Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
1.
Cell Stem Cell ; 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38996472

RESUMEN

Recent advances have made modeling human small intestines in vitro possible, but it remains a challenge to recapitulate fully their structural and functional characteristics. We suspected interstitial flow within the intestine, powered by circulating blood plasma during embryonic organogenesis, to be a vital factor. We aimed to construct an in vivo-like multilayered small intestinal tissue by incorporating interstitial flow into the system and, in turn, developed the micro-small intestine system by differentiating definitive endoderm and mesoderm cells from human pluripotent stem cells simultaneously on a microfluidic device capable of replicating interstitial flow. This approach enhanced cell maturation and led to the development of a three-dimensional small intestine-like tissue with villi-like epithelium and an aligned mesenchymal layer. Our micro-small intestine system not only overcomes the limitations of conventional intestine models but also offers a unique opportunity to gain insights into the detailed mechanisms underlying intestinal tissue development.

2.
Nat Commun ; 15(1): 458, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38302444

RESUMEN

In the central nervous system, astrocytes enable appropriate synapse function through glutamate clearance from the synaptic cleft; however, it remains unclear how astrocytic glutamate transporters function at peri-synaptic contact. Here, we report that Down syndrome cell adhesion molecule (DSCAM) in Purkinje cells controls synapse formation and function in the developing cerebellum. Dscam-mutant mice show defects in CF synapse translocation as is observed in loss of function mutations in the astrocytic glutamate transporter GLAST expressed in Bergmann glia. These mice show impaired glutamate clearance and the delocalization of GLAST away from the cleft of parallel fibre (PF) synapse. GLAST complexes with the extracellular domain of DSCAM. Riluzole, as an activator of GLAST-mediated uptake, rescues the proximal impairment in CF synapse formation in Purkinje cell-selective Dscam-deficient mice. DSCAM is required for motor learning, but not gross motor coordination. In conclusion, the intercellular association of synaptic and astrocyte proteins is important for synapse formation and function in neural transmission.


Asunto(s)
Neuroglía , Neuronas , Animales , Ratones , Sistema de Transporte de Aminoácidos X-AG/metabolismo , Cerebelo/metabolismo , Ácido Glutámico/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Células de Purkinje/metabolismo , Sinapsis/metabolismo
3.
Cancer Med ; 12(18): 18611-18621, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37649318

RESUMEN

BACKGROUND: Resectable pancreatic cancer (RPC) is potentially resectable on admission, and the impact of neoadjuvant therapy on these tumors is controversial. Moreover, the safety and efficacy of neoadjuvant chemoradiotherapy with moderately hypofractionated intensity-modulated radiation therapy (NACIMRT) for RPC have not been studied. Here, we conducted a phase II study to evaluate the safety and efficacy of hypofractionated NACIMRT for RPC. METHODS: A total of 54 RPC patients were enrolled and treated according to the study protocol. We used moderately hypofractionated (45 Gy in 15 fractions) IMRT with gemcitabine to shorten the duration of radiotherapy and reduce gastrointestinal toxicity. The primary endpoint was overall survival (OS), and we subsequently analyzed the microscopically margin-negative resection (R0) rate, disease-free survival (DFS), and histologic effects and safety of NACIMRT. RESULTS: Median OS for the cohort was 40.0 months. Forty-two patients (77.8%) underwent pancreatectomy after NACIMRT. Median DFS was 20.3 months. The R0 resection rate was 95.2% (40/42) per protocol and 85.2% (46/54) for the cohort. There were no intervention-related deaths during the study period. Local treatment response, as assessed by the CAP classification, showed no residual tumor in 4.8% of patients. Overall, 23.9% of patients experienced CTCAE grade 3 or 4 during NACIMRT. Adjuvant therapy was initiated in 88% of patients undergoing resection. Postoperative complications grade ≥3b on the Clavien-Dindo scale occurred in 4.8% of patients. CA19-9 level at enrollment was an independent prognostic factor for OS and DFS. CONCLUSIONS: This is the first prospective study of hypofractionated IMRT as neoadjuvant therapy for RPC. Hypofractionated NACIMRT for RPC could be safely introduced with a high induction rate of adjuvant chemotherapy, with an overall survival of 40.0 months.

5.
PLoS One ; 17(11): e0275600, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36378656

RESUMEN

Cell therapies using human induced pluripotent stem cell (hiPSC)-derived nephron progenitor cells (NPCs) are expected to ameliorate acute kidney injury (AKI). However, using hiPSC-derived NPCs clinically is a challenge because hiPSCs themselves are tumorigenic. LIN28A, ESRG, CNMD and SFRP2 transcripts have been used as a marker of residual hiPSCs for a variety of cell types undergoing clinical trials. In this study, by reanalyzing public databases, we found a baseline expression of LIN28A, ESRG, CNMD and SFRP2 in hiPSC-derived NPCs and several other cell types, suggesting LIN28A, ESRG, CNMD and SFRP2 are not always reliable markers for iPSC detection. As an alternative, we discovered a lncRNA marker gene, MIR302CHG, among many known and unknown iPSC markers, as highly differentially expressed between hiPSCs and NPCs, by RNA sequencing and quantitative RT-PCR (qRT-PCR) analyses. Using MIR302CHG as an hiPSC marker, we constructed two assay methods, a combination of magnetic bead-based enrichment and qRT-PCR and digital droplet PCR alone, to detect a small number of residual hiPSCs in NPC populations. The use of these in vitro assays could contribute to patient safety in treatments using hiPSC-derived cells.


Asunto(s)
Células Madre Pluripotentes Inducidas , Neuroblastoma , ARN Largo no Codificante , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Diferenciación Celular/genética , Técnicas In Vitro , Nefronas , ARN Largo no Codificante/metabolismo , Neuroblastoma/metabolismo
6.
BMC Cancer ; 22(1): 119, 2022 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-35093003

RESUMEN

BACKGROUND: Borderline resectable pancreatic cancer (BRPC) is a category of pancreatic cancer that is anatomically widely spread, and curative resection is uncommon with upfront surgery. Intensity-modulated radiation therapy (IMRT) is a form of radiation therapy that delivers precise radiation to a tumor while minimizing the dose to surrounding normal tissues. Here, we conducted a phase 2 study to estimate the curability and efficacy of neoadjuvant chemoradiotherapy using IMRT (NACIMRT) for patients with BRPC with arterial abutment (BRPC-A). METHODS: A total of 49 BRPC-A patients were enrolled in this study and were treated at our hospital according to the study protocol between June 2013 and March 2021. The primary endpoint was microscopically margin-negative resection (R0) rates and we subsequently analyzed safety, histological effect of the treatment as well as survivals among patients with NACIMRT. RESULTS: Twenty-nine patients (59.2%) received pancreatectomy after NACIMRT. The R0 rate in resection patients was 93.1% and that in the whole cohort was 55.1%. No mortality was encountered. Local therapeutic effects as assessed by Evans classification showed good therapeutic effect (Grade 1, 3.4%; Grade 2a, 31.0%; Grade 2b, 48.3%; Grade 3, 3.4%; Grade 4, 3.4%). Median disease-free survival was 15.5 months. Median overall survival in the whole cohort was 35.1 months. The only independent prognostic pre-NACIMRT factor identified was serum carbohydrate antigen 19-9 (CA19-9) > 400 U/ml before NACIMRT. CONCLUSIONS: NACIMRT showed preferable outcome without significant operative morbidity for BRPC-A patients. NACIMRT contributes to good local tumor control, but a high initial serum CA19-9 implies poor prognosis even after neoadjuvant treatment. TRIAL REGISTRATION: UMIN-CTR Clinical Trial: https://upload.umin.ac.jp/cgi-open-bin/ctr_e/ctr_view.cgi?recptno=R000011776 Registration number: UMIN000010113. Date of first registration: 01/03/2013.


Asunto(s)
Terapia Neoadyuvante , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/terapia , Radioterapia de Intensidad Modulada , Anciano , Antígenos de Carbohidratos Asociados a Tumores/sangre , Arterias , Femenino , Humanos , Masculino , Márgenes de Escisión , Persona de Mediana Edad , Pancreatectomía , Estudios Prospectivos , Resultado del Tratamiento
7.
EMBO J ; 40(14): e105712, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34057742

RESUMEN

During development, neural progenitors are in proliferative and immature states; however, the molecular machinery that cooperatively controls both states remains elusive. Here, we report that cyclin D1 (CCND1) directly regulates both proliferative and immature states of cerebellar granule cell progenitors (GCPs). CCND1 not only accelerates cell cycle but also upregulates ATOH1 protein, an essential transcription factor that maintains GCPs in an immature state. In cooperation with CDK4, CCND1 directly phosphorylates S309 of ATOH1, which inhibits additional phosphorylation at S328 and consequently prevents S328 phosphorylation-dependent ATOH1 degradation. Additionally, PROX1 downregulates Ccnd1 expression by histone deacetylation of Ccnd1 promoter in GCPs, leading to cell cycle exit and differentiation. Moreover, WNT signaling upregulates PROX1 expression in GCPs. These findings suggest that WNT-PROX1-CCND1-ATOH1 signaling cascade cooperatively controls proliferative and immature states of GCPs. We revealed that the expression and phosphorylation levels of these molecules dynamically change during cerebellar development, which are suggested to determine appropriate differentiation rates from GCPs to GCs at distinct developmental stages. This study contributes to understanding the regulatory mechanism of GCPs as well as neural progenitors.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Cerebelo/crecimiento & desarrollo , Cerebelo/metabolismo , Ciclina D1/metabolismo , Gránulos Citoplasmáticos/metabolismo , Fosforilación/fisiología , Células Madre/metabolismo , Animales , Ciclo Celular/genética , Diferenciación Celular/fisiología , División Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Proteínas Hedgehog/metabolismo , Ratones , Neurogénesis/fisiología , Transducción de Señal/fisiología , Factores de Transcripción
8.
Nat Commun ; 12(1): 560, 2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33495473

RESUMEN

The squamous-columnar junction (SCJ) is a boundary consisting of precisely positioned transitional epithelium between the squamous and columnar epithelium. Transitional epithelium is a hotspot for precancerous lesions, and is therefore clinically important; however, the origins and physiological properties of transitional epithelium have not been fully elucidated. Here, by using mouse genetics, lineage tracing, and organoid culture, we examine the development of the SCJ in the mouse stomach, and thus define the unique features of transitional epithelium. We find that two transcription factors, encoded by Sox2 and Gata4, specify primitive transitional epithelium into squamous and columnar epithelium. The proximal-distal segregation of Sox2 and Gata4 expression establishes the boundary of the unspecified transitional epithelium between committed squamous and columnar epithelium. Mechanistically, Gata4-mediated expression of the morphogen Fgf10 in the distal stomach and Sox2-mediated Fgfr2 expression in the proximal stomach induce the intermediate regional activation of MAPK/ERK, which prevents the differentiation of transitional epithelial cells within the SCJ boundary. Our results have implications for tissue regeneration and tumorigenesis, which are related to the SCJ.


Asunto(s)
Células Epiteliales/metabolismo , Factor de Transcripción GATA4/genética , Regulación de la Expresión Génica , Uniones Intercelulares/genética , Sistema de Señalización de MAP Quinasas/genética , Factores de Transcripción SOXB1/genética , Animales , Células Cultivadas , Femenino , Factor de Transcripción GATA4/metabolismo , Mucosa Gástrica/metabolismo , Queratina-7/genética , Queratina-7/metabolismo , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Ratones Transgénicos , Factores de Transcripción SOXB1/metabolismo
9.
Endocr Relat Cancer ; 27(7): 431-439, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32369772

RESUMEN

Although pancreatic neuroendocrine neoplasms (PanNENs) are generally indolent, patients with distant metastasis have a dismal prognosis. Recently, the autophagy inhibitor chloroquine (CQ) has been shown to suppress the tumour growth of PanNENs, but the detailed mechanisms have not been elucidated. Furthermore, these results were obtained from poorly differentiated cell lines rather than well-differentiated cell lines, which is the most prevalent type in this tumour. To explore the mechanism and efficacy of CQ on PanNENs, we applied CQ to cell lines and evaluated the resulting apoptosis and endoplasmic reticulum (ER) stress. CQ treatment induced ER stress, and an unfolded protein response was activated through the PERK-eIF2α-ATF4 pathway, resulting in the expression of the pro-apoptotic protein C/EBP homologous protein (CHOP), which reflects ER-stress-mediated apoptotic cell death. Furthermore, hydroxychloroquine (HCQ) was effective in Men1 heterozygous-deficient (Men1+/ΔN3-8) mice, a mouse PanNEN model that is considered to correspond to human low-grade PanNEN. HCQ administration decreased tumour size in Men1+/ΔN3-8 mice. In the HCQ group, histological analyses revealed that proliferative activity was unchanged, but apoptosis was accelerated, accompanied by CHOP expression. These results suggest that autophagy inhibition by CQ/HCQ could be used for the treatment of PanNEN, including the well-differentiated type.


Asunto(s)
Antirreumáticos/uso terapéutico , Carcinoma Neuroendocrino/tratamiento farmacológico , Cloroquina/uso terapéutico , Estrés del Retículo Endoplásmico/inmunología , Animales , Antirreumáticos/farmacología , Apoptosis , Autofagia , Cloroquina/farmacología , Humanos , Ratones
10.
Scand J Gastroenterol ; 55(4): 479-484, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32223573

RESUMEN

Background: In the treatment of metastatic pancreatic neuroendocrine tumors (PanNETs), surgical resection is the first choice if curative resection is expected. However, most patients develop recurrence after resection of liver metastasis. Because one of the benefits of resection is to gain a tumor-free period for the patients, it is important to identify which patients achieve longer recurrence-free survival (RFS) by resection. In this study, the clinicopathological factors associated with RFS after resection of metastatic PanNETs in the liver were evaluated to identify the patient group that is suitable for resection.Methods: Consecutively diagnosed patients with PanNET liver metastasis with resection at our hospital from January 2000 to July 2019 were evaluated. A total of 26 metastatic PanNET patients with primary liver resections were evaluated. The median follow-up time was 48.3 months.Results: There were 18 NET recurrences of the total 26 resections, with a median RFS of 17.9 months. Independent risk factors for short RFS were a high Ki67 index (p = .009) and the number of resected tumors (p = .045). When the cut-off value for the Ki67 index was 5.0% and that for the number of resected tumors was 6, Ki67 > 5.0% tumors had shorter RFS (4.9 months vs. 38.2 months p = .006), and patients with tumors > = 7 tumors had shorter RFS (4.7 months vs. 27.5 months p = .001).Conclusions: These findings indicate that good candidates for resection of metastatic tumors of PanNETs could be patients with low Ki67 tumors and a small number of metastatic tumors.


Asunto(s)
Antígeno Ki-67/metabolismo , Neoplasias Hepáticas/secundario , Recurrencia Local de Neoplasia/mortalidad , Tumores Neuroendocrinos/mortalidad , Neoplasias Pancreáticas/mortalidad , Adulto , Anciano , Femenino , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/cirugía , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/patología , Tumores Neuroendocrinos/cirugía , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/cirugía , Factores de Riesgo , Tasa de Supervivencia
11.
Stem Cell Res ; 44: 101754, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32179491

RESUMEN

In the multi-step differentiation protocol used to generate pancreatic endocrine cells from human pluripotent stem cells, the induction of NGN3+ endocrine precursors from the PDX1+/NKX6.1+ pancreatic endoderm is crucial for efficient endocrine cell production. Here, we demonstrate that transient, not prolonged FOXO1 inhibition results in enhanced NGN3+ endocrine precursors and hormone-producing cell production. FOXO1 inhibition does not directly induce NGN3 expression but stimulates PDX1+/NKX6.1+ cell proliferation. NOTCH activity, whose suppression is important for Ngn3 expression, is not suppressed but Wnt signaling is stimulated by FOXO1 inhibition. Reversely, Wnt inhibition suppresses the effects of FOXO1 inhibitor. These findings indicate that FOXO1 and Wnt are involved in regulating the proliferation of PDX1+/NKX6.1+ pancreatic endoderm that gives rise to NGN3+ endocrine precursors.


Asunto(s)
Endodermo , Proteína Forkhead Box O1 , Células Madre Pluripotentes Inducidas , Vía de Señalización Wnt , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diferenciación Celular , Proteína Forkhead Box O1/genética , Proteínas de Homeodominio/genética , Humanos , Páncreas , Transactivadores
12.
FEBS Lett ; 593(16): 2226-2234, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31240701

RESUMEN

In this study, we used tamoxifen-inducible Elastase-Cre-mediated inactivation of pancreatic and duodenal homeobox1 (Pdx1), an indispensable gene during embryonic pancreatogenesis, to investigate the role of Pdx1 in adult pancreatic exocrine tissue. We found that Pdx1 depletion in approximately 50% of acinar cell mass did not show any macroscopic phenotype. Lineage tracing experiments revealed that the percentage of Pdx1-depleted cells did not change initially but gradually decreased, while the proliferation of Pdx1-preserved cells increased. Electron microscopic analysis showed the emergence of round-shaped mitochondria with less cristae, dilated ER lumen and increased number of autophagosomes but no apoptosis. Instead, Pdx1-depleted acinar cells became senescent. These findings indicate that intracellular stress caused by Pdx1 inactivation triggers the senescence-associated secretory phenotype to maintain organ homeostasis in this model.


Asunto(s)
Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Páncreas/citología , Tamoxifeno/farmacología , Transactivadores/genética , Transactivadores/metabolismo , Células Acinares/citología , Células Acinares/metabolismo , Animales , Proliferación Celular , Senescencia Celular , Silenciador del Gen , Ratones , Microscopía Electrónica , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Páncreas/metabolismo , Elastasa Pancreática/genética , Elastasa Pancreática/metabolismo
13.
Surg Today ; 49(7): 593-600, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30838442

RESUMEN

BACKGROUND: We evaluated the clinicopathological factors associated with lymph node metastasis in patients with non-functioning pancreatic neuroendocrine neoplasms (PanNENs), focusing on the risk factors and range of lymph node metastasis for tumors ≤ 2 cm in diameter. METHODS: The subjects of this study were patients with PanNENs consecutively diagnosed at our hospital between January, 2000 and June, 2018. We analyzed 69 patients who underwent R0 resection of a non-functioning sporadic PanNEN with no distant metastasis, as well as 43 patients with tumors ≤ 20 mm in radiological diameter. RESULTS: Nineteen patients (27.5%), including 7 (16.3%) with a small PanNEN, had lymph node metastasis. A large radiological diameter, a high Ki67 index, and cyst formation correlated significantly with positive lymph node metastasis. In patients with tumors ≤ 20 mm in diameter, a high Ki67 index correlated significantly with lymph node metastasis. When we set the cut-off Ki67 index as 3.3%, 2 of 43 patients had lymph node metastasis. Tumors in the uncinate process readily metastasized to the region around the superior mesenteric artery. CONCLUSIONS: These findings suggest that a high Ki67 index indicates a risk of lymph node metastasis for tumors ≤ 20 mm in diameter and that lymphadenectomy should be performed in the region spatially adjacent to the primary tumor.


Asunto(s)
Biomarcadores de Tumor/análisis , Antígeno Ki-67/análisis , Tumores Neuroendocrinos/diagnóstico , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/patología , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Escisión del Ganglio Linfático , Metástasis Linfática , Masculino , Arteria Mesentérica Superior/patología , Persona de Mediana Edad , Tumores Neuroendocrinos/cirugía , Neoplasias Pancreáticas/cirugía , Valor Predictivo de las Pruebas , Tomografía Computarizada por Rayos X
14.
Sci Rep ; 9(1): 1636, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30733468

RESUMEN

During embryogenesis, exocrine and endocrine pancreatic tissues are formed in distinct regions within the branched ductal structure in mice. We previously reported that exocrine-specific inactivation of Pdx1 by Elastase-Cre caused not only hypoplastic exocrine formation but also substantial endocrine defects resulting in diabetic phenotype, indicating the existence of an exocrine-driven factor(s) that regulates proper endocrine development. In this study, we identified Trefoil Factor 2 (TFF2) as an exocrine gene expressed from embryonic day 16.5 to adulthood in normal mice but significantly less in our Pdx1 mutants. Using in vitro explant culture of embryonic pancreatic tissue, we demonstrated that TFF2 prevented the apoptosis of insulin-producing cells but that antagonizing CXCR4, a known TFF2 receptor, suppressed this anti-apoptotic effect in the mutants. Furthermore, the antagonist in normal pancreatic tissue accelerated the apoptosis of insulin-producing cells, indicating that the TFF2/CXCR4 axis maintains embryonic insulin-producing cells in normal development. TFF2 also suppressed the apoptosis of Nkx6.1+ endocrine precursors in mutant pancreata, but this effect was unperturbed by the CXCR4 antagonist, suggesting the existence of an unknown receptor for TFF2. These findings suggest TFF2 is a novel exocrine factor that supports the survival of endocrine cells in the multiple stages of organogenesis through distinct receptors.


Asunto(s)
Páncreas/citología , Páncreas/embriología , Factor Trefoil-2/genética , Animales , Apoptosis/fisiología , Linaje de la Célula , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Insulina/metabolismo , Ratones Noqueados , Organogénesis , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Transactivadores/genética , Factor Trefoil-2/metabolismo
15.
Sci Rep ; 8(1): 15812, 2018 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-30361559

RESUMEN

Pancreas transcription factor 1 subunit alpha (PTF1A) is one of the key regulators in pancreatogenesis. In adults, it transcribes digestive enzymes, but its other functions remain largely unknown. Recent conditional knockout studies using Ptf1aCreER/floxed heterozygous mouse models have found PTF1A contributes to the identity maintenance of acinar cells and prevents tumorigenesis caused by the oncogenic gene Kras. However, Ptf1a heterozygote is known to behave differently from homozygote. To elucidate the effects of Ptf1a homozygous loss, we prepared Elastase-CreERTM; Ptf1afloxed/floxed mice and found that homozygous Ptf1a deletion in adult acinar cells causes severe apoptosis. Electron microscopy revealed endoplasmic reticulum (ER) stress, a known cause of unfolded protein responses (UPR). We confirmed that UPR was upregulated by the activating transcription factor 6 (ATF6) and protein kinase RNA (PKR)-like endoplasmic reticulum kinase (PERK) pathways, but not the inositol requiring enzyme 1 (IRE1) pathway. Furthermore, we detected the expression of CCAAT-enhancer-binding protein (C/EBP) homologous protein (CHOP), a pro-apoptotic factor, indicating the apoptosis was induced through UPR. Our homozygous model helps clarify the role PTF1A has on the homeostasis and pathogenesis of exocrine pancreas in mice.


Asunto(s)
Células Acinares/metabolismo , Apoptosis , Estrés del Retículo Endoplásmico , Páncreas Exocrino/patología , Factores de Transcripción/metabolismo , Factor de Transcripción Activador 6/metabolismo , Animales , Linaje de la Célula , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/ultraestructura , Ratones Noqueados , Empalme del ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Transcripción CHOP/metabolismo , Factores de Transcripción/deficiencia , Regulación hacia Arriba/genética , Proteína 1 de Unión a la X-Box/genética , Proteína 1 de Unión a la X-Box/metabolismo
16.
Cell Rep ; 24(1): 79-94, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29972793

RESUMEN

The mammalian brain undergoes sexual differentiation by gonadal hormones during the perinatal critical period. However, the machinery at earlier stages has not been well studied. We found that Ptf1a is expressed in certain neuroepithelial cells and immature neurons around the third ventricle that give rise to various neurons in several hypothalamic nuclei. We show that conditional Ptf1a-deficient mice (Ptf1a cKO) exhibit abnormalities in sex-biased behaviors and reproductive organs in both sexes. Gonadal hormone administration to gonadectomized animals revealed that the abnormal behavior is caused by disorganized sexual development of the knockout brain. Accordingly, expression of sex-biased genes was severely altered in the cKO hypothalamus. In particular, Kiss1, important for sexual differentiation of the brain, was drastically reduced in the cKO hypothalamus, which may contribute to the observed phenotypes in the Ptf1a cKO. These findings suggest that forebrain Ptf1a is one of the earliest regulators for sexual differentiation of the brain.


Asunto(s)
Prosencéfalo/embriología , Diferenciación Sexual , Factores de Transcripción/metabolismo , Animales , Linaje de la Célula , Embrión de Mamíferos/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Gónadas/anomalías , Hipotálamo/embriología , Hipotálamo/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Diferenciación Sexual/genética , Conducta Sexual Animal , Factores de Transcripción/deficiencia
17.
Nat Commun ; 9(1): 2081, 2018 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-29802314

RESUMEN

The faithful shutdown of the somatic program occurs in the early stage of reprogramming. Here, we examined the effect of in vivo reprogramming on Kras-induced cancer development. We show that the transient expression of reprogramming factors (1-3 days) in pancreatic acinar cells results in the transient repression of acinar cell enhancers, which are similarly observed in pancreatitis. We next demonstrate that Kras and p53 mutations are insufficient to induce ERK signaling in the pancreas. Notably, the transient expression of reprogramming factors in Kras mutant mice is sufficient to induce the robust and persistent activation of ERK signaling in acinar cells and rapid formation of pancreatic ductal adenocarcinoma. In contrast, the forced expression of acinar cell-related transcription factors inhibits the pancreatitis-induced activation of ERK signaling and development of precancerous lesions in Kras-mutated acinar cells. These results underscore a crucial role of dedifferentiation-associated epigenetic regulations in the initiation of pancreatic cancers.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Transformación Celular Neoplásica/patología , Regulación Neoplásica de la Expresión Génica , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Células Acinares/metabolismo , Células Acinares/patología , Animales , Carcinoma Ductal Pancreático/patología , Transformación Celular Neoplásica/genética , Reprogramación Celular/genética , Epigénesis Genética , Femenino , Humanos , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Transgénicos , Células Madre Embrionarias de Ratones , Mutación , Páncreas/citología , Páncreas/patología , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas p21(ras)/genética , Estómago/patología , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , alfa-Fetoproteínas/metabolismo
18.
Ann Surg Oncol ; 25(2): 528-534, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29164413

RESUMEN

BACKGROUND: The primary site of a neuroendocrine neoplasms (NEN) around the head of the pancreas is sometimes difficult to assess before resection, and the characteristics of the primary site around this region have not been elucidated for recurrence after curative resection. In this study, the clinicopathologic characteristics of pancreatic NEN (PanNEN) and duodenal NEN (DuNEN) were evaluated, and the risk factors as well as the recurrence types after resection were investigated. METHODS: Consecutively diagnosed NEN patients at the authors' hospital from January 2000 to July 2016 were evaluated in this study. For 117 PanNEN patients and 31 non-ampullary DuNEN patients, R0 resection was achieved. The median follow-up period was 8.1 years. RESULTS: In this study, 27 PanNEN patients (23.1%) had recurrences, with a median disease-free survival (DFS) of 133 months, whereas 11 DuNEN patients (35.5%) had recurrences, with a median DFS of 156 months. The PanNEN patients tended to have primary recurrence in the liver (85.2%), followed by lymph nodes (11.1%). The independent risk factors for short DFS were lymph node metastasis at resection (p = 0.001) and microvascular invasion (p = 0.048). In contrast, the DuNEN patients were likely to have lymph node metastasis (81.8%). The independent risk factors for short DFS were lymph node metastasis at resection (p = 0.003) and large diameter (p = 0.013). CONCLUSIONS: Most initial recurrences of PanNEN occurred in the liver, whereas those of DuNEN appeared in lymph nodes, suggesting that proper diagnosis of the primary site and appropriate imaging methods for surveillance after resection are necessary.


Asunto(s)
Neoplasias Duodenales/cirugía , Recurrencia Local de Neoplasia/diagnóstico , Tumores Neuroendocrinos/cirugía , Pancreatectomía/mortalidad , Neoplasias Pancreáticas/cirugía , Pancreaticoduodenectomía/mortalidad , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Neoplasias Duodenales/patología , Femenino , Estudios de Seguimiento , Humanos , Metástasis Linfática , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Recurrencia Local de Neoplasia/mortalidad , Tumores Neuroendocrinos/secundario , Neoplasias Pancreáticas/patología , Pronóstico , Estudios Retrospectivos , Tasa de Supervivencia , Adulto Joven
19.
Anticancer Res ; 37(12): 7023-7030, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29187490

RESUMEN

BACKGROUND/AIM: Intensity-modulated radiation therapy (IMRT) is a form of radiation therapy that allows accurate irradiation with reduced damage to surrounding tissues. Here, we analyzed borderline-resectable pancreatic cancer (BRPC) with arterial abutment (BR-A) patients with IMRT as neoadjuvant therapy and performed comparisons with patients with conventional RT to clarify the advantages of IMRT as a neoadjuvant therapy. PATIENTS AND METHODS: Thirty BR-A patients treated at our hospital between January 2012 and December 2015 were divided into two groups: 12 patients underwent conventional 3D-RT before resection (RT group); and 18 patients underwent IMRT before resection (IMRT group). We analyzed safety, tumor resection rate, histological classification of the tumor and overall survival. RESULTS: The R0 rate was 84% for the IMRT group and 83% for the RT group. Local therapeutic effects as assessed by Evans classification showed a higher local control rate in the IMRT group (Grade: 1, 0%; 2a, 25%; 2b, 41.6%; 3, 17%; 4, 8%) than in the RT group (Grade: 1, 17%; 2a, 50%; 2b, 17%; 3, 17%; 4, 0%). The cumulative dose of S1 treatment as adjuvant therapy was much smaller in the RT group (18.3%) compared to that in the IMRT group (57.1%, p=0.047), and with better subsequent overall survival rate (MST 32 months vs. 13.8 months, p=0.0273). CONCLUSION: The IMRT group showed a better control rate than the RT group. The neoadjuvant IMRT has advantages of higher completion rate of adjuvant chemotherapy with better nutritional status and better subsequent overall survival rate (OS).


Asunto(s)
Arterias/efectos de la radiación , Neoplasias Pancreáticas/radioterapia , Radioterapia Conformacional/métodos , Radioterapia de Intensidad Modulada/métodos , Anciano , Arterias/patología , Quimioradioterapia , Femenino , Humanos , Masculino , Persona de Mediana Edad , Terapia Neoadyuvante , Páncreas/efectos de los fármacos , Páncreas/efectos de la radiación , Páncreas/cirugía , Neoplasias Pancreáticas/irrigación sanguínea , Estudios Prospectivos , Análisis de Supervivencia , Resultado del Tratamiento
20.
Surg Case Rep ; 3(1): 82, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28730577

RESUMEN

BACKGROUND: Although pancreatic neuroendocrine tumors generally have a far better prognosis relative to pancreatic cancer, the varied manifestations lead to treatment-related challenges. Everolimus therapy is generally recommended for patients with advanced pancreatic neuroendocrine tumors; however, its efficacy in a neoadjuvant setting remains unclear. Here we present a case of a giant pancreatic neuroendocrine tumor with a portal tumor thrombus that became resectable after everolimus therapy. CASE PRESENTATION: A 62-year-old woman was admitted to our hospital for surgical resection of a giant pancreatic neuroendocrine tumor in the left upper abdomen. Unfortunately, she was ineligible for surgery because the tumor had extended near the hepatic hilus in the portal vein, and she was administered everolimus (10 mg/day). After 2 years of this therapy, the extent of portal vein involvement had decreased, despite the lack of significant changes in the tumor size, and the hepatic hilus became free of disease. She was subsequently referred to us for resection via distal pancreatectomy with portal vein reconstruction because the tumor had begun to grow slowly. Pathological review identified a grade 2 neuroendocrine tumor with no lymph node metastasis. The patient's postoperative course was uneventful, and she has remained recurrence-free for 27 months, despite a lack of additional treatment. CONCLUSIONS: Our experience suggests that everolimus could be useful for neoadjuvant therapy in cases of locally advanced pancreatic neuroendocrine tumor.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA