Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Cell ; 175(2): 416-428.e13, 2018 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-30245014

RESUMO

The anti-cancer immune response against mutated peptides of potential immunological relevance (neoantigens) is primarily attributed to MHC-I-restricted cytotoxic CD8+ T cell responses. MHC-II-restricted CD4+ T cells also drive anti-tumor responses, but their relation to neoantigen selection and tumor evolution has not been systematically studied. Modeling the potential of an individual's MHC-II genotype to present 1,018 driver mutations in 5,942 tumors, we demonstrate that the MHC-II genotype constrains the mutational landscape during tumorigenesis in a manner complementary to MHC-I. Mutations poorly bound to MHC-II are positively selected during tumorigenesis, even more than mutations poorly bound to MHC-I. This emphasizes the importance of CD4+ T cells in anti-tumor immunity. In addition, we observed less inter-patient variation in mutation presentation for MHC-II than for MHC-I. These differences were reflected by age at diagnosis, which was correlated with presentation by MHC-I only. Collectively, our results emphasize the central role of MHC-II presentation in tumor evolution.


Assuntos
Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Neoplasias/genética , Fatores Etários , Animais , Antígenos de Neoplasias/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Evolução Molecular , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Imunoterapia/métodos , Mutação/genética
2.
Cell ; 171(6): 1272-1283.e15, 2017 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-29107334

RESUMO

MHC-I molecules expose the intracellular protein content on the cell surface, allowing T cells to detect foreign or mutated peptides. The combination of six MHC-I alleles each individual carries defines the sub-peptidome that can be effectively presented. We applied this concept to human cancer, hypothesizing that oncogenic mutations could arise in gaps in personal MHC-I presentation. To validate this hypothesis, we developed and applied a residue-centric patient presentation score to 9,176 cancer patients across 1,018 recurrent oncogenic mutations. We found that patient MHC-I genotype-based scores could predict which mutations were more likely to emerge in their tumor. Accordingly, poor presentation of a mutation across patients was correlated with higher frequency among tumors. These results support that MHC-I genotype-restricted immunoediting during tumor formation shapes the landscape of oncogenic mutations observed in clinically diagnosed tumors and paves the way for predicting personal cancer susceptibilities from knowledge of MHC-I genotype.


Assuntos
Apresentação de Antígeno , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Mutação , Neoplasias/imunologia , Linhagem Celular Tumoral , Simulação por Computador , Feminino , Células HeLa , Humanos , Masculino , Monitorização Imunológica , Proteoma
3.
Cell ; 162(4): 766-79, 2015 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-26276631

RESUMO

Compensatory proliferation triggered by hepatocyte loss is required for liver regeneration and maintenance but also promotes development of hepatocellular carcinoma (HCC). Despite extensive investigation, the cells responsible for hepatocyte restoration or HCC development remain poorly characterized. We used genetic lineage tracing to identify cells responsible for hepatocyte replenishment following chronic liver injury and queried their roles in three distinct HCC models. We found that a pre-existing population of periportal hepatocytes, located in the portal triads of healthy livers and expressing low amounts of Sox9 and other bile-duct-enriched genes, undergo extensive proliferation and replenish liver mass after chronic hepatocyte-depleting injuries. Despite their high regenerative potential, these so-called hybrid hepatocytes do not give rise to HCC in chronically injured livers and thus represent a unique way to restore tissue function and avoid tumorigenesis. This specialized set of pre-existing differentiated cells may be highly suitable for cell-based therapy of chronic hepatocyte-depleting disorders.


Assuntos
Hepatócitos/transplante , Fígado/citologia , Fígado/fisiologia , Animais , Ductos Biliares/citologia , Proliferação de Células , Transplante de Células/métodos , Hepatócitos/classificação , Hepatócitos/citologia , Fígado/lesões , Neoplasias Hepáticas , Camundongos , Regeneração , Fatores de Transcrição SOX9/genética , Transcriptoma
5.
Cell ; 155(2): 384-96, 2013 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-24120137

RESUMO

Hepatocellular carcinoma (HCC) is a slowly developing malignancy postulated to evolve from premalignant lesions in chronically damaged livers. However, it was never established that premalignant lesions actually contain tumor progenitors that give rise to cancer. Here, we describe isolation and characterization of HCC progenitor cells (HcPCs) from different mouse HCC models. Unlike fully malignant HCC, HcPCs give rise to cancer only when introduced into a liver undergoing chronic damage and compensatory proliferation. Although HcPCs exhibit a similar transcriptomic profile to bipotential hepatobiliary progenitors, the latter do not give rise to tumors. Cells resembling HcPCs reside within dysplastic lesions that appear several months before HCC nodules. Unlike early hepatocarcinogenesis, which depends on paracrine IL-6 production by inflammatory cells, due to upregulation of LIN28 expression, HcPCs had acquired autocrine IL-6 signaling that stimulates their in vivo growth and malignant progression. This may be a general mechanism that drives other IL-6-producing malignancies.


Assuntos
Comunicação Autócrina , Regulação Neoplásica da Expressão Gênica , Interleucina-6/metabolismo , Neoplasias Hepáticas/patologia , Células-Tronco Neoplásicas/metabolismo , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Progressão da Doença , Hepacivirus , Hepatite C/genética , Hepatite C/metabolismo , Hepatite C/patologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
6.
Proc Natl Acad Sci U S A ; 120(25): e2304055120, 2023 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-37310998

RESUMO

The polymorphic nature and intrinsic instability of class I major histocompatibility complex (MHC-I) and MHC-like molecules loaded with suboptimal peptides, metabolites, or glycolipids presents a fundamental challenge for identifying disease-relevant antigens and antigen-specific T cell receptors (TCRs), hindering the development of autologous therapeutics. Here, we leverage the positive allosteric coupling between the peptide and light chain (ß2 microglobulin, ß2m) subunits for binding to the MHC-I heavy chain (HC) through an engineered disulfide bond bridging conserved epitopes across the HC/ß2m interface, to generate conformationally stable, peptide-receptive molecules named "open MHC-I." Biophysical characterization shows that open MHC-I molecules are properly folded protein complexes of enhanced thermal stability compared to the wild type when loaded with low- to moderate-affinity peptides. Using solution NMR, we characterize the effects of the disulfide bond on the conformation and dynamics of the MHC-I structure, ranging from local changes in ß2m-interacting sites of the peptide-binding groove to long-range effects on the α2-1 helix and α3 domain. The interchain disulfide bond stabilizes MHC-I molecules in an open conformation to promote peptide exchange across multiple human leukocyte antigen (HLA) allotypes, covering representatives from five HLA-A supertypes, six HLA-B supertypes, and oligomorphic HLA-Ib molecules. Our structure-guided design, combined with conditional ß-peptide ligands, provides a universal platform to generate ready-to-load MHC-I systems of enhanced stability, enabling a range of approaches to screen antigenic epitope libraries and probe polyclonal TCR repertoires covering highly polymorphic HLA-I allotypes, as well as oligomorphic nonclassical molecules.


Assuntos
Antígenos de Histocompatibilidade Classe II , Antígenos de Histocompatibilidade , Humanos , Peptídeos/genética , Complexo Principal de Histocompatibilidade , Epitopos , Dissulfetos
7.
Nature ; 521(7550): 94-8, 2015 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-25924065

RESUMO

Cancer-associated genetic alterations induce expression of tumour antigens that can activate CD8(+) cytotoxic T cells (CTLs), but the microenvironment of established tumours promotes immune tolerance through poorly understood mechanisms. Recently developed therapeutics that overcome tolerogenic mechanisms activate tumour-directed CTLs and are effective in some human cancers. Immune mechanisms also affect treatment outcome, and certain chemotherapeutic drugs stimulate cancer-specific immune responses by inducing immunogenic cell death and other effector mechanisms. Our previous studies revealed that B cells recruited by the chemokine CXCL13 into prostate cancer tumours promote the progression of castrate-resistant prostate cancer by producing lymphotoxin, which activates an IκB kinase α (IKKα)-BMI1 module in prostate cancer stem cells. Because castrate-resistant prostate cancer is refractory to most therapies, we examined B cell involvement in the acquisition of chemotherapy resistance. Here we focus on oxaliplatin, an immunogenic chemotherapeutic agent that is effective in aggressive prostate cancer. We show that mouse B cells modulate the response to low-dose oxaliplatin, which promotes tumour-directed CTL activation by inducing immunogenic cell death. Three different mouse prostate cancer models were refractory to oxaliplatin unless genetically or pharmacologically depleted of B cells. The crucial immunosuppressive B cells are plasmocytes that express IgA, interleukin (IL)-10 and programmed death ligand 1 (PD-L1), the appearance of which depends on TGFß receptor signalling. Elimination of these cells, which also infiltrate human-therapy-resistant prostate cancer, allows CTL-dependent eradication of oxaliplatin-treated tumours.


Assuntos
Compostos Organoplatínicos/farmacologia , Plasmócitos/efeitos dos fármacos , Plasmócitos/imunologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/imunologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia , Transferência Adotiva , Animais , Anticorpos Antineoplásicos/imunologia , Antineoplásicos/imunologia , Antineoplásicos/farmacologia , Antígeno B7-H1/metabolismo , Células Cultivadas , Quimiocina CXCL13/metabolismo , Humanos , Quinase I-kappa B/metabolismo , Imunoglobulina A/imunologia , Interleucina-10/imunologia , Ativação Linfocitária/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neoplásicas/patologia , Compostos Organoplatínicos/administração & dosagem , Compostos Organoplatínicos/imunologia , Compostos Organoplatínicos/uso terapêutico , Oxaliplatina , Plasmócitos/citologia , Neoplasias da Próstata/patologia , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Linfócitos T Citotóxicos/citologia , Fator de Crescimento Transformador beta/imunologia
8.
Proc Natl Acad Sci U S A ; 115(42): E9879-E9888, 2018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-30287485

RESUMO

Cancer genomics has enabled the exhaustive molecular characterization of tumors and exposed hepatocellular carcinoma (HCC) as among the most complex cancers. This complexity is paralleled by dozens of mouse models that generate histologically similar tumors but have not been systematically validated at the molecular level. Accurate models of the molecular pathogenesis of HCC are essential for biomedical progress; therefore we compared genomic and transcriptomic profiles of four separate mouse models [MUP transgenic, TAK1-knockout, carcinogen-driven diethylnitrosamine (DEN), and Stelic Animal Model (STAM)] with those of 987 HCC patients with distinct etiologies. These four models differed substantially in their mutational load, mutational signatures, affected genes and pathways, and transcriptomes. STAM tumors were most molecularly similar to human HCC, with frequent mutations in Ctnnb1, similar pathway alterations, and high transcriptomic similarity to high-grade, proliferative human tumors with poor prognosis. In contrast, TAK1 tumors better reflected the mutational signature of human HCC and were transcriptionally similar to low-grade human tumors. DEN tumors were least similar to human disease and almost universally carried the Braf V637E mutation, which is rarely found in human HCC. Immune analysis revealed that strain-specific MHC-I genotype can influence the molecular makeup of murine tumors. Thus, different mouse models of HCC recapitulate distinct aspects of HCC biology, and their use should be adapted to specific questions based on the molecular features provided here.


Assuntos
Biomarcadores Tumorais/genética , Carcinoma Hepatocelular/genética , Perfilação da Expressão Gênica , Genômica/métodos , Neoplasias Hepáticas Experimentais/genética , Neoplasias Hepáticas/genética , Animais , Carcinoma Hepatocelular/patologia , Modelos Animais de Doenças , Humanos , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas Experimentais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Transcriptoma
9.
Proc Natl Acad Sci U S A ; 111(38): 13870-5, 2014 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-25189770

RESUMO

Metastatic spread is the leading cause of cancer mortality. Breast cancer (BCa) metastatic recurrence can happen years after removal of the primary tumor. Here we show that Ubc13, an E2 enzyme that catalyzes K63-linked protein polyubiquitination, is largely dispensable for primary mammary tumor growth but is required for metastatic spread and lung colonization by BCa cells. Loss of Ubc13 inhibited BCa growth and survival only at metastatic sites. Ubc13 was dispensable for transforming growth factor ß (TGFß)-induced SMAD activation but was required for activation of non-SMAD signaling via TGFß-activating kinase 1 (TAK1) and p38, whose activity controls expression of numerous metastasis promoting genes. p38 activation restored metastatic activity to Ubc13-deficient cells, and its pharmacological inhibition attenuated BCa metastasis in mice, suggesting it is a therapeutic option for metastatic BCa.


Assuntos
Neoplasias da Mama/enzimologia , MAP Quinase Quinase Quinases/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas de Neoplasias/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Xenoenxertos , Humanos , MAP Quinase Quinase Quinases/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Metástase Neoplásica , Proteínas de Neoplasias/genética , Transplante de Neoplasias , Enzimas de Conjugação de Ubiquitina/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética
10.
Proc Natl Acad Sci U S A ; 111(3): 1090-5, 2014 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-24395807

RESUMO

E-cadherin is an important adhesion molecule whose loss is associated with progression and poor prognosis of liver cancer. However, it is unclear whether the loss of E-cadherin is a real culprit or a bystander in liver cancer progression. In addition, the precise role of E-cadherin in maintaining liver homeostasis is also still unknown, especially in vivo. Here we demonstrate that liver-specific E-cadherin knockout mice develop spontaneous periportal inflammation via an impaired intrahepatic biliary network, as well as periductal fibrosis, which resembles primary sclerosing cholangitis. Inducible gene knockout studies identified E-cadherin loss in biliary epithelial cells as a causal factor of cholangitis induction. Furthermore, a few of the E-cadherin knockout mice developed spontaneous liver cancer. When knockout of E-cadherin is combined with Ras activation or chemical carcinogen administration, E-cadherin knockout mice display markedly accelerated carcinogenesis and an invasive phenotype associated with epithelial-mesenchymal transition, up-regulation of stem cell markers, and elevated ERK activation. Also in human hepatocellular carcinoma, E-cadherin loss correlates with increased expression of mesenchymal and stem cell markers, and silencing of E-cadherin in hepatocellular carcinoma cell lines causes epithelial-mesenchymal transition and increased invasiveness, suggesting that E-cadherin loss can be a causal factor of these phenotypes. Thus, E-cadherin plays critical roles in maintaining homeostasis and suppressing carcinogenesis in the liver.


Assuntos
Caderinas/metabolismo , Carcinogênese , Colangite Esclerosante/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Colangite/metabolismo , Transição Epitelial-Mesenquimal , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hepatócitos/citologia , Inflamação , Fígado/patologia , Proteínas Luminescentes/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Metástase Neoplásica , Fenótipo , Prognóstico , Células-Tronco/citologia
11.
Nucleic Acids Res ; 42(4): 2126-37, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24271395

RESUMO

Development of tools to jointly visualize the genome and the epigenome remains a challenge. chroGPS is a computational approach that addresses this question. chroGPS uses multidimensional scaling techniques to represent similarity between epigenetic factors, or between genetic elements on the basis of their epigenetic state, in 2D/3D reference maps. We emphasize biological interpretability, statistical robustness, integration of genetic and epigenetic data from heterogeneous sources, and computational feasibility. Although chroGPS is a general methodology to create reference maps and study the epigenetic state of any class of genetic element or genomic region, we focus on two specific kinds of maps: chroGPS(factors), which visualizes functional similarities between epigenetic factors, and chroGPS(genes), which describes the epigenetic state of genes and integrates gene expression and other functional data. We use data from the modENCODE project on the genomic distribution of a large collection of epigenetic factors in Drosophila, a model system extensively used to study genome organization and function. Our results show that the maps allow straightforward visualization of relationships between factors and elements, capturing relevant information about their functional properties that helps to interpret epigenetic information in a functional context and derive testable hypotheses.


Assuntos
Cromatina/metabolismo , Epigênese Genética , Epigenômica/métodos , Software , Animais , Linhagem Celular , Gráficos por Computador , Drosophila/genética , Expressão Gênica , Genes de Insetos , Transdução de Sinais/genética
12.
Nutrients ; 12(4)2020 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-32230953

RESUMO

The liver plays an outsized role in oncology. Liver tumors are one of the most frequently found tumors in cancer patients and these arise from either primary or metastatic disease. Hepatocellular carcinoma (HCC), the most prevalent form of primary liver cancer and the 6th most common cancer type overall, is expected to become the 3rd leading cause of cancer mortality in the US by the year 2030. The liver is also the most common site of distant metastasis from solid tumors. For instance, colorectal cancer (CRC) metastasizes to the liver in two-thirds of cases, and CRC liver metastasis is the leading cause of mortality in these patients. The interplay between inflammation and cancer is unmistakably evident in the liver. In nearly every case, HCC is diagnosed in chronic liver disease (CLD) and cirrhosis background. The consumption of a Western-style high-fat diet is a major risk factor for the development of non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH), both of which are becoming more prevalent in parallel with the obesity epidemic. Excessive alcohol intake also contributes significantly to the CLD burden in the form of alcoholic liver disease (ALD). Inflammation is a key component in the development of all CLDs. Additionally, during the development of liver metastasis, pro-inflammatory signaling is crucial in eliminating invading cancer cells but ironically also helps foster a pro-metastatic environment that supports metastatic seeding and colonization. Here we review how Westernized high-fat diets and excessive alcohol intake can influence inflammation within the liver microenvironment, stimulating both primary and metastatic liver tumorigenesis.


Assuntos
Carcinoma Hepatocelular , Dieta Hiperlipídica , Inflamação , Hepatopatias Alcoólicas , Neoplasias Hepáticas , Carcinogênese , Neoplasias Colorretais , Dieta , Microbioma Gastrointestinal/fisiologia , Humanos , Inflamação/metabolismo , Inflamação/fisiopatologia , Fígado/metabolismo , Fígado/fisiologia
13.
Methods Mol Biol ; 2131: 185-198, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32162254

RESUMO

MHC class I proteins present intracellular peptides on the cell's surface, enabling the immune system to recognize tumor-specific neoantigens of early neoplastic cells and eliminate them before the tumor develops further. However, variability in peptide-MHC-I affinity results in variable presentation of oncogenic peptides, leading to variable likelihood of immune evasion across both individuals and mutations. Since the major determinant of peptide-MHC-I affinity in patients is individual MHC-I genotype, we developed a residue-centric presentation score taking both mutated residues and MHC-I genotype into account and hypothesized that high scores (which correspond to poor presentation) would correlate to high mutation frequencies within tumors. We applied our scoring system to 9176 tumor samples from TCGA across 1018 recurrent mutations and found that, indeed, presentation scores predicted mutation probability. These findings open the door to more personalized treatment plans based on simple genotyping. Here, we outline the computational tools and statistical methods used to arrive at this conclusion.


Assuntos
Biologia Computacional/métodos , Antígenos de Histocompatibilidade Classe II/genética , Mutação , Neoplasias/genética , Bases de Dados Genéticas , Predisposição Genética para Doença , Técnicas de Genotipagem , Humanos , Funções Verossimilhança , Taxa de Mutação , Medicina de Precisão , Evasão Tumoral , Sequenciamento do Exoma
14.
Cell Mol Gastroenterol Hepatol ; 9(2): 295-312, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31606566

RESUMO

BACKGROUND AND AIMS: Tumor necrosis factor (TNF) is a major pathogenic effector and a therapeutic target in inflammatory bowel disease (IBD), yet the basis for TNF-induced intestinal epithelial cell (IEC) death is unknown, because TNF does not kill normal IECs. Here, we investigated how chronic nuclear factor (NF)- κB activation, which occurs in human IBD, promotes TNF-dependent IEC death in mice. METHODS: Human IBD specimens were stained for p65 and cleaved caspase-3. C57BL/6 mice with constitutively active IKKß in IEC (Ikkß(EE)IEC), Ripk1D138N/D138N knockin mice, and Ripk3-/- mice were injected with TNF or lipopolysaccharide. Enteroids were also isolated from these mice and challenged with TNF with or without RIPK1 and RIPK3 inhibitors or butylated hydroxyanisole. Ripoptosome-mediated caspase-8 activation was assessed by immunoprecipitation. RESULTS: NF-κB activation in human IBD correlated with appearance of cleaved caspase-3. Congruently, unlike normal mouse IECs that are TNF-resistant, IECs in Ikkß(EE)IEC mice and enteroids were susceptible to TNF-dependent apoptosis, which depended on the protein kinase function of RIPK1. Constitutively active IKKß facilitated ripoptosome formation, a RIPK1 signaling complex that mediates caspase-8 activation by TNF. Butylated hydroxyanisole treatment and RIPK1 inhibitors attenuated TNF-induced and ripoptosome-mediated caspase-8 activation and IEC death in vitro and in vivo. CONCLUSIONS: Contrary to common expectations, chronic NF-κB activation induced intestinal crypt apoptosis after TNF stimulation, resulting in severe mucosal erosion. RIPK1 kinase inhibitors selectively inhibited TNF destructive properties while preserving its survival and proliferative properties, which do not require RIPK1 kinase activity. RIPK1 kinase inhibition could be a potential treatment for IBD.


Assuntos
Apoptose/imunologia , Colite Ulcerativa/imunologia , Doença de Crohn/imunologia , Mucosa Intestinal/patologia , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Fator de Transcrição RelA/metabolismo , Adulto , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Células Cultivadas , Colite Ulcerativa/tratamento farmacológico , Colite Ulcerativa/patologia , Colo/efeitos dos fármacos , Colo/imunologia , Colo/patologia , Colonoscopia , Doença de Crohn/tratamento farmacológico , Doença de Crohn/patologia , Modelos Animais de Doenças , Células Epiteliais/imunologia , Células Epiteliais/patologia , Técnicas de Introdução de Genes , Humanos , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Íleo/imunologia , Íleo/patologia , Imidazóis/farmacologia , Imidazóis/uso terapêutico , Indóis/farmacologia , Indóis/uso terapêutico , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Camundongos Knockout , Organoides , Cultura Primária de Células , RNA-Seq , Proteína Serina-Treonina Quinases de Interação com Receptores/antagonistas & inibidores , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Fator de Necrose Tumoral alfa/metabolismo
15.
Methods Mol Biol ; 1905: 59-70, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30536090

RESUMO

The liver is unmatched in regenerative capacity. However, when exhausted, the liver is predisposed to various diseases based on injury types and causal agents. Although hepatocytes have been proposed to be the main source of new hepatocytes during regeneration, the existence of specialized liver stem cells has been long debated. In mice, oval cells or ductal cells have been postulated as such stem/progenitor pool. Exhaustive works from different laboratories have shown that in genetically unmodified mice, oval cells, or by extension ductal cells, only contribute marginally in producing new hepatocytes during liver regeneration, thus indicating that hepatocytes are the main regenerative cell source. In this debated context, we identified a new population of periportal hepatocytes in the normal mouse liver. These cells we termed hybrid hepatocytes (HybHP) express low levels of the transcription factor Sox9. Using complementary lineage tracing tools, we demonstrated that HybHP regenerate the liver after chronic hepatocyte depleting injuries. Here, we describe the two-step genetic recombination method that allowed us to study HybHP's lineage in two established models of liver injury.


Assuntos
Hepatócitos/citologia , Fígado/lesões , Recombinação Genética , Fatores de Transcrição SOX9/metabolismo , Células-Tronco/citologia , Animais , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Modelos Animais de Doenças , Regulação para Baixo , Feminino , Hepatócitos/metabolismo , Fígado/citologia , Fígado/metabolismo , Regeneração Hepática , Masculino , Camundongos , Células-Tronco/metabolismo
16.
Mol Cell Oncol ; 5(2): e1409863, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29487895

RESUMO

MHC-I exposes the intracellular contents to immune cells for surveillance of cellular health. Due to high genomic variation, individuals' immune systems differ in their ability to expose and eliminate cancer-causing mutations. These personalized immune blind spots create specific oncogenic mutation predispositions within patients and influence their prevalence across populations.

17.
Cell Metab ; 23(1): 48-62, 2016 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-26771116

RESUMO

Although discussion of the obesity epidemic had become a cocktail party cliché, its impact on public health cannot be dismissed. In the past decade, cancer had joined the list of chronic debilitating diseases whose risk is substantially increased by hypernutrition. Here we discuss recent advances in understanding how obesity increases cancer risk and propose a unifying hypothesis according to which the major tumor-promoting mechanism triggered by hypernutrition is the indolent inflammation that takes place at particular organ sites, including liver, pancreas, and gastrointestinal tract. The mechanisms by which excessive fat deposition feeds this tumor-promoting inflammatory flame are diverse and tissue specific.


Assuntos
Neoplasias do Sistema Digestório/etiologia , Obesidade/complicações , Animais , Transformação Celular Neoplásica/imunologia , Neoplasias do Sistema Digestório/imunologia , Neoplasias do Sistema Digestório/metabolismo , Predisposição Genética para Doença , Humanos , Inflamação/etiologia , Inflamação/metabolismo , Mediadores da Inflamação/fisiologia , Obesidade/imunologia , Obesidade/metabolismo , Especificidade de Órgãos , Fatores de Risco
18.
Cancer Cell ; 29(6): 935-948, 2016 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-27211490

RESUMO

p62 is a ubiquitin-binding autophagy receptor and signaling protein that accumulates in premalignant liver diseases and most hepatocellular carcinomas (HCCs). Although p62 was proposed to participate in the formation of benign adenomas in autophagy-deficient livers, its role in HCC initiation was not explored. Here we show that p62 is necessary and sufficient for HCC induction in mice and that its high expression in non-tumor human liver predicts rapid HCC recurrence after curative ablation. High p62 expression is needed for activation of NRF2 and mTORC1, induction of c-Myc, and protection of HCC-initiating cells from oxidative stress-induced death.


Assuntos
Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Células-Tronco Neoplásicas/citologia , Proteína Sequestossoma-1/genética , Regulação para Cima , Animais , Carcinoma Hepatocelular/patologia , Sobrevivência Celular , Dietilnitrosamina/efeitos adversos , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Transgênicos , Complexos Multiproteicos/genética , Fator 2 Relacionado a NF-E2/genética , Neoplasias Experimentais , Células-Tronco Neoplásicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/genética , Serina-Treonina Quinases TOR/genética
19.
Nat Commun ; 6: 7049, 2015 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-25916810

RESUMO

dDsk2 is a conserved extraproteasomal ubiquitin receptor that targets ubiquitylated proteins for degradation. Here we report that dDsk2 plays a nonproteolytic function in transcription regulation. dDsk2 interacts with the dHP1c complex, localizes at promoters of developmental genes and is required for transcription. Through the ubiquitin-binding domain, dDsk2 interacts with H2Bub1, a modification that occurs at dHP1c complex-binding sites. H2Bub1 is not required for binding of the complex; however, dDsk2 depletion strongly reduces H2Bub1. Co-depletion of the H2Bub1 deubiquitylase dUbp8/Nonstop suppresses this reduction and rescues expression of target genes. RNA polymerase II is strongly paused at promoters of dHP1c complex target genes and dDsk2 depletion disrupts pausing. Altogether, these results suggest that dDsk2 prevents dUbp8/Nonstop-dependent H2Bub1 deubiquitylation at promoters of dHP1c complex target genes and regulates RNA polymerase II pausing. These results expand the catalogue of nonproteolytic functions of ubiquitin receptors to the epigenetic regulation of chromatin modifications.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimologia , Drosophila melanogaster/genética , Histonas/metabolismo , RNA Polimerase II/metabolismo , Animais , Sítios de Ligação , Proteínas de Transporte/química , Proteínas de Ciclo Celular/química , Imunoprecipitação da Cromatina , Proteínas de Drosophila/química , Histonas/química , Complexos Multiproteicos/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Estrutura Terciária de Proteína , Proteólise , Sítio de Iniciação de Transcrição , Transcrição Gênica , Ubiquitinação
20.
Cancer Cell ; 26(3): 331-343, 2014 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-25132496

RESUMO

Endoplasmic reticulum (ER) stress has been implicated in the pathogenesis of viral hepatitis, insulin resistance, hepatosteatosis, and nonalcoholic steatohepatitis (NASH), disorders that increase risk of hepatocellular carcinoma (HCC). To determine whether and how ER stress contributes to obesity-driven hepatic tumorigenesis we fed wild-type (WT) and MUP-uPA mice, in which hepatocyte ER stress is induced by plasminogen activator expression, with high-fat diet. Although both strains were equally insulin resistant, the MUP-uPA mice exhibited more liver damage, more immune infiltration, and increased lipogenesis and, as a result, displayed classical NASH signs and developed typical steatohepatitic HCC. Both NASH and HCC development were dependent on TNF produced by inflammatory macrophages that accumulate in the MUP-uPA liver in response to hepatocyte ER stress.


Assuntos
Carcinoma Hepatocelular/metabolismo , Estresse do Retículo Endoplasmático , Neoplasias Hepáticas Experimentais/metabolismo , Hipernutrição/complicações , Fator de Necrose Tumoral alfa/fisiologia , Animais , Carcinoma Hepatocelular/etiologia , Carcinoma Hepatocelular/patologia , Células Cultivadas , Dieta Hiperlipídica/efeitos adversos , Chaperona BiP do Retículo Endoplasmático , Fígado Gorduroso/etiologia , Fígado Gorduroso/metabolismo , Proteínas de Choque Térmico/metabolismo , Lipogênese , Neoplasias Hepáticas Experimentais/etiologia , Neoplasias Hepáticas Experimentais/patologia , Masculino , Camundongos , Camundongos Transgênicos , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Transdução de Sinais , Carga Tumoral , Ativador de Plasminogênio Tipo Uroquinase/genética , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA