Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-38077056

RESUMO

Under chronic stress, cells must balance competing demands between cellular survival and tissue function. In metabolic dysfunction-associated steatotic liver disease (MASLD, formerly NAFLD/NASH), hepatocytes cooperate with structural and immune cells to perform crucial metabolic, synthetic, and detoxification functions despite nutrient imbalances. While prior work has emphasized stress-induced drivers of cell death, the dynamic adaptations of surviving cells and their functional repercussions remain unclear. Namely, we do not know which pathways and programs define cellular responses, what regulatory factors mediate (mal)adaptations, and how this aberrant activity connects to tissue-scale dysfunction and long-term disease outcomes. Here, by applying longitudinal single-cell multi -omics to a mouse model of chronic metabolic stress and extending to human cohorts, we show that stress drives survival-linked tradeoffs and metabolic rewiring, manifesting as shifts towards development-associated states in non-transformed hepatocytes with accompanying decreases in their professional functionality. Diet-induced adaptations occur significantly prior to tumorigenesis but parallel tumorigenesis-induced phenotypes and predict worsened human cancer survival. Through the development of a multi -omic computational gene regulatory inference framework and human in vitro and mouse in vivo genetic perturbations, we validate transcriptional (RELB, SOX4) and metabolic (HMGCS2) mediators that co-regulate and couple the balance between developmental state and hepatocyte functional identity programming. Our work defines cellular features of liver adaptation to chronic stress as well as their links to long-term disease outcomes and cancer hallmarks, unifying diverse axes of cellular dysfunction around core causal mechanisms.

2.
Nat Commun ; 12(1): 4995, 2021 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-34404785

RESUMO

A cell's phenotype and function are influenced by dynamic interactions with its microenvironment. To examine cellular spatiotemporal activity, we developed SPACECAT-Spatially PhotoActivatable Color Encoded Cell Address Tags-to annotate, track, and isolate cells while preserving viability. In SPACECAT, samples are stained with photocaged fluorescent molecules, and cells are labeled by uncaging those molecules with user-patterned near-UV light. SPACECAT offers single-cell precision and temporal stability across diverse cell and tissue types. Illustratively, we target crypt-like regions in patient-derived intestinal organoids to enrich for stem-like and actively mitotic cells, matching literature expectations. Moreover, we apply SPACECAT to ex vivo tissue sections from four healthy organs and an autochthonous lung tumor model. Lastly, we provide a computational framework to identify spatially-biased transcriptome patterns and enriched phenotypes. This minimally perturbative and broadly applicable method links cellular spatiotemporal and/or behavioral phenotypes with diverse downstream assays, enabling insights into the connections between tissue microenvironments and (dys)function.


Assuntos
Rastreamento de Células/psicologia , Corantes , Transcriptoma , Animais , Bioensaio , Citocinas , Feminino , Fluoresceínas , Corantes Fluorescentes , Células HEK293 , Nível de Saúde , Humanos , Neoplasias Pulmonares , Masculino , Camundongos , Células Mieloides , Organoides , Fenótipo , Células-Tronco , Microambiente Tumoral , Raios Ultravioleta
3.
Cell Rep ; 35(10): 109212, 2021 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-34107251

RESUMO

Obesity is an established risk factor for cancer in many tissues. In the mammalian intestine, a pro-obesity high-fat diet (HFD) promotes regeneration and tumorigenesis by enhancing intestinal stem cell (ISC) numbers, proliferation, and function. Although PPAR (peroxisome proliferator-activated receptor) nuclear receptor activity has been proposed to facilitate these effects, their exact role is unclear. Here we find that, in loss-of-function in vivo models, PPARα and PPARδ contribute to the HFD response in ISCs. Mechanistically, both PPARs do so by robustly inducing a downstream fatty acid oxidation (FAO) metabolic program. Pharmacologic and genetic disruption of CPT1A (the rate-controlling enzyme of mitochondrial FAO) blunts the HFD phenotype in ISCs. Furthermore, inhibition of CPT1A dampens the pro-tumorigenic consequences of a HFD on early tumor incidence and progression. These findings demonstrate that inhibition of a HFD-activated FAO program creates a therapeutic opportunity to counter the effects of a HFD on ISCs and intestinal tumorigenesis.


Assuntos
Carcinogênese/patologia , Dieta Hiperlipídica/efeitos adversos , Ácidos Graxos/metabolismo , Intestinos/patologia , Obesidade/fisiopatologia , PPAR alfa/metabolismo , Células-Tronco/metabolismo , Animais , Humanos , Camundongos , Oxirredução
4.
Mol Syst Biol ; 15(8): e8849, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31464373

RESUMO

Obesity-associated type 2 diabetes and accompanying diseases have developed into a leading human health risk across industrialized and developing countries. The complex molecular underpinnings of how lipid overload and lipid metabolites lead to the deregulation of metabolic processes are incompletely understood. We assessed hepatic post-translational alterations in response to treatment of cells with saturated and unsaturated free fatty acids and the consumption of a high-fat diet by mice. These data revealed widespread tyrosine phosphorylation changes affecting a large number of enzymes involved in metabolic processes as well as canonical receptor-mediated signal transduction networks. Targeting two of the most prominently affected molecular features in our data, SRC-family kinase activity and elevated reactive oxygen species, significantly abrogated the effects of saturated fat exposure in vitro and high-fat diet in vivo. In summary, we present a comprehensive view of diet-induced alterations of tyrosine signaling networks, including proteins involved in fundamental metabolic pathways.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Dieta Hiperlipídica/efeitos adversos , Fígado/efeitos dos fármacos , Obesidade/metabolismo , Fosfotirosina/metabolismo , Processamento de Proteína Pós-Traducional , Animais , Linhagem Celular Tumoral , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Modelos Animais de Doenças , Ácidos Graxos/farmacologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Hepatócitos/patologia , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Obesidade/etiologia , Obesidade/genética , Obesidade/patologia , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteômica/métodos , Ratos , Espécies Reativas de Oxigênio/agonistas , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Quinases da Família src/genética , Quinases da Família src/metabolismo
5.
Am J Physiol Cell Physiol ; 317(3): C405-C419, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31216420

RESUMO

In vitro, cell cultures are essential tools in the study of intestinal function and disease. For the past few decades, monolayer cellular cultures, such as cancer cell lines or immortalized cell lines, have been widely applied in gastrointestinal research. Recently, the development of three-dimensional cultures known as organoids has permitted the growth of normal crypt-villus units that recapitulate many aspects of intestinal physiology. Organoid culturing has also been applied to study gastrointestinal diseases, intestinal-microbe interactions, and colorectal cancer. These models are amenable to CRISPR gene editing and drug treatments, including high-throughput small-molecule testing. Three-dimensional intestinal cultures have been transplanted into mice to develop versatile in vivo models of intestinal disease, particularly cancer. Limitations of currently available organoid models include cost and challenges in modeling nonepithelial intestinal cells, such as immune cells and the microbiota. Here, we describe the development of organoid models of intestinal biology and the applications of organoids for study of the pathophysiology of intestinal diseases and cancer.


Assuntos
Gastroenteropatias/patologia , Trato Gastrointestinal/patologia , Trato Gastrointestinal/fisiologia , Organoides/patologia , Organoides/fisiologia , Animais , Células Cultivadas , Gastroenteropatias/fisiopatologia , Neoplasias Gastrointestinais/patologia , Neoplasias Gastrointestinais/fisiopatologia , Trato Gastrointestinal/fisiopatologia , Humanos , Mucosa Intestinal/patologia , Mucosa Intestinal/fisiologia , Mucosa Intestinal/fisiopatologia , Organoides/fisiopatologia
6.
Nature ; 560(7717): E26, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29849139

RESUMO

In Fig. 4e of this Article, the labels for 'Control' and 'HFD' were reversed ('Control' should have been labelled blue rather than purple, and 'HFD' should have been labelled purple rather than blue). Similarly, in Fig. 4f of this Article, the labels for 'V' and 'GW' were reversed ('V' should have been labelled blue rather than purple, and 'GW' should have been labelled purple instead of blue). The original figure has been corrected online.

7.
Curr Stem Cell Rep ; 3(1): 1-8, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28966904

RESUMO

PURPOSE OF REVIEW: Dietary intake is a critical regulator of organismal physiology and health. Tissue homeostasis and regeneration are dependent on adult tissue stem cells that self-renew and differentiate into the specialized cell types. As stem cells respond to cues from their environment, dietary signals and nutrients influence tissue biology by altering the function and activity of adult stem cells. In this review, we highlight recent studies that illustrate how diverse diets such as caloric restriction, fasting, high fat diets, and ketogenic diets impact stem cell function and their microenvironments. RECENT FINDINGS: Caloric restriction generally exerts positive effects on adult stem cells, notably increasing stem cell functionality in the intestine and skeletal muscle as well as increasing hematopoietic stem cell quiescence. Similarly, fasting confers protection of intestinal, hematopoietic, and neuronal stem cells against injury. High fat diets induce intestinal stem cell niche independence and stem-like properties in intestinal progenitors, while high fat diets impair hematopoiesis and neurogenesis. SUMMARY: Caloric restriction and fasting are generally beneficial to adult stem cell function, while high fat diets impair stem cell function or create opportunities for tumorigenesis. However, the effects of each diet on stem cell biology are complex and vary greatly between tissues. Given the recent interest in developing dietary interventions or mimetics as therapeutics, further studies, including on ketogenic diets, will be essential to understand how adult stem cells respond to diet-induced signals and physiology.

8.
Cell Stem Cell ; 19(2): 205-216, 2016 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-27184401

RESUMO

The E3 ubiquitin ligase Mule is often overexpressed in human colorectal cancers, but its role in gut tumorigenesis is unknown. Here, we show in vivo that Mule controls murine intestinal stem and progenitor cell proliferation by modulating Wnt signaling via c-Myc. Mule also regulates protein levels of the receptor tyrosine kinase EphB3 by targeting it for proteasomal and lysosomal degradation. In the intestine, EphB/ephrinB interactions position cells along the crypt-villus axis and compartmentalize incipient colorectal tumors. Our study thus unveils an important new avenue by which Mule acts as an intestinal tumor suppressor by regulation of the intestinal stem cell niche.


Assuntos
Efrina-B3/metabolismo , Intestinos/citologia , Lisossomos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Nicho de Células-Tronco , Ubiquitina-Proteína Ligases/metabolismo , Via de Sinalização Wnt , Polipose Adenomatosa do Colo/patologia , Alelos , Animais , Carcinogênese/metabolismo , Carcinogênese/patologia , Proliferação de Células , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Endocitose , Células HEK293 , Humanos , Camundongos Knockout , Modelos Biológicos , Mutação/genética , Celulas de Paneth/patologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Supressoras de Tumor , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/deficiência
9.
Nature ; 531(7592): 53-8, 2016 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-26935695

RESUMO

Little is known about how pro-obesity diets regulate tissue stem and progenitor cell function. Here we show that high-fat diet (HFD)-induced obesity augments the numbers and function of Lgr5(+) intestinal stem cells of the mammalian intestine. Mechanistically, a HFD induces a robust peroxisome proliferator-activated receptor delta (PPAR-δ) signature in intestinal stem cells and progenitor cells (non-intestinal stem cells), and pharmacological activation of PPAR-δ recapitulates the effects of a HFD on these cells. Like a HFD, ex vivo treatment of intestinal organoid cultures with fatty acid constituents of the HFD enhances the self-renewal potential of these organoid bodies in a PPAR-δ-dependent manner. Notably, HFD- and agonist-activated PPAR-δ signalling endow organoid-initiating capacity to progenitors, and enforced PPAR-δ signalling permits these progenitors to form in vivo tumours after loss of the tumour suppressor Apc. These findings highlight how diet-modulated PPAR-δ activation alters not only the function of intestinal stem and progenitor cells, but also their capacity to initiate tumours.


Assuntos
Transformação Celular Neoplásica/efeitos dos fármacos , Neoplasias do Colo/patologia , Dieta Hiperlipídica/efeitos adversos , Intestinos/patologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/patologia , Animais , Contagem de Células , Autorrenovação Celular/efeitos dos fármacos , Feminino , Genes APC , Humanos , Masculino , Camundongos , Obesidade/induzido quimicamente , Obesidade/patologia , Organoides/efeitos dos fármacos , Organoides/metabolismo , Organoides/patologia , PPAR delta/metabolismo , Transdução de Sinais/efeitos dos fármacos , Nicho de Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , beta Catenina/metabolismo
10.
Magn Reson Med ; 74(6): 1750-7, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25522343

RESUMO

PURPOSE: Manganese (Mn) is an effective contrast agent and biologically active metal, which has been widely used for Mn-enhanced MRI (MEMRI). The purpose of this study was to develop and test a Mn binding protein for use as a genetic reporter for MEMRI. METHODS: The bacterial Mn-binding protein, MntR was identified as a candidate reporter protein. MntR was engineered for expression in mammalian cells, and targeted to different subcellular organelles, including the Golgi Apparatus where cellular Mn is enriched. Transfected HEK293 cells and B16 melanoma cells were tested in vitro and in vivo, using immunocytochemistry, MR imaging and relaxometry. RESULTS: Subcellular targeting of MntR to the cytosol, endoplasmic reticulum and Golgi apparatus was verified with immunocytochemistry. After targeting to the Golgi, MntR expression produced robust R1 changes and T1 contrast in cells, in vitro and in vivo. Co-expression with the divalent metal transporter DMT1, a previously described Mn-based reporter, further enhanced contrast in B16 cells in culture, but in the in vivo B16 tumor model tested was not significantly better than MntR alone. CONCLUSION: This second-generation reporter system both expands the capabilities of genetically encoded reporters for imaging with MEMRI and provides important insights into the mechanisms of Mn biology which create endogenous MEMRI contrast.


Assuntos
Proteínas de Bactérias/metabolismo , Genes Reporter/genética , Imageamento por Ressonância Magnética/métodos , Manganês/metabolismo , Neoplasias Experimentais/metabolismo , Proteínas Repressoras/metabolismo , Frações Subcelulares/metabolismo , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Linhagem Celular Tumoral , Meios de Contraste/metabolismo , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Sonda Molecular , Sondas Moleculares/genética , Sondas Moleculares/farmacocinética , Neoplasias Experimentais/patologia , Ligação Proteica , Engenharia de Proteínas/métodos , Proteínas Repressoras/química , Proteínas Repressoras/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA