Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Horm Metab Res ; 55(6): 426-431, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36758614

RESUMEN

Repressor element-1 silencing transcription factor (Rest) is not expressed in pancreatic beta cells and neuronal cells. However, Rest4, a truncated form of Rest, is expressed in high passaged MIN6 (HP-MIN6) cells, a pancreatic beta cell line that lost glucose-responsive insulin secretion. Rest4 is also expressed in injured MIN6 cells and isolated islets. Herein, the forced expression of dominant negative form of Rest in HP-MIN6 cells was subjected to microarray analysis of gene expression to investigate the role of Rest4 gene in MIN6 cells. Furthermore, the forced expression of Rest4 gene in MIN6 cells was subjected to microarray analysis of gene expression to investigate the function of Rest4 in normal insulin-producing cells. The results showed that Rest4 inhibits cell proliferation and DNA and RNA metabolism and stimulates secretory mechanisms and nervous system gene expression. These findings suggest that Rest4 may act defensively against cellular injury in pancreatic beta cells.


Asunto(s)
Células Secretoras de Insulina , Proteínas Represoras , Animales , Ratones , Regulación de la Expresión Génica , Glucosa , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas Represoras/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
2.
Genes Cells ; 17(9): 758-67, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22845550

RESUMEN

Several reports have suggested that Foxo1, a key regulator in differentiation, growth and metabolism, is involved in pancreatic ß-cell function. However, detailed analyses have been hampered by a lack of Foxo1-deficient ß cells. To elucidate Foxo1's function in ß cells, we produced a ß-cell line with inducible Foxo1 deletion. We generated a conditional knockout mouse line, in which Cre recombinase deletes the Foxo1 gene. We then established a ß-cell line from an insulinoma induced in this knockout mouse by the ß-cell-specific expression of simian virus 40 T antigen. In this cell line, designated MIN6-Foxo1flox/flox, adenovirus-mediated Cre expression ablates the Foxo1 gene, generating MIN6-Foxo1-KO cells. Using these knockout and floxed cell lines, we found that Foxo1 ablation enhanced the glucose-stimulated insulin secretion (GSIS) at high glucose concentrations and enhanced ß-cell proliferation. We also conducted DNA microarray analyses of MIN6-Foxo1-KO cells infected with either an adenovirus vector expressing a constitutively active FOXO1 or a control vector and identified several Foxo1-regulated genes, including some known to be related to ß-cell function. These cells should be useful for further studies on Foxo1's roles in ß-cells and may lead to novel strategies for treating the impaired insulin secretion in type 2 diabetes mellitus.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Alelos , Animales , Antígenos Transformadores de Poliomavirus/genética , Antígenos Transformadores de Poliomavirus/metabolismo , Apoptosis , Línea Celular , Proliferación Celular , Quimera/genética , Quimera/metabolismo , Femenino , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Glucosa/farmacología , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Integrasas/genética , Integrasas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Virus 40 de los Simios/genética , Virus 40 de los Simios/metabolismo
3.
Genes Cells ; 15(8): 813-28, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20590823

RESUMEN

In a search for genes specifically expressed in mouse embryonic stem cells, we identified one we called Ces5. We found that it corresponded to the Ooep gene, which was recently reported to be expressed specifically in oocytes. Mouse Ces5/Ooep, also called Moep19 or Floped, encoded a 164-amino acid protein, which was detected in the cytoplasm of developing and mature oocytes and in embryos throughout the preimplantation period. To examine its function, we carried out targeted disruption of this gene. The Ces5/Ooep-null mice were grossly normal, but the females were infertile. Although the ovaries and ovulation appeared normal, the embryos from Ces5/Ooep-null females mated with wild-type males showed developmental arrest at the two- or four-cell stage. In addition, their first cleavage was considerably delayed and often asymmetrical. Thus, Ces5/Ooep is a maternal-effect gene. By electron microscopy, we found that the eggs from Ces5/Ooep-null females lacked oocyte cytoplasmic lattices (CPLs), which have long been predicted to function as a storage form for components that are maternally contributed to the early embryo. Further analysis showed that CES5/OOEP was directly associated with the CPLs. These results indicate that CES5/OOEP is an essential component of the CPLs and is required for embryonic development at the maternal-zygotic stage transition.


Asunto(s)
Citoplasma/metabolismo , Embrión de Mamíferos/embriología , Oocitos/citología , Oocitos/metabolismo , Proteínas de Unión al ARN/metabolismo , Cigoto/metabolismo , Secuencia de Aminoácidos , Animales , Embrión de Mamíferos/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Transcripción Genética/genética , Cigoto/citología
4.
Dev Biol ; 335(1): 216-27, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19735653

RESUMEN

We recently reported that the Gtsf1/Cue110 gene, a member of the evolutionarily conserved UPF0224 family, is expressed predominantly in male germ cells, and that the GTSF1/CUE110 protein is localized to the cytoplasm of these cells in the adult testis. Here, to analyze the roles of the Gtsf1/Cue110 gene in spermatogenesis, we produced Gtsf1/Cue110-null mice by gene targeting. The Gtsf1/Cue110-null mice grew normally and appeared healthy; however, the males were sterile due to massive apoptotic death of their germ cells after postnatal day 14. In contrast, the null females were fertile. Detailed analyses revealed that the Gtsf1/Cue110-null male meiocytes ceased meiotic progression before the zygotene stage. Thus, the Gtsf1/Cue110 gene is essential for spermatogenesis beyond the early meiotic phase. Furthermore, the loss of the Gtsf1/Cue110 gene caused increased transcription of the long interspersed nucleotide element (Line-1) and the intracisternal A-particle (IAP) retrotransposons, accompanied by demethylation of their promoter regions. These observations indicate that Gtsf1/Cue110 is required for spermatogenesis and involved in retrotransposon suppression in male germ cells.


Asunto(s)
Proteínas , Retroelementos , Espermatogénesis/fisiología , Testículo , Dedos de Zinc , Animales , Metilación de ADN , Femenino , Fertilidad/fisiología , Regulación de la Expresión Génica , Marcación de Gen , Péptidos y Proteínas de Señalización Intracelular , Masculino , Meiosis/fisiología , Ratones , Ratones Noqueados , Proteínas/genética , Proteínas/metabolismo , Testículo/citología , Testículo/fisiología
5.
Dev Biol ; 325(1): 238-48, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19014927

RESUMEN

The differentiation programs of spermatogenesis and oogenesis are largely independent. In the early stages, however, the mechanisms partly overlap. Here we demonstrated that a germ-cell-specific basic helix-loop-helix (bHLH) transcription factor gene, Sohlh2, is required for early spermatogenesis and oogenesis. SOHLH2 was expressed in mouse spermatogonia from the undifferentiated stage through differentiation and in primordial-to-primary oocytes. Sohlh2-null mice, produced by gene targeting, showed both male and female sterility, owing to the disrupted differentiation of mature (KIT(+)) spermatogonia and oocytes. The Sohlh2-null mice also showed the downregulation of genes involved in spermatogenesis and oogenesis, including the Sohlh1 gene, which is essential for these processes. Furthermore, we showed that SOHLH2 and SOHLH1 could form heterodimers. These observations suggested that SOHLH2 might coordinate with SOHLH1 to control spermatogonial and oocyte genes, including Sohlh1, to promote the differentiation of KIT(+) germ cells in vivo. This study lays the foundation for further dissection of the bHLH network that regulates early spermatogenesis and oogenesis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Oocitos/citología , Oocitos/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Espermatogonias/citología , Espermatogonias/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Western Blotting , Línea Celular , Femenino , Técnica del Anticuerpo Fluorescente , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Ratones , Oogénesis/genética , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espermatogénesis/genética , Testículo/citología , Testículo/metabolismo
6.
Antioxid Redox Signal ; 10(1): 43-9, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17949261

RESUMEN

The authors previously established a transgenic mouse line in the type 1 diabetes model, NOD mouse, in which thioredoxin (TRX), a redox protein, is overexpressed in pancreatic beta cells, and found that TRX overexpression slows the progression of type 1 diabetes. Recent reports on type 2 diabetes suggest that oxidative stress also degrades the function of beta cells. To elucidate whether TRX overexpression can prevent progressive beta cell failure from oxidative stress in type 2 diabetes, the authors transferred the TRX transgene from the NOD mouse onto a mouse model of type 2 diabetes, the db/db mouse. The progression of hyperglycemia and the reduction of body weight gain and insulin content of the db/db mouse were significantly suppressed by the TRX expression. Furthermore, TRX suppressed the reduction of Pdx-1 and MafA expression in the beta cells, which may be one of the cellular mechanisms for protecting beta cells from losing their insulin-secreting capacity. These results showed that TRX can protect beta cells from destruction not only in type 1 but also in type 2 diabetes, and that they provide evidence that oxidative stress plays a crucial role in the deterioration of beta cell function during the progression of type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Islotes Pancreáticos/metabolismo , Tiorredoxinas/metabolismo , Animales , Glucemia/análisis , Peso Corporal , Diabetes Mellitus Tipo 2/fisiopatología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Proteínas de Homeodominio/metabolismo , Humanos , Insulina/sangre , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Tiorredoxinas/genética , Transactivadores/metabolismo
7.
Endocr Regul ; 52(1): 21-26, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-29453918

RESUMEN

OBJECTIVE: Histone deacytylase inhibitors (HDACis) inhibit the deacetylation of the lysine residue of proteins, including histones, and regulate the transcription of a variety of genes. Recently, HDACis have been used clinically as anti-cancer drugs and possible anti-diabetic drugs. Even though HDACis have been proven to protect the cytokine-induced damage of pancreatic beta cells, evidence also shows that high doses of HDACis are cytotoxic. In the present study, we, therefore, investigated the eff ect of HDACis on insulin secretion in a pancreatic beta cell line. METHODS: Pancreatic beta cells MIN6 were treated with selected HDACis (trichostatin A, TSA; valproic acid, VPA; and sodium butyrate, NaB) in medium supplemented with 25 mM glucose and 13% heat-inactivated fetal bovine serum (FBS) for indicated time intervals. Protein expression of Pdx1 and Mafa in MIN6 cells was demonstrated by immunohistochemistry and immunocytochemistry, expression of Pdx1 and Mafa genes was measured by quantitative RT-PCR method. Insulin release from MIN6 cells and insulin cell content were estimated by ELISA kit. Superoxide production in MIN6 cells was measured using a Total ROS/Superoxide Detection System. RESULTS: TSA, VPA, and NaB inhibited the expression of Pdx1 and Mafa genes and their products. TSA treatment led to beta cell malfunction, characterized by enhanced insulin secretion at 3 and 9 mM glucose, but impaired insulin secretion at 15 and 25 mM glucose. Th us, TSA induced dysregulation of the insulin secretion mechanism. TSA also enhanced reactive oxygen species production in pancreatic beta cells. CONCLUSIONS: Our results showed that HDACis caused failure to suppress insulin secretion at low glucose concentrations and enhance insulin secretion at high glucose concentrations. In other words, when these HDACis are used clinically, high doses of HDACis may cause hypoglycemia in the fasting state and hyperglycemia in the fed state. When using HDACis, physicians should, therefore, be aware of the capacity of these drugs to modulate the insulin secretory capacity of pancreatic beta cells.


Asunto(s)
Ácido Butírico/farmacología , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Ácido Valproico/farmacología , Animales , Bovinos , Línea Celular , Humanos
8.
Gene Expr Patterns ; 8(1): 27-35, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17919994

RESUMEN

The large number of expressed sequence tags (ESTs) now available in databases has enabled the analysis of gene expression profiles in silico. We searched public databases for uncharacterized transcripts specifically expressed in germ cells, in an attempt to identify genes involved in gametogenesis. We found a transcript that is expressed in unfertilized eggs, ovaries, and testes of the mouse. It has an open reading frame (ORF) encoding a 167-amino acid protein belonging to the UPF0224 (unknown protein family 0224) family. We called the novel gene Cue110. We examined the Pfam database for other members of the UPF0224 family, and found a conserved N-terminal portion among members of various species. To study the cellular localization of the Cue110 transcript and protein, we performed in situ hybridization and immunohistochemical analysis of the adult mouse ovary and testis. In the testis, specific hybridization signals were observed weakly in preleptotene spermatocytes but maximally in late round spermatids. Immunostaining showed that Cue110 protein was present predominantly in the cytoplasm of pachytene spermatocytes and round spermatids. In the ovary, weak hybridization signals were observed in primary oocytes in the primordial, primary, and secondary follicles, but Cue110 protein was not detected in oocytes by immunostaining. We next examined the developmental expression pattern of the Cue110 gene using RT-PCR and western blotting, and found its increasing expression coincided with the appearance of spermatocytes. Thus, the Cue110 gene is expressed predominantly in male germ cells at stages from the pachytene spermatocytes to round spermatids.


Asunto(s)
Gametogénesis/genética , Células Germinativas/química , Proteínas/genética , Espermatocitos/química , Animales , Bases de Datos de Ácidos Nucleicos , Femenino , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones , Ovario/química , Proteínas/análisis , ARN Mensajero/análisis , Testículo/química
9.
Diabetes Res Clin Pract ; 77 Suppl 1: S138-42, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17451835

RESUMEN

Embryonic stem (ES) cells can differentiate into many cell types. Recent reports have shown that ES cells can differentiate into insulin-producing cells. We have established an ES cell line in which exogenous Pdx-1 expression was precisely regulated by the Tet-off system integrated into the ROSA26 locus and succeeded to produce insulin-producing cells. The Pdx-1 expressing final differentiated insulin-positive cells can be maintained for more than 2 months. However, in spite of their induced expression of Pdx-1, the repeated passages of cells lost their capacity to express insulin and NeuroD1 gene. Forced expression of NeuroD1 gene by adenoviral vector in these cells restored the expression of insulin. These results suggested that maintenance of the property of insulin-producing cells derived from ES cells could be achieved by synergistic expression of Pdx-1 and NeuroD1.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Insulina/genética , Transactivadores/genética , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Humanos , Ratones , ARN/genética , ARN/aislamiento & purificación , Transfección
11.
PLoS One ; 11(3): e0151927, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26986842

RESUMEN

Elucidating the regulation of glucose-stimulated insulin secretion (GSIS) in pancreatic ß cells is important for understanding and treating diabetes. The pancreatic ß cell line, MIN6, retains GSIS but gradually loses it in long-term culture. The MIN6 subclone, MIN6c4, exhibits well-regulated GSIS even after prolonged culture. We previously used DNA microarray analysis to compare gene expression in the parental MIN6 cells and MIN6c4 cells and identified several differentially regulated genes that may be involved in maintaining GSIS. Here we investigated the potential roles of six of these genes in GSIS: Tmem59l (Transmembrane protein 59 like), Scgn (Secretagogin), Gucy2c (Guanylate cyclase 2c), Slc29a4 (Solute carrier family 29, member 4), Cdhr1 (Cadherin-related family member 1), and Celsr2 (Cadherin EGF LAG seven-pass G-type receptor 2). These genes were knocked down in MIN6c4 cells using lentivirus vectors expressing gene-specific short hairpin RNAs (shRNAs), and the effects of the knockdown on insulin expression and secretion were analyzed. Suppression of Tmem59l, Scgn, and Gucy2c expression resulted in significantly decreased glucose- and/or KCl-stimulated insulin secretion from MIN6c4 cells, while the suppression of Slc29a4 expression resulted in increased insulin secretion. Tmem59l overexpression rescued the phenotype of the Tmem59l knockdown MIN6c4 cells, and immunostaining analysis indicated that the TMEM59L protein colocalized with insulin and GM130, a Golgi complex marker, in MIN6 cells. Collectively, our findings suggested that the proteins encoded by Tmem59l, Scgn, Gucy2c, and Slc29a4 play important roles in regulating GSIS. Detailed studies of these proteins and their functions are expected to provide new insights into the molecular mechanisms involved in insulin secretion.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animales , Western Blotting , Cadherinas/fisiología , Línea Celular , Femenino , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Genes Reguladores/fisiología , Glucosa/fisiología , Insulina/fisiología , Secreción de Insulina , Células Secretoras de Insulina/fisiología , Proteínas de la Membrana/fisiología , Proteínas de Transporte de Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Receptores de Enterotoxina , Receptores Acoplados a la Guanilato-Ciclasa/fisiología , Receptores de Péptidos/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Secretagoginas/fisiología
12.
Diabetes ; 53(4): 1030-7, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15047618

RESUMEN

Embryonic stem (ES) cells can differentiate into many cell types. Recent reports have shown that ES cells can differentiate into insulin-producing cells. However, the differentiation is not efficient enough to produce insulin-secreting cells for future therapeutic use. Pdx-1, a homeodomain-containing transcription factor, is a crucial regulator for pancreatic development. We established an ES cell line in which exogenous pdx-1 expression was precisely regulated by the Tet-off system integrated into the ROSA26 locus. Using this cell line, we examined the effect of pdx-1 expression during in vitro differentiation via embryoid body formation. The results showed that pdx-1 expression clearly enhanced the expression of the insulin 2, somatostatin, Kir6.2, glucokinase, neurogenin3, p48, Pax6, PC2, and HNF6 genes in the resulting differentiated cells. Immunohistochemical examination also revealed that insulin was highly produced in most of the differentiated ES cells. Thus, exogenous expression of pdx-1 should provide a promising approach for efficiently producing insulin-secreting cells from human ES cells for future therapeutic use in diabetic patients.


Asunto(s)
Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de Homeodominio , Insulina/metabolismo , Islotes Pancreáticos/embriología , Células Madre/fisiología , Transactivadores/genética , Actinas/genética , Animales , Secreción de Insulina , Islotes Pancreáticos/citología , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/citología
13.
Diabetes ; 52(5): 1163-8, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12716747

RESUMEN

Embryonic stem (ES) cells can differentiate into many cell types and are expected to be useful for tissue engineering. Recent reports have shown that ES cells can differentiate into insulin-producing cells in response to the transient expression of the pdx-1 gene, after the removal of feeder cells. To investigate the lineage of insulin-producing cells and their in vitro differentiation, we introduced the betageo gene, encoding a beta-galactosidase-neomycin phosphotransferase fusion protein under the control of the mouse insulin 2 promoter, into ES cells that had been adapted to feeder-free culture, and analyzed insulin gene expression during their in vitro differentiation. We also examined the expression of transcription factors that are related to the differentiation of the pancreas. X-gal staining analysis revealed beta-galactosidase-positive cells on the surface and in the center of the embryoid body that proliferated during differentiation. Glucose-responsive insulin-producing cells, derived from our feeder-free ES cells, expressed insulin 2, pdx-1, Pax4, and Isl1 and also the glucagon, somatostatin, and PP genes. Moreover, the genes encoding p48, amylase, and carboxypeptidase A were also expressed. These results suggest that ES cells can differentiate not only into endocrine cells but also into exocrine cells of the pancreas, without the initiation of pdx-1 expression.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas de Homeodominio , Insulina/genética , Células Madre/citología , Células Madre/fisiología , Animales , Secuencia de Bases , Cartilla de ADN , Regulación del Desarrollo de la Expresión Génica , Glucosa/farmacología , Insulina/biosíntesis , Kanamicina Quinasa/genética , Ratones , Regiones Promotoras Genéticas , Proteínas Recombinantes de Fusión/biosíntesis , Células Madre/efectos de los fármacos , Transactivadores/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , beta-Glucosidasa/genética
14.
Transplantation ; 74(11): 1603-8, 2002 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-12490795

RESUMEN

BACKGROUND: Viral interleukin (vIL)-10, encoded in the Epstein-Barr virus genome, shares many of the anti-inflammatory properties of cellular IL-10 but is supposed to lack IL-10's immunostimulatory properties. Thus, vIL-10 is expected to offer superior immunosuppression. METHODS: We established transgenic mice (vIL-10 Tg) that express vIL-10 systemically and transplanted their hearts as vascularized allografts into unmodified major histocompatibility complex (MHC) full-mismatch or MHC class II-disparate mice. RESULTS: The vIL-10 Tg mice revealed high-level expression of vIL-10 in major organs including the heart. However, the heart grafts from the vIL-10 Tg mice failed to exhibit prolonged survival in combination with either the MHC full-mismatch or the class II-disparate mice. In the MHC class II-disparate mice, the vIL-10 Tg heart grafts showed severe CD8 T-cell infiltration and increased interferon (IFN)-gamma mRNA expression compared with non-Tg grafts. CONCLUSION: High level expression of vIL-10 in grafts can exacerbate immunological rejection in an allogenic transplantation model.


Asunto(s)
Supervivencia de Injerto/efectos de los fármacos , Trasplante de Corazón , Herpesvirus Humano 4/metabolismo , Interleucina-10/metabolismo , Interleucina-10/uso terapéutico , Miocardio/metabolismo , Animales , Femenino , Supervivencia de Injerto/fisiología , Interleucina-10/genética , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos/genética , Factores de Tiempo , Trasplante Homólogo , Insuficiencia del Tratamiento
15.
Autoimmunity ; 35(1): 63-6, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11908709

RESUMEN

Linkage analysis and congenic mapping have localized 18 loci (Idd1-18) that contribute to the development of autoimmune type 1 diabetes in the nonobese diabetic (NOD) mouse. By using a congenic NOD strain which possesses recombinant MHC from a closely related CTS strain, a susceptible region (Idd16) was mapped to the segment adjacent to, but distinct from class II A and E genes (Idd1). The tumor necrosis factor alpha gene (Tnf), which is located within the Idd16 region, has been suspected to be a candidate gene for type 1 diabetes in the NOD mouse. Although the protein-coding region in Tnf has been sequenced in the NOD mouse and its related strains, the complete upstream region (approximately 1400 bp, including the 5'-untranslated region) has not yet been studied. To study the possible contribution of the transcriptional regulation of Tnf to susceptibility to type 1 diabetes, we determined the complete nucleotide sequences of the NOD strain and its related strain, CTS, in comparison with the non-diabetic control strain, C57BL/6. The nucleotide sequence of the 5'-upstream region in the NOD mouse was identical to that in the C57BL/6 mouse, but different from that in the CTS mouse. In particular, a C to A substitution at position 3408 in the CTS mouse creates a new GATA family binding site, which may be responsible for the lower incidence of type 1 diabetes in the NOD. CTS-H-2 congenic strain despite the presence of the same class II MHC.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Factor de Necrosis Tumoral alfa/genética , Regiones no Traducidas 5' , Animales , Secuencia de Bases , Mapeo Cromosómico , ADN/genética , Genes MHC Clase II , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Datos de Secuencia Molecular , Homología de Secuencia de Ácido Nucleico
16.
J Biochem ; 133(4): 423-7, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12761288

RESUMEN

We previously demonstrated that cytokine expression following intramuscular gene transfer of a naked plasmid is increased 2 logs by in vivo electroporation, but the relatively low expression levels of the encoded protein is still a limitation for successful gene therapy and gene function studies. We recently reported that the serum viral IL-10 levels achieved by electroporation-mediated intramuscular delivery of pCAGGS-vIL10, a viral IL-10-expressing plasmid, can be further enhanced by modifying the plasmid into an immunoglobulin fusion protein expression plasmid, pCAGGS-vIL10/Fc. Here we examined the applicability of this approach to the expression of an endogenous cytokine, IL-10, in two different inbred mouse strains. We obtained sustained high serum levels of IL-10 in C3H/HeJ mice (C3H), but the level and duration of the gene expression was mouse-strain dependent. Although the serum IL-10 level was also increased by using the IL-10/Fc gene plasmid in C57BL/6 mice (B6), IL-10/Fc and a luciferase reporter showed significantly lower levels in B6 than in C3H mice, and the persistence of pCAGGS-IL10/Fc expression ranged from several days in B6 mice to more than one month in C3H mice. These results suggest that the electroporation-mediated intramuscular delivery of the immunoglobulin fusion protein expression plasmid is simple and very efficient, but mouse strain differences in transgene expression should be taken into consideration in its use.


Asunto(s)
Citocinas/biosíntesis , Fragmentos Fc de Inmunoglobulinas/biosíntesis , Interleucina-10/biosíntesis , Animales , Citocinas/genética , Electroporación , Expresión Génica , Técnicas de Transferencia de Gen , Fragmentos Fc de Inmunoglobulinas/genética , Interleucina-10/sangre , Interleucina-10/genética , Luciferasas/análisis , Luciferasas/genética , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Plásmidos , Proteínas Recombinantes de Fusión/biosíntesis , Especificidad de la Especie
17.
J Biosci Bioeng ; 94(1): 81-3, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-16233274

RESUMEN

In search of an efficient nonviral vector, polyethyleneimine (PEI)-based vectors were examined. In general, the transfection efficiency of nonviral vectors is suppressed by serum. Here we show that PEI based vectors, particularly, the chitosan hexamer-PEI vector, could perform efficient gene transfection into intestinal epithelial cells (IEC-6) in the presence of serum. The conjugation order of the two polymers with a plasmid (first, chitosan hexamer; second, PEI) was found to be an important factor in enhancing transfection efficiency.

18.
PLoS One ; 8(4): e61211, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23560115

RESUMEN

Elucidating the regulation of glucose-stimulated insulin secretion (GSIS) in pancreatic islet ß cells is important for understanding and treating diabetes. MIN6 cells, a transformed ß-cell line derived from a mouse insulinoma, retain GSIS and are a popular in vitro model for insulin secretion. However, in long-term culture, MIN6 cells' GSIS capacity is lost. We previously isolated a subclone, MIN6 clone 4, from the parental MIN6 cells, that shows well-regulated insulin secretion in response to glucose, glybenclamide, and KCl, even after prolonged culture. To investigate the molecular mechanisms responsible for preserving GSIS in this subclone, we compared four groups of MIN6 cells: Pr-LP (parental MIN6, low passage number), Pr-HP (parental MIN6, high passage number), C4-LP (MIN6 clone 4, low passage number), and C4-HP (MIN6 clone 4, high passage number). Based on their capacity for GSIS, we designated the Pr-LP, C4-LP, and C4-HP cells as "responder cells." In a DNA microarray analysis, we identified a group of genes with high expression in responder cells ("responder genes"), but extremely low expression in the Pr-HP cells. Another group of genes ("non-responder genes") was expressed at high levels in the Pr-HP cells, but at extremely low levels in the responder cells. Some of the responder genes were involved in secretory machinery or glucose metabolism, including Chrebp, Scgn, and Syt7. Among the non-responder genes were Car2, Maf, and Gcg, which are not normally expressed in islet ß cells. Interestingly, we found a disproportionate number of known imprinted genes among the responder genes. Our findings suggest that the global expression profiling of GSIS-competent and GSIS-incompetent MIN6 cells will help delineate the gene regulatory networks for insulin secretion.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Anhidrasa Carbónica II/genética , Anhidrasa Carbónica II/metabolismo , Línea Celular , Proliferación Celular , Células Clonales , Redes Reguladoras de Genes , Glucosa/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/citología , Ratones , Análisis por Micromatrices , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Especificidad de Órganos , Proglucagón/genética , Proglucagón/metabolismo , Proteínas Proto-Oncogénicas c-maf/genética , Proteínas Proto-Oncogénicas c-maf/metabolismo , Secretagoginas , Sinaptotagminas/genética , Sinaptotagminas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
J Diabetes Investig ; 3(1): 41-51, 2012 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-24843545

RESUMEN

UNLABELLED: Aims/Introduction: We recently established a strategy for isolating multipotential duct-like cells, called pdx-1-positive pancreatic cell-derived (PPPD) cells, from the pancreas. To analyze the molecular mechanisms of pancreatic cell differentiation, we introduced a polyoma-based episomal vector system into PPPD cells. MATERIALS AND METHODS: PPPD cells were stably transfected with a polyoma large T (PLT)-expressing plasmid vector, which included the polyoma origin of replication, to generate PLT-PPPD cells. Various cDNA for pancreas-related transcription factors were subcloned into the expression plasmid pPyCAG, which included the polyoma origin of replication. PLT-PPPD cells were stably transfected with the resulting plasmid vectors and then subjected to gene and protein expression analyses. RESULTS: The coexpression of Mafa, Neurod1 and Ipf1 induced Ins1 and Ins2 expression in PLT-PPPD cells. The forced expression of Pax6 alone induced the expression of glucagon. The coexpression of Neurod1 and Isl1 induced Ins2 and Sst expression. In contrast, the expression of Ptf1a and Foxa2 induced the expression of exocrine markers Cpa1 and Amy2. Transfections with multiple transcription factors showed that Isl1 is required for the differentiation of both insulin-positive cells and somatostatin-positive cells. In addition, Foxa2 induced the differentiation of glucagon-positive cells and inhibited the differentiation of insulin-positive and somatostatin-positive cells. PLT-PPPD cells allow episomal vector-based gene expression and should be useful for studying the transcription factor cascades involved in the differentiation of pancreatic cell types in vitro. CONCLUSIONS: Our coexpression study showed novel critical roles for Isl1 and Foxa2 in the differentiation of PPPD cells into endocrine cells. (J Diabetes Invest, doi: 10.1111/j.2040-1124.2011.00136.x, 2012).

20.
PLoS One ; 7(10): e47536, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23077629

RESUMEN

Tubular complexes (TCs) are aggregates of duct-like monolayered cells in the developing and regenerating pancreas. Recent studies showed that TCs have regenerative potential, including islet neogenesis. We previously delivered adenovirus vector (AdV) into exocrine cells of the pancreas by intra-common bile ductal (ICBD) injection, and found that AdV expressing Pdx1, a pancreas-specific transcription factor, causes TC formation and islet neogenesis. We also established RTF-Pdx1-EGFP mice, which ubiquitously express Pdx1 when tetracycline is removed from the drinking water. However, exogenous Pdx1 expression in adult RTF-Pdx1-EGFP mice did not cause any pathological changes in the pancreas during three weeks of observation after tetracycline withdrawal. To examine whether the host immune response induced by AdV was involved in TC formation, we delivered AdVs expressing pancreas-related transcription factors or an irrelevant protein into the pancreas of RTF-Pdx1-EGFP mice. Histological analyses showed that both AdV injection and Pdx1 expression are required for TC formation. We also analyzed the effects of these ICBD-injected AdVs. AdV expressing Isl1, a proendocrine transcription factor, effectively induced TC formation through acinar-to-ductal metaplasia, and exogenous Pdx1 expression facilitated this process. Considering the regenerative potential of TCs, a strategy that efficiently induces TC formation may lead to novel therapies for diabetes.


Asunto(s)
Proteínas de Homeodominio , Proteínas con Homeodominio LIM , Metaplasia , Páncreas , Transactivadores , Factores de Transcripción , Adenoviridae , Animales , Diabetes Mellitus/genética , Diabetes Mellitus/terapia , Regulación Neoplásica de la Expresión Génica , Vectores Genéticos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Metaplasia/genética , Metaplasia/metabolismo , Ratones , Páncreas/metabolismo , Páncreas/patología , Regeneración/genética , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA