Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
J Natl Cancer Inst ; 116(3): 421-433, 2024 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-37847647

RESUMO

BACKGROUND: Although the fusion of the transmembrane serine protease 2 gene (TMPRSS2) with the erythroblast transformation-specific-related gene (ERG), or TMPRSS2-ERG, occurs frequently in prostate cancer, its impact on clinical outcomes remains controversial. Roughly half of TMPRSS2-ERG fusions occur through intrachromosomal deletion of interstitial genes and the remainder via insertional chromosomal rearrangements. Because prostate cancers with deletion-derived TMPRSS2-ERG fusions are more aggressive than those with insertional fusions, we investigated the impact of interstitial gene loss on prostate cancer progression. METHODS: We conducted an unbiased analysis of transcriptome data from large collections of prostate cancer samples and employed diverse in vitro and in vivo models combined with genetic approaches to characterize the interstitial gene loss that imposes the most important impact on clinical outcome. RESULTS: This analysis identified FAM3B as the top-ranked interstitial gene whose loss is associated with a poor prognosis. The association between FAM3B loss and poor clinical outcome extended to fusion-negative prostate cancers where FAM3B downregulation occurred through epigenetic imprinting. Importantly, FAM3B loss drives disease progression in prostate cancer. FAM3B acts as an intermediator of a self-governing androgen receptor feedback loop. Specifically, androgen receptor upregulates FAM3B expression by binding to an intronic enhancer to induce an enhancer RNA and facilitate enhancer-promoter looping. FAM3B, in turn, attenuates androgen receptor signaling. CONCLUSION: Loss of FAM3B in prostate cancer, whether through the TMPRSS2-ERG translocation or epigenetic imprinting, causes an exit from this autoregulatory loop to unleash androgen receptor activity and prostate cancer progression. These findings establish FAM3B loss as a new driver of prostate cancer progression and support the utility of FAM3B loss as a biomarker to better define aggressive prostate cancer.


Assuntos
Neoplasias da Próstata , Receptores Androgênicos , Masculino , Humanos , Receptores Androgênicos/genética , Retroalimentação , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Transcriptoma , Proteínas de Fusão Oncogênica/genética , Regulador Transcricional ERG/genética , Regulador Transcricional ERG/metabolismo , Proteínas de Neoplasias/genética , Citocinas/genética
2.
FEBS Open Bio ; 14(3): 434-443, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38129973

RESUMO

Type 1 diabetes (T1D) is an autoimmune disease initiated by genetic predisposition and environmental influences, which result in the specific destruction of insulin-producing pancreatic ß-cells. Currently, there are over 1.6 million cases of T1D in the United States with a worldwide incidence rate that has been increasing since 1990. Here, we examined the effect of Cornus officinalis (CO), a well-known ethnopharmacological agent, on a T1D model of the non-obese diabetic (NOD) mouse. A measured dose of CO extract was delivered into 10-week-old NOD mice by oral gavage for 15 weeks. T1D incidence and hyperglycemia were significantly lower in the CO-treated group as compared to the water gavage (WT) and a no handling or treatment control group (NHT) following treatment. T1D onset per group was 30%, 60% and 86% for the CO, WT and NHT groups, respectively. Circulating C-peptide was higher, and pancreatic insulitis was decreased in non-T1D CO-treated mice. Our findings suggest that CO may have therapeutic potential as both a safe and effective interventional agent to slow early stage T1D progression.


Assuntos
Cornus , Diabetes Mellitus Tipo 1 , Hiperglicemia , Células Secretoras de Insulina , Camundongos , Animais , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/genética , Camundongos Endogâmicos NOD , Hiperglicemia/tratamento farmacológico
3.
J Diabetes Complications ; 37(7): 108494, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37209505

RESUMO

AIMS: This cross-sectional analysis explored the relationships between periodontal disease (PD) and subclinical CVD in a cohort of patients with type 1 diabetes and non-diabetic controls. METHODS: Data were collected from adults enrolled in the Coronary Artery Calcification in Type 1 Diabetes (CACTI) study or enrolled through the Barbara Davis Center for Diabetes Adult Clinic. A clinical periodontal exam measured attachment loss and probing depth. Brachial artery distensibility (brachD), carotid intima-media thickness (cIMT), and pulse wave velocity (PWV) were assessed as measures of subclinical cardiovascular structure and function. RESULTS: 144 participants with T1D and 148 non-diabetics were enrolled. Compared to non-diabetic controls, T1D participants had a higher probing depth (2.6 mm vs. 2.5 mm; p = 0.04), higher attachment loss (2.7 mm vs. 2.4 mm; p < 0.01), lower brachD (mean 5.8 vs. 6.4 mmHg; p < 0.01), a higher cIMT (mean 0.68 vs. 0.64 mm; p < 0.01), and a higher PWV (mean 8.3 vs. 7.8 m/s; p < 0.01). There were no significant associations between PD and CVD metrics. CONCLUSIONS: Periodontal and cardiovascular health was worse in participants with T1D compared to non-diabetics. No significant associations between PD measures and CVD were identified.


Assuntos
Doenças Cardiovasculares , Diabetes Mellitus Tipo 1 , Doenças Periodontais , Periodontite , Adulto , Humanos , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 1/epidemiologia , Estudos Transversais , Fatores de Risco , Espessura Intima-Media Carotídea , Doenças Cardiovasculares/complicações , Doenças Cardiovasculares/epidemiologia , Análise de Onda de Pulso , Fatores de Risco de Doenças Cardíacas , Doenças Periodontais/complicações , Doenças Periodontais/epidemiologia
4.
Mol Cell Endocrinol ; 557: 111773, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36100124

RESUMO

Type 1 diabetes (T1D) is an autoimmune disease initiated by genetic predisposition and environmental influences culminating in the immunologically mediated destruction of pancreatic ß-cells with eventual loss of insulin production. Although T1D can be accurately predicted via autoantibodies, therapies are lacking that can intercede autoimmunity and protect pancreatic ß-cells. There are no approved interventional modalities established for this purpose. One such potential source for clinical agents of this use is from the frequently utilized Cornus officinalis (CO) in the field of ethnopharmacology. Studies by our lab and others have demonstrated that CO has robust proliferative, metabolic, and cytokine protective effects on pancreatic ß-cells. To identify the molecular mechanism of the biological effects of CO, we performed a proteomic and phosphoproteomic analysis examining the cellular networks impacted by CO application on the 1.1B4 pancreatic ß-cell line. Our label-free mass spectrometry approach has demonstrated significant increased phosphorylation of the selective autophagy receptor of p62 (Sequestosome-1/SQSTM1/p62) and predicted activation of the antioxidant Kelch-like ECH-associated protein 1 (Keap1)/Nuclear factor-erythroid factor 2-related factor 2 (Nrf2) pathway. Further validation by immunoblotting and immunofluorescence revealed markers of autophagy such as increased LC3-II and decreased total p62 along with nuclear localization of Nrf2. Both autophagy and the Keap1/Nrf2 pathways have been shown to be impaired in human and animal models of T1D and may serve as an excellent potential therapeutic target stimulated by CO.


Assuntos
Cornus , Diabetes Mellitus Tipo 1 , Insulinas , Animais , Antioxidantes/metabolismo , Autoanticorpos , Autofagia/fisiologia , Citocinas/metabolismo , Humanos , Insulinas/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Proteômica , Proteína Sequestossoma-1/metabolismo
5.
Sci Rep ; 11(1): 2658, 2021 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-33514800

RESUMO

Saliva has immense potential as a diagnostic fluid for identification and monitoring of several systemic diseases. Composition of the microbiome and inflammation has been associated and reflective of oral and overall health. In addition, the relative ease of collection of saliva further strengthens large-scale diagnostic purposes. However, the future clinical utility of saliva cannot be fully determined without a detailed examination of daily fluctuations that may occur within the oral microbiome and inflammation due to circadian rhythm. In this study, we explored the association between the salivary microbiome and the concentration of IL-1ß, IL-6 and IL-8 in the saliva of 12 healthy adults over a period of 24 h by studying the 16S rRNA gene followed by negative binomial mixed model regression analysis. To determine the periodicity and oscillation patterns of both the oral microbiome and inflammation (represented by the cytokine levels), two of the twelve subjects were studied for three consecutive days. Our results indicate that the Operational Taxonomic Units (OTUs) belonging to Prevotella, SR1 and Ruminococcaceae are significantly associated to IL-1ß while Prevotella and Granulicatella were associated with IL-8. Our findings have also revealed a periodicity of both the oral microbiome (OTUs) and inflammation (cytokine levels) with identifiable patterns between IL-1ß and Prevotella, and IL-6 with Prevotella, Neisseria and Porphyromonas. We believe that this study represents the first measure and demonstration of simultaneous periodic fluctuations of cytokine levels and specific populations of the oral microbiome.


Assuntos
Bactérias/metabolismo , Ritmo Circadiano , Citocinas/metabolismo , Microbiota , Saliva/metabolismo , Proteínas e Peptídeos Salivares/metabolismo , Adulto , Feminino , Humanos , Estudos Longitudinais , Masculino
6.
Data Brief ; 25: 104401, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31497634

RESUMO

This article displays raw data linked to the research article "Compositional Analysis and Biological Characterization of Cornus officinalis on Human 1.1B4 Pancreatic ß Cells" [1]. This data was generated by utilizing HPLC/(+and -)ESI-MSn on Cornus officinalis (CO) from four independent sources [1]. The aim was to identify the chemical profile of CO from multiple sources to compare the similarities and differences resulting from various processing methods, and compile a list of known and novel constituents to elucidate the bioactive ingredients. This report contains the full chromatogram and a raw list of the constituents found in CO including chemical name, retention time, and molecular weight from all four sources. All data from HPLC/MS analysis is raw and unprocessed.

7.
Mol Cell Endocrinol ; 494: 110491, 2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31255730

RESUMO

Type 1 diabetes (T1D) is an autoimmune disease resulting from the loss of pancreatic ß cells and subsequent insulin production. Novel interventional therapies are urgently needed that can protect existing ß cells from cytokine-induced death and enhance their function before symptomatic onset. Our initial evidence is suggesting that bioactive ingredients within Cornus officinalis (CO) may be able to serve in this function. CO has been extensively used in Traditional Chinese Medicine (TCM) and reported to possess both anti-inflammatory and pro-metabolic effects. We hypothesize that CO treatment may provide a future potential candidate for interventional therapy for early stage T1D prior to significant ß cell loss. Our data demonstrated that CO can inhibit cytokine-mediated ß cell death, increase cell viability and oxidative capacity, and increase expression of NFATC2 (Nuclear Factor of Activated T Cells, Cytoplasmic 2). We have also profiled the bioactive components in CO from multiple sources by HPLC/MS (High Performance Liquid Chromatography/Mass Spectrometry) analysis. Altogether, CO significantly increases the energy metabolism of ß cells while inducing the NFAT pathway to signal for increased proliferation and endocrine function.


Assuntos
Cornus/química , Células Secretoras de Insulina/metabolismo , Extratos Vegetais/química , Extratos Vegetais/farmacologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Respiração Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citocinas/farmacologia , Glicólise/efeitos dos fármacos , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Fatores de Transcrição NFATC/metabolismo , Fenótipo , Compostos Fitoquímicos/química , Compostos Fitoquímicos/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células Th1/efeitos dos fármacos , Fatores de Tempo , Transcriptoma/genética , Regulação para Cima/efeitos dos fármacos
8.
FEBS Open Bio ; 8(7): 1155-1166, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29988574

RESUMO

Pancreatic cancer is one of the most recalcitrant and lethal of all cancers. We examined the effects of Anemarrhena asphodeloides (AA) and timosaponin-AIII (TAIII), a steroidal saponin present in AA, on pancreatic cancer cell proliferation and aimed to elucidate their potential apoptotic mechanisms of action. Viability assays and cell cycle analysis revealed that both AA and TAIII significantly inhibited pancreatic cancer cell proliferation and cell cycle progression compared to treatment with gemcitabine, the standard chemotherapeutic agent for advanced pancreatic cancer. We identified a dose-dependent increase in caspase-dependent apoptosis and activation of pro-apoptotic PI3K/Akt pathway proteins, with a subsequent downregulation of pro-survival PI3K/Akt pathway proteins, in pancreatic cancer cells treated with AA or TAIII over those treated with gemcitabine.

9.
J Clin Transl Endocrinol ; 11: 26-30, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29686968

RESUMO

AIM: PANcreatic-DERived factor (PANDER, FAM3B) is a novel hormone that regulates glucose levels via interaction with both the endocrine pancreas and liver. Prior studies examining PANDER were primarily conducted in murine models or in vitro but little is known regarding the circulating concentration of PANDER in humans, especially with regard to the association of type 2 diabetes (T2D) or overall glycemic regulation. To address this limitation, we performed a cross-sectional analysis of circulating serum PANDER concentration in association with other hormones that serve as either markers of insulin resistance (insulin and adiponectin) or to metabolic parameters of glycemic control such as fasting HbA1c and blood glucose (FBG). METHODS: Fasting serum was obtained from a commercial biorepository from 300 de-identified adult subjects with 150 T2D and non-T2D adult subjects collected from a population within the United States, respectively, matched on gender, age group and race/ethnicity. Concentration of PANDER, insulin and adiponectin were measured for all samples as determined by commercial ELISA. Metadata was provided for each subject including demography, anthropometry, and cigarette and alcohol use. In addition, fasting blood glucose (FBG) and HbA1c were available on T2D subjects. RESULTS: Multiple linear regression analyses were performed to examine the relationships between circulating log PANDER concentration on HbA1c, fasting glucose, log insulin, log HOMA-ß and log HOMA-IR among T2D subjects and for insulin and adiponectin in non-T2D subjects. A significant linear association was identified between PANDER with fasting HbA1c (ß 0.832 ±â€¯SE 0.22, p = 0.0003) and FBG (ß 20.66 ±â€¯SE 7.43, p = 0.006) within T2D subjects. However, insulin, HOMA-ß, HOMA-IR and adiponectin (p > 0.05) were not found to be linearly associated with PANDER concentration. CONCLUSION: Within T2D subjects, PANDER is modestly linearly associated with increased HbA1c and FBG in a US population. In addition, highest circulating PANDER levels were measured in T2D subjects with HbA1c above 9.9. No association was identified with PANDER and insulin resistance or pancreatic ß-cell function in T2D subjects.

10.
Diabetologia ; 61(1): 254, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29080005

RESUMO

The authors regret that the SNP in SH2B3 was incorrectly referred to as rs3184505 instead of rs3184504 on both mentions in this paper (Methods section and Table 1).

11.
Diabetologia ; 60(10): 1931-1940, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28770319

RESUMO

AIMS/HYPOTHESIS: Respiratory infections and onset of islet autoimmunity are reported to correlate positively in two small prospective studies. The Environmental Determinants of Diabetes in the Young (TEDDY) study is the largest prospective international cohort study on the environmental determinants of type 1 diabetes that regularly monitors both clinical infections and islet autoantibodies. The aim was to confirm the influence of reported respiratory infections and to further characterise the temporal relationship with autoantibody seroconversion. METHODS: During the years 2004-2009, 8676 newborn babies with HLA genotypes conferring an increased risk of type 1 diabetes were enrolled at 3 months of age to participate in a 15 year follow-up. In the present study, the association between parent-reported respiratory infections and islet autoantibodies at 3 month intervals up to 4 years of age was evaluated in 7869 children. Time-dependent proportional hazard models were used to assess how the timing of respiratory infections related to persistent confirmed islet autoimmunity, defined as autoantibody positivity against insulin, GAD and/or insulinoma antigen-2, concordant at two reference laboratories on two or more consecutive visits. RESULTS: In total, 87,327 parent-reported respiratory infectious episodes were recorded while the children were under study surveillance for islet autoimmunity, and 454 children seroconverted. The number of respiratory infections occurring in a 9 month period was associated with the subsequent risk of autoimmunity (p < 0.001). For each 1/year rate increase in infections, the hazard of islet autoimmunity increased by 5.6% (95% CI 2.5%, 8.8%). The risk association was linked primarily to infections occurring in the winter (HR 1.42 [95% CI 1.16, 1.74]; p < 0.001). The types of respiratory infection independently associated with autoimmunity were common cold, influenza-like illness, sinusitis, and laryngitis/tracheitis, with HRs (95% CI) of 1.38 (1.11, 1.71), 2.37 (1.35, 4.15), 2.63 (1.22, 5.67) and 1.76 (1.04, 2.98), respectively. CONCLUSIONS/INTERPRETATION: Recent respiratory infections in young children correlate with an increased risk of islet autoimmunity in the TEDDY study. Further studies to identify the potential causative viruses with pathogen-specific assays should focus especially on the 9 month time window leading to autoantibody seroconversion.


Assuntos
Autoimunidade/imunologia , Diabetes Mellitus Tipo 1/imunologia , Ilhotas Pancreáticas/imunologia , Infecções Respiratórias/imunologia , Adolescente , Autoanticorpos/imunologia , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Fatores de Risco
12.
Obesity (Silver Spring) ; 25(7): 1284-1291, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28558132

RESUMO

OBJECTIVE: To examine the association between pericardial adipose tissue (PAT) and the ratio of PAT to subcutaneous adipose tissue (SAT) with insulin resistance in adults with and without type 1 diabetes (T1D). METHODS: Data for this report came from a substudy of the Coronary Artery Calcification in Type 1 Diabetes cohort (n = 83; 38 with T1D, 45 without T1D). Insulin resistance was measured by hyperinsulinemic-euglycemic clamp. Abdominal computed tomography (CT) was used to measure visceral adipose tissue (VAT) and SAT. PAT was measured from CT scans of the heart. RESULTS: PAT and the ratio of PAT to SAT was higher in males compared to females. After adjustment for demographics, diabetes, blood pressure and lipid factors, BMI, VAT, and log PAT/SAT ratio, log PAT was positively associated with the glucose infusion rate (GIR) in females only (ß = 3.36 ± 1.96, P = 0.097, P for sex interaction = 0.055). Conversely, the log PAT/SAT ratio was significantly associated with decreased GIR in both males and females (ß = -2.08 ± 1.03, P = 0.047, P for sex interaction = 0.768). CONCLUSIONS: A significant association between the PAT/SAT ratio and insulin resistance was found, independent of BMI, VAT, and PAT. These results highlight the importance of considering fat distribution independent of volume.


Assuntos
Distribuição da Gordura Corporal , Resistência à Insulina , Pericárdio/metabolismo , Gordura Subcutânea/metabolismo , Adulto , Glicemia/metabolismo , Pressão Sanguínea , Índice de Massa Corporal , Estudos de Casos e Controles , Diabetes Mellitus Tipo 1/diagnóstico , Feminino , Seguimentos , Hemoglobinas Glicadas/metabolismo , Humanos , Gordura Intra-Abdominal/metabolismo , Masculino , Pessoa de Meia-Idade , Tomógrafos Computadorizados
13.
Data Brief ; 9: 159-62, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27642623

RESUMO

This article contains raw and processed data related to research published in "Quantitative Proteomic Profiling Reveals Hepatic Lipogenesis and Liver X Receptor Activation in the PANDER Transgenic Model" (M.G. Athanason, W.A. Ratliff, D. Chaput, C.B. MarElia, M.N. Kuehl, S.M., Jr. Stevens, B.R. Burkhardt (2016)) [1], and was generated by "spike-in" SILAC-based proteomic analysis of livers obtained from the PANcreatic-Derived factor (PANDER) transgenic mouse (PANTG) under various metabolic conditions [1]. The mass spectrometry output of the PANTG and wild-type B6SJLF mice liver tissue and resulting proteome search from MaxQuant 1.2.2.5 employing the Andromeda search algorithm against the UniprotKB reference database for Mus musculus has been deposited to the ProteomeXchange Consortium (http://www.proteomexchange.org) via the PRIDE partner repository with dataset identifiers PRIDE: PXD004171 and doi:10.6019/PXD004171. Protein ratio values representing PANTG/wild-type obtained by MaxQuant analysis were input into the Perseus processing suite to determine statistical significance using the Significance A outlier test (p<0.05). Differentially expressed proteins using this approach were input into Ingenuity Pathway Analysis to determined altered pathways and upstream regulators that were altered in PANTG mice.

14.
Mol Cell Endocrinol ; 436: 41-9, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27394190

RESUMO

PANcreatic-DERived factor (PANDER) is a member of a superfamily of FAM3 proteins modulating glycemic levels by metabolic regulation of the liver and pancreas. The precise PANDER-induced hepatic signaling mechanism is still being elucidated and has been very complex due to the pleiotropic nature of this novel hormone. Our PANDER transgenic (PANTG) mouse displays a selective hepatic insulin resistant (SHIR) phenotype whereby insulin signaling is blunted yet lipogenesis is increased, a phenomena observed in type 2 diabetes. To examine the complex PANDER-induced mechanism of SHIR, we utilized quantitative mass spectrometry-based proteomic analysis using Stable Isotope Labeling by Amino Acids in Cell Culture (SILAC) to reveal the global hepatic proteome differences within the PANTG under the metabolic states of fasting, fed and insulin-stimulated conditions. Proteomic analysis identified lipid metabolism as one of the top cellular functions differentially altered in all metabolic states. Differentially expressed proteins within the PANTG having a lipid metabolic role included ACC, ACLY, CD36, CYP7A1, FASN and SCD1. Central to the differentially expressed proteins involved in lipid metabolism was the predicted activation of the liver X receptor (LXR) pathway. Western analysis validated the increased hepatic expression of LXRα along with LXR-directed targets such as FASN and CYP7A1 within the PANTG liver. Furthermore, recombinant PANDER was capable of inducing LXR promoter activity in-vitro as determined by luciferase reporter assays. Taken together, PANDER strongly impacts hepatic lipid metabolism across metabolic states and may induce a SHIR phenotype via the LXR pathway.


Assuntos
Citocinas/genética , Lipogênese , Receptores X do Fígado/metabolismo , Fígado/metabolismo , Proteômica/métodos , Animais , Western Blotting , Ácido Graxo Sintases/metabolismo , Feminino , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Marcação por Isótopo , Lipogênese/genética , Receptores X do Fígado/genética , Masculino , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Reprodutibilidade dos Testes , Transcrição Gênica
15.
Mol Cell Proteomics ; 14(12): 3173-84, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26424600

RESUMO

Microglia, the resident immune cells of the brain, have been shown to display a complex spectrum of roles that span from neurotrophic to neurotoxic depending on their activation status. Microglia can be classified into four stages of activation, M1, which most closely matches the classical (pro-inflammatory) activation stage, and the alternative activation stages M2a, M2b, and M2c. The alternative activation stages have not yet been comprehensively analyzed through unbiased, global-scale protein expression profiling. In this study, BV2 mouse immortalized microglial cells were stimulated with agonists specific for each of the four stages and total protein expression for 4644 protein groups was quantified using SILAC-based proteomic analysis. After validating induction of the various stages through a targeted cytokine assay and Western blotting of activation states, the data revealed novel insights into the similarities and differences between the various states. The data identify several protein groups whose expression in the anti-inflammatory, pro-healing activation states are altered presumably to curtail inflammatory activation through differential protein expression, in the M2a state including CD74, LYN, SQST1, TLR2, and CD14. The differential expression of these proteins promotes healing, limits phagocytosis, and limits activation of reactive nitrogen species through toll-like receptor cascades. The M2c state appears to center around the down-regulation of a key member in the formation of actin-rich phagosomes, SLP-76. In addition, the proteomic data identified a novel activation marker, DAB2, which is involved in clathrin-mediated endocytosis and is significantly different between M2a and either M1 or M2b states. Western blot analysis of mouse primary microglia stimulated with the various agonists of the classical and alternative activation states revealed a similar trend of DAB2 expression compared with BV2 cells.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Aminoácidos/química , Microglia/citologia , Proteômica/métodos , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas Reguladoras de Apoptose , Técnicas de Cultura de Células , Linhagem Celular , Regulação da Expressão Gênica , Marcação por Isótopo , Lipopolissacarídeos/farmacologia , Camundongos , Microglia/efeitos dos fármacos , Microglia/metabolismo
16.
Mol Cell Endocrinol ; 413: 101-12, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26123584

RESUMO

PANcreatic-DERived factor (PANDER, FAM3B) has been shown to regulate glycemic levels via interactions with both pancreatic islets and the liver. Although PANDER is predominantly expressed from the endocrine pancreas, recent work has provided sufficient evidence that the liver may also be an additional tissue source of PANDER production. At physiological levels, PANDER is capable of disrupting insulin signaling and promoting increased hepatic glucose production. As shown in some animal models, strong expression of PANDER, induced by viral delivery within the liver, induces hepatic steatosis. However, no studies to date have explicitly characterized the transcriptional regulation of PANDER from the liver. Therefore, our investigation elucidated the nutrient and hormonal regulation of the hepatic PANDER promoter. Initial RNA-ligated rapid amplification of cDNA ends identified a novel transcription start site (TSS) approximately 26 bp upstream of the PANDER translational start codon not previously revealed in pancreatic ß-cell lines. Western evaluation of various murine tissues demonstrated robust expression in the liver and brain. Promoter analysis identified strong tissue-specific activity of the PANDER promoter in both human and murine liver-derived cell lines. The minimal element responsible for maximal promoter activity within hepatic cell lines was located between -293 and -3 of the identified TSS. PANDER promoter activity was inhibited by both insulin and palmitate, whereas glucose strongly increased expression. The minimal element was responsible for maximal glucose-responsive and basal activity. Co-transfection reporter assays, chromatin-immunoprecipitation (ChIP) and site-directed mutagenesis revealed that the carbohydrate-responsive element binding protein (ChREBP) increased PANDER promoter activity and interacted with the PANDER promoter. E-box 3 was shown to be critical for basal and glucose responsive expression. In summary, in-vitro and in-vivo glucose is a potent stimulator of the PANDER promoter within the liver and this response may be facilitated by ChREBP.


Assuntos
Citocinas/biossíntese , Glucose/metabolismo , Insulina/metabolismo , Fígado/metabolismo , Proteínas de Neoplasias/biossíntese , Elementos de Resposta/fisiologia , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Citocinas/genética , Células Hep G2 , Humanos , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Fígado/citologia , Camundongos , Células NIH 3T3 , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
17.
PLoS One ; 10(4): e0125320, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25915398

RESUMO

BACKGROUND: Type 1 diabetes (T1D) is an autoimmune disease resulting in the targeted destruction of pancreatic ß-cells and permanent loss of insulin production. Proper glucose management results in better clinical outcomes for T1D and provides a strong rationale to identify non-invasive biomarkers indicative or predictive of glycemic control. Therefore, we investigated the association of salivary inflammation with HbA1c in a T1D cohort. METHODS: Unstimulated saliva was collected from 144 subjects with T1D at the USF Diabetes Center. BMI, duration of diabetes, and HbA1c were recorded during clinical visit. Levels of interleukin (IL)-1ß, -6, -8, -10, IFN-γ, TNF-α, MMP-3, -8, and -9 were measured using multiplexing immunoassay analysis. To account for smoking status, salivary cotinine levels were also determined. RESULTS: Multiple linear (HbA1c) and logistic (self-reported gingival condition) regression analyses were performed to examine the relationships between the Principal Component Analysis (PCA) components and HbA1c and gingival condition (adjusted for age, duration of diabetes, BMI, and sex; model for HbA1c also adjusted for gingival condition and model for gingival condition also adjusted for HbA1c). PCA components 1 (MMP-8 and MMP-9) and 3 (TNF-α) were significantly associated with HbA1c (ß = 0.28 ±0.14, p = 0.045; ß = 0.31 ±0.14, p = 0.029), while PCA component 2 (IL-6, IL-1ß, and IL-8) was significantly associated with gingival condition (OR 1.60 95% CI 1.09-2.34, p = 0.016). In general, increased salivary inflammatory burden is associated with decreased glycemic control and self-reported gingival condition. CONCLUSIONS: The saliva may represent a useful reservoir of novel noninvasive inflammatory biomarkers predictive of the progression and control of T1D.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , Hemoglobinas Glicadas/metabolismo , Metaloproteinase 8 da Matriz/metabolismo , Saliva/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Biomarcadores/metabolismo , Humanos , Modelos Logísticos , Metaloproteinase 9 da Matriz/metabolismo , Análise de Componente Principal
18.
PLoS One ; 10(2): e0117156, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25679806

RESUMO

OBJECTIVE: Pancreatic-derived factor (PANDER, also named as FAM3B) is secreted by pancreatic α and ß cells. Increasing evidence suggests that it may serve a hormonal function related to glycemic and lipid metabolism. In this study, we investigated the effects of PANDER overexpression on hepatic and adipose triglyceride metabolism in high-fat diet-fed male C57BL/6 mice. METHODS: PANDER overexpression was achieved by tail-vein injection of recombinant Ad-PANDER and Ad-GFP injected mice served as a control. The TG metabolism in both groups were compared. RESULTS: Adenoviral-mediated overexpression of PANDER did not affect body weight, food consumption, or liver enzymes. The triglyceride (TG) content of both liver and adipose tissue was significantly decreased in Ad-PANDER mice (liver: 6.16±1.89 mg/g vs. control 14.95±2.27 mg/g, P<0.05; adipose: 39.31±1.99 mg/100mg vs. 47.22±2.21 mg/100mg, P<0.05). The free fatty acid (FFA) content of adipose tissue in Ad-PANDER mice was also decreased (1.38±0.18 mg/g vs. 2.77±0.31 mg/g, P<0.01). The investigation of key enzymes of triglyceride hydrolysis and FFA oxidation in liver and adipose tissue showed that p-HSL/HSL was significantly increased and that DGAT1 gene and protein expression were significantly reduced in the liver of PANDER-overexpressing mice. PKA phosphorylation was also significantly increased in the livers of Ad-PANDER mice. No differences in ATGL, CPT1, ACOX1, or DGAT2 expression were observed. CONCLUSION: Overexpression of PANDER is associated with observable decreases in TG, increases in PKA phosphorylation, and decreased DGAT1 expression, suggesting a possible interrelationship. The mechanisms by which this occurs remain to be elucidated.


Assuntos
Citocinas/genética , Diacilglicerol O-Aciltransferase/genética , Regulação da Expressão Gênica , Fígado/metabolismo , Triglicerídeos/metabolismo , Adenoviridae/genética , Tecido Adiposo/metabolismo , Animais , Peso Corporal , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Citocinas/metabolismo , Dieta Hiperlipídica , Vetores Genéticos/genética , Lipídeos/sangue , Lipólise , Masculino , Camundongos , Fosforilação
19.
Dis Model Mech ; 7(11): 1307-15, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25217499

RESUMO

Pancreatic-derived factor (PANDER; also known as FAM3B) is a uniquely structured protein strongly expressed within and secreted from the endocrine pancreas. PANDER has been hypothesized to regulate fasting and fed glucose homeostasis, hepatic lipogenesis and insulin signaling, and to serve a potential role in the onset or progression of type 2 diabetes (T2D). Despite having potentially pivotal pleiotropic roles in glycemic regulation and T2D, there has been limited generation of stable animal models for the investigation of PANDER function, and there are no models on well-established genetic murine backgrounds for T2D. Our aim was to generate an enhanced murine model to further elucidate the biological function of PANDER. Therefore, a pure-bred PANDER knockout C57BL/6 (PANKO-C57) model was created and phenotypically characterized with respect to glycemic regulation and hepatic insulin signaling. The PANKO-C57 model exhibited an enhanced metabolic phenotype, particularly with regard to enhanced glucose tolerance. Male PANKO-C57 mice displayed decreased fasting plasma insulin and C-peptide levels, whereas leptin levels were increased as compared with matched C57BL/6J wild-type mice. Despite similar peripheral insulin sensitivity between both groups, hepatic insulin signaling was significantly increased during fasting conditions, as demonstrated by increased phosphorylation of hepatic PKB/Akt and AMPK, along with mature SREBP-1 expression. Insulin stimulation of PANKO-C57 mice resulted in increased hepatic triglyceride and glycogen content as compared with wild-type C57BL/6 mice. In summary, the PANKO-C57 mouse represents a suitable model for the investigation of PANDER in multiple metabolic states and provides an additional tool to elucidate the biological function and potential role in T2D.


Assuntos
Citocinas/metabolismo , Teste de Tolerância a Glucose , Animais , Citocinas/genética , Feminino , Insulina/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
20.
J Endocrinol ; 220(3): 219-31, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24468680

RESUMO

PANcreatic-DERived factor (PANDER, FAM3B) is a novel protein that is highly expressed within the endocrine pancreas and to a lesser degree in other tissues. Under glucose stimulation, PANDER is co-secreted with insulin from the ß-cell. Despite prior creation and characterization of acute hepatic PANDER animal models, the physiologic function remains to be elucidated from pancreas-secreted PANDER. To determine this, in this study, a transgenic mouse exclusively overexpressing PANDER from the endocrine pancreas was generated. PANDER was selectively expressed by the pancreatic-duodenal homeobox-1 (PDX1) promoter. The PANDER transgenic (PANTG) mice were metabolically and proteomically characterized to evaluate effects on glucose homeostasis, insulin sensitivity, and lipid metabolism. Fasting glucose, insulin and C-peptide levels were elevated in the PANTG compared with matched WT mice. Younger PANTG mice also displayed glucose intolerance in the absence of peripheral insulin sensitivity. Hyperinsulinemic-euglycemic clamp studies revealed that hepatic glucose production and insulin resistance were significantly increased in the PANTG with no difference in either glucose infusion rate or rate of disappearance. Fasting glucagon, corticosterones, resistin and leptin levels were also similar between PANTG and WT. Stable isotope labeling of amino acids in cell culture revealed increased gluconeogenic and lipogenic proteomic profiles within the liver of the PANTG with phosphoenol-pyruvate carboxykinase demonstrating a 3.5-fold increase in expression. This was matched with increased hepatic triglyceride content and decreased p-AMPK and p-acetyl coenzyme A carboxylase-1 signaling in the PANTG. Overall, our findings support a role of pancreatic ß-cell-secreted PANDER in the regulation of hepatic insulin and lipogenenic signaling with subsequent impact on overall glycemia.


Assuntos
Citocinas/metabolismo , Hiperglicemia/metabolismo , Resistência à Insulina , Insulina/metabolismo , Fígado/metabolismo , Proteínas de Neoplasias/metabolismo , Animais , Citocinas/genética , Modelos Animais de Doenças , Jejum/metabolismo , Feminino , Glucose/metabolismo , Humanos , Hiperglicemia/genética , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas de Neoplasias/genética , Triglicerídeos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...