Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cell Mol Med ; 26(15): 4183-4194, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35801494

RESUMO

Excessive dietary intake of fat results in its storage in white adipose tissue (WAT). Energy expenditure through lipid oxidation occurs in brown adipose tissue (BAT). Certain WAT depots can undergo a change termed beiging where markers that BAT express are induced. Little is known about signalling pathways inducing beiging. Here, inhibition of a signalling pathway regulating alternative pre-mRNA splicing is involved in adipocyte beiging. Clk1/2/4 kinases regulate splicing by phosphorylating factors that process pre-mRNA. Clk1 inhibition by TG003 results in beige-like adipocytes highly expressing PGC1α and UCP1. SiRNA for Clk1, 2 and 4, demonstrated that Clk1 depletion increased UCP1 and PGC1α expression, whereas Clk2/4 siRNA did not. TG003-treated adipocytes contained fewer lipid droplets, are smaller, and contain more mitochondria, resulting in proton leak increases. Additionally, inhibition of PKCßII activity, a splice variant regulated by Clk1, increased beiging. PGC1α is a substrate for both Clk1 and PKCßII kinases, and we surmised that inhibition of PGC1α phosphorylation resulted in beiging of adipocytes. We show that TG003 binds Clk1 more than Clk2/4 through direct binding, and PGC1α binds to Clk1 at a site close to TG003. Furthermore, we show that TG003 is highly specific for Clk1 across hundreds of kinases in our activity screen. Hence, Clk1 inhibition becomes a target for induction of beige adipocytes.


Assuntos
Adipócitos , Precursores de RNA , Células 3T3-L1 , Adipócitos/metabolismo , Animais , Biomarcadores/metabolismo , Camundongos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Proteína Quinase C beta/metabolismo , Precursores de RNA/metabolismo , RNA Interferente Pequeno/metabolismo
2.
Endocrinology ; 158(1): 183-195, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27841943

RESUMO

Brain injury may be caused by trauma or may occur in stroke and neurodegenerative diseases. Because the central nervous system is unable to regenerate efficiently, there is utmost interest in the use of stem cells to promote neuronal survival. Of interest here are human adipose-derived stem cells (hASCs), which secrete factors that enhance regeneration and survival of neurons in sites of injury. We evaluated the effect of hASC secretome on immortalized mouse hippocampal cell line (HT22) after injury. Protein kinase C δ (PKCδ) activates survival and proliferation in neurons and is implicated in memory. We previously showed that alternatively spliced PKCδII enhances neuronal survival via B-cell lymphoma 2 Bcl2 in HT22 neuronal cells. Our results demonstrate that following injury, treatment with exosomes from the hASC secretome increases expression of PKCδII in HT22 cells and increases neuronal survival and proliferation. Specifically, we demonstrate that metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), a long noncoding RNA contained in the hASC exosomes mediates PKCδII splicing, thereby increasing neuronal survival. Using antisense oligonucleotides for MALAT1 and RNA immunoprecipitation assays, we demonstrate that MALAT1 recruits splice factor serine-arginine-rich splice factor 2 (SRSF2) to promote alternative splicing of PKCδII. Finally, we evaluated the role of insulin in enhancing hASC-mediated neuronal survival and demonstrated that insulin treatment dramatically increases the association of MALAT1 and SRSF2 and substantially increases survival and proliferation after injury in HT22 cells. In conclusion, we demonstrate the mechanism of action of hASC exosomes in increasing neuronal survival. This effect of hASC exosomes to promote wound healing can be further enhanced by insulin treatment in HT22 cells.


Assuntos
Células-Tronco Adultas/metabolismo , Neurônios/fisiologia , Proteína Quinase C-delta/metabolismo , RNA Longo não Codificante/fisiologia , Processamento Alternativo , Animais , Linhagem Celular , Proliferação de Células , Sobrevivência Celular , Exossomos/metabolismo , Humanos , Insulina , Camundongos , Fatores de Processamento de Serina-Arginina/metabolismo
3.
Stem Cell Investig ; 3: 2, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27358894

RESUMO

BACKGROUND: Adipose-derived stem cells (ASC) and its exosomes are gaining utmost importance in the field of regenerative medicine. The ASCs tested for their potential in wound healing are predominantly derived from the subcutaneous depot of lean donors. However, it is important to characterize the ASC derived from different adipose depots as these depots have clinically distinct roles. METHODS: We characterized the ASC derived from subcutaneous and omental depots from a lean donor (sc-ASCn and om-ASCn) and compared it to the ASC derived from an obese donor (sc-ASCo and om-ASCo) using flow cytometry and real time qPCR. RESULTS: We show that stem cell markers Oct4, Sal4, Sox15, KLF4 and BMI1 have distinct expression patterns in each ASC. We evaluated the secretome of the ASC and characterized their secreted exosomes. We show long noncoding RNAs (lncRNAs) are secreted by ASC and their expression varied between the ASC's derived from different depots. Protein kinase C delta (PKCδ) regulates the mitogenic signals in stem cells. We evaluated the effect of silencing PKCδ in sc-ASCn, om-ASCn, sc-ASCo and om-ASCo. Using ß-galactosidase staining, we evaluated the percentage of senescent cells in sc-ASCn, om-ASCn, sc-ASCo and om-ASCo. Our results also indicated that silencing PKCδ increases the percentage of senescent cells. CONCLUSIONS: Our case-specific study demonstrates a role of PKCδ in maintaining the adipose stem cell niche and importantly demonstrates depot-specific differences in adipose stem cells and their exosome content.

4.
BBA Clin ; 4: 102-7, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26675493

RESUMO

BACKGROUND: Diabetes mellitus (DM), a metabolic disease, is characterized by impaired fasting glucose levels. Type 2 DM is adult onset diabetes. Long non-coding RNAs (lncRNAs) regulate gene expression and multiple studies have linked lncRNAs to human diseases. METHODS: Serum samples obtained from 96 participating veterans at JAH VA were deposited in the Research Biospecimen Repository. We used a two-stage strategy to identify an lncRNA whose levels correlated with T2DM. Initially we screened five serum samples from diabetic and non-diabetic individuals using lncRNA arrays. Next, GAS5 lncRNA levels were analyzed in 96 serum samples using quantitative PCR. Receiver operating characteristic (ROC) analysis was performed to determine the optimal cutoff GAS5 for diagnosis of DM. RESULTS: Our results demonstrate that decreased GAS5 levels in serum were associated with diabetes in a cohort of US military veterans. The ROC analysis revealed an optimal cutoff GAS5 value of less than or equal to 10. qPCR results indicated that individuals with absolute GAS5 < 10 ng/µl have almost twelve times higher odds of having diabetes (Exact Odds Ratio [OR] = 11.79 (95% CI: 3.97, 37.26), p < 0.001). Analysis indicated area under curve (AUC) of ROC of 0.81 with 85.1% sensitivity and 67.3% specificity in distinguishing non-diabetic from diabetic subjects. The positive predictive value is 71.4%. CONCLUSION: lncRNA GAS5 levels are correlated to prevalence of T2DM. GENERAL SIGNIFICANCE: Assessment of GAS5 in serum along with other parameters offers greater accuracy in identifying individuals at-risk for diabetes.

5.
Genes (Basel) ; 5(4): 1050-63, 2014 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-25437750

RESUMO

Long non-coding (lnc) RNAs serve a multitude of functions in cells. NEAT1 RNA is a highly abundant 4 kb lncRNA in nuclei, and coincides with paraspeckles, nuclear domains that control sequestration of paraspeckle proteins. We examined NEAT1 RNA levels and its function in 3T3-L1 cells during differentiation to adipocytes. Levels of NEAT1 transcript, measured by RT-PCR, fluctuated in a temporal manner over the course of differentiation that suggested its role in alternative splicing of PPARγ mRNA, the major transcription factor driving adipogenesis. When cells were induced to differentiate by a media cocktail of insulin, dexamethasone, and isobutylmethyxanthine (IBMX) on Day 0, NEAT1 levels dropped on Day 4, when the PPARγ2 variant was spliced and when terminal differentiation occurs The appearance of PPARγ2 coordinates with the PPARγ1 variant to drive differentiation of adipocytes. SiRNA used to deplete NEAT1 resulted in the inability of cells to phosphorylate the serine/arginine-rich splicing protein, SRp40. SiRNA treatment for SRp40 resulted in dysregulation of PPARγ1 and, primarily, PPARγ2 mRNA levels. SRp40 associated with NEAT1, as shown by RNA-IP on days 0 and 8, but decreased on day 4, and concentrations increased over that of IgG control. Overexpression of SRp40 increased PPARγ2, but not γ1. Although lncRNA MALAT1 has been investigated in SR protein function, NEAT1 has not been shown to bind SR proteins for phosphorylation such that alternative splicing results. The ability of cells to increase phosphorylated SR proteins for PPARγ2 splicing suggests that fluxes in NEAT1 levels during adipogenesis regulate alternative splicing events.

6.
J Biol Chem ; 289(46): 31662-31672, 2014 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-25261467

RESUMO

Obesity is characterized by adipocyte hyperplasia and hypertrophy. We previously showed that PKCδ expression is dysregulated in obesity (Carter, G., Apostolatos, A., Patel, R., Mathur, A., Cooper, D., Murr, M., and Patel, N. A. (2013) ISRN Obes. 2013, 161345). Using 3T3L1 preadipocytes, we studied adipogenesis in vitro and showed that expression of PKCδ splice variants, PKCδI and PKCδII, have different expression patterns during adipogenesis (Patel, R., Apostolatos, A., Carter, G., Ajmo, J., Gali, M., Cooper, D. R., You, M., Bisht, K. S., and Patel, N. A. (2013) J. Biol. Chem. 288, 26834-26846). Here, we evaluated the role of PKCδI splice variant during adipogenesis. Our results indicate that PKCδI expression level is high in preadipocytes and decreasing PKCδI accelerated terminal differentiation. Our results indicate that PKCδI is required for mitotic clonal expansion of preadipocytes. We next evaluated the splice factor regulating the expression of PKCδI during 3T3L1 adipogenesis. Our results show TRA2B increased PKCδI expression. To investigate the molecular mechanism, we cloned a heterologous splicing PKCδ minigene and showed that inclusion of PKCδ exon 9 is increased by TRA2B. Using mutagenesis and a RNA-immunoprecipitation assay, we evaluated the binding of Tra2ß on PKCδI exon 9 and show that its association is required for PKCδI splicing. These results provide a better understanding of the role of PKCδI in adipogenesis. Determination of this molecular mechanism of alternative splicing presents a novel therapeutic target in the management of obesity and its co-morbidities.


Assuntos
Ciclo Celular , Regulação da Expressão Gênica , Proteínas Nucleares/metabolismo , Proteína Quinase C-delta/metabolismo , Proteínas de Ligação a RNA/metabolismo , Células 3T3-L1 , Adipócitos/citologia , Adipogenia , Processamento Alternativo , Animais , Apoptose , Diferenciação Celular , Proliferação de Células , Camundongos , Mutação , Fatores de Processamento de Serina-Arginina
7.
Adv Wound Care (New Rochelle) ; 3(3): 219-228, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24669358

RESUMO

Objective: Adipose tissue is a robust source of adipose-derived stem cells (ADSCs) that may be able to provide secreted factors that promote the ability of wounded tissue to heal. However, adipocytes also have the potential to dedifferentiate in culture to cells with stem cell-like properties that may improve their behavior and functionality for certain applications. Approach: ADSCs are adult mesenchymal stem cells that are cultured from the stromal vascular fraction of adipose tissue. However, adipocytes are capable of dedifferentiating into cells with stem cell properties. In this case study, we compare ADSC and dedifferentiated fat (DFAT) cells from the same patient and fat depot for mesenchymal cell markers, embryonic stem cell markers, ability to differentiate to adipocytes and osteoblasts, senescence and telomerase levels, and ability of conditioned media (CM) to stimulate migration of human dermal fibroblasts (HDFs). Innovation and Conclusions: ADSCs and DFAT cells displayed identical levels of CD90, CD44, CD105, and were CD34- and CD45-negative. They also expressed similar levels of Oct4, BMI1, KLF4, and SALL4. DFAT cells, however, showed higher efficiency in adipogenic and osteogenic capacity. Telomerase levels of DFAT cells were double those of ADSCs, and senescence declined in DFAT cells. CM from both cell types altered the migration of fibroblasts. Despite reports of ADSCs from a number of human depots, there have been no comparisons of the ability of dedifferentiated DFAT cells from the same donor and depot to differentiate or modulate migration of HDFs. Since ADSCs were from an obese diabetic donor, reprogramming of DFAT cells may help improve a patient's cells for regenerative medicine applications.

8.
Stem Cell Investig ; 1: 3, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-27358850

RESUMO

BACKGROUND: Adipose-derived stem cells (ADSC) were isolated and characterized from lean and obese subjects. We previously reported that distinct differences were observed in differentiating lean and obese preadipocytes. Protein kinase C delta (PKCδ) is alternatively spliced and has important roles in apoptosis. PKCδI promotes apoptosis and PKCδVIII promotes survival. Our previous data indicated an increase in the survival kinase, PKCδVIII in ADSC derived from an obese donor. We also determined that obese adipocytes were resistant to apoptosis. Here, we determine the relationship between a survival kinase PKCδVIII and hTERT expression in adipose derived stem cells from a lean and obese subject. METHODS: We evaluated the telomerase activity and human telomerase reverse transcriptase (hTERT) expression in lean and obese ADSC. The lean and obese ADSC were purchased as cryopreserved cells from ZenBio™ (Research Triangle Park, NC, USA). Analyses were performed using PRISM™ software and analyzed using two-tailed Student's t-test. RESULTS: We observed an increase in telomerase in differentiating obese ADSC using western blot analysis. We determined the levels of hTERT splice variants. hTERT α+/ß+ splice variant was increased after transfected of PKCδVIII. We next determined whether PKCδVIII over-expression affected the levels of telomerase. The results indicate an increase in telomerase with PKCδVIII over-expression. CONCLUSIONS: Over-expression of PKCδVIII in lean ADSC substantially increased expression of hTERT and telomerase. The decreased senescence seen in obese ADSC may in part be attributed to PKCδVIII. Obese ADSC undergo lower senescence and may have increased growth potential. These results propose a larger epigenetic modification in obese ADSC compared to lean ADSC.

9.
J Biol Chem ; 288(37): 26834-46, 2013 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-23902767

RESUMO

Increased food intake and lack of physical activity results in excess energy stored in adipocytes, and this imbalance contributes to obesity. New adipocytes are required for storage of energy in the white adipose tissue. This process of adipogenesis is widely studied in differentiating 3T3L1 preadipocytes in vitro. We have identified a key signaling kinase, protein kinase C delta (PKCδ), whose alternative splice variant expression is modulated during adipogenesis. We demonstrate that PKCδII splice variant promotes survival in differentiating 3T3L1 cells through the Bcl2 pathway. Here we demonstrate that resveratrol, a naturally occurring polyphenol, increases apoptosis and inhibits adipogenesis along with disruption of PKCδ alternative splicing during 3T3L1 differentiation. Importantly, we have identified a PKCδII splice variant inhibitor. This inhibitor may be a valuable tool with therapeutic implications in obesity.


Assuntos
Adipogenia , Processamento Alternativo , Apoptose , Proteína Quinase C-delta/antagonistas & inibidores , Estilbenos/química , Células 3T3-L1 , Animais , Diferenciação Celular , Regulação Enzimológica da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Polifenóis/química , Proteína Quinase C-delta/genética , Inibidores de Proteínas Quinases/química , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Resveratrol , Transfecção
10.
PLoS One ; 8(1): e53268, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23308182

RESUMO

The development of adipocytes from their progenitor cells requires the action of growth factors signaling to transcription factors to induce the expression of adipogenic proteins leading to the accumulation of lipid droplets, induction of glucose transport, and secretion of adipokines signaling metabolic events throughout the body. Murine 3T3-L1 pre-adipocytes sequentially express all the proteins necessary to become mature adipocytes throughout an 8-10 day process initiated by a cocktail of hormones. We examined the role of Clk/STY or Clk1, a cdc2-like kinase, in adipogenesis since it is known to be regulated by Akt, a pivotal kinase in development. Inhibition of Clk1 by a specific inhibitor, TG003, blocked alternative splicing of PKCßII and expression of PPARγ1 and PPARγ2. SiRNA depletion of Clk1 resulted in early expression of PKCßII and sustained PKCßI expression. Since Clk1 is a preferred Akt substrate, required for phosphorylation of splicing factors, mutation of Clk1 Akt phosphorylation sites was undertaken. Akt sites on Clk1 are in the serine/arginine-rich domain and not the kinase domain. Mutation of single and multiple sites resulted in dysregulation of PKCßII, PKCßI, and PPARγ1&2 expression. Additionally, adipogenesis was blocked as assessed by Oil Red O staining, adiponectin, and Glut1 and 4 expression. Immunofluorescence microscopy revealed that Clk1 triple mutant cDNA, transfected into pre-adipocytes, resulted in excluding SRp40 (SFSR6) from co-localizing to the nucleus with PFS, a perispeckle specific protein. This study demonstrates the role of Akt and Clk1 kinases in the early differentiation of 3T3-L1 cells to adipocytes.


Assuntos
Células 3T3-L1/citologia , Adipogenia , Processamento Alternativo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células 3T3-L1/metabolismo , Adipócitos/citologia , Adipócitos/metabolismo , Animais , Sítios de Ligação , Regulação Neoplásica da Expressão Gênica , Camundongos , Mutação , Proteínas Nucleares/metabolismo , Fosforilação , Proteína Quinase C/metabolismo , Proteína Quinase C beta , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/química , Proteínas Tirosina Quinases/genética , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/metabolismo , Fatores de Processamento de Serina-Arginina , Tiazóis/farmacologia
11.
ISRN Obes ; 2013: 161345, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24533217

RESUMO

Obesity and its comorbidities affect millions of people. Here, we demonstrate that human preadipocytes are susceptible to programmed cell death (apoptosis) while mature adipocytes are resistant to apoptosis. The molecular mechanisms underlying the phenotype of apoptosis-resistant adipocytes are lesser known. To study the role of apoptosis and define molecular differences in the developmental process of adipogenesis, human preadipocytes were differentiated in vitro to mature adipocytes. Many genes in the apoptosis pathway are alternatively spliced. Our data demonstrates that during differentiation PKC δ , Bclx, and Caspase9 switch to their prosurvival splice variants along with an increase in Bcl2 expression when the cells terminally differentiate into mature adipocytes. Next we determined the expression pattern of these genes in obesity. Our data indicated high expression of PKC δ VIII in adipose tissue of obese patient in different depots. We demonstrate a shift in the in vitro expression of these splice variants in differentiating preadipocytes derived from obese patients along with a decrease in adipogenesis markers. Hence, the programmed splicing of antiapoptotic proteins is a pivotal switch in differentiation that commits adipocytes to a prosurvival pathway. The expression pattern of these genes is dysregulated in obesity and may contribute to adipose tissue dysfunction.

12.
Histochem Cell Biol ; 132(1): 95-103, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19352691

RESUMO

Atrial natriuretic peptide (ANP), brain type natriuretic peptide (BNP) and C-type natriuretic peptide (CNP) comprise a family of natriuretic peptides that mediate their biological effects through three natriuretic peptide receptor subtypes, NPR-A (ANP, BNP), NPR-B (CNP) and NPR-C (ANP, BNP, CNP). Several reports have provided evidence for the expression of ANP and specific binding sites for ANP in the pancreas. The purpose of this study was to identify the ANP receptor subtype and to localize its expression to a specific cell type in the human pancreas. NPR-C immunoreactivity, but neither ANP nor NPR-A, was detected in human islets by immunofluorescent staining. No immunostaining was observed in the exocrine pancreas or ductal structures. Double-staining revealed that NPR-C was expressed mainly in the glucagon-containing alpha cells. NPR-C mRNA and protein were detected in isolated human islets by RT-PCR and Western blot analysis, respectively. NPR-C expression was also detected by immunofluorescent staining in glucagonoma but not in insulinoma. ANP, as well as BNP and CNP, stimulated glucagon secretion from perifused human islets (1,111 +/- 55% vs. basal [7.3 fmol/min]; P < 0.001). This response was mimicked by cANP(4-23), a selective agonist of NPR-C. In conclusion, the NPR-C receptor is expressed in normal and neoplastic human alpha cells. These findings suggest a role for natriuretic peptides in the regulation of glucagon secretion from human alpha cells.


Assuntos
Fator Natriurético Atrial/metabolismo , Células Secretoras de Glucagon/metabolismo , Receptores do Fator Natriurético Atrial/biossíntese , Adulto , Glucagonoma/metabolismo , Glucagonoma/patologia , Humanos , Insulinoma/metabolismo , Insulinoma/patologia , Masculino , Pessoa de Meia-Idade , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , RNA Mensageiro/biossíntese , Adulto Jovem
13.
Mol Cell Biochem ; 293(1-2): 103-18, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16786190

RESUMO

Evidence suggests that functional atrial natriuretic peptide (ANP) receptors occur in surface gastric mucosal epithelial cells. To evaluate functional aspects of ANP in a model of these cells we examined the expression of natriuretic peptide receptors (NPR) subtypes A and C in the non-transformed rat gastric mucosal epithelial cell line RGM1. Transcripts for NPR-A and NPR-C were detected in RGM1 cells by RT-PCR. However, only NPR-C protein was detected by Western blot and immunohistochemical analyses. Specific saturable binding of (125)I-ANP to RGM1 cells revealed a single class of high affinity binding sites (K (d) = 208 +/- 71pM, B (max) = 110,000 +/- 14,000 sites/cell, Hill coefficient = 0.97 +/- 0.05). ANP (IC(50) 130 +/- 47pM), BNP (IC(50) 716 +/- 26 pM), CNP (IC(50) 356 +/- 85pM) and C-ANP (IC(50) 134 +/- 13pM), a specific ligand for NPR-C, effectively displaced (125)I-ANP binding. Cross-linking of (125)I-ANP to cells labeled predominantly a protein of 66,000 Da. These data suggest that (125)I-ANP binding was primarily to NPR-C. ANP and C-ANP inhibited forskolin- and prostaglandin E(2) (PGE(2))-stimulated cAMP in a PTx-sensitive fashion. PGE(2), transforming growth factor-+/-1 (TGF-+/-1), forskolin, 8-bromo-cyclic AMP, and phorbol-12-myristate-13-acetate (PMA) caused a dose-dependent decrease in specific (125)I-ANP binding, whereas epidermal growth factor (EGF), 8-bromo-cyclic GMP and 4+/--phorbol didecanoate had no effect. PGE(2), forskolin, TGF-+/-1 and PMA significantly decreased (125)I-ANP B (max) values, NPR-C protein and steady-state NPR-C transcript levels. H89, a protein kinase A inhibitor, blocked the reduction of NPR-C mRNA produced by both forskolin and PGE(2.) GF109203X, a protein kinase C inhibitor, abolished the PMA-induced decrease in NPR-C transcripts but only partially blocked that produced by TGF-+/-1. RGM1 cells exhibited a dose-dependent decrease in both DNA synthesis and cell proliferation when cultured in the presence of ANP or C-ANP. These findings indicate that RGM1 cells express functional NPR-C receptors that can influence RGM1 cell proliferation and are down-regulated by PGE(2) and TGF-+/-1.


Assuntos
Células Epiteliais/metabolismo , Mucosa Gástrica/metabolismo , Receptores do Fator Natriurético Atrial/metabolismo , Adenilil Ciclases/metabolismo , Animais , Fator Natriurético Atrial/metabolismo , Proliferação de Células , Colforsina/farmacologia , Dinoprostona/farmacologia , Relação Dose-Resposta a Droga , Regulação para Baixo , Imunofluorescência , Mucosa Gástrica/citologia , Masculino , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptores do Fator Natriurético Atrial/genética , Acetato de Tetradecanoilforbol/farmacologia , Fator de Crescimento Transformador beta1/farmacologia
14.
Mol Cell Biochem ; 252(1-2): 263-71, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14577601

RESUMO

The present investigation was designed to determine if the mechanism for the increased atrial natriuretic peptides within the circulation of diabetic animals involves atrial natriuretic hormone prohormone (proANH) gene expression upregulation. The tissue specificity of this potential upregulation of the proANH gene was investigated in a spontaneous model of type 2 diabetes, i.e. the Goto-Kakizaki (GK) rat with comparison to age-matched non-diabetic Wistar rats from which the GK colony was originally derived. Reverse transcription-polymerase chain reaction revealed that proANH gene expression was increased 3.1-fold in the left heart ventricle, 5-fold in lung, 2-fold in kidney, 3-fold within mucosa and 1.8-fold within muscle of gastric antrum (p < 0.05 for each) of GK rats compared to Wistar rats. There was no significant increase in proANH gene expression in atria and right ventricle of the heart of GK rats compared to Wistars. These results indicate that steady-state ANH prohormone mRNA levels increase within the left ventricle and extracardiac tissues in type 2 diabetic animals. This enhanced gene expression is a functional increase with its expressed proteins (4 peptide hormones; ANPs) increasing 2-6 fold within the circulation of GKs. The greater increase in proANH messenger RNA in the extracardiac tissues compared to the amount of increase within the heart and the greater tissue mass of these combined extra cardiac tissues suggests the majority of the increase in ANPs within the circulation of diabetics is secondary to increased synthesis in extracardiac tissues. This also suggests that there is a systemic regulatory mechanism of proANH gene expression not only within the heart but also within the lung, gastrointestinal tract and kidney. Diabetes is the first disease in which there is more upregulation of ANH prohormone in extracardiac tissues compared to upregulation within the heart itself.


Assuntos
Fator Natriurético Atrial/genética , Diabetes Mellitus Experimental/genética , Expressão Gênica , Miocárdio/metabolismo , Precursores de Proteínas/genética , Animais , Sequência de Bases , Southern Blotting , Primers do DNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
J Surg Res ; 111(1): 70-7, 2003 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-12842450

RESUMO

BACKGROUND: Our in vivo model of tolerance, sublethal hemorrhage (SLH), alters cytokine production, nuclear factor-kappaB mobilization, mitogen-activated protein (MAP) kinase activity, and makes rats tolerant to shock. Heat shock protein (HSP) protects animals from stress. This study investigated if SLH induces in vivo HSP72 expression and whether in vitro HSP72 induction by sodium arsenite (NaArs) alters intracellular signal transduction and cytokine production similar to SLH. METHODS: Sprague-Dawley rats were made tolerant by SLH (MAP = 30 mmHg for 15 min, shed blood returned) and given lipopolysaccharide (LPS; 40 mg/kg i.p.) 24 h later. Lung was harvested 1, 12, and 24 h after SLH (n = 4) and 1 h after LPS (n = 8). Other rats underwent bronchoalveolar lavage 24 h after SLH, and macrophages (mphi) were treated with LPS (10 microg/ml). The NR8383 alveolar mphi cell line was treated with 50 microM NaArsx 12 h and LPS. Reverse transcription polymerase chain reaction, Western blots, and enzyme-linked immunosorbent assay were performed for gene, MAPK, and protein expression (tumor necrosis factor [TNF], HSP, p38). RESULTS: SLH induced significantly more lung HSP72 mRNA and protein. SLH mphi had more HSP72 protein before and after LPS compared with shams. NaArs induced HSP72 mRNA and protein in NR8383 mphi, and these cells made less TNF compared with controls. NaArs significantly increased p38 activation vs control. SB203580 inhibition of p38 activity did not affect HSP72 expression, or reverse NaArs inhibition of LPS induced TNF production. CONCLUSION: SLH induces HSP72 in vivo. In vitro HSP72 induction is associated with increased p38 phosphorylation. Like SLH, mphi with induced HSP72 expression, have an attenuated TNF response. HSP72 acts independently from p38 in inducing tolerance.


Assuntos
Proteínas de Choque Térmico/fisiologia , Hemorragia/fisiopatologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Choque Séptico/prevenção & controle , Animais , Arsenitos/farmacologia , Northern Blotting , Western Blotting , Líquido da Lavagem Broncoalveolar , Modelos Animais de Doenças , Ativação Enzimática , Expressão Gênica/efeitos dos fármacos , Proteínas de Choque Térmico HSP72 , Proteínas de Choque Térmico/análise , Proteínas de Choque Térmico/genética , Lipopolissacarídeos/farmacologia , Pulmão/química , Macrófagos Alveolares/fisiologia , Masculino , Fosforilação , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Compostos de Sódio/farmacologia , Fator de Necrose Tumoral alfa/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno
16.
J Gastrointest Surg ; 7(1): 20-5, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12559181

RESUMO

We have demonstrated that Kupffer cell-derived tumor necrosis factor (TNF) mediates pancreatitis-associated liver injury. The aim of this study was to determine the role of p38 mitogen-activated protein kinase (MAPK), extracellular stress-related kinase 1/2 (ERK1/2), stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK), and nuclear factor-kappaB (NF-kappaB) in TNF gene expression within Kupffer cells. TNF and TNF-mRNA were measured in rat livers perfused with elastase. TNF, TNF-mRNA, NF-kappaB activation, and phosphorylated p38-MAPK, SAPK/JNK, and ERK1/2 were determined in Kupffer cells treated with elastase. Elastase increased TNF and upregulated TNF-mRNA in livers (P<0.03) and Kupffer cells (P<0.001). Phosphorylated p38-MAPK, SAPK/JNK, and ERK1/2 and activated NF-kappaB were detected in Kupffer cells at 7 minutes; at 60 minutes, TNF-mRNA peaked and NF-kappaB returned to baseline, whereas all three kinases remained activated. Gadolinium inhibited elastase-induced upregulation of TNF-mRNA (P < 0.001), TNF production (P<0.001), and attenuated SAPK/JNK, as well as ERK1/2, but not p38-MAPK. Both UO126 and SB203580 significantly inhibited elastase-induced upregulation of TNF-mRNA and TNF production (P<0.001), but only UO126 inhibited activation of NF-kappaB. It was concluded that pretranscriptional regulation of TNF gene expression in Kupffer cells follows an orderly activation of p38-MAPK, ERK1/2, and SAPK/JNK that may not converge on NF-kappaB. The seemingly limited duration of NF-kappaB activation may be important in "switching off" the cytokine cascade during acute pancreatitis.


Assuntos
Regulação da Expressão Gênica , Células de Kupffer/metabolismo , Proteínas Quinases Ativadas por Mitógeno/fisiologia , NF-kappa B/fisiologia , Pancreatite/genética , Fator de Necrose Tumoral alfa/genética , Doença Aguda , Animais , Células Cultivadas , Ensaio de Desvio de Mobilidade Eletroforética , Gadolínio/farmacologia , Immunoblotting , Proteínas Quinases JNK Ativadas por Mitógeno , Masculino , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase 3 Ativada por Mitógeno , NF-kappa B/efeitos dos fármacos , Elastase Pancreática/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ativação Transcricional/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...