Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Gastroenterology ; 162(2): 604-620.e20, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34695382

RESUMEN

BACKGROUND & AIMS: Acinar to ductal metaplasia (ADM) occurs in the pancreas in response to tissue injury and is a potential precursor for adenocarcinoma. The goal of these studies was to define the populations arising from ADM, the associated transcriptional changes, and markers of disease progression. METHODS: Acinar cells were lineage-traced with enhanced yellow fluorescent protein (EYFP) to follow their fate post-injury. Transcripts of more than 13,000 EYFP+ cells were determined using single-cell RNA sequencing (scRNA-seq). Developmental trajectories were generated. Data were compared with gastric metaplasia, KrasG12D-induced neoplasia, and human pancreatitis. Results were confirmed by immunostaining and electron microscopy. KrasG12D was expressed in injury-induced ADM using several inducible Cre drivers. Surgical specimens of chronic pancreatitis from 15 patients were evaluated by immunostaining. RESULTS: scRNA-seq of ADM revealed emergence of a mucin/ductal population resembling gastric pyloric metaplasia. Lineage trajectories suggest that some pyloric metaplasia cells can generate tuft and enteroendocrine cells (EECs). Comparison with KrasG12D-induced ADM identifies populations associated with disease progression. Activation of KrasG12D expression in HNF1B+ or POU2F3+ ADM populations leads to neoplastic transformation and formation of MUC5AC+ gastric-pit-like cells. Human pancreatitis samples also harbor pyloric metaplasia with a similar transcriptional phenotype. CONCLUSIONS: Under conditions of chronic injury, acinar cells undergo a pyloric-type metaplasia to mucinous progenitor-like populations, which seed disparate tuft cell and EEC lineages. ADM-derived EEC subtypes are diverse. KrasG12D expression is sufficient to drive neoplasia when targeted to injury-induced ADM populations and offers an alternative origin for tumorigenesis. This program is conserved in human pancreatitis, providing insight into early events in pancreas diseases.


Asunto(s)
Células Acinares/metabolismo , Carcinoma Ductal Pancreático/genética , Metaplasia/genética , Conductos Pancreáticos/metabolismo , Neoplasias Pancreáticas/genética , Células Acinares/citología , Plasticidad de la Célula/genética , Células Enteroendocrinas/citología , Células Enteroendocrinas/metabolismo , Perfilación de la Expresión Génica , Humanos , Metaplasia/metabolismo , Mucina 5AC/genética , Páncreas/citología , Páncreas/metabolismo , Conductos Pancreáticos/citología , Pancreatitis/genética , Pancreatitis/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Análisis de la Célula Individual
2.
Diabetologia ; 63(10): 2095-2101, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32894320

RESUMEN

Efforts to phenotype pancreatic islets have contributed tremendously to our present understanding of endocrine function and diabetes. A continued evolution in approaches to study islet physiology is important given the need to establish reference points for mature islet functionality, understanding biological variation amongst individuals and cells, and the ongoing appreciation of the role for islets in diabetes susceptibility. Recent efforts in islet biology have focused on technological improvements in imaging, molecular profiling and data analysis, along with a push for enhanced transparency and reporting. The integration of these approaches within a classical islet physiology framework, and approaches to link these data with in vivo human phenotypes, will be critical as we move towards a better understanding of islet function in health and disease. Here we discuss what we feel are important issues and useful approaches to consider as we move forward as a field in islet and beta cell phenotyping. Graphical abstract.


Asunto(s)
Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Calcio/metabolismo , Genotipo , Humanos , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/fisiología , Fenotipo , Análisis de la Célula Individual , Transcriptoma
3.
Proc Natl Acad Sci U S A ; 111(49): 17612-7, 2014 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-25404292

RESUMEN

Pancreatic islets secrete hormones that play a key role in regulating blood glucose levels (glycemia). Age-dependent impairment of islet function and concomitant dysregulation of glycemia are major health threats in aged populations. However, the major causes of the age-dependent decline of islet function are still disputed. Here we demonstrate that aging of pancreatic islets in mice and humans is notably associated with inflammation and fibrosis of islet blood vessels but does not affect glucose sensing and the insulin secretory capacity of islet beta cells. Accordingly, when transplanted into the anterior chamber of the eye of young mice with diabetes, islets from old mice are revascularized with healthy blood vessels, show strong islet cell proliferation, and fully restore control of glycemia. Our results indicate that beta cell function does not decline with age and suggest that islet function is threatened by an age-dependent impairment of islet vascular function. Strategies to mitigate age-dependent dysregulation in glycemia should therefore target systemic and/or local inflammation and fibrosis of the aged islet vasculature.


Asunto(s)
Envejecimiento , Glucemia/metabolismo , Capilares/fisiología , Islotes Pancreáticos/fisiología , Adolescente , Adulto , Anciano , Animales , Proliferación Celular , Fibrosis , Glucosa/metabolismo , Homeostasis , Humanos , Inflamación , Insulina/metabolismo , Islotes Pancreáticos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Perfusión , Factores de Tiempo , Adulto Joven
4.
Diabetologia ; 58(10): 2218-28, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26215305

RESUMEN

The human genome project and its search for factors underlying human diseases has fostered a major human research effort. Therefore, unsurprisingly, in recent years we have observed an increasing number of studies on human islet cells, including disease approaches focusing on type 1 and type 2 diabetes. Yet, the field of islet and diabetes research relies on the legacy of rodent-based investigations, which have proven difficult to translate to humans, particularly in type 1 diabetes. Whole islet physiology and pathology may differ between rodents and humans, and thus a comprehensive cross-species as well as species-specific view on islet research is much needed. In this review we summarise the current knowledge of interspecies islet cytoarchitecture, and discuss its potential impact on islet function and future perspectives in islet pathophysiology research.


Asunto(s)
Islotes Pancreáticos/anatomía & histología , Islotes Pancreáticos/fisiología , Animales , Humanos , Especificidad de la Especie
5.
Biochim Biophys Acta ; 1830(7): 3956-64, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22967761

RESUMEN

BACKGROUND: Thyroid hormone signaling is critical for development, growth and metabolic control in vertebrates. Although serum concentration of thyroid hormone is remarkable stable, deiodinases modulate thyroid hormone signaling on a time- and cell-specific fashion by controlling the activation and inactivation of thyroid hormone. SCOPE OF THE REVIEW: This review covers the recent advances in D2 biology, a member of the iodothyronine deiodinase family, thioredoxin fold-containing selenoenzymes that modify thyroid hormone signaling in a time- and cell-specific manner. MAJOR CONCLUSIONS: D2-catalyzed T3 production increases thyroid hormone signaling whereas blocking D2 activity or disruption of the Dio2 gene leads to a state of localized hypothyroidism. D2 expression is regulated by different developmental, metabolic or environmental cues such as the hedgehog pathway, the adrenergic- and the TGR5-activated cAMP pathway, by xenobiotic molecules such as flavonols and by stress in the endoplasmic reticulum, which specifically reduces de novo synthesis of D2 via an eIF2a-mediated mechanism. Thus, D2 plays a central role in important physiological processes such as determining T3 content in developing tissues and in the adult brain, and promoting adaptive thermogenesis in brown adipose tissue. Notably, D2 is critical in the T4-mediated negative feed-back at the pituitary and hypothalamic levels, whereby T4 inhibits TSH and TRH expression, respectively. Notably, ubiquitination is a major step in the control of D2 activity, whereby T4 binding to and/or T4 catalysis triggers D2 inactivation by ubiquitination that is mediated by the E3 ubiquitin ligases WSB-1 and/or TEB4. Ubiquitinated D2 can be either targeted to proteasomal degradation or reactivated by deubiquitination, a process that is mediated by the deubiquitinases USP20/33 and is important in adaptive thermogenesis. GENERAL SIGNIFICANCE: Here we review the recent advances in the understanding of D2 biology focusing on the mechanisms that regulate its expression and their biological significance in metabolically relevant tissues. This article is part of a Special Issue entitled Thyroid hormone signalling.


Asunto(s)
Yoduro Peroxidasa/metabolismo , Hormonas Tiroideas/metabolismo , Animales , Regulación del Desarrollo de la Expresión Génica , Humanos , Yoduro Peroxidasa/genética , Transducción de Señal , Hormonas Tiroideas/genética , Yodotironina Deyodinasa Tipo II
6.
J Neurosci ; 32(25): 8491-500, 2012 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-22723689

RESUMEN

In neurons, the type 3 deiodinase (D3) inactivates thyroid hormone and reduces oxygen consumption, thus creating a state of cell-specific hypothyroidism. Here we show that hypoxia leads to nuclear import of D3 in neurons, without which thyroid hormone signaling and metabolism cannot be reduced. After unilateral hypoxia in the rat brain, D3 protein level is increased predominantly in the nucleus of the neurons in the pyramidal and granular ipsilateral layers, as well as in the hilus of the dentate gyrus of the hippocampal formation. In hippocampal neurons in culture as well as in a human neuroblastoma cell line (SK-N-AS), a 24 h hypoxia period redirects active D3 from the endoplasmic reticulum to the nucleus via the cochaperone Hsp40 pathway. Preventing nuclear D3 import by Hsp40 knockdown resulted an almost doubling in the thyroid hormone-dependent glycolytic rate and quadrupling the transcription of thyroid hormone target gene ENPP2. In contrast, Hsp40 overexpression increased nuclear import of D3 and minimized thyroid hormone effects in cell metabolism. In conclusion, ischemia/hypoxia induces an Hsp40-mediated translocation of D3 to the nucleus, facilitating thyroid hormone inactivation proximal to the thyroid hormone receptors. This adaptation decreases thyroid hormone signaling and may function to reduce ischemia-induced hypoxic brain damage.


Asunto(s)
Hipoxia de la Célula/fisiología , Núcleo Celular/metabolismo , Proteínas del Choque Térmico HSP40/fisiología , Yoduro Peroxidasa/metabolismo , Neuronas/metabolismo , Animales , Isquemia Encefálica/metabolismo , Núcleo Celular/enzimología , Células Cultivadas , ADN/genética , Retículo Endoplásmico/metabolismo , Glicosilación , Hipocampo/citología , Hipocampo/metabolismo , Inmunohistoquímica , Inmunoprecipitación , Masculino , Microscopía Electrónica , Arteria Cerebral Media/fisiología , Consumo de Oxígeno/fisiología , Reacción en Cadena de la Polimerasa , Ratas , Ratas Sprague-Dawley , Receptores de Hormona Tiroidea/metabolismo , Transducción de Señal/fisiología , Hormonas Tiroideas/fisiología
7.
Diabetes ; 71(11): 2313-2330, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-35802127

RESUMEN

In the setting of obesity and insulin resistance, glycemia is controlled in part by ß-cell compensation and subsequent hyperinsulinemia. Weight loss improves glycemia and decreases hyperinsulinemia, whereas weight cycling worsens glycemic control. The mechanisms responsible for weight cycling-induced deterioration in glucose homeostasis are poorly understood. Thus, we aimed to pinpoint the main regulatory junctions at which weight cycling alters glucose homeostasis in mice. Using in vivo and ex vivo procedures we show that despite having worsened glucose tolerance, weight-cycled mice do not manifest impaired whole-body insulin action. Instead, weight cycling reduces insulin secretory capacity in vivo during clamped hyperglycemia and ex vivo in perifused islets. Islets from weight-cycled mice have reduced expression of factors essential for ß-cell function (Mafa, Pdx1, Nkx6.1, Ucn3) and lower islet insulin content, compared with those from obese mice, suggesting inadequate transcriptional and posttranscriptional response to repeated nutrient overload. Collectively, these data support a model in which pancreatic plasticity is challenged in the face of large fluctuations in body weight resulting in a mismatch between glycemia and insulin secretion in mice.


Asunto(s)
Hiperinsulinismo , Resistencia a la Insulina , Islotes Pancreáticos , Ratones , Animales , Insulina/metabolismo , Secreción de Insulina , Ciclo del Peso , Obesidad/metabolismo , Resistencia a la Insulina/fisiología , Glucemia/metabolismo , Dieta , Hiperinsulinismo/metabolismo , Insulina Regular Humana , Islotes Pancreáticos/metabolismo , Glucosa/metabolismo
8.
Sci Adv ; 8(40): eabo3932, 2022 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-36197983

RESUMEN

Pancreatic islet beta cells are essential for maintaining glucose homeostasis. To understand the impact of aging on beta cells, we performed meta-analysis of single-cell RNA sequencing datasets, transcription factor (TF) regulon analysis, high-resolution confocal microscopy, and measured insulin secretion from nondiabetic donors spanning most of the human life span. This revealed the range of molecular and functional changes that occur during beta cell aging, including the transcriptional deregulation that associates with cellular immaturity and reorganization of beta cell TF networks, increased gene transcription rates, and reduced glucose-stimulated insulin release. These alterations associate with activation of endoplasmic reticulum (ER) stress and autophagy pathways. We propose that a chronic state of ER stress undermines old beta cell structure function to increase the risk of beta cell failure and type 2 diabetes onset as humans age.

9.
Cell Metab ; 34(2): 256-268.e5, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35108513

RESUMEN

In diabetes, glucagon secretion from pancreatic α cells is dysregulated. The underlying mechanisms, and whether dysfunction occurs uniformly among cells, remain unclear. We examined α cells from human donors and mice using electrophysiological, transcriptomic, and computational approaches. Rising glucose suppresses α cell exocytosis by reducing P/Q-type Ca2+ channel activity, and this is disrupted in type 2 diabetes (T2D). Upon high-fat feeding of mice, α cells shift toward a "ß cell-like" electrophysiological profile in concert with indications of impaired identity. In human α cells we identified links between cell membrane properties and cell surface signaling receptors, mitochondrial respiratory chain complex assembly, and cell maturation. Cell-type classification using machine learning of electrophysiology data demonstrated a heterogenous loss of "electrophysiologic identity" in α cells from donors with type 2 diabetes. Indeed, a subset of α cells with impaired exocytosis is defined by an enrichment in progenitor and lineage markers and upregulation of an immature transcriptomic phenotype, suggesting important links between α cell maturation state and dysfunction.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Glucagón , Islotes Pancreáticos , Animales , Diabetes Mellitus Tipo 2/metabolismo , Exocitosis/fisiología , Glucagón/metabolismo , Células Secretoras de Glucagón/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Ratones
10.
Diabetes ; 70(10): 2163-2173, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34593534

RESUMEN

ß-Cells in the islet of Langerhans have a central role in maintaining energy homeostasis. Understanding the physiology of ß-cells and other islet cells requires a deep understanding of their structural and functional organization, their interaction with vessels and nerves, the layout of paracrine interactions, and the relationship between subcellular compartments and protein complexes inside each cell. These elements are not static; they are dynamic and exert their biological actions at different scales of time. Therefore, scientists must be able to investigate (and visualize) short- and long-lived events within the pancreas and ß-cells. Current technological advances in microscopy are able to bridge multiple spatiotemporal scales in biology to reveal the complexity and heterogeneity of ß-cell biology. Here, I briefly discuss the historical discoveries that leveraged microscopes to establish the basis of ß-cell anatomy and structure, the current imaging platforms that allow the study of islet and ß-cell biology at multiple scales of resolution, and their challenges and implications. Lastly, I outline how the remarkable longevity of structural elements at different scales in biology, from molecules to cells to multicellular structures, could represent a previously unrecognized organizational pattern in developing and adult ß-cells and pancreas biology.


Asunto(s)
Células Secretoras de Insulina/fisiología , Animales , Autorrenovación de las Células/fisiología , Supervivencia Celular/fisiología , Homeostasis , Humanos , Células Secretoras de Insulina/citología , Islotes Pancreáticos/anatomía & histología , Islotes Pancreáticos/citología , Páncreas/anatomía & histología , Páncreas/citología , Factores de Tiempo
11.
Dev Cell ; 56(21): 2952-2965.e9, 2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34715012

RESUMEN

In order to combat molecular damage, most cellular proteins undergo rapid turnover. We have previously identified large nuclear protein assemblies that can persist for years in post-mitotic tissues and are subject to age-related decline. Here, we report that mitochondria can be long lived in the mouse brain and reveal that specific mitochondrial proteins have half-lives longer than the average proteome. These mitochondrial long-lived proteins (mitoLLPs) are core components of the electron transport chain (ETC) and display increased longevity in respiratory supercomplexes. We find that COX7C, a mitoLLP that forms a stable contact site between complexes I and IV, is required for complex IV and supercomplex assembly. Remarkably, even upon depletion of COX7C transcripts, ETC function is maintained for days, effectively uncoupling mitochondrial function from ongoing transcription of its mitoLLPs. Our results suggest that modulating protein longevity within the ETC is critical for mitochondrial proteome maintenance and the robustness of mitochondrial function.


Asunto(s)
Transporte de Electrón/fisiología , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/metabolismo , Animales , Complejo I de Transporte de Electrón/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Ratones , Fosforilación Oxidativa
12.
Adv Biosyst ; 4(5): e2000044, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32402127

RESUMEN

Aging of the circulatory system correlates with the pathogenesis of a large spectrum of diseases. However, it is largely unknown which factors drive the age-dependent or pathological decline of the vasculature and how vascular defects relate to tissue aging. The goal of the study is to design a multianalytical approach to identify how the cellular microenvironment (i.e., fibroblasts) and serum from healthy donors of different ages or Alzheimer disease (AD) patients can modulate the functionality of organ-specific vascular endothelial cells (VECs). Long-living human microvascular networks embedding VECs and fibroblasts from skin biopsies are generated. RNA-seq, secretome analyses, and microfluidic assays demonstrate that fibroblasts from young donors restore the functionality of aged endothelial cells, an effect also achieved by serum from young donors. New biomarkers of vascular aging are validated in human biopsies and it is shown that young serum induces angiopoietin-like-4, which can restore compromised vascular barriers. This strategy is then employed to characterize transcriptional/functional changes induced on the blood-brain barrier by AD serum, demonstrating the importance of PTP4A3 in the regulation of permeability. Features of vascular degeneration during aging and AD are recapitulated, and a tool to identify novel biomarkers that can be exploited to develop future therapeutics modulating vascular function is established.


Asunto(s)
Envejecimiento/metabolismo , Enfermedad de Alzheimer/metabolismo , Células Endoteliales/metabolismo , Fibroblastos/metabolismo , Microvasos/metabolismo , Anciano , Femenino , Humanos , Masculino , Técnicas Analíticas Microfluídicas
13.
J Cell Biol ; 218(2): 433-444, 2019 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-30552100

RESUMEN

Many adult tissues contain postmitotic cells as old as the host organism. The only organelle that does not turn over in these cells is the nucleus, and its maintenance represents a formidable challenge, as it harbors regulatory proteins that persist throughout adulthood. Here we developed strategies to visualize two classes of such long-lived proteins, histones and nucleoporins, to understand the function of protein longevity in nuclear maintenance. Genome-wide mapping of histones revealed specific enrichment of long-lived variants at silent gene loci. Interestingly, nuclear pores are maintained by piecemeal replacement of subunits, resulting in mosaic complexes composed of polypeptides with vastly different ages. In contrast, nondividing quiescent cells remove old nuclear pores in an ESCRT-dependent manner. Our findings reveal distinct molecular strategies of nuclear maintenance, linking lifelong protein persistence to gene regulation and nuclear integrity.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Histonas/metabolismo , Mitosis/fisiología , Poro Nuclear/metabolismo , Animales , Línea Celular , Estudio de Asociación del Genoma Completo , Ratones , Factores de Tiempo
14.
Cell Metab ; 30(2): 343-351.e3, 2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31178361

RESUMEN

Most neurons are not replaced during an animal's lifetime. This nondividing state is characterized by extreme longevity and age-dependent decline of key regulatory proteins. To study the lifespans of cells and proteins in adult tissues, we combined isotope labeling of mice with a hybrid imaging method (MIMS-EM). Using 15N mapping, we show that liver and pancreas are composed of cells with vastly different ages, many as old as the animal. Strikingly, we also found that a subset of fibroblasts and endothelial cells, both known for their replicative potential, are characterized by the absence of cell division during adulthood. In addition, we show that the primary cilia of beta cells and neurons contains different structural regions with vastly different lifespans. Based on these results, we propose that age mosaicism across multiple scales is a fundamental principle of adult tissue, cell, and protein complex organization.


Asunto(s)
Envejecimiento/genética , Senescencia Celular/genética , Mosaicismo , Especificidad de Órganos/genética , Animales , Cilios/metabolismo , Células Endoteliales/metabolismo , Femenino , Fibroblastos/metabolismo , Células Secretoras de Insulina/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos , Neuronas/metabolismo , Páncreas/metabolismo
15.
Nat Commun ; 10(1): 3700, 2019 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-31420552

RESUMEN

Little is known about the role of islet delta cells in regulating blood glucose homeostasis in vivo. Delta cells are important paracrine regulators of beta cell and alpha cell secretory activity, however the structural basis underlying this regulation has yet to be determined. Most delta cells are elongated and have a well-defined cell soma and a filopodia-like structure. Using in vivo optogenetics and high-speed Ca2+ imaging, we show that these filopodia are dynamic structures that contain a secretory machinery, enabling the delta cell to reach a large number of beta cells within the islet. This provides for efficient regulation of beta cell activity and is modulated by endogenous IGF-1/VEGF-A signaling. In pre-diabetes, delta cells undergo morphological changes that may be a compensation to maintain paracrine regulation of the beta cell. Our data provides an integrated picture of how delta cells can modulate beta cell activity under physiological conditions.


Asunto(s)
Islotes Pancreáticos/ultraestructura , Comunicación Paracrina , Estado Prediabético/patología , Seudópodos/ultraestructura , Células Secretoras de Somatostatina/ultraestructura , Animales , Glucemia/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/ultraestructura , Microscopía Intravital , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Ratones , Ratones Transgénicos , Microscopía Electrónica , Imagen Óptica , Optogenética , Estado Prediabético/metabolismo , Seudópodos/metabolismo , Células Secretoras de Somatostatina/citología , Células Secretoras de Somatostatina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
Nat Med ; 24(1): 39-49, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29200204

RESUMEN

Thyroid hormone (TH) is critical for the maintenance of cellular homeostasis during stress responses, but its role in lung fibrosis is unknown. Here we found that the activity and expression of iodothyronine deiodinase 2 (DIO2), an enzyme that activates TH, were higher in lungs from patients with idiopathic pulmonary fibrosis than in control individuals and were correlated with disease severity. We also found that Dio2-knockout mice exhibited enhanced bleomycin-induced lung fibrosis. Aerosolized TH delivery increased survival and resolved fibrosis in two models of pulmonary fibrosis in mice (intratracheal bleomycin and inducible TGF-ß1). Sobetirome, a TH mimetic, also blunted bleomycin-induced lung fibrosis. After bleomycin-induced injury, TH promoted mitochondrial biogenesis, improved mitochondrial bioenergetics and attenuated mitochondria-regulated apoptosis in alveolar epithelial cells both in vivo and in vitro. TH did not blunt fibrosis in Ppargc1a- or Pink1-knockout mice, suggesting dependence on these pathways. We conclude that the antifibrotic properties of TH are associated with protection of alveolar epithelial cells and restoration of mitochondrial function and that TH may thus represent a potential therapy for pulmonary fibrosis.


Asunto(s)
Mitocondrias/fisiología , Fibrosis Pulmonar/prevención & control , Hormonas Tiroideas/fisiología , Animales , Células Cultivadas , Epitelio/fisiología , Femenino , Humanos , Yoduro Peroxidasa/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Imitación Molecular , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Proteínas Quinasas/genética , Fibrosis Pulmonar/fisiopatología , Yodotironina Deyodinasa Tipo II
17.
Endocrinology ; 148(3): 954-60, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17138654

RESUMEN

For T(3) to mediate its biological effects, the prohormone T(4) must be activated by removal of an outer-ring iodine by the type 1 or 2 deiodinases (D1 and D2) with approximately 60% of the daily T(3) production in rodents being produced extrathyroidally through this pathway. To further define the role of these enzymes in thyroid hormone homeostasis, we backcrossed the targeted disruption of the Dio2 gene into C3H/HeJ (C3H) mice with genetically low D1 expression to create the C3H-D2KO mouse. Remarkably, these mice maintain euthyroid serum T(3) levels with normal growth and no decrease in expression of hepatic T(3)-responsive genes. However, serum T(4) is increased 1.2-fold relative to the already elevated C3H levels, and serum TSH is increased 1.4-fold. Despite these increases, thyroidal (125)I uptake indicates no difference in thyroidal activity between C3H-D2KO and C3H mice. Although C3H-D2KO hepatic and renal D1 activities were well below those observed in wild-type mice (approximately 0.1-fold for both), they were 8-fold and 2-fold higher, respectively, relative to C3H mice. Thyroidal D1 and cerebral cortical type 3 deiodinase activity were unchanged between C3H-D2KO and C3H mice. In conclusion, C3H-D2KO mice have notably elevated serum T(4) levels, and this, in conjunction with residual D1 activity, is likely an important role in the maintenance of euthyroid serum T(3) concentrations.


Asunto(s)
Yoduro Peroxidasa/genética , Tiroxina/metabolismo , Triyodotironina/sangre , Triyodotironina/metabolismo , Animales , Cruzamientos Genéticos , Femenino , Yoduro Peroxidasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Yodotironina Deyodinasa Tipo II
18.
Sci Rep ; 7: 44261, 2017 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-28303903

RESUMEN

Thanks to the development of new 3D Imaging techniques, volumetric data of thick samples, especially tissues, are commonly available. Several algorithms were proposed to analyze cells or nuclei in tissues, however these tools are limited to two dimensions. Within any given tissue, cells are not likely to be organized randomly and as such have specific patterns of cell-cell interaction forming complex communication networks. In this paper, we propose a new set of tools as an approach to segment and analyze tissues in 3D with single cell resolution. This new tool box can identify and compute the geographical location of single cells and analyze the potential physical interactions between different cell types and in 3D. As a proof-of-principle, we applied our methodology to investigation of the cyto-architecture of the islets of Langerhans in mice and monkeys. The results obtained here are a significant improvement in current methodologies and provides new insight into the organization of alpha cells and their cellular interactions within the islet's cellular framework.


Asunto(s)
Algoritmos , Núcleo Celular/ultraestructura , Procesamiento de Imagen Asistido por Computador/métodos , Islotes Pancreáticos/citología , Análisis de la Célula Individual/métodos , Animales , Comunicación Celular , Núcleo Celular/metabolismo , Expresión Génica , Glucagón/genética , Glucagón/metabolismo , Haplorrinos , Procesamiento de Imagen Asistido por Computador/estadística & datos numéricos , Imagenología Tridimensional , Insulina/genética , Insulina/metabolismo , Islotes Pancreáticos/anatomía & histología , Ratones , Ratones Endogámicos C57BL , Imagen Óptica/métodos , Somatostatina/genética , Somatostatina/metabolismo
19.
Cell Rep ; 20(6): 1490-1501, 2017 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-28793270

RESUMEN

Blood flow regulation in pancreatic islets is critical for function but poorly understood. Here, we establish an in vivo imaging platform in a non-human primate where islets transplanted autologously into the anterior chamber of the eye are monitored non-invasively and longitudinally at single-cell resolution. Engrafted islets were vascularized and innervated and maintained the cytoarchitecture of in situ islets in the pancreas. Blood flow velocity in the engrafted islets was not affected by increasing blood glucose levels and/or the GLP-1R agonist liraglutide. However, islet blood flow was dynamic in nature and fluctuated in various capillaries. This was associated with vasoconstriction events resembling a sphincter-like action, most likely regulated by adrenergic signaling. These observations suggest a mechanism in primate islets that diverts blood flow to cell regions with higher metabolic demand. The described imaging technology applied in non-human primate islets may contribute to a better understanding of human islet pathophysiology.


Asunto(s)
Velocidad del Flujo Sanguíneo , Islotes Pancreáticos/irrigación sanguínea , Animales , Glucemia/metabolismo , Capilares/fisiología , Células Cultivadas , Hipoglucemiantes/farmacología , Islotes Pancreáticos/efectos de los fármacos , Liraglutida/farmacología , Macaca fascicularis , Masculino , Flujo Sanguíneo Regional , Vasoconstricción
20.
Mol Endocrinol ; 27(12): 2105-15, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24196352

RESUMEN

The type II iodothyronine deiodinase (D2) is a type I endoplasmic reticulum (ER)-resident thioredoxin fold-containing selenoprotein that activates thyroid hormone. D2 is inactivated by ER-associated ubiquitination and can be reactivated by two ubiquitin-specific peptidase-class D2-interacting deubiquitinases (DUBs). Here, we used D2-expressing cell models to define that D2 ubiquitination (UbD2) occurs via K48-linked ubiquitin chains and that exposure to its natural substrate, T4, accelerates UbD2 formation and retrotranslocation to the cytoplasm via interaction with the p97-ATPase complex. D2 retrotranslocation also includes deubiquitination by the p97-associated DUB Ataxin-3 (Atx3). Inhibiting Atx3 with eeyarestatin-I did not affect D2:p97 binding but decreased UbD2 retrotranslocation and caused ER accumulation of high-molecular weight UbD2 bands possibly by interfering with the D2-ubiquitin-specific peptidases binding. Once in the cytosol, D2 is delivered to the proteasomes as evidenced by coprecipitation with 19S proteasome subunit S5a and increased colocalization with the 20S proteasome. We conclude that interaction between UbD2 and p97/Atx3 mediates retranslocation of UbD2 to the cytoplasm for terminal degradation in the proteasomes, a pathway that is accelerated by exposure to T4.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Citoplasma/enzimología , Yoduro Peroxidasa/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Represoras/metabolismo , Ataxina-3 , Retículo Endoplásmico/metabolismo , Degradación Asociada con el Retículo Endoplásmico , Estabilidad de Enzimas , Células HEK293 , Humanos , Lisina/metabolismo , Transporte de Proteínas , Especificidad por Sustrato , Ubiquitina/metabolismo , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA