1
|
Pérez-Regalado S, Leon J, Padial P, Benavente C, Almeida F, Bonitch-Góngora J, de la Fuente B, Feriche B. Exploring the Impact of Resistance Training at Moderate Altitude on Metabolic Cytokines in Humans: Implications for Adipose Tissue Dynamics. Int J Mol Sci 2024; 25:11418. [PMID: 39518972 PMCID: PMC11546518 DOI: 10.3390/ijms252111418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/16/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Hypobaric hypoxia (HH) limits oxygen supply to tissues and increases metabolic demands, especially during exercise. We studied the influence of HH exposure on the subcutaneous adipose tissue (SAT) thickness and circulating metabolic-related cytokines levels after a resistance training (RT) program. Twenty trained men participated in a traditional hypertrophy RT for 8 weeks (three sessions/week) under intermittent terrestrial HH (2320 m) or normoxia (N, 690 m) conditions. Before, at week 6, and after the RT, SAT, and vastus lateralis (VL) muscle thickness were measured by ultrasound. Blood samples were taken to analyse serum cytokines (IL-6, IL-15, irisin, and myostatin) by multiplex immunoassay. Our findings revealed a moderate reduction in IL-6 and irisin in HH following the RT (ES < -0.64; p < 0.05). Additionally, RT in HH promoted serum IL-15 release (ES = 0.890; p = 0.062), which exhibited a trivial inverse association with the reductions observed on SAT (-17.69%; p < 0.001) compared with N. RT in HH explained ~50% of SAT variance (p < 0.001). These results highlight the benefit of stressor factors linked to RT in HH on SAT through the modulation of serum metabolic cytokine profiles, suggesting a potential effect on overall body composition.
Collapse
Affiliation(s)
- Sergio Pérez-Regalado
- Department of Physical Education and Sport, Faculty of Sport Sciences, University of Granada, 18011 Granada, Spain; (S.P.-R.); (P.P.); (C.B.); (F.A.); (J.B.-G.); (B.F.)
| | - Josefa Leon
- Biosanitary Research Institute of Granada, ibs. Granada, 18012 Granada, Spain
- Clinical Management Unit of Digestive System, San Cecilio University Clinical Hospital, 18007 Granada, Spain
| | - Paulino Padial
- Department of Physical Education and Sport, Faculty of Sport Sciences, University of Granada, 18011 Granada, Spain; (S.P.-R.); (P.P.); (C.B.); (F.A.); (J.B.-G.); (B.F.)
| | - Cristina Benavente
- Department of Physical Education and Sport, Faculty of Sport Sciences, University of Granada, 18011 Granada, Spain; (S.P.-R.); (P.P.); (C.B.); (F.A.); (J.B.-G.); (B.F.)
| | - Filipa Almeida
- Department of Physical Education and Sport, Faculty of Sport Sciences, University of Granada, 18011 Granada, Spain; (S.P.-R.); (P.P.); (C.B.); (F.A.); (J.B.-G.); (B.F.)
| | - Juan Bonitch-Góngora
- Department of Physical Education and Sport, Faculty of Sport Sciences, University of Granada, 18011 Granada, Spain; (S.P.-R.); (P.P.); (C.B.); (F.A.); (J.B.-G.); (B.F.)
| | - Blanca de la Fuente
- High-Performance Centre of Sierra Nevada, Spanish Sports Council, 18196 Granada, Spain;
| | - Belén Feriche
- Department of Physical Education and Sport, Faculty of Sport Sciences, University of Granada, 18011 Granada, Spain; (S.P.-R.); (P.P.); (C.B.); (F.A.); (J.B.-G.); (B.F.)
| |
Collapse
|
2
|
Wang KY, Gao MX, Qi HB, An WT, Lin JY, Ning SL, Yang F, Xiao P, Cheng J, Pan W, Cheng QX, Wang J, Fang L, Sun JP, Yu X. Differential contributions of G protein- or arrestin subtype-mediated signalling underlie urocortin 3-induced somatostatin secretion in pancreatic δ cells. Br J Pharmacol 2024; 181:2600-2621. [PMID: 38613153 DOI: 10.1111/bph.16351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/29/2023] [Accepted: 02/05/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND AND PURPOSE Pancreatic islets are modulated by cross-talk among different cell types and paracrine signalling plays important roles in maintaining glucose homeostasis. Urocortin 3 (UCN3) secreted by pancreatic β cells activates the CRF2 receptor (CRF2R) and downstream pathways mediated by different G protein or arrestin subtypes in δ cells to cause somatostatin (SST) secretion, and constitutes an important feedback circuit for glucose homeostasis. EXPERIMENTAL APPROACH Here, we used Arrb1-/-, Arrb2-/-, Gsfl/fl and Gqfl/fl knockout mice, the G11-shRNA-GFPfl/fl lentivirus, as well as functional assays and pharmacological characterization to study how the coupling of Gs, G11 and β-arrestin1 to CRF2R contributed to UCN3-induced SST secretion in pancreatic δ cells. KEY RESULTS Our study showed that CRF2R coupled to a panel of G protein and arrestin subtypes in response to UCN3 engagement. While RyR3 phosphorylation by PKA at the S156, S2706 and S4697 sites may underlie the Gs-mediated UCN3- CRF2R axis for SST secretion, the interaction of SYT1 with β-arrestin1 is also essential for efficient SST secretion downstream of CRF2R. The specific expression of the transcription factor Stat6 may contribute to G11 expression in pancreatic δ cells. Furthermore, we found that different UCN3 concentrations may have distinct effects on glucose homeostasis, and these effects may depend on different CRF2R downstream effectors. CONCLUSIONS AND IMPLICATIONS Collectively, our results provide a landscape view of signalling mediated by different G protein or arrestin subtypes downstream of paracrine UCN3- CRF2R signalling in pancreatic β-δ-cell circuits, which may facilitate the understanding of fine-tuned glucose homeostasis networks.
Collapse
Affiliation(s)
- Kai-Yu Wang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Ming-Xin Gao
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Hai-Bo Qi
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Wen-Tao An
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jing-Yu Lin
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Shang-Lei Ning
- Department of Hepatobiliary Surgery, General surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Fan Yang
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Peng Xiao
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Jie Cheng
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Pan
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Qiu-Xia Cheng
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Jin Wang
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Le Fang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jin-Peng Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| |
Collapse
|
3
|
Li C, Peng Y, Zhu X, Liu Y, Zou J, Zhu H, Li X, Yi H, Guan J, Zhang X, Xu H, Yin S. Independent relationship between sleep apnea-specific hypoxic burden and glucolipid metabolism disorder: a cross-sectional study. Respir Res 2024; 25:214. [PMID: 38762509 PMCID: PMC11102635 DOI: 10.1186/s12931-024-02846-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 05/12/2024] [Indexed: 05/20/2024] Open
Abstract
OBJECTIVES Obstructive sleep apnea (OSA) is associated with abnormal glucose and lipid metabolism. However, whether there is an independent association between Sleep Apnea-Specific Hypoxic Burden (SASHB) and glycolipid metabolism disorders in patients with OSA is unknown. METHODS We enrolled 2,173 participants with suspected OSA from January 2019 to July 2023 in this study. Polysomnographic variables, biochemical indicators, and physical measurements were collected from each participant. Multiple linear regression analyses were used to evaluate independent associations between SASHB, AHI, CT90 and glucose as well as lipid profile. Furthermore, logistic regressions were used to determine the odds ratios (ORs) for abnormal glucose and lipid metabolism across various SASHB, AHI, CT90 quartiles. RESULTS The SASHB was independently associated with fasting blood glucose (FBG) (β = 0.058, P = 0.016), fasting insulin (FIN) (β = 0.073, P < 0.001), homeostasis model assessment of insulin resistance (HOMA-IR) (β = 0.058, P = 0.011), total cholesterol (TC) (β = 0.100, P < 0.001), total triglycerides (TG) (β = 0.063, P = 0.011), low-density lipoprotein cholesterol (LDL-C) (β = 0.075, P = 0.003), apolipoprotein A-I (apoA-I) (β = 0.051, P = 0.049), apolipoprotein B (apoB) (β = 0.136, P < 0.001), apolipoprotein E (apoE) (β = 0.088, P < 0.001) after adjustments for confounding factors. Furthermore, the ORs for hyperinsulinemia across the higher SASHB quartiles were 1.527, 1.545, and 2.024 respectively, compared with the lowest quartile (P < 0.001 for a linear trend); the ORs for hyper-total cholesterolemia across the higher SASHB quartiles were 1.762, 1.998, and 2.708, compared with the lowest quartile (P < 0.001 for a linear trend) and the ORs for hyper-LDL cholesterolemia across the higher SASHB quartiles were 1.663, 1.695, and 2.316, compared with the lowest quartile (P < 0.001 for a linear trend). Notably, the ORs for hyper-triglyceridemia{1.471, 1.773, 2.099} and abnormal HOMA-IR{1.510, 1.492, 1.937} maintained a consistent trend across the SASHB quartiles. CONCLUSIONS We found SASHB was independently associated with hyperinsulinemia, abnormal HOMA-IR, hyper-total cholesterolemia, hyper-triglyceridemia and hyper-LDL cholesterolemia in Chinese Han population. Further prospective studies are needed to confirm that SASHB can be used as a predictor of abnormal glycolipid metabolism disorders in patients with OSA. TRIAL REGISTRATION ChiCTR1900025714 { http://www.chictr.org.cn/ }; Prospectively registered on 6 September 2019; China.
Collapse
Affiliation(s)
- Chenyang Li
- Department of Otolaryngology-Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Yu Peng
- Department of Otolaryngology-Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Xiaoyue Zhu
- Department of Otolaryngology-Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Yupu Liu
- Department of Otolaryngology-Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Jianyin Zou
- Department of Otolaryngology-Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Huaming Zhu
- Department of Otolaryngology-Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Xinyi Li
- Department of Otolaryngology-Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Hongliang Yi
- Department of Otolaryngology-Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Jian Guan
- Department of Otolaryngology-Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai, China.
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China.
| | - Xu Zhang
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China.
| | - Huajun Xu
- Department of Otolaryngology-Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai, China.
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China.
| | - Shankai Yin
- Department of Otolaryngology-Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| |
Collapse
|
4
|
Wang J, Liu S, Sun L, Kong Z, Chai J, Wen J, Tian X, Chen N, Xu C. Association of attenuated leptin signaling pathways with impaired cardiac function under prolonged high-altitude hypoxia. Sci Rep 2024; 14:10206. [PMID: 38702334 PMCID: PMC11068766 DOI: 10.1038/s41598-024-59559-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 04/12/2024] [Indexed: 05/06/2024] Open
Abstract
Cardiovascular function and adipose metabolism were markedly influenced under high altitudes. However, the interplay between adipokines and heart under hypoxia remains to be elucidated. We aim to explore alterations of adipokines and underlying mechanisms in regulating cardiac function under high altitudes. We investigated the cardiopulmonary function and five adipokines in Antarctic expeditioners at Kunlun Station (4,087 m) for 20 days and established rats exposed to hypobaric hypoxia (5,000 m), simulating Kunlun Station. Antarctic expeditioners exhibited elevated heart rate, blood pressure, systemic vascular resistance, and decreased cardiac pumping function. Plasma creatine phosphokinase-MB (CK-MB) and platelet-endothelial cell adhesion molecule-1 (sPecam-1) increased, and leptin, resistin, and lipocalin-2 decreased. Plasma leptin significantly correlated with altered cardiac function indicators. Additionally, hypoxic rats manifested impaired left ventricular systolic and diastolic function, elevated plasma CK-MB and sPecam-1, and decreased plasma leptin. Chronic hypoxia for 14 days led to increased myocyte hypertrophy, fibrosis, apoptosis, and mitochondrial dysfunction, coupled with reduced protein levels of leptin signaling pathways in myocardial tissues. Cardiac transcriptome analysis revealed leptin was associated with downregulated genes involved in rhythm, Na+/K+ transport, and cell skeleton. In conclusion, chronic hypoxia significantly reduced leptin signaling pathways in cardiac tissues along with significant pathological changes, thus highlighting the pivotal role of leptin in regulation of cardiac function under high altitudes.
Collapse
Affiliation(s)
- Jianan Wang
- Institute of Basic Medical Sciences, School of Basic Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Shiying Liu
- Institute of Basic Medical Sciences, School of Basic Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Lihong Sun
- Center for Experimental Animal Research, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Zhanping Kong
- Qinghai Provincial People's Hospital, Xining, 810000, Qinghai, China
| | - Jiamin Chai
- Institute of Basic Medical Sciences, School of Basic Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Jigang Wen
- Institute of Basic Medical Sciences, School of Basic Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Xuan Tian
- Institute of Basic Medical Sciences, School of Basic Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Nan Chen
- Institute of Basic Medical Sciences, School of Basic Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Chengli Xu
- Institute of Basic Medical Sciences, School of Basic Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China.
- Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China.
| |
Collapse
|
5
|
Flaherty SE, Bezy O, Zheng W, Yan D, Li X, Jagarlapudi S, Albuquerque B, Esquejo RM, Peloquin M, Semache M, Mancini A, Kang L, Drujan D, Breitkopf SB, Griffin JD, Jean Beltran PM, Xue L, Stansfield J, Pashos E, Shakey Q, Pehmøller C, Monetti M, Birnbaum MJ, Fortin JP, Wu Z. Chronic UCN2 treatment desensitizes CRHR2 and improves insulin sensitivity. Nat Commun 2023; 14:3953. [PMID: 37402735 DOI: 10.1038/s41467-023-39597-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 06/21/2023] [Indexed: 07/06/2023] Open
Abstract
Urocortin 2 (UCN2) acts as a ligand for the G protein-coupled receptor corticotropin-releasing hormone receptor 2 (CRHR2). UCN2 has been reported to improve or worsen insulin sensitivity and glucose tolerance in vivo. Here we show that acute dosing of UCN2 induces systemic insulin resistance in male mice and skeletal muscle. Inversely, chronic elevation of UCN2 by injection with adenovirus encoding UCN2 resolves metabolic complications, improving glucose tolerance. CRHR2 recruits Gs in response to low concentrations of UCN2, as well as Gi and β-Arrestin at high concentrations of UCN2. Pre-treating cells and skeletal muscle ex vivo with UCN2 leads to internalization of CRHR2, dampened ligand-dependent increases in cAMP, and blunted reductions in insulin signaling. These results provide mechanistic insights into how UCN2 regulates insulin sensitivity and glucose metabolism in skeletal muscle and in vivo. Importantly, a working model was derived from these results that unifies the contradictory metabolic effects of UCN2.
Collapse
Affiliation(s)
- Stephen E Flaherty
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Olivier Bezy
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Wei Zheng
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Dong Yan
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Xiangping Li
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Srinath Jagarlapudi
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Bina Albuquerque
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Ryan M Esquejo
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Matthew Peloquin
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | | | | | - Liya Kang
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Doreen Drujan
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Susanne B Breitkopf
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - John D Griffin
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Pierre M Jean Beltran
- Machine Learning and Computational Sciences, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Liang Xue
- Machine Learning and Computational Sciences, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - John Stansfield
- Biostatistics, Early Clinical Development, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Evanthia Pashos
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Quazi Shakey
- Biomedicine design, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Christian Pehmøller
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Mara Monetti
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Morris J Birnbaum
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Jean-Philippe Fortin
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA
| | - Zhidan Wu
- Internal Medicine Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, MA, USA.
| |
Collapse
|
6
|
Song Q, Liu S, Wang J, Chai J, Wen J, Xu C. Hypoxia promotes white adipose tissues browning in rats under simulated environment at altitude of 5000 m. Biochem Biophys Res Commun 2023; 666:146-153. [PMID: 37187092 DOI: 10.1016/j.bbrc.2023.05.015] [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: 04/09/2023] [Revised: 04/24/2023] [Accepted: 05/03/2023] [Indexed: 05/17/2023]
Abstract
People living in plains tend to decrease in body weight or body fat percentage after entering the plateau. Previous studies have found that plateau animals can burn fat and release calories through white adipose tissues (WATs) browning. However, these studies have focused on the effect of cold stimulation that induced WATs browning while there's hardly study on the effect of hypoxia. In this study, we investigate that whether and how hypoxia contributes to WATs browning in rats from acute to chronic hypoxia. We constructed hypobaric hypoxic rat models by exposing 9-week-old male SD rats to a hypobaric hypoxic chamber for 1, 3, 14 and 28 days (Group H) under simulated environment at altitude of 5000 m. We also established normoxic control groups for each time period (Group C), as well as paired 1-day and 14-day normoxic food-restriction rats that were fed the same amount of food as the hypoxic group ate (Group R). We then observed the growth status of rats and recorded dynamic changes in histologic, cellular and molecular levels of perirenal WATs (PWAT), epididymal WATs (EWAT) and subcutaneous WATs (SWAT) in each group. Results showed that (1) Hypoxic rats had lower food intake, significantly lower body weight than control rats, and showed lower WATs index. (2) In group H14, ASC1 mRNA expressions of PWAT and EWAT in rats were lower than that in group C14, and PAT2 mRNA expression of EWAT was higher than that in both group C14 and R14. In group R14, however, ASC1 mRNA expressions of PWAT and EWAT in rats were higher than both group C14 and H14, and that of SWAT was also significantly higher than group C14. (3) In group H3, both the mRNA and protein levels of uncoupling protein 1 (UCP1) of PWAT in rats were significantly increased than group C3. And in group H14, those of EWAT in rats were significantly increased than group C14. (4) In plasma of rats, norepinephrine (NE) level was significantly increased in group H3 than group C3, and free fatty acids (FFAs) level was significantly increased in group H14 than both group C14 and R14. In group R1, FASN mRNA expressions of PWAT and EWAT in rats were down-regulated than group C1. In group H3, FASN mRNA expressions of PWAT and EWAT in rats were down-regulated while ATGL mRNA expression of EWAT was up-regulated than group C3. Conversely, in group R14, FASN mRNA expressions of PWAT and EWAT in rats were significantly up-regulated than group C14 and H14. These results suggested that hypoxia promoted different WATs browning in rats under simulated environment at altitude of 5000 m and changed the lipid metabolism in WATs. Furthermore, rats in the chronic hypoxic group showed a completely different lipid metabolism of WATs from that in paired food-restriction group.
Collapse
Affiliation(s)
- Qiaoyue Song
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Shiying Liu
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Jianan Wang
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Jiamin Chai
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Jigang Wen
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Chengli Xu
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China.
| |
Collapse
|
7
|
Xiong Y, Wang Y, Xiong Y, Teng L. 4-PBA inhibits hypoxia-induced lipolysis in rat adipose tissue and lipid accumulation in the liver through regulating ER stress. Food Sci Nutr 2023; 11:1223-1231. [PMID: 36911831 PMCID: PMC10002945 DOI: 10.1002/fsn3.3156] [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: 02/21/2022] [Revised: 11/05/2022] [Accepted: 11/10/2022] [Indexed: 02/12/2023] Open
Abstract
High-altitude hypoxia may disturb the metabolic modulation and function of both adipose tissue and liver. The endoplasmic reticulum (ER) is a crucial organelle in lipid metabolism and ER stress is closely correlated with lipid metabolism dysfunction. The aim of this study is to elucidate whether the inhibition of ER stress could alleviate hypoxia-induced white adipose tissue (WAT) lipolysis and liver lipid accumulation-mediated hepatic injury. A rat model of high-altitude hypoxia (5500 m) was established using hypobaric chamber. The response of ER stress and lipolysis-related pathways were analyzed in WAT under hypoxia exposure with or without 4-phenylbutyric acid (PBA) treatment. Liver lipid accumulation, liver injury, and apoptosis were evaluated. Hypoxia evoked significant ER stress in WAT, evidenced by increased GRP78, CHOP, and phosphorylation of IRE1α, PERK. Moreover, Lipolysis in perirenal WAT significantly increased under hypoxia, accompanied with increased phosphorylation of hormone-sensitive lipase (HSL) and perilipin. Treatment with 4-PBA, inhibitor of ER stress, effectively attenuated hypoxia-induced lipolysis via cAMP-PKA-HSL/perilipin pathway. In addition, 4-PBA treatment significantly inhibited the increase in fatty acid transporters (CD36, FABP1, FABP4) and ameliorated liver FFA accumulation. 4-PBA treatment significantly attenuated liver injury and apoptosis, which is likely resulting from decreased liver lipid accumulation. Our results highlight the importance of ER stress in hypoxia-induced WAT lipolysis and liver lipid accumulation.
Collapse
Affiliation(s)
- Yanlei Xiong
- Department of PathologyXuanwu Hospital, Capital Medical UniversityBeijingChina
- Department of PathophysiologyInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS); School of Basic Medicine, Peking Union Medical College (PUMC)BeijingChina
| | - Yueming Wang
- Department of AnatomySchool of Basic Medicine, Binzhou Medical UniversityYantaiChina
| | - Yanlian Xiong
- Department of AnatomySchool of Basic Medicine, Binzhou Medical UniversityYantaiChina
| | - Lianghong Teng
- Department of PathologyXuanwu Hospital, Capital Medical UniversityBeijingChina
| |
Collapse
|
8
|
Zhao LH, Lin J, Ji SY, Zhou XE, Mao C, Shen DD, He X, Xiao P, Sun J, Melcher K, Zhang Y, Yu X, Xu HE. Structure insights into selective coupling of G protein subtypes by a class B G protein-coupled receptor. Nat Commun 2022; 13:6670. [PMID: 36335102 PMCID: PMC9637140 DOI: 10.1038/s41467-022-33851-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 10/05/2022] [Indexed: 11/07/2022] Open
Abstract
The ability to couple with multiple G protein subtypes, such as Gs, Gi/o, or Gq/11, by a given G protein-coupled receptor (GPCR) is critical for many physiological processes. Over the past few years, the cryo-EM structures for all 15 members of the medically important class B GPCRs, all in complex with Gs protein, have been determined. However, no structure of class B GPCRs with Gq/11 has been solved to date, limiting our understanding of the precise mechanisms of G protein coupling selectivity. Here we report the structures of corticotropin releasing factor receptor 2 (CRF2R) bound to Urocortin 1 (UCN1), coupled with different classes of heterotrimeric G proteins, G11 and Go. We compare these structures with the structure of CRF2R in complex with Gs to uncover the structural differences that determine the selective coupling of G protein subtypes by CRF2R. These results provide important insights into the structural basis for the ability of CRF2R to couple with multiple G protein subtypes.
Collapse
Affiliation(s)
- Li-Hua Zhao
- grid.9227.e0000000119573309The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Jingyu Lin
- grid.27255.370000 0004 1761 1174Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, 250012 China
| | - Su-Yu Ji
- grid.13402.340000 0004 1759 700XDepartment of Biophysics and Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058 China
| | - X. Edward Zhou
- grid.251017.00000 0004 0406 2057Department of Structural Biology, Van Andel Research Institute, Grand Rapids, MI 49503 USA
| | - Chunyou Mao
- grid.13402.340000 0004 1759 700XDepartment of Biophysics and Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058 China
| | - Dan-Dan Shen
- grid.13402.340000 0004 1759 700XDepartment of Biophysics and Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058 China
| | - Xinheng He
- grid.9227.e0000000119573309The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Peng Xiao
- grid.27255.370000 0004 1761 1174Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, 250012 China
| | - Jinpeng Sun
- grid.27255.370000 0004 1761 1174Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, 250012 China
| | - Karsten Melcher
- grid.251017.00000 0004 0406 2057Department of Structural Biology, Van Andel Research Institute, Grand Rapids, MI 49503 USA
| | - Yan Zhang
- grid.13402.340000 0004 1759 700XDepartment of Biophysics and Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058 China ,grid.13402.340000 0004 1759 700XLiangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121 China ,grid.13402.340000 0004 1759 700XMOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, 310058 China ,Zhejiang Provincial Key Laboratory of Immunity and Inflammatory diseases, Hangzhou, 310058 China
| | - Xiao Yu
- grid.27255.370000 0004 1761 1174Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, 250012 China
| | - H. Eric Xu
- grid.9227.e0000000119573309The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| |
Collapse
|
9
|
He J, Zhong Y, Sun Y, Xie C, Yu T. Construction of an immune-related prognostic model by exploring the tumor microenvironment of clear cell renal cell carcinoma. Anal Biochem 2022; 643:114567. [PMID: 35122734 DOI: 10.1016/j.ab.2022.114567] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/05/2022] [Accepted: 01/19/2022] [Indexed: 11/19/2022]
Abstract
OBJECTIVE In this study, bioinformatics methods were performed to screen the candidate prognosis-related genes of clear cell renal cell carcinoma (ccRCC) by analyzing the tumor microenvironment (TME). METHODS Gene expression and clinical data of ccRCC patients were accessed from TCGA, and R package ESTIMATE was applied to calculate immune, stromal, and ESTIMATE scores of the patients. Survival analysis was conducted per median of these three scores. Based on the scoring results, differentially expressed genes (DEGs) were screened. Regression algorithms were utilized to screen prognostic genes and establish a risk model. Finally, pathway activity differences were analyzed through GSEA. RESULTS Patients with the unfavorable prognosis had high immune scores. 619 DEGs (499 up-regulated and 120 down-regulated) were screened based on the differences in gene expression of the patients with high and low immune scores. These genes mainly participated in immune-related signaling pathways. A prognostic risk model for ccRCC patients was constructed and 7 immune-related signature genes (RORB, TNFSF14, UCN2, USP2, TOX3, KLRC2, SLAMF9) were obtained through regression analysis. The constructed prognostic risk model could be used for determining prognoses of patients with ccRCC. CONCLUSION We unraveled the association between TME and prognosis of ccRCC patients and established a prognostic risk model based on the differentially expressed genes. These results contributed to understanding of TME that affected patients' prognosis and progression of ccRCC and conduced to finding potential biomarkers of ccRCC.
Collapse
Affiliation(s)
- Jia He
- Department of Urology, Zhejiang Rongjun Hospital, Jiaxing, 314000, China
| | - Yun Zhong
- Department of Urology, Zhejiang Rongjun Hospital, Jiaxing, 314000, China
| | - Yanli Sun
- Department of Urology, Zhejiang Rongjun Hospital, Jiaxing, 314000, China
| | - Chao Xie
- Department of Urology, Zhejiang Rongjun Hospital, Jiaxing, 314000, China
| | - Tianqiang Yu
- Department of Urology, Zhejiang Rongjun Hospital, Jiaxing, 314000, China.
| |
Collapse
|
10
|
Protective Effects of Adiponectin against Cobalt Chloride-Induced Apoptosis of Smooth Muscle Cells via cAMP/PKA Pathway. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7169348. [PMID: 33102590 PMCID: PMC7576343 DOI: 10.1155/2020/7169348] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022]
Abstract
Adiponectin (APN) is an adipokine secreted from adipose tissue and exhibits biological functions such as microcirculation-regulating, hearing-protective, and antiapoptotic. However, the effect of APN on the apoptosis of spiral arterial smooth muscle cells (SMCs) under hypoxic conditions in vitro is not clear. We used cobalt chloride (CoCl2) to simulate chemical hypoxia in vitro, and the SMCs were pretreated with APN and then stimulated with CoCl2. The viability of cells and apoptosis were assessed by CCK-8 and flow cytometry, respectively. Superoxide dismutase (SOD) activity, malondialdehyde (MDA) levels, cAMP level, and the activity of PKA were detected by ELISA. Protein expression and localization were studied by Western blot and immunofluorescence analysis. In the present study, we found that APN exhibits antiapoptosis effects. CoCl2 exhibited decreased cell viability, increased apoptosis and MDA levels, and decreased SOD activity in a concentration-dependent manner, compared with the control group. Moreover, CoCl2 upregulated the expression levels of Bax and cleaved caspase-3 and then downregulated Bcl-2 levels in a time-dependent manner. Compared with the CoCl2 group, the group pretreated with APN had increased cell viability, SOD activity, PKA activity, cAMP level, and PKA expression, but decreased MDA levels and apoptosis. Lastly, the protective effect of APN was blocked by cAMP inhibitor SQ22536 and PKA inhibitor H 89. These results showed that APN protected SMCs against CoCl2-induced hypoxic injury via the cAMP/PKA signaling pathway.
Collapse
|
11
|
Hu T, Huang W, Li Z, Kane MA, Zhang L, Huang SM, Wang H. Comparative proteomic analysis of SLC13A5 knockdown reveals elevated ketogenesis and enhanced cellular toxic response to chemotherapeutic agents in HepG2 cells. Toxicol Appl Pharmacol 2020; 402:115117. [PMID: 32634519 DOI: 10.1016/j.taap.2020.115117] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 06/09/2020] [Accepted: 06/12/2020] [Indexed: 12/29/2022]
Abstract
Solute carrier family 13 member 5 (SLC13A5) is an uptake transporter mainly expressed in the liver and transports citrate from blood circulation into hepatocytes. Accumulating evidence suggests that SLC13A5 is involved in hepatic lipogenesis, cell proliferation, epilepsy, and bone development in mammals. However, the molecular mechanisms behind SLC13A5-mediated physiological/pathophysiological changes are largely unknown. In this regard, we conducted a differential proteome analysis in HepG2 and SLC13A5-knockdown (KD) HepG2 cells. A total of 3826 proteins were quantified and 330 proteins showed significant alterations (fold change ≥1.5; p < .05) in the knockdown cells. Gene ontology enrichment analysis reveals that 38 biological processes were significantly changed, with ketone body biosynthetic process showing the most significant upregulation following SLC13A5-KD. Catalytic activity and binding activity were the top two molecular functions associated with differentially expressed proteins, while HMG-CoA lyase activity showed the highest fold enrichment. Further ingenuity pathway analysis predicted 40 canonical pathways and 28 upstream regulators (p < .01), of which most were associated with metabolism, cell proliferation, and stress response. In line with these findings, functional validation demonstrated increased levels of two key ketone bodies, acetoacetate and β-hydroxybutyrate, in the SLC13A5-KD cells. Additional experiments showed that SLC13A5-KD sensitizes HepG2 cells to cellular stress caused by a number of chemotherapeutic agents. Together, our findings demonstrate that knockdown of SLC13A5 promotes hepatic ketogenesis and enhances cellular stress response in HepG2 cells, suggesting a potential role of this transporter in metabolic disorders and liver cancer.
Collapse
Affiliation(s)
- Tao Hu
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, United States of America
| | - Weiliang Huang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, United States of America
| | - Zhihui Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, United States of America
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, United States of America
| | - Lei Zhang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration (FDA), Silver Spring, MD 20993, United States of America
| | - Shiew-Mei Huang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration (FDA), Silver Spring, MD 20993, United States of America
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, United States of America.
| |
Collapse
|
12
|
Barros D, García-Río F. Obstructive sleep apnea and dyslipidemia: from animal models to clinical evidence. Sleep 2020; 42:5204276. [PMID: 30476296 DOI: 10.1093/sleep/zsy236] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 10/02/2018] [Accepted: 11/22/2018] [Indexed: 01/11/2023] Open
Abstract
Lipid metabolism deregulation constitutes the pathogenic basis for the development of atherosclerosis and justifies a high incidence of cardiovascular-related morbidity and mortality. Some data suggest that dyslipidemia may be associated with sleep-disordered breathing, mainly obstructive sleep apnea (OSA), due to alterations in fundamental biochemical processes, such as intermittent hypoxia (IH). The aim of this systematic review was to identify and critically evaluate the current evidence supporting the existence of a possible relationship between OSA and alterations in lipid metabolism. Much evidence shows that, during the fasting state, OSA and IH increase lipid delivery from the adipose tissue to the liver through an up-regulation of the sterol regulatory element-binding protein-1 and stearoyl-CoA desaturase-1, increasing the synthesis of cholesterol esters and triglycerides. In the postprandial state, lipoprotein clearance is delayed due to lower lipoprotein lipase activity, probably secondary to IH-up-regulation of angiopoietin-like protein 4 and decreased activity of the peroxisome proliferator-activated receptor alpha. Moreover, oxidative stress can generate dysfunctional oxidized lipids and reduce the capacity of high-density lipoproteins (HDL) to prevent low-density lipoprotein (LDL) oxidation. In the clinical field, several observational studies and a meta-regression analysis support the existence of a link between OSA and dyslipidemia. Although there is evidence of improved lipid profile after apnea-hypopnea suppression with continuous positive airway pressure (CPAP), the majority of the data come from observational studies. In contrast, randomized controlled trials evaluating the effects of CPAP on lipid metabolism present inconclusive results and two meta-analyses provide contradictory evidence.
Collapse
Affiliation(s)
- David Barros
- Servicio de Neumología, Hospital Montecelo, Pontevedra, Spain
| | - Francisco García-Río
- Servicio de Neumología, Hospital Universitario La Paz-IdiPAZ, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
13
|
Gioldasi S, Karvela A, Rojas-Gil AP, Rodi M, de Lastic AL, Thomas I, Spiliotis BE, Mouzaki A. Metabolic Association between Leptin and the Corticotropin Releasing Hormone. Endocr Metab Immune Disord Drug Targets 2020; 19:458-466. [PMID: 30727936 PMCID: PMC7360915 DOI: 10.2174/1871530319666190206165626] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 10/31/2018] [Accepted: 12/27/2018] [Indexed: 01/29/2023]
Abstract
Objective In healthy individuals, leptin is produced from adipose tissue and is secreted into the circulation to communicate energy balance status to the brain and control fat metabolism. Corticotropin-Releasing Hormone (CRH) is synthesized in the hypothalamus and regulates stress responses. Among the many adipokines and hormones that control fat metabolism, leptin and CRH both curb appetite and inhibit food intake. Despite numerous reports on leptin and CRH properties and function, little has been actually shown about their association in the adipose tissue environment. Methods In this article, we summarized the salient information on leptin and CRH in relation to metabolism. We also investigated the direct effect of recombinant CRH on leptin secretion by primary cultures of human adipocytes isolated from subcutaneous abdominal adipose tissue of 7 healthy children and adolescents, and measured CRH and leptin levels in plasma collected from peripheral blood of 24 healthy children and adolescents to assess whether a correlation exists between CRH and leptin levels in the periphery. Results and Conclusion The available data indicate that CRH exerts a role in the regulation of leptin in human adipocytes. We show that CRH downregulates leptin production by mature adipocytes and that a strong negative correlation exists between CRH and leptin levels in the periphery, and suggest the possible mechanisms of CRH control of leptin. Delineation of CRH control of leptin production by adipocytes may explain unknown pathogenic mechanisms linking stress and metabolism.
Collapse
Affiliation(s)
- Sofia Gioldasi
- Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| | - Alexia Karvela
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, Medical School, University of Patras, Patras, Greece
| | | | - Maria Rodi
- Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| | - Anne-Lise de Lastic
- Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| | - Iason Thomas
- Department of Allergy, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Bessie E Spiliotis
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, Medical School, University of Patras, Patras, Greece
| | - Athanasia Mouzaki
- Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| |
Collapse
|
14
|
Borg ML, Massart J, Schönke M, De Castro Barbosa T, Guo L, Wade M, Alsina-Fernandez J, Miles R, Ryan A, Bauer S, Coskun T, O'Farrell E, Niemeier EM, Chibalin AV, Krook A, Karlsson HK, Brozinick JT, Zierath JR. Modified UCN2 Peptide Acts as an Insulin Sensitizer in Skeletal Muscle of Obese Mice. Diabetes 2019; 68:1403-1414. [PMID: 31010957 DOI: 10.2337/db18-1237] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 04/08/2019] [Indexed: 11/13/2022]
Abstract
The neuropeptide urocortin 2 (UCN2) and its receptor corticotropin-releasing hormone receptor 2 (CRHR2) are highly expressed in skeletal muscle and play a role in regulating energy balance and glucose metabolism. We investigated a modified UCN2 peptide as a potential therapeutic agent for the treatment of obesity and insulin resistance, with a specific focus on skeletal muscle. High-fat-fed mice (C57BL/6J) were injected daily with a PEGylated UCN2 peptide (compound A) at 0.3 mg/kg subcutaneously for 14 days. Compound A reduced body weight, food intake, whole-body fat mass, and intramuscular triglycerides compared with vehicle-treated controls. Furthermore, whole-body glucose tolerance was improved by compound A treatment, with increased insulin-stimulated Akt phosphorylation at Ser473 and Thr308 in skeletal muscle, concomitant with increased glucose transport into extensor digitorum longus and gastrocnemius muscle. Mechanistically, this is linked to a direct effect on skeletal muscle because ex vivo exposure of soleus muscle from chow-fed lean mice to compound A increased glucose transport and insulin signaling. Moreover, exposure of GLUT4-Myc-labeled L6 myoblasts to compound A increased GLUT4 trafficking. Our results demonstrate that modified UCN2 peptides may be efficacious in the treatment of type 2 diabetes by acting as an insulin sensitizer in skeletal muscle.
Collapse
Affiliation(s)
- Melissa L Borg
- Department of Physiology and Pharmacology, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Julie Massart
- Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Milena Schönke
- Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Thais De Castro Barbosa
- Department of Physiology and Pharmacology, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Lili Guo
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, IN
| | - Mark Wade
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, IN
| | | | - Rebecca Miles
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, IN
| | - Andrew Ryan
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, IN
| | - Steve Bauer
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, IN
| | - Tamer Coskun
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, IN
| | - Elizabeth O'Farrell
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, IN
| | - Evan M Niemeier
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, IN
| | - Alexander V Chibalin
- Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Anna Krook
- Department of Physiology and Pharmacology, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Håkan K Karlsson
- Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Joseph T Brozinick
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, IN
| | - Juleen R Zierath
- Department of Physiology and Pharmacology, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
15
|
Kavalakatt S, Khadir A, Madhu D, Hammad M, Devarajan S, Abubaker J, Al-Mulla F, Tuomilehto J, Tiss A. Urocortin 3 Levels Are Impaired in Overweight Humans With and Without Type 2 Diabetes and Modulated by Exercise. Front Endocrinol (Lausanne) 2019; 10:762. [PMID: 31781037 PMCID: PMC6851015 DOI: 10.3389/fendo.2019.00762] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 10/21/2019] [Indexed: 12/26/2022] Open
Abstract
Urocortin3 (UCN3) regulates metabolic functions and is involved in cellular stress response. Although UCN3 is expressed in human adipose tissue, the association of UCN3 with obesity and diabetes remains unclear. This study investigated the effects of Type 2 diabetes (T2D) and increased body weight on the circulatory and subcutaneous adipose tissue (SAT) levels of UCN3 and assessed UCN3 modulation by a regular physical exercise. Normal-weight (n = 37) and overweight adults with and without T2D (n = 98 and n = 107, respectively) were enrolled in the study. A subset of the overweight subjects (n = 39 for each group) underwent a supervised 3-month exercise program combining both moderate intensity aerobic exercise and resistance training with treadmill. UCN3 levels in SAT were measured by immunofluorescence and RT-PCR. Circulatory UCN3 in plasma was assessed by ELISA and was correlated with various clinical and metabolic markers. Our data revealed that plasma UCN3 levels decreased in overweight subjects without T2D compared with normal-weight controls [median; 11.99 (0.78-86.07) and 6.27 (0.64-77.04), respectively; p < 0.001], whereas plasma UCN3 levels increased with concomitant T2D [median; 9.03 (0.77-104.92) p < 0.001]. UCN3 plasma levels were independently associated with glycemic index; fasting plasma glucose and hemoglobin A1c (r = 0.16 and r = 0.20, p < 0.05, respectively) and were significantly different between both overweight, with and without T2D, and normal-weight individuals (OR = 2.11 [1.84-4.11, 95% CI] and OR = 2.12 [1.59-3.10, 95% CI], p < 0.01, respectively). Conversely, the UCN3 patterns observed in SAT were opposite to those in circulation; UCN3 levels were significantly increased with body weight and decreased with T2D. After a 3-month supervised exercise protocol, UCN3 expression showed a significant reduction in SAT of both overweight groups (2.3 and 1.6-fold change; p < 0.01, respectively). In conclusion, UCN levels are differentially dysregulated in obesity in a tissue-dependent manner and can be mitigated by regular moderate physical exercise.
Collapse
Affiliation(s)
- Sina Kavalakatt
- Research Division, Biochemistry and Molecular Biology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Abdelkrim Khadir
- Research Division, Biochemistry and Molecular Biology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Dhanya Madhu
- Research Division, Biochemistry and Molecular Biology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Maha Hammad
- Research Division, Biochemistry and Molecular Biology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | | | - Jehad Abubaker
- Research Division, Biochemistry and Molecular Biology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Fahd Al-Mulla
- Research Division, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Jaakko Tuomilehto
- Research Division, Dasman Diabetes Institute, Kuwait City, Kuwait
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Department of Public Health Solutions, National Institute for Health and Welfare, Helsinki, Finland
| | - Ali Tiss
- Research Division, Biochemistry and Molecular Biology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
- *Correspondence: Ali Tiss
| |
Collapse
|
16
|
Gao SX, Guo J, Fan GQ, Qiao Y, Zhao RQ, Yang XJ. ZAG alleviates HFD-induced insulin resistance accompanied with decreased lipid depot in skeletal muscle in mice. J Lipid Res 2018; 59:2277-2286. [PMID: 30318474 PMCID: PMC6277152 DOI: 10.1194/jlr.m082180] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 09/24/2018] [Indexed: 12/25/2022] Open
Abstract
Over the past two decades, intramuscular lipids have been viewed as a cause of insulin resistance due to their ability to suppress insulin-stimulated glucose uptake in skeletal muscle. Zinc-α2-glycoprotein (ZAG) is an adipokine involved in lipolysis of white adipose tissue (WAT). To investigate the action of ZAG on insulin resistance induced by a high-fat diet (HFD), which affects the intramuscular fat, mice were divided into three groups, normal diet, HFD, and ZAG treatment under HFD (HFZ). The results showed that the insulin sensitivity of ZAG-treated mice was significantly improved. The body weight, WAT weight, and intramuscular fat were significantly decreased in the HFZ group compared with the HFD group. The lipolytic enzymes, including phosphorylation of hormone-sensitive lipase and adipose triglyceride lipase, were significantly upregulated in the skeletal muscle of mice that received the ZAG treatment compared with the HFD group. Insulin signaling proteins, such as phosphorylation of insulin receptor substrate 1 and cell membrane glucose transporter type 4, were also significantly increased in the skeletal muscle of the ZAG-treated group. Furthermore, a metabolic rate study showed that ZAG overexpression increases the respiratory exchange ratio and heat production. In vitro, ZAG treatment promotes glucose uptake and decreases intracellular lipids in C2C12 myotubes. Taken together, these data showed that overexpression of ZAG alleviates HFD-induced insulin resistance in mice, along with decreasing the lipid content of skeletal muscle.
Collapse
Affiliation(s)
- Shi-Xing Gao
- Key Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Jun Guo
- Key Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Guo-Qiang Fan
- Key Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Yu Qiao
- Key Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Ru-Qian Zhao
- Key Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Xiao-Jing Yang
- Key Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| |
Collapse
|
17
|
Adão R, Mendes-Ferreira P, Santos-Ribeiro D, Maia-Rocha C, Pimentel LD, Monteiro-Pinto C, Mulvaney EP, Reid HM, Kinsella BT, Potus F, Breuils-Bonnet S, Rademaker MT, Provencher S, Bonnet S, Leite-Moreira AF, Brás-Silva C. Urocortin-2 improves right ventricular function and attenuates pulmonary arterial hypertension. Cardiovasc Res 2018; 114:1165-1177. [DOI: 10.1093/cvr/cvy076] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 03/22/2018] [Indexed: 12/26/2022] Open
Abstract
Abstract
Aims
Pulmonary arterial hypertension (PAH) is a devastating disease and treatment options are limited. Urocortin-2 (Ucn-2) has shown promising therapeutic effects in experimental and clinical left ventricular heart failure (HF). Our aim was to analyse the expression of Ucn-2 in human and experimental PAH, and to investigate the effects of human Ucn-2 (hUcn-2) administration in rats with monocrotaline (MCT)-induced pulmonary hypertension (PH).
Methods and results
Tissue samples were collected from patients with and without PAH and from rats with MCT-induced PH. hUcn-2 (5 μg/kg, bi-daily, i.p., for 10 days) or vehicle was administered to male wistar rats subjected to MCT injection or to pulmonary artery banding (PAB) to induce right ventricular (RV) overload without PAH. Expression of Ucn-2 and its receptor was increased in the RV of patients and rats with PAH. hUcn-2 treatment reduced PAH in MCT rats, resulting in decreased morbidity, improved exercise capacity and attenuated pulmonary arterial and RV remodelling and dysfunction. Additionally, RV gene expression of hypertrophy and failure signalling pathways were attenuated. hUcn-2 treatment also attenuated PAB-induced RV hypertrophy.
Conclusions
Ucn-2 levels are altered in human and experimental PAH. hUcn-2 treatment attenuates PAH and RV dysfunction in MCT-induced PH, has direct anti-remodelling effects on the pressure-overloaded RV, and improves pulmonary vascular function.
Collapse
Affiliation(s)
- Rui Adão
- Department of Surgery and Physiology, Cardiovascular Research and Development Center - UnIC, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Pedro Mendes-Ferreira
- Department of Surgery and Physiology, Cardiovascular Research and Development Center - UnIC, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Diana Santos-Ribeiro
- Department of Surgery and Physiology, Cardiovascular Research and Development Center - UnIC, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Carolina Maia-Rocha
- Department of Surgery and Physiology, Cardiovascular Research and Development Center - UnIC, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Luís D Pimentel
- Department of Surgery and Physiology, Cardiovascular Research and Development Center - UnIC, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Cláudia Monteiro-Pinto
- Department of Surgery and Physiology, Cardiovascular Research and Development Center - UnIC, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Eamon P Mulvaney
- UCD Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin,Ireland
| | - Helen M Reid
- UCD Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin,Ireland
| | - B Therese Kinsella
- UCD Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin,Ireland
| | - François Potus
- Pulmonary Hypertension Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Laval University, Quebec City, Canada
| | - Sandra Breuils-Bonnet
- Pulmonary Hypertension Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Laval University, Quebec City, Canada
| | - Miriam T Rademaker
- Department of Medicine, Christchurch Heart Institute, University of Otago-Christchurch, Christchurch, New Zealand
| | - Steeve Provencher
- Pulmonary Hypertension Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Laval University, Quebec City, Canada
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Laval University, Quebec City, Canada
| | - Adelino F Leite-Moreira
- Department of Surgery and Physiology, Cardiovascular Research and Development Center - UnIC, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Carmen Brás-Silva
- Department of Surgery and Physiology, Cardiovascular Research and Development Center - UnIC, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- Faculty of Nutrition and Food Sciences, University of Porto, 4200-319 Porto, Portugal
| |
Collapse
|
18
|
Esnault S, Bernau K, Torr EE, Bochkov YA, Jarjour NN, Sandbo N. RNA-sequencing analysis of lung primary fibroblast response to eosinophil-degranulation products predicts downstream effects on inflammation, tissue remodeling and lipid metabolism. Respir Res 2017; 18:188. [PMID: 29126429 PMCID: PMC5681771 DOI: 10.1186/s12931-017-0669-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 10/30/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The association of eosinophils with inflammation and tissue remodeling is at least partially due to their release of toxic granule proteins and other mediators, including cytokines. Tissue remodeling and consequent functional defects are affected by activity of connective tissue fibroblasts. Exaggerated fibroblast activation, accumulation and change of phenotype may lead to fibrosis and loss of tissue function. So far, little information has been reported on how eosinophils affect inflammation and tissue remodeling via the activation of fibroblasts. We have recently shown that eosinophil activation with IL-3 led to a robust eosinophil degranulation on immunoglobin-G (IgG) coated plates. Thus, in the present study, we analyze the effects of IL-3-activated eosinophil degranulation products on primary human lung fibroblasts (HLF) using whole transcriptome sequencing. METHODS Conditioned media was obtained from eosinophils that were pre-activated with IL-3 or IL-5 and subsequently cultured for 6 h on IgG to induce degranulation. This conditioned media was added on human lung fibroblasts (HLF) for 24 h and the cell lysates were then subjected to whole transcriptome sequencing to identify global changes in gene expression. Differentially expressed genes were analyzed using the Ingenuity Pathway Analysis (IPA), and validated by qPCR. RESULTS In HLF, the expression level of 300 genes was changed by conditioned media from IL-3-activated eosinophils compared to control fibroblast cultures. Among these 300 genes, the expression level of 35 genes coding for known proteins was upregulated by IL-3- versus IL-5-pre-activated eosinophils. Of the 35 upregulated genes, IPA identified C3, CH25H, CXCL1, CXCL8, CYP1A1, ICAM1, IL6 and UCN2 as having downstream functions on inflammation, tissue remodeling and lipid synthesis. This analysis combined with previous RNA sequencing analyses of eosinophils suggest IL-1ß, OSM and TNFSF12 as potential upstream regulators of fibroblasts. CONCLUSIONS This study has identified several novel pro-inflammatory and pro-remodeling mediators produced by fibroblasts in response to activated eosinophils. These findings may have significant implications on the role of eosinophil/fibroblast interactions in eosinophilic disorders.
Collapse
Affiliation(s)
- Stephane Esnault
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, The University of Wisconsin-Madison School of Medicine and Public Health, K4/928 Clinical Science Center MC 9988, 600 Highland Avenue, Madison, WI, 53792, USA.
| | - Ksenija Bernau
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, The University of Wisconsin-Madison School of Medicine and Public Health, K4/928 Clinical Science Center MC 9988, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Elizabeth E Torr
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, The University of Wisconsin-Madison School of Medicine and Public Health, K4/928 Clinical Science Center MC 9988, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Yury A Bochkov
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53792, USA
| | - Nizar N Jarjour
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, The University of Wisconsin-Madison School of Medicine and Public Health, K4/928 Clinical Science Center MC 9988, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Nathan Sandbo
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, The University of Wisconsin-Madison School of Medicine and Public Health, K4/928 Clinical Science Center MC 9988, 600 Highland Avenue, Madison, WI, 53792, USA
| |
Collapse
|
19
|
Weiszenstein M, Shimoda LA, Koc M, Seda O, Polak J. Inhibition of Lipolysis Ameliorates Diabetic Phenotype in a Mouse Model of Obstructive Sleep Apnea. Am J Respir Cell Mol Biol 2017; 55:299-307. [PMID: 26978122 DOI: 10.1165/rcmb.2015-0315oc] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Obstructive sleep apnea (OSA) is associated with insulin resistance, glucose intolerance, and type 2 diabetes. Causal mechanisms mediating this association are not well defined; however, augmented lipolysis in adipose might be involved. Here, we investigated the effect of acipimox treatment (lipolysis inhibitor) on glucose tolerance and insulin sensitivity in mice exposed to intermittent hypoxia (IH). C57BL6/J mice were exposed for 14 days to IH or control conditions. IH was created by decreasing the fraction of inspired oxygen from 20.9 to 6.5%, 60 times/h. Control exposure was air (fraction of inspired oxygen, 20.9%) delivered at an identical flow rate. Acipimox was provided in drinking water (0.5 g/ml) during exposures. After exposures, intraperitoneal insulin (0.5 IU/kg) and glucose (1 g/kg) tolerance tests were performed, and primary adipocytes were isolated for lipolysis experiments. IH elevated fasting glucose by 51% and worsened glucose tolerance and insulin sensitivity by 33 and 102%, respectively. In parallel, IH increased spontaneous lipolysis by 264%, and reduced epididymal fat mass by 15% and adipocyte size by 8%. Acipimox treatment prevented IH-induced lipolysis and increased epididymal fat mass and adipocyte size by 19 and 10%, respectively. Acipimox fully prevented IH-induced impairments in fasting glycemia, glucose tolerance, and insulin sensitivity. For all reported results, P less than 0.05 was considered significant. Augmented lipolysis contributes to insulin resistance and glucose intolerance observed in mice exposed to IH. Acipimox treatment ameliorated the metabolic consequences of IH and might represent a novel treatment option for patients with obstructive sleep apnea.
Collapse
Affiliation(s)
- Martin Weiszenstein
- 1 Center for Research on Diabetes, Metabolism, and Nutrition, Third Faculty of Medicine.,2 Center of Toxicology and Health Safety, National Institute of Public Health, Prague, Czech Republic
| | - Larissa A Shimoda
- 3 Division of Pulmonary and Critical Care Medicine, Asthma and Allergy Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Michal Koc
- 4 Sports Medicine Department, Third Faculty of Medicine, and
| | - Ondrej Seda
- 5 Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic; and
| | - Jan Polak
- 1 Center for Research on Diabetes, Metabolism, and Nutrition, Third Faculty of Medicine.,2 Center of Toxicology and Health Safety, National Institute of Public Health, Prague, Czech Republic.,3 Division of Pulmonary and Critical Care Medicine, Asthma and Allergy Center, Johns Hopkins School of Medicine, Baltimore, Maryland.,6 Second Internal Medicine Department, Vinohrady Teaching Hospital, Prague, Czech Republic
| |
Collapse
|
20
|
Lefere S, Van Steenkiste C, Verhelst X, Van Vlierberghe H, Devisscher L, Geerts A. Hypoxia-regulated mechanisms in the pathogenesis of obesity and non-alcoholic fatty liver disease. Cell Mol Life Sci 2016; 73:3419-31. [PMID: 27091156 PMCID: PMC11108443 DOI: 10.1007/s00018-016-2222-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 04/05/2016] [Accepted: 04/07/2016] [Indexed: 02/06/2023]
Abstract
The pandemic rise in obesity has resulted in an increased incidence of metabolic complications. Non-alcoholic fatty liver disease is the hepatic manifestation of the metabolic syndrome and has become the most common chronic liver disease in large parts of the world. The adipose tissue expansion and hepatic fat accumulation characteristics of these disorders compromise local oxygen homeostasis. The resultant tissue hypoxia induces adaptive responses to restore oxygenation and tissue metabolism and cell survival. Hypoxia-inducible factors (HIFs) function as master regulators of this hypoxia adaptive response, and are in turn hydroxylated by prolyl hydroxylases (PHDs). PHDs are the main cellular oxygen sensors and regulate HIF proteasomal degradation in an oxygen-dependent manner. HIFs and PHDs are implicated in numerous physiological and pathological conditions. Extensive research using genetic models has revealed that hypoxia signaling is also a key mechanism in adipose tissue dysfunction, leading to adipose tissue fibrosis, inflammation and insulin resistance. Moreover, hypoxia affects liver lipid metabolism and deranges hepatic lipid accumulation. This review summarizes the molecular mechanisms through which the hypoxia adaptive response affects adipocyte and hepatic metabolism, and the therapeutic possibilities of modulating HIFs and PHDs in obesity and fatty liver disease.
Collapse
Affiliation(s)
- Sander Lefere
- Department of Gastroenterology and Hepatology, Ghent University Hospital, De Pintelaan 185, 1K12IE, 9000, Ghent, Belgium.
| | - Christophe Van Steenkiste
- Department of Gastroenterology and Hepatology, Ghent University Hospital, De Pintelaan 185, 1K12IE, 9000, Ghent, Belgium
- Department of Gastroenterology and Hepatology, Maria Middelares Hospital, Ghent, Belgium
| | - Xavier Verhelst
- Department of Gastroenterology and Hepatology, Ghent University Hospital, De Pintelaan 185, 1K12IE, 9000, Ghent, Belgium
| | - Hans Van Vlierberghe
- Department of Gastroenterology and Hepatology, Ghent University Hospital, De Pintelaan 185, 1K12IE, 9000, Ghent, Belgium
| | - Lindsey Devisscher
- Department of Gastroenterology and Hepatology, Ghent University Hospital, De Pintelaan 185, 1K12IE, 9000, Ghent, Belgium
| | - Anja Geerts
- Department of Gastroenterology and Hepatology, Ghent University Hospital, De Pintelaan 185, 1K12IE, 9000, Ghent, Belgium
| |
Collapse
|
21
|
Bozec A, Hannemann N. Mechanism of Regulation of Adipocyte Numbers in Adult Organisms Through Differentiation and Apoptosis Homeostasis. J Vis Exp 2016. [PMID: 27284940 PMCID: PMC4927764 DOI: 10.3791/53822] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Considering that adipose tissue (AT) is an endocrine organ, it can influence whole body metabolism. Excessive energy storage leads to the dysregulation of adipocytes, which in turn induces abnormal secretion of adipokines, triggering metabolic syndromes such as obesity, dyslipidemia, hyperglycemia, hyperinsulinemia, insulin resistance and type 2 diabetes. Therefore, investigating the molecular mechanisms behind adipocyte dysregulation could help to develop novel therapeutic strategies. Our protocol describes methods for evaluating the molecular mechanism affected by hypoxic conditions of the AT, which correlates with adipocyte apoptosis in adult mice. This protocol describes how to analyze AT in vivo through gene expression profiling as well as histological analysis of adipocyte differentiation, proliferation and apoptosis during hypoxia exposure, ascertained through staining of hypoxic cells or HIF-1α protein. Furthermore, in vitro analysis of adipocyte differentiation and its responses to various stimuli completes the characterization of the molecular pathways behind possible adipocyte dysfunction leading to metabolic syndromes.
Collapse
Affiliation(s)
- Aline Bozec
- Department of Medicine 3, Rheumatology and Immunology, Universitätsklinikum Erlangen; Nikolaus Fiebiger Center of Molecular Medicine, Universitätsklinikum Erlangen;
| | - Nicole Hannemann
- Department of Medicine 3, Rheumatology and Immunology, Universitätsklinikum Erlangen; Nikolaus Fiebiger Center of Molecular Medicine, Universitätsklinikum Erlangen
| |
Collapse
|
22
|
Yoo JM, Lee M, Kwon HO, Choi SG, Bae MH, Kim OK. Effects of Sinetrol-XPur on Leptin-Deficient Obese Mice and Activation of cAMP-Dependent UCP-2. ACTA ACUST UNITED AC 2016. [DOI: 10.3746/jkfn.2016.45.4.484] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|