Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 36(10): 109674, 2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34496236

RESUMEN

Tumor-initiating stem cells (TSCs) are critical for drug resistance and immune escape. However, the mutual regulations between TSC and tumor microenvironment (TME) remain unclear. Using DNA-label retaining, single-cell RNA sequencing (scRNA-seq), and other approaches, we investigated intestinal adenoma in response to chemoradiotherapy (CRT), thus identifying therapy-resistant TSCs (TrTSCs). We find bidirectional crosstalk between TSCs and TME using CellPhoneDB analysis. An intriguing finding is that TSCs shape TME into a landscape that favors TSCs for immunosuppression and propagation. Using adenoma-organoid co-cultures, niche-cell depletion, and lineaging tracing, we characterize a functional role of cyclooxygenase-2 (Cox-2)-dependent signaling, predominantly occurring between tumor-associated monocytes and macrophages (TAMMs) and TrTSCs. We show that TAMMs promote TrTSC proliferation through prostaglandin E2 (PGE2)-PTGER4(EP4) signaling, which enhances ß-catenin activity via AKT phosphorylation. Thus, our study shows that the bidirectional crosstalk between TrTSC and TME results in a pro-tumorigenic and immunosuppressive contexture.


Asunto(s)
Carcinogénesis/patología , Forma de la Célula/fisiología , Células Madre Neoplásicas/patología , Microambiente Tumoral/fisiología , Animales , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Humanos , Intestinos/metabolismo , Ratones , Organoides/metabolismo
2.
Nat Cell Biol ; 22(6): 689-700, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32313104

RESUMEN

Leukaemia stem cells (LSCs) underlie cancer therapy resistance but targeting these cells remains difficult. The Wnt-ß-catenin and PI3K-Akt pathways cooperate to promote tumorigenesis and resistance to therapy. In a mouse model in which both pathways are activated in stem and progenitor cells, LSCs expanded under chemotherapy-induced stress. Since Akt can activate ß-catenin, inhibiting this interaction might target therapy-resistant LSCs. High-throughput screening identified doxorubicin (DXR) as an inhibitor of the Akt-ß-catenin interaction at low doses. Here we repurposed DXR as a targeted inhibitor rather than a broadly cytotoxic chemotherapy. Targeted DXR reduced Akt-activated ß-catenin levels in chemoresistant LSCs and reduced LSC tumorigenic activity. Mechanistically, ß-catenin binds multiple immune-checkpoint gene loci, and targeted DXR treatment inhibited expression of multiple immune checkpoints specifically in LSCs, including PD-L1, TIM3 and CD24. Overall, LSCs exhibit distinct properties of immune resistance that are reduced by inhibiting Akt-activated ß-catenin. These findings suggest a strategy for overcoming cancer therapy resistance and immune escape.


Asunto(s)
Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Leucemia Mieloide Aguda/patología , Células Madre Neoplásicas/patología , Fosfohidrolasa PTEN/fisiología , Proteínas Wnt/fisiología , beta Catenina/fisiología , Animales , Antibióticos Antineoplásicos/farmacología , Apoptosis , Proliferación Celular , Femenino , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Masculino , Ratones , Ratones Noqueados , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Blood ; 120(9): 1831-42, 2012 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-22802336

RESUMEN

Previous studies have shown that fibroblast growth factor (FGF) signaling promotes hematopoietic stem and progenitor cell (HSPC) expansion in vitro. However, it is unknown whether FGF promotes HSPC expansion in vivo. Here we examined FGF receptor 1 (FGFR1) expression and investigated its in vivo function in HSPCs. Conditional knockout (CKO) of Fgfr1 did not affect phenotypical number of HSPCs and homeostatic hematopoiesis, but led to a reduced engraftment only in the secondary transplantation. When treated with 5-fluorouracil (5FU), the Fgfr1 CKO mice showed defects in both proliferation and subsequent mobilization of HSPCs. We identified megakaryocytes (Mks) as a major resource for FGF production, and further discovered a novel mechanism by which Mks underwent FGF-FGFR signaling dependent expansion to accelerate rapid FGF production under stress. Within HSPCs, we observed an up-regulation of nuclear factor κB and CXCR4, a receptor for the chemoattractant SDF-1, in response to bone marrow damage only in control but not in Fgfr1 CKO model, accounting for the corresponding defects in proliferation and migration of HSPCs. This study provides the first in vivo evidence that FGF signaling facilitates postinjury recovery of the mouse hematopoietic system by promoting proliferation and facilitating mobilization of HSPCs.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Sistema Hematopoyético/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Animales , Antimetabolitos Antineoplásicos/farmacología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Movimiento Celular/genética , Proliferación Celular , Células Cultivadas , Femenino , Citometría de Flujo , Fluorouracilo/farmacología , Expresión Génica/efectos de los fármacos , Sistema Hematopoyético/citología , Sistema Hematopoyético/efectos de los fármacos , Inmunohistoquímica , Masculino , Megacariocitos/efectos de los fármacos , Megacariocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , FN-kappa B/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptores CXCR4/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Cell Stem Cell ; 2(4): 367-79, 2008 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-18397756

RESUMEN

Osteoblasts expressing the homophilic adhesion molecule N-cadherin form a hematopoietic stem cell (HSC) niche. Therefore, we examined how N-cadherin expression in HSCs relates to their function. We found that bone marrow (BM) cells highly expressing N-cadherin (N-cadherin(hi)) are not stem cells, being largely devoid of a Lineage(-)Sca1(+)cKit(+) population and unable to reconstitute hematopoietic lineages in irradiated recipient mice. Instead, long-term HSCs form distinct populations expressing N-cadherin at intermediate (N-cadherin(int)) or low (N-cadherin(lo)) levels. The minority N-cadherin(lo) population can robustly reconstitute the hematopoietic system, express genes that may prime them to mobilize, and predominate among HSCs mobilized from BM to spleen. The larger N-cadherin(int) population performs poorly in reconstitution assays when freshly isolated but improves in response to overnight in vitro culture. Their expression profile and lower cell-cycle entry rate suggest N-cadherin(int) cells are being held in reserve. Thus, differential N-cadherin expression reflects functional distinctions between two HSC subpopulations.


Asunto(s)
Biomarcadores/metabolismo , Cadherinas/metabolismo , Células Madre Hematopoyéticas/citología , Animales , Antimetabolitos Antineoplásicos/farmacología , Ataxina-1 , Ataxinas , Secuencia de Bases , Células de la Médula Ósea/metabolismo , Cadherinas/genética , Diferenciación Celular , Linaje de la Célula , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Cartilla de ADN/química , Citometría de Flujo , Fluorouracilo/farmacología , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/fisiología , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Osteoblastos/citología , Osteoblastos/fisiología , Proteínas Proto-Oncogénicas c-kit/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Ácido Nucleico , Bazo/citología , Bazo/metabolismo
6.
Dev Biol ; 305(2): 508-21, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17418116

RESUMEN

Transforming growth factor-beta (TGF-beta) superfamily signaling has been implicated in many developmental processes, including pancreatic development. Previous studies are conflicting with regard to an exact role for TGF-beta signaling in various aspects of pancreatic organogenesis. Here we have investigated the role of TGF-beta isoform signaling in embryonic pancreas differentiation and lineage selection. The TGF-beta isoform receptors (RI, RII and ALK1) were localized mainly to both the pancreatic epithelium and mesenchyme at early stages of development, but then with increasing age localized to the pancreatic islets and ducts. To determine the specific role of TGF-beta isoforms, we functionally inactivated TGF-beta signaling at different points in the signaling cascade. Disruption of TGF-beta signaling at the receptor level using mice overexpressing the dominant-negative TGF-beta type II receptor showed an increase in endocrine precursors and proliferating endocrine cells, with an abnormal accumulation of endocrine cells around the developing ducts of mid-late stage embryonic pancreas. This pattern suggested that TGF-beta isoform signaling may suppress the origination of secondary transition endocrine cells from the ducts. Secondly, TGF-beta isoform ligand inhibition with neutralizing antibody in pancreatic organ culture also led to an increase in the number of endocrine-positive cells. Thirdly, hybrid mix-and-match in vitro recombinations of transgenic pancreatic mesenchyme and wild-type epithelium also led to increased endocrine cell differentiation, but with different patterns depending on the directionality of the epithelial-mesenchymal signaling. Together these results suggest that TGF-beta signaling is important for restraining the growth and differentiation of pancreatic epithelial cells, particularly away from the endocrine lineage. Inhibition of TGF-beta signaling in the embryonic period may thus allow pancreatic epithelial cells to progress towards the endocrine lineage unchecked, particularly as part of the secondary transition of pancreatic endocrine cell development. TGF-beta RII in the ducts and islets may normally serve to downregulate the production of beta cells from embryonic ducts.


Asunto(s)
Islotes Pancreáticos/embriología , Mesodermo/fisiología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Islotes Pancreáticos/metabolismo , Ratones , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Isoformas de Proteínas/fisiología , Proteínas Serina-Treonina Quinasas , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética
7.
Nat Genet ; 39(2): 189-98, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17237784

RESUMEN

Intestinal polyposis, a precancerous neoplasia, results primarily from an abnormal increase in the number of crypts, which contain intestinal stem cells (ISCs). In mice, widespread deletion of the tumor suppressor Phosphatase and tensin homolog (PTEN) generates hamartomatous intestinal polyps with epithelial and stromal involvement. Using this model, we have established the relationship between stem cells and polyp and tumor formation. PTEN helps govern the proliferation rate and number of ISCs and loss of PTEN results in an excess of ISCs. In PTEN-deficient mice, excess ISCs initiate de novo crypt formation and crypt fission, recapitulating crypt production in fetal and neonatal intestine. The PTEN-Akt pathway probably governs stem cell activation by helping control nuclear localization of the Wnt pathway effector beta-catenin. Akt phosphorylates beta-catenin at Ser552, resulting in a nuclear-localized form in ISCs. Our observations show that intestinal polyposis is initiated by PTEN-deficient ISCs that undergo excessive proliferation driven by Akt activation and nuclear localization of beta-catenin.


Asunto(s)
Mucosa Intestinal/metabolismo , Pólipos Intestinales/genética , Fosfohidrolasa PTEN/fisiología , Células Madre/metabolismo , Animales , Recuento de Células , Ciclo Celular , Núcleo Celular/metabolismo , Ratones , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , beta Catenina/metabolismo
8.
J Pediatr Surg ; 41(7): 1208-13, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16818050

RESUMEN

BACKGROUND: The organogenesis of esophageal atresia with tracheoesophageal fistula remains unclear. We have previously demonstrated that the fistula tract develops from a trifurcation of the embryonic lung bud and displays pulmonary lineage traits. Unlike the lung, the fistula grows without branching. Bone morphogenetic proteins (BMPs) are known to be important in lung branching. We studied possible BMP signaling defects as a potential cause for the absence of branching in the fistula tract. METHODS: Adriamycin was administered to pregnant rats on days 6-9 of gestation to induce tracheoesophageal fistula. Microdissection was performed at E13 and E17 isolating the foregut. Tissues were analyzed using immunohistochemistry for BMP ligand (BMP2, BMP4, BMP7) and receptor (BMPRIA, BMPRIB, BMPRII) expression. RESULTS: Immunohistochemistry revealed the presence of all 3 BMP ligands at E13, localized specifically to the esophageal mucosa but absent in the fistula and lung. At E17, the ligands were again present in the esophageal mucosa, and additionally in the fistula tract mucosa, but remained absent in the lung. At E17, all of the BMP receptors were also localized to the luminal surface of esophagus and fistula. However, in the lung epithelium, only BMPRII was found, whereas BMPRIA and BMPRIB remained absent. CONCLUSIONS: The normal expression pattern of BMP4 was increased at the branch tips and low between branches. Among other results, we show here a constant expression level of BMP ligands throughout the entire epithelium of the fistula tract. This diffuse expression suggests defective BMP signaling in the fistula tract and explains its nonbranching phenotype.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Atresia Esofágica/fisiopatología , Transducción de Señal/fisiología , Fístula Traqueoesofágica/fisiopatología , Anomalías Inducidas por Medicamentos/fisiopatología , Animales , Modelos Animales de Enfermedad , Doxorrubicina/efectos adversos , Atresia Esofágica/complicaciones , Femenino , Inmunohistoquímica , Embarazo , Ratas , Ratas Sprague-Dawley , Teratógenos/farmacología , Fístula Traqueoesofágica/inducido químicamente , Fístula Traqueoesofágica/complicaciones
9.
Dev Growth Differ ; 48(2): 65-72, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16512851

RESUMEN

Epithelial-mesenchymal interactions are crucial for the proper development of many organs, including the pancreas. Within the pancreas, the ducts are thought to harbor stem/progenitor cells, and possibly to give rise to pancreatic ductal carcinoma. Little is known about the mechanism of formation of pancreatic ducts in the embryo. Pancreatic mesenchyme contains numerous soluble factors which help to sustain the growth and differentiation of exocrine and endocrine structures. Here, we report that one such morphoregulatory mesenchymal protein, epimorphin, plays an important role during pancreatic ductal proliferation and differentiation. We found that epimorphin is expressed in pancreatic mesenchyme during early stages of development, and at mesenchymal-epithelial interfaces surrounding the ducts at later stages. Strong upregulation of epimorphin expression was seen during in vitro pancreatic duct differentiation. Similarly, in vitro pancreatic duct formation was inhibited by a neutralizing antibody against epimorphin, whereas addition of recombinant epimorphin partially rescued duct formation. Together, our study demonstrates the role of epimorphin in pancreatic ductal morphogenesis.


Asunto(s)
Glicoproteínas de Membrana/fisiología , Mesodermo/fisiología , Conductos Pancreáticos/embriología , Animales , Diferenciación Celular/fisiología , Femenino , Masculino , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Mesodermo/citología , Ratones , Ratones Endogámicos ICR , Técnicas de Cultivo de Órganos , Conductos Pancreáticos/citología , Transducción de Señal/fisiología , Regulación hacia Arriba/fisiología
10.
Pediatr Surg Int ; 22(2): 154-7, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16315037

RESUMEN

The organogenesis of esophageal atresia with tracheoesophageal fistula (EA/TEF) remains unknown. The fistula tract appears to develop from a non-branching trifurcation of the embryonic lung bud. The non-branching growth of the fistula differs from the other lung buds and suggests a deficiency in bone morphogenetic protein (BMP) signaling, since BMPs are critical to proper lung development and branching. With IRB approval, portions of newborn human proximal esophageal pouch and distal fistula samples were recovered at the time of surgical repair of EA/TEF. The tissues were processed for immunohistochemistry. Commercially available fetal tissues were used as controls. In control tissues, BMP ligands (BMP 2, 4, and 7) were all present in the esophagus but absent in the trachea. BMPRIA was absent in both tissues. BMPRIB was detected in trachea but not in esophagus and BMPRII was detected in esophagus but not in trachea. In the EA/TEF specimens, all BMP ligands were present in the proximal esophageal pouch but absent in the fistula tract. BMPRIA and BMPRIB were not detected in either tissue. However, BMPRII was found in both fistula tract and proximal pouch. The submucosa of the fistula appears to maintain a mixed (identical neither to lung, esophagus, or trachea) BMP signaling pattern, providing one mechanism which could potentially explain the esophageal dismotility and lack of lung branching seen in the fistula/distal esophagus.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Atresia Esofágica/embriología , Fístula Traqueoesofágica/embriología , Estudios de Casos y Controles , Atresia Esofágica/patología , Humanos , Inmunohistoquímica , Recién Nacido , Ligandos , Transducción de Señal , Fístula Traqueoesofágica/patología
11.
Pancreas ; 31(2): 138-41, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16025000

RESUMEN

OBJECTIVES: Glucagon-like peptide-1 (GLP-1) is known to stimulate glucose-dependent insulin production and secretion by pancreatic beta-cells. Preliminary evidence suggests that GLP-1 may also influence endocrine differentiation from pancreatic progenitor cells. Additionally, TGF-beta signaling can also control endocrine differentiation by both inhibiting proliferation and enhancing differentiation of endocrine progenitor cells to become mature beta-cells. Here we document synergy of these two signaling pathways in the differentiation of endocrine cells in the developing pancreas. METHODS: Embryonic pancreas was harvested from mice at day 11.5 and cultured for six days with GLP-1 agonist, exendin-4, and/or TGF-beta1 ligand. Also, a pan-neutralizing TGF-beta isoform antibody was used alone or with exendin-4 to study TGF-beta inhibition in this system. Pancreatic cultures were processed for immunohistochemistry. RESULTS: Exogenous TGF-beta1 and exendin-4 each individually enhanced both insulin and glucagon differentiation dose-dependently. However, when combined there was an additive effect to a 4.5-fold increase in insulin-positive differentiation. We also saw suppression of amylase-positive differentiation. Surprisingly, TGF-beta pan-neutralizing antibody also gave an augmentation of endocrine differentiation by 1.5 to 2-fold, but no synergistic effect was seen with exendin-4. CONCLUSION: We conclude that TGF-beta isoforms have a specific synergistic role with GLP-1 pathway signaling in early pancreatic development, toward endocrine differentiation and away from acinar differentiation.


Asunto(s)
Péptido 1 Similar al Glucagón/farmacología , Islotes Pancreáticos , Factor de Crecimiento Transformador beta/farmacología , Animales , Anticuerpos/farmacología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Sinergismo Farmacológico , Exenatida , Péptido 1 Similar al Glucagón/agonistas , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/embriología , Ratones , Ratones Endogámicos , Páncreas Exocrino/citología , Páncreas Exocrino/embriología , Péptidos/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Células Madre/citología , Células Madre/efectos de los fármacos , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta1 , Ponzoñas/farmacología
12.
J Biol Chem ; 280(37): 32209-17, 2005 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-16020542

RESUMEN

A key goal of cellular engineering is to manipulate progenitor cells to become beta-cells, allowing cell replacement therapy to cure diabetes mellitus. As a paradigm for cell engineering, we have studied the molecular mechanisms by which AR42J cells become beta-cells. Bone morphogenetic proteins (BMPs), implicated in a myriad of developmental pathways, have not been well studied in insulin-positive differentiation. We found that the canonical intracellular mediators of BMP signaling, Smad-1 and Smad-8, were significantly elevated in AR42J cells undergoing insulin-positive differentiation in response to exendin-4 treatment, suggesting a role for BMP signaling in beta-cell formation. Similarly, endogenous BMP-2 ligand and ALK-1 receptor (activin receptor-like kinase-1; known to activate Smads 1 and 8) mRNAs were specifically up-regulated in exendin-4-treated AR42J cells. Surprisingly, Smad-1 and Smad-8 levels were suppressed by the addition of BMP-soluble receptor inhibition of BMP ligand binding to its receptor. Here, insulin-positive differentiation was also ablated. BMP-2 ligand antisense also strongly inhibited Smad-1 and Smad-8 expression, again with the abolition of insulin-positive differentiation. These results demonstrate a previously unrecognized key role for BMP signaling in mediating insulin-positive differentiation through the intracellular Smad signaling pathway. In short, BMP signaling may represent a novel downstream target of exendin-4 (glucagon-like peptide 1) signaling and potentially serve as an upstream regulator of transforming growth factor-beta isoform signaling to differentiate the acinar-like AR42J cells into insulin-secreting cells.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Insulina/metabolismo , Péptidos/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Ponzoñas/metabolismo , Animales , Benzotiazoles , Western Blotting , Proteína Morfogenética Ósea 2 , Diferenciación Celular , Cartilla de ADN/química , Proteínas de Unión al ADN/metabolismo , Diaminas , Relación Dosis-Respuesta a Droga , Exenatida , Glucagón/metabolismo , Péptido 1 Similar al Glucagón , Islotes Pancreáticos , Ligandos , Compuestos Orgánicos/farmacología , Fragmentos de Péptidos/metabolismo , Fosfoproteínas/metabolismo , Reacción en Cadena de la Polimerasa , Unión Proteica , Isoformas de Proteínas , Precursores de Proteínas/metabolismo , Quinolinas , ARN Mensajero/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Smad , Proteína Smad1 , Proteína Smad5 , Proteína Smad8 , Transactivadores/metabolismo
13.
Diabetes ; 53(11): 2824-35, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15504962

RESUMEN

The differentiation of pancreatic exocrine AR42J cells into insulin-expressing endocrine cells has served as an important model for both endogenous in vivo beta-cell differentiation as well as potential application to beta-cell engineering of progenitor cells. Exogenous activin, possibly working through intracellular smad 2 and/or smad 3, as well as exogenous exendin-4 (a long-acting glucagon-like peptide-1 agonist) have both been shown to induce insulin-positive/endocrine differentiation in AR42J cells. In this study, we present evidence of significant interplay and interdependence of these two pathways as well as potential synergy between the pathways. In particular, insulin-positive differentiation seems to entail an exendin-4-induced drop in smad 2 and elevation in smad 3 in RNA levels. The latter appears to be dependent on endogenous transforming growth factor (TGF)-beta isoform release by the AR42J cells and may serve as a mechanism to promote beta-cell maturation. The drop in smad 2 may mediate early endocrine commitment. The coapplication of exogenous exendin-4 and, specifically, low-dose exogenous TGF-beta1 led to a dramatic 20-fold increase in insulin mRNA levels, supporting a novel synergistic and codependent relationship between exendin-4 signaling and TGF-beta isoform signaling.


Asunto(s)
Diferenciación Celular/fisiología , Glucagón/fisiología , Insulina/farmacología , Fragmentos de Péptidos/fisiología , Precursores de Proteínas/fisiología , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/fisiología , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Cartilla de ADN , Equidae , Exenatida , Péptido 1 Similar al Glucagón , Oligonucleótidos Antisentido/farmacología , Péptidos/farmacología , Reacción en Cadena de la Polimerasa , Ponzoñas/farmacología
14.
Dev Biol ; 269(1): 252-63, 2004 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15081371

RESUMEN

Mouse pancreatic development is critically dependent on epithelial-mesenchymal interactions. The pancreas differs from other epithelial-mesenchymal organs in that the epithelium gives rise to both epithelial exocrine cells and non-epithelial endocrine cells. We studied the nature of the interactions between the epithelium and mesenchyme with respect to the decision between exocrine and endocrine lineages. We show here a tripartite influence of mesenchyme on the developing epithelium. First, close proximity or contact of mesenchyme with the epithelium induces exocrine differentiation. Second, this mesenchymal proximity to the epithelium suppresses endocrine differentiation. Third, mesenchyme has an overall enhancing effect on the degree of insulin differentiation, suggesting a pro-endocrine effect in those epithelial cells at a distance from the mesenchyme. Proximity or contact between the mesenchyme and epithelium appeared to be necessary for the pro-exocrine effects of mesenchyme. We found that, in a co-culture system, NIH3T3 cells were able to substitute for mesenchyme in exocrine induction as well as in both the endocrine induction and endocrine inhibition, implying that the responsible molecules are not unique to pancreatic mesenchyme. Laminin appears to be a key molecule mediating the epithelial-mesenchymal interactions that lead to exocrine differentiation, since inhibition of laminin expression resulted in blockage of the pro-exocrine induction of mesenchyme.


Asunto(s)
Diferenciación Celular/fisiología , Mesodermo/metabolismo , Páncreas/embriología , Animales , Epitelio/embriología , Epitelio/metabolismo , Técnicas In Vitro , Islotes Pancreáticos/embriología , Islotes Pancreáticos/metabolismo , Laminina/metabolismo , Ratones , Células 3T3 NIH , Páncreas/metabolismo
15.
J Pediatr Surg ; 39(4): 537-9, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15065023

RESUMEN

BACKGROUND: Although the pathogenesis of esophageal atresia with tracheoesophageal fistula (EA/TEF) remains unknown, it has been shown that despite its esophageal appearance, the fistula tract originates from respiratory epithelium. The authors now hypothesize that defects in fibroblast growth factor (FGF) signaling contribute to the esophaguslike phenotype of the fistula tract. FGF2R is critical to normal lung morphogenesis and occurs in 2 isoforms (FGF2RIIIb and FGF2RIIIc), each with different ligand-binding specificity. To characterize FGF signaling in the developing EA/TEF, the authors analyzed levels of FGF2R splice variants in experimental EA/TEF. METHODS: The standard Adriamycin-induced EA/TEF model in rats was used. Individual foregut components from Adriamycin-treated and control embryos were processed for real-time, fluorescence-activated semiquantitative reverse transcriptase polymerase chain reaction on gestational days 12.5 and 13.5. RESULTS: Both fistula tract and Adriamycin-treated or normal esophagus showed significantly lower levels of FGF2RIIIb than either Adriamycin-treated lung buds (E12.5, P =.02; E13.5, P <.005) or normal lung buds (E12.5, P <.005; E13.5, P <.01). At E13.5, the fistula tract had lower levels of FGF2RIIIc than either treated (P <.01) or normal lung (P <.05). CONCLUSIONS: Levels of FGF2R in the developing fistula tract resemble that of distal esophagus rather than developing lung. This defect in FGF2RIIIb signaling may account for the nonbranching, esophaguslike phenotype of the fistula, despite its respiratory origin.


Asunto(s)
Atresia Esofágica/embriología , Receptores de Factores de Crecimiento de Fibroblastos/deficiencia , Fístula Traqueoesofágica/embriología , Animales , Doxorrubicina/toxicidad , Atresia Esofágica/inducido químicamente , Atresia Esofágica/metabolismo , Atresia Esofágica/patología , Factores de Crecimiento de Fibroblastos/fisiología , Pulmón/embriología , Modelos Animales , Morfogénesis/efectos de los fármacos , Fenotipo , Reacción en Cadena de la Polimerasa , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Transducción de Señal , Fístula Traqueoesofágica/inducido químicamente , Fístula Traqueoesofágica/metabolismo , Fístula Traqueoesofágica/patología
16.
Surgery ; 134(2): 345-50, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12947339

RESUMEN

BACKGROUND: The pathogenesis of esophageal atresia and tracheoesophageal fistula (EA/TEF) remains unknown. We have found previously that an initial esophageal atresia, followed by an abnormal (absent) branching pattern of the middle branch of a trifurcation of the lung/tracheal bud, leads to the neonatal finding of TEF. Mice null mutant for hedgehog signaling can experience the development of EA/TEF, but the mechanism for this development is also unknown. Given that EA/TEF in humans appears not to be due to genetic defects, a hedgehog mutation cause seems very unlikely. However, defective hedgehog signaling that is caused by environmental effects in the human embryo likely could be implicated. We studied a teratogen-induced model of EA/TEF to determine the mechanism by which defective hedgehog signaling may lead to EA/TEF. METHODS: We injected Adriamycin into pregnant rats to induce EA/TEF in rat embryos. We first quantified sonic hedgehog (Shh) signaling pathway molecule expression using real-time, semiquantitative reverse-transcriptase polymerase chain reaction for Shh, Shh receptors (patched and smoothened), and downstream intracellular targets of those receptors (Gli family members). On the basis of these findings, we then developed an in vitro culture system for the day-12 embryonic TEF and manipulated Shh signaling using either exogenous Shh or Shh inhibitors. RESULTS: By reverse transcriptase-polymerase chain reaction, a unique difference between the fistula tract and control tissues was that Gli-2 (downstream signaling molecule of Shh) messenger RNA levels were much lower in the fistula tract than in the adjacent esophagus (P =.002). Surprisingly, in the culture experiments, the fistula tract was induced to branch by exogenous Shh. Such branching of the fistula was unexpected and further supports the presumed respiratory origin of the fistula tract because the normal lung, but not normal esophagus, branched in response to Shh. The Shh inhibitor had no effect, which indicated that defective signaling, rather than hyperfunctioning Shh, is critical to the nonbranching phenotype of the fistula tract in TEF. CONCLUSIONS: The recapitulation of respiratory developmental morphogenesis by the fistula tract of TEF in the presence of exogenous Shh, together with the quantitative reduction in normal, endogenous levels of Gli-2, strongly suggests that 1 mechanism for the formation of the fistula tract is the lack of proper Shh signaling because of Gli-2 deficiency, with subsequent straight, nonbranching caudal growth of the fistula tract. This deficiency can be rescued by excess exogenous Shh, thus reestablishing respiratory morphogenesis.


Asunto(s)
Atresia Esofágica/embriología , Atresia Esofágica/etiología , Transducción de Señal , Fístula Traqueoesofágica/embriología , Fístula Traqueoesofágica/etiología , Transactivadores/metabolismo , Animales , Doxorrubicina , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario y Fetal/efectos de los fármacos , Atresia Esofágica/inducido químicamente , Femenino , Proteínas Hedgehog , Factores de Transcripción de Tipo Kruppel , Técnicas de Cultivo de Órganos , Embarazo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Fístula Traqueoesofágica/inducido químicamente , Transactivadores/farmacología , Factores de Transcripción/genética , Proteína Gli2 con Dedos de Zinc
17.
J Pediatr Surg ; 38(3): 474-7; discussion 474-7, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12632370

RESUMEN

BACKGROUND/PURPOSE: The Adriamycin-induced rat model of esophageal atresia and tracheoesophageal fistula (EA/TEF) provides a reliable system for the study of EA/TEF pathogenesis. The authors previously hypothesized that faulty branching lung morphogenesis pathways were a critical component of its pathogenesis. The authors have found evidence for faulty fibroblast growth factor (FGF) signaling related to epithelial-mesenchymal interactions in the fistula tract. To better define FGF signaling, the differential expression of FGF ligands and their receptors between lung, fistula tract, and esophagus are described. METHODS: Time-dated pregnant, Sprague-Dawley rats were injected with Adriamycin (2 mg/kg intraperitoneally) on days 6 through 9 of gestation. Tissues were processed for histology and reverse transcriptase polymerase chain reaction. FGF-1, -7 and -10 were measured from whole lung, fistula tract, and esophagus of TEF or normal embryos. Expression of FGF2RIIIb and FGF2RIIIc receptors was measured in isolated epithelium and mesenchyme of lung and fistula tract of TEF embryos as well as lung and esophagus from normal controls. RESULTS: FGF-1 mRNA was present in the fistula tract and normal and Adriamycin-exposed lung but absent from whole esophagus. Interestingly, FGF-7 mRNA was present only in normal lung. FGF-10 was present in all tissues examined. FGF2RIIIb mRNA was absent in fistula mesenchyme but present in all other tissues examined. However, the splice variant FGF2RIIIc mRNA was present in all tissues examined. CONCLUSIONS: These findings support defective FGF signaling in the rat model of EA/TEF. Absence of FGF-7 mRNA in Adriamycin-exposed tissues suggests the primary effect of Adriamycin may be to inhibit FGF-7 expression. Moreover, absence of FGF2RIIIb in fistula mesenchyme may be caused by loss of positive feedback from FGF-7, its normal obligate ligand. Understanding these specific defects in FGF signaling may provide insight into faulty mechanisms of EA/TEF.


Asunto(s)
Anomalías Inducidas por Medicamentos/genética , Anomalías Múltiples/genética , Doxorrubicina/toxicidad , Atresia Esofágica/genética , Proteínas Fetales/fisiología , Factores de Crecimiento de Fibroblastos/fisiología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Fístula Traqueoesofágica/genética , Anomalías Inducidas por Medicamentos/etiología , Anomalías Inducidas por Medicamentos/metabolismo , Anomalías Inducidas por Medicamentos/patología , Anomalías Múltiples/inducido químicamente , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología , Animales , Modelos Animales de Enfermedad , Epitelio/metabolismo , Atresia Esofágica/inducido químicamente , Atresia Esofágica/embriología , Esófago/embriología , Esófago/metabolismo , Femenino , Proteínas Fetales/biosíntesis , Proteínas Fetales/genética , Factor 7 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/biosíntesis , Factores de Crecimiento de Fibroblastos/deficiencia , Factores de Crecimiento de Fibroblastos/genética , Pulmón/embriología , Pulmón/metabolismo , Mesodermo/química , Morfogénesis/efectos de los fármacos , Embarazo , Ratas , Ratas Sprague-Dawley , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/biosíntesis , Receptores de Factores de Crecimiento de Fibroblastos/deficiencia , Receptores de Factores de Crecimiento de Fibroblastos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tráquea/embriología , Tráquea/metabolismo , Fístula Traqueoesofágica/inducido químicamente , Fístula Traqueoesofágica/embriología
18.
Diabetes ; 51(11): 3229-36, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12401714

RESUMEN

The embryonic pancreas is thought to develop from pluripotent endodermal cells that give rise to endocrine and exocrine cells. A key guidance mechanism for pancreatic development has previously been found to be epithelial-mesenchymal interaction. Interactions within the epithelium, however, have not been well studied. Glucagon is the earliest peptide hormone present at appreciable levels in the developing pancreatic epithelium (embryonic day [E]-9.5 in mouse). Insulin accumulation begins slightly later (E11 in mouse), followed by a rapid accumulation during the "second wave" of insulin differentiation ( approximately E15). Here we found that blocking early expression and function of glucagon, but not GLP-1, an alternate gene product of preproglucagon mRNA, prevented insulin-positive differentiation in early embryonic (E11) pancreas. These results suggest a novel concept and a key role for glucagon in the paracrine induction of differentiation of other pancreatic components in the early embryonic pancreas.


Asunto(s)
Insulina/genética , Islotes Pancreáticos/embriología , Páncreas/embriología , Animales , Secuencia de Bases , Diferenciación Celular/efectos de los fármacos , Cartilla de ADN , Desarrollo Embrionario y Fetal , Femenino , Edad Gestacional , Glucagón/genética , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Ratones , Oligodesoxirribonucleótidos Antisentido/farmacología , Páncreas/citología , Reacción en Cadena de la Polimerasa , Embarazo , Proglucagón , Precursores de Proteínas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Gastroenterology ; 123(4): 1331-40, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12360493

RESUMEN

BACKGROUND & AIMS: The early embryonic pancreas gives rise to exocrine (ducts and acini) and endocrine lineages. Control of exocrine differentiation is poorly understood, but may be a critical avenue through which to manipulate pancreatic ductal carcinoma. Retinoids have been shown to change the character of pancreatic ductal cancer cells to a less malignant phenotype. We have shown that 9-cis retinoic acid (9cRA) inhibits acinar differentiation in the developing pancreas, in favor of ducts, and we wanted to determine the role of retinoids in duct versus acinar differentiation. METHODS: We used multiple culture systems for the 11-day embryonic mouse pancreas. RESULTS: Retinoic acid receptor (RAR)-selective agonists mimicked the acinar suppressive effect of 9cRA, suggesting that RAR-RXR heterodimers were critical to ductal differentiation. RARalpha was only expressed in mesenchyme, whereas RXRalpha was expressed in epithelium and mesenchyme. Retinaldehyde dehydrogenase 2, a critical enzyme in retinoid synthesis, was expressed only in pancreatic epithelium. 9cRA did not induce ductal differentiation in the absence of mesenchyme, implicating a requirement for mesenchyme in 9cRA effects. Mesenchymal laminin is necessary for duct differentiation, and retinoids are known to enhance laminin expression. In 9cRA-treated pancreas, immunohistochemistry for laminin showed a strong band of staining around ducts, and blockage of laminin signaling blocked all 9cRA effects. Western blot and RT-PCR of pancreatic mesenchyme showed laminin-beta1 protein and mRNA induction by 9cRA. CONCLUSIONS: Retinoids regulate exocrine lineage selection through epithelial-mesenchymal interactions, mediated through up-regulation of mesenchymal laminin-1.


Asunto(s)
Antineoplásicos/farmacología , Páncreas/citología , Páncreas/embriología , Transducción de Señal/fisiología , Tretinoina/farmacología , Alitretinoína , Animales , Comunicación Celular/efectos de los fármacos , Comunicación Celular/fisiología , Diferenciación Celular/fisiología , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/fisiología , Células Cultivadas , Células Epiteliales/citología , Regulación del Desarrollo de la Expresión Génica/fisiología , Técnicas In Vitro , Laminina/genética , Laminina/metabolismo , Mesodermo/citología , Ratones , ARN Mensajero/análisis , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Receptor alfa de Ácido Retinoico , Receptores X Retinoide , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Regulación hacia Arriba/fisiología
20.
Anat Rec ; 267(3): 220-4, 2002 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-12115271

RESUMEN

The embryogenesis of tracheoesophageal anomalies remains controversial. The purpose of this study was to better define the embryogenesis of developing esophageal atresia with tracheoesophageal fistula (EA/TEF), with specific attention to the controversial issue of whether a discontinuity exists in the foregut during its development of EA/TEF. Pregnant outbred rats were injected with adriamycin (2 mg/kg i.p.) on days 6-9 of gestation (E6-E9). At E12.5 and 13.5, microdissection of the entire foregut was performed. Foreguts were examined by phase microscopy, and serial, precisely transverse sections were created for hematoxylin and eosin (H&E) staining. Gross microdissection of the developing foregut at E12.5 (n = 9) revealed a blind-ending, bulbous fistula tract arising from the middle branch of the tracheal trifurcation (as seen by direct and phase microscopy). No connection with the gut could be appreciated at E12.5, but by E13.5 (n = 10) there was an obvious connection between the fistula and the stomach. Serial H&E transverse sections also demonstrated a blind-ending fistula tract arising from the trachea at E12.5. This fistula tract was clearly discontinuous from the developing stomach, which appeared much further caudal to the end of the fistula tract. These results strongly support a model of experimental TEF wherein the fistula tract arises from a trifurcation of the trachea, and (only during a specific gestational window between days 12.5 and 13.5) there is discontinuity between the fistula tract and the stomach. By day 13.5, the fistula joins with the stomach anlage. These observations in the developing EA/TEF should help to resolve the controversy about the mechanism of EA/TEF formation.


Asunto(s)
Atresia Esofágica/embriología , Fístula Traqueoesofágica/embriología , Animales , Modelos Animales de Enfermedad , Doxorrubicina , Desarrollo Embrionario y Fetal , Atresia Esofágica/complicaciones , Atresia Esofágica/patología , Esófago/embriología , Femenino , Embarazo , Ratas , Ratas Sprague-Dawley , Fístula Traqueoesofágica/complicaciones , Fístula Traqueoesofágica/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...