had a good safety profile for recurrent GBM. Developing combinatory treatment may help outcomes. In addition, we found significant immune modulation of cytokines and immune cells by bevacizumab, which may enhance the effect of ICI.


Assuntos
Glioblastoma , Humanos , Pessoa de Meia-Idade , Bevacizumab/efeitos adversos , Glioblastoma/tratamento farmacológico , Anticorpos Monoclonais , Fator A de Crescimento do Endotélio Vascular , Antígeno B7-H1
2.
Gut ; 72(4): 654-662, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36191961

RESUMO

OBJECTIVE: Loss-of-function mutations in genes generating reactive oxygen species (ROS), such as NOX1, are associated with IBD. Mechanisms whereby loss of ROS drive IBD are incompletely defined. DESIGN: ROS measurements and single-cell transcriptomics were performed on colonoids stratified by NOX1 genotype and TNFα stimulation. Clustering of epithelial cells from human UC (inflamed and uninflamed) scRNASeq was performed. Validation of M cell induction was performed by immunohistochemistry using UEA1 (ulex europaeus agglutin-1 lectin) and in vivo with DSS injury. RESULTS: TNFα induces ROS production more in NOX1-WT versus NOX1-deficient murine colonoids under a range of Wnt-mediated and Notch-mediated conditions. scRNASeq from inflamed and uninflamed human colitis versus TNFα stimulated, in vitro colonoids defines substantially shared, induced transcription factors; NOX1-deficient colonoids express substantially lower levels of STAT3 (signal transducer and activator of transcription 3), CEBPD (CCAAT enhancer-binding protein delta), DNMT1 (DNA methyltransferase) and HIF1A (hypoxia-inducible factor) baseline. Subclustering unexpectedly showed marked TNFα-mediated induction of M cells (sentinel cells overlying lymphoid aggregates) in NOX1-deficient colonoids. M cell induction by UEA1 staining is rescued with H2O2 and paraquat, defining extra- and intracellular ROS roles in maintenance of LGR5+ stem cells. DSS injury demonstrated GP2 (glycoprotein-2), basal lymphoplasmacytosis and UEA1 induction in NOX1-deficiency. Principal components analyses of M cell genes and decreased DNMT1 RNA velocity correlate with UC inflammation. CONCLUSIONS: NOX1 deficiency plus TNFα stimulation contribute to colitis through dysregulation of the stem cell niche and altered cell differentiation, enhancing basal lymphoplasmacytosis. Our findings prioritise ROS modulation for future therapies.


Assuntos
Colite , Doenças Inflamatórias Intestinais , Camundongos , Humanos , Animais , Espécies Reativas de Oxigênio/metabolismo , Fator de Necrose Tumoral alfa/efeitos adversos , Células M , NADPH Oxidase 1/genética , NADPH Oxidase 1/metabolismo , Peróxido de Hidrogênio/efeitos adversos , Colite/induzido quimicamente
3.
Cancer Cell ; 40(9): 1027-1043.e9, 2022 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-36099881

RESUMO

Programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1)-blockade immunotherapies have limited efficacy in the treatment of bladder cancer. Here, we show that NKG2A associates with improved survival and responsiveness to PD-L1 blockade immunotherapy in bladder tumors that have high abundance of CD8+ T cells. In bladder tumors, NKG2A is acquired on CD8+ T cells later than PD-1 as well as other well-established immune checkpoints. NKG2A+ PD-1+ CD8+ T cells diverge from classically defined exhausted T cells through their ability to react to human leukocyte antigen (HLA) class I-deficient tumors using T cell receptor (TCR)-independent innate-like mechanisms. HLA-ABC expression by bladder tumors is progressively diminished as disease progresses, framing the importance of targeting TCR-independent anti-tumor functions. Notably, NKG2A+ CD8+ T cells are inhibited when HLA-E is expressed by tumors and partly restored upon NKG2A blockade in an HLA-E-dependent manner. Overall, our study provides a framework for subsequent clinical trials combining NKG2A blockade with other T cell-targeted immunotherapies, where tumors express higher levels of HLA-E.


Assuntos
Subfamília C de Receptores Semelhantes a Lectina de Células NK/metabolismo , Neoplasias da Bexiga Urinária , Antígeno B7-H1/metabolismo , Linfócitos T CD8-Positivos , Antígenos de Histocompatibilidade Classe I , Humanos , Receptor de Morte Celular Programada 1 , Neoplasias da Bexiga Urinária/terapia
4.
Gastroenterology ; 160(5): 1709-1724, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33421512

RESUMO

BACKGROUND & AIMS: Recent literature has implicated a key role for mast cells in murine models of colonic inflammation, but their role in human ulcerative colitis (UC) is not well established. A major advance has been the identification of mrgprb2 (human orthologue, MRGPX2) as mediating IgE-independent mast cell activation. We sought to define mechanisms of mast cell activation and MRGPRX2 in human UC. METHODS: Colon tissues were collected from patients with UC for bulk RNA sequencing and lamina propria cells were isolated for MRGPRX2 activation studies and single-cell RNA sequencing. Genetic association of all protein-altering G-protein coupled receptor single-nucleotide polymorphism was performed in an Ashkenazi Jewish UC case-control cohort. Variants of MRGPRX2 were transfected into Chinese hamster ovary (CHO) and human mast cell (HMC) 1.1 cells to detect genotype-dependent effects on ß-arrestin recruitment, IP-1 accumulation, and phosphorylated extracellular signal-regulated kinase. RESULTS: Mast cell-specific mediators and adrenomedullin (proteolytic precursor of PAMP-12, an MRGPRX2 agonist) are up-regulated in inflamed compared to uninflamed UC. MRGPRX2 stimulation induces carboxypeptidase secretion from inflamed UC. Of all protein-altering GPCR alleles, a unique variant of MRGPRX2, Asn62Ser, was most associated with and was bioinformatically predicted to alter arrestin recruitment. We validated that the UC protective serine allele enhances ß-arrestin recruitment, decreases IP-1, and increases phosphorylated extracellular signal-regulated kinase with MRGPRX2 agonists. Single-cell RNA sequencing defines that adrenomedullin is expressed by activated fibroblasts and epithelial cells and that interferon gamma is a key upstream regulator of mast cell gene expression. CONCLUSION: Inflamed UC regions are distinguished by MRGPRX2-mediated activation of mast cells, with decreased activation observed with a UC-protective genetic variant. These results define cell modules of UC activation and a new therapeutic target.


Assuntos
Degranulação Celular , Colite Ulcerativa/metabolismo , Colo/metabolismo , Mastócitos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropeptídeos/metabolismo , Adrenomedulina/genética , Adrenomedulina/metabolismo , Animais , Células CHO , Estudos de Casos e Controles , Colite Ulcerativa/genética , Colite Ulcerativa/imunologia , Colo/imunologia , Cricetulus , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Variação Genética , Humanos , Fosfatos de Inositol/metabolismo , Ligantes , Mastócitos/imunologia , Proteínas do Tecido Nervoso/genética , Fosforilação , Receptores Acoplados a Proteínas G/genética , Receptores de Neuropeptídeos/genética , beta-Arrestina 2/genética , beta-Arrestina 2/metabolismo
5.
Dis Model Mech ; 12(8)2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31337664

RESUMO

Genome-wide association studies have identified over 200 genomic loci associated with inflammatory bowel disease (IBD). High-effect risk alleles define key roles for genes involved in bacterial response and innate defense. More high-throughput in vivo systems are required to rapidly evaluate therapeutic agents. We visualize, in zebrafish, the effects on epithelial barrier function and intestinal autophagy of one-course and repetitive injury. Repetitive injury induces increased mortality, impaired recovery of intestinal barrier function, failure to contain bacteria within the intestine and impaired autophagy. Prostaglandin E2 (PGE2) administration protected against injury by enhancing epithelial barrier function and limiting systemic infection. Effects of IBD therapeutic agents were defined: mesalamine showed protective features during injury, whereas 6-mercaptopurine displayed marked induction of autophagy during recovery. Given the highly conserved nature of innate defense in zebrafish, it represents an ideal model system with which to test established and new IBD therapies targeted to the epithelial barrier.This article has an associated First Person interview with the first author of the paper.


Assuntos
Epitélio/patologia , Doenças Inflamatórias Intestinais/microbiologia , Doenças Inflamatórias Intestinais/patologia , Intestinos/lesões , Peixe-Zebra/fisiologia , Ácidos/metabolismo , Animais , Autofagia , Proteínas de Bactérias/metabolismo , Sulfato de Dextrana , Dinoprostona/metabolismo , Modelos Animais de Doenças , Enterócitos/metabolismo , Humanos , Doenças Inflamatórias Intestinais/tratamento farmacológico , Intestinos/patologia , Lisossomos/metabolismo , Modelos Biológicos , Mucinas/metabolismo , Muco/metabolismo
6.
J Clin Invest ; 129(2): 786-801, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30480549

RESUMO

Tumor cure with conventional fractionated radiotherapy is 65%, dependent on tumor cell-autonomous gradual buildup of DNA double-strand break (DSB) misrepair. Here we report that single-dose radiotherapy (SDRT), a disruptive technique that ablates more than 90% of human cancers, operates a distinct dual-target mechanism, linking acid sphingomyelinase-mediated (ASMase-mediated) microvascular perfusion defects to DNA unrepair in tumor cells to confer tumor cell lethality. ASMase-mediated microcirculatory vasoconstriction after SDRT conferred an ischemic stress response within parenchymal tumor cells, with ROS triggering the evolutionarily conserved SUMO stress response, specifically depleting chromatin-associated free SUMO3. Whereas SUMO3, but not SUMO2, was indispensable for homology-directed repair (HDR) of DSBs, HDR loss of function after SDRT yielded DSB unrepair, chromosomal aberrations, and tumor clonogen demise. Vasoconstriction blockade with the endothelin-1 inhibitor BQ-123, or ROS scavenging after SDRT using peroxiredoxin-6 overexpression or the SOD mimetic tempol, prevented chromatin SUMO3 depletion, HDR loss of function, and SDRT tumor ablation. We also provide evidence of mouse-to-human translation of this biology in a randomized clinical trial, showing that 24 Gy SDRT, but not 3×9 Gy fractionation, coupled early tumor ischemia/reperfusion to human cancer ablation. The SDRT biology provides opportunities for mechanism-based selective tumor radiosensitization via accessing of SDRT/ASMase signaling, as current studies indicate that this pathway is tractable to pharmacologic intervention.


Assuntos
Recombinação Homóloga , Neoplasias , Traumatismo por Reperfusão , Transdução de Sinais , Animais , Linhagem Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Humanos , Camundongos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/radioterapia , Transdução de Sinais/genética , Transdução de Sinais/efeitos da radiação , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Ubiquitinas/genética , Ubiquitinas/metabolismo
7.
J Clin Invest ; 129(1): 215-222, 2019 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-30475228

RESUMO

Recurrent broad-scale heterozygous deletions are frequently observed in human cancer. Here we tested the hypothesis that compound haploinsufficiency of neighboring genes at chromosome 8p promotes tumorigenesis. By targeting the mouse orthologs of human DOK2 and DUSP4 genes, which were co-deleted in approximately half of human lung adenocarcinomas, we found that compound-heterozygous deletion of Dok2 and Dusp4 in mice resulted in lung tumorigenesis with short latency and high incidence, and that their co-deletion synergistically activated MAPK signaling and promoted cell proliferation. Conversely, restoration of DOK2 and DUSP4 in lung cancer cells suppressed MAPK activation and cell proliferation. Importantly, in contrast to downregulation of DOK2 or DUSP4 alone, concomitant downregulation of DOK2 and DUSP4 was associated with poor survival in human lung adenocarcinoma. Therefore, our findings lend in vivo experimental support to the notion that compound haploinsufficiency, due to broad-scale chromosome deletions, constitutes a driving force in tumorigenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Transformação Celular Neoplásica , Haploinsuficiência , Neoplasias Pulmonares , Proteínas de Neoplasias , Fosfoproteínas , Proteínas Tirosina Fosfatases , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Regulação para Baixo , Feminino , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Sistema de Sinalização das MAP Quinases/genética , Masculino , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Fosfoproteínas/biossíntese , Fosfoproteínas/genética , Proteínas Tirosina Fosfatases/biossíntese , Proteínas Tirosina Fosfatases/genética
8.
AIDS ; 32(16): 2309-2316, 2018 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-30005024

RESUMO

OBJECTIVES: The aim of this study was to determine whether biomarker P16 predicts progression risk for anal low-grade squamous intraepithelial lesions (LSILs). DESIGN: A retrospective study. METHODS: One hundred and nine HIV-infected and 18 HIV-uninfected patients with biopsy-proven anal LSIL at initial screening underwent surveillance high-resolution anoscopy and biopsy within 2 years of diagnosis. P16 immunohistochemistry was performed on index lesions and evaluated using a semi-quantitative scoring system. The association of predictors and lesional outcomes (progression, persistence or regression) was analysed using ordinal logistic regression models. A subset of p16-positive LSILs was tested for high-risk human papillomavirus (HR-HPV) DNA using real-time PCR. RESULTS: Upon follow-up, 46 (36%) LSILs progressed to high-grade squamous intraepithelial lesion (HSIL), 50 (40%) persisted as LSIL and 31 (24%) regressed to benign mucosa (median 16 months, range 5-24 months). Age, sex, race, history of condylomata, CD4 T-cell count and HIV plasma viral load were similar regardless of clinical outcome. P16 immunoreactivity of index lesion was classified as block-positive (n = 36), focal-positive (n = 49) or negative (n = 42). Sixty-four percent of block-positive lesions progressed, as opposed to 35% of focal-positive and 14% of negative lesions (P < 0.001). HR-HPV DNA was detected in 90% of p16 block-positive lesions vs. 55% of focal-positive lesions. In unadjusted analyses, positive p16, HIV and former smoker status were significantly associated with lesional persistence and progression. P16 remained the only significant predictor in an adjusted model. CONCLUSION: Biomarker p16 is the strongest predictor for anal LSIL-to-HSIL progression, outperforming other risk factors. To enhance the overall effectiveness of surveillance, we propose using p16 immunohistochemistry to help stratify patients at high vs. low risk of progression.


Assuntos
Neoplasias do Ânus/diagnóstico , Biomarcadores/análise , Inibidor p16 de Quinase Dependente de Ciclina/análise , Lesões Intraepiteliais Escamosas Cervicais/diagnóstico , Adulto , Idoso , Biópsia , Progressão da Doença , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Adulto Jovem
9.
Clin Cancer Res ; 24(20): 4937-4948, 2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-29950349

RESUMO

Purpose: Polyinosinic-polycytidylic acid-poly-l-lysine carboxymethylcellulose (poly-ICLC), a synthetic double-stranded RNA complex, is a ligand for toll-like receptor-3 and MDA-5 that can activate immune cells, such as dendritic cells, and trigger natural killer cells to kill tumor cells.Patients and Methods: In this pilot study, eligible patients included those with recurrent metastatic disease in whom prior systemic therapy (head and neck squamous cell cancer and melanoma) failed. Patients received 2 treatment cycles, each cycle consisting of 1 mg poly-ICLC 3× weekly intratumorally (IT) for 2 weeks followed by intramuscular (IM) boosters biweekly for 7 weeks, with a 1-week rest period. Immune response was evaluated by immunohistochemistry (IHC) and RNA sequencing (RNA-seq) in tumor and blood.Results: Two patients completed 2 cycles of IT treatments, and 1 achieved clinical benefit (stable disease, progression-free survival 6 months), whereas the remainder had progressive disease. Poly-ICLC was well tolerated, with principal side effects of fatigue and inflammation at injection site (

Assuntos
Carboximetilcelulose Sódica/análogos & derivados , Fatores Imunológicos/administração & dosagem , Imunomodulação/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Poli I-C/administração & dosagem , Polilisina/análogos & derivados , Idoso , Biópsia , Carboximetilcelulose Sódica/administração & dosagem , Carboximetilcelulose Sódica/efeitos adversos , Esquema de Medicação , Feminino , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Imuno-Histoquímica , Fatores Imunológicos/efeitos adversos , Injeções Intralesionais , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Neoplasias/diagnóstico , Neoplasias/mortalidade , Projetos Piloto , Poli I-C/efeitos adversos , Polilisina/administração & dosagem , Polilisina/efeitos adversos , Prognóstico , Tomografia Computadorizada por Raios X , Resultado do Tratamento
10.
Methods Mol Biol ; 1388: 23-37, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27033068

RESUMO

Immunohistochemistry (IHC) is an excellent technique used routinely to define the phenotype in pathology laboratories through the analysis of molecular expression in cells and tissues. The PTEN protein is ubiquitously expressed in the majority of human tissues, and allelic or complete loss of PTEN is frequently observed in different types of malignancies leading to an activation of the AKT/mTOR pathways. IHC-based analyses are best to determine the level of PTEN expression in histological samples, but not to assess partial or heterozygous deletions, for which FISH analyses are more appropriate. Interpretation of the IHC results is the most critical point in the assessment of PTEN expression, since it is used both as a prognostic factor and as a tool to guide therapeutic intervention and response to therapy. Importantly, analyses of well-known downstream markers, such as AKT or mTOR, may be used to further analyze PTEN functional status.


Assuntos
Imunofluorescência/métodos , PTEN Fosfo-Hidrolase/análise , Animais , Técnicas Citológicas , Humanos
11.
Nat Neurosci ; 17(1): 121-30, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24270187

RESUMO

Using the Illumina 450K array and a stringent statistical analysis with age and gender correction, we report genome-wide differences in DNA methylation between pathology-free regions derived from human multiple sclerosis-affected and control brains. Differences were subtle, but widespread and reproducible in an independent validation cohort. The transcriptional consequences of differential DNA methylation were further defined by genome-wide RNA-sequencing analysis and validated in two independent cohorts. Genes regulating oligodendrocyte survival, such as BCL2L2 and NDRG1, were hypermethylated and expressed at lower levels in multiple sclerosis-affected brains than in controls, while genes related to proteolytic processing (for example, LGMN, CTSZ) were hypomethylated and expressed at higher levels. These results were not due to differences in cellular composition between multiple sclerosis and controls. Thus, epigenomic changes in genes affecting oligodendrocyte susceptibility to damage are detected in pathology-free areas of multiple sclerosis-affected brains.


Assuntos
Encéfalo/metabolismo , Epigênese Genética , Esclerose Múltipla/patologia , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Encéfalo/patologia , Proteínas de Ciclo Celular/metabolismo , Estudos de Coortes , Ilhas de CpG/genética , Metilação de DNA/genética , Epigenômica/métodos , Ontologia Genética , Antígenos HLA-DR/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fator de Transcrição 2 de Oligodendrócitos , Oligodendroglia/patologia
12.
Gastroenterology ; 143(5): 1266-1276, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22841781

RESUMO

BACKGROUND & AIMS: Adult stem cells have been proposed to be quiescent and radiation resistant, repairing DNA double-strand breaks by nonhomologous end joining. However, the population of putative small intestinal stem cells (ISCs) at position +4 from the crypt base contradicts this model, in that they are highly radiosensitive. Cycling crypt base columnar cells (CBCs) at crypt positions +1-3 recently were defined as an alternative population of ISCs. Little is known about the sensitivity of this stem cell population to radiation. METHODS: Radiation-induced lethality of CBCs was quantified kinetically in Lgr5-lacZ transgenic mice. γ-H2AX, BRCA1, RAD51, and DNA-PKcs foci were used as DNA repair surrogates to investigate the inherent ability of CBCs to recognize and repair double-strand breaks. 5-ethynyl-2'-deoxyuridine and 5-bromo-2'-deoxyuridine incorporation assays were used to study patterns of CBC growth arrest and re-initiation of cell cycling. Apoptosis was evaluated by caspase-3 staining. RESULTS: CBCs are relatively radioresistant, repairing DNA by homologous recombination significantly more efficiently than transit amplifying progenitors or villus cells. CBCs undergo apoptosis less than 24 hours after irradiation (32% ± 2% of total lethality) or mitotic death at 24-48 hours. Survival of CBCs at 2 days predicts crypt regeneration at 3.5 days and lethality from gastrointestinal syndrome. Crypt repopulation originates from CBCs that survive irradiation. CONCLUSIONS: Adult ISCs in mice can cycle rapidly yet still be radioresistant. Importantly, homologous recombination can protect adult stem cell populations from genotoxic stress. These findings broaden and refine concepts of the phenotype of adult stem cells.


Assuntos
Células-Tronco Adultas/efeitos da radiação , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA , Jejuno/efeitos da radiação , Tolerância a Radiação , Animais , Apoptose/efeitos da radiação , Medula Óssea/efeitos da radiação , Pontos de Checagem do Ciclo Celular/efeitos da radiação , Jejuno/patologia , Camundongos , Camundongos Transgênicos
13.
Radiat Res ; 177(4): 467-82, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22348249

RESUMO

Prediction of response to therapy has been identified as an important tool to obtain a more customized treatment. It allows the selection of those patients who will benefit most from a particular therapy and prevents the exposure of patients to toxic, noneffective regimens. Recent technical advances and the introduction of novel markers in anatomical and functional imaging have created exciting opportunities for in vivo visualization and quantification of cell death. This review will focus on in vivo apoptosis imaging as a predictive marker for tumor response after radiation.


Assuntos
Apoptose/efeitos da radiação , Imagem Molecular/métodos , Neoplasias/radioterapia , Animais , Anexina A5 , Apoptose/fisiologia , Caspase 3/fisiologia , Quimiorradioterapia , Ensaios Clínicos como Assunto , Meios de Contraste , Imagem de Difusão por Ressonância Magnética , Fluoresceína-5-Isotiocianato/análogos & derivados , Corantes Fluorescentes , Humanos , Camundongos , Microbolhas , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Compostos de Organotecnécio , Tomografia por Emissão de Pósitrons/métodos , Lesões por Radiação/etiologia , Lesões por Radiação/patologia , Compostos Radiofarmacêuticos , Radioterapia/efeitos adversos , Dosagem Radioterapêutica , Receptores de Morte Celular/fisiologia , Tecnécio Tc 99m Sestamibi , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Resultado do Tratamento , Ultrassonografia/métodos
14.
J Clin Invest ; 121(12): 4700-11, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22105174

RESUMO

Advanced human thyroid cancers, particularly those that are refractory to treatment with radioiodine (RAI), have a high prevalence of BRAF (v-raf murine sarcoma viral oncogene homolog B1) mutations. However, the degree to which these cancers are dependent on BRAF expression is still unclear. To address this question, we generated mice expressing one of the most commonly detected BRAF mutations in human papillary thyroid carcinomas (BRAF(V600E)) in thyroid follicular cells in a doxycycline-inducible (dox-inducible) manner. Upon dox induction of BRAF(V600E), the mice developed highly penetrant and poorly differentiated thyroid tumors. Discontinuation of dox extinguished BRAF(V600E) expression and reestablished thyroid follicular architecture and normal thyroid histology. Switching on BRAF(V600E) rapidly induced hypothyroidism and virtually abolished thyroid-specific gene expression and RAI incorporation, all of which were restored to near basal levels upon discontinuation of dox. Treatment of mice with these cancers with small molecule inhibitors of either MEK or mutant BRAF reduced their proliferative index and partially restored thyroid-specific gene expression. Strikingly, treatment with the MAPK pathway inhibitors rendered the tumor cells susceptible to a therapeutic dose of RAI. Our data show that thyroid tumors carrying BRAF(V600E) mutations are exquisitely dependent on the oncoprotein for viability and that genetic or pharmacological inhibition of its expression or activity is associated with tumor regression and restoration of RAI uptake in vivo in mice. These findings have potentially significant clinical ramifications.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma Papilar/tratamento farmacológico , Indóis/uso terapêutico , Radioisótopos do Iodo/farmacocinética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas de Neoplasias/fisiologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas B-raf/fisiologia , Sulfonamidas/uso terapêutico , Neoplasias da Glândula Tireoide/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidade , Apoptose/efeitos dos fármacos , Benzamidas/administração & dosagem , Benzamidas/farmacologia , Benzamidas/uso terapêutico , Benzamidas/toxicidade , Carcinoma Papilar/genética , Carcinoma Papilar/metabolismo , Carcinoma Papilar/patologia , Dano ao DNA , Difenilamina/administração & dosagem , Difenilamina/análogos & derivados , Difenilamina/farmacologia , Difenilamina/uso terapêutico , Difenilamina/toxicidade , Doxorrubicina/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Ativação Enzimática/efeitos dos fármacos , Genes Sintéticos/efeitos dos fármacos , Humanos , Indóis/administração & dosagem , Indóis/farmacologia , Indóis/toxicidade , MAP Quinase Quinase 1/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Camundongos Transgênicos , Mutação de Sentido Incorreto , Proteínas de Neoplasias/genética , Mutação Puntual , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/toxicidade , Proteínas Proto-Oncogênicas B-raf/genética , Sulfonamidas/administração & dosagem , Sulfonamidas/farmacologia , Sulfonamidas/toxicidade , Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/patologia , Vemurafenib
15.
Cancer Res ; 70(20): 8179-86, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20924105

RESUMO

Severe combined immunodeficient (SCID) mice carry a germ-line mutation in DNA-PK, associated with deficiency in recognition and repair DNA double-strand breaks. Thus, SCID cells and tissues display increased sensitivity to radiation-induced postmitotic (clonogenic) cell death. Nonetheless, the single-radiation doses required for 50% permanent local control (TCD(50)) of tumors implanted in SCID mice are not significantly different from the TCD(50) values of the same tumors in wild-type hosts. Whereas the tumor stroma is derived from the host, the observation that tumors implanted in SCID mice do not exhibit hypersensitivity to radiation might imply that stromal endothelial elements do not contribute substantially to tumor cure by ionizing radiation. Here, we challenge this notion, testing the hypothesis that ASMase-mediated endothelial apoptosis, which results from plasma membrane alterations, not DNA damage, is a crucial element in the cure of tumors in SCID mice by single-dose radiotherapy (SDRT). We show that the endothelium in MCA/129 fibrosarcomas and B16 melanomas exhibits a wild-type apoptotic phenotype in SCID hosts, abrogated in tumors in SCID(asmase-/-) littermates, which also acquire resistance to SDRT. Conversion into a radioresistant tumor phenotype when implanted in SCID(asmase-/-) hosts provides compelling evidence that cell membrane ASMase-mediated microvascular dysfunction, rather than DNA damage-mediated endothelial clonogenic lethality, plays a mandatory role in the complex pathophysiologic mechanism of tumor cure by SDRT, and provides an explanation for the wild-type SDRT responses reported in tumors implanted in SCID mice.


Assuntos
Morte Celular/efeitos da radiação , Dano ao DNA/efeitos da radiação , Animais , Apoptose/efeitos da radiação , Divisão Celular/efeitos da radiação , Cruzamentos Genéticos , Fibrossarcoma/patologia , Marcação In Situ das Extremidades Cortadas , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Mutação , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/patologia
16.
J Cell Biol ; 177(4): 647-57, 2007 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-17502421

RESUMO

Satellite cells play a critical role in skeletal muscle regeneration in response to injury. Notch signaling is vital for satellite cell activation and myogenic precursor cell expansion but inhibits myogenic differentiation. Thus, precise spatial and temporal regulation of Notch activity is necessary for efficient muscle regeneration. We report that the basic helix-loop-helix transcription factor Stra13 modulates Notch signaling in regenerating muscle. Upon injury, Stra13(-/-) mice exhibit increased cellular proliferation, elevated Notch signaling, a striking regeneration defect characterized by degenerated myotubes, increased mononuclear cells, and fibrosis. Stra13(-/-) primary myoblasts also exhibit enhanced Notch activity, increased proliferation, and defective differentiation. Inhibition of Notch signaling ex vivo and in vivo ameliorates the phenotype of Stra13(-/-) mutants. We demonstrate in vitro that Stra13 antagonizes Notch activity and reverses the Notch-imposed inhibition of myogenesis. Thus, Stra13 plays an important role in postnatal myogenesis by attenuating Notch signaling to reduce myoblast proliferation and promote myogenic differentiation.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Proteínas de Homeodomínio/fisiologia , Receptores Notch/antagonistas & inibidores , Receptores Notch/fisiologia , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/metabolismo , Transdução de Sinais/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/genética , Linhagem Celular , Proliferação de Células , Células Cultivadas , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Transdução de Sinais/genética
17.
EMBO Rep ; 8(4): 401-7, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17347673

RESUMO

In response to a number of genotoxic stimuli that induce DNA damage in cells, the tumour suppressor p53 is activated resulting in cell cycle arrest or apoptosis. In this study, we have identified stimulated with retinoic acid 13 (Stra13), a basic helix-loop-helix transcription factor, as a regulator of ionizing-radiation-induced apoptosis. We show that Stra13 is induced in response to several DNA-damaging agents in a p53-independent manner. Stra13-/- thymocytes show impaired apoptosis in response to ionizing radiation, and consistently, p53 levels and also expression of its key transcriptional targets Puma and Noxa are reduced in the mutant thymocytes. In vitro, Stra13 regulates p53 levels in a mouse double mutant 2 (Mdm2)-dependent manner by physically interacting with p53 and preventing Mdm2-mediated ubiquitination and nuclear export. Together, our studies provide evidence that Stra13 is involved in DNA-damage-induced apoptosis and indicate its role in tumorigenesis.


Assuntos
Apoptose/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Dano ao DNA , Proteínas de Homeodomínio/fisiologia , Transporte Ativo do Núcleo Celular , Animais , Proteínas Reguladoras de Apoptose , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Núcleo Celular/química , Núcleo Celular/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Transporte Proteico , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-mdm2/análise , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Radiação Ionizante , Timo/citologia , Timo/efeitos dos fármacos , Timo/efeitos da radiação , Tretinoína/farmacologia , Proteína Supressora de Tumor p53/análise , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Regulação para Cima
18.
J Biol Chem ; 278(9): 7699-708, 2003 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-12499384

RESUMO

Estrogens, primarily 17beta-estradiol (E(2)), may play important roles in male physiology via the androgen receptor (AR). It has already been shown that E(2) modulates AR function in LNCaP prostate cancer cells and xenograft CWR22 prostate cancer tissues. Using a molecular model of E(2) bound-AR-ligand binding domain (LBD) and employing site-directed mutagenesis strategies, we screened several AR mutants that were mutated at E(2)-AR contact sites. We found a mutation at amino acid 749, AR(M749L), which confers AR hypersensitivity to E(2). The reporter assays demonstrate that E(2) can function, like androgen, to induce AR(M749L) transactivation. This E(2)-induced AR mutant transactivation is a direct effect of the AR(M749L), because the transactivation was blocked by antiandrogens. The hypersensitivity of AR(M749L) to E(2) is not due to increased affinity of AR(M749L) for E(2), rather it may be due to the existence of the proper conformation necessary to maintain E(2) binding to the AR-LBD long enough to result in E(2)-induced transactivation. AR(M749L) transactivation can be further enhanced in the presence of AR coregulators, such as ARA70 and SRC-1. Therefore, amino acid 749 may represent an important site within the AR-LBD that is involved in interaction with E(2) that, when mutated, allows E(2) induction of AR transactivation.


Assuntos
Estradiol/farmacologia , Mutação , Receptores Androgênicos/genética , Animais , Sítios de Ligação , Células COS , Dexametasona/farmacologia , Relação Dose-Resposta a Droga , Genes Reporter , Humanos , Cinética , Ligantes , Modelos Moleculares , Mutagênese Sítio-Dirigida , Reação em Cadeia da Polimerase , Progesterona/farmacologia , Ligação Proteica , Estrutura Terciária de Proteína , Transdução de Sinais , Esteroides/metabolismo , Ativação Transcricional , Tripsina/farmacologia , Células Tumorais Cultivadas
19.
J Biol Chem ; 277(39): 36499-508, 2002 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-12068007

RESUMO

The influence of estrogen on the development of the male reproductive system may be interrupted in a subset of partial androgen insensitivity syndrome (PAIS) patients. PAIS describes a wide range of male undermasculinization resulting from mutations in the androgen receptor (AR) or steroid metabolism enzymes that perturb androgen-AR regulation of male sex organ development. In this study, we are interested in determining if PAIS-derived AR mutants that respond normally to androgen have altered responses to estrogen in the presence of ARA70, a coregulator previously shown to enhance 17beta-estradiol E2-induced AR transactivation. The wild-type AR (wtAR) and two PAIS AR mutants, AR(S703G) and AR(E709K), all bind to androgen and E2 and subsequently translocate to the nucleus. Whereas ARA70 functionally interacts with the wtAR and the PAIS AR mutants in response to androgen, E2 only promotes the functional interaction between ARA70 and the wtAR but not the PAIS AR mutants. ARA70 increases E2 competitive binding to the wtAR in the presence of low level androgen and also retards E2 dissociation from the wtAR. ARA70 is present in both the cytoplasm and the nucleus of various mouse testicular cells during early embryogenesis day 16, at postpartum day 0 during estradiol synthesis and in the Leydig cells at postpartum day 49. ARA70 may be unable to modulate the PAIS AR mutants-E2 binding, diminishing the effect of E2 via AR during male reproductive system development in patients with such mutations. Therefore, the presence of ARA70 in the testosterone and E2-producing Leydig cells may enhance the overall activity of AR during critical stages of male sex organ development.


Assuntos
Estradiol/metabolismo , Mutação , Proteínas Oncogênicas , Receptores Androgênicos/metabolismo , Transativadores/genética , Fatores de Transcrição , Ativação Transcricional , Animais , Células COS , Núcleo Celular/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Células Intersticiais do Testículo/metabolismo , Ligantes , Masculino , Microscopia de Fluorescência , Coativadores de Receptor Nuclear , Plasmídeos/metabolismo , Testes de Precipitina , Ligação Proteica , Receptores Androgênicos/genética , Fatores de Tempo , Transativadores/metabolismo , Transfecção , Células Tumorais Cultivadas , Técnicas do Sistema de Duplo-Híbrido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...