Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Front Pharmacol ; 14: 1297596, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38026988

RESUMEN

Atypical chemokine receptors (ACKRs) play pivotal roles in immune regulation by binding chemokines and regulating their spatial distribution without inducing G-protein activation. Recently, GPR182, provisionally named ACKR5, was identified as a novel ACKR expressed in microvascular and lymphatic endothelial cells, with functions in hematopoietic stem cell homeostasis. Here, we comprehensively investigated the chemokine binding profile of human and mouse GPR182. Competitive binding assays using flow cytometry revealed that besides CXCL10, CXCL12 and CXCL13, also human and mouse CXCL11, CXCL14 and CCL25, as well as human CCL1, CCL11, CCL19, CCL26, XCL1 and mouse CCL22, CCL24, CCL27 and CCL28 bind with an affinity of less than 100 nM to GPR182. In line with the binding affinity observed in vitro, elevated serum levels of CCL22, CCL24, CCL25, and CCL27 were observed in GPR182-deficient mice, underscoring the role of GPR182 in chemokine scavenging. These data show a broader chemokine binding repertoire of GPR182 than previously reported and they will be important for future work exploring the physiological and pathophysiological roles of GPR182, which we propose to be renamed atypical chemokine receptor 5 (ACKR5).

2.
Nat Commun ; 14(1): 1980, 2023 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-37031213

RESUMEN

Activation of endothelial YAP/TAZ signaling is crucial for physiological and pathological angiogenesis. The mechanisms of endothelial YAP/TAZ regulation are, however, incompletely understood. Here we report that the protocadherin FAT1 acts as a critical upstream regulator of endothelial YAP/TAZ which limits the activity of these transcriptional cofactors during developmental and tumor angiogenesis by promoting their degradation. We show that loss of endothelial FAT1 results in increased endothelial cell proliferation in vitro and in various angiogenesis models in vivo. This effect is due to perturbed YAP/TAZ protein degradation, leading to increased YAP/TAZ protein levels and expression of canonical YAP/TAZ target genes. We identify the E3 ubiquitin ligase Mind Bomb-2 (MIB2) as a FAT1-interacting protein mediating FAT1-induced YAP/TAZ ubiquitination and degradation. Loss of MIB2 expression in endothelial cells in vitro and in vivo recapitulates the effects of FAT1 depletion and causes decreased YAP/TAZ degradation and increased YAP/TAZ signaling. Our data identify a pivotal mechanism of YAP/TAZ regulation involving FAT1 and its associated E3 ligase MIB2, which is essential for YAP/TAZ-dependent angiogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Transactivadores , Humanos , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteolisis , Transactivadores/metabolismo , Células Endoteliales/metabolismo , Proteínas Señalizadoras YAP , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Neovascularización Patológica/metabolismo , Fosfoproteínas/metabolismo , Cadherinas/metabolismo
3.
Neuron ; 110(24): 4074-4089.e6, 2022 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-36549270

RESUMEN

How the vascular and neural compartment cooperate to achieve such a complex and highly specialized structure as the central nervous system is still unclear. Here, we reveal a crosstalk between motor neurons (MNs) and endothelial cells (ECs), necessary for the coordinated development of MNs. By analyzing cell-to-cell interaction profiles of the mouse developing spinal cord, we uncovered semaphorin 3C (Sema3C) and PlexinD1 as a communication axis between MNs and ECs. Using cell-specific knockout mice and in vitro assays, we demonstrate that removal of Sema3C in MNs, or its receptor PlexinD1 in ECs, results in premature and aberrant vascularization of MN columns. Those vascular defects impair MN axon exit from the spinal cord. Impaired PlexinD1 signaling in ECs also causes MN maturation defects at later stages. This study highlights the importance of a timely and spatially controlled communication between MNs and ECs for proper spinal cord development.


Asunto(s)
Células Endoteliales , Neuronas Motoras , Animales , Ratones , Neuronas Motoras/fisiología , Médula Espinal , Transducción de Señal , Axones , Ratones Noqueados
5.
Nat Commun ; 12(1): 6798, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34815397

RESUMEN

G-protein-coupled receptors (GPCRs), especially chemokine receptors, play a central role in the regulation of T cell migration. Various GPCRs are upregulated in activated CD4 T cells, including P2Y10, a putative lysophospholipid receptor that is officially still considered an orphan GPCR, i.e., a receptor with unknown endogenous ligand. Here we show that in mice lacking P2Y10 in the CD4 T cell compartment, the severity of experimental autoimmune encephalomyelitis and cutaneous contact hypersensitivity is reduced. P2Y10-deficient CD4 T cells show normal activation, proliferation and differentiation, but reduced chemokine-induced migration, polarization, and RhoA activation upon in vitro stimulation. Mechanistically, CD4 T cells release the putative P2Y10 ligands lysophosphatidylserine and ATP upon chemokine exposure, and these mediators induce P2Y10-dependent RhoA activation in an autocrine/paracrine fashion. ATP degradation impairs RhoA activation and migration in control CD4 T cells, but not in P2Y10-deficient CD4 T cells. Importantly, the P2Y10 pathway appears to be conserved in human T cells. Taken together, P2Y10 mediates RhoA activation in CD4 T cells in response to auto-/paracrine-acting mediators such as LysoPS and ATP, thereby facilitating chemokine-induced migration and, consecutively, T cell-mediated diseases.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Esclerosis Múltiple/inmunología , Receptores Purinérgicos P2Y/metabolismo , Receptores Purinérgicos P2/metabolismo , Adenosina Trifosfato/metabolismo , Adulto , Anciano , Animales , Comunicación Autocrina/inmunología , Linfocitos T CD4-Positivos/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Quimiocinas/metabolismo , Quimiotaxis de Leucocito/inmunología , Encefalomielitis Autoinmune Experimental/sangre , Femenino , Técnicas de Silenciamiento del Gen , Técnicas de Inactivación de Genes , Humanos , Lisofosfolípidos/metabolismo , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Esclerosis Múltiple/sangre , Comunicación Paracrina/inmunología , Cultivo Primario de Células , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2Y/genética , Proteína de Unión al GTP rhoA/metabolismo
6.
Proc Natl Acad Sci U S A ; 118(17)2021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33875597

RESUMEN

G protein-coupled receptor 182 (GPR182) has been shown to be expressed in endothelial cells; however, its ligand and physiological role has remained elusive. We found GPR182 to be expressed in microvascular and lymphatic endothelial cells of most organs and to bind with nanomolar affinity the chemokines CXCL10, CXCL12, and CXCL13. In contrast to conventional chemokine receptors, binding of chemokines to GPR182 did not induce typical downstream signaling processes, including Gq- and Gi-mediated signaling or ß-arrestin recruitment. GPR182 showed relatively high constitutive activity in regard to ß-arrestin recruitment and rapidly internalized in a ligand-independent manner. In constitutive GPR182-deficient mice, as well as after induced endothelium-specific loss of GPR182, we found significant increases in the plasma levels of CXCL10, CXCL12, and CXCL13. Global and induced endothelium-specific GPR182-deficient mice showed a significant decrease in hematopoietic stem cells in the bone marrow as well as increased colony-forming units of hematopoietic progenitors in the blood and the spleen. Our data show that GPR182 is a new atypical chemokine receptor for CXCL10, CXCL12, and CXCL13, which is involved in the regulation of hematopoietic stem cell homeostasis.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , Animales , Quimiocina CXCL10 , Quimiocina CXCL12 , Quimiocina CXCL13 , Quimiocinas/metabolismo , Células Endoteliales/metabolismo , Femenino , Células HEK293 , Células Madre Hematopoyéticas/metabolismo , Homeostasis , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Quimiocina/metabolismo , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/fisiología , beta-Arrestinas/metabolismo
7.
Oncogene ; 40(6): 1118-1127, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33323967

RESUMEN

Dysregulated androgen receptor (AR) plays a crucial role in prostate cancer (PCa) development, though further factors involved in its regulation remain to be identified. Recently, paradoxical results were reported on the implication of the MEN1 gene in PCa. To dissect its role in prostate luminal cells, we generated a mouse model with inducible Men1 disruption in Nkx3.1-deficient mice in which mouse prostatic intraepithelial neoplasia (mPIN) occur. Prostate glands from mutant and control mice were analyzed pathologically and molecularly; cellular and molecular analyses were carried out in PCa cell lines after MEN1 knockdown (KD) by siRNA. Double-mutant mice developed accelerated mPIN and later displayed microinvasive adenocarcinoma. Markedly, early-stage lesions exhibited a decreased expression of AR and its target genes, accompanied by reduced CK18 and E-cadherin expression, suggesting a shift from a luminal to a dedifferentiated epithelial phenotype. Intriguingly, over 60% of menin-deficient cells expressed CD44 at a later stage. Furthermore, MEN1 KD led to the increase in CD44 expression in PC3 cells re-expressing AR. Menin bound to the proximal AR promoter and regulated AR transcription via the H3K4me3 histone mark. Interestingly, the cell proliferation of AR-dependent cells (LNCaP, 22Rv1, and VCaP), but not of AR-independent cells (DU145, PC3), responded strongly to MEN1 silencing. Finally, menin expression was found reduced in some human PCa. These findings highlight the regulation of the AR promoter by menin and the crosstalk between menin and the AR pathway. Our data could be useful for better understanding the increasingly reported AR-negative/NE-negative subtype of PCa and the mechanisms underlying its development.


Asunto(s)
Proteínas de Homeodominio/genética , Receptores de Hialuranos/genética , Neoplasia Intraepitelial Prostática/genética , Proteínas Proto-Oncogénicas/genética , Receptores Androgénicos/genética , Factores de Transcripción/genética , Animales , Proliferación Celular/genética , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Masculino , Ratones , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Próstata/metabolismo , Próstata/patología , Neoplasia Intraepitelial Prostática/patología , Transducción de Señal
8.
ACS Pharmacol Transl Sci ; 3(4): 676-689, 2020 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-32832870

RESUMEN

The G protein-coupled receptor 182 (GPR182) is an orphan GPCR, the expression of which is enriched in embryonic endothelial cells (ECs). However, the physiological role and molecular mechanism of action of GPR182 are unknown. Here, we show that GPR182 negatively regulates definitive hematopoiesis in zebrafish and mice. In zebrafish, gpr182 expression is enriched in the hemogenic endothelium (HE), and gpr182 -/- display an increased expression of HE and hematopoietic stem cell (HSC) marker genes. Notably, we find an increased number of myeloid cells in gpr182 -/- compared to wild-type. Further, by time-lapse imaging of zebrafish embryos during the endothelial-to-hematopoietic transition, we find that HE/HSC cell numbers are increased in gpr182 -/- compared to wild-type. GPR182 -/- mice also exhibit an increased number of myeloid cells compared to wild-type, indicating a conserved role for GPR182 in myelopoiesis. Using cell-based small molecule screening and transcriptomic analyses, we further find that GPR182 regulates the leukotriene B4 (LTB4) biosynthesis pathway. Taken together, these data indicate that GPR182 is a negative regulator of definitive hematopoiesis in zebrafish and mice, and provide further evidence for LTB4 signaling in HSC biology.

9.
Elife ; 82019 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-31549965

RESUMEN

Myogenic vasoconstriction is an autoregulatory function of small arteries. Recently, G-protein-coupled receptors have been involved in myogenic vasoconstriction, but the downstream signalling mechanisms and the in-vivo-function of this myogenic autoregulation are poorly understood. Here, we show that small arteries from mice with smooth muscle-specific loss of G12/G13 or the Rho guanine nucleotide exchange factor ARHGEF12 have lost myogenic vasoconstriction. This defect was accompanied by loss of RhoA activation, while vessels showed normal increases in intracellular [Ca2+]. In the absence of myogenic vasoconstriction, perfusion of peripheral organs was increased, systemic vascular resistance was reduced and cardiac output and left ventricular mass were increased. In addition, animals with defective myogenic vasoconstriction showed aggravated hypotension in response to endotoxin. We conclude that G12/G13- and Rho-mediated signaling plays a key role in myogenic vasoconstriction and that myogenic tone is required to maintain local and systemic vascular resistance under physiological and pathological condition.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Resistencia Vascular , Vasoconstricción , Animales , Subunidades alfa de la Proteína de Unión al GTP G12-G13/deficiencia , Ratones Endogámicos C57BL , Factores de Intercambio de Guanina Nucleótido Rho/deficiencia
10.
Development ; 146(14)2019 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-31142539

RESUMEN

An early step in pancreas development is marked by the expression of the transcription factor Pdx1 within the pancreatic endoderm, where it is required for the specification of all endocrine cell types. Subsequently, Pdx1 expression becomes restricted to the ß-cell lineage, where it plays a central role in ß-cell function. This pivotal role of Pdx1 at various stages of pancreas development makes it an attractive target to enhance pancreatic ß-cell differentiation and increase ß-cell function. In this study, we used a newly generated zebrafish reporter to screen over 8000 small molecules for modulators of pdx1 expression. We found four hit compounds and validated their efficacy at different stages of pancreas development. Notably, valproic acid treatment increased pancreatic endoderm formation, while inhibition of TGFß signaling led to α-cell to ß-cell transdifferentiation. HC toxin, another HDAC inhibitor, enhances ß-cell function in primary mouse and human islets. Thus, using a whole organism screening strategy, this study identified new pdx1 expression modulators that can be used to influence different steps in pancreas and ß-cell development.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Islotes Pancreáticos/embriología , Modelos Animales , Organogénesis/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/análisis , Pez Cebra , Animales , Animales Modificados Genéticamente , Células COS , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Transdiferenciación Celular/efectos de los fármacos , Transdiferenciación Celular/genética , Células Cultivadas , Chlorocebus aethiops , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/aislamiento & purificación , Inhibidores de Histona Desacetilasas/farmacología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/crecimiento & desarrollo , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos C57BL , Organogénesis/genética , Bibliotecas de Moléculas Pequeñas/aislamiento & purificación , Transactivadores/genética , Transactivadores/metabolismo , Ácido Valproico/aislamiento & purificación , Ácido Valproico/farmacología , Pez Cebra/embriología , Pez Cebra/genética
11.
Nat Commun ; 9(1): 177, 2018 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-29330456

RESUMEN

The long-chain fatty acid receptor FFAR1 is highly expressed in pancreatic ß-cells. Synthetic FFAR1 agonists can be used as antidiabetic drugs to promote glucose-stimulated insulin secretion (GSIS). However, the physiological role of FFAR1 in ß-cells remains poorly understood. Here we show that 20-HETE activates FFAR1 and promotes GSIS via FFAR1 with higher potency and efficacy than dietary fatty acids such as palmitic, linoleic, and α-linolenic acid. Murine and human ß-cells produce 20-HETE, and the ω-hydroxylase-mediated formation and release of 20-HETE is strongly stimulated by glucose. Pharmacological inhibition of 20-HETE formation and blockade of FFAR1 in islets inhibits GSIS. In islets from type-2 diabetic humans and mice, glucose-stimulated 20-HETE formation and 20-HETE-dependent stimulation of GSIS are strongly reduced. We show that 20-HETE is an FFAR1 agonist, which functions as an autocrine positive feed-forward regulator of GSIS, and that a reduced glucose-induced 20-HETE formation contributes to inefficient GSIS in type-2 diabetes.


Asunto(s)
Glucosa/farmacología , Ácidos Hidroxieicosatetraenoicos/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Insulina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Adulto , Animales , Comunicación Autocrina/efectos de los fármacos , Células COS , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Chlorocebus aethiops , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Humanos , Ácidos Hidroxieicosatetraenoicos/sangre , Ácidos Hidroxieicosatetraenoicos/farmacología , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Masculino , Ratones Noqueados , Ratones Obesos , Persona de Mediana Edad , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Adulto Joven
12.
J Pathol ; 242(1): 90-101, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28188614

RESUMEN

Foxa2, known as one of the pioneer factors, plays a crucial role in islet development and endocrine functions. Its expression and biological functions are regulated by various factors, including, in particular, insulin and glucagon. However, its expression and biological role in adult pancreatic α-cells remain elusive. In the current study, we showed that Foxa2 was overexpressed in islets from α-cell-specific Men1 mutant mice, at both the transcriptional level and the protein level. More importantly, immunostaining analyses showed its prominent nuclear accumulation, specifically in α-cells, at a very early stage after Men1 disruption. Similar nuclear FOXA2 expression was also detected in a substantial proportion (12/19) of human multiple endocrine neoplasia type 1 (MEN1) glucagonomas. Interestingly, our data revealed an interaction between Foxa2 and menin encoded by the Men1 gene. Furthermore, using several approaches, we demonstrated the relevance of this interaction in the regulation of two tested Foxa2 target genes, including the autoregulation of the Foxa2 promoter by Foxa2 itself. The current study establishes menin, a novel protein partner of Foxa2, as a regulator of Foxa2, the biological functions of which extend beyond the pancreatic endocrine cells. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Glucagonoma/metabolismo , Factor Nuclear 3-beta del Hepatocito/biosíntesis , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Neoplasias Pancreáticas/metabolismo , Animales , Regulación Neoplásica de la Expresión Génica , Glucagonoma/genética , Factor Nuclear 3-beta del Hepatocito/genética , Humanos , Ratones Transgénicos , Neoplasia Endocrina Múltiple Tipo 1/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/genética , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Transfección , Células Tumorales Cultivadas
13.
Mol Cell Biol ; 36(5): 756-64, 2015 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-26711255

RESUMEN

Loss of pancreatic ß-cell maturity occurs in diabetes and insulinomas. Although both physiological and pathological stresses are known to promote ß-cell dedifferentiation, little is known about the molecules involved in this process. Here we demonstrate that activinB, a transforming growth factor ß (TGF-ß)-related ligand, is upregulated during tumorigenesis and drives the loss of insulin expression and ß-cell maturity in a mouse insulinoma model. Our data further identify Pax4 as a previously unknown activinB target and potent contributor to the observed ß-cell dedifferentiation. More importantly, using compound mutant mice, we found that deleting activinB expression abolishes tumor ß-cell dedifferentiation and, surprisingly, increases survival without significantly affecting tumor growth. Hence, this work reveals an unexpected role for activinB in the loss of ß-cell maturity, islet plasticity, and progression of insulinoma through its participation in ß-cell dedifferentiation.


Asunto(s)
Activinas/metabolismo , Desdiferenciación Celular , Células Secretoras de Insulina/patología , Insulinoma/patología , Páncreas/patología , Neoplasias Pancreáticas/patología , Activinas/genética , Animales , Regulación Neoplásica de la Expresión Génica , Insulina/genética , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Insulinoma/genética , Insulinoma/metabolismo , Ratones Endogámicos C57BL , Páncreas/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo
14.
Mol Cell Biol ; 35(19): 3274-83, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26169832

RESUMEN

The cells of origin of pancreatic gastrinomas remain an enigma, since no gastrin-expressing cells are found in the normal adult pancreas. It was proposed that the cellular origin of pancreatic gastrinomas may come from either the pancreatic cells themselves or gastrin-expressing cells which have migrated from the duodenum. In the current study, we further characterized previously described transient pancreatic gastrin-expressing cells using cell lineage tracing in a pan-pancreatic progenitor and a pancreatic endocrine progenitor model. We provide evidence showing that pancreatic gastrin-expressing cells, found from embryonic day 12.5 until postnatal day 7, are derived from pancreatic Ptf1a(+) and neurogenin 3-expressing (Ngn3(+)) progenitors. Importantly, the majority of them coexpress glucagon, with 4% coexpressing insulin, indicating that they are a temporary subpopulation of both alpha and beta cells. Interestingly, Men1 disruption in both Ngn3 progenitors and beta and alpha cells resulted in the development of pancreatic gastrin-expressing tumors, suggesting that the latter developed from islet cells. Finally, we detected gastrin expression using three human cohorts with pancreatic endocrine tumors (pNETs) that have not been diagnosed as gastrinomas (in 9/34 pNETs from 6/14 patients with multiple endocrine neoplasia type 1, in 5/35 sporadic nonfunctioning pNETs, and in 2/20 sporadic insulinomas), consistent with observations made in mouse models. Our work provides insight into the histogenesis of pancreatic gastrin-expressing tumors.


Asunto(s)
Gastrinas/metabolismo , Islotes Pancreáticos/patología , Neoplasia Endocrina Múltiple/patología , Neoplasias Pancreáticas/patología , Animales , Carcinogénesis/patología , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasia Endocrina Múltiple/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo
15.
World J Diabetes ; 5(6): 847-53, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25512786

RESUMEN

Pancreatic insulin-secreting ß-cells are essential regulators of glucose metabolism. New strategies are currently being investigated to create insulin-producing ß cells to replace deficient ß cells, including the differentiation of either stem or progenitor cells, and the newly uncovered transdifferentiation of mature non-ß islet cell types. However, in order to correctly drive any cell to adopt a new ß-cell fate, a better understanding of the in vivo mechanisms involved in the plasticity and biology of islet cells is urgently required. Here, we review the recent studies reporting the phenomenon of transdifferentiation of α cells into ß cells by focusing on the major candidates and contexts revealed to be involved in adult ß-cell regeneration through this process. The possible underlying mechanisms of transdifferentiation and the interactions between several key factors involved in the process are also addressed. We propose that it is of importance to further study the molecular and cellular mechanisms underlying α- to ß-cell transdifferentiation, in order to make ß-cell regeneration from α cells a relevant and realizable strategy for developing cell-replacement therapy.

16.
PLoS One ; 8(8): e72194, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24013263

RESUMEN

Pax4 and MafA (v-maf musculoaponeurotic fibrosarcoma oncogene homolog A) are two transcription factors crucial for normal functions of islet beta cells in the mouse. Intriguingly, recent studies indicate the existence of notable difference between human and rodent islet in terms of gene expression and functions. To better understand the biological role of human PAX4 and MAFA, we investigated their expression in normal and diseased human islets, using validated antibodies. PAX4 was detected in 43.0±5.0% and 39.1±4.0% of normal human alpha and beta cells respectively. We found that MAFA, detected in 88.3±6.3% insulin(+)cells as in the mouse, turned out to be also expressed in 61.2±6.4% of human glucagons(+) cells with less intensity than in insulin(+) cells, whereas MAFB expression was found not only in the majority of glucagon(+) cells (67.2±7.6%), but also in 53.6±10.5% of human insulin(+) cells. Interestingly, MAFA nuclear expression in both alpha and beta cells, and the percentage of alpha cells expressing PAX4 were found altered in a substantial proportion of patients with type 2 diabetes. Both MAFA and PAX4 display, therefore, a distinct expression pattern in human islet cells, suggesting more potential plasticity of human islets as compared with rodent islets.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Glucagón/metabolismo , Proteínas de Homeodominio/metabolismo , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Factores de Transcripción Paired Box/metabolismo , Adolescente , Adulto , Anciano , Animales , Estudios de Casos y Controles , Núcleo Celular/metabolismo , Células Cultivadas , Diabetes Mellitus Tipo 2/patología , Femenino , Expresión Génica , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Células Secretoras de Insulina/metabolismo , Factores de Transcripción Maf de Gran Tamaño/genética , Masculino , Ratones , Persona de Mediana Edad , Obesidad/metabolismo , Factores de Transcripción Paired Box/genética , Páncreas/metabolismo , Páncreas/patología
17.
J Pathol ; 229(4): 546-58, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23180448

RESUMEN

Mutations of the MEN1 tumour suppressor gene predispose patients to the development of multiple endocrine neoplasia type 1 (MEN1) syndrome, which is characterized by multiple endocrine tumours, including prolactinomas. The recent findings of the interaction between menin, encoded by the MEN1 gene, and the oestrogen receptor, as well as the observation of rare cases of mammary carcinomas in our heterozygous Men1 mutant mice, led us to investigate a putative tumour suppressor function of the Men1 gene in mouse mammary cells by disrupting the gene in luminal epithelial cells. A significantly higher incidence of mammary intraepithelial neoplasia (MIN) was observed in mutant WapCre-Men1(F/F) mice (51.5%) than in WapCre-Men1(+/+) (0%) or Men1(F/F) (7.1%) control mice. The majority of MIN observed in the mutant mice displayed complete menin inactivation. Because of the leakage of WapCre transgene expression, prolactinomas were observed in 83.3% of mutant mice, leading to premature death. As there was no correlation between MIN development and elevated serum prolactin levels, and phospho-STAT5 expression was decreased in mammary lesions, the increased incidence of MIN lesions was most likely due to Men1 disruption rather than to prolactinoma development. Interestingly, in MIN lesions, we found a decrease in membrane-associated E-cadherin and beta-catenin expression, the latter of which is a menin partner. Finally, reduced menin expression was found in a large proportion of two independent cohorts of patients with breast carcinomas. Taken together, the current work indicates a role of Men1 inactivation in the development of mammary pre-cancerous lesions in mice and a potential role in human mammary cancer.


Asunto(s)
Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/genética , Lesiones Precancerosas/patología , Proteínas Proto-Oncogénicas/metabolismo , beta Catenina/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Transformación Celular Neoplásica , Estudios de Cohortes , Células Epiteliales , Femenino , Estudios de Seguimiento , Humanos , Incidencia , Integrasas/genética , Integrasas/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas de la Leche/genética , Proteínas de la Leche/metabolismo , Mutación , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Embarazo , Proteínas Proto-Oncogénicas/genética , Análisis de Matrices Tisulares
18.
Gastroenterology ; 138(5): 1954-65, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20138042

RESUMEN

BACKGROUND & AIMS: The tumor suppressor menin is recognized as a key regulator of pancreatic islet development, proliferation, and beta-cell function, whereas its role in alpha cells remains poorly understood. The purpose of the current study was to address this issue in relation to islet tumor histogenesis. METHODS: We generated alpha cell-specific Men1 mutant mice with Cre/loxP technology and carried out analyses of pancreatic lesions developed in the mutant mice during aging. RESULTS: We showed that, despite the alpha-cell specificity of the GluCre transgene, both glucagonomas and a large amount of insulinomas developed in mutant mice older than 6 months, accompanied by mixed islet tumors. Interestingly, the cells sharing characteristics of both alpha and beta cells were identified shortly after the appearance of menin-deficient alpha cells but well before the tumor onset. Using a genetic cell lineage tracing analysis, we demonstrated that insulinoma cells were directly derived from transdifferentiating glucagon-expressing cells. Furthermore, our data indicated that the expression of Pdx1, MafA, Pax4, and Ngn3 did not seem to be required for the initiation of this transdifferentiation. CONCLUSIONS: Our work shows cell transdifferentiation as a novel mechanism involved in islet tumor development and provides evidence showing that menin regulates the plasticity of differentiated pancreatic alpha cells in vivo, shedding new light on the mechanisms of islet tumorigenesis.


Asunto(s)
Transdiferenciación Celular , Transformación Celular Neoplásica/metabolismo , Células Secretoras de Glucagón/metabolismo , Glucagón/metabolismo , Glucagonoma/metabolismo , Insulinoma/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas/deficiencia , Factores de Edad , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Biomarcadores/metabolismo , Fusión Celular , Linaje de la Célula , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Eliminación de Gen , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genotipo , Células Secretoras de Glucagón/patología , Glucagonoma/genética , Glucagonoma/patología , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Insulinoma/genética , Insulinoma/patología , Ratones , Ratones Noqueados , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Fenotipo , Proteínas Proto-Oncogénicas/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA