1
|
Moniruzzaman M, Bezerra AB, Mohibullah M, Judd RL, Granneman JG, Easley CJ. Dynamic sampling from ex vivo adipose tissue using droplet-based microfluidics supports separate mechanisms for glycerol and fatty acid secretion. LAB ON A CHIP 2024; 24:5020-5031. [PMID: 39344798 DOI: 10.1039/d4lc00664j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Pathologies in adipose (fat) tissue function are linked with human diseases such as diabetes, obesity, metabolic syndrome, and cancer. Dynamic, rapid release of metabolites has been observed in adipocyte cells and tissue, yet higher temporal resolution is needed to adequately study this process. In this work, a microfluidic device with precise and regular valve-automated droplet sampling, termed a microfluidic analog-to-digital converter (μADC), was used to sample secretions from ∼0.75 mm diameter adipose explants from mice, and on-chip salt water electrodes were used to merge sampled droplets with reagent droplets from two different fluorometric coupled enzyme assays. By integrating sampling and assays on-chip, either glycerol or non-esterified fatty acids (NEFA), or both, were quantified optically within merged 12 nanoliter droplets using a fluorescence microscope with as high as 20 second temporal resolution. Limits of detection were 6 μM for glycerol (70 fmol) and 0.9 μM for NEFA (10 fmol). Multiple ex vivo adipose tissue explants were analyzed with this system, all showing clear increases in lipolytic function after switching from feeding to fasting conditions. Enabled by high temporal resolution, lipolytic oscillations of both glycerol and NEFA were observed for the first time in the range of 0.2 to 1.6 min-1. Continuous wavelet transform (CWT) spectrograms and burst analyses (0.1 to 4.0 pmol bursts) revealed complex dynamics, with multiplexed assays (duplex for glycerol and NEFA) from the same explants showing mostly discordant bursts. These data support separate mechanisms of NEFA and glycerol release, although the connection to intracellular metabolic oscillations remains unknown. Overall, this device allowed automated and highly precise temporal sampling of tissue explants at high resolution and programmable downstream merging with multiple assay reagents, revealing unique biological information. Such device features should be applicable to various other tissue or spheroid types and to other assay formats.
Collapse
Affiliation(s)
- Md Moniruzzaman
- Department of Chemistry and Biochemistry, Auburn University, Auburn, AL, USA.
| | - Andresa B Bezerra
- Department of Chemistry and Biochemistry, Auburn University, Auburn, AL, USA.
| | - Md Mohibullah
- Department of Chemistry and Biochemistry, Auburn University, Auburn, AL, USA.
| | - Robert L Judd
- Department of Anatomy, Physiology, and Pharmacology, Auburn University, Auburn, AL, USA
| | - James G Granneman
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | | |
Collapse
|
2
|
Okasha HH, Hegazy MA, Shaker O, Elfatah YA, El-Sawy SS, Abdelfatah D, Abdellatef A. Study of non-alcoholic fatty pancreatic disease among the Egyptian population and the value of serum fatty acid binding protein-1 (FABP-1) as a non-invasive biomarker. Clin Res Hepatol Gastroenterol 2024; 48:102364. [PMID: 38788255 DOI: 10.1016/j.clinre.2024.102364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 04/10/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND Non-alcoholic fatty pancreas disease (NAFPD) can be detected using various imaging techniques, but accurately measuring the amount of fat in the pancreas remains difficult. Fatty acid binding protein-1 (FABP-1) is a marker specific to certain tissues and can aid in diagnosing NAFPD. However, this study aimed to investigate the prevalence of NAFPD among obese and non-obese people with and without diabetes mellitus (DM). Additionally, it aimed to evaluate the associated risk factors for NAFPD and the utility of the FABP-1 level as a simple, non-invasive biomarker for diagnosing NAFPD. METHODS This study is a prospective cross-sectional study. RESULTS Ninety-five patients were enrolled in the study, comprising 35 males and 60 females, with a mean age of 44 years and a standard deviation (SD) of 11 years. However, 26.3 % were morbidly obese, 22.1 % were severely obese, 31.6 % were obese, 12.6 % were overweight, and 7.4 % were normal. Additionally, 35.8 % had diabetes mellitus, while 26.3 % of patients had hypertension. Regarding the ultrasonographic findings, 94.7 % of the patients had fatty liver, with the majority (41.1 %) classified as grade II, followed by 38.9 % classified as grade I, and 14.7 % classified as grade III fatty liver. Among these patients, 78.9 % had fatty pancreas, with 38.9 % classified as grade II, 31.6 % classified as grade I, and 8.4 % classified as grade III fatty pancreas. The median FABP-1 level among patients with fatty pancreas was 3.3 ng/ml, which exhibited a significant fair negative correlation with total bilirubin and a fair, positive correlation with alkaline phosphatase and portal vein diameter. A statistically substantial distinction was observed between the levels of AFABP-1 and the presence or grading of the fatty pancreas (p-value = 0.048 and < 0.001, respectively). Using multivariate analysis, FABP-1 was the only significant predictor of a fatty pancreas. The receiver operating characteristic (ROC) curve analysis indicated that at a cut-off point of FABP-1 of ≤ 3.7, it had a sensitivity of 58 %, specificity of 80 %, positive predictive value (PPV) of 96.6 %, negative predictive value (NPV) of 17 %, and an area under the curve (AUC) of 0.77. CONCLUSION NAFPD is becoming an increasingly significant challenge. FABP-1 can potentially be a straightforward and non-invasive predictor of the fatty pancreas.
Collapse
Affiliation(s)
- Hussein Hassan Okasha
- Internal Medicine Department, Division of Gastroenterology and Hepatology, Kasr Al-Aini School of Medicine, Cairo University, Cairo, Egypt
| | - Mona A Hegazy
- Internal Medicine Department, Division of Gastroenterology and Hepatology, Kasr Al-Aini School of Medicine, Cairo University, Cairo, Egypt
| | - Olfat Shaker
- Medical Biochemistry and Molecular Biology Department, Faculty of medicine, Cairo University, Cairo, Egypt
| | - Yasmine Abd Elfatah
- Internal Medicine Department, DM and endocrinology Division, Kasr Al-Aini School of Medicine, Cairo University, Cairo, Egypt
| | - Shereen Sadik El-Sawy
- Internal Medicine Department, DM and endocrinology Division, Kasr Al-Aini School of Medicine, Cairo University, Cairo, Egypt
| | - Dalia Abdelfatah
- Cancer epidemiology and Biostatistics department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Abeer Abdellatef
- Internal Medicine Department, Division of Gastroenterology and Hepatology, Kasr Al-Aini School of Medicine, Cairo University, Cairo, Egypt.
| |
Collapse
|
3
|
Pagkali A, Makris A, Brofidi K, Agouridis AP, Filippatos TD. Pathophysiological Mechanisms and Clinical Associations of Non-Alcoholic Fatty Pancreas Disease. Diabetes Metab Syndr Obes 2024; 17:283-294. [PMID: 38283640 PMCID: PMC10813232 DOI: 10.2147/dmso.s397643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024] Open
Abstract
Non-Alcoholic Fatty Pancreas disease (NAFPD), characterized by fat accumulation in pancreatic tissue, is an emerging clinical entity. However, the clinical associations, the underlying molecular drivers, and the pathophysiological mechanisms of NAFPD have not yet been characterized in detail. The NAFPD spectrum not only includes infiltration and accumulation of fat within and between pancreatic cells but also involves several inflammatory processes, dysregulation of physiological metabolic pathways, and hormonal defects. A deeper understanding of the underlying molecular mechanisms is key to correlate NAFPD with clinical entities including non-alcoholic fatty liver disease, metabolic syndrome, diabetes mellitus, atherosclerosis, as well as pancreatic cancer and pancreatitis. The aim of this review is to examine the pathophysiological mechanisms of NAFPD and to assess the possible causative/predictive risk factors of NAFPD-related clinical syndromes.
Collapse
Affiliation(s)
- Antonia Pagkali
- School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasios Makris
- School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Kalliopi Brofidi
- Department of Internal Medicine, School of Medicine, University of Crete, Heraklion, Greece
| | - Aris P Agouridis
- School of Medicine, European University Cyprus, Nicosia, Cyprus
- Department of Internal Medicine, German Oncology Center, Limassol, Cyprus
| | | |
Collapse
|
4
|
Fang X, Ye H, Zhang S, Guo L, Xu Y, Zhang D, Nie Q. Investigation of potential genetic factors for growth traits in yellow-feather broilers using weighted single-step genome-wide association study. Poult Sci 2023; 102:103034. [PMID: 37657249 PMCID: PMC10480639 DOI: 10.1016/j.psj.2023.103034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/04/2023] [Accepted: 08/11/2023] [Indexed: 09/03/2023] Open
Abstract
Yellow-feather broilers take a large portion of poultry industry in China due to its meat characteristics. Improving the growth traits of yellow-feathered broilers will have great significance for the Chinese poultry market. The current study was designed to investigate the potential genetic factors using the weighted single-step genome-wide association study (wssGWAS) method, which takes consideration of more factors including pedigree, sex, environment and has more accuracy than traditional GWAS. The yellow-feather dwarf chickens from Wens Nanfang Poultry Breeding Co. Ltd. were revolved to recode 9 growth traits: Average daily gain (ADG), body weight (BW) at 45 d, 49 d, 56 d, 63 d, 70 d, 77 d, 84 d, 91 d for analysis. For the results, the region 4.63 to 5.03 Mb on chromosome 15, which was the QTL overlapped in BW45, BW49, BW56, BW63, BW84, might be the crucial genetic region for growth traits. Seven GO terms and 3 KEGG pathways, GO:0005200, GO:0005882, GO:0045111, GO:0099513, GO:0099081, GO:0099512, GO:0099080, KEGG:gga04020, KEGG:gga04540, KEGG:gga04210, were detected to relevant with growth traits. The genes enriched in these biological processes (NRAS, TUBB1, ADORA2B, NTRK3, NGF, TNNC2, F-KER, LOC429492, LOC431325, LOC431324, LOC396480) might have the function in growth of yellow-feather broilers.
Collapse
Affiliation(s)
- Xiang Fang
- State Key Laboratory of Livestock and Poultry Breeding, & Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou 510642, China
| | - Haoqiang Ye
- State Key Laboratory of Livestock and Poultry Breeding, & Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou 510642, China
| | - Siyu Zhang
- State Key Laboratory of Livestock and Poultry Breeding, & Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou 510642, China
| | - Lijin Guo
- State Key Laboratory of Livestock and Poultry Breeding, & Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou 510642, China; Henry Fok School of Biology and Agriculture, Shaoguan University, Shaoguan 512005, China
| | - Yibin Xu
- State Key Laboratory of Livestock and Poultry Breeding, & Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou 510642, China
| | - Dexiang Zhang
- State Key Laboratory of Livestock and Poultry Breeding, & Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou 510642, China
| | - Qinghua Nie
- State Key Laboratory of Livestock and Poultry Breeding, & Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou 510642, China; Henry Fok School of Biology and Agriculture, Shaoguan University, Shaoguan 512005, China.
| |
Collapse
|
5
|
Chen YC, Lutkewitte AJ, Basavarajappa HD, Fueger PT. Glucolipotoxic Stress-Induced Mig6 Desensitizes EGFR Signaling and Promotes Pancreatic Beta Cell Death. Metabolites 2023; 13:627. [PMID: 37233668 PMCID: PMC10222246 DOI: 10.3390/metabo13050627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 05/27/2023] Open
Abstract
A loss of functional beta cell mass is a final etiological event in the development of frank type 2 diabetes (T2D). To preserve or expand beta cells and therefore treat/prevent T2D, growth factors have been considered therapeutically but have largely failed to achieve robust clinical success. The molecular mechanisms preventing the activation of mitogenic signaling pathways from maintaining functional beta cell mass during the development of T2D remain unknown. We speculated that endogenous negative effectors of mitogenic signaling cascades impede beta cell survival/expansion. Thus, we tested the hypothesis that a stress-inducible epidermal growth factor receptor (EGFR) inhibitor, mitogen-inducible gene 6 (Mig6), regulates beta cell fate in a T2D milieu. To this end, we determined that: (1) glucolipotoxicity (GLT) induces Mig6, thereby blunting EGFR signaling cascades, and (2) Mig6 mediates molecular events regulating beta cell survival/death. We discovered that GLT impairs EGFR activation, and Mig6 is elevated in human islets from T2D donors as well as GLT-treated rodent islets and 832/13 INS-1 beta cells. Mig6 is essential for GLT-induced EGFR desensitization, as Mig6 suppression rescued the GLT-impaired EGFR and ERK1/2 activation. Further, Mig6 mediated EGFR but not insulin-like growth factor-1 receptor nor hepatocyte growth factor receptor activity in beta cells. Finally, we identified that elevated Mig6 augmented beta cell apoptosis, as Mig6 suppression reduced apoptosis during GLT. In conclusion, we established that T2D and GLT induce Mig6 in beta cells; the elevated Mig6 desensitizes EGFR signaling and induces beta cell death, suggesting Mig6 could be a novel therapeutic target for T2D.
Collapse
Affiliation(s)
- Yi-Chun Chen
- Department of Pediatrics and Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Andrew J. Lutkewitte
- Department of Pediatrics and Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Halesha D. Basavarajappa
- Department of Molecular and Cellular Endocrinology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Patrick T. Fueger
- Department of Pediatrics and Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Molecular and Cellular Endocrinology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
6
|
Chan TT, Tse YK, Lui RNS, Wong GLH, Chim AML, Kong APS, Woo J, Yeung DKW, Abrigo JM, Chu WCW, Wong VWS, Tang RSY. Fatty Pancreas Is Independently Associated With Subsequent Diabetes Mellitus Development: A 10-Year Prospective Cohort Study. Clin Gastroenterol Hepatol 2022; 20:2014-2022.e4. [PMID: 34571257 DOI: 10.1016/j.cgh.2021.09.027] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/12/2021] [Accepted: 09/16/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Although the association between fatty pancreas and metabolic syndrome has been suggested in retrospective studies, long-term prospective data on the effect of fatty pancreas on various metabolic outcomes are lacking. We aimed to prospectively investigate the association between fatty pancreas and the development of major metabolic outcomes. METHODS A total of 631 subjects from a population study using fat-water magnetic resonance imaging to quantify pancreatic and liver fat content during 2008 to 2010 were followed up prospectively until December 2020 (mean follow-up time, 11.1 ± 1.1 y). Subjects with significant alcohol intake and diabetes mellitus (DM) at baseline were excluded. Incidence of newly diagnosed DM, hypertension, dyslipidemia, ischemic heart disease, cardiovascular accidents, pancreatic cancer, and mortality were evaluated. RESULTS Among the 631 subjects (mean age, 48 ± 11 y), 93 (14.7%) had fatty pancreas. The fatty pancreas group had a higher incidence of DM (33.3% vs 10.4%; P < .001), hypertension (37.7% vs 22.7%; P = .003), and dyslipidemia (37.7% vs 14.6%; P < .001) during long-term follow-up evaluation. Individuals with both fatty liver and pancreas had the highest DM incidence, followed by fatty liver only and fatty pancreas only groups (P < .001). Fatty pancreas was associated independently with DM (adjusted hazard ratio, 1.81; 95% CI, 1.10-3.00; P = .020), but not hypertension or dyslipidemia on multivariate analysis. Each percentage increase of pancreatic fat increased the risk of incident DM by 7% (adjusted hazard ratio, 1.07; 95% CI, 1.01-1.13; P = .016). No participants developed pancreatic cancer during the follow-up period. CONCLUSIONS Fatty pancreas is associated independently with subsequent DM development, but not hypertension or dyslipidemia.
Collapse
Affiliation(s)
- Ting Ting Chan
- Department of Medicine and Therapeutics, Hong Kong Special Administrative Region, China; Institute of Digestive Disease, Hong Kong Special Administrative Region, China
| | - Yee Kit Tse
- Department of Medicine and Therapeutics, Hong Kong Special Administrative Region, China; Medical Data Analytic Centre, Hong Kong Special Administrative Region, China
| | - Rashid Nok-Shun Lui
- Department of Medicine and Therapeutics, Hong Kong Special Administrative Region, China; Institute of Digestive Disease, Hong Kong Special Administrative Region, China
| | - Grace Lai-Hung Wong
- Department of Medicine and Therapeutics, Hong Kong Special Administrative Region, China; Institute of Digestive Disease, Hong Kong Special Administrative Region, China; Medical Data Analytic Centre, Hong Kong Special Administrative Region, China
| | - Angel Mei-Ling Chim
- Department of Medicine and Therapeutics, Hong Kong Special Administrative Region, China; Institute of Digestive Disease, Hong Kong Special Administrative Region, China
| | - Alice Pik-Shan Kong
- Department of Medicine and Therapeutics, Hong Kong Special Administrative Region, China
| | - Jean Woo
- Department of Medicine and Therapeutics, Hong Kong Special Administrative Region, China
| | - David Ka-Wai Yeung
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jill M Abrigo
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Winnie Chiu-Wing Chu
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Vincent Wai-Sun Wong
- Department of Medicine and Therapeutics, Hong Kong Special Administrative Region, China; Institute of Digestive Disease, Hong Kong Special Administrative Region, China; Medical Data Analytic Centre, Hong Kong Special Administrative Region, China.
| | - Raymond Shing-Yan Tang
- Department of Medicine and Therapeutics, Hong Kong Special Administrative Region, China; Institute of Digestive Disease, Hong Kong Special Administrative Region, China.
| |
Collapse
|
7
|
Zhang M, Yu F, Xue Y, Song L, Du M, Li X, Li W. The Relationship of 25(OH)D 3 with Diabetes Mellitus and the Mediation Effect of Lipid Profile in Chinese Rural Population of Henan Province. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:85. [PMID: 35056393 PMCID: PMC8781849 DOI: 10.3390/medicina58010085] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 01/02/2022] [Indexed: 12/24/2022]
Abstract
Background and Objectives: Studies suggest that vitamin D is involved in the development of type 2 diabetes mellitus (T2DM) and influences serum lipids levels, while lipid disorders are also closely related to T2DM. This study attempts to explore the complex relationship of serum 25(OH)D3, serum lipids, and T2DM among Chinese population. Materials and Methods: A cross-sectional study was carried out among 2326 subjects. The chi-square (χ2) test was applied to compare the prevalence of T2DM or dyslipidemia between two serum 25(OH)D3 levels. Linear regression was applied to analyze the correlation between serum lipids and 25(OH)D3 contents. Univariate and logistic analysis were used to explore the relationship between two lipid levels and T2DM. Mediation analysis was used to explore whether serum lipids mediate the relationship between two serum 25(OH)D3 levels and T2DM. Results: Compared to subjects with 25(OH)D3 ≥ 30 ng/mL, subjects with 25(OH)D3 < 30 ng/mL were higher in the prevalence of T2DM. The occurrences of high TG and low HDL-C were significantly higher in vitamin D deficiency and insufficiency than those in vitamin D sufficiency. Serum 25(OH)D3 content showed a reverse correlation with TC, TG, and LDL-C, but positive correlation with HDL-C. The odds ratios (ORs) (95% confidence intervals, 95%CI) of T2DM by comparing TG ≥ 2.26 mmol/L vs. TG < 2.26 mmol/L and HDL-C < 1.04 mmol/L vs. HDL-C ≥ 1.04 mmol/L in all participants were 2.48 (1.94-3.18) and 1.37 (1.07-1.75), respectively. Serum TG or HDL-C level partially mediated the relationship between two 25(OH)D3 level and T2DM. Conclusions: Serum 25(OH)D3 < 30 ng/mL seems to be associated with T2DM or dyslipidemia (high TG and low HDL-C) in our study, but there is still no proof of a cause-effect relationship. Moreover, serum TG or HDL-C level partially mediated the relationship between 25(OH)D3 levels and T2DM.
Collapse
Affiliation(s)
| | - Fei Yu
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China; (M.Z.); (Y.X.); (L.S.); (M.D.); (W.L.)
| | | | | | | | - Xing Li
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China; (M.Z.); (Y.X.); (L.S.); (M.D.); (W.L.)
| | | |
Collapse
|
8
|
Murao N, Yokoi N, Takahashi H, Hayami T, Minami Y, Seino S. Increased glycolysis affects β-cell function and identity in aging and diabetes. Mol Metab 2022; 55:101414. [PMID: 34871777 PMCID: PMC8732780 DOI: 10.1016/j.molmet.2021.101414] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 11/25/2021] [Accepted: 12/01/2021] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Age is a risk factor for type 2 diabetes (T2D). We aimed to elucidate whether β-cell glucose metabolism is altered with aging and contributes to T2D. METHODS We used senescence-accelerated mice (SAM), C57BL/6J (B6) mice, and ob/ob mice as aging models. As a diabetes model, we used db/db mice. The glucose responsiveness of insulin secretion and the [U-13C]-glucose metabolic flux were examined in isolated islets. We analyzed the expression of β-cell-specific genes in isolated islets and pancreatic sections as molecular signatures of β-cell identity. β cells defective in the malate-aspartate (MA) shuttle were previously generated from MIN6-K8 cells by the knockout of Got1, a component of the shuttle. We analyzed Got1 KO β cells as a model of increased glycolysis. RESULTS We identified hyperresponsiveness to glucose and compromised cellular identity as dysfunctional phenotypes shared in common between aged and diabetic mouse β cells. We also observed a metabolic commonality between aged and diabetic β cells: hyperactive glycolysis through the increased expression of nicotinamide mononucleotide adenylyl transferase 2 (Nmnat2), a cytosolic nicotinamide adenine dinucleotide (NAD)-synthesizing enzyme. Got1 KO β cells showed increased glycolysis, β-cell dysfunction, and impaired cellular identity, phenocopying aging and diabetes. Using Got1 KO β cells, we show that attenuation of glycolysis or Nmnat2 activity can restore β-cell function and identity. CONCLUSIONS Our study demonstrates that hyperactive glycolysis is a metabolic signature of aged and diabetic β cells, which may underlie age-related β-cell dysfunction and loss of cellular identity. We suggest Nmnat2 suppression as an approach to counteract age-related T2D.
Collapse
Affiliation(s)
- Naoya Murao
- Division of Molecular and Metabolic Medicine, Graduate School of Medicine, Kobe University, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Norihide Yokoi
- Division of Molecular and Metabolic Medicine, Graduate School of Medicine, Kobe University, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan; Laboratory of Animal Breeding and Genetics, Graduate School of Agriculture, Kyoto University, Sakyo-ku, Kyoto, Kyoto 606-8502, Japan
| | - Harumi Takahashi
- Division of Molecular and Metabolic Medicine, Graduate School of Medicine, Kobe University, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan.
| | - Tomohide Hayami
- Division of Molecular and Metabolic Medicine, Graduate School of Medicine, Kobe University, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan; Division of Diabetes, Department of Internal Medicine, Aichi Medical University, Nagakute, Aichi 480-1195, Japan
| | - Yasuhiro Minami
- Division of Cell Physiology, Graduate School of Medicine, Kobe University, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Susumu Seino
- Division of Molecular and Metabolic Medicine, Graduate School of Medicine, Kobe University, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| |
Collapse
|
9
|
Aggarwal R, Peng Z, Zeng N, Silva J, He L, Chen J, Debebe A, Tu T, Alba M, Chen CY, Stiles EX, Hong H, Stiles BL. Chronic Exposure to Palmitic Acid Down-Regulates AKT in Beta-Cells through Activation of mTOR. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:130-145. [PMID: 34619135 PMCID: PMC8759041 DOI: 10.1016/j.ajpath.2021.09.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/09/2021] [Accepted: 09/22/2021] [Indexed: 01/03/2023]
Abstract
High circulating lipids occurring in obese individuals and insulin-resistant patients are considered a contributing factor to type 2 diabetes. Exposure to high lipid concentration is proposed to both protect and damage beta-cells under different circumstances. Here, by feeding mice a high-fat diet (HFD) for 2 weeks to up to 14 months, the study showed that HFD initially causes the beta-cells to expand in population, whereas long-term exposure to HFD is associated with failure of beta-cells and the inability of animals to respond to glucose challenge. To prevent the failure of beta-cells and the development of type 2 diabetes, the molecular mechanisms that underlie this biphasic response of beta-cells to lipid exposure were explored. Using palmitic acid (PA) in cultured beta-cells and islets, the study demonstrated that chronic exposure to lipids leads to reduced viability and inhibition of cell cycle progression concurrent with down-regulation of a pro-growth/survival kinase AKT, independent of glucose. This AKT down-regulation by PA is correlated with the induction of mTOR/S6K activity. Inhibiting mTOR activity with rapamycin induced Raptor and restored AKT activity, allowing beta-cells to gain proliferation capacity that was lost after HFD exposure. In summary, a novel mechanism in which lipid exposure may cause the dipole effects on beta-cell growth was elucidated, where mTOR acts as a lipid sensor. These mechanisms can be novel targets for future therapeutic developments.
Collapse
Affiliation(s)
- Richa Aggarwal
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Zhechu Peng
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Ni Zeng
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Joshua Silva
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Lina He
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Jingyu Chen
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Anketse Debebe
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Taojian Tu
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Mario Alba
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Chien-Yu Chen
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Eileen X. Stiles
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Handan Hong
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Bangyan L. Stiles
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California,Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California,Address correspondence to Bangyan L. Stiles, Ph.D., Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033.
| |
Collapse
|
10
|
Imai Y, Cousins RS, Liu S, Phelps BM, Promes JA. Connecting pancreatic islet lipid metabolism with insulin secretion and the development of type 2 diabetes. Ann N Y Acad Sci 2019; 1461:53-72. [PMID: 30937918 DOI: 10.1111/nyas.14037] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/24/2019] [Accepted: 01/30/2019] [Indexed: 02/06/2023]
Abstract
Obesity is the major contributing factor for the increased prevalence of type 2 diabetes (T2D) in recent years. Sustained positive influx of lipids is considered to be a precipitating factor for beta cell dysfunction and serves as a connection between obesity and T2D. Importantly, fatty acids (FA), a key building block of lipids, are a double-edged sword for beta cells. FA acutely increase glucose-stimulated insulin secretion through cell-surface receptor and intracellular pathways. However, chronic exposure to FA, combined with elevated glucose, impair the viability and function of beta cells in vitro and in animal models of obesity (glucolipotoxicity), providing an experimental basis for the propensity of beta cell demise under obesity in humans. To better understand the two-sided relationship between lipids and beta cells, we present a current view of acute and chronic handling of lipids by beta cells and implications for beta cell function and health. We also discuss an emerging role for lipid droplets (LD) in the dynamic regulation of lipid metabolism in beta cells and insulin secretion, along with a potential role for LD under nutritional stress in beta cells, and incorporate recent advancement in the field of lipid droplet biology.
Collapse
Affiliation(s)
- Yumi Imai
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa.,Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa
| | - Ryan S Cousins
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia
| | - Siming Liu
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa.,Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa
| | - Brian M Phelps
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia
| | - Joseph A Promes
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa.,Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa
| |
Collapse
|
11
|
Popp D, Aertsen S, Luetke-Daldrup C, Coppenrath E, Hetterich H, Saam T, Rottenkolber M, Seissler J, Lechner A, Sommer NN. No Correlation of Pancreatic Fat and β-Cell Function in Young Women With and Without a History of Gestational Diabetes. J Clin Endocrinol Metab 2018; 103:3260-3266. [PMID: 29947782 DOI: 10.1210/jc.2018-00187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 06/21/2018] [Indexed: 01/11/2023]
Abstract
CONTEXT Pancreatic steatosis may contribute to β-cell dysfunction in type 2 diabetes (T2D), but data are controversial. Women who had gestational diabetes mellitus (GDM) are at high risk for developing T2D. OBJECTIVE To examine the association of pancreatic fat content with early/first-phase insulin secretion (as markers of β-cell function). DESIGN Cross-sectional analysis of a subcohort of the monocentric, prospective cohort study titled Prediction, Prevention, and Subclassification of Type 2 Diabetes. SETTING Ludwig Maximilians University Hospital, Munich, Germany. PARTICIPANTS Ninety-seven women, 3 to 16 months after pregnancy [41 normoglycemic women post-GDM, 19 women post-GDM with pathological glucose metabolism, and 37 normoglycemic women after a normoglycemic pregnancy (controls)]. MAIN OUTCOME MEASURES Correlation of MRI-measured pancreatic fat content with early insulin release in an oral glucose tolerance test (OGGT) [insulin increment within the first 30 minutes of the OGTT (IR30)] and first-phase insulin response (FPIR) in an intravenous glucose tolerance test (n = 65), both adjusted for insulin sensitivity index (ISI). RESULTS Pancreatic fat content did not correlate with IR30 and FPIR adjusted for ISI. It correlated positively with body mass index, waist circumference, liver fat, and intraabdominal fat volume. CONCLUSION Pancreatic fat content does not correlate with β-cell function in a cohort of young women with different degrees of T2D risk.
Collapse
Affiliation(s)
- Daniel Popp
- Klinik und Poliklinik für Radiologie, Ludwig Maximilians University Hospital, Munich, Germany
| | - Stephanie Aertsen
- Diabetes Research Group, Medizinische Klinik und Poliklinik IV, Ludwig Maximilians University Hospital, Munich, Germany
- Clinical Cooperation Group Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, Germany
- Deutsches Zentrum für Diabetesforschung, Neuherberg, Germany
| | | | - Eva Coppenrath
- Klinik und Poliklinik für Radiologie, Ludwig Maximilians University Hospital, Munich, Germany
| | - Holger Hetterich
- Klinik und Poliklinik für Radiologie, Ludwig Maximilians University Hospital, Munich, Germany
| | - Tobias Saam
- Klinik und Poliklinik für Radiologie, Ludwig Maximilians University Hospital, Munich, Germany
| | - Marietta Rottenkolber
- Diabetes Research Group, Medizinische Klinik und Poliklinik IV, Ludwig Maximilians University Hospital, Munich, Germany
- Clinical Cooperation Group Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, Germany
- Deutsches Zentrum für Diabetesforschung, Neuherberg, Germany
| | - Jochen Seissler
- Diabetes Research Group, Medizinische Klinik und Poliklinik IV, Ludwig Maximilians University Hospital, Munich, Germany
- Clinical Cooperation Group Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, Germany
- Deutsches Zentrum für Diabetesforschung, Neuherberg, Germany
| | - Andreas Lechner
- Diabetes Research Group, Medizinische Klinik und Poliklinik IV, Ludwig Maximilians University Hospital, Munich, Germany
- Clinical Cooperation Group Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, Germany
- Deutsches Zentrum für Diabetesforschung, Neuherberg, Germany
| | - Nora N Sommer
- Klinik und Poliklinik für Radiologie, Ludwig Maximilians University Hospital, Munich, Germany
| |
Collapse
|
12
|
Chakraborty TR, Gomez V, Adhikari D, Chakraborty S. The Synergism in Hormonal and Cellular Changes in Male Mice on Long Term High Fat Exposure. J Am Coll Nutr 2018. [PMID: 29533142 DOI: 10.1080/07315724.2017.1412276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
OBJECTIVE To determine the hormonal changes that occur as a result of the long-term intake of a very-high-fat diet (VHFD) that leads to simultaneous changes in the islets of Langerhans and adipocyte cell size. METHODS Male mice were fed with a normal chow diet (ND, n = 15) and a VHFD (n = 30) for 2, 12, and 24 weeks. Body weight, food intake, caloric intake (fat [saturated and unsaturated], protein, and carbohydrate), hormone levels (leptin and insulin), and islet of Langerhans/adipocyte size were quantitatively recorded. RESULTS In VHFD-fed animals, body weight showed a significant percent increase within the first 12 weeks and then plateaued with time. VHFD-fed animals consumed significantly less food than ND at all time periods, indicating that it was the quality of food and not the quantity that caused this increase in body weight. Male mice on VHFD showed a significant increase in leptin and insulin levels, along with accompanying growth in islet and adipocyte size within the first 12 weeks, which plateaued as the mice aged. The increases in the islet and adipocyte size in VHFD-fed animals were similar to the analogous increases in hormonal levels (2 vs. 12 vs. 24 weeks). These results, therefore, suggest that in diet-induced obesity changes, shifts in hormonal levels works hand-in-hand with metabolic adjustments at the cellular level to combat the effect of fat. CONCLUSION Thus, mechanisms like hormonal resistance, changes in adiposity, islet size, and caloric intake with prolonged exposure to high fat are probably defensive mechanisms employed to protect against diabetes. In order to understand these complicated and nuanced effects of high fat and to comprehend the underlying mechanism associated with it, it is important to focus on long-term studies that emphasize the synergy between cellular and hormonal changes, in addition to an analysis of individual components.
Collapse
Affiliation(s)
- Tandra R Chakraborty
- a Department of Biology , Adelphi University , Garden City, New York , New York , USA
| | - Victoria Gomez
- a Department of Biology , Adelphi University , Garden City, New York , New York , USA
| | | | - Sanjoy Chakraborty
- c Department of Biological Sciences , New York City College of Technology / CUNY , New York , New York , USA
| |
Collapse
|
13
|
Abstract
OBJECTIVE Glucosamine (GlcN), which has been reported to induce insulin resistance (IR), is a popular nutritional supplement used to treat osteoarthritis in menopausal women. We previously demonstrated that GlcN treatment caused IR in ovariectomized rats by reducing the expression of glucose transport protein subtype 4 (GLUT-4) in skeletal muscle. In the present study, we hypothesized that endurance exercise training can reverse GlcN-induced IR. METHODS Fifty female rats were randomly divided into five groups with 10 rats in each group: (1) sham-operated group; (2) sham-operated group with GlcN treatment for 14 days; (3) ovariectomy (OVX) group; (4) OVX with GlcN treatment; and (5) OVX with GlcN treatment followed by exercise training (running program) for 8 weeks. RESULTS Fasting plasma glucose increased in the OVX + GlcN group, and fasting plasma insulin and the homeostasis model assessment-insulin resistance (HOMA-IR) were significantly higher only in this group. After the rats received exercise training for 8 weeks, no increase in the fasting plasma glucose, insulin, or HOMA-IR was observed. In an intraperitoneal glucose tolerance test, the plasma glucose, plasma insulin, HOMA-IR, and glucose-insulin index were significantly elevated only in the OVX with GlcN treatment group. However, the plasma glucose, plasma insulin, HOMA-IR, and glucose-insulin index decreased after exercise training for 8 weeks, implying that GlcN-induced IR in OVX rats could be reversed through exercise. A histological analysis revealed that exercise training can reduce islet hypertrophy and maintain GLUT-4 in skeletal muscle. CONCLUSIONS Exercise training can alleviate IR in OVX rats treated with GlcN. Islet hyperplasia was subsequently prevented. Preserving GLUT-4 expression may be one of the mechanisms by which exercise prevents IR.
Collapse
|
14
|
Petry SF, Sun LM, Knapp A, Reinl S, Linn T. Distinct Shift in Beta-Cell Glutaredoxin 5 Expression Is Mediated by Hypoxia and Lipotoxicity Both In Vivo and In Vitro. Front Endocrinol (Lausanne) 2018; 9:84. [PMID: 29593651 PMCID: PMC5857561 DOI: 10.3389/fendo.2018.00084] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 02/22/2018] [Indexed: 01/04/2023] Open
Abstract
Histomorphological and functional alterations in pancreatic islet composition directly correlate with hyperglycemia severity. Progressive deterioration of metabolic control in subjects suffering from type 2 diabetes is predominantly caused by impaired beta-cell functionality. The glutaredoxin system is supposed to wield protective properties for beta-cells. Therefore, we sought to identify a correlation between the structural changes observed in diabetic pancreatic islets with altered glutaredoxin 5 expression, in order to determine an underlying mechanism of beta-cell impairment. Islets of db/db mice presenting with uncontrolled diabetes were assessed in terms of morphological structure and insulin, glucagon, and glutaredoxin 5 expression. MIN6 cell function and glutaredoxin 5 expression were analyzed after exposure to oleic acid and hypoxia. Islets of diabese mice were marked by typical remodeling and distinct reduction of, and shifts, in localization of glutaredoxin 5-positive cells. These islets featured decreased glutaredoxin 5 as well as insulin and glucagon content. In beta-cell culture, glutaredoxin 5 protein and mRNA expression were decreased by hypoxia and oleic acid but not by leptin treatment. Our study demonstrates that glutaredoxin 5 expression patterns are distinctively altered in islets of rodents presenting with uncontrolled diabesity. In vitro, reduction of islet-cell glutaredoxin 5 expression was mediated by hypoxia and oleic acid. Thus, glutaredoxin 5-deficiency in islets during diabetes may be caused by lipotoxicity and hypoxia.
Collapse
Affiliation(s)
- Sebastian Friedrich Petry
- Clinical Research Unit, Center of Internal Medicine, Justus Liebig University, Giessen, Germany
- *Correspondence: Sebastian Friedrich Petry,
| | - Lia Mingzhe Sun
- Clinical Research Unit, Center of Internal Medicine, Justus Liebig University, Giessen, Germany
| | - Anna Knapp
- Clinical Research Unit, Center of Internal Medicine, Justus Liebig University, Giessen, Germany
| | - Sabrina Reinl
- Clinical Research Unit, Center of Internal Medicine, Justus Liebig University, Giessen, Germany
| | - Thomas Linn
- Clinical Research Unit, Center of Internal Medicine, Justus Liebig University, Giessen, Germany
| |
Collapse
|
15
|
Sampson M, Lathen DR, Dallon BW, Draney C, Ray JD, Kener KB, Parker BA, Gibbs JL, Gropp JS, Tessem JS, Bikman BT. β-Hydroxybutyrate improves β-cell mitochondrial function and survival. JOURNAL OF INSULIN RESISTANCE 2017. [DOI: 10.4102/jir.v2i1.25] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Pharmacological interventions aimed at improving outcomes in type 2 diabetes and achieving normoglycaemia, including insulin therapy, are increasingly common, despite the potential for substantial side effects. Carbohydrate-restricted diets that result in increased ketogenesis have effectively been used to improve insulin resistance, a fundamental feature of type 2 diabetes. In addition, limited evidence suggests that states of ketogenesis may also improve β-cell function in type 2 diabetics. Considering how little is known regarding the effects of ketones on β-cell function, we sought to determine the specific effects of β-Hydroxybutyrate (βHB) on pancreatic β-cell physiology and mitochondrial function. βHB treatment increased β-cell survival and proliferation, while also increasing mitochondrial mass, respiration and adenosine triphosphate (ATP) production. Despite these improvements, were unable to detect an increase in β-cell or islet insulin production and secretion. Collectively, these findings have two implications. Firstly, they indicate that β-cells have improved survival and proliferation in the midst of βHB, the circulating form of ketones. Secondly, insulin secretion does not appear to be directly related to apparent improvements in mitochondrial function and cellular proliferation.
Collapse
|
16
|
Seo D, Faintuch BL, Aparecida de Oliveira E, Faintuch J. Pancreas and liver uptake of new radiolabeled incretins (GLP-1 and Exendin-4) in models of diet-induced and diet-restricted obesity. Nucl Med Biol 2017; 49:57-64. [DOI: 10.1016/j.nucmedbio.2017.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 02/02/2017] [Accepted: 03/15/2017] [Indexed: 01/19/2023]
|
17
|
Choi JS, Kim JW, Park JB, Pyo SE, Hong YK, Ku SK, Kim MR. Blood glycemia-modulating effects of melanian snail protein hydrolysates in mice with type II diabetes. Int J Mol Med 2017; 39:1437-1451. [PMID: 28487991 PMCID: PMC5428967 DOI: 10.3892/ijmm.2017.2967] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 03/29/2017] [Indexed: 12/23/2022] Open
Abstract
Freshwater animal proteins have long been used as nutrient supplements. In this study, melanian snail (Semisulcospira libertina) protein hydrolysates (MPh) were found to exert anti-diabetic and protective effects against liver and kidney damage in mice with type II diabetes adapted to a 45% kcal high-fat diet (HFD). The hypoglycemic, hepatoprotective and nephroprotective effects of MPh were analyzed after 12 weeks of the continuous oral administration of MPh at 125, 250 and 500 mg/kg. Diabetic control mice exhibited an increase in body weight, and blood glucose and insulin levels, with a decrease in serum high-density lipoprotein (HDL) levels. In addition, an increase in the regions of steatohepatitis, hepatocyte hypertrophy, and lipid droplet deposit-related renal tubular vacuolation degenerative lesions were detected, with noticeable expansion and hyperplasia of the pancreatic islets, and an increase in glucagon- and insulin-producing cells, insulin/glucagon cell ratios in the endocrine pancreas and hepatic lipid peroxidation, as well as decreased zymogen contents. Furthermore, a deterioration of the endogenous antioxidant defense system was observed, with reduced glucose utilization related hepatic glucokinase (GK) activity and an increase in hepatic gluconeogenesis-related phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphatase (G6pase) activity. However, all of these diabetic complications were significantly inhibited by oral treatment with MPh in a dose-dependent manner. In addition, the marked dose-dependent inhibition of hepatic lipid peroxidation, the depletion of the liver endogenous antioxidant defense system, and changes in hepatic glucose-regulating enzyme activities were also observed. The results of this study suggest that MPh exerts potent anti-diabetic effects, along with the amelioration of related complications in mice with type II diabetes. The overall effects of MPh at a dose of 125 mg/kg on HFD-induced diabetes and related complications were similar or more potent than those of metformin (250 mg/kg).
Collapse
Affiliation(s)
- Jae-Suk Choi
- Major in Food Biotechnology, Division of Bioindustry, College of Medical and Life Sciences, Silla University, Sasang-gu, Busan 46958, Republic of Korea
| | - Joo-Wan Kim
- Aribio Inc., Byeoksan Digital Valley, Yeongdeungpo-gu, Seoul 07286, Republic of Korea
| | - Jeong Been Park
- Major in Food Biotechnology, Division of Bioindustry, College of Medical and Life Sciences, Silla University, Sasang-gu, Busan 46958, Republic of Korea
| | - Sang Eun Pyo
- Major in Food Biotechnology, Division of Bioindustry, College of Medical and Life Sciences, Silla University, Sasang-gu, Busan 46958, Republic of Korea
| | - Yong-Ki Hong
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Nam-Gu, Busan 48513, Republic of Korea
| | - Sae Kwang Ku
- Department of Anatomy and Histology, College of Oriental Medicine, Daegu Haany University, Gyeongsan-si, Gyeongsangbuk-do 38610, Republic of Korea
| | - Mi-Ryung Kim
- Major in Food Biotechnology, Division of Bioindustry, College of Medical and Life Sciences, Silla University, Sasang-gu, Busan 46958, Republic of Korea
| |
Collapse
|
18
|
Jordaens L, Van Hoeck V, Maillo V, Gutierrez-Adan A, Marei WFA, Vlaeminck B, Thys S, Sturmey RG, Bols PEJ, Leroy JLMR. Maternal metabolic stress may affect oviduct gatekeeper function. Reproduction 2017; 153:759-773. [PMID: 28258156 DOI: 10.1530/rep-16-0569] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 02/23/2017] [Accepted: 03/03/2017] [Indexed: 01/01/2023]
Abstract
We hypothesized that elevated non-esterified fatty acids (NEFA) modify in vitro bovine oviduct epithelial cell (BOEC) metabolism and barrier function. Hereto, BOECs were studied in a polarized system with 24-h treatments at Day 9: (1) control (0 µM NEFA + 0% EtOH), (2) solvent control (0 µM NEFA + 0.45% EtOH), (3) basal NEFA (720 µM NEFA + 0.45% EtOH in the basal compartment) and (4) apical NEFA (720 µM NEFA + 0.45% EtOH in the apical compartment). FITC-albumin was used for monolayer permeability assessment and related to transepithelial electric resistance (TER). Fatty acid (FA), glucose, lactate and pyruvate concentrations were measured in spent medium. Intracellular lipid droplets (LD) and FA uptake were studied using Bodipy 493/503 and immunolabelling of FA transporters (FAT/CD36, FABP3 and CAV1). BOEC-mRNA was retrieved for qRT-PCR. Results revealed that apical NEFA reduced relative TER increase (46.85%) during treatment and increased FITC-albumin flux (27.59%) compared to other treatments. In basal NEFA, FAs were transferred to the apical compartment as free FAs: mostly palmitic and oleic acid increased respectively 56.0 and 33.5% of initial FA concentrations. Apical NEFA allowed no FA transfer, but induced LD accumulation and upregulated FA transporter expression (↑CD36, ↑FABP3 and ↑CAV1). Gene expression in apical NEFA indicated increased anti-apoptotic (↑BCL2) and anti-oxidative (↑SOD1) capacity, upregulated lipid metabolism (↑CPT1, ↑ACSL1 and ↓ACACA) and FA uptake (↑CAV1). All treatments had similar carbohydrate metabolism and oviduct function-specific gene expression (OVGP1, ESR1 and FOXJ1). Overall, elevated NEFAs affected BOEC metabolism and barrier function differently depending on NEFA exposure side. Data substantiate the concept of the oviduct as a gatekeeper that may actively alter early embryonic developmental conditions.
Collapse
Affiliation(s)
- L Jordaens
- Laboratory for Veterinary Physiology and BiochemistryGamete Research Center, University of Antwerp, Wilrijk, Belgium
| | - V Van Hoeck
- Laboratory for Veterinary Physiology and BiochemistryGamete Research Center, University of Antwerp, Wilrijk, Belgium
| | - V Maillo
- INIAInstituto Nacional de Investigacion y Tecnologia Agraria y Alimentaria, Madrid, Spain
| | - A Gutierrez-Adan
- INIAInstituto Nacional de Investigacion y Tecnologia Agraria y Alimentaria, Madrid, Spain
| | - W F A Marei
- Laboratory for Veterinary Physiology and BiochemistryGamete Research Center, University of Antwerp, Wilrijk, Belgium.,Department of TheriogenologyFaculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - B Vlaeminck
- Laboratory for Animal Nutrition and Animal Product QualityGhent University, Ghent, Belgium
| | - S Thys
- Laboratory for Cell Biology and HistologyCore Facility for Biomedical Microscopic Imaging, University of Antwerp, Antwerp, Belgium
| | - R G Sturmey
- Hull York Medical SchoolCenter for Cardiovascular and Metabolic Research, University of Hull, Hull, UK
| | - P E J Bols
- Laboratory for Veterinary Physiology and BiochemistryGamete Research Center, University of Antwerp, Wilrijk, Belgium
| | - J L M R Leroy
- Laboratory for Veterinary Physiology and BiochemistryGamete Research Center, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
19
|
Hayes HL, Peterson BS, Haldeman JM, Newgard CB, Hohmeier HE, Stephens SB. Delayed apoptosis allows islet β-cells to implement an autophagic mechanism to promote cell survival. PLoS One 2017; 12:e0172567. [PMID: 28212395 PMCID: PMC5315295 DOI: 10.1371/journal.pone.0172567] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 01/24/2017] [Indexed: 01/09/2023] Open
Abstract
Increased β-cell death coupled with the inability to replicate existing β-cells drives the decline in β-cell mass observed in the progression of both major forms of diabetes. Understanding endogenous mechanisms of islet cell survival could have considerable value for the development of novel strategies to limit β-cell loss and thereby promote β-cell recovery. Insulinoma cells have provided useful insight into β-cell death pathways but observations made in cell lines sometimes fail to translate to primary islets. Here, we report dramatic differences in the temporal regulation and engagement of the apoptotic program in primary rodent islets relative to the INS-1 derived 832/13 cell line. As expected, 832/13 cells rapidly induced cell stress markers in response to ER stress or DNA damage and were fully committed to apoptosis, resulting in >80% cell death within 24 h. In contrast, primary rat islets were largely refractory to cell death in response to ER stress and DNA damage, despite rapid induction of stress markers, such as XBP-1(s), CHOP, and PUMA. Gene expression profiling revealed a general suppression of pro-apoptotic machinery, such as Apaf-1 and caspase 3, and sustained levels of pro-survival factors, such as cIAP-1, cIAP-2, and XIAP, in rat islets. Furthermore, we observed sustained induction of autophagy following chronic ER stress and found that inhibition of autophagy rendered islet β-cells highly vulnerable to ER stress-induced cell death. We propose that islet β-cells dampen the apoptotic response to delay the onset of cell death, providing a temporal window in which autophagy can be activated to limit cellular damage and promote survival.
Collapse
Affiliation(s)
- Heather L. Hayes
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Brett S. Peterson
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina, United States of America
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Jonathan M. Haldeman
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina, United States of America
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Christopher B. Newgard
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina, United States of America
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Medicine, Division of Endocrinology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Hans E. Hohmeier
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina, United States of America
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Medicine, Division of Endocrinology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Samuel B. Stephens
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina, United States of America
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
20
|
Du T, Lu S, Jiang Q, Li Y, Ma K. Quantitative Proteomic Analysis of Hepatic Tissue of T2DM Rhesus Macaque. J Diabetes Res 2017; 2017:3601708. [PMID: 29404372 PMCID: PMC5748286 DOI: 10.1155/2017/3601708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/09/2017] [Indexed: 12/19/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic disorder that severely affects human health, but the pathogenesis of the disease remains unknown. The high-fat/high-sucrose diets combined with streptozotocin- (STZ-) induced nonhuman primate animal model of diabetes are a valuable research source of T2DM. Here, we present a study of a STZ rhesus macaque model of T2DM that utilizes quantitative iTRAQ-based proteomic method. We compared the protein profiles in the liver of STZ-treated macaques as well as age-matched healthy controls. We identified 171 proteins differentially expressed in the STZ-treated groups, about 70 of which were documented as diabetes-related gene in previous studies. Pathway analyses indicated that the biological functions of differentially expressed proteins were related to glycolysis/gluconeogenesis, fatty acid metabolism, complements, and coagulation cascades. Expression change in tryptophan metabolism pathway was also found in this study which may be associations with diabetes. This study is the first to explore genome-wide protein expression in hepatic tissue of diabetes macaque model using HPLC-Q-TOF/MS technology. In addition to providing potential T2DM biomarkers, this quantitative proteomic study may also shed insights regarding the molecular pathogenesis of T2DM.
Collapse
Affiliation(s)
- Tingfu Du
- Center for Drug Safety Evaluation and Research, Institute of Medical Biology, Chinese Academy of Medical Sciences, Kunming 650118, China
- Medical Primate Research Center & Neuroscience Center, Chinese Academy of Medical Sciences, Beijing 100005, China
- Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Diseases, Kunming 650118, China
| | - Shuaiyao Lu
- Center for Drug Safety Evaluation and Research, Institute of Medical Biology, Chinese Academy of Medical Sciences, Kunming 650118, China
- Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Diseases, Kunming 650118, China
| | - Qinfang Jiang
- Center for Drug Safety Evaluation and Research, Institute of Medical Biology, Chinese Academy of Medical Sciences, Kunming 650118, China
- Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Diseases, Kunming 650118, China
| | - Yun Li
- Center for Drug Safety Evaluation and Research, Institute of Medical Biology, Chinese Academy of Medical Sciences, Kunming 650118, China
- Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Diseases, Kunming 650118, China
| | - Kaili Ma
- Center for Drug Safety Evaluation and Research, Institute of Medical Biology, Chinese Academy of Medical Sciences, Kunming 650118, China
- Medical Primate Research Center & Neuroscience Center, Chinese Academy of Medical Sciences, Beijing 100005, China
- Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Diseases, Kunming 650118, China
| |
Collapse
|
21
|
A Pdx-1-Regulated Soluble Factor Activates Rat and Human Islet Cell Proliferation. Mol Cell Biol 2016; 36:2918-2930. [PMID: 27620967 DOI: 10.1128/mcb.00103-16] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 09/02/2016] [Indexed: 12/23/2022] Open
Abstract
The homeodomain transcription factor Pdx-1 has important roles in pancreas and islet development as well as in β-cell function and survival. We previously reported that Pdx-1 overexpression stimulates islet cell proliferation, but the mechanism remains unclear. Here, we demonstrate that overexpression of Pdx-1 triggers proliferation largely by a non-cell-autonomous mechanism mediated by soluble factors. Consistent with this idea, overexpression of Pdx-1 under the control of a β-cell-specific promoter (rat insulin promoter [RIP]) stimulates proliferation of both α and β cells, and overexpression of Pdx-1 in islets separated by a Transwell membrane from islets lacking Pdx-1 overexpression activates proliferation in the untreated islets. Microarray and gene ontology (GO) analysis identified inhibin beta-B (Inhbb), an activin subunit and member of the transforming growth factor β (TGF-β) superfamily, as a Pdx-1-responsive gene. Overexpression of Inhbb or addition of activin B stimulates rat islet cell and β-cell proliferation, and the activin receptors RIIA and RIIB are required for the full proliferative effects of Pdx-1 in rat islets. In human islets, Inhbb overexpression stimulates total islet cell proliferation and potentiates Pdx-1-stimulated proliferation of total islet cells and β cells. In sum, this study identifies a mechanism by which Pdx-1 induces a soluble factor that is sufficient to stimulate both rat and human islet cell proliferation.
Collapse
|
22
|
Wortham M, Sander M. Mechanisms of β-cell functional adaptation to changes in workload. Diabetes Obes Metab 2016; 18 Suppl 1:78-86. [PMID: 27615135 PMCID: PMC5021190 DOI: 10.1111/dom.12729] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 05/09/2016] [Indexed: 11/28/2022]
Abstract
Insulin secretion must be tightly coupled to nutritional state to maintain blood glucose homeostasis. To this end, pancreatic β-cells sense and respond to changes in metabolic conditions, thereby anticipating insulin demands for a given physiological context. This is achieved in part through adjustments of nutrient metabolism, which is controlled at several levels including allosteric regulation, post-translational modifications, and altered expression of metabolic enzymes. In this review, we discuss mechanisms of β-cell metabolic and functional adaptation in the context of two physiological states that alter glucose-stimulated insulin secretion: fasting and insulin resistance. We review current knowledge of metabolic changes that occur in the β-cell during adaptation and specifically discuss transcriptional mechanisms that underlie β-cell adaptation. A more comprehensive understanding of how β-cells adapt to changes in nutrient state could identify mechanisms to be co-opted for therapeutically modulating insulin secretion in metabolic disease.
Collapse
Affiliation(s)
- M Wortham
- Departments of Pediatrics and Cellular and Molecular Medicine, Pediatric Diabetes Research Center, University of California San Diego, La Jolla
| | - M Sander
- Departments of Pediatrics and Cellular and Molecular Medicine, Pediatric Diabetes Research Center, University of California San Diego, La Jolla.
| |
Collapse
|
23
|
Mutua MP, Muya S, Gicheru MM. Protective roles of free avian respiratory macrophages in captive birds. Biol Res 2016; 49:29. [PMID: 27306902 PMCID: PMC4910195 DOI: 10.1186/s40659-016-0090-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 06/09/2016] [Indexed: 11/10/2022] Open
Abstract
In the mammalian lung, respiratory macrophages provide front line defense against invading pathogens and particulate matter. In birds, respiratory macrophages are known as free avian respiratory macrophages (FARM) and a dearth of the cells in the avian lung has been purported to foreordain a weak first line of pulmonary defense, a condition associated with high mortality of domestic birds occasioned by respiratory inflictions. Avian pulmonary mechanisms including a three tiered aerodynamic filtration system, tight epithelial junctions and an efficient mucociliary escalator system have been known to supplement FARM protective roles. Current studies, however, report FARM to exhibit an exceptionally efficient phagocytic capacity and are effective in elimination of invading pathogens. In this review, we also report on effects of selective synthetic peroxisome proliferator activated receptor gamma (PPAR γ) agonists on non phlogistic phagocytic properties in the FARM. To develop effective therapeutic interventions targeting FARM in treatment and management of respiratory disease conditions in the poultry, further studies are required to fully understand the role of FARM in innate and adaptive immune responses.
Collapse
Affiliation(s)
- Mbuvi P Mutua
- Department of Zoological Sciences, Kenyatta University, P.O. Box 43844-00100, Nairobi, Kenya.
| | - Shadrack Muya
- Department of Zoology, Jomo Kenyatta University of Agriculture and Technology, P.O. Box 62000-00200, Nairobi, Kenya
| | - Muita M Gicheru
- Department of Zoological Sciences, Kenyatta University, P.O. Box 43844-00100, Nairobi, Kenya
| |
Collapse
|
24
|
Draney C, Hobson AE, Grover SG, Jack BO, Tessem JS. Cdk5r1 Overexpression Induces Primary β-Cell Proliferation. J Diabetes Res 2016; 2016:6375804. [PMID: 26788519 PMCID: PMC4691621 DOI: 10.1155/2016/6375804] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 08/15/2015] [Accepted: 08/18/2015] [Indexed: 02/07/2023] Open
Abstract
Decreased β-cell mass is a hallmark of type 1 and type 2 diabetes. Islet transplantation as a method of diabetes therapy is hampered by the paucity of transplant ready islets. Understanding the pathways controlling islet proliferation may be used to increase functional β-cell mass through transplantation or by enhanced growth of endogenous β-cells. We have shown that the transcription factor Nkx6.1 induces β-cell proliferation by upregulating the orphan nuclear hormone receptors Nr4a1 and Nr4a3. Using expression analysis to define Nkx6.1-independent mechanisms by which Nr4a1 and Nr4a3 induce β-cell proliferation, we demonstrated that cyclin-dependent kinase 5 regulatory subunit 1 (Cdk5r1) is upregulated by Nr4a1 and Nr4a3 but not by Nkx6.1. Overexpression of Cdk5r1 is sufficient to induce primary rat β-cell proliferation while maintaining glucose stimulated insulin secretion. Overexpression of Cdk5r1 in β-cells confers protection against apoptosis induced by etoposide and thapsigargin, but not camptothecin. The Cdk5 kinase complex inhibitor roscovitine blocks islet proliferation, suggesting that Cdk5r1 mediated β-cell proliferation is a kinase dependent event. Overexpression of Cdk5r1 results in pRb phosphorylation, which is inhibited by roscovitine treatment. These data demonstrate that activation of the Cdk5 kinase complex is sufficient to induce β-cell proliferation while maintaining glucose stimulated insulin secretion.
Collapse
Affiliation(s)
- Carrie Draney
- Nutrition, Dietetics and Food Science Department, College of Life Sciences, Brigham Young University, Provo, UT 84602, USA
| | - Amanda E. Hobson
- Nutrition, Dietetics and Food Science Department, College of Life Sciences, Brigham Young University, Provo, UT 84602, USA
| | - Samuel G. Grover
- Nutrition, Dietetics and Food Science Department, College of Life Sciences, Brigham Young University, Provo, UT 84602, USA
| | - Benjamin O. Jack
- Nutrition, Dietetics and Food Science Department, College of Life Sciences, Brigham Young University, Provo, UT 84602, USA
| | - Jeffery S. Tessem
- Nutrition, Dietetics and Food Science Department, College of Life Sciences, Brigham Young University, Provo, UT 84602, USA
- *Jeffery S. Tessem:
| |
Collapse
|
25
|
Mutua MP, Steinaa L, Shadrack MM, Muita GM. Activation of peroxisome proliferator-activated receptor gamma induces anti-inflammatory properties in the chicken free avian respiratory macrophages. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2015; 57:40. [PMID: 26594385 PMCID: PMC4653948 DOI: 10.1186/s40781-015-0073-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 11/15/2015] [Indexed: 11/10/2022]
Abstract
Background Activation of peroxisome proliferator activated receptor gamma (PPAR γ) in the alveolar macrophages (AM) by selective synthetic PPAR γ ligands, improves the ability of the cells to resolve inflammation. In birds, respiratory macrophages are known as free avian respiratory macrophages (FARM) and show distinct functional differences from AM. The effects of treating FARM with PPAR γ ligands are unclear. Methods FARM were harvested by lavage of chicken respiratory tract and their morphology assessed at microscopic level. The effects of PPAR γ agonists on the FARM in vitro viability, phagocytic capacity and proinflammatory cytokine (TNF-α) production were assessed. Results FARM had eccentric nucleus and plasma membrane ruffled with filopodial extensions. Ultrastructurally, numerous vesicular bodies presumed to be lysosomes were present. FARM treated with troglitazone, a selective PPAR γ agonist, had similar in vitro viability with untreated FARM. However, treated FARM co-cultured with polystyrene particles, internalized more particles with a mean volume density of 41 % compared to that of untreated FARM of 21 %. Further, treated FARM significantly decreased LPS-induced TNF-α production in a dose dependent manner. Conclusion Results from this study show that PPAR γ synthetic ligands enhance phagocytic ability of FARM. Further the ligands attenuate production of proinflammatory cytokines in the FARM, suggesting potential therapeutic application of PPAR γ ligands in the management of respiratory inflammatory disorders in the poultry industry.
Collapse
Affiliation(s)
- Mbuvi P Mutua
- Department of Zoological Sciences, Kenyatta University, P.O Box 43844-00100, Nairobi, Kenya
| | - Lucilla Steinaa
- International Livestock Research Institute, P.O Box 30709-00100, Nairobi, Kenya
| | - Muya M Shadrack
- Department of Zoology, Jomo Kenyatta University of Agriculture and Technology, P.O Box 62000-0200, Nairobi, Kenya
| | - Gicheru M Muita
- Department of Zoological Sciences, Kenyatta University, P.O Box 43844-00100, Nairobi, Kenya
| |
Collapse
|
26
|
Fermentation of Green Tea with 2% Aquilariae lignum Increases the Anti-Diabetic Activity of Green Tea Aqueous Extracts in the High Fat-Fed Mouse. Nutrients 2015; 7:9046-78. [PMID: 26540072 PMCID: PMC4663575 DOI: 10.3390/nu7115447] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 09/18/2015] [Accepted: 10/23/2015] [Indexed: 12/21/2022] Open
Abstract
Anti-diabetic effects on the metabolomic differences between green tea (GT) and Aquilariae lignum-fermented green tea (fGT) were investigated in the high fat-fed mouse. To prove the differences, hypoglycemic (blood glucose, insulin and glycated hemoglobin levels, pancreas weights and histopathological-immunohistochemistrical analysis of pancreas–insulin/glucagon cells), hepato- and nephron-protective (the changes in liver and kidney weight, histopathology of liver and kidney, serum aminotransferases (AST and ALT) levels, blood urea nitrogen, and serum creatinine levels), and hypolipidemic (the changes of serum total cholesterol, triglyceride, low- and high-density lipoprotein levels with fecal TC and TG contents) effects were evaluated. In addition, liver lipid peroxidation, the glutathione contents, and catalase and superoxide dismutase activities were measured according to the hepatic glucose-regulating enzyme activities of glucokinase (GK), glucose-6-phosphatase (G6pase) and phosphoenolpyruvate carboxykinase (PEPCK) for action mechanisms. As a result, fGT showed a stronger hypoglycemic, hepato- and nephron-protective, hypolipidemic, and anti-oxidant effect than GT in high fat-fed mice. In addition, fGT-treated mice exerted more favorable inhibitory activities against GK, G6pase, PERCK activities as compared to GT-treated mice. Taken together, fGT fermented with Aquilariae lignum, 1:49 (2%; g/g) has a stronger effect compared with GT. Therefore, fGT has the potential to increase bioactivity against type 2 diabetics.
Collapse
|
27
|
Transcriptional regulation of chemokine genes: a link to pancreatic islet inflammation? Biomolecules 2015; 5:1020-34. [PMID: 26018641 PMCID: PMC4496708 DOI: 10.3390/biom5021020] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 05/12/2015] [Indexed: 12/18/2022] Open
Abstract
Enhanced expression of chemotactic cytokines (aka chemokines) within pancreatic islets likely contributes to islet inflammation by regulating the recruitment and activation of various leukocyte populations, including macrophages, neutrophils, and T-lymphocytes. Because of the powerful actions of these chemokines, precise transcriptional control is required. In this review, we highlight what is known about the signals and mechanisms that govern the transcription of genes encoding specific chemokine proteins in pancreatic islet β-cells, which include contributions from the NF-κB and STAT1 pathways. We further discuss increased chemokine expression in pancreatic islets during autoimmune-mediated and obesity-related development of diabetes.
Collapse
|
28
|
Simental-Mendía LE, Castañeda-Chacón A, Rodriguez-Morán M, Aradillas-García C, Guerrero-Romero F. Relationship between elevated triglyceride levels with the increase of HOMA-IR and HOMA-β in healthy children and adolescents with normal weight. Eur J Pediatr 2015; 174:597-605. [PMID: 25339422 DOI: 10.1007/s00431-014-2426-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 09/10/2014] [Accepted: 09/15/2014] [Indexed: 11/26/2022]
Abstract
UNLABELLED To test the hypothesis that mildly elevated triglyceride levels are associated with the increase of homeostasis model assessment of insulin resistance (HOMA-IR) and β-cell function (HOMA-β) indices in healthy children and adolescents with normal weight, we conducted a cross-sectional population study. Based on fasting triglyceride levels, participants were allocated into groups with and without triglyceride levels ≥1.2 mmol/L. Normal weight was defined by body mass index between the 15th and 85th percentiles, for age and gender. Insulin resistance and insulin secretion were estimated using HOMA-IR and HOMA-β indices. A total of 1660 children and adolescents were enrolled, of them 327 (19.7%) with mildly elevated triglycerides. The multivariate linear regression analysis showed that mildly elevated triglyceride levels in children were associated with HOMA-IR (β = 0.214, p < 0.001), HOMA-β (β = 0.139, p = 0.001), systolic (β = 0.094, p = 0.01), and diastolic blood pressure (β = 0.102, p = 0.007), whereas in adolescents, HOMA-IR (β = 0.267, p < 0.001) and HOMA-β (β = 0.154, p < 0.001), but not systolic (β = 0.029, p = 0.38) and diastolic blood pressure (β = 0.015, p = 0.642), showed association with mildly elevated triglycerides. CONCLUSION Mildly elevated triglyceride levels are associated with increased HOMA-IR and HOMA-β indices in healthy children and adolescents with normal weight.
Collapse
Affiliation(s)
- Luis E Simental-Mendía
- Biomedical Research Unit of the Mexican Social Security Institute, Siqueiros 225 esq/Castañeda, 34000, Durango, DGO, Mexico,
| | | | | | | | | |
Collapse
|
29
|
Garcia Barrado MJ, Iglesias Osma MC, Blanco EJ, Carretero Hernández M, Sánchez Robledo V, Catalano Iniesta L, Carrero S, Carretero J. Dopamine modulates insulin release and is involved in the survival of rat pancreatic beta cells. PLoS One 2015; 10:e0123197. [PMID: 25886074 PMCID: PMC4401745 DOI: 10.1371/journal.pone.0123197] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 02/18/2015] [Indexed: 12/30/2022] Open
Abstract
The local synthesis of dopamine and its effects on insulin release have been described in isolated islets. Thus, it may be accepted that dopamine exerts an auto-paracrine regulation of insulin secretion from pancreatic beta cells. The aim of the present study is to analyze whether dopamine is a regulator of the proliferation and apoptosis of rat pancreatic beta cells after glucose-stimulated insulin secretion. Glucose stimulated pancreatic islets obtained from male Wistar rats were cultured with 1 or 10 μM dopamine from 1 to 12 h. Insulin secretion was analyzed by RIA. The cellular proliferation rate of pancreatic islets and beta cells was studied with immunocytochemical double labelling for both insulin and PCNA (proliferating cell nuclear antigen), and active caspase-3 was detected to evaluate apoptosis. The secretion of insulin from isolated islets was significantly inhibited (p<0.01), by treatment with 1 and 10 μM dopamine, with no differences between either dose as early as 1 h after treatment. The percentage of insulin-positive cells in the islets decreased significantly (p<0.01) after 1 h of treatment up to 12 h. The proliferation rate of insulin-positive cells in the islets decreased significantly (p<0.01) following treatment with dopamine. Apoptosis in pancreatic islets and beta cells was increased by treatment with 1 and 10 μM dopamine along 12 h. In conclusion, these results suggest that dopamine could modulate the proliferation and apoptosis of pancreatic beta cells and that dopamine may be involved in the maintenance of pancreatic islets.
Collapse
Affiliation(s)
- Maria Jose Garcia Barrado
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Salamanca, Salamanca, Spain
- Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León, and Laboratory of Neuroendocrinology and Obesity of IBSAL, University of Salamanca, Salamanca, Spain
| | - Maria Carmen Iglesias Osma
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Salamanca, Salamanca, Spain
- Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León, and Laboratory of Neuroendocrinology and Obesity of IBSAL, University of Salamanca, Salamanca, Spain
| | - Enrique J. Blanco
- Department of Human Anatomy and Histology, Faculty of Medicine, University of Salamanca, Salamanca, Spain
- Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León, and Laboratory of Neuroendocrinology and Obesity of IBSAL, University of Salamanca, Salamanca, Spain
| | - Marta Carretero Hernández
- Department of Human Anatomy and Histology, Faculty of Medicine, University of Salamanca, Salamanca, Spain
| | - Virginia Sánchez Robledo
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Salamanca, Salamanca, Spain
| | - Leonardo Catalano Iniesta
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Salamanca, Salamanca, Spain
| | - Sixto Carrero
- Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León, and Laboratory of Neuroendocrinology and Obesity of IBSAL, University of Salamanca, Salamanca, Spain
| | - Jose Carretero
- Department of Human Anatomy and Histology, Faculty of Medicine, University of Salamanca, Salamanca, Spain
- Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León, and Laboratory of Neuroendocrinology and Obesity of IBSAL, University of Salamanca, Salamanca, Spain
- * E-mail:
| |
Collapse
|
30
|
Hobson A, Draney C, Stratford A, Becker TC, Lu D, Arlotto M, Tessem JS. Aurora Kinase A is critical for the Nkx6.1 mediated β-cell proliferation pathway. Islets 2015; 7:e1027854. [PMID: 26030060 PMCID: PMC4588548 DOI: 10.1080/19382014.2015.1027854] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Type 1 and type 2 diabetes are ultimately characterized by depleted β-cell mass. Characterization of the molecular pathways that control β-cell proliferation could be harnessed to restore these cells. The homeobox β-cell transcription factor Nkx6.1 induces β-cell proliferation by activating the orphan nuclear receptors Nr4a1 and Nr4a3. Here, we demonstrate that Nkx6.1 localizes to the promoter of the mitotic kinase AURKA (Aurora Kinase A) and induces its expression. Adenovirus mediated overexpression of AURKA is sufficient to induce proliferation in primary rat islets while maintaining glucose stimulated insulin secretion. Furthermore, AURKA is necessary for Nkx6.1 mediated β-cell proliferation as demonstrated by shRNA mediated knock down and pharmacological inhibition of AURKA kinase activity. AURKA preferentially induces DNA replication in β-cells as measured by BrdU incorporation, and enhances the rate of histone H3 phosphorylation in primary β-cells, demonstrating that AURKA induces the replicative and mitotic cell cycle phases in rat β-cells. Finally, overexpression of AURKA results in phosphorylation of the cell cycle regulator p53, which targets p53 for degradation and permits cell cycle progression. These studies define a pathway by which AURKA upregulation by Nkx6.1 results in phosphorylation and degradation of p53, thus removing a key inhibitory factor and permitting engagement of the β-cell proliferation pathway.
Collapse
Key Words
- AURKA
- AURKA, Aurora Kinase A
- BrdU, bromodeoxyuridine
- ChIP, chromatin immunoprecipitation
- Nkx6.1
- Nkx6.1, NK Homeobox 1
- Nr4a1, Nuclear receptor subfamily 4, group A, member 1
- Nr4a3, Nuclear receptor subfamily 4, group A, member 3
- cell cycle
- islet
- p53
- proliferation
Collapse
Affiliation(s)
- Amanda Hobson
- Nutrition; Dietetics and Food Science Department; College of Life Sciences; Brigham Young University, Provo, Utah USA
| | - Carrie Draney
- Nutrition; Dietetics and Food Science Department; College of Life Sciences; Brigham Young University, Provo, Utah USA
| | - Andrew Stratford
- Nutrition; Dietetics and Food Science Department; College of Life Sciences; Brigham Young University, Provo, Utah USA
| | - Thomas C Becker
- Duke Molecular Physiology Institute; Duke University Medical Center; Durham, NC USA
| | - Danhong Lu
- Duke Molecular Physiology Institute; Duke University Medical Center; Durham, NC USA
| | - Michelle Arlotto
- Duke Molecular Physiology Institute; Duke University Medical Center; Durham, NC USA
| | - Jeffery S Tessem
- Nutrition; Dietetics and Food Science Department; College of Life Sciences; Brigham Young University, Provo, Utah USA
- Correspondence to: Jeffery Sivert Tessem;
| |
Collapse
|
31
|
Ross JS, Russo SB, Chavis GC, Cowart LA. Sphingolipid regulators of cellular dysfunction in Type 2 diabetes mellitus: a systems overview. ACTA ACUST UNITED AC 2014. [DOI: 10.2217/clp.14.37] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
32
|
Ballak DB, van Diepen JA, Moschen AR, Jansen HJ, Hijmans A, Groenhof GJ, Leenders F, Bufler P, Boekschoten MV, Müller M, Kersten S, Li S, Kim S, Eini H, Lewis EC, Joosten LAB, Tilg H, Netea MG, Tack CJ, Dinarello CA, Stienstra R. IL-37 protects against obesity-induced inflammation and insulin resistance. Nat Commun 2014; 5:4711. [DOI: 10.1038/ncomms5711] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 07/16/2014] [Indexed: 12/16/2022] Open
|
33
|
Cadavez L, Montane J, Alcarraz-Vizán G, Visa M, Vidal-Fàbrega L, Servitja JM, Novials A. Chaperones ameliorate beta cell dysfunction associated with human islet amyloid polypeptide overexpression. PLoS One 2014; 9:e101797. [PMID: 25010593 PMCID: PMC4092029 DOI: 10.1371/journal.pone.0101797] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 06/10/2014] [Indexed: 12/14/2022] Open
Abstract
In type 2 diabetes, beta-cell dysfunction is thought to be due to several causes, one being the formation of toxic protein aggregates called islet amyloid, formed by accumulations of misfolded human islet amyloid polypeptide (hIAPP). The process of hIAPP misfolding and aggregation is one of the factors that may activate the unfolded protein response (UPR), perturbing endoplasmic reticulum (ER) homeostasis. Molecular chaperones have been described to be important in regulating ER response to ER stress. In the present work, we evaluate the role of chaperones in a stressed cellular model of hIAPP overexpression. A rat pancreatic beta-cell line expressing hIAPP exposed to thapsigargin or treated with high glucose and palmitic acid, both of which are known ER stress inducers, showed an increase in ER stress genes when compared to INS1E cells expressing rat IAPP or INS1E control cells. Treatment with molecular chaperone glucose-regulated protein 78 kDa (GRP78, also known as BiP) or protein disulfite isomerase (PDI), and chemical chaperones taurine-conjugated ursodeoxycholic acid (TUDCA) or 4-phenylbutyrate (PBA), alleviated ER stress and increased insulin secretion in hIAPP-expressing cells. Our results suggest that the overexpression of hIAPP induces a stronger response of ER stress markers. Moreover, endogenous and chemical chaperones are able to ameliorate induced ER stress and increase insulin secretion, suggesting that improving chaperone capacity can play an important role in improving beta-cell function in type 2 diabetes.
Collapse
Affiliation(s)
- Lisa Cadavez
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Joel Montane
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Gema Alcarraz-Vizán
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Montse Visa
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Laia Vidal-Fàbrega
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Spain
| | - Joan-Marc Servitja
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Anna Novials
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
- * E-mail:
| |
Collapse
|
34
|
Yokota K, Fukushima M, Takahashi Y, Igaki N, Seino S. Insulin secretion and computed tomography values of the pancreas in the early stage of the development of diabetes. J Diabetes Investig 2014; 3:371-6. [PMID: 24843592 PMCID: PMC4019257 DOI: 10.1111/j.2040-1124.2012.00212.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Aims/Introduction: The computed tomography (CT) value of the pancreas was examined across the range of glucose tolerance, and the relationships between pancreatic CT values and factors responsible for glucose intolerance were analyzed. Materials and Methods: A total of 167 health‐check examinees were classified into normal glucose tolerance (NGT), impaired glucose tolerance (IGT) and diabetes mellitus (DM) according to 75 g oral glucose tolerance test (OGTT). Pancreatic and hepatic CT values were estimated at decreasing stages of glucose tolerance. The association of CT values of the pancreas and the indices of glucose tolerance were analyzed. Results: Insulinogenic index (II) was decreased from NGT through IGT to DM. Mean pancreatic CT value was decreased significantly from NGT through IGT to DM. Mean area under the curves of glucose (AUC‐G) was significantly associated with II and insulin sensitivity index (ISI) composite in univariate analysis. In multiple regression analysis, II was most strongly inversely correlated with mean AUC‐G, suggesting that II is the strongest determinant of glucose tolerance in Japanese. In addition, II was significantly associated with mean pancreatic CT value in univariate analysis. In multiple regression analysis, mean pancreatic CT value was strongly correlated with II. Conclusions: Pancreatic CT values were significantly decreased from NGT through IGT to DM. II was the strongest determinant of glucose tolerance, and was significantly influenced by pancreatic CT values. Thus, pancreatic fat deposition might impair insulin secretion in the early stage of development of type 2 diabetes, before overt deterioration of glucose tolerance. (J Diabetes Invest, doi: 10.1111/j.2040‐1124.2012.00212.x, 2012)
Collapse
Affiliation(s)
- Kazuki Yokota
- Division of Diabetes and Endocrinology, Department of Internal Medicine ; Takasago Municipal Hospital, Hyogo
| | - Mitsuo Fukushima
- Division of Diabetes and Endocrinology, Department of Internal Medicine ; Division of Clinical Nutrition and Internal Medicine, Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, Okayama, Japan ; Health Informatics Research Group, Foundation for Biomedical Research and Innovation, Kobe
| | - Yoshihisa Takahashi
- Health Informatics Research Group, Foundation for Biomedical Research and Innovation, Kobe
| | | | - Susumu Seino
- Division of Diabetes and Endocrinology, Department of Internal Medicine ; Division of Cellular and Molecular Medicine, Department of Physiology and Cellular Biology, Kobe University Graduate School of Medicine
| |
Collapse
|
35
|
Cohen M, Syme C, Deforest M, Wells G, Detzler G, Cheng HL, McCrindle B, Hanley A, Hamilton J. Ectopic fat in youth: the contribution of hepatic and pancreatic fat to metabolic disturbances. Obesity (Silver Spring) 2014; 22:1280-6. [PMID: 24402863 DOI: 10.1002/oby.20674] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 11/15/2013] [Accepted: 11/30/2013] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To study the relationships between parameters of glucose and insulin metabolism and visceral and abdominal ectopic fat in youth. METHODS A cross sectional study of 50 children (24 females), 8-18 years old. Anthropometrics, body composition, blood-work and visceral and ectopic fat by magnetic resonance imaging were assessed. Insulin secretion, insulin sensitivity and beta cell function were calculated from an oral glucose tolerance test. RESULTS BMI z-scores ranged between -1.3 and 4.5. The hepatic fat fraction (HFF) ranged between 0 and 36% and pancreatic fat fraction (PFF) between 0 and 14%. Visceral fat, HFF and PFF were associated with clinical and biochemical metabolic abnormalities, and correlated with markers of insulin sensitivity (r = -0.60, P < 0.01; r = -0.64, P < 0.01; r = -0.48, P < 0.01, respectively) insulin secretion (r = 0.55, P < 0.01; r = 0.57, P < 0.01; r = 0.41, P < 0.01, respectively), and beta cell function (r = -0.49, P < 0.01; r = -0.59, P < 0.01; r = -0.39, P < 0.01, respectively). CONCLUSIONS Accumulations of pancreatic and hepatic fat have complementary clinical consequences in youth. While visceral and hepatic fat demonstrated a dominant effect, even relatively small degrees of pancreatic fat deposition may contribute to metabolic alterations.
Collapse
Affiliation(s)
- Michal Cohen
- Division of Endocrinology, the Hospital for Sick Children, and the University of Toronto, ON, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Montane J, Cadavez L, Novials A. Stress and the inflammatory process: a major cause of pancreatic cell death in type 2 diabetes. Diabetes Metab Syndr Obes 2014; 7:25-34. [PMID: 24520198 PMCID: PMC3917922 DOI: 10.2147/dmso.s37649] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Type 2 diabetes (T2D) is a complex metabolic disorder characterized by hyperglycemia in the context of insulin resistance, which precedes insulin deficiency as a result of β-cell failure. Accumulating evidence indicates that β-cell loss in T2D results as a response to the combination of oxidative stress and endoplasmic reticulum (ER) stress. Failure of the ER's adaptive capacity and further activation of the unfolded protein response may trigger macroautophagy (hereafter referred as autophagy) as a process of self-protection and inflammation. Many studies have shown that inflammation plays a very important role in the pathogenesis of T2D. Inflammatory mechanisms and cytokine production activated by stress via the inflammasome may further alter the normal structure of β-cells by inducing pancreatic islet cell apoptosis. Thus, the combination of oxidative and ER stress, together with autophagy insufficiency and inflammation, may contribute to β-cell death or dysfunction in T2D. Therapeutic approaches aimed at ameliorating stress and inflammation may therefore prove to be promising targets for the development of new diabetes treatment methods. Here, we discuss different mechanisms involved in stress and inflammation, and the role of antioxidants, endogenous and chemical chaperones, and autophagic pathways, which may shift the tendency from ER stress and apoptosis toward cell survival. Strategies targeting cell survival can be essential for relieving ER stress and reestablishing homeostasis, which may diminish inflammation and prevent pancreatic β-cell death associated with T2D.
Collapse
Affiliation(s)
- Joel Montane
- Diabetes and Obesity Research Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Lisa Cadavez
- Diabetes and Obesity Research Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Anna Novials
- Diabetes and Obesity Research Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
- Correspondence: Anna Novials, Diabetes and Obesity Research Laboratory, Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic de Barcelona, c/Rosello, 149-153 08036, Barcelona, Spain, Tel +34 93 227 5400, Fax +34 93 312 9403, Email
| |
Collapse
|
37
|
Guan HP, Chen G. Factors affecting insulin-regulated hepatic gene expression. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 121:165-215. [PMID: 24373238 DOI: 10.1016/b978-0-12-800101-1.00006-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Obesity has become a major concern of public health. A common feature of obesity and related metabolic disorders such as noninsulin-dependent diabetes mellitus is insulin resistance, wherein a given amount of insulin produces less than normal physiological responses. Insulin controls hepatic glucose and fatty acid metabolism, at least in part, via the regulation of gene expression. When the liver is insulin-sensitive, insulin can stimulate the expression of genes for fatty acid synthesis and suppress those for gluconeogenesis. When the liver becomes insulin-resistant, the insulin-mediated suppression of gluconeogenic gene expression is lost, whereas the induction of fatty acid synthetic gene expression remains intact. In the past two decades, the mechanisms of insulin-regulated hepatic gene expression have been studied extensively and many components of insulin signal transduction pathways have been identified. Factors that alter these pathways, and the insulin-regulated hepatic gene expression, have been revealed and the underlying mechanisms have been proposed. This chapter summarizes the recent progresses in our understanding of the effects of dietary factors, drugs, bioactive compounds, hormones, and cytokines on insulin-regulated hepatic gene expression. Given the large amount of information and progresses regarding the roles of insulin, this chapter focuses on findings in the liver and hepatocytes and not those described for other tissues and cells. Typical insulin-regulated hepatic genes, such as insulin-induced glucokinase and sterol regulatory element-binding protein-1c and insulin-suppressed cytosolic phosphoenolpyruvate carboxyl kinase and insulin-like growth factor-binding protein 1, are used as examples to discuss the mechanisms such as insulin regulatory element-mediated transcriptional regulation. We also propose the potential mechanisms by which these factors affect insulin-regulated hepatic gene expression and discuss potential future directions of the area of research.
Collapse
Affiliation(s)
- Hong-Ping Guan
- Department of Diabetes, Merck Research Laboratories, Kenilworth, New Jersey, USA
| | - Guoxun Chen
- Department of Nutrition, University of Tennessee at Knoxville, Knoxville, Tennessee, USA
| |
Collapse
|
38
|
Dalbøge LS, Almholt DLC, Neerup TSR, Vassiliadis E, Vrang N, Pedersen L, Fosgerau K, Jelsing J. Characterisation of age-dependent beta cell dynamics in the male db/db mice. PLoS One 2013; 8:e82813. [PMID: 24324833 PMCID: PMC3855780 DOI: 10.1371/journal.pone.0082813] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 11/06/2013] [Indexed: 12/20/2022] Open
Abstract
Aim To characterise changes in pancreatic beta cell mass during the development of diabetes in untreated male C57BLKS/J db/db mice. Methods Blood samples were collected from a total of 72 untreated male db/db mice aged 5, 6, 8, 10, 12, 14, 18, 24 and 34 weeks, for measurement of terminal blood glucose, HbA1c, plasma insulin, and C-peptide. Pancreata were removed for quantification of beta cell mass, islet numbers as well as proliferation and apoptosis by immunohistochemistry and stereology. Results Total pancreatic beta cell mass increased significantly from 2.1 ± 0.3 mg in mice aged 5 weeks to a peak value of 4.84 ± 0.26 mg (P < 0.05) in 12-week-old mice, then gradually decreased to 3.27 ± 0.44 mg in mice aged 34 weeks. Analysis of islets in the 5-, 10-, and 24-week age groups showed increased beta cell proliferation in the 10-week-old animals whereas a low proliferation is seen in older animals. The expansion in beta cell mass was driven by an increase in mean islet mass as the total number of islets was unchanged in the three groups. Conclusions/Interpretation The age-dependent beta cell dynamics in male db/db mice has been described from 5-34 weeks of age and at the same time alterations in insulin/glucose homeostasis were assessed. High beta cell proliferation and increased beta cell mass occur in young animals followed by a gradual decline characterised by a low beta cell proliferation in older animals. The expansion of beta cell mass was caused by an increase in mean islet mass and not islet number.
Collapse
Affiliation(s)
| | | | - Trine S. R. Neerup
- Department of Research and Development, Zealand Pharma A/S, Glostrup, Denmark
| | | | - Niels Vrang
- Department of Histology, Gubra ApS, Hørsholm, Denmark
| | - Lars Pedersen
- Department of Stereology, Visiopharm, Hørsholm, Denmark
| | - Keld Fosgerau
- Department of Research and Development, Zealand Pharma A/S, Glostrup, Denmark
| | - Jacob Jelsing
- Department of Histology, Gubra ApS, Hørsholm, Denmark
- * E-mail:
| |
Collapse
|
39
|
ER stress in rodent islets of Langerhans is concomitant with obesity and β-cell compensation but not with β-cell dysfunction and diabetes. Nutr Diabetes 2013; 3:e93. [PMID: 24145577 PMCID: PMC3817349 DOI: 10.1038/nutd.2013.35] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 08/15/2013] [Accepted: 09/06/2013] [Indexed: 12/20/2022] Open
Abstract
Objective: The objective of this study was to determine whether ER stress correlates with β-cell dysfunction in obesity-associated diabetes. Methods: Quantitative RT-PCR and western blot analysis were used to investigate changes in the expression of markers of ER stress, the unfolded protein response (UPR) and β-cell function in islets isolated from (1) non-diabetic Zucker obese (ZO) and obese female Zucker diabetic fatty (fZDF) rats compared with their lean littermates and from (2) high-fat-diet-fed fZDF rats (HF-fZDF), to induce diabetes, compared with age-matched non-diabetic obese fZDF rats. Results: Markers of an adaptive ER stress/UPR and β-cell function are elevated in islets isolated from ZO and fZDF rats compared with their lean littermates. In islets isolated from HF-fZDF rats, there was no significant change in the expression of markers of ER stress compared with age matched, obese, non-diabetic fZDF rats. Conclusions: These results provide evidence that obesity-induced activation of the UPR is an adaptive response for increasing the ER folding capacity to meet the increased demand for insulin. As ER stress is not exacerbated in high-fat-diet-induced diabetes, we suggest that failure of the islet to mount an effective adaptive UPR in response to an additional increase in insulin demand, rather than chronic ER stress, may ultimately lead to β-cell failure and hence diabetes.
Collapse
|
40
|
Glucose homeostasis and the enteroinsular axis in the horse: a possible role in equine metabolic syndrome. Vet J 2013; 199:11-8. [PMID: 24287206 DOI: 10.1016/j.tvjl.2013.09.064] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 09/02/2013] [Accepted: 09/29/2013] [Indexed: 02/06/2023]
Abstract
One of the principal components of equine metabolic syndrome (EMS) is hyperinsulinaemia combined with insulin resistance. It has long been known that hyperinsulinaemia occurs after the development of insulin resistance. But it is also known that hyperinsulinaemia itself can induce insulin resistance and obesity and might play a key role in the development of metabolic syndrome. This review focuses on the physiology of glucose and insulin metabolism and the pathophysiological mechanisms in glucose homeostasis in the horse (compared with what is already known in humans) in order to gain insight into the pathophysiological principles underlying EMS. The review summarizes new insights on the oral uptake of glucose by the gut and the enteroinsular axis, the role of diet in incretin hormone and postprandial insulin responses, the handling of glucose by the liver, muscle and fat tissue, and the production and secretion of insulin by the pancreas under healthy and disrupted glucose homeostatic conditions in horses.
Collapse
|
41
|
Burke SJ, Updegraff BL, Bellich RM, Goff MR, Lu D, Minkin SC, Karlstad MD, Collier JJ. Regulation of iNOS gene transcription by IL-1β and IFN-γ requires a coactivator exchange mechanism. Mol Endocrinol 2013; 27:1724-42. [PMID: 24014650 DOI: 10.1210/me.2013-1159] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The proinflammatory cytokines IL-1β and IFN-γ decrease functional islet β-cell mass in part through the increased expression of specific genes, such as inducible nitric oxide synthase (iNOS). Dysregulated iNOS protein accumulation leads to overproduction of nitric oxide, which induces DNA damage, impairs β-cell function, and ultimately diminishes cellular viability. However, the transcriptional mechanisms underlying cytokine-mediated expression of the iNOS gene are not completely understood. Herein, we demonstrated that individual mutations within the proximal and distal nuclear factor-κB sites impaired cytokine-mediated transcriptional activation. Surprisingly, mutating IFN-γ-activated site (GAS) elements in the iNOS gene promoter, which are classically responsive to IFN-γ, modulated transcriptional sensitivity to IL-1β. Transcriptional sensitivity to IL-1β was increased by generation of a consensus GAS element and decreased correspondingly with 1 or 2 nucleotide divergences from the consensus sequence. The nuclear factor-κB subunits p65 and p50 bound to the κB response elements in an IL-1β-dependent manner. IL-1β also promoted binding of serine-phosphorylated signal transducer and activator of transcription-1 (STAT1) (Ser727) but not tyrosine-phosphorylated STAT1 (Tyr701) to GAS elements. However, phosphorylation at Tyr701 was required for IFN-γ to potentiate the IL-1β response. Furthermore, coactivator p300 and coactivator arginine methyltransferase were recruited to the iNOS gene promoter with concomitant displacement of the coactivator CREB-binding protein in cells exposed to IL-1β. Moreover, these coordinated changes in factor recruitment were associated with alterations in acetylation, methylation, and phosphorylation of histone proteins. We conclude that p65 and STAT1 cooperate to control iNOS gene transcription in response to proinflammatory cytokines by a coactivator exchange mechanism. This increase in transcription is also associated with signal-specific chromatin remodeling that leads to RNA polymerase II recruitment and phosphorylation.
Collapse
Affiliation(s)
- Susan J Burke
- Department of Nutrition, University of Tennessee, 1215 Cumberland Avenue, 229 JHB, Knoxville, Tennessee 37996-1920.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Pdx-1 activates islet α- and β-cell proliferation via a mechanism regulated by transient receptor potential cation channels 3 and 6 and extracellular signal-regulated kinases 1 and 2. Mol Cell Biol 2013; 33:4017-29. [PMID: 23938296 DOI: 10.1128/mcb.00469-13] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The homeodomain transcription factor Pdx-1 has important roles in pancreatic development and β-cell function and survival. In the present study, we demonstrate that adenovirus-mediated overexpression of Pdx-1 in rat or human islets also stimulates cell replication. Moreover, cooverexpression of Pdx-1 with another homeodomain transcription factor, Nkx6.1, has an additive effect on proliferation compared to either factor alone, implying discrete activating mechanisms. Consistent with this, Nkx6.1 stimulates mainly β-cell proliferation, whereas Pdx-1 stimulates both α- and β-cell proliferation. Furthermore, cyclins D1/D2 are upregulated by Pdx-1 but not by Nkx6.1, and inhibition of cdk4 blocks Pdx-1-stimulated but not Nkx6.1-stimulated islet cell proliferation. Genes regulated by Pdx-1 but not Nkx6.1 were identified by microarray analysis. Two members of the transient receptor potential cation (TRPC) channel family, TRPC3 and TRPC6, are upregulated by Pdx-1 overexpression, and small interfering RNA (siRNA)-mediated knockdown of TRPC3/6 or TRPC6 alone inhibits Pdx-1-induced but not Nkx6.1-induced islet cell proliferation. Pdx-1 also stimulates extracellular signal-regulated kinase 1 and 2 (ERK1/2) phosphorylation, an effect partially blocked by knockdown of TRPC3/6, and blockade of ERK1/2 activation with a MEK1/2 inhibitor partially impairs Pdx-1-stimulated proliferation. These studies define a pathway by which overexpression of Pdx-1 activates islet cell proliferation that is distinct from and additive to a pathway activated by Nkx6.1.
Collapse
|
43
|
Jensen MV, Haldeman JM, Zhang H, Lu D, Huising MO, Vale WW, Hohmeier HE, Rosenberg P, Newgard CB. Control of voltage-gated potassium channel Kv2.2 expression by pyruvate-isocitrate cycling regulates glucose-stimulated insulin secretion. J Biol Chem 2013; 288:23128-40. [PMID: 23788641 DOI: 10.1074/jbc.m113.491654] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Recent studies have shown that the pyruvate-isocitrate cycling pathway, involving the mitochondrial citrate/isocitrate carrier and the cytosolic NADP-dependent isocitrate dehydrogenase (ICDc), is involved in control of glucose-stimulated insulin secretion (GSIS). Here we demonstrate that pyruvate-isocitrate cycling regulates expression of the voltage-gated potassium channel family member Kv2.2 in islet β-cells. siRNA-mediated suppression of ICDc, citrate/isocitrate carrier, or Kv2.2 expression impaired GSIS, and the effect of ICDc knockdown was rescued by re-expression of Kv2.2. Moreover, chronic exposure of β-cells to elevated fatty acids, which impairs GSIS, resulted in decreased expression of Kv2.2. Surprisingly, knockdown of ICDc or Kv2.2 increased rather than decreased outward K(+) current in the 832/13 β-cell line. Immunoprecipitation studies demonstrated interaction of Kv2.1 and Kv2.2, and co-overexpression of the two channels reduced outward K(+) current compared with overexpression of Kv2.1 alone. Also, siRNA-mediated knockdown of ICDc enhanced the suppressive effect of the Kv2.1-selective inhibitor stromatoxin1 on K(+) currents. Our data support a model in which a key function of the pyruvate-isocitrate cycle is to maintain levels of Kv2.2 expression sufficient to allow it to serve as a negative regulator of Kv channel activity.
Collapse
Affiliation(s)
- Mette V Jensen
- Duke Institute of Molecular Physiology, Duke University Medical Center, Durham, North Carolina 27704, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Hernandez AM, Colvin ES, Chen YC, Geiss SL, Eller LE, Fueger PT. Upregulation of p21 activates the intrinsic apoptotic pathway in β-cells. Am J Physiol Endocrinol Metab 2013; 304:E1281-90. [PMID: 23592481 PMCID: PMC3680698 DOI: 10.1152/ajpendo.00663.2012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Diabetes manifests from a loss in functional β-cell mass, which is regulated by a dynamic balance of various cellular processes, including β-cell growth, proliferation, and death as well as secretory function. The cell cycle machinery comprised of cyclins, kinases, and inhibitors regulates proliferation. However, their involvement during β-cell stress during the development of diabetes is not well understood. Interestingly, in a screen of multiple cell cycle inhibitors, p21 was dramatically upregulated in INS-1-derived 832/13 cells and rodent islets by two pharmacological inducers of β-cell stress, dexamethasone and thapsigargin. We hypothesized that β-cell stress upregulates p21 to activate the apoptotic pathway and suppress cell survival signaling. To this end, p21 was adenovirally overexpressed in pancreatic rat islets and 832/13 cells. As expected, p21 overexpression resulted in decreased [(3)H]thymidine incorporation. Flow cytometry analysis in p21-transduced 832/13 cells verified lower replication, as indicated by a decreased cell population in the S phase and a block in G2/M transition. The sub-G0 cell population was higher with p21 overexpression and was attributable to apoptosis, as demonstrated by increased annexin-positive stained cells and cleaved caspase-3 protein. p21-mediated caspase-3 cleavage was inhibited by either overexpression of the antiapoptotic mitochondrial protein Bcl-2 or siRNA-mediated suppression of the proapoptotic proteins Bax and Bak. Therefore, an intact intrinsic apoptotic pathway is central for p21-mediated cell death. In summary, our findings indicate that β-cell apoptosis can be triggered by p21 during stress and is thus a potential target to inhibit for protection of functional β-cell mass.
Collapse
|
45
|
Wu WC, Wang CY. Association between non-alcoholic fatty pancreatic disease (NAFPD) and the metabolic syndrome: case-control retrospective study. Cardiovasc Diabetol 2013; 12:77. [PMID: 23688357 PMCID: PMC3682938 DOI: 10.1186/1475-2840-12-77] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 05/06/2013] [Indexed: 02/07/2023] Open
Abstract
Background Fatty liver is associated with insulin resistance, dyslipidemia, and obesity and is therefore considered a phenotype of metabolic syndrome. However, less is known regarding the metabolic abnormalities associated with non-alcoholic fatty pancreatic disease (NAFPD; fatty pancreas). The present study was performed to ascertain whether fatty pancreas is associated with specific metabolic risk factors and with metabolic syndrome as defined by the Adult Treatment Panel III. Methods Five-hundred-fifty-seven healthy and consecutive subjects without known hypertension or diabetes and who received a health investigation at the National Taiwan University Hospital Health Management Center were enrolled in this retrospective study. Fatty pancreas was diagnosed via trans-abdominal ultrasonographic findings. Results Seventy-two (12.9%) subjects diagnosed with fatty pancreas comprised the fatty pancreas group, and remaining subjects comprised the normal pancreas group. The presence of various demographic and metabolic risk factors was recorded for all subjects, and the two groups were examined for statistically significant differences in these factors. As compared to the absence of fatty pancreas, the presence of the disease was associated with older age and with higher values for each of the following: BMI, abdominal girth/height, abdominal girth (both genders), fasting and postprandial blood glucose, HbA1c, total cholesterol, triglycerides, LDL-cholesterol, systolic blood pressure, and platelet count. In contrast to previously reported findings, serum amylase values were lower in the fatty pancreas as compared to the control group. Conclusion The presence of fatty pancreas represents a meaningful manifestation of metabolic syndrome together with obesity.
Collapse
Affiliation(s)
- Wan-Chen Wu
- Division of Endocrinology, Department of Internal Medicine, National Taiwan University, 7, Chung-Shang South Road, Taipei, Taiwan
| | | |
Collapse
|
46
|
Toledo-Corral CM, Alderete TL, Hu HH, Nayak K, Esplana S, Liu T, Goran MI, Weigensberg MJ. Ectopic fat deposition in prediabetic overweight and obese minority adolescents. J Clin Endocrinol Metab 2013; 98:1115-21. [PMID: 23386647 PMCID: PMC3590481 DOI: 10.1210/jc.2012-3806] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
CONTEXT Optimizing effective prevention and treatment of type 2 diabetes in youth is limited by incomplete understanding of its pathophysiology and how this varies across ethnicities with high risk. OBJECTIVE The aim of this study was to examine the contribution of visceral adipose tissue (VAT), hepatic fat fraction (HFF), and pancreatic fat fraction (PFF) to prediabetes in overweight/obese African American (AA) and Latino youth. DESIGN AND SETTING We conducted a cross-sectional study in an academic pediatric care facility. SUBJECTS A total of 148 healthy, overweight/obese adolescents (56 AA, 92 Latino; 72 males, 76 females; age, 15.5 ± 1.2 y; BMI z-score, 2.1 ± 0.5) participated in the study. They were normal glucose tolerant (n = 106) and prediabetic (n = 42), based on fasting glucose of 100-125 mg/dL and/or 2-hour glucose of 140-199 mg/dL, and/or hemoglobin A1C 6.0-6.4%. MAIN OUTCOME MEASURES We measured sc abdominal adipose tissue, VAT, HFF, and PFF by 3-Tesla magnetic resonance imaging and measured total body fat by dual-energy x-ray absorptiometry. RESULTS Adolescents with prediabetes had 30% higher HFF (P = .001) and 31% higher PFF (P = .042), compared to those with normal glucose tolerance after controlling for age, sex, pubertal stage, ethnicity, total percentage body fat, and VAT. Logistic regression showed that PFF predicted prediabetes in AAs and HFF predicted prediabetes in Latinos, with the odds of having prediabetes increased by 66% for every 1% increase in PFF in African Americans, and increased by 22% for every 1% increase in HFF in Latinos. CONCLUSION These data demonstrate that ectopic fat phenotypes associated with prediabetes are established by adolescence. Ethnic differences in the deposition of ectopic fat in adolescents with prediabetes may differ, with pancreatic fat in AAs, vs hepatic fat in Latino adolescents, being associated with diabetes risk.
Collapse
Affiliation(s)
- Claudia M Toledo-Corral
- Department of Preventive Medicine, University of Southern California, 2250 Alcazar Street, CSC 200, Los Angeles, California 90033, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Colvin ES, Ma HY, Chen YC, Hernandez AM, Fueger PT. Glucocorticoid-induced suppression of β-cell proliferation is mediated by Mig6. Endocrinology 2013; 154:1039-46. [PMID: 23384834 PMCID: PMC3578994 DOI: 10.1210/en.2012-1923] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Glucocorticoids can cause steroid-induced diabetes or accelerate the progression to diabetes by creating systemic insulin resistance and decreasing functional β-cell mass, which is influenced by changes in β-cell function, growth, and death. The synthetic glucocorticoid agonist dexamethasone (Dex) is deleterious to functional β-cell mass by decreasing β-cell function, survival, and proliferation. However, the mechanism by which Dex decreases β-cell proliferation is unknown. Interestingly, Dex induces the transcription of an antiproliferative factor and negative regulator of epidermal growth factor receptor signaling, Mig6 (also known as gene 33, RALT, and Errfi1). We, therefore, hypothesized that Dex impairs β-cell proliferation by increasing the expression of Mig6 and thereby decreasing downstream signaling of epidermal growth factor receptor. We found that Dex induced Mig6 and decreased [(3)H]thymidine incorporation, an index of cellular replication, in mouse, rat, and human islets. Using adenovirally delivered small interfering RNA targeted to Mig6 in rat islets, we were able to limit the induction of Mig6 upon exposure to Dex, compared with islets treated with a control virus, and completely rescued the Dex-mediated impairment in replication. We demonstrated that both Dex and overexpression of Mig6 attenuated the phosphorylation of ERK1/2 and blocked the G(1)/S transition of the cell cycle. In conclusion, Mig6 functions as a molecular brake for β-cell proliferation during glucocorticoid treatment in β-cells, and thus, Mig6 may be a novel target for preventing glucocorticoid-induced impairments in functional β-cell mass.
Collapse
Affiliation(s)
- E Scott Colvin
- Department of Pediatrics and Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | |
Collapse
|
48
|
Simental-Mendía LE, Rodríguez-Morán M, Simental-Saucedo L, Guerrero-Romero F. Insulin secretion is increased in non-diabetic subjects with fasting hypertriglyceridaemia. Diabetes Metab Res Rev 2013; 29:214-9. [PMID: 23225554 DOI: 10.1002/dmrr.2379] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Revised: 11/08/2012] [Accepted: 11/15/2012] [Indexed: 11/08/2022]
Abstract
BACKGROUND The elevation of triglycerides is strongly linked with insulin resistance, but it has not been evaluated in relationship to insulin secretion. The aim of this study was to determine whether hypertriglyceridaemia is associated with abnormal insulin secretion. METHODS A cross-sectional study was carried out. Eligible subjects, apparently healthy men and non-pregnant women aged 20-65 years were recruited. According to the triglyceride levels, subjects were allocated in the groups with hypertriglyceridaemia and normotriglyceridaemia. Hypertriglyceridaemia was defined by serum triglyceride levels ≥150 mg/dL. Insulin secretion was evaluated by the first phase of insulin secretion (1st PIS) and the second phase of insulin secretion (2nd PIS). A regression linear analysis was performed to evaluate the association between hypertriglyceridaemia (independent variable) and the first and second phase insulin secretion (dependent variables). RESULTS A total of 247 apparently healthy subjects were enrolled; 113 (45.7%) with hypertriglyceridaemia and 134 (54.3%) in the control group. The simple regression linear analysis showed a significant association between hypertriglyceridaemia and the 1st PIS [B = 207.0; 95% confidence interval (CI) 33.5-380.5, p = 0.02] and the 2nd PIS (B = 48.7; 95% CI 9.2-88.2, p = 0.01). A multiple regression linear analysis adjusted by age, sex, body mass index and waist circumference was performed showing that fasting hypertriglyceridaemia remained significantly associated with the 1st PIS (B = 184.3; 95% CI 13.0-355.7, p = 0.03) and the 2nd PIS (B = 43.1; 95% CI 4.2-81.9, p = 0.03). CONCLUSIONS The results of this study show that hypertriglyceridaemia is associated with the increase of the 1st PIS and the 2nd PIS in apparently healthy subjects.
Collapse
|
49
|
Russo SB, Ross JS, Cowart LA. Sphingolipids in obesity, type 2 diabetes, and metabolic disease. Handb Exp Pharmacol 2013:373-401. [PMID: 23563667 DOI: 10.1007/978-3-7091-1511-4_19] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Metabolic disease, including obesity and type 2 diabetes, constitutes a major emerging health crisis in Western nations. Although the symptoms and clinical pathology and physiology of these conditions are well understood, the molecular mechanisms underlying the disease process have largely remained obscure. Sphingolipids, a lipid class with both signaling and structural properties, have recently emerged as key players in most major tissues affected by diabetes and are required components in the molecular etiology of this disease. Indeed, sphingolipids have been shown to mediate loss of insulin sensitivity, to promote the characteristic diabetic proinflammatory state, and to induce cell death and dysfunction in important organs such as the pancreas and heart. Furthermore, plasma sphingolipid levels are emerging as potential biomarkers for the decompensation of insulin resistance to frank type 2 diabetes. Despite these discoveries, the roles of specific sphingolipid species and sphingolipid metabolic pathways remain obscure, and newly developed experimental approaches must be employed to elucidate the detailed molecular mechanisms necessary for rational drug development and other clinical applications.
Collapse
Affiliation(s)
- S B Russo
- Department of Biochemistry and Molecular Biology, The Medical University of South Carolina, Charleston, SC, USA
| | | | | |
Collapse
|
50
|
Chen YC, Colvin ES, Maier BF, Mirmira RG, Fueger PT. Mitogen-inducible gene 6 triggers apoptosis and exacerbates ER stress-induced β-cell death. Mol Endocrinol 2012. [PMID: 23204325 DOI: 10.1210/me.2012-1174] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The increased insulin secretory burden placed on pancreatic β-cells during obesity and insulin resistance can ultimately lead to β-cell dysfunction and death and the development of type 2 diabetes. Mitogen-inducible gene 6 (Mig6) is a cellular stress-responsive protein that can negatively regulate the duration and intensity of epidermal growth factor receptor signaling and has been classically viewed as a molecular brake for proliferation. In this study, we used Mig6 heterozygous knockout mice (Mig6(+/-)) to study the role of Mig6 in regulating β-cell proliferation and survival. Surprisingly, the proliferation rate of Mig6(+/-) pancreatic islets was lower than wild-type islets despite having comparable β-cell mass and glucose tolerance. We thus speculated that Mig6 regulates cellular death. Using adenoviral vectors to overexpress or knockdown Mig6, we found that caspase 3 activation during apoptosis was dependent on the level of Mig6. Interestingly, Mig6 expression was induced during endoplasmic reticulum (ER) stress, and its protein levels were maintained throughout ER stress. Using polyribosomal profiling, we identified that Mig6 protein translation was maintained, whereas the global protein translation was inhibited during ER stress. In addition, Mig6 overexpression exacerbated ER stress-induced caspase 3 activation in vitro. In conclusion, Mig6 is transcriptionally up-regulated and resistant to global translational inhibition during stressed conditions in β-cells and mediates apoptosis in the form of caspase 3 activation. The sustained production of Mig6 protein exacerbates ER stress-induced β-cell death. Thus, preventing the induction, translation, and/or function of Mig6 is warranted for increasing β-cell survival.
Collapse
Affiliation(s)
- Yi-Chun Chen
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | |
Collapse
|