Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Surg Res ; 279: 712-721, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35933789

RESUMO

INTRODUCTION: Resuscitative endovascular balloon occlusion of the aorta (REBOA) causes a severe ischemia-reperfusion injury. Endovascular Perfusion Augmentation for Critical Care (EPACC) has emerged as a hemodynamic/mechanical adjunct to vasopressors and crystalloid for the treatment of post-REBOA ischemia-reperfusion injury. The objective of the study is to examine the impact of EPACC as a tool for a wean from complete REBOA compared to standard resuscitation techniques. METHODS: Nine swine underwent anesthesia and then a controlled 30% blood volume hemorrhage with 30 min of supraceliac total aortic occlusion to create an ischemia-reperfusion injury. Animals were randomized to standardized critical care (SCC) or 90 min of EPACC followed by SCC. The critical care phase lasted 270 min after injury. Hemodynamic markers and laboratory values of ischemia were recorded. RESULTS: During the first 90 min the intervention phase SCC spent 60% (54%-73%) and EPACC spent 91% (88%-92%) of the time avoiding proximal hypotension (<60 mm Hg), P = 0.03. There was also a statistically significant decrease in cumulative norepinephrine dose at the end of the experiment between SCC (80.89 mcg/kg) versus EPACC (22.03 mcg/kg), P = 0.03. Renal artery flow during EPACC was similar compared to SCC during EPACC, P = 0.19. But during the last hour of the experiment (after removal of aortic balloon) the renal artery flow in EPACC (2.9 mL/kg/min) was statistically significantly increased compared to SCC (1.57 mL/min/kg), P = 0.03. There was a statistically significant decrease in terminal creatinine in the EPACC (1.7 mg/dL) compared to SCC (2.1 mg/dL), P = 0.03. CONCLUSIONS: The 90 min of EPACC as a weaning adjunct in the setting of a severe ischemia-reperfusion injury after complete supraceliac REBOA provides improved renal flow with improvement in terminal creatinine compared to SCC with stabilized proximal hemodynamics and decreased vasopressor dose.


Assuntos
Oclusão com Balão , Procedimentos Endovasculares , Traumatismo por Reperfusão , Choque Hemorrágico , Animais , Aorta , Oclusão com Balão/métodos , Creatinina , Soluções Cristaloides , Modelos Animais de Doenças , Procedimentos Endovasculares/métodos , Norepinefrina , Perfusão , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/prevenção & controle , Ressuscitação/métodos , Choque Hemorrágico/terapia , Suínos
3.
Breast Cancer Res ; 22(1): 125, 2020 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-33187540

RESUMO

BACKGROUND: CRIPTO is a multi-functional signaling protein that promotes stemness and oncogenesis. We previously developed a CRIPTO antagonist, ALK4L75A-Fc, and showed that it causes loss of the stem cell phenotype in normal mammary epithelia suggesting it may similarly inhibit CRIPTO-dependent plasticity in breast cancer cells. METHODS: We focused on two triple negative breast cancer cell lines (MDA-MB-231 and MDA-MB-468) to measure the effects of ALK4L75A-Fc on cancer cell behavior under nutrient deprivation and endoplasmic reticulum stress. We characterized the proliferation and migration of these cells in vitro using time-lapse microscopy and characterized stress-dependent changes in the levels and distribution of CRIPTO signaling mediators and cancer stem cell markers. We also assessed the effects of ALK4L75A-Fc on proliferation, EMT, and stem cell markers in vivo as well as on tumor growth and metastasis using inducible lentiviral delivery or systemic administration of purified ALK4L75A-Fc, which represents a candidate therapeutic approach. RESULTS: ALK4L75A-Fc inhibited adaptive responses of breast cancer cells under conditions of nutrient and ER stress and reduced their proliferation, migration, clonogenicity, and expression of EMT and cancer stem cell markers. ALK4L75A-Fc also inhibited proliferation of human breast cancer cells in stressed tumor microenvironments in xenografts and reduced both primary tumor size and metastatic burden. CONCLUSIONS: Cancer cell adaptation to stresses such as nutrient deprivation, hypoxia, and chemotherapy can critically contribute to dormancy, metastasis, therapy resistance, and recurrence. Identifying mechanisms that govern cellular adaptation, plasticity, and the emergence of stem-like cancer cells may be key to effective anticancer therapies. Results presented here indicate that targeting CRIPTO with ALK4L75A-Fc may have potential as such a therapy since it inhibits breast cancer cell adaptation to microenvironmental challenges and associated stem-like and EMT phenotypes.


Assuntos
Proteínas Ligadas por GPI/antagonistas & inibidores , Proteínas de Neoplasias/antagonistas & inibidores , Células-Tronco Neoplásicas/efeitos dos fármacos , Proteínas Recombinantes de Fusão/farmacologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Receptores de Ativinas Tipo I/genética , Animais , Linhagem Celular Tumoral , Plasticidade Celular/efeitos dos fármacos , Estresse do Retículo Endoplasmático , Feminino , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Recidiva Local de Neoplasia , Células-Tronco Neoplásicas/patologia , Mutação Puntual , Ligação Proteica/genética , Domínios Proteicos/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/uso terapêutico , Neoplasias de Mama Triplo Negativas/patologia , Hipóxia Tumoral , Microambiente Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Immunity ; 53(1): 143-157.e8, 2020 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-32640256

RESUMO

Regulatory T (Treg) cells play a pivotal role in suppressing auto-reactive T cells and maintaining immune homeostasis. Treg cell development and function are dependent on the transcription factor Foxp3. Here, we performed a genome-wide CRISPR loss-of-function screen to identify Foxp3 regulators in mouse primary Treg cells. Foxp3 regulators were enriched in genes encoding subunits of the SWI/SNF nucleosome-remodeling and SAGA chromatin-modifying complexes. Among the three SWI/SNF-related complexes, the Brd9-containing non-canonical (nc) BAF complex promoted Foxp3 expression, whereas the PBAF complex was repressive. Chemical-induced degradation of Brd9 led to reduced Foxp3 expression and reduced Treg cell function in vitro. Brd9 ablation compromised Treg cell function in inflammatory disease and tumor immunity in vivo. Furthermore, Brd9 promoted Foxp3 binding and expression of a subset of Foxp3 target genes. Our findings provide an unbiased analysis of the genetic networks regulating Foxp3 and reveal ncBAF as a target for therapeutic manipulation of Treg cell function.


Assuntos
Sistemas CRISPR-Cas/genética , Fatores de Transcrição Forkhead/metabolismo , Neoplasias/imunologia , Linfócitos T Reguladores/metabolismo , Fatores de Transcrição/metabolismo , Animais , Autoimunidade/imunologia , Linhagem Celular Tumoral , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Modelos Animais de Doenças , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica/genética , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nucleossomos/imunologia , RNA Guia de Cinetoplastídeos/genética , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Fatores de Transcrição/genética
5.
Nature ; 569(7754): 131-135, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30996350

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis largely owing to inefficient diagnosis and tenacious drug resistance. Activation of pancreatic stellate cells (PSCs) and consequent development of dense stroma are prominent features accounting for this aggressive biology1,2. The reciprocal interplay between PSCs and pancreatic cancer cells (PCCs) not only enhances tumour progression and metastasis but also sustains their own activation, facilitating a vicious cycle to exacerbate tumorigenesis and drug resistance3-7. Furthermore, PSC activation occurs very early during PDAC tumorigenesis8-10, and activated PSCs comprise a substantial fraction of the tumour mass, providing a rich source of readily detectable factors. Therefore, we hypothesized that the communication between PSCs and PCCs could be an exploitable target to develop effective strategies for PDAC therapy and diagnosis. Here, starting with a systematic proteomic investigation of secreted disease mediators and underlying molecular mechanisms, we reveal that leukaemia inhibitory factor (LIF) is a key paracrine factor from activated PSCs acting on cancer cells. Both pharmacologic LIF blockade and genetic Lifr deletion markedly slow tumour progression and augment the efficacy of chemotherapy to prolong survival of PDAC mouse models, mainly by modulating cancer cell differentiation and epithelial-mesenchymal transition status. Moreover, in both mouse models and human PDAC, aberrant production of LIF in the pancreas is restricted to pathological conditions and correlates with PDAC pathogenesis, and changes in the levels of circulating LIF correlate well with tumour response to therapy. Collectively, these findings reveal a function of LIF in PDAC tumorigenesis, and suggest its translational potential as an attractive therapeutic target and circulating marker. Our studies underscore how a better understanding of cell-cell communication within the tumour microenvironment can suggest novel strategies for cancer therapy.


Assuntos
Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/patologia , Fator Inibidor de Leucemia/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Comunicação Parácrina , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Carcinogênese/genética , Carcinoma Ductal Pancreático/diagnóstico , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Linhagem Celular Tumoral , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos , Transição Epitelial-Mesenquimal , Feminino , Humanos , Fator Inibidor de Leucemia/antagonistas & inibidores , Fator Inibidor de Leucemia/sangue , Masculino , Espectrometria de Massas , Camundongos , Neoplasias Pancreáticas/diagnóstico , Comunicação Parácrina/efeitos dos fármacos , Receptores de OSM-LIF/deficiência , Receptores de OSM-LIF/genética , Receptores de OSM-LIF/metabolismo , Microambiente Tumoral
6.
Cell ; 176(5): 1098-1112.e18, 2019 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-30794774

RESUMO

Increased levels of intestinal bile acids (BAs) are a risk factor for colorectal cancer (CRC). Here, we show that the convergence of dietary factors (high-fat diet) and dysregulated WNT signaling (APC mutation) alters BA profiles to drive malignant transformations in Lgr5-expressing (Lgr5+) cancer stem cells and promote an adenoma-to-adenocarcinoma progression. Mechanistically, we show that BAs that antagonize intestinal farnesoid X receptor (FXR) function, including tauro-ß-muricholic acid (T-ßMCA) and deoxycholic acid (DCA), induce proliferation and DNA damage in Lgr5+ cells. Conversely, selective activation of intestinal FXR can restrict abnormal Lgr5+ cell growth and curtail CRC progression. This unexpected role for FXR in coordinating intestinal self-renewal with BA levels implicates FXR as a potential therapeutic target for CRC.


Assuntos
Neoplasias Intestinais/metabolismo , Células-Tronco Neoplásicas/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Ácidos e Sais Biliares/metabolismo , Linhagem Celular , Proliferação de Células/genética , Neoplasias Colorretais/metabolismo , Ácido Desoxicólico/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Neoplasias Intestinais/genética , Intestinos , Fígado , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neoplásicas/fisiologia , Organoides/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Risco , Transdução de Sinais , Ácido Taurocólico/análogos & derivados , Ácido Taurocólico/metabolismo , Via de Sinalização Wnt/genética , Via de Sinalização Wnt/fisiologia
7.
Cell Metab ; 29(2): 285-302.e7, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30415923

RESUMO

AMPK, a conserved sensor of low cellular energy, can either repress or promote tumor growth depending on the context. However, no studies have examined AMPK function in autochthonous genetic mouse models of epithelial cancer. Here, we examine the role of AMPK in murine KrasG12D-mediated non-small-cell lung cancer (NSCLC), a cancer type in humans that harbors frequent inactivating mutations in the LKB1 tumor suppressor-the predominant upstream activating kinase of AMPK and 12 related kinases. Unlike LKB1 deletion, AMPK deletion in KrasG12D lung tumors did not accelerate lung tumor growth. Moreover, deletion of AMPK in KrasG12D p53f/f tumors reduced lung tumor burden. We identified a critical role for AMPK in regulating lysosomal gene expression through the Tfe3 transcription factor, which was required to support NSCLC growth. Thus, AMPK supports the growth of KrasG12D-dependent lung cancer through the induction of lysosomes, highlighting an unrecognized liability of NSCLC.


Assuntos
Proteínas Quinases Ativadas por AMP/fisiologia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Animais , Linhagem Celular Tumoral , Mutação com Perda de Função , Camundongos
8.
Mol Cancer Res ; 16(5): 825-832, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29523765

RESUMO

Small cell lung cancer (SCLC) is the most deadly subtype of lung cancer due to its dismal prognosis. We have developed a lentiviral vector-mediated SCLC mouse model and have explored the role of both the NF-κB and CREB families of transcription factors in this model. Surprisingly, induction of NF-κB activity, which promotes tumor progression in many cancer types including non-small cell lung carcinoma (NSCLC), is dispensable in SCLC. Instead, suppression of NF-κB activity in SCLC tumors moderately accelerated tumor development. Examination of gene expression signatures of both mouse and human SCLC tumors revealed overall low NF-κB but high CREB activity. Blocking CREB activation by a dominant-negative form of PKA (dnPKA) completely abolished the development of SCLC. Similarly, expression of dnPKA or treatment with PKA inhibitor H89 greatly reduced the growth of SCLC tumors in syngeneic transplantation models. Altogether, our results strongly suggest that targeting CREB is a promising therapeutic strategy against SCLC.Implications: Activity of the transcription factor CREB is elevated in SCLC tumors, which helps to maintain its neuroendocrine signature and cell proliferation. Our results highlight the importance of targeting the CREB pathway to develop new therapeutics to combat SCLC. Mol Cancer Res; 16(5); 825-32. ©2018 AACR.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Neoplasias Pulmonares/genética , Carcinoma de Pequenas Células do Pulmão/genética , Animais , Modelos Animais de Doenças , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Transdução de Sinais , Carcinoma de Pequenas Células do Pulmão/patologia
9.
Comp Med ; 67(6): 529-536, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-29212586

RESUMO

A 10-y-old cranially implanted rhesus macaque (Macaca mulatta) involved in visual research was presented for dull mentation and weight loss. Physical examination revealed alopecia and poor body conditioning, and bloodwork revealed marked hypercortisolemia (23 µg/dL). Differential diagnoses for hypercortisolemia, weight loss, and alopecia included Cushing and pseudo-Cushing syndromes. To further evaluate hypercortisolemia, we compared the urine cortisol:creatinine ratio (UCCR) at baseline and after low-dose dexamethasone suppression (LDDS) testing in the presenting animal and healthy naïve and implanted working monkeys. At baseline, UCCR was 10 times higher in the presenting macaque (118.1 ± 7.1) than in naïve animals (12.5 ± 12.8) and 3 times higher than in healthy implanted working macaques (44.4 ± 6.9); however, levels were suppressed similarly by dexamethasone in both the presenting animal and healthy controls. In addition, healthy implanted working macaques had significantly higher baseline UCCR levels than naïve controls, suggesting chronic stress in working animals. Abdominal ultrasonography and radiographs of the presenting animal revealed marked bilateral adrenal mineralization but no overt adrenal tumor or hyperplasia. Overall, these results excluded endogenous Cushing syndrome and prompted us to evaluate different causes of pseudo-Cushing syndrome, including depression. Using videorecordings to evaluate behavior, we used published criteria for macaque models of depression models, including huddling, to make a presumptive diagnosis of depression. The macaque was treated with fluoxetine (2 mg/kg PO daily), provided increased environmental enrichment, and followed over time by regular UCCR assessment and videorecordings. The animal improved clinically and behaviorally, and UCCR returned to levels observed in working implanted macaques (44.4) after 8 wk of treatment. This case highlights the potential effect of research-related work on stress and pathologic behaviors in macaques and demonstrates the utility of UCCR and LDDS for screening behavioral and hypothalamic-pituitary-adrenal abnormalities in these animals.


Assuntos
Síndrome de Cushing/veterinária , Depressão , Macaca mulatta , Doenças dos Macacos , Estresse Fisiológico , Glândulas Suprarrenais/diagnóstico por imagem , Alopecia/etiologia , Alopecia/veterinária , Animais , Antidepressivos de Segunda Geração/administração & dosagem , Creatinina/urina , Síndrome de Cushing/complicações , Síndrome de Cushing/psicologia , Depressão/complicações , Dexametasona/sangue , Diagnóstico Diferencial , Fluoxetina/administração & dosagem , Hidrocortisona/urina , Masculino , Doenças dos Macacos/psicologia , Radiografia/veterinária , Ultrassonografia/veterinária
10.
Proc Natl Acad Sci U S A ; 114(44): 11739-11744, 2017 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-29078378

RESUMO

Populations of circulating immune cells are maintained in equilibrium through signals that enhance the retention or egress of hematopoietic stem cells (HSCs) from bone marrow (BM). Prostaglandin E2 (PGE2) stimulates HSC renewal and engraftment through, for example, induction of the cAMP pathway. Triggering of PGE2 receptors increases HSC survival in part via the PKA-mediated induction of the cAMP response element-binding protein (CREB) signaling pathway. PKA stimulates cellular gene expression by phosphorylating CREB at Ser133 and by promoting the dephosphorylation of the cAMP- responsive transcriptional coactivators (CRTCs). We show here that disruption of both CRTC2 and CRTC3 causes embryonic lethality, and that a single allele of either CRTC2 or CRTC3 is sufficient for viability. CRTC2 knockout mice that express one CRTC3 allele (CRTC2/3m mice) develop neutrophilia and splenomegaly in adulthood due to the up-regulation of granulocyte-colony stimulating factor (G-CSF); these effects are reversed following administration of neutralizing anti-G-CSF antiserum. Adoptive transfer of CRTC2/3m BM conferred the splenomegaly/neutrophilia phenotype in WT recipients. Targeted disruption of both CRTC2 and CRTC3 in stromal cells with a mesenchymal Prx1-Cre transgene also promoted this phenotype. Depletion of CRTC2/3 was found to decrease the expression of Suppressor of Cytokine Signaling 3 (SOCS3), leading to increases in STAT3 phosphorylation and to the induction of CEBPß, a key regulator of the G-CSF gene. As small molecule inhibition of JAK activity disrupted CEBPß induction and reduced G-CSF expression in CRTC2/3m stromal cells, our results demonstrate how cross-coupling between the CREB/CRTC and JAK/STAT pathways contributes to BM homeostasis.


Assuntos
Medula Óssea/fisiologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Hematopoese/fisiologia , Fatores de Transcrição/metabolismo , Animais , Transplante de Medula Óssea , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fator Estimulador de Colônias de Granulócitos/metabolismo , Janus Quinases/genética , Janus Quinases/metabolismo , Camundongos , Camundongos Knockout , NF-kappa B/genética , NF-kappa B/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Fatores de Transcrição/genética
11.
Proc Natl Acad Sci U S A ; 114(5): 1129-1134, 2017 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-28096419

RESUMO

A fibroinflammatory stromal reaction cooperates with oncogenic signaling to influence pancreatic ductal adenocarcinoma (PDAC) initiation, progression, and therapeutic outcome, yet the mechanistic underpinning of this crosstalk remains poorly understood. Here we show that stromal cues elicit an adaptive response in the cancer cell including the rapid mobilization of a transcriptional network implicated in accelerated growth, along with anabolic changes of an altered metabolome. The close overlap of stroma-induced changes in vitro with those previously shown to be regulated by oncogenic Kras in vivo suggests that oncogenic Kras signaling-a hallmark and key driver of PDAC-is contingent on stromal inputs. Mechanistically, stroma-activated cancer cells show widespread increases in histone acetylation at transcriptionally enhanced genes, implicating the PDAC epigenome as a presumptive point of convergence between these pathways and a potential therapeutic target. Notably, inhibition of the bromodomain and extraterminal (BET) family of epigenetic readers, and of Bromodomain-containing protein 2 (BRD2) in particular, blocks stroma-inducible transcriptional regulation in vitro and tumor progression in vivo. Our work suggests the existence of a molecular "AND-gate" such that tumor activation is the consequence of mutant Kras and stromal cues, providing insight into the role of the tumor microenvironment in the origin and treatment of Ras-driven tumors.


Assuntos
Carcinoma Ductal Pancreático/fisiopatologia , Fibroblastos/fisiologia , Regulação Neoplásica da Expressão Gênica/genética , Código das Histonas , Metaboloma , Neoplasias Pancreáticas/fisiopatologia , Células Estromais/fisiologia , Microambiente Tumoral/fisiologia , Acetilação , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Citocinas/metabolismo , Metabolismo Energético , Elementos Facilitadores Genéticos , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/fisiologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Células Estreladas do Pâncreas/fisiologia , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/fisiologia , Fatores de Transcrição , Células Tumorais Cultivadas
12.
Proc Natl Acad Sci U S A ; 112(51): 15713-8, 2015 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-26644586

RESUMO

Liver fibrosis is characterized by the persistent deposition of extracellular matrix components by hepatic stellate cell (HSC)-derived myofibroblasts. It is the histological manifestation of progressive, but reversible wound-healing processes. An unabated fibrotic response results in chronic liver disease and cirrhosis, a pathological precursor of hepatocellular carcinoma. We report here that JQ1, a small molecule inhibitor of bromodomain-containing protein 4 (BRD4), a member of bromodomain and extraterminal (BET) proteins, abrogate cytokine-induced activation of HSCs. Cistromic analyses reveal that BRD4 is highly enriched at enhancers associated with genes involved in multiple profibrotic pathways, where BRD4 is colocalized with profibrotic transcription factors. Furthermore, we show that JQ1 is not only protective, but can reverse the fibrotic response in carbon tetrachloride-induced fibrosis in mouse models. Our results implicate that BRD4 can act as a global genomic regulator to direct the fibrotic response through its coordinated regulation of myofibroblast transcription. This suggests BRD4 as a potential therapeutic target for patients with fibrotic complications.


Assuntos
Cirrose Hepática Experimental/tratamento farmacológico , Proteínas Nucleares/fisiologia , Fatores de Transcrição/fisiologia , Animais , Azepinas/farmacologia , Azepinas/uso terapêutico , Células Cultivadas , Células Estreladas do Fígado/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/antagonistas & inibidores , Fatores de Transcrição/antagonistas & inibidores , Triazóis/farmacologia , Triazóis/uso terapêutico
13.
Nature ; 528(7580): 137-41, 2015 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-26580014

RESUMO

Age-associated insulin resistance (IR) and obesity-associated IR are two physiologically distinct forms of adult-onset diabetes. While macrophage-driven inflammation is a core driver of obesity-associated IR, the underlying mechanisms of the obesity-independent yet highly prevalent age-associated IR are largely unexplored. Here we show, using comparative adipo-immune profiling in mice, that fat-resident regulatory T cells, termed fTreg cells, accumulate in adipose tissue as a function of age, but not obesity. Supporting the existence of two distinct mechanisms underlying IR, mice deficient in fTreg cells are protected against age-associated IR, yet remain susceptible to obesity-associated IR and metabolic disease. By contrast, selective depletion of fTreg cells via anti-ST2 antibody treatment increases adipose tissue insulin sensitivity. These findings establish that distinct immune cell populations within adipose tissue underlie ageing- and obesity-associated IR, and implicate fTreg cells as adipo-immune drivers and potential therapeutic targets in the treatment of age-associated IR.


Assuntos
Tecido Adiposo/citologia , Tecido Adiposo/imunologia , Envelhecimento/imunologia , Resistência à Insulina/imunologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Animais , Diabetes Mellitus Tipo 2/metabolismo , Glucose/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Macrófagos/imunologia , Masculino , Síndrome Metabólica/imunologia , Síndrome Metabólica/metabolismo , Camundongos , Obesidade/metabolismo
14.
Science ; 350(6260): 558-63, 2015 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-26516283

RESUMO

Infections and inflammation can lead to cachexia and wasting of skeletal muscle and fat tissue by as yet poorly understood mechanisms. We observed that gut colonization of mice by a strain of Escherichia coli prevents wasting triggered by infections or physical damage to the intestine. During intestinal infection with the pathogen Salmonella Typhimurium or pneumonic infection with Burkholderia thailandensis, the presence of this E. coli did not alter changes in host metabolism, caloric uptake, or inflammation but instead sustained signaling of the insulin-like growth factor 1/phosphatidylinositol 3-kinase/AKT pathway in skeletal muscle, which is required for prevention of muscle wasting. This effect was dependent on engagement of the NLRC4 inflammasome. Therefore, this commensal promotes tolerance to diverse diseases.


Assuntos
Escherichia coli/imunologia , Inflamassomos/imunologia , Fator de Crescimento Insulin-Like I/metabolismo , Intestinos/microbiologia , Microbiota , Músculo Esquelético/metabolismo , Síndrome de Emaciação/imunologia , Síndrome de Emaciação/microbiologia , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Vias Biossintéticas , Burkholderia , Infecções por Burkholderia/complicações , Proteínas de Ligação ao Cálcio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Infecções por Salmonella/complicações , Salmonella typhimurium , Síndrome de Emaciação/etiologia
15.
Cell Metab ; 21(4): 628-36, 2015 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-25863252

RESUMO

Neurons utilize mitochondrial oxidative phosphorylation (OxPhos) to generate energy essential for survival, function, and behavioral output. Unlike most cells that burn both fat and sugar, neurons only burn sugar. Despite its importance, how neurons meet the increased energy demands of complex behaviors such as learning and memory is poorly understood. Here we show that the estrogen-related receptor gamma (ERRγ) orchestrates the expression of a distinct neural gene network promoting mitochondrial oxidative metabolism that reflects the extraordinary neuronal dependence on glucose. ERRγ(-/-) neurons exhibit decreased metabolic capacity. Impairment of long-term potentiation (LTP) in ERRγ(-/-) hippocampal slices can be fully rescued by the mitochondrial OxPhos substrate pyruvate, functionally linking the ERRγ knockout metabolic phenotype and memory formation. Consistent with this notion, mice lacking neuronal ERRγ in cerebral cortex and hippocampus exhibit defects in spatial learning and memory. These findings implicate neuronal ERRγ in the metabolic adaptations required for memory formation.


Assuntos
Hipocampo/fisiologia , Potenciação de Longa Duração/fisiologia , Mitocôndrias/metabolismo , Neurônios/metabolismo , Receptores de Estrogênio/metabolismo , Análise de Variância , Animais , Imunoprecipitação da Cromatina , Galactosídeos , Técnicas de Inativação de Genes , Glicólise/fisiologia , Hipocampo/metabolismo , Indóis , Memória/fisiologia , Camundongos , Análise em Microsséries , Ácido Pirúvico , Reação em Cadeia da Polimerase em Tempo Real , Aprendizagem Espacial/fisiologia
17.
Mol Cell Biol ; 35(7): 1281-98, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25624346

RESUMO

Almost all cellular functions are powered by a continuous energy supply derived from cellular metabolism. However, it is little understood how cellular energy production is coordinated with diverse energy-consuming cellular functions. Here, using the cardiac muscle system, we demonstrate that nuclear receptors estrogen-related receptor α (ERRα) and ERRγ are essential transcriptional coordinators of cardiac energy production and consumption. On the one hand, ERRα and ERRγ together are vital for intact cardiomyocyte metabolism by directly controlling expression of genes important for mitochondrial functions and dynamics. On the other hand, ERRα and ERRγ influence major cardiomyocyte energy consumption functions through direct transcriptional regulation of key contraction, calcium homeostasis, and conduction genes. Mice lacking both ERRα and cardiac ERRγ develop severe bradycardia, lethal cardiomyopathy, and heart failure featuring metabolic, contractile, and conduction dysfunctions. These results illustrate that the ERR transcriptional pathway is essential to couple cellular energy metabolism with energy consumption processes in order to maintain normal cardiac function.


Assuntos
Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Receptores de Estrogênio/metabolismo , Animais , Cardiomiopatias/genética , Cardiomiopatias/fisiopatologia , Células Cultivadas , Feminino , Deleção de Genes , Regulação da Expressão Gênica , Coração/fisiologia , Coração/fisiopatologia , Canais Iônicos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Receptores de Estrogênio/genética , Receptor ERRalfa Relacionado ao Estrogênio
18.
Cell ; 158(4): 734-748, 2014 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-25126782

RESUMO

The homeostasis of multicellular organisms requires terminally differentiated cells to preserve their lineage specificity. However, it is unclear whether mechanisms exist to actively protect cell identity in response to environmental cues that confer functional plasticity. Regulatory T (Treg) cells, specified by the transcription factor Foxp3, are indispensable for immune system homeostasis. Here, we report that conserved noncoding sequence 2 (CNS2), a CpG-rich Foxp3 intronic cis-element specifically demethylated in mature Tregs, helps maintain immune homeostasis and limit autoimmune disease development by protecting Treg identity in response to signals that shape mature Treg functions and drive their initial differentiation. In activated Tregs, CNS2 helps protect Foxp3 expression from destabilizing cytokine conditions by sensing TCR/NFAT activation, which facilitates the interaction between CNS2 and Foxp3 promoter. Thus, epigenetically marked cis-elements can protect cell identity by sensing key environmental cues central to both cell identity formation and functional plasticity without interfering with initial cell differentiation.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Linfócitos T Reguladores/citologia , Animais , Sequência de Bases , Diferenciação Celular , Sequência Conservada , Ilhas de CpG , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Expressão Gênica , Transtornos Linfoproliferativos/imunologia , Transtornos Linfoproliferativos/metabolismo , Camundongos , Dados de Sequência Molecular , Sequências Reguladoras de Ácido Nucleico , Linfócitos T Reguladores/metabolismo
19.
Comp Med ; 63(4): 367-72, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24209974

RESUMO

We report a case of brain abscess after craniotomy and the placement of a recording chamber for electrophysiologic records in an adult rhesus macaque (Macaca mulatta) enrolled in visual research. Approximately 2 wk after surgery, the macaque presented with nonspecific gastrointestinal signs and showed no evidence of fever, neurologic deficits, increased intracranial pressure, suggestive alterations in the CBC, or abnormal changes in the recording chamber. The macaque responded to symptomatic and antibiotic treatment and showed no behavioral or abnormal clinical signs for 3 wk before collapsing suddenly. The macaque was euthanized, and pathologic evaluation revealed a large brain abscess immediately under the original craniotomy.


Assuntos
Abscesso Encefálico/veterinária , Craniotomia/veterinária , Implantes Experimentais/efeitos adversos , Doenças dos Macacos/patologia , Animais , Encéfalo/microbiologia , Encéfalo/patologia , Abscesso Encefálico/etiologia , Abscesso Encefálico/patologia , Craniotomia/efeitos adversos , Implantes Experimentais/microbiologia , Macaca mulatta , Doenças dos Macacos/microbiologia
20.
Proc Natl Acad Sci U S A ; 110(17): 6991-6, 2013 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-23559372

RESUMO

Wnt signaling in mouse mammary development and tumorigenesis has been heavily studied and characterized, but its role in human breast cancer remains elusive. Although Wnt inhibitors are in early clinical development, it is unclear whether they will be of therapeutic benefit to breast cancer patients, and subsequently, to which ones. To address this, we generated a panel of Wnt reporting human breast cancer cell lines and identified a previously unrecognized enrichment for the ability to respond to Wnt in the basal B or claudin-low subtype, which has a poor prognosis and no available targeted therapies. By co-injecting Wnt3A expressing human mammary fibroblasts with human breast cancer cell lines into mouse mammary fat pads, we showed that elevated paracrine Wnt signaling was correlated with accelerated tumor growth. Using this heterotypic system and a dual lentiviral reporter system that enables simultaneous real-time measurement of both Wnt-responsive cells and bulk tumor cells, we analyzed the outcome of elevated Wnt signaling in patient-derived xenograft (PDX) models. Interestingly, the PDX models exhibited responses not observed in the cell lines analyzed. Exogenous WNT3A promoted tumor growth in one human epidermal growth factor receptor 2-overexpressing PDX line but inhibited growth in a second PDX line obtained from a patient with triple-negative breast cancer. Tumor suppression was associated with squamous differentiation in the latter. Thus, our work suggests that paracrine Wnt signaling can either fuel or repress the growth of human breast cancers depending on yet to be determined aspects of the molecular pathways they express.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/fisiopatologia , Comunicação Parácrina/fisiologia , Via de Sinalização Wnt/fisiologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Fibroblastos/metabolismo , Fibroblastos/transplante , Humanos , Imuno-Histoquímica , Luciferases , Proteínas Luminescentes , Camundongos , Camundongos SCID , Receptor ErbB-2/metabolismo , Fatores de Tempo , Transplante Heterólogo , Proteína Wnt3A/metabolismo , Proteína Vermelha Fluorescente
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA