Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Adv ; 10(25): eadm9228, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38905339

RESUMEN

Transient receptor potential melastatin 8 (TRPM8) is a temperature- and menthol-sensitive ion channel that contributes to diverse physiological roles, including cold sensing and pain perception. Clinical trials targeting TRPM8 have faced repeated setbacks predominantly due to the knowledge gap in unraveling the molecular underpinnings governing polymodal activation. A better understanding of the molecular foundations between the TRPM8 activation modes may aid the development of mode-specific, thermal-neutral therapies. Ancestral sequence reconstruction was used to explore the origins of TRPM8 activation modes. By resurrecting key TRPM8 nodes along the human evolutionary trajectory, we gained valuable insights into the trafficking, stability, and function of these ancestral forms. Notably, this approach unveiled the differential emergence of cold and menthol sensitivity over evolutionary time, providing a fresh perspective on complex polymodal behavior. These studies provide a paradigm for understanding polymodal behavior in TRPM8 and other proteins with the potential to enhance our understanding of sensory receptor biology and pave the way for innovative therapeutic interventions.


Asunto(s)
Frío , Mentol , Canales Catiónicos TRPM , Canales Catiónicos TRPM/metabolismo , Canales Catiónicos TRPM/genética , Humanos , Mentol/farmacología , Evolución Molecular , Filogenia , Sensación Térmica
2.
bioRxiv ; 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-38077056

RESUMEN

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.

3.
Gastroenterology ; 164(7): 1137-1151.e15, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36871599

RESUMEN

BACKGROUND & AIMS: Fibrosis and tissue stiffening are hallmarks of inflammatory bowel disease (IBD). We have hypothesized that the increased stiffness directly contributes to the dysregulation of the epithelial cell homeostasis in IBD. Here, we aim to determine the impact of tissue stiffening on the fate and function of the intestinal stem cells (ISCs). METHODS: We developed a long-term culture system consisting of 2.5-dimensional intestinal organoids grown on a hydrogel matrix with tunable stiffness. Single-cell RNA sequencing provided stiffness-regulated transcriptional signatures of the ISCs and their differentiated progeny. YAP-knockout and YAP-overexpression mice were used to manipulate YAP expression. In addition, we analyzed colon samples from murine colitis models and human IBD samples to assess the impact of stiffness on ISCs in vivo. RESULTS: We demonstrated that increasing the stiffness potently reduced the population of LGR5+ ISCs and KI-67+-proliferating cells. Conversely, cells expressing the stem cell marker, olfactomedin-4, became dominant in the crypt-like compartments and pervaded the villus-like regions. Concomitantly, stiffening prompted the ISCs to preferentially differentiate toward goblet cells. Mechanistically, stiffening increased the expression of cytosolic YAP, driving the extension of olfactomedin-4+ cells into the villus-like regions, while it induced the nuclear translocation of YAP, leading to preferential differentiation of ISCs toward goblet cells. Furthermore, analysis of colon samples from murine colitis models and patients with IBD demonstrated cellular and molecular remodeling reminiscent of those observed in vitro. CONCLUSIONS: Collectively, our findings highlight that matrix stiffness potently regulates the stemness of ISCs and their differentiation trajectory, supporting the hypothesis that fibrosis-induced gut stiffening plays a direct role in epithelial remodeling in IBD.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Humanos , Ratones , Animales , Células Caliciformes , Células Madre/fisiología , Mucosa Intestinal/metabolismo , Diferenciación Celular/genética , Enfermedades Inflamatorias del Intestino/metabolismo , Colitis/metabolismo
5.
Nat Commun ; 12(1): 4995, 2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34404785

RESUMEN

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.


Asunto(s)
Rastreo Celular/psicología , Colorantes , Transcriptoma , Animales , Bioensayo , Citocinas , Femenino , Fluoresceínas , Colorantes Fluorescentes , Células HEK293 , Estado de Salud , Humanos , Neoplasias Pulmonares , Masculino , Ratones , Células Mieloides , Organoides , Fenotipo , Células Madre , Microambiente Tumoral , Rayos Ultravioleta
6.
Cell Rep ; 35(10): 109212, 2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-34107251

RESUMEN

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.


Asunto(s)
Carcinogénesis/patología , Dieta Alta en Grasa/efectos adversos , Ácidos Grasos/metabolismo , Intestinos/patología , Obesidad/fisiopatología , PPAR alfa/metabolismo , Células Madre/metabolismo , Animales , Humanos , Ratones , Oxidación-Reducción
8.
Clin Cancer Res ; 26(6): 1277-1287, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-31852835

RESUMEN

PURPOSE: Sessile serrated lesions (SSL) are precursors to colon carcinoma, and their distinction from other polyps, in particular hyperplastic polyps (HP), presents significant diagnostic challenges. We evaluated expression patterns in colonic polyps of previously identified colon carcinoma-associated extracellular matrix (ECM) proteins to identify markers distinguishing SSLs from other polyps. EXPERIMENTAL DESIGN: Gene-expression analyses of ECM proteins were performed using publicly available data on preneoplastic colonic polyps. In parallel, we evaluated by IHC the expression of agrin (AGRN) in over 400 colonic polyps, including HP, SSL with and without dysplasia, traditional serrated adenomas (TSA), and tubular adenomas (TA), and compared the consistency of standard histologic diagnosis of SSLs by experienced gastrointestinal pathologists with that of AGRN IHC. RESULTS: Differential gene expression analysis and IHC identified AGRN, serine peptidase inhibitor (SERPINE2), and TIMP metallopeptidase inhibitor 1 (TIMP1) elevated in SSLs and HPs but decreased in TAs and absent in normal colon. AGRN-positive basal laminae were noted in all TA, TSA, HP, and SSL in distinguishable patterns, whereas other polyps and normal mucosa were negative. SSL with or without dysplasia consistently showed IHC staining for AGRN in the muscularis mucosae, which was absent in HP, TSA, TA, and other polyps. In contrast, histologic evaluation showed only weak interobserver agreement (kappa value = 0.493) in distinguishing SSLs. CONCLUSIONS: Muscularis mucosae-based AGRN immunostaining is a novel biomarker to distinguish SSL from HP, TSA, and TA, with a specificity of 97.1% and sensitivity of 98.9% and can assist in diagnosis of morphologically challenging colonic polyps.


Asunto(s)
Agrina/metabolismo , Biomarcadores de Tumor/metabolismo , Pólipos del Colon/diagnóstico , Neoplasias Colorrectales/diagnóstico , Regulación Neoplásica de la Expresión Génica , Hiperplasia/diagnóstico , Membrana Mucosa/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Agrina/genética , Niño , Preescolar , Pólipos del Colon/genética , Pólipos del Colon/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Diagnóstico Diferencial , Humanos , Hiperplasia/genética , Hiperplasia/metabolismo , Persona de Mediana Edad , Membrana Mucosa/patología , Adulto Joven
9.
Ann N Y Acad Sci ; 1462(1): 5-13, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31792987

RESUMEN

Tumor cells have devised unique metabolic strategies to garner enough nutrients to sustain continuous growth and cell division. Oncogenic mutations may alter metabolic pathways to unlock new sources of energy, and cells take the advantage of various scavenging pathways to ingest material from their environment. These changes in metabolism result in a metabolic profile that, in addition to providing the building blocks for macromolecules, can also influence cell signaling pathways to promote tumor initiation and progression. Understanding what pathways tumor cells use to synthesize the materials necessary to support metabolic growth can pave the way for new cancer therapeutics. Potential strategies include depriving tumors of the materials needed to grow or targeting pathways involved in dependencies that arise by virtue of their altered metabolis.


Asunto(s)
Congresos como Asunto/tendencias , Metabolismo Energético/fisiología , Neoplasias/metabolismo , Informe de Investigación/tendencias , Animales , Transformación Celular Neoplásica/metabolismo , Humanos , Redes y Vías Metabólicas/fisiología , Ciudad de Nueva York
10.
Mol Syst Biol ; 15(8): e8849, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31464373

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa/efectos adversos , Hígado/efectos de los fármacos , Obesidad/metabolismo , Fosfotirosina/metabolismo , Procesamiento Proteico-Postraduccional , Animales , Línea Celular Tumoral , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Modelos Animales de Enfermedad , Ácidos Grasos/farmacología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/etiología , Obesidad/genética , Obesidad/patología , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteómica/métodos , Ratas , Especies Reactivas de Oxígeno/agonistas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
11.
Nature ; 571(7765): 398-402, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31292548

RESUMEN

A decline in stem cell function impairs tissue regeneration during ageing, but the role of the stem-cell-supporting niche in ageing is not well understood. The small intestine is maintained by actively cycling intestinal stem cells that are regulated by the Paneth cell niche1,2. Here we show that the regenerative potential of human and mouse intestinal epithelium diminishes with age owing to defects in both stem cells and their niche. The functional decline was caused by a decrease in stemness-maintaining Wnt signalling due to production of Notum, an extracellular Wnt inhibitor, in aged Paneth cells. Mechanistically, high activity of mammalian target of rapamycin complex 1 (mTORC1) in aged Paneth cells inhibits activity of peroxisome proliferator activated receptor α (PPAR-α)3, and lowered PPAR-α activity increased Notum expression. Genetic targeting of Notum or Wnt supplementation restored function of aged intestinal organoids. Moreover, pharmacological inhibition of Notum in mice enhanced the regenerative capacity of aged stem cells and promoted recovery from chemotherapy-induced damage. Our results reveal a role of the stem cell niche in ageing and demonstrate that targeting of Notum can promote regeneration of aged tissues.


Asunto(s)
Envejecimiento , Senescencia Celular , Esterasas/metabolismo , Mucosa Intestinal/patología , Células de Paneth/metabolismo , Regeneración , Envejecimiento/fisiología , Animales , Senescencia Celular/fisiología , Esterasas/antagonistas & inhibidores , Esterasas/biosíntesis , Femenino , Humanos , Mucosa Intestinal/fisiología , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , PPAR alfa/metabolismo , Células de Paneth/patología , Receptores Acoplados a Proteínas G/metabolismo , Nicho de Células Madre , Células Madre/patología , Proteínas Wnt/antagonistas & inhibidores , Vía de Señalización Wnt
12.
Am J Physiol Cell Physiol ; 317(3): C405-C419, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31216420

RESUMEN

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.


Asunto(s)
Enfermedades Gastrointestinales/patología , Tracto Gastrointestinal/patología , Tracto Gastrointestinal/fisiología , Organoides/patología , Organoides/fisiología , Animales , Células Cultivadas , Enfermedades Gastrointestinales/fisiopatología , Neoplasias Gastrointestinales/patología , Neoplasias Gastrointestinales/fisiopatología , Tracto Gastrointestinal/fisiopatología , Humanos , Mucosa Intestinal/patología , Mucosa Intestinal/fisiología , Mucosa Intestinal/fisiopatología , Organoides/fisiopatología
13.
Nature ; 560(7717): E26, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29849139

RESUMEN

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.

14.
Cell Stem Cell ; 22(5): 769-778.e4, 2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29727683

RESUMEN

Diet has a profound effect on tissue regeneration in diverse organisms, and low caloric states such as intermittent fasting have beneficial effects on organismal health and age-associated loss of tissue function. The role of adult stem and progenitor cells in responding to short-term fasting and whether such responses improve regeneration are not well studied. Here we show that a 24 hr fast augments intestinal stem cell (ISC) function in young and aged mice by inducing a fatty acid oxidation (FAO) program and that pharmacological activation of this program mimics many effects of fasting. Acute genetic disruption of Cpt1a, the rate-limiting enzyme in FAO, abrogates ISC-enhancing effects of fasting, but long-term Cpt1a deletion decreases ISC numbers and function, implicating a role for FAO in ISC maintenance. These findings highlight a role for FAO in mediating pro-regenerative effects of fasting in intestinal biology, and they may represent a viable strategy for enhancing intestinal regeneration.


Asunto(s)
Envejecimiento , Ayuno/metabolismo , Ácidos Grasos/metabolismo , Homeostasis , Intestinos/citología , Células Madre/citología , Células Madre/metabolismo , Animales , Células Cultivadas , Ratones , Ratones Endogámicos , Oxidación-Reducción
15.
Curr Stem Cell Rep ; 3(1): 1-8, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28966904

RESUMEN

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.

16.
Cell Stem Cell ; 20(6): 801-816.e7, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28285904

RESUMEN

Highly proliferative Lgr5+ stem cells maintain the intestinal epithelium and are thought to be largely homogeneous. Although quiescent intestinal stem cell (ISC) populations have been described, the identity and features of such a population remain controversial. Here we report unanticipated heterogeneity within the Lgr5+ ISC pool. We found that expression of the RNA-binding protein Mex3a labels a slowly cycling subpopulation of Lgr5+ ISCs that contribute to all intestinal lineages with distinct kinetics. Single-cell transcriptome profiling revealed that Lgr5+ cells adopt two discrete states, one of which is defined by a Mex3a expression program and relatively low levels of proliferation genes. During homeostasis, Mex3a+ cells continually shift into the rapidly dividing, self-renewing ISC pool. Chemotherapy and radiation preferentially target rapidly dividing Lgr5+ cells but spare the Mex3a-high/Lgr5+ population, helping to promote regeneration of the intestinal epithelium following toxic insults. Thus, Mex3a defines a reserve-like ISC population within the Lgr5+ compartment.


Asunto(s)
Proliferación Celular/fisiología , Mucosa Intestinal/metabolismo , Proteínas de Unión al ARN/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células Madre/metabolismo , Animales , Mucosa Intestinal/citología , Ratones , Ratones Transgénicos , Proteínas de Unión al ARN/genética , Receptores Acoplados a Proteínas G/genética , Células Madre/citología
17.
Cell Stem Cell ; 19(2): 205-216, 2016 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-27184401

RESUMEN

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.


Asunto(s)
Efrina-B3/metabolismo , Intestinos/citología , Lisosomas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Nicho de Células Madre , Ubiquitina-Proteína Ligasas/metabolismo , Vía de Señalización Wnt , Poliposis Adenomatosa del Colon/patología , Alelos , Animales , Carcinogénesis/metabolismo , Carcinogénesis/patología , Proliferación Celular , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Endocitosis , Células HEK293 , Humanos , Ratones Noqueados , Modelos Biológicos , Mutación/genética , Células de Paneth/patología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Supresoras de Tumor , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/deficiencia
18.
Nature ; 531(7592): 53-8, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-26935695

RESUMEN

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.


Asunto(s)
Transformación Celular Neoplásica/efectos de los fármacos , Neoplasias del Colon/patología , Dieta Alta en Grasa/efectos adversos , Intestinos/patología , Células Madre/efectos de los fármacos , Células Madre/patología , Animales , Recuento de Células , Autorrenovación de las Células/efectos de los fármacos , Femenino , Genes APC , Humanos , Masculino , Ratones , Obesidad/inducido químicamente , Obesidad/patología , Organoides/efectos de los fármacos , Organoides/metabolismo , Organoides/patología , PPAR delta/metabolismo , Transducción de Señal/efectos de los fármacos , Nicho de Células Madre/efectos de los fármacos , Células Madre/metabolismo , beta Catenina/metabolismo
19.
Magn Reson Med ; 74(6): 1750-7, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25522343

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Genes Reporteros/genética , Imagen por Resonancia Magnética/métodos , Manganeso/metabolismo , Neoplasias Experimentales/metabolismo , Proteínas Represoras/metabolismo , Fracciones Subcelulares/metabolismo , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Línea Celular Tumoral , Medios de Contraste/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Técnicas de Sonda Molecular , Sondas Moleculares/genética , Sondas Moleculares/farmacocinética , Neoplasias Experimentales/patología , Unión Proteica , Ingeniería de Proteínas/métodos , Proteínas Represoras/química , Proteínas Represoras/genética
20.
Proc Natl Acad Sci U S A ; 108(26): 10508-13, 2011 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-21670261

RESUMEN

We present a model of cytoplasmically driven microtubule-based pronuclear motion in the single-celled Caenorhabditis elegans embryo. In this model, a centrosome pair at the male pronucleus initiates stochastic microtubule (MT) growth. These MTs encounter motor proteins, distributed throughout the cytoplasm, that attach and exert a pulling force. The consequent MT-length-dependent pulling forces drag the pronucleus through the cytoplasm. On physical grounds, we assume that the motor proteins also exert equal and opposite forces on the surrounding viscous cytoplasm, here modeled as an incompressible Newtonian fluid constrained within an ellipsoidal eggshell. This naturally leads to streaming flows along the MTs. Our computational method is based on an immersed boundary formulation that allows for the simultaneous treatment of fluid flow and the dynamics of structures immersed within. Our simulations demonstrate that the balance of MT pulling forces and viscous nuclear drag is sufficient to move the pronucleus, while simultaneously generating minus-end directed flows along MTs that are similar to the observed movement of yolk granules toward the center of asters. Our simulations show pronuclear migration, and moreover, a robust pronuclear centration and rotation very similar to that observed in vivo. We find also that the confinement provided by the eggshell significantly affects the internal dynamics of the cytoplasm, increasing by an order of magnitude the forces necessary to translocate and center the pronucleus.


Asunto(s)
Caenorhabditis elegans/embriología , Citoplasma/fisiología , Microtúbulos/fisiología , Animales , Biofisica , Núcleo Celular/fisiología , Centrosoma , Modelos Biológicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...