1
|
Malta A, Matiusso CCI, de Lima LDS, Ribeiro TA, Tófolo LP, Almeida DL, Moreira VM, Martins IP, Pavanello A, de Freitas Mathias PC. Early exposure to a cholinergic receptor blocking agent mitigates adult obesity and protects pancreatic islet function in male rats. Am J Physiol Endocrinol Metab 2025; 328:E34-E43. [PMID: 39535172 DOI: 10.1152/ajpendo.00191.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
We tested the hypothesis that attenuation of the circulating insulin level in rats during early life can provide sustained protection against diet-induced obesity and metabolic dysfunction in adulthood. Male Wistar rats received intraperitoneal scopolamine butylbromide (SB) during the first 12 days of suckling, whereas control rats received 0.9% saline injections. The animals were weaned on day 21 and fed a normal chow diet. At 60 days of age, the control and SB groups were fed a normal chow diet (ND; 4.5% fat) or a high-fat diet (HF; 35% fat) until 90 days of age to induce obesity and metabolic dysfunction. Insulin secretion, food intake, and body weight were measured. Pancreatic islet function, autonomic nervous system function, and glucose homeostasis were evaluated at 90 days of age. During lactation, the plasma insulin concentration was significantly lower in the SB groups than in the control group. SB rats also exhibited reduced body weight. The HF diet resulted in obesity, glucose intolerance, insulin resistance, disruption of insulin secretion, and vagal hyperactivity in adult control rats. Remarkably, SB-treated rats fed the HF diet showed attenuated body weight and adiposity and did not develop diet-induced glucose/insulin imbalance. In addition, vagal activity and adequate pancreatic islet insulin secretion were preserved. Offspring exposed to SB during early life are provided with long-lasting protection against obesity and metabolic complications induced by an HF diet. An attenuated circulating insulin level in early life may have far-reaching consequences on metabolic programming.NEW & NOTEWORTHY High insulin levels during early life may lead to the late development of obesity and diabetes. We investigated whether attenuation of insulin levels by using an antimuscarinic agent could prevent diet-induced obesity and diabetes. Rats' early exposure to an antimuscarinic agent reduced insulin levels during the lactation period and promoted protection against obesity and metabolic dysfunctions. Independent of the programming mechanisms, insulin levels during early life may be a defining factor of health or diseases later in life.
Collapse
Affiliation(s)
- Ananda Malta
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringa, Maringa, Brazil
| | | | - Lucas da Silva de Lima
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringa, Maringa, Brazil
| | - Tatiane Aparecida Ribeiro
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringa, Maringa, Brazil
- Department of Biochemistry and Biomedical Science, McMaster University, Hamilton, Ontario, Canada
| | - Laize Peron Tófolo
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringa, Maringa, Brazil
- Physical Education Department, Paraná State University, Paranavai, Brazil
| | - Douglas Lopes Almeida
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringa, Maringa, Brazil
| | - Veridiana Mota Moreira
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringa, Maringa, Brazil
| | - Isabela Peixoto Martins
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringa, Maringa, Brazil
- Health Sciences Center, Unicesumar, Maringa, Brazil
| | - Audrei Pavanello
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringa, Maringa, Brazil
- Health Sciences Center, Unicesumar, Maringa, Brazil
| | | |
Collapse
|
2
|
Janssen JAMJL. The Causal Role of Ectopic Fat Deposition in the Pathogenesis of Metabolic Syndrome. Int J Mol Sci 2024; 25:13238. [PMID: 39769002 PMCID: PMC11675790 DOI: 10.3390/ijms252413238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/03/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
Consuming a "modern" Western diet and overnutrition may increase insulin secretion. Additionally, nutrition-mediated hyperinsulinemia is a major driver of ectopic fat deposition. The global prevalence of metabolic syndrome is high and growing. Within this context, people with congenital lipodystrophy often experience a severe form of metabolic syndrome. Evidence is increasingly supporting that subtle partial lipodystrophy plays an important role in the development of metabolic syndrome in the general population. In individuals in the general population with subtle partial lipodystrophy, as well as in those with congenital lipodystrophy, the subcutaneous adipose tissues are unable to accommodate surplus energy intake. In both conditions, (excess) fat is directed toward the liver, pancreas, and muscles, where it is deposited as ectopic fat, as this fat can no longer be stored in the "safe" subcutaneous fat depots. Ectopic fat depositions cause insulin resistance in the liver and muscles, as well as β-cell dysfunction in the pancreas. Support of a direct pathological role of ectopic fat deposition in this condition is further provided by the rapid normalization of hepatic insulin sensitivity and improvement in pancreatic β-cell function after marked reductions in ectopic fat depositions. Thus, ectopic fat deposition in the liver, pancreas, and muscles may play a causal role in the pathogenesis of metabolic syndrome even in the general population. As such, the prevention of ectopic fat deposition may reduce the risk of metabolic syndrome and mitigate its effects.
Collapse
Affiliation(s)
- Joseph A M J L Janssen
- Department of Internal Medicine, Erasmus Medical Center (Erasmus MC), Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
3
|
Al Harake SN, Abedin Y, Hatoum F, Nassar NZ, Ali A, Nassar A, Kanaan A, Bazzi S, Azar S, Harb F, Ghadieh HE. Involvement of a battery of investigated genes in lipid droplet pathophysiology and associated comorbidities. Adipocyte 2024; 13:2403380. [PMID: 39329369 PMCID: PMC11445895 DOI: 10.1080/21623945.2024.2403380] [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: 12/07/2023] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
Lipid droplets (LDs) are highly specialized energy storage organelles involved in the maintenance of lipid homoeostasis by regulating lipid flux within white adipose tissue (WAT). The physiological function of adipocytes and LDs can be compromised by mutations in several genes, leading to NEFA-induced lipotoxicity, which ultimately manifests as metabolic complications, predominantly in the form of dyslipidemia, ectopic fat accumulation, and insulin resistance. In this review, we delineate the effects of mutations and deficiencies in genes - CIDEC, PPARG, BSCL2, AGPAT2, PLIN1, LIPE, LMNA, CAV1, CEACAM1, and INSR - involved in lipid droplet metabolism and their associated pathophysiological impairments, highlighting their roles in the development of lipodystrophies and metabolic dysfunction.
Collapse
Affiliation(s)
- Sami N. Al Harake
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Yasamin Abedin
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Fatema Hatoum
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Nour Zahraa Nassar
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Ali Ali
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Aline Nassar
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Amjad Kanaan
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Samer Bazzi
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Sami Azar
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Frederic Harb
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Hilda E. Ghadieh
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| |
Collapse
|
4
|
Nguyen MLT, Bui KC, Ngo TH, Nguyen TS, Nham PLT, Pham C, Hoang TM, Huynh QT, Tang TT, Dang TC, Dang TL, Nguyen LT, Can VM. Early impaired insulin tolerance among Vietnamese diabetes with or without dyslipidemia. Medicine (Baltimore) 2024; 103:e40212. [PMID: 39495996 PMCID: PMC11537581 DOI: 10.1097/md.0000000000040212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 10/04/2024] [Indexed: 11/06/2024] Open
Abstract
This study aims to evaluate impaired insulin tolerance among Vietnamese diabetes with or without dyslipidemia. Diabetes mellitus (DM) remains the serious global health and social burden that has increased over the past few decades. It progresses silently to vascular injury and disability of injured vascular-perfused tissues/organs. Insulin intolerance and dyslipidemia exacerbate and accelerate the implications of DM. Thus, early detection and more evidence of early insulin intolerance and dyslipidemia is needed for proactive management. This cross-sectional descriptive study recruited 100 healthy control (HC) and 297 DM patients in Military Hospital 103 from 2021 to 2023. Patients with DM were subgrouped into lipid metabolism disorder (LMD, n = 98) and non-LMD (NLMD, n = 99). The biochemists' serum levels were measured automatically and the accuracy of the test result was strictly controlled. Insulin tolerance indices (HOMA2-IR, HOMA2-%S and HOMA2-%B) were compared between HC, DM with or without dyslipidemia as well as correlated with lipid ingredients (total Cholesterol, triglyceride, LDL-C and HDL-C). Among DM patients, HOMA2-IR was significantly high and HOMA2-%S and HOMA2-%B were significantly low. HOMA2-IR was higher and HOMA2-%S and HOMA2-%B were lower in DM with LMD than in DM without LMD. In addition, HOMA2-IR was positively correlated with serum cholesterol, triglyceride and LDL-C concentration, and negatively correlated to HDL-C concentration. In contrast, HOMA2-%S and HOMA2-%B was negatively correlated with serum cholesterol, triglyceride and LDL-C, and positively correlated with HDL-C. Impaired insulin intolerance occurred in early stage of DM, and more serious among DM with LMD, compared to DM with NLMD.
Collapse
Affiliation(s)
- Mai Ly Thi Nguyen
- Department of Biochemistry, Military Hospital 103, Vietnam Military Medical University Hanoi, Vietnam
| | - Khac Cuong Bui
- Laboratory Animal Research Center, Vietnam Military Medical University, Hanoi, Vietnam
- Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam
- Vietnamese-German Center for Medical Research (VG-CARE), 108 Military Central Hospital, Hanoi, Vietnam
| | - Thu Hang Ngo
- Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam
| | - Tai Suc Nguyen
- Laboratory Animal Research Center, Vietnam Military Medical University, Hanoi, Vietnam
| | - Phuong Linh Thi Nham
- Laboratory Animal Research Center, Vietnam Military Medical University, Hanoi, Vietnam
| | - Chi Pham
- Laboratory Animal Research Center, Vietnam Military Medical University, Hanoi, Vietnam
| | - Thi Minh Hoang
- Department of Biochemistry, Military Hospital 103, Vietnam Military Medical University Hanoi, Vietnam
| | - Quang Thuan Huynh
- Department of Biochemistry, Military Hospital 103, Vietnam Military Medical University Hanoi, Vietnam
| | - Thanh Thuy Tang
- Department of Biochemistry, Military Hospital 103, Vietnam Military Medical University Hanoi, Vietnam
| | - Thanh Chung Dang
- Department of Pathology, Military Hospital 103, Vietnam Military Medical University Hanoi, Vietnam
| | - Thuy Linh Dang
- Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam
| | - Linh Toan Nguyen
- Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam
- Vietnamese-German Center for Medical Research (VG-CARE), 108 Military Central Hospital, Hanoi, Vietnam
| | - Van Mao Can
- Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam
| |
Collapse
|
5
|
Cunningham CR, Mehrsheikh AL, Aswani Y, Shetty AS, Itani M, Ballard DH, Khot R, Moshiri M, Picard MM, Northrup BE. Off the wall: incidental paraspinal and pelvic muscle pathology on abdominopelvic imaging. Abdom Radiol (NY) 2024; 49:4016-4041. [PMID: 38831073 DOI: 10.1007/s00261-024-04365-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 04/27/2024] [Accepted: 04/28/2024] [Indexed: 06/05/2024]
Abstract
As the use of cross-sectional abdominal and pelvic imaging has increased exponentially in the past several decades, incidental musculoskeletal findings have become commonplace. These are often unrelated to the indication for the examination and are frequently referred to as the "radiologist's blind spot" on these studies. The differential diagnosis for abnormalities of the paraspinal and pelvic musculature is, in many cases, quite different from the anterior abdominal wall muscles. Furthermore, due to their relatively deep location, pathology involving the former muscle groups is more likely to be clinically occult, often presenting only incidentally when the patient undergoes cross-sectional imaging. Effective treatment of diseases of these muscles is dependent on adherence to a diverse set of diagnostic and treatment algorithms. The purpose of this review article is to familiarize the radiologist with the unique pathology of these often-overlooked muscles of the abdomen and pelvis.
Collapse
Affiliation(s)
- Christopher R Cunningham
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, 510 S. Kingshighway Blvd., Campus Box 8131, St. Louis, MO, 63110, USA
| | - Amanda L Mehrsheikh
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, 510 S. Kingshighway Blvd., Campus Box 8131, St. Louis, MO, 63110, USA
| | - Yashant Aswani
- Department of Radiology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Anup S Shetty
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, 510 S. Kingshighway Blvd., Campus Box 8131, St. Louis, MO, 63110, USA
| | - Malak Itani
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, 510 S. Kingshighway Blvd., Campus Box 8131, St. Louis, MO, 63110, USA
| | - David H Ballard
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, 510 S. Kingshighway Blvd., Campus Box 8131, St. Louis, MO, 63110, USA
| | - Rachita Khot
- Department of Radiology and Medical Imaging, University of Virginia Health System, Charlottesville, VA, USA
| | - Mariam Moshiri
- Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC, USA
| | - Melissa M Picard
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Benjamin E Northrup
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, 510 S. Kingshighway Blvd., Campus Box 8131, St. Louis, MO, 63110, USA.
| |
Collapse
|
6
|
Zheng H, Bai Y, Wu S, Jiang Z, Pei Q, Yao M. Transcriptome dataset of mouse adipose tissue across estrous cycles. Sci Data 2024; 11:1090. [PMID: 39368986 PMCID: PMC11455834 DOI: 10.1038/s41597-024-03942-5] [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: 06/20/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024] Open
Abstract
Adipose tissue is crucial for energy storage and release, ensuring energy homeostasis within the body. Disturbances in the physiology of adipose tissue have been associated with various health disorders, such as obesity and diabetes. The reproductive cycle represents a fundamental biological pattern in female physiology. Although previous research has highlighted the substantial regulatory influence of ovarian hormones on adipose tissue, our understanding of the comprehensive changes in adipose tissue throughout the reproductive cycle remains limited. In this study, we examined the transcriptomic profile of female mouse-adipose tissue across their complete estrous cycles. The findings provided detailed descriptions of the datasets generated, including information on data collection, processing, and quality control. The study also demonstrated the robustness of these data through various validation steps. These findings serve as crucial resources for investigating the role of estrous cycle rhythmicity in important adipose tissue processes in the future.
Collapse
Affiliation(s)
- Hongjie Zheng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Yier Bai
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Shan Wu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Zhixuan Jiang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Qing Pei
- Department of Burn, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.
| | - Min Yao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China.
| |
Collapse
|
7
|
Chiveto DT, Musarurwa C, Mapira HT, Kaseke F, Nyengerai T, Kaseke T, Gori E. Glycemic Control and Cardiometabolic Risk in Black Zimbabweans with Type 2 Diabetes Mellitus. Diabetes Metab Syndr Obes 2024; 17:3187-3196. [PMID: 39220799 PMCID: PMC11365488 DOI: 10.2147/dmso.s473042] [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: 04/27/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
Purpose Type 2 diabetes mellitus (T2DM) frequently presents with modified cardiometabolic risk profiles, indicative of an elevated susceptibility to cardiovascular disease (CVD). Cardiometabolic risk factors such as obesity, hyperglycemia, hypertension, insulin resistance and dyslipidemia are known contributors to increased CVD hazard in individuals with T2DM. This study evaluated the glycemic control-based cardiometabolic risk profiles of black Zimbabweans with T2DM. Patients and Methods A cross-sectional study of 116 T2DM patients recruited from diabetic clinics at Parirenyatwa and Sally Mugabe Hospitals, Harare, Zimbabwe, was conducted. Blood samples were collected for glycated hemoglobin (HbA1c) and lipid profile assessment. The Framingham risk scores (FRS) based on body mass index (BMI) and lipid profile were used to determine CVD risk. Parametric variables were analyzed using one-way analysis of variance (ANOVA) with post hoc Bonferroni correction, while non-parametric variables were compared using the Kruskal-Wallis test with post hoc Dunn test for multiple comparisons. Results The overall frequency of dyslipidemia was 83.6% (n=97) and hypoalphalipoproteinemia was the most prevalent dyslipidemia (79.3%). Median HDLC levels were significantly lower in participants with poor glycemic control (1.12 mmol/L) compared to those with good glycemic control group (1.37 mmol/L) (p=0.011). Despite lack of significant variations in Framingham Risk Scores, there was a trend towards lower FRS-BMI in the good control group (29.8%) compared to the inadequate control (35.4%) and poor control (32.7%) groups (p=0.078). Conclusion Duration since DM diagnosis was observed to be an important risk factor for poor glycemic control being significantly shorter in those with good glycemic control compared to those with inadequate and poor control. Overall, there was no significant difference in HbA1c status by age but individuals with poor glycemic control were significantly older than those with good control. The most prevalent dyslipidemia among the study participants was hypoalphalipoproteinemia which is reportedly associated with genetic predisposition, warranting further investigations.
Collapse
Affiliation(s)
- Dexter Tadiwanashe Chiveto
- Department of Laboratory Diagnostic and Investigative Sciences - Chemical Pathology Unit, Faculty of Medicine and Health Sciences, University of Zimbabwe, Harare, Zimbabwe
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Cuthbert Musarurwa
- Department of Biomedical Laboratory Sciences, School of Health Sciences, College of Medicine and Health Sciences, University of Rwanda, Kigali, Rwanda
| | - Herbert T Mapira
- Department of Biomedical Laboratory Sciences, School of Health Sciences, College of Medicine and Health Sciences, University of Rwanda, Kigali, Rwanda
| | - Farayi Kaseke
- Department of Physiotherapy, School of Health Sciences, College of Medicine and Health Sciences, University of Rwanda, Kigali, Rwanda
| | | | - Timothy Kaseke
- Zimbabwe Aids Prevention Project, ZAPP, Harare, Zimbabwe
| | - Elizabeth Gori
- Department of Medical Biochemistry, Molecular Biology and Genetics, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Huye, Rwanda
| |
Collapse
|
8
|
Deng H, Rao X, Zhang S, Chen L, Zong Y, Zhou R, Meng R, Dong X, Wu G, Li Q. Protein kinase CK2: An emerging regulator of cellular metabolism. Biofactors 2024; 50:624-633. [PMID: 38158592 DOI: 10.1002/biof.2032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/02/2023] [Indexed: 01/03/2024]
Abstract
The protein kinase casein kinase 2 (CK2) exerts its influence on the metabolism of three major cellular substances by phosphorylating essential protein molecules involved in various cellular metabolic pathways. These substances include hormones, especially insulin, rate-limiting enzymes, transcription factors of key genes, and cytokines. This regulatory role of CK2 is closely tied to important cellular processes such as cell proliferation and apoptosis. Additionally, tumor cells undergo metabolic reprogramming characterized by aerobic glycolysis, accelerated lipid β-oxidation, and abnormally active glutamine metabolism. In this context, CK2, which is overexpressed in various tumors, also plays a pivotal role. Hence, this review aims to summarize the regulatory mechanisms of CK2 in diverse metabolic pathways and tumor development, providing novel insights for the diagnosis, treatment, and prognosis of metabolism-related diseases and cancers.
Collapse
Affiliation(s)
- Huilin Deng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinrui Rao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sijia Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Leichong Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Zong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Zhou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Meng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaorong Dong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qianwen Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
9
|
Guo PP, Yao XR, Xu YN, Jin X, Li Q, Yan CG, Kim NH, Li XZ. Insulin interacts with PPARγ agonists to promote bovine adipocyte differentiation. Domest Anim Endocrinol 2024; 88:106848. [PMID: 38574690 DOI: 10.1016/j.domaniend.2024.106848] [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: 01/11/2024] [Revised: 02/29/2024] [Accepted: 03/19/2024] [Indexed: 04/06/2024]
Abstract
Insulin is a potent adipogenic hormone that triggers a series of transcription factors that regulate the differentiation of preadipocytes into mature adipocytes. Ciglitazone specifically binds to peroxisome proliferator-activated receptor-γ (PPARγ), thereby promoting adipocyte differentiation. As a natural ligand of PPARγ, oleic acid (OA) can promote the translocation of PPARγ into the nucleus, regulate the expression of downstream genes, and promote adipocyte differentiation. We hypothesized that ciglitazone and oleic acid interact with insulin to enhance bovine preadipocyte differentiation. Preadipocytes were cultured 96 h in differentiation medium containing 10 mg/L insulin (I), 10 mg/L insulin + 10 µM cycloglitazone (IC), 10 mg/L insulin + 100 µM oleic acid (IO), or 10 mg/L insulin + 10 µM cycloglitazone+100 µM oleic acid (ICO). Control preadipocytes (CON) were cultured in differentiation medium (containing 5% fetal calf serum). The effects on the differentiation of Yanbian cattle preadipocytes were examined using molecular and transcriptomic techniques, including differentially expressed genes (DEGs) and Kyoto Encyclopaedia of Genes and Genomes (KEGG) pathway analysis. I, IC, IO, and ICO treatments produced higher concentrations of triglycerides (TAG) and lipid droplet accumulation in preadipocytes compared with CON treatment (P < 0.05). Co-treatment of insulin and PPARγ agonists significantly increased the expression of genes involved in regulating adipogenesis and fatty acid synthesis. (P < 0.05). Differential expression analysis identified 1488, 1764, 1974 and 1368 DEGs in the I, IC, IO and ICO groups, respectively. KEGG pathway analysis revealed DEGs mainly enriched in PPAR signalling, FOXO signaling pathway and fatty acid metabolism. These results indicate that OA, as PPARγ agonist, can more effectively promote the expression of bovine lipogenesis genes and the content of TAG and adiponectin when working together with insulin, and stimulate the differentiation of bovine preadipocytes. These findings provide a basis for further screening of relevant genes and transcription factors in intramuscular fat deposition and meat quality to enhance breeding programs.
Collapse
Affiliation(s)
- Pan Pan Guo
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, PR China; Healthcare International Innovation Institute, Jiangmen 529020, PR China; Guangdong University of Technology, Guangzhou 510000, PR China; Engineering Research Centre of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Department of Animal Science, College of Agriculture, Yanbian University, Yanji 133002, PR China
| | - Xue Rui Yao
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, PR China; Healthcare International Innovation Institute, Jiangmen 529020, PR China; Guangdong University of Technology, Guangzhou 510000, PR China
| | - Yong Nan Xu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, PR China
| | - Xin Jin
- Engineering Research Centre of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Department of Animal Science, College of Agriculture, Yanbian University, Yanji 133002, PR China; Laboratory Animal Center, Yanbian University, Yanji 133002, PR China
| | - Qiang Li
- Engineering Research Centre of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Department of Animal Science, College of Agriculture, Yanbian University, Yanji 133002, PR China
| | - Chang Guo Yan
- Engineering Research Centre of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Department of Animal Science, College of Agriculture, Yanbian University, Yanji 133002, PR China; Yanbian Hongchao Wisdom Animal Husbandry Co., LTD, Yanji 133002, PR China
| | - Nam Hyung Kim
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, PR China
| | - Xiang Zi Li
- Engineering Research Centre of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Department of Animal Science, College of Agriculture, Yanbian University, Yanji 133002, PR China.
| |
Collapse
|
10
|
Gong W, Chen W, Dong J, Liao L. Rabson-Mendenhall Syndrome: Analysis of the Clinical Characteristics and Gene Mutations in 42 Patients. J Endocr Soc 2024; 8:bvae123. [PMID: 38957655 PMCID: PMC11216847 DOI: 10.1210/jendso/bvae123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Indexed: 07/04/2024] Open
Abstract
Aims Rabson-Mendenhall syndrome (RMS) is a rare autosomal, recessive disorder characterized by severe insulin resistance due to mutations in the insulin receptor (INSR) gene. This study aims to analyze the clinical features and gene mutations in RMS, which have not been extensively studied. Methods PubMed, Embase, the China National Knowledge Infrastructure, and Wanfang were searched for "Rabson-Mendenhall syndrome" or "Black acanthosis hirsutism insulin resistance syndrome." Results A total of 42 cases from 33 articles were included. The body mass index ranged from 18.50 to 20.00 kg/m2 with an average of 16.00 kg/m2. There were no overweight (25.00∼29.90 kg/m2) or obese (≥30.00 kg/m2) patients. Acanthosis was present in 29 cases (29/42, 69.05%); growth retardation in 25 cases (25/42, 59.52%); dental anomalies including absence of teeth, crowding, and malocclusion in 23 cases (23/42, 54.76%); and hirsutism in 17 cases (17/42, 40.48%). The average glycosylated hemoglobin was 9.35%, and the average fasting blood-glucose was 8.44 mmol/L; the mean fasting insulin was 349.96 μIU/mL, and the average fasting C-peptide was 6.00 ng/mL. Diabetes was reported in 25 cases (25/33, 75.76%) all of which were diagnosed before 23 years old. All 42 patients had recorded gene mutations, with 22 patients (22/42, 52.38%) having ≥ 2 mutations and 20 cases (20/42, 47.62%) having only 1 mutation. No statistical differences were found in clinical features and laboratory parameters between patients with different mutations. Conclusion The study indicates that RMS should be considered in young patients with hyperinsulinemia, hyperglycemia with low weight, acanthosis nigricans, growth retardation, dental anomalies, and hirsutism.
Collapse
Affiliation(s)
- Wenfeng Gong
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Jinan, 250014, China
| | - Wenzhe Chen
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Jinan, 250014, China
| | - Jianjun Dong
- Division of Endocrinology, Department of Internal Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Lin Liao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Jinan, 250014, China
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Jinan, 250014, China
| |
Collapse
|
11
|
Cortés-Camacho F, Zambrano-Vásquez OR, Aréchaga-Ocampo E, Castañeda-Sánchez JI, Gonzaga-Sánchez JG, Sánchez-Gloria JL, Sánchez-Lozada LG, Osorio-Alonso H. Sodium-Glucose Cotransporter Inhibitors: Cellular Mechanisms Involved in the Lipid Metabolism and the Treatment of Chronic Kidney Disease Associated with Metabolic Syndrome. Antioxidants (Basel) 2024; 13:768. [PMID: 39061837 PMCID: PMC11274291 DOI: 10.3390/antiox13070768] [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: 05/09/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Metabolic syndrome (MetS) is a multifactorial condition that significantly increases the risk of cardiovascular disease and chronic kidney disease (CKD). Recent studies have emphasized the role of lipid dysregulation in activating cellular mechanisms that contribute to CKD progression in the context of MetS. Sodium-glucose cotransporter 2 inhibitors (SGLT2i) have demonstrated efficacy in improving various components of MetS, including obesity, dyslipidemia, and insulin resistance. While SGLT2i have shown cardioprotective benefits, the underlying cellular mechanisms in MetS and CKD remain poorly studied. Therefore, this review aims to elucidate the cellular mechanisms by which SGLT2i modulate lipid metabolism and their impact on insulin resistance, mitochondrial dysfunction, oxidative stress, and CKD progression. We also explore the potential benefits of combining SGLT2i with other antidiabetic drugs. By examining the beneficial effects, molecular targets, and cytoprotective mechanisms of both natural and synthetic SGLT2i, this review provides a comprehensive understanding of their therapeutic potential in managing MetS-induced CKD. The information presented here highlights the significance of SGLT2i in addressing the complex interplay between metabolic dysregulation, lipid metabolism dysfunction, and renal impairment, offering clinicians and researchers a valuable resource for developing improved treatment strategies and personalized approaches for patients with MetS and CKD.
Collapse
Affiliation(s)
- Fernando Cortés-Camacho
- Doctorado en Ciencias Biologicas y de la Salud, Universidad Autónoma Metropolitana, Mexico City 04960, Mexico; (F.C.-C.); (O.R.Z.-V.)
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico; (J.G.G.-S.); (L.G.S.-L.)
| | - Oscar René Zambrano-Vásquez
- Doctorado en Ciencias Biologicas y de la Salud, Universidad Autónoma Metropolitana, Mexico City 04960, Mexico; (F.C.-C.); (O.R.Z.-V.)
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico; (J.G.G.-S.); (L.G.S.-L.)
| | - Elena Aréchaga-Ocampo
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana, Unidad Cuajimalpa, Mexico City 05348, Mexico;
| | | | - José Guillermo Gonzaga-Sánchez
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico; (J.G.G.-S.); (L.G.S.-L.)
| | - José Luis Sánchez-Gloria
- Department of Internal Medicine, Division of Nephrology, Rush University Medical Center, Chicago, IL 60612, USA;
| | - Laura Gabriela Sánchez-Lozada
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico; (J.G.G.-S.); (L.G.S.-L.)
| | - Horacio Osorio-Alonso
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico; (J.G.G.-S.); (L.G.S.-L.)
| |
Collapse
|
12
|
Kaczmarek I, Suchý T, Strnadová M, Thor D. Qualitative and quantitative analysis of lipid droplets in mature 3T3-L1 adipocytes using oil red O. STAR Protoc 2024; 5:102977. [PMID: 38875117 PMCID: PMC11225905 DOI: 10.1016/j.xpro.2024.102977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/31/2024] [Accepted: 03/08/2024] [Indexed: 06/16/2024] Open
Abstract
By differentiating into mature adipocytes, 3T3-L1 cells can be utilized as a model cell line to investigate (pre)adipocyte function in vitro. Here, we present a protocol for combining qualitative and quantitative analysis of lipid droplets in mature 3T3-L1 adipocytes using oil red O. We describe steps to differentiate 3T3-L1 preadipocytes to adipocytes and give detailed procedures to determine total lipid amount as well as lipid droplet size and number using microscopic devices and an ImageJ macro. For complete details on the use and execution of this protocol, please refer to Kaczmarek et al.1.
Collapse
Affiliation(s)
- Isabell Kaczmarek
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, 04103 Saxony, Germany.
| | - Tomáš Suchý
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, 04103 Saxony, Germany
| | - Martina Strnadová
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, 04103 Saxony, Germany
| | - Doreen Thor
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, 04103 Saxony, Germany.
| |
Collapse
|
13
|
Strnadová M, Thor D, Kaczmarek I. Protocol for changing gene expression in 3T3-L1 (pre)adipocytes using siRNA-mediated knockdown. STAR Protoc 2024; 5:103075. [PMID: 38805394 PMCID: PMC11153903 DOI: 10.1016/j.xpro.2024.103075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 03/21/2024] [Accepted: 04/26/2024] [Indexed: 05/30/2024] Open
Abstract
3T3-L1 is a model cell line which can be differentiated from preadipocytes into mature adipocytes. Here, we present a protocol for changing gene expression in 3T3-L1 (pre)adipocytes using small interfering RNA (siRNA)-mediated knockdown. We describe steps to perform the knockdown of a certain gene prior to differentiation (day 4) to analyze the impact on adipogenesis. We then detail procedures for knockdown on day 8 of differentiation to study the role of a certain gene in mature adipocyte function. For complete details on the use and execution of this protocol, please refer to Kaczmarek et al.1.
Collapse
Affiliation(s)
- Martina Strnadová
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, 04103 Saxony, Germany
| | - Doreen Thor
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, 04103 Saxony, Germany.
| | - Isabell Kaczmarek
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, 04103 Saxony, Germany.
| |
Collapse
|
14
|
Ngema M, Xulu ND, Ngubane PS, Khathi A. A Review of Fetal Development in Pregnancies with Maternal Type 2 Diabetes Mellitus (T2DM)-Associated Hypothalamic-Pituitary-Adrenal (HPA) Axis Dysregulation: Possible Links to Pregestational Prediabetes. Biomedicines 2024; 12:1372. [PMID: 38927579 PMCID: PMC11201628 DOI: 10.3390/biomedicines12061372] [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: 04/30/2024] [Revised: 06/15/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Research has identified fetal risk factors for adult diseases, forming the basis for the Developmental Origins of Health and Disease (DOHaD) hypothesis. DOHaD suggests that maternal insults during pregnancy cause structural and functional changes in fetal organs, increasing the risk of chronic diseases like type 2 diabetes mellitus (T2DM) in adulthood. It is proposed that altered maternal physiology, such as increased glucocorticoid (GC) levels associated with a dysregulated hypothalamic-pituitary-adrenal (HPA) axis in maternal stress and T2DM during pregnancy, exposes the fetus to excess GC. Prenatal glucocorticoid exposure reduces fetal growth and programs the fetal HPA axis, permanently altering its activity into adulthood. This programmed HPA axis is linked to increased risks of hypertension, cardiovascular diseases, and mental disorders in adulthood. With the global rise in T2DM, particularly among young adults of reproductive age, it is crucial to prevent its onset. T2DM is often preceded by a prediabetic state, a condition that does not show any symptoms, causing many to unknowingly progress to T2DM. Studying prediabetes is essential, as it is a reversible stage that may help prevent T2DM-related pregnancy complications. The existing literature focuses on HPA axis dysregulation in T2DM pregnancies and its link to fetal programming. However, the effects of prediabetes on HPA axis function, specifically glucocorticoid in pregnancy and fetal outcomes, are not well understood. This review consolidates research on T2DM during pregnancy, its impact on fetal programming via the HPA axis, and possible links with pregestational prediabetes.
Collapse
Affiliation(s)
| | | | | | - Andile Khathi
- School of Laboratory Medicine & Medical Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4001, South Africa; (M.N.); (N.D.X.); (P.S.N.)
| |
Collapse
|
15
|
Lee S, Lee J, Choi S, Kim E, Kwon H, Lee J, Kim SM, Shin H. Biofabrication of 3D adipose tissue via assembly of composite stem cell spheroids containing adipo-inductive dual-signal delivery nanofibers. Biofabrication 2024; 16:035018. [PMID: 38739412 DOI: 10.1088/1758-5090/ad4a67] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/03/2024] [Indexed: 05/14/2024]
Abstract
Reconstruction of large 3D tissues based on assembly of micro-sized multi-cellular spheroids has gained attention in tissue engineering. However, formation of 3D adipose tissue from spheroids has been challenging due to the limited adhesion capability and restricted cell mobility of adipocytes in culture media. In this study, we addressed this problem by developing adipo-inductive nanofibers enabling dual delivery of indomethacin and insulin. These nanofibers were introduced into composite spheroids comprising human adipose-derived stem cells (hADSCs). This approach led to a significant enhancement in the formation of uniform lipid droplets, as evidenced by the significantly increased Oil red O-stained area in spheroids incorporating indomethacin and insulin dual delivery nanofibers (56.9 ± 4.6%) compared to the control (15.6 ± 3.5%) with significantly greater gene expression associated with adipogenesis (C/EBPA, PPARG, FABP4, and adiponectin) of hADSCs. Furthermore, we investigated the influence of culture media on the migration and merging of spheroids and observed significant decrease in migration and merging of spheroids in adipogenic differentiation media. Conversely, the presence of adipo-inductive nanofibers promoted spheroid fusion, allowing the formation of macroscopic 3D adipose tissue in the absence of adipogenic supplements while facilitating homogeneous adipogenesis of hADSCs. The approach described here holds promise for the generation of 3D adipose tissue constructs by scaffold-free assembly of stem cell spheroids with potential applications in clinical and organ models.
Collapse
Affiliation(s)
- Sangmin Lee
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
- BK21 FOUR, Human-Tech Convergence Program, Hanyang University, Seoul 04763, Republic of Korea
| | - Jeongbok Lee
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation, Hanyang University, Seoul 04763, Republic of Korea
| | - Soomi Choi
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Eunhyung Kim
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation, Hanyang University, Seoul 04763, Republic of Korea
| | - Hyunseok Kwon
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation, Hanyang University, Seoul 04763, Republic of Korea
| | - Jinkyu Lee
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Sung Min Kim
- BK21 FOUR, Human-Tech Convergence Program, Hanyang University, Seoul 04763, Republic of Korea
- Major in Sport Science, Collage of Performing Arts and Sport, Hanyang University, Seoul 04763, Republic of Korea
- Center for Artificial Intelligence Muscle, Hanyang University, Seoul 04743, Republic of Korea
| | - Heungsoo Shin
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation, Hanyang University, Seoul 04763, Republic of Korea
- Institute of Nano Science and Technology, Hanyang University, Seoul 04763, Republic of Korea
| |
Collapse
|
16
|
Rahman MS, Hosen ME, Faruqe MO, Khalekuzzaman M, Islam MA, Acharjee UK, Bin Jardan YA, Nafidi HA, Mekonnen AB, Bourhia M, Zaman R. Evaluation of Adenanthera pavonina-derived compounds against diabetes mellitus: insight into the phytochemical analysis and in silico assays. Front Mol Biosci 2024; 10:1278701. [PMID: 38601799 PMCID: PMC11004346 DOI: 10.3389/fmolb.2023.1278701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 12/19/2023] [Indexed: 04/12/2024] Open
Abstract
Adenanthera pavonina is a medicinal plant with numerous potential secondary metabolites showing a significant level of antidiabetic activity. The objective of the current study was to identify potential phytochemicals from the methanolic leaf extract of Adenanthera pavonina as therapeutic agents against diabetes mellitus using GC-MS and in silico methods. The GC-MS analysis of the leaf extract revealed a total of 17 phytochemicals. Molecular docking was performed using these phytochemicals, targeting the mutated insulin receptor tyrosine kinase (5hhw), which inhibits glucose uptake by cells. Diazoprogesterone (-9.2 kcal/mol), 2,4,4,7a-Tetramethyl-1-(3-oxobutyl)octahydro-1H-indene-2-carboxylic acid (-6.9 kcal/mol), and 2-Naphthalenemethanol, decahydro-.alpha.,.alpha.,4a-trimethyl-8-methylene-, [2R-(2.alpha.,4a.alpha.,8a.beta.)] (-6.6 kcal/mol) exhibited better binding with the target protein. The ADMET analysis was performed for the top three compounds with the best docking scores, which showed positive results with no observed toxicity in the AMES test. Furthermore, the molecular dynamics study confirmed the favorable binding of Diazoprogesterone, 2,4,4,7a-Tetramethyl-1-(3-oxobutyl)octahydro-1H-indene-2-carboxylic acid and 2-Naphthalenemethanol, decahydro-.alpha.,.alpha.,4a-trimethyl-8-methylene-, [2R-(2.alpha.,4a.alpha.,8a.beta.)] with the receptor throughout the 100 ns simulation period.
Collapse
Affiliation(s)
- Md. Sojiur Rahman
- Professor Joarder DNA and Chromosome Research Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi, Bangladesh
| | - Md. Eram Hosen
- Professor Joarder DNA and Chromosome Research Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi, Bangladesh
| | - Md. Omar Faruqe
- Department of Computer Science and Engineering, University of Rajshahi, Rajshahi, Bangladesh
| | - Md. Khalekuzzaman
- Professor Joarder DNA and Chromosome Research Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi, Bangladesh
| | - Md. Asadul Islam
- Professor Joarder DNA and Chromosome Research Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi, Bangladesh
| | - Uzzal Kumar Acharjee
- Professor Joarder DNA and Chromosome Research Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi, Bangladesh
| | - Yousef A. Bin Jardan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hiba-Allah Nafidi
- Department of Food Science, Faculty of Agricultural and Food Sciences, Laval University, Quebec City, QC, Canada
| | | | - Mohammed Bourhia
- Laboratory of Biotechnology and Natural Resources Valorization, Faculty of Sciences, Ibn Zohr University, Agadir, Morocco
| | - Rashed Zaman
- Professor Joarder DNA and Chromosome Research Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi, Bangladesh
| |
Collapse
|
17
|
Dai J, Chen T, Meng R, Jardi F, Kourula S, Pham L, De Jonghe S, De Smedt A, Frisk AL, Xie J. Species differences in small intestinal exposure-related epithelial vacuolation in rats and dogs treated with a heteroaryldihydropyrimidine molecule. J Appl Toxicol 2024; 44:400-414. [PMID: 37814191 DOI: 10.1002/jat.4550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/11/2023]
Abstract
Small intestinal epithelial vacuolation induced by a heteroaryldihydropyrimidine compound (HAP-1) was observed in rats but not in dogs at termination in screening toxicity studies, despite the plasma exposure being higher in dogs. To understand the species differences, investigational studies with multiple time points following single dose (SD) and 7-day repeated dose (RD) were conducted in both species at doses resulting in comparable plasma exposures. In rats, epithelial vacuolation in the duodenum and jejunum were observed at all time points. In dogs, transient vacuolation was noted at 8 h post-SD (SD_8h) and 4 h post-RD (RD_4 h), but not at termination (RD_24 h). Special stains demonstrated lipid accumulation within enterocytes in both species and intracytoplasmic inclusion bodies in rats. Transmission electron microscopy identified these inclusion bodies as endoplasmic reticulum (ER) membranous structures. Transcriptomic analysis on jejunal mucosa at SD_8 h and RD_24 h revealed perturbations of lipid metabolism-related genes at SD_8 h in both species, but not at RD_24 h in dogs. ER stress-related gene changes at both time points were observed in rats only. Despite comparable HAP-1 plasma exposures, the duodenum and jejunum tissue concentrations of HAP-1 and acyl glucuronide metabolite were >5- and >30-fold higher in rats than in dogs, respectively. In vitro, similar cytotoxicity was observed in rat and dog duodenal organoids treated with HAP-1. In conclusion, HAP-1-induced intestinal epithelial vacuolation was related to lipid metabolism dysregulation in both species and ER-related injuries in rats only. The species differences were likely related to the difference in intestinal exposure to HAP-1 and its reactive metabolite.
Collapse
Affiliation(s)
- Jieyu Dai
- Preclinical Sciences and Translational Safety (PSTS), Janssen R&D, Shanghai, China
| | - Tao Chen
- Preclinical Sciences and Translational Safety (PSTS), Janssen R&D, Shanghai, China
| | - Ryan Meng
- Preclinical Sciences and Translational Safety (PSTS), Janssen R&D, Shanghai, China
| | | | | | - Ly Pham
- PSTS, Janssen Pharmaceuticals Inc., Spring House, Pennsylvania, USA
| | | | | | | | - Jianxun Xie
- Preclinical Sciences and Translational Safety (PSTS), Janssen R&D, Shanghai, China
| |
Collapse
|
18
|
Song R, Yao X, Jing F, Yang W, Wu J, Zhang H, Zhang P, Xie Y, Pan X, Zhao L, Wu C. Effects of Five Lipid Sources on Growth, Hematological Parameters, Immunity and Muscle Quality in Juvenile Largemouth Bass ( Micropterus salmoides). Animals (Basel) 2024; 14:781. [PMID: 38473166 DOI: 10.3390/ani14050781] [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: 02/04/2024] [Revised: 02/25/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
This study investigated the effects of fish oil (FO), soybean oil (SO), rapeseed oil (RO), peanut oil (PO) and lard oil (LO) on growth, immunity and muscle quality in juvenile largemouth bass. After 8 weeks, the results showed that FO and RO could increase weight gain and serum alkaline phosphatase and apelin values compared with LO (p < 0.05). Except lower crude lipid contents, higher amounts of n-3 polyunsaturated fatty acids (15.83% and 14.64%) were present in the dorsal muscle of the FO and RO groups. Meanwhile, FO and RO could heighten mRNA levels of immune defense molecules (lysozyme, hepcidin, and transforming growth factor β1) compared with PO (p < 0.05). While SO could increase potential inflammatory risk via rising counts of white blood cells, platelets, neutrophils and monocytes, and mRNA levels of interleukins (IL-1β, IL-8, IL-12 and IL-15), FO and RO could improve hardness, chewiness and springiness through increasing amounts of hydroxyproline, collagen and lysyl oxidase, and mRNA levels of collagen 1α2 and prolyl hydroxylase in the fish dorsal muscle. Moreover, FO and RO could improve firmness through increasing glycogen and glycogen synthase 1 levels when compared with LO (p < 0.05). Therefore, these results could provide dietary lipid source references during the feeding process of adult largemouth bass.
Collapse
Affiliation(s)
- Rui Song
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), Huzhou University, 759 East 2nd Road, Huzhou 313000, China
| | - Xinfeng Yao
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), Huzhou University, 759 East 2nd Road, Huzhou 313000, China
| | - Futao Jing
- Shandong Fisheries Development and Resources Conservation Center, 162 Jiefang Road, Jinan 250013, China
| | - Wenxue Yang
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), Huzhou University, 759 East 2nd Road, Huzhou 313000, China
| | - Jiaojiao Wu
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), Huzhou University, 759 East 2nd Road, Huzhou 313000, China
| | - Hao Zhang
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), Huzhou University, 759 East 2nd Road, Huzhou 313000, China
| | - Penghui Zhang
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), Huzhou University, 759 East 2nd Road, Huzhou 313000, China
| | - Yuanyuan Xie
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), Huzhou University, 759 East 2nd Road, Huzhou 313000, China
| | - Xuewen Pan
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), Huzhou University, 759 East 2nd Road, Huzhou 313000, China
| | - Long Zhao
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), Huzhou University, 759 East 2nd Road, Huzhou 313000, China
| | - Chenglong Wu
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), Huzhou University, 759 East 2nd Road, Huzhou 313000, China
| |
Collapse
|
19
|
Mallick R, Basak S, Das RK, Banerjee A, Paul S, Pathak S, Duttaroy AK. Fatty Acids and their Proteins in Adipose Tissue Inflammation. Cell Biochem Biophys 2024; 82:35-51. [PMID: 37794302 PMCID: PMC10867084 DOI: 10.1007/s12013-023-01185-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2023] [Indexed: 10/06/2023]
Abstract
Chronic low-grade adipose tissue inflammation is associated with metabolic disorders. Inflammation results from the intertwined cross-talks of pro-inflammatory and anti-inflammatory pathways in the immune response of adipose tissue. In addition, adipose FABP4 levels and lipid droplet proteins are involved in systemic and tissue inflammation. Dysregulated adipocytes help infiltrate immune cells derived from bone marrow responsible for producing cytokines and chemokines. When adipose tissue expands in excess, adipocyte exhibits increased secretion of adipokines and is implicated in metabolic disturbances due to the release of free fatty acids. This review presents an emerging concept in adipose tissue fat metabolism, fatty acid handling and binding proteins, and lipid droplet proteins and their involvement in inflammatory disorders.
Collapse
Affiliation(s)
- Rahul Mallick
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sanjay Basak
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Ranjit K Das
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Antara Banerjee
- Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chennai, India
| | - Sujay Paul
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc, San Pablo, Queretaro, 76130, Mexico
| | - Surajit Pathak
- Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chennai, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, POB 1046 Blindern, Oslo, Norway.
| |
Collapse
|
20
|
Kim SY, Park SY, Kim JE. GULP1 deficiency reduces adipogenesis and glucose uptake via downregulation of PPAR signaling and disturbing of insulin/ERK signaling in 3T3-L1 cells. J Cell Physiol 2024; 239:e31173. [PMID: 38214103 DOI: 10.1002/jcp.31173] [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: 07/30/2023] [Revised: 11/10/2023] [Accepted: 12/06/2023] [Indexed: 01/13/2024]
Abstract
Obesity and metabolic disorders caused by alterations in lipid metabolism are major health issues in developed, affluent societies. Adipose tissue is the only organ that stores lipids and prevents lipotoxicity in other organs. Mature adipocytes can affect themselves and distant metabolism-related tissues by producing various adipokines, including adiponectin and leptin. The engulfment adaptor phosphotyrosine-binding domain-containing 1 (GULP1) regulates intracellular trafficking of glycosphingolipids and cholesterol, suggesting its close association with lipid metabolism. However, the role of GULP1 in adipocytes remains unknown. Therefore, this study aimed to investigate the function of GULP1 in adipogenesis, glucose uptake, and the insulin signaling pathway in adipocytes. A 3T3-L1 cell line with Gulp1 knockdown (shGulp1) and a 3T3-L1 control group (U6) were established. Changes in shGulp1 cells due to GULP1 deficiency were examined and compared to those in U6 cells using microarray analysis. Glucose uptake was monitored via insulin stimulation in shGulp1 and U6 cells using a 2-NBDG glucose uptake assay, and the insulin signaling pathway was investigated by western blot analysis. Adipogenesis was significantly delayed, lipid metabolism was altered, and several adipogenesis-related genes were downregulated in shGulp1 cells compared to those in U6 cells. Microarray analysis revealed significant inhibition of peroxisome proliferator-activated receptor signaling in shGulp1 cells compared with U6 cells. The production and secretion of adiponectin as well as the expression of adiponectin receptor were decreased in shGulp1 cells. In particular, compared with U6 cells, glucose uptake via insulin stimulation was significantly decreased in shGulp1 cells through the disturbance of ERK1/2 phosphorylation. This is the first study to identify the role of GULP1 in adipogenesis and insulin-stimulated glucose uptake by adipocytes, thereby providing new insights into the differentiation and functions of adipocytes and the metabolism of lipids and glucose, which can help better understand metabolic diseases.
Collapse
Affiliation(s)
- Soon-Young Kim
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Seung-Yoon Park
- Department of Biochemistry, School of Medicine, Dongguk University, Gyeongju, Republic of Korea
| | - Jung-Eun Kim
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Biomedical Science, BK21 Four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
21
|
Liu G, Yang L, Tang Y, Lin J, Wang F, Shen J, Chang B, Kong X. Study on the action mechanism of the Polygonum perfoliatum L. on non-alcoholic fatty liver disease, based on network pharmacology and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117330. [PMID: 37863399 DOI: 10.1016/j.jep.2023.117330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese medicine (TCM) holds that non-alcoholic fatty liver disease (NAFLD) belong to the category of "thoracic fullness". Polygonum perfoliatum L. (PPL), a Chinese medicinal herb with the effect of treating thoracic fullness, was recorded in the ancient Chinese medicine book "Supplements to Compendium of Materia Medica". It has been used since ancient times to treat NAFLD. However, the underlying mechanism and active components of PPL against NAFLD remains unclear. AIM OF STUDY To identify the main active components and the anti-NAFLD mechanism of PPL. MATERIALS AND METHODS Network pharmacology, UPLC/QE-HFX analysis, and molecular docking were employed to determine the main bioactive compounds and key targets of PPL for the NAFLD treatment. This effect was further validated with administration of PPL (200 mg/kg and 400 mg/kg) to NAFLD model mice for 5 weeks. Systemic signs of obesity, biochemical parameters, and histological changes were characterized. Immunohistochemistry, western blot, and PCR analysis were conducted to elucidate the mechanistic pathways through which PPL exerts its effects. RESULTS Network pharmacology revealed 77 crossover genes between the PPL and NAFLD. The kyoto encyclopedia of genes and genomes (KEGG) analysis show that PPL treat NAFLD mainly regulating glucose-lipid metabolism mediated by PI3K/AKT signal pathway. The Gene Ontology (GO) enrichment analysis show that PPL treat NAFLD mainly regulating inflammation mediated by cytokine-mediated signaling pathway. In accordance with the anticipated outcomes, administration of PPL in a dose-dependent manner effectively mitigated insulin resistance induced by a high-fat diet (HFD) by activating the PI3K/AKT signaling pathway. Histopathological evaluation corroborated the hepatoprotective effects of PPL against HFD-induced hepatic steatosis, as evidenced by the inhibition of de novo fatty acid synthesis and promotion of fatty acid β-oxidation (FAO). Further research showed that PPL blocked cytokine production by inhibiting the NF-κB pathway, thereby reducing immune cell infiltration. Furthermore, five flavonoids from PPL, including quercetin, baicalein, galangin, apigenin, and genistein were identified as key compounds based on ingredient-target-pathway network analysis. Molecular docking show that these active compounds have favorable binding interactions with AKT1, PIK3R1, and MAPK1, further confirming the impact of PPL on the PI3K/AKT pathway. CONCLUSIONS Through the combination of network pharmacology prediction and experimental validation, this work determined that therapeutic effect of PPL on NAFLD, and such protective effect is mediated by activating PI3K/AKT-mediated glucolipid metabolism pathway and hepatic NF-κB-mediated cytokine signaling pathway.
Collapse
Affiliation(s)
- Guanjie Liu
- Central Laboratory, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Liu Yang
- Central Laboratory, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Yifei Tang
- Department of Liver Diseases, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Jiacheng Lin
- Central Laboratory, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Fang Wang
- Central Laboratory, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Jie Shen
- Department of pharmacy, The SATCM Third Grade Laboratory of Traditional Chinese Medicine Preparations, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Bin Chang
- Department of Pathology, Shuguang Hospital, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China.
| | - Xiaoni Kong
- Central Laboratory, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China.
| |
Collapse
|
22
|
Ye RZ, Montastier E, Frisch F, Noll C, Allard-Chamard H, Gévry N, Tchernof A, Carpentier AC. Adipocyte hypertrophy associates with in vivo postprandial fatty acid metabolism and adipose single-cell transcriptional dynamics. iScience 2024; 27:108692. [PMID: 38226167 PMCID: PMC10788217 DOI: 10.1016/j.isci.2023.108692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/07/2023] [Accepted: 12/05/2023] [Indexed: 01/17/2024] Open
Abstract
Adipocyte hypertrophy is associated with metabolic complications independent of obesity. We aimed to determine: 1) the association between adipocyte size and postprandial fatty acid metabolism; 2) the potential mechanisms driving the obesity-independent, hypertrophy-associated dysmetabolism in vivo and at a single-cell resolution. Tracers with positron emission tomography were used to measure fatty acid metabolism in 40 men and women with normal or impaired glucose tolerance (NCT02808182), and single nuclei RNA-sequencing (snRNA-seq) to determine transcriptional dynamics of subcutaneous adipose tissue (AT) between individuals with AT hypertrophy vs. hyperplasia matched for sex, ethnicity, glucose-tolerance status, BMI, total and percent body fat, and waist circumference. Adipocyte size was associated with high postprandial total cardiac fatty acid uptake and higher visceral AT dietary fatty acid uptake, but lower lean tissue dietary fatty acid uptake. We found major shifts in cell transcriptomal dynamics with AT hypertrophy that were consistent with in vivo metabolic changes.
Collapse
Affiliation(s)
- Run Zhou Ye
- Division of Endocrinology, Department of Medicine, Centre de recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Emilie Montastier
- Division of Endocrinology, Department of Medicine, Centre de recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Frédérique Frisch
- Division of Endocrinology, Department of Medicine, Centre de recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Christophe Noll
- Division of Endocrinology, Department of Medicine, Centre de recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Hugues Allard-Chamard
- Division of Endocrinology, Department of Medicine, Centre de recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Nicolas Gévry
- Department of Biology, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - André Tchernof
- Québec Heart and Lung Research Institute, Laval University, Québec, QC G1V 4G5, Canada
| | - André C. Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Department of Nuclear Medicine and Radiobiology, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| |
Collapse
|
23
|
Engin AB, Engin A. Next-Cell Hypothesis: Mechanism of Obesity-Associated Carcinogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:727-766. [PMID: 39287871 DOI: 10.1007/978-3-031-63657-8_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Higher body fat content is related to a higher risk of mortality, and obesity-related cancer represents approximately 40% of all cancer patients diagnosed each year. Furthermore, epigenetic mechanisms are involved in cellular metabolic memory and can determine one's predisposition to being overweight. Low-grade chronic inflammation, a well-established characteristic of obesity, is a central component of tumor development and progression. Cancer-associated adipocytes (CAA), which enhance inflammation- and metastasis-related gene sets within the cancer microenvironment, have pro-tumoral effects. Adipose tissue is a major source of the exosomal micro ribonucleic acids (miRNAs), which modulate pathways involved in the development of obesity and obesity-related comorbidities. Owing to their composition of cargo, exosomes can activate receptors at the target cell or transfer molecules to the target cells and thereby change the phenotype of these cells. Exosomes that are released into the extracellular environment are internalized with their cargo by neighboring cells. The tumor-secreted exosomes promote organ-specific metastasis of tumor cells that normally lack the capacity to metastasize to a specific organ. Therefore, the communication between neighboring cells via exosomes is defined as the "next-cell hypothesis." The reciprocal interaction between the adipocyte and tumor cell is realized through the adipocyte-derived exosomal miRNAs and tumor cell-derived oncogenic miRNAs. The cargo molecules of adipocyte-derived exosomes are important messengers for intercellular communication involved in metabolic responses and have very specific signatures that direct the metabolic activity of target cells. RNA-induced silencing regulates gene expression through various mechanisms. Destabilization of DICER enzyme, which catalyzes the conversion of primary miRNA (pri-miRNA) to precursor miRNA (pre-miRNA), is an important checkpoint in cancer development and progression. Interestingly, adipose tissue in obesity and tumors share similar pathogenic features, and the local hypoxia progress in both. While hypoxia in obesity leads to the adipocyte dysfunction and metabolic abnormalities, in obesity-related cancer cases, it is associated with worsened prognosis, increased metastatic potential, and resistance to chemotherapy. Notch-interleukin-1 (IL-1)-Leptin crosstalk outcome is referred to as "NILCO effect." In this chapter, obesity-related cancer development is discussed in the context of "next-cell hypothesis," miRNA biogenesis, and "NILCO effect."
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| | - Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey
| |
Collapse
|
24
|
Zhou Y, Liu K, Tang W, Zhang Y, Sun Y, Wu Y, Shi Y, Yao Z, Li Y, Bai R, Liang R, Sun P, Chang X, Wang S, Zhu Y, Han X. β-Cell miRNA-503-5p Induced by Hypomethylation and Inflammation Promotes Insulin Resistance and β-Cell Decompensation. Diabetes 2024; 73:57-74. [PMID: 37847900 DOI: 10.2337/db22-1044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 10/02/2023] [Indexed: 10/19/2023]
Abstract
Chronic inflammation promotes pancreatic β-cell decompensation to insulin resistance because of local accumulation of supraphysiologic interleukin 1β (IL-1β) levels. However, the underlying molecular mechanisms remain elusive. We show that miR-503-5p is exclusively upregulated in islets from humans with type 2 diabetes and diabetic rodents because of its promoter hypomethylation and increased local IL-1β levels. β-Cell-specific miR-503 transgenic mice display mild or severe diabetes in a time- and expression-dependent manner. By contrast, deletion of the miR-503 cluster protects mice from high-fat diet-induced insulin resistance and glucose intolerance. Mechanistically, miR-503-5p represses c-Jun N-terminal kinase-interacting protein 2 (JIP2) translation to activate mitogen-activated protein kinase signaling cascades, thus inhibiting glucose-stimulated insulin secretion (GSIS) and compensatory β-cell proliferation. In addition, β-cell miR-503-5p is packaged in nanovesicles to dampen insulin signaling transduction in liver and adipose tissues by targeting insulin receptors. Notably, specifically blocking the miR-503 cluster in β-cells effectively remits aging-associated diabetes through recovery of GSIS capacity and insulin sensitivity. Our findings demonstrate that β-cell miR-503-5p is required for the development of insulin resistance and β-cell decompensation, providing a potential therapeutic target against diabetes. ARTICLE HIGHLIGHTS Promoter hypomethylation during natural aging permits miR-503-5p overexpression in islets under inflammation conditions, conserving from rodents to humans. Impaired β-cells release nanovesicular miR-503-5p to accumulate in liver and adipose tissue, leading to their insulin resistance via the miR-503-5p/insulin receptor/phosphorylated AKT axis. Accumulated miR-503-5p in β-cells impairs glucose-stimulated insulin secretion via the JIP2-coordinated mitogen-activated protein kinase signaling cascades. Specific blockage of β-cell miR-503-5p improves β-cell function and glucose tolerance in aging mice.
Collapse
Affiliation(s)
- Yuncai Zhou
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Kerong Liu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wei Tang
- Department of Endocrinology, Geriatric Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yan Zhang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yi Sun
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yangyang Wu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ying Shi
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhengjian Yao
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yating Li
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rongjie Bai
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rui Liang
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Peng Sun
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoai Chang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shusen Wang
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Yunxia Zhu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao Han
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
25
|
Benedet PO, Safikhan NS, Pereira MJ, Lum BM, Botezelli JD, Kuo CH, Wu HL, Craddock BP, Miller WT, Eriksson JW, Yue JTY, Conway EM. CD248 promotes insulin resistance by binding to the insulin receptor and dampening its insulin-induced autophosphorylation. EBioMedicine 2024; 99:104906. [PMID: 38061240 PMCID: PMC10750038 DOI: 10.1016/j.ebiom.2023.104906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 11/24/2023] [Accepted: 11/25/2023] [Indexed: 12/29/2023] Open
Abstract
BACKGROUND In spite of new treatments, the incidence of type 2 diabetes (T2D) and its morbidities continue to rise. The key feature of T2D is resistance of adipose tissue and other organs to insulin. Approaches to overcome insulin resistance are limited due to a poor understanding of the mechanisms and inaccessibility of drugs to relevant intracellular targets. We previously showed in mice and humans that CD248, a pre/adipocyte cell surface glycoprotein, acts as an adipose tissue sensor that mediates the transition from healthy to unhealthy adipose, thus promoting insulin resistance. METHODS Molecular mechanisms by which CD248 regulates insulin signaling were explored using in vivo insulin clamp studies and biochemical analyses of cells/tissues from CD248 knockout (KO) and wild-type (WT) mice with diet-induced insulin resistance. Findings were validated with human adipose tissue specimens. FINDINGS Genetic deletion of CD248 in mice, overcame diet-induced insulin resistance with improvements in glucose uptake and lipolysis in white adipose tissue depots, effects paralleled by increased adipose/adipocyte GLUT4, phosphorylated AKT and GSK3β, and reduced ATGL. The insulin resistance of the WT mice could be attributed to direct interaction of the extracellular domains of CD248 and the insulin receptor (IR), with CD248 acting to block insulin binding to the IR. This resulted in dampened insulin-mediated autophosphorylation of the IR, with reduced downstream signaling/activation of intracellular events necessary for glucose and lipid homeostasis. INTERPRETATION Our discovery of a cell-surface CD248-IR complex that is accessible to pharmacologic intervention, opens research avenues toward development of new agents to prevent/reverse insulin resistance. FUNDING Funded by Canadian Institutes of Health Research (CIHR), Natural Sciences and Engineering Research Council of Canada (NSERC), Canada Foundations for Innovation (CFI), the Swedish Diabetes Foundation, Family Ernfors Foundation and Novo Nordisk Foundation.
Collapse
Affiliation(s)
- Patricia O Benedet
- Centre for Blood Research, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, Canada; Departments of Medicine and Pathology and Laboratory Medicine, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Nooshin S Safikhan
- Centre for Blood Research, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, Canada; Departments of Medicine and Pathology and Laboratory Medicine, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Maria J Pereira
- Department of Medical Sciences, Clinical Diabetology & Metabolism, Uppsala University, Sweden
| | - Bryan M Lum
- Department of Physiology, Alberta Diabetes Institute and Group on Molecular and Cell Biology of Lipids, University of Alberta, Canada
| | - José Diego Botezelli
- Centre for Blood Research, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, Canada; Departments of Medicine and Pathology and Laboratory Medicine, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Cheng-Hsiang Kuo
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
| | - Hua-Lin Wu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Barbara P Craddock
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, USA
| | - W Todd Miller
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, USA; Veterans Affairs Medical Center, Northport, NY, USA
| | - Jan W Eriksson
- Department of Medical Sciences, Clinical Diabetology & Metabolism, Uppsala University, Sweden
| | - Jessica T Y Yue
- Department of Physiology, Alberta Diabetes Institute and Group on Molecular and Cell Biology of Lipids, University of Alberta, Canada
| | - Edward M Conway
- Centre for Blood Research, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, Canada; Departments of Medicine and Pathology and Laboratory Medicine, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
26
|
Umbayev B, Saliev T, Safarova (Yantsen) Y, Yermekova A, Olzhayev F, Bulanin D, Tsoy A, Askarova S. The Role of Cdc42 in the Insulin and Leptin Pathways Contributing to the Development of Age-Related Obesity. Nutrients 2023; 15:4964. [PMID: 38068822 PMCID: PMC10707920 DOI: 10.3390/nu15234964] [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: 10/25/2023] [Revised: 11/22/2023] [Accepted: 11/26/2023] [Indexed: 12/18/2023] Open
Abstract
Age-related obesity significantly increases the risk of chronic diseases such as type 2 diabetes, cardiovascular diseases, hypertension, and certain cancers. The insulin-leptin axis is crucial in understanding metabolic disturbances associated with age-related obesity. Rho GTPase Cdc42 is a member of the Rho family of GTPases that participates in many cellular processes including, but not limited to, regulation of actin cytoskeleton, vesicle trafficking, cell polarity, morphology, proliferation, motility, and migration. Cdc42 functions as an integral part of regulating insulin secretion and aging. Some novel roles for Cdc42 have also been recently identified in maintaining glucose metabolism, where Cdc42 is involved in controlling blood glucose levels in metabolically active tissues, including skeletal muscle, adipose tissue, pancreas, etc., which puts this protein in line with other critical regulators of glucose metabolism. Importantly, Cdc42 plays a vital role in cellular processes associated with the insulin and leptin signaling pathways, which are integral elements involved in obesity development if misregulated. Additionally, a change in Cdc42 activity may affect senescence, thus contributing to disorders associated with aging. This review explores the complex relationships among age-associated obesity, the insulin-leptin axis, and the Cdc42 signaling pathway. This article sheds light on the vast molecular web that supports metabolic dysregulation in aging people. In addition, it also discusses the potential therapeutic implications of the Cdc42 pathway to mitigate obesity since some new data suggest that inhibition of Cdc42 using antidiabetic drugs or antioxidants may promote weight loss in overweight or obese patients.
Collapse
Affiliation(s)
- Bauyrzhan Umbayev
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (Y.S.); (A.Y.); (F.O.); (A.T.); (S.A.)
| | - Timur Saliev
- S.D. Asfendiyarov Kazakh National Medical University, Almaty 050012, Kazakhstan;
| | - Yuliya Safarova (Yantsen)
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (Y.S.); (A.Y.); (F.O.); (A.T.); (S.A.)
| | - Aislu Yermekova
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (Y.S.); (A.Y.); (F.O.); (A.T.); (S.A.)
| | - Farkhad Olzhayev
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (Y.S.); (A.Y.); (F.O.); (A.T.); (S.A.)
| | - Denis Bulanin
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan;
| | - Andrey Tsoy
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (Y.S.); (A.Y.); (F.O.); (A.T.); (S.A.)
| | - Sholpan Askarova
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (Y.S.); (A.Y.); (F.O.); (A.T.); (S.A.)
| |
Collapse
|
27
|
Luna-Ramirez RI, Kelly AC, Anderson MJ, Bidwell CA, Goyal R, Limesand SW. Elevated Norepinephrine Stimulates Adipocyte Hyperplasia in Ovine Fetuses With Placental Insufficiency and IUGR. Endocrinology 2023; 165:bqad177. [PMID: 38035825 PMCID: PMC10726312 DOI: 10.1210/endocr/bqad177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/01/2023] [Accepted: 11/22/2023] [Indexed: 12/02/2023]
Abstract
Prevailing hypoxemia and hypoglycemia in near-term fetuses with placental insufficiency-induced intrauterine growth restriction (IUGR) chronically increases norepinephrine concentrations, which lower adrenergic sensitivity and lipid mobilization postnatally, indicating a predisposition for adiposity. To determine adrenergic-induced responses, we examined the perirenal adipose tissue transcriptome from IUGR fetuses with or without hypercatecholaminemia. IUGR was induced in sheep with maternal hyperthermia, and hypercatecholaminemia in IUGR was prevented with bilateral adrenal demedullation. Adipose tissue was collected from sham-operated control (CON) and IUGR fetuses and adrenal-demedullated control (CAD) and IUGR (IAD) fetuses. Norepinephrine concentrations were lower in IAD fetuses than in IUGR fetuses despite both being hypoxemic and hypoglycemic. In IUGR fetuses, perirenal adipose tissue mass relative to body mass was greater compared with the CON, adrenal-demedullated control, and IAD groups. Transcriptomic analysis identified 581 differentially expressed genes (DEGs) in CON vs IUGR adipose tissue and 193 DEGs in IUGR vs IAD adipose tissue. Integrated functional analysis of these 2 comparisons showed enrichment for proliferator-activated receptor signaling and metabolic pathways and identified adrenergic responsive genes. Within the adrenergic-regulated DEGs, we identified transcripts that regulate adipocyte proliferation and differentiation: adipogenesis regulatory factor, C/CCAAT/enhancer binding protein α, and sterol carrier protein 2. DEGs associated with the metabolic pathway included pyruvate dehydrogenase kinase 4, 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 4, IGF-binding proteins (IGFBP-5 and IGFBP-7). Sex-specific expression differences were also found for adipogenesis regulatory factor, pyruvate dehydrogenase kinase 4, IGFBP5, and IGFBP7. These findings indicate that sustained adrenergic stimulation during IUGR leads to adipocyte hyperplasia with alterations in metabolism, proliferation, and preadipocyte differentiation pathways.
Collapse
Affiliation(s)
- Rosa I Luna-Ramirez
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85719, USA
| | - Amy C Kelly
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85719, USA
| | - Miranda J Anderson
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85719, USA
| | | | - Ravi Goyal
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85719, USA
| | - Sean W Limesand
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85719, USA
| |
Collapse
|
28
|
He M, Li Z, Tung VSK, Pan M, Han X, Evgrafov O, Jiang XC. Inhibiting Phosphatidylcholine Remodeling in Adipose Tissue Increases Insulin Sensitivity. Diabetes 2023; 72:1547-1559. [PMID: 37625119 PMCID: PMC10588299 DOI: 10.2337/db23-0317] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023]
Abstract
Cell membrane phosphatidylcholine (PC) composition is regulated by lysophosphatidylcholine acyltransferase (LPCAT); changes in membrane PC saturation are implicated in metabolic disorders. Here, we identified LPCAT3 as the major isoform of LPCAT in adipose tissue and created adipocyte-specific Lpcat3-knockout mice to study adipose tissue lipid metabolism. Transcriptome sequencing and plasma adipokine profiling were used to investigate how LPCAT3 regulates adipose tissue insulin signaling. LPCAT3 deficiency reduced polyunsaturated PCs in adipocyte plasma membranes, increasing insulin sensitivity. LPCAT3 deficiency influenced membrane lipid rafts, which activated insulin receptors and AKT in adipose tissue, and attenuated diet-induced insulin resistance. Conversely, higher LPCAT3 activity in adipose tissue from ob/ob, db/db, and high-fat diet-fed mice reduced insulin signaling. Adding polyunsaturated PCs to mature human or mouse adipocytes in vitro worsened insulin signaling. We suggest that targeting LPCAT3 in adipose tissue to manipulate membrane phospholipid saturation is a new strategy to treat insulin resistance. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Mulin He
- Department of Cell Biology, The State University of New York Downstate Health Sciences University, Brooklyn, NY
| | - Zhiqiang Li
- Department of Cell Biology, The State University of New York Downstate Health Sciences University, Brooklyn, NY
| | - Victoria Sook Keng Tung
- Department of Cell Biology, The State University of New York Downstate Health Sciences University, Brooklyn, NY
| | - Meixia Pan
- Lipidomics Core, The University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Xianlin Han
- Lipidomics Core, The University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Oleg Evgrafov
- Department of Cell Biology, The State University of New York Downstate Health Sciences University, Brooklyn, NY
| | - Xian-Cheng Jiang
- Department of Cell Biology, The State University of New York Downstate Health Sciences University, Brooklyn, NY
- Molecular and Cellular Cardiology Program, Veterans Affairs New York Harbor Healthcare System, New York, NY
| |
Collapse
|
29
|
Genchi VA, Palma G, Sorice GP, D'Oria R, Caccioppoli C, Marrano N, Biondi G, Caruso I, Cignarelli A, Natalicchio A, Laviola L, Giorgino F, Perrini S. Pharmacological modulation of adaptive thermogenesis: new clues for obesity management? J Endocrinol Invest 2023; 46:2213-2236. [PMID: 37378828 PMCID: PMC10558388 DOI: 10.1007/s40618-023-02125-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023]
Abstract
BACKGROUND Adaptive thermogenesis represents the main mechanism through which the body generates heat in response to external stimuli, a phenomenon that includes shivering and non-shivering thermogenesis. The non-shivering thermogenesis is mainly exploited by adipose tissue characterized by a brown aspect, which specializes in energy dissipation. A decreased amount of brown adipose tissue has been observed in ageing and chronic illnesses such as obesity, a worldwide health problem characterized by dysfunctional adipose tissue expansion and associated cardiometabolic complications. In the last decades, the discovery of a trans-differentiation mechanism ("browning") within white adipose tissue depots, leading to the generation of brown-like cells, allowed to explore new natural and synthetic compounds able to favour this process and thus enhance thermogenesis with the aim of counteracting obesity. Based on recent findings, brown adipose tissue-activating agents could represent another option in addition to appetite inhibitors and inhibitors of nutrient absorption for obesity treatment. PURPOSE This review investigates the main molecules involved in the physiological (e.g. incretin hormones) and pharmacological (e.g. β3-adrenergic receptors agonists, thyroid receptor agonists, farnesoid X receptor agonists, glucagon-like peptide-1, and glucagon receptor agonists) modulation of adaptive thermogenesis and the signalling mechanisms involved.
Collapse
Affiliation(s)
- V A Genchi
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - G Palma
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - G P Sorice
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - R D'Oria
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - C Caccioppoli
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - N Marrano
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - G Biondi
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - I Caruso
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - A Cignarelli
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - A Natalicchio
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - L Laviola
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - F Giorgino
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy.
| | - S Perrini
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| |
Collapse
|
30
|
Bahramzadeh A, Bolandnazar K, Meshkani R. Resveratrol as a potential protective compound against skeletal muscle insulin resistance. Heliyon 2023; 9:e21305. [PMID: 38027557 PMCID: PMC10660041 DOI: 10.1016/j.heliyon.2023.e21305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
The increasing prevalence of type 2 diabetes has become a major global problem. Insulin resistance has a central role in pathophysiology of type 2 diabetes. Skeletal muscle is responsible for the disposal of most of the glucose under conditions of insulin stimulation, and insulin resistance in skeletal muscle causes dysregulation of glucose homeostasis in the whole body. Despite the current pharmaceutical and non-pharmacological treatment strategies to combat diabetes, there is still a need for new therapeutic agents due to the limitations of the therapeutic agents. Meanwhile, plant polyphenols have attracted the attention of researchers for their use in the treatment of diabetes and have gained popularity. Resveratrol, a stilbenoid polyphenol, exists in various plant sources, and a growing body of evidence suggests its beneficial properties, including antidiabetic activities. The present review aimed to provide a summary of the role of resveratrol in insulin resistance in skeletal muscle and its related mechanisms. To achieve the objectives, by searching the PubMed, Scopus and Web of Science databases, we have summarized the results of all cell culture, animal, and human studies that have investigated the effects of resveratrol in different models on insulin resistance in skeletal muscle.
Collapse
Affiliation(s)
- Arash Bahramzadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kosar Bolandnazar
- Department of Biological Sciences and Technology, Islamic Azad University of Mashhad, Mashhad, Iran
| | - Reza Meshkani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Fajardo CM, Cerda A, Bortolin RH, de Oliveira R, Stefani TIM, Dos Santos MA, Braga AA, Dorea EL, Bernik MMS, Bastos GM, Sampaio MF, Damasceno NRT, Verlengia R, de Oliveira MRM, Hirata MH, Hirata RDC. Influence of polymorphisms in IRS1, IRS2, MC3R, and MC4R on metabolic and inflammatory status and food intake in Brazilian adults: An exploratory pilot study. Nutr Res 2023; 119:21-32. [PMID: 37716291 DOI: 10.1016/j.nutres.2023.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/18/2023]
Abstract
Polymorphisms in genes of leptin-melanocortin and insulin pathways have been associated with obesity and type 2 diabetes. We hypothesized that polymorphisms in IRS1, IRS2, MC3R, and MC4R influence metabolic and inflammatory markers and food intake composition in Brazilian subjects. This exploratory pilot study included 358 adult subjects. Clinical, anthropometric, and laboratory data were obtained through interview and access to medical records. The variants IRS1 rs2943634 A˃C, IRS2 rs1865434 C>T, MC3R rs3746619 C>A, and MC4R rs17782313 T>C were analyzed by real-time polymerase chain reaction. Food intake composition was assessed in a group of subjects with obesity (n = 84) before and after a short-term nutritional counseling program (9 weeks). MC4R rs17782313 was associated with increased risk of obesity (P = .034). Multivariate linear regression analysis adjusted by covariates indicated associations of IRS2 rs1865434 with reduced low-density lipoprotein cholesterol and resistin, MC3R rs3746619 with high glycated hemoglobin, and IRS1 rs2943634 and MC4R rs17782313 with increased high-sensitivity C-reactive protein (P < .05). Energy intake and carbohydrate and total fat intakes were reduced after the diet-oriented program (P < .05). Multivariate linear regression analysis showed associations of IRS2 rs1865434 with high basal fiber intake, IRS1 rs2943634 with low postprogram carbohydrate intake, and MC4R rs17782313 with low postprogram total fat and saturated fatty acid intakes (P < .05). Although significant associations did not survive correction for multiple comparisons using the Benjamini-Hochberg method in this exploratory study, polymorphisms in IRS1, IRS2, MC3R, and MC4R influence metabolic and inflammatory status in Brazilian adults. IRS1 and MC4R variants may influence carbohydrate, total fat, and saturated fatty acid intakes in response to a diet-oriented program in subjects with obesity.
Collapse
MESH Headings
- Adult
- Humans
- Pilot Projects
- Diabetes Mellitus, Type 2/genetics
- Polymorphism, Single Nucleotide
- Brazil
- Obesity/genetics
- Obesity/metabolism
- Eating
- Carbohydrates
- Fatty Acids
- Receptor, Melanocortin, Type 4/genetics
- Receptor, Melanocortin, Type 4/metabolism
- Insulin Receptor Substrate Proteins/genetics
- Insulin Receptor Substrate Proteins/metabolism
- Receptor, Melanocortin, Type 3/genetics
- Receptor, Melanocortin, Type 3/metabolism
Collapse
Affiliation(s)
- Cristina Moreno Fajardo
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Alvaro Cerda
- Department of Basic Sciences, Center of Excellence in Translational Medicine, CEMT-BIOREN, Universidad de La Frontera, Temuco 4810296, Chile
| | - Raul Hernandes Bortolin
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil; Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, United States
| | - Raquel de Oliveira
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Tamires Invencioni Moraes Stefani
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Marina Aparecida Dos Santos
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Aécio Assunção Braga
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Egídio Lima Dorea
- Medical Clinic Division, University Hospital, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | | | - Gisele Medeiros Bastos
- Laboratory of Molecular Research in Cardiology, Institute of Cardiology Dante Pazzanese, Sao Paulo 04012-909, Brazil; Hospital Beneficiencia Portuguesa de Sao Paulo, Sao Paulo 01323-001, Brazil
| | - Marcelo Ferraz Sampaio
- Hospital Beneficiencia Portuguesa de Sao Paulo, Sao Paulo 01323-001, Brazil; Medical Clinic Division, Institute of Cardiology Dante Pazzanese, Sao Paulo 04012-909, Brazil
| | | | - Rozangela Verlengia
- Research Laboratory in Human Performance, Methodist University of Piracicaba, Piracicaba 13400-901, Brazil
| | | | - Mario Hiroyuki Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Rosario Dominguez Crespo Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil.
| |
Collapse
|
32
|
Su J, Tang L, Luo Y, Xu J, Ouyang S. Research progress on drugs for diabetes based on insulin receptor/insulin receptor substrate. Biochem Pharmacol 2023; 217:115830. [PMID: 37748666 DOI: 10.1016/j.bcp.2023.115830] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 09/27/2023]
Abstract
The number of people with diabetes worldwide is increasing annually, resulting in a serious economic burden. Insulin resistance is a major pathology in the early onset of diabetes mellitus, and therefore, related drug studies have attracted research attention. The insulin receptor/insulin receptor substrate (INSR/IRS) serves as the primary conduit in the insulin signal transduction cascade, and dysregulation of this pathway can lead to insulin resistance. Currently, there exist a plethora of hypoglycemic drugs in the market; however, drugs that specifically target INSR/IRS are comparatively limited. The literature was collected by direct access to the PubMed database, and was searched using the terms "diabetes mellitus; insulin resistance; insulin receptor; insulin receptor substrate; diabetes drug" as the main keywords for literature over the last decade. This article provides a comprehensive analysis of the structure and function of INSR and IRS proteins, as well as the drugs used for the treatment of diabetes. Additionally, it serves as a valuable reference for the advancement of novel therapeutic agents for diabetes management.
Collapse
Affiliation(s)
- Jingqian Su
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China; Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China.
| | - Lu Tang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China; Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Yingsheng Luo
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China; Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Jingran Xu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China; Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Songying Ouyang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China; Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China; Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, Fujian Normal University, Fuzhou 350117, China.
| |
Collapse
|
33
|
Xu J, Shao X, Zeng H, Wang C, Li J, Peng X, Zhuo Y, Hua L, Meng F, Han X. Hepatic-Specific FGF21 Knockout Abrogates Ovariectomy-Induced Obesity by Reversing Corticosterone Production. Int J Mol Sci 2023; 24:14922. [PMID: 37834368 PMCID: PMC10573867 DOI: 10.3390/ijms241914922] [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: 09/04/2023] [Revised: 09/25/2023] [Accepted: 10/04/2023] [Indexed: 10/15/2023] Open
Abstract
Increased glucocorticoid (GC) levels act as a master contributor to central obesity in estrogen-depleted females; however, what factors cause their increased GC production is unclear. Given (1) liver fibroblast growth factor 21 (FGF21) and GCs regulate each other's production in a feed-forward loop, and (2) circulating FGF21 and GCs are parallelly increased in menopausal women and ovariectomized mice, we thus hypothesized that elevation of hepatic FGF21 secretion causes increased GGs production in estrogen-depleted females. Using the ovariectomized mice as a model for menopausal women, we found that ovariectomy (OVX) increased circulating corticosterone levels, which in turn increased visceral adipose Hsd11b1 expression, thus causing visceral obesity in females. In contrast, liver-specific FGF21 knockout (FGF21 LKO) completely reversed OVX-induced high GCs and high visceral adipose Hsd11b1 expression, thus abrogating OVX-induced obesity in females. Even though FGF21 LKO failed to rescue OVX-induced dyslipidemia, hepatic steatosis, and insulin resistance. What's worse, FGF21 LKO even further exacerbated whole-body glucose metabolic dysfunction as evidenced by more impaired glucose and pyruvate tolerance and worsened insulin resistance. Mechanically, we found that FGF21 LKO reduced circulating insulin levels, thus causing the dissociation between decreased central obesity and the improvement of obesity-related metabolic syndromes in OVX mice. Collectively, our results suggest that liver FGF21 plays an essential role in mediating OVX-induced central obesity by promoting GC production. However, lack of liver FGF21 signaling reduces insulin production and in turn causes the dissociation between decreased central obesity and the improvement of obesity-related metabolic syndromes, highlighting a detrimental role for hepatic FGF21 signals in mediating the development of central obesity but a beneficial role in preventing metabolic abnormality from further exacerbation in estrogen-depleted females.
Collapse
Affiliation(s)
- Jiayu Xu
- College of Life Science, Sichuan Agricultural University, Ya’an 625014, China; (J.X.); (X.S.); (H.Z.); (C.W.); (J.L.); (X.P.); (F.M.)
| | - Xinyu Shao
- College of Life Science, Sichuan Agricultural University, Ya’an 625014, China; (J.X.); (X.S.); (H.Z.); (C.W.); (J.L.); (X.P.); (F.M.)
| | - Haozhe Zeng
- College of Life Science, Sichuan Agricultural University, Ya’an 625014, China; (J.X.); (X.S.); (H.Z.); (C.W.); (J.L.); (X.P.); (F.M.)
| | - Chengxi Wang
- College of Life Science, Sichuan Agricultural University, Ya’an 625014, China; (J.X.); (X.S.); (H.Z.); (C.W.); (J.L.); (X.P.); (F.M.)
| | - Jiayi Li
- College of Life Science, Sichuan Agricultural University, Ya’an 625014, China; (J.X.); (X.S.); (H.Z.); (C.W.); (J.L.); (X.P.); (F.M.)
| | - Xiaoqin Peng
- College of Life Science, Sichuan Agricultural University, Ya’an 625014, China; (J.X.); (X.S.); (H.Z.); (C.W.); (J.L.); (X.P.); (F.M.)
| | - Yong Zhuo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611134, China (L.H.)
| | - Lun Hua
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611134, China (L.H.)
| | - Fengyan Meng
- College of Life Science, Sichuan Agricultural University, Ya’an 625014, China; (J.X.); (X.S.); (H.Z.); (C.W.); (J.L.); (X.P.); (F.M.)
| | - Xingfa Han
- College of Life Science, Sichuan Agricultural University, Ya’an 625014, China; (J.X.); (X.S.); (H.Z.); (C.W.); (J.L.); (X.P.); (F.M.)
| |
Collapse
|
34
|
Chen Y, Du X, Kuppa A, Feitosa MF, Bielak LF, O'Connell JR, Musani SK, Guo X, Kahali B, Chen VL, Smith AV, Ryan KA, Eirksdottir G, Allison MA, Bowden DW, Budoff MJ, Carr JJ, Chen YDI, Taylor KD, Oliveri A, Correa A, Crudup BF, Kardia SLR, Mosley TH, Norris JM, Terry JG, Rotter JI, Wagenknecht LE, Halligan BD, Young KA, Hokanson JE, Washko GR, Gudnason V, Province MA, Peyser PA, Palmer ND, Speliotes EK. Genome-wide association meta-analysis identifies 17 loci associated with nonalcoholic fatty liver disease. Nat Genet 2023; 55:1640-1650. [PMID: 37709864 PMCID: PMC10918428 DOI: 10.1038/s41588-023-01497-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 08/07/2023] [Indexed: 09/16/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is common and partially heritable and has no effective treatments. We carried out a genome-wide association study (GWAS) meta-analysis of imaging (n = 66,814) and diagnostic code (3,584 cases versus 621,081 controls) measured NAFLD across diverse ancestries. We identified NAFLD-associated variants at torsin family 1 member B (TOR1B), fat mass and obesity associated (FTO), cordon-bleu WH2 repeat protein like 1 (COBLL1)/growth factor receptor-bound protein 14 (GRB14), insulin receptor (INSR), sterol regulatory element-binding transcription factor 1 (SREBF1) and patatin-like phospholipase domain-containing protein 2 (PNPLA2), as well as validated NAFLD-associated variants at patatin-like phospholipase domain-containing protein 3 (PNPLA3), transmembrane 6 superfamily 2 (TM6SF2), apolipoprotein E (APOE), glucokinase regulator (GCKR), tribbles homolog 1 (TRIB1), glycerol-3-phosphate acyltransferase (GPAM), mitochondrial amidoxime-reducing component 1 (MARC1), microsomal triglyceride transfer protein large subunit (MTTP), alcohol dehydrogenase 1B (ADH1B), transmembrane channel like 4 (TMC4)/membrane-bound O-acyltransferase domain containing 7 (MBOAT7) and receptor-type tyrosine-protein phosphatase δ (PTPRD). Implicated genes highlight mitochondrial, cholesterol and de novo lipogenesis as causally contributing to NAFLD predisposition. Phenome-wide association study (PheWAS) analyses suggest at least seven subtypes of NAFLD. Individuals in the top 10% and 1% of genetic risk have a 2.5-fold to 6-fold increased risk of NAFLD, cirrhosis and hepatocellular carcinoma. These genetic variants identify subtypes of NAFLD, improve estimates of disease risk and can guide the development of targeted therapeutics.
Collapse
Affiliation(s)
- Yanhua Chen
- Department of Internal Medicine, Division of Gastroenterology and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Xiaomeng Du
- Department of Internal Medicine, Division of Gastroenterology and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Annapurna Kuppa
- Department of Internal Medicine, Division of Gastroenterology and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Lawrence F Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Jeffrey R O'Connell
- Department of Endocrinology, Diabetes and Nutrition, University of Maryland - Baltimore, Baltimore, MD, USA
| | - Solomon K Musani
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Bratati Kahali
- Department of Internal Medicine, Division of Gastroenterology and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
| | - Vincent L Chen
- Department of Internal Medicine, Division of Gastroenterology and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Albert V Smith
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Kathleen A Ryan
- Department of Endocrinology, Diabetes and Nutrition, University of Maryland - Baltimore, Baltimore, MD, USA
| | | | - Matthew A Allison
- Department of Family Medicine, University of California San Diego, San Diego, CA, USA
| | - Donald W Bowden
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Matthew J Budoff
- Department of Internal Medicine, Lundquist Institute at Harbor-UCLA, Torrance, CA, USA
| | - John Jeffrey Carr
- Department of Radiology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Yii-Der I Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Antonino Oliveri
- Department of Internal Medicine, Division of Gastroenterology and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Adolfo Correa
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Breland F Crudup
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Thomas H Mosley
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jill M Norris
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
| | - James G Terry
- Department of Radiology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Lynne E Wagenknecht
- Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Brian D Halligan
- Department of Internal Medicine, Division of Gastroenterology and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Kendra A Young
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
| | - John E Hokanson
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
| | - George R Washko
- Department of Medicine, Division of Pulmonary and Critical Care, Brigham and Women's Hospital, Boston, MA, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Department of Medicine, University of Iceland, Reykjavik, Iceland
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Patricia A Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Nicholette D Palmer
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Elizabeth K Speliotes
- Department of Internal Medicine, Division of Gastroenterology and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
35
|
Bourebaba N, Sikora M, Qasem B, Bourebaba L, Marycz K. Sex hormone-binding globulin (SHBG) mitigates ER stress and improves viability and insulin sensitivity in adipose-derived mesenchymal stem cells (ASC) of equine metabolic syndrome (EMS)-affected horses. Cell Commun Signal 2023; 21:230. [PMID: 37697311 PMCID: PMC10496240 DOI: 10.1186/s12964-023-01254-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/01/2023] [Indexed: 09/13/2023] Open
Abstract
BACKGROUND Equine metabolic syndrome (EMS), which encompasses insulin resistance, low-grade inflammation and predisposition to laminitis is a critical endocrine disorder among the most prevalent conditions affecting horses from different breeds. According to the most recent research, low human sex hormone-binding globulin (SHBG) serum levels correlate with an increased risk of obesity, insulin resistance and diabetes, and may contribute to overall metabolic dysregulations. This study aimed to test whether exogenous SHBG could protect EMS affected adipose-derived stromal stem cells (EqASCEMS) from apoptosis, oxidative stress, ER stress and thus improve insulin sensitivity. METHODS EqASCEMS wells were treated with two different concentrations (50 and 100 nM) of exogenous SHBG, whose biocompatibility was tested after 24, 48 and 72 h of incubation. Several parameters including cell viability, apoptosis, cell cycle, reactive oxygen species levels, ER stress, Pi3K/MAPK activation and insulin transducers expression were analysed. RESULTS Obtained data demonstrated that exogenous SHBG treatment significantly promoted ASCs cells proliferation, cell cycle and survival with reduced expression of p53 and p21 pro-apoptotic mediators. Furthermore, SHBG alleviated the oxidative stress caused by EMS and reduced the overaccumulation of intracellular ROS, by reducing ROS + cell percentage and regulating gene expression of endogenous antioxidant enzymes (Sod 1, Cat, GPx), SHBG treatment exhibited antioxidant activity by modulating total nitric oxide (NO) levels in EMS cells as well. SHBG treatment dampened the activation of ER stress sensors and effectors in EqASCEMS cells via the upregulation of MiR-7a-5p, the decrease in the expression levels of ATF-6, CHOP and eiF2A and the restoration of PDIA3 chaperone protein levels. As a consequence, SHBG application substantially improved insulin sensitivity through the modulation of Pi3K/Akt/Glut4 insulin signalling cascades. CONCLUSION Our results suggest that the SHBG is endowed with crucial beneficial effects on ASCs metabolic activities and could serve as a valuable therapeutic target for the development of efficient EMS treatment protocols. Video Abstract.
Collapse
Affiliation(s)
- Nabila Bourebaba
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland
| | - Mateusz Sikora
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland
| | - Badr Qasem
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland
| | - Lynda Bourebaba
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland
| | - Krzysztof Marycz
- Department of Medicine and Epidemiology, UC Davis School of Veterinary Medicine, Davis, CA, 95516, USA.
- Department of Veterinary Medicine and Epidemiology, Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, CA, USA.
| |
Collapse
|
36
|
Hong S, Park SK, Lee J, Park SH, Kim YS, Park JH, Yu S, Lee YG. Patulin Ameliorates Hypertrophied Lipid Accumulation and Lipopolysaccharide-Induced Inflammatory Response by Modulating Mitochondrial Respiration. Antioxidants (Basel) 2023; 12:1750. [PMID: 37760053 PMCID: PMC10526082 DOI: 10.3390/antiox12091750] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/06/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
Patulin (PAT) is a natural mycotoxin found in decaying pome fruits. Although some toxicological studies have been conducted on PAT, recent research has highlighted its anticancer and antifungal effects. However, studies have yet to examine the effects and molecular mechanisms of PAT in other metabolic diseases. Obesity is a chronic disease caused by excessive food intake and abnormal lifestyle, leading to low-grade inflammation. Therefore, this study aimed to elucidate the effect of PAT on obesity at the cellular level. PAT treatment reduced lipid accumulation, suppressed glucose and LDL uptake, inhibited lipid deposition and triglyceride synthesis, upregulated fatty acid oxidation-related genes (Pgc1α), and downregulated adipogenic/lipogenic genes (Pparγ and C/ebpα) in hypertrophied 3T3-L1 adipocytes. Additionally, PAT treatment enhanced mitochondrial respiration and mass in differentiated adipocytes and alleviated inflammatory response in activated RAW 264.7 macrophages. Moreover, PAT treatment downregulated pro-inflammatory genes (il-6, Tnf-α, Cox-2, and inos), suppressed lipopolysaccharide (LPS)-induced increase in inflammatory mediators (IL-6, TNF-α, and NO), and restored mitochondrial oxidative function in LPS-stimulated macrophages by improving oxygen consumption and mitochondrial integrity and suppressing ROS generation. Overall, these findings suggest a potential for PAT in the prevention of lipid accumulation and inflammation-related disorders.
Collapse
Affiliation(s)
- Seulmin Hong
- Personalized Diet Research Group, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; (S.H.); (S.K.P.); (J.L.); (S.H.P.); (J.-H.P.)
- Department of Food Science & Technology, Jeonbuk National University, Jeonju 54896, Republic of Korea;
| | - Seon Kyeong Park
- Personalized Diet Research Group, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; (S.H.); (S.K.P.); (J.L.); (S.H.P.); (J.-H.P.)
| | - Jangho Lee
- Personalized Diet Research Group, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; (S.H.); (S.K.P.); (J.L.); (S.H.P.); (J.-H.P.)
| | - Soo Hyun Park
- Personalized Diet Research Group, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; (S.H.); (S.K.P.); (J.L.); (S.H.P.); (J.-H.P.)
| | - Young-Soo Kim
- Department of Food Science & Technology, Jeonbuk National University, Jeonju 54896, Republic of Korea;
| | - Jae-Ho Park
- Personalized Diet Research Group, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; (S.H.); (S.K.P.); (J.L.); (S.H.P.); (J.-H.P.)
| | - Seungmin Yu
- Personalized Diet Research Group, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; (S.H.); (S.K.P.); (J.L.); (S.H.P.); (J.-H.P.)
| | - Yu Geon Lee
- Personalized Diet Research Group, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; (S.H.); (S.K.P.); (J.L.); (S.H.P.); (J.-H.P.)
| |
Collapse
|
37
|
Monmai C, Kim JS, Sim HB, Yun DW, Oh SD, Rha ES, Kim JJ, Baek SH. Protopanaxadiol-Enriched Rice Exerted Antiadipogenic Activity during 3T3-L1 Differentiation and Anti-Inflammatory Activity in 3T3-L1 Adipocytes. Pharmaceutics 2023; 15:2123. [PMID: 37631337 PMCID: PMC10458103 DOI: 10.3390/pharmaceutics15082123] [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: 07/18/2023] [Revised: 07/28/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Ginseng is a traditional medicine with health benefits for humans. Protopanaxadiol (PPD) is an important bioactive compound found in ginseng. Transgenic rice containing PPD has been generated previously. In the present study, extracts of this transgenic rice were evaluated to assess their antiadipogenic and anti-inflammatory activities. During adipogenesis, cells were treated with transgenic rice seed extracts. The results revealed that the concentrations of the rice seed extracts tested in this study did not affect cell viability at 3 days post-treatment. However, the rice seed extracts significantly reduced the accumulation of lipids in cells and suppressed the activation of CCAAT/enhancer-binding protein α (C/EBPα) and peroxisome proliferator-activated receptor γ (PPARγ), which in turn inhibited the expression of adipogenesis-related mRNAs, such as adiponectin, PPARγ, C/EBPα, sterol regulatory element-binding protein 1, glucose transport member 4, and fatty acid synthase. In adipocytes, the extracts significantly reduced the mRNA expression of inflammation-related factors following LPS treatment. The activation of NF-κB p65 and ERK 1/2 was inhibited in extract-treated adipocytes. Moreover, treatment with extract #8 markedly reduced the cell population of the G2/M phase. Collectively, these results indicate that transgenic rice containing PPD may act as an obesity-reducing and/or -preventing agent.
Collapse
Affiliation(s)
- Chaiwat Monmai
- Department of Agricultural Life Science, Sunchon National University, Sunchon 59722, Republic of Korea; (C.M.); (J.-S.K.); (E.-S.R.)
| | - Jin-Suk Kim
- Department of Agricultural Life Science, Sunchon National University, Sunchon 59722, Republic of Korea; (C.M.); (J.-S.K.); (E.-S.R.)
| | - Hyun Bo Sim
- Department of Biomedical Science, Sunchon National University, Sunchon 57922, Republic of Korea;
| | - Doh-Won Yun
- National Institute of Agricultural Sciences, Rural Development Administration, Jeonju 54874, Republic of Korea; (D.-W.Y.); (S.-D.O.)
| | - Sung-Dug Oh
- National Institute of Agricultural Sciences, Rural Development Administration, Jeonju 54874, Republic of Korea; (D.-W.Y.); (S.-D.O.)
| | - Eui-Shik Rha
- Department of Agricultural Life Science, Sunchon National University, Sunchon 59722, Republic of Korea; (C.M.); (J.-S.K.); (E.-S.R.)
| | - Jong-Jin Kim
- Department of Biomedical Science, Sunchon National University, Sunchon 57922, Republic of Korea;
| | - So-Hyeon Baek
- Department of Agricultural Life Science, Sunchon National University, Sunchon 59722, Republic of Korea; (C.M.); (J.-S.K.); (E.-S.R.)
| |
Collapse
|
38
|
Mubtasim N, Gollahon L. Characterizing 3T3-L1 MBX Adipocyte Cell Differentiation Maintained with Fatty Acids as an In Vitro Model to Study the Effects of Obesity. Life (Basel) 2023; 13:1712. [PMID: 37629569 PMCID: PMC10455818 DOI: 10.3390/life13081712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
The increasing prevalence of obesity has prompted intensive research into understanding its role in pathogenesis and designing appropriate treatments. To determine the signals generated from the interaction of fat cells with a target organ, a reliable white adipocyte model in vitro is needed. Differentiated fibroblasts are the most extensively studied using in vitro cell models of white adipocytes. However, it can be argued that differentiated fibroblasts minimally recapitulate the consequences of obesity. Here, we describe 3T3-L1 MBX cells as a culture model for studying obese adipocytes and their effects. Differentiation of 3T3-L1 MBX cells was at first optimized and then maintained in the presence of fatty acids cocktail combination to induce the obese condition. Lipid accumulation and adipokine secretion profiles were analyzed. Results showed that fatty acid-maintained, differentiated 3T3-L1 MBX cells had significantly greater accumulation of lipids and significant changes in the adipokine secretions in comparison to differentiated 3T3-L1 MBX cells maintained in medium without fatty acids. To elucidate the molecular changes associated with adipogenesis and lipid accumulation profile of 3T3-L1 MBX cells, we have also explored the expression of some of the regulatory proteins related to the development and maintenance of adipocytes from the preadipocyte lineage.
Collapse
Affiliation(s)
| | - Lauren Gollahon
- Department of Biological Sciences, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA;
| |
Collapse
|
39
|
Duan YY, Chen XF, Zhu RJ, Jia YY, Huang XT, Zhang M, Yang N, Dong SS, Zeng M, Feng Z, Zhu DL, Wu H, Jiang F, Shi W, Hu WX, Ke X, Chen H, Liu Y, Jing RH, Guo Y, Li M, Yang TL. High-throughput functional dissection of noncoding SNPs with biased allelic enhancer activity for insulin resistance-relevant phenotypes. Am J Hum Genet 2023; 110:1266-1288. [PMID: 37506691 PMCID: PMC10432149 DOI: 10.1016/j.ajhg.2023.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Most of the single-nucleotide polymorphisms (SNPs) associated with insulin resistance (IR)-relevant phenotypes by genome-wide association studies (GWASs) are located in noncoding regions, complicating their functional interpretation. Here, we utilized an adapted STARR-seq to evaluate the regulatory activities of 5,987 noncoding SNPs associated with IR-relevant phenotypes. We identified 876 SNPs with biased allelic enhancer activity effects (baaSNPs) across 133 loci in three IR-relevant cell lines (HepG2, preadipocyte, and A673), which showed pervasive cell specificity and significant enrichment for cell-specific open chromatin regions or enhancer-indicative markers (H3K4me1, H3K27ac). Further functional characterization suggested several transcription factors (TFs) with preferential allelic binding to baaSNPs. We also incorporated multi-omics data to prioritize 102 candidate regulatory target genes for baaSNPs and revealed prevalent long-range regulatory effects and cell-specific IR-relevant biological functional enrichment on them. Specifically, we experimentally verified the distal regulatory mechanism at IRS1 locus, in which rs952227-A reinforces IRS1 expression by long-range chromatin interaction and preferential binding to the transcription factor HOXC6 to augment the enhancer activity. Finally, based on our STARR-seq screening data, we predicted the enhancer activity of 227,343 noncoding SNPs associated with IR-relevant phenotypes (fasting insulin adjusted for BMI, HDL cholesterol, and triglycerides) from the largest available GWAS summary statistics. We further provided an open resource (http://www.bigc.online/fnSNP-IR) for better understanding genetic regulatory mechanisms of IR-relevant phenotypes.
Collapse
Affiliation(s)
- Yuan-Yuan Duan
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Xiao-Feng Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Ren-Jie Zhu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Ying-Ying Jia
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Xiao-Ting Huang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Meng Zhang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Ning Yang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Shan-Shan Dong
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Mengqi Zeng
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Zhihui Feng
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Dong-Li Zhu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Hao Wu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Feng Jiang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Wei Shi
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Wei-Xin Hu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Xin Ke
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Hao Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Rui-Hua Jing
- Department of Ophthalmology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710000, China
| | - Yan Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Meng Li
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| | - Tie-Lin Yang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
40
|
Efthymiou V, Ding L, Balaz M, Sun W, Balazova L, Straub LG, Dong H, Simon E, Ghosh A, Perdikari A, Keller S, Ghoshdastider U, Horvath C, Moser C, Hamilton B, Neubauer H, Wolfrum C. Inhibition of AXL receptor tyrosine kinase enhances brown adipose tissue functionality in mice. Nat Commun 2023; 14:4162. [PMID: 37443109 PMCID: PMC10344962 DOI: 10.1038/s41467-023-39715-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
The current obesity epidemic and high prevalence of metabolic diseases necessitate efficacious and safe treatments. Brown adipose tissue in this context is a promising target with the potential to increase energy expenditure, however no pharmacological treatments activating brown adipose tissue are currently available. Here, we identify AXL receptor tyrosine kinase as a regulator of adipose function. Pharmacological and genetic inhibition of AXL enhance thermogenic capacity of brown and white adipocytes, in vitro and in vivo. Mechanistically, these effects are mediated through inhibition of PI3K/AKT/PDE signaling pathway, resulting in induction of nuclear FOXO1 localization and increased intracellular cAMP levels via PDE3/4 inhibition and subsequent stimulation of the PKA-ATF2 pathway. In line with this, both constitutive Axl deletion as well as inducible adipocyte-specific Axl deletion protect animals from diet-induced obesity concomitant with increases in energy expenditure. Based on these data, we propose AXL receptor as a target for the treatment of obesity.
Collapse
Affiliation(s)
- Vissarion Efthymiou
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
- Joslin Diabetes Center, Section of Integrative Physiology and Metabolism, Research Division, Harvard Medical School, Boston, MA, USA
| | - Lianggong Ding
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
| | - Miroslav Balaz
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
- Laboratory of Cellular and Molecular Metabolism, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Wenfei Sun
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lucia Balazova
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
- Laboratory of Cellular and Molecular Metabolism, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Leon G Straub
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
- Institute of Child Nutrition, Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Karlsruhe, Germany
| | - Hua Dong
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Eric Simon
- Department of Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Adhideb Ghosh
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
| | - Aliki Perdikari
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
| | - Svenja Keller
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
- Mechanisms of Inherited Kidney Diseases Group, Institute of Physiology, University of Zurich, 8057, Zurich, Switzerland
| | - Umesh Ghoshdastider
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
| | - Carla Horvath
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
| | - Caroline Moser
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland
| | - Bradford Hamilton
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Heike Neubauer
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Christian Wolfrum
- ETH Zürich - Swiss Federal Institute of Technology, Department of Health Sciences and Technology, Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Schwerzenbach, Switzerland.
| |
Collapse
|
41
|
Hariri A, Mirian M, Zarrabi A, Kohandel M, Amini-Pozveh M, Aref AR, Tabatabaee A, Prabhakar PK, Sivakumar PM. The circadian rhythm: an influential soundtrack in the diabetes story. Front Endocrinol (Lausanne) 2023; 14:1156757. [PMID: 37441501 PMCID: PMC10333930 DOI: 10.3389/fendo.2023.1156757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/03/2023] [Indexed: 07/15/2023] Open
Abstract
Type 2 Diabetes Mellitus (T2DM) has been the main category of metabolic diseases in recent years due to changes in lifestyle and environmental conditions such as diet and physical activity. On the other hand, the circadian rhythm is one of the most significant biological pathways in humans and other mammals, which is affected by light, sleep, and human activity. However, this cycle is controlled via complicated cellular pathways with feedback loops. It is widely known that changes in the circadian rhythm can alter some metabolic pathways of body cells and could affect the treatment process, particularly for metabolic diseases like T2DM. The aim of this study is to explore the importance of the circadian rhythm in the occurrence of T2DM via reviewing the metabolic pathways involved, their relationship with the circadian rhythm from two perspectives, lifestyle and molecular pathways, and their effect on T2DM pathophysiology. These impacts have been demonstrated in a variety of studies and led to the development of approaches such as time-restricted feeding, chronotherapy (time-specific therapies), and circadian molecule stabilizers.
Collapse
Affiliation(s)
- Amirali Hariri
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mina Mirian
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, Türkiye
| | - Mohammad Kohandel
- Department of Applied Mathematics, Faculty of Mathematics, University of Waterloo, Waterloo, ON, Canada
| | - Maryam Amini-Pozveh
- Department of Prosthodontics Dentistry, Dental Materials Research Center, Dental Research Institute, School of Dentistry, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana Farber Cancer Institute, Boston, MA, United States
- Translational Sciences, Xsphera Biosciences Inc., Boston, MA, United States
| | - Aliye Tabatabaee
- School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Pranav Kumar Prabhakar
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Lovely Professional University, Phagwara, Punjab, India
- Division of Research and Development, Lovely Professional University, Phagwara Punjab, India
| | - Ponnurengam Malliappan Sivakumar
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam
- School of Medicine and Pharmacy, Duy Tan University, Da Nang, Vietnam
| |
Collapse
|
42
|
Shijina BN, Radhika A, Sherin S, Biju PG. Vindoline Exhibits Anti-Diabetic Potential in Insulin-Resistant 3T3-L1 Adipocytes and L6 Skeletal Myoblasts. Nutrients 2023; 15:2865. [PMID: 37447192 DOI: 10.3390/nu15132865] [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: 06/01/2023] [Revised: 06/18/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) emerged as a major health care concern in modern society, primarily due to lifestyle changes and dietary habits. Obesity-induced insulin resistance is considered as the major pathogenic factor in T2DM. In this study, we investigated the effect of vindoline, an indole alkaloid of Catharanthus roseus on insulin resistance (IR), oxidative stress and inflammatory responses in dexamethasone (IR inducer)-induced dysfunctional 3T3-L1 adipocytes and high-glucose-induced insulin-resistant L6-myoblast cells. Results showed that dexamethasone-induced dysfunctional 3T3-L1 adipocytes treated with different concentrations of vindoline significantly enhanced basal glucose consumption, accompanied by increased expression of GLUT-4, IRS-1 and adiponectin. Similarly, vindoline-treated insulin-resistant L6 myoblasts exhibited significantly enhanced glycogen content accompanied with upregulation of IRS-1 and GLUT-4. Thus, in vitro studies of vindoline in insulin resistant skeleton muscle and dysfunctional adipocytes confirmed that vindoline treatment significantly mitigated insulin resistance in myotubes and improved functional status of adipocytes. These results demonstrated that vindoline has the potential to be used as a therapeutic agent to ameliorate obesity-induced T2DM-associated insulin resistance profile in adipocytes and skeletal muscles.
Collapse
Affiliation(s)
- Beegum Noorjahan Shijina
- Department of Biochemistry, University of Kerala, Kariavattom Campus, Thiruvananthapuram 695581, Kerala, India
| | - Achuthan Radhika
- Department of Biochemistry, Government College, Kariavattom, Thiruvananthapuram 695581, Kerala, India
| | - Sainulabdeen Sherin
- Department of Biochemistry, PMS College of Dental Science and Research, Vattappara, Thiruvananthapuram 695028, Kerala, India
| | - Prabath Gopalakrishnan Biju
- Department of Biochemistry, University of Kerala, Kariavattom Campus, Thiruvananthapuram 695581, Kerala, India
| |
Collapse
|
43
|
Rivas-Domínguez A, Mohamed-Mohamed H, Jimenez-Palomares M, García-Morales V, Martinez-Lopez L, Orta ML, Ramos-Rodriguez JJ, Bermudez-Pulgarin B. Metabolic Disturbance of High-Saturated Fatty Acid Diet in Cognitive Preservation. Int J Mol Sci 2023; 24:ijms24098042. [PMID: 37175748 PMCID: PMC10178694 DOI: 10.3390/ijms24098042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/16/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Aging continues to be the main cause of the development of Alzheimer's, although it has been described that certain chronic inflammatory pathologies can negatively influence the progress of dementia, including obesity and hyperlipidemia. In this sense, previous studies have shown a relationship between low-density lipoprotein receptor (LDLR) and the amyloid-beta (Aβ) binding activity, one of the main neuropathological features of Alzheimer's disease (AD). LDLR is involved in several processes, including lipid transport, regulation of inflammatory response and lipid metabolism. From this perspective, LDLR-/- mice are a widely accepted animal model for the study of pathologies associated with alterations in lipid metabolism, such as familial hypercholesterolemia, cardiovascular diseases, metabolic syndrome, or early cognitive decline. In this context, we induced hyperlipidemia in LDLR-/- mice after feeding with a high-saturated fatty acid diet (HFD) for 44 weeks. LDLR-/--HFD mice exhibited obesity, hypertriglyceridemia, higher glucose levels, and early hepatic steatosis. In addition, HFD increased plasmatic APOE and ubiquitin 60S levels. These proteins are related to neuronal integrity and health maintenance. In agreement, we detected mild cognitive dysfunctions in mice fed with HFD, whereas LDLR-/--HFD mice showed a more severe and evident affectation. Our data suggest central nervous system dysfunction is associated with a well-established metabolic syndrome. As a late consequence, metabolic syndrome boots many behavioral and pathological alterations recognized in dementia, supporting that the control of metabolic parameters could improve cognitive preservation and prognosis.
Collapse
Affiliation(s)
| | - Himan Mohamed-Mohamed
- Department of Physiology, Faculty of Health Sciences (Ceuta), University of Granada, 51001 Ceuta, Spain
| | | | - Victoria García-Morales
- Department of Biomedicine, Biotechnology and Public Health, University of Cádiz, 11003 Cádiz, Spain
| | | | - Manuel Luis Orta
- Department of Cellular Biology, University of Seville, 41009 Seville, Spain
| | - Juan José Ramos-Rodriguez
- Department of Physiology, Faculty of Health Sciences (Ceuta), University of Granada, 51001 Ceuta, Spain
| | | |
Collapse
|
44
|
Jang WY, Hwang JY, Cho JY. Ginsenosides from Panax ginseng as Key Modulators of NF-κB Signaling Are Powerful Anti-Inflammatory and Anticancer Agents. Int J Mol Sci 2023; 24:6119. [PMID: 37047092 PMCID: PMC10093821 DOI: 10.3390/ijms24076119] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
Nuclear factor kappa B (NF-κB) signaling pathways progress inflammation and immune cell differentiation in the host immune response; however, the uncontrollable stimulation of NF-κB signaling is responsible for several inflammatory illnesses regardless of whether the conditions are acute or chronic. Innate immune cells, such as macrophages, microglia, and Kupffer cells, secrete pro-inflammatory cytokines, such as TNF-α, IL-6, and IL-1β, via the activation of NF-κB subunits, which may lead to the damage of normal cells, including neurons, cardiomyocytes, hepatocytes, and alveolar cells. This results in the occurrence of neurodegenerative disorders, cardiac infarction, or liver injury, which may eventually lead to systemic inflammation or cancer. Recently, ginsenosides from Panax ginseng, a historical herbal plant used in East Asia, have been used as possible options for curing inflammatory diseases. All of the ginsenosides tested target different steps of the NF-κB signaling pathway, ameliorating the symptoms of severe illnesses. Moreover, ginsenosides inhibit the NF-κB-mediated activation of cancer metastasis and immune resistance, significantly attenuating the expression of MMPs, Snail, Slug, TWIST1, and PD-L1. This review introduces current studies on the therapeutic efficacy of ginsenosides in alleviating NF-κB responses and emphasizes the critical role of ginsenosides in severe inflammatory diseases as well as cancers.
Collapse
Affiliation(s)
| | | | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
45
|
Knocking Down CDKN2A in 3D hiPSC-Derived Brown Adipose Progenitors Potentiates Differentiation, Oxidative Metabolism and Browning Process. Cells 2023; 12:cells12060870. [PMID: 36980212 PMCID: PMC10047013 DOI: 10.3390/cells12060870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 03/16/2023] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) have the potential to be differentiated into any cell type, making them a relevant tool for therapeutic purposes such as cell-based therapies. In particular, they show great promise for obesity treatment as they represent an unlimited source of brown/beige adipose progenitors (hiPSC-BAPs). However, the low brown/beige adipocyte differentiation potential in 2D cultures represents a strong limitation for clinical use. In adipose tissue, besides its cell cycle regulator functions, the cyclin-dependent kinase inhibitor 2A (CDKN2A) locus modulates the commitment of stem cells to the brown-like type fate, mature adipocyte energy metabolism and the browning of adipose tissue. Here, using a new method of hiPSC-BAPs 3D culture, via the formation of an organoid-like structure, we silenced CDKN2A expression during hiPSC-BAP adipogenic differentiation and observed that knocking down CDKN2A potentiates adipogenesis, oxidative metabolism and the browning process, resulting in brown-like adipocytes by promoting UCP1 expression and beiging markers. Our results suggest that modulating CDKN2A levels could be relevant for hiPSC-BAPs cell-based therapies.
Collapse
|
46
|
Abstract
Brown adipose tissue (BAT) displays the unique capacity to generate heat through uncoupled oxidative phosphorylation that makes it a very attractive therapeutic target for cardiometabolic diseases. Here, we review BAT cellular metabolism, its regulation by the central nervous and endocrine systems and circulating metabolites, the plausible roles of this tissue in human thermoregulation, energy balance, and cardiometabolic disorders, and the current knowledge on its pharmacological stimulation in humans. The current definition and measurement of BAT in human studies relies almost exclusively on BAT glucose uptake from positron emission tomography with 18F-fluorodeoxiglucose, which can be dissociated from BAT thermogenic activity, as for example in insulin-resistant states. The most important energy substrate for BAT thermogenesis is its intracellular fatty acid content mobilized from sympathetic stimulation of intracellular triglyceride lipolysis. This lipolytic BAT response is intertwined with that of white adipose (WAT) and other metabolic tissues, and cannot be independently stimulated with the drugs tested thus far. BAT is an interesting and biologically plausible target that has yet to be fully and selectively activated to increase the body's thermogenic response and shift energy balance. The field of human BAT research is in need of methods able to directly, specifically, and reliably measure BAT thermogenic capacity while also tracking the related thermogenic responses in WAT and other tissues. Until this is achieved, uncertainty will remain about the role played by this fascinating tissue in human cardiometabolic diseases.
Collapse
Affiliation(s)
- André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, J1H 5N4, Canada
| | - Denis P Blondin
- Division of Neurology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, J1H 5N4, Canada
| | | | - Denis Richard
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Quebec City, Quebec, G1V 4G5, Canada
| |
Collapse
|
47
|
Panic A, Sudar-Milovanovic E, Stanimirovic J, Obradovic M, Zafirovic S, Soskic S, Isenovic ER. Does oestradiol treatment alleviate obesity-induced oxidative stress in the male liver? Med Hypotheses 2023. [DOI: 10.1016/j.mehy.2023.111049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
|
48
|
Zhao B, Zhang H, Zhao D, Liang Y, Qiao L, Liu J, Pan Y, Yang K, Liu W. circINSR Inhibits Adipogenic Differentiation of Adipose-Derived Stromal Vascular Fractions through the miR-152/ MEOX2 Axis in Sheep. Int J Mol Sci 2023; 24:ijms24043501. [PMID: 36834919 PMCID: PMC9964708 DOI: 10.3390/ijms24043501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/12/2023] Open
Abstract
Adipose tissue plays a crucial role in energy metabolism. Several studies have shown that circular RNA (circRNA) is involved in the regulation of fat development and lipid metabolism. However, little is known about their involvement in the adipogenic differentiation of ovine stromal vascular fractions (SVFs). Here, based on previous sequencing data and bioinformatics analysis, a novel circINSR was identified in sheep, which acts as a sponge to promote miR-152 in inhibiting the adipogenic differentiation of ovine SVFs. The interactions between circINSR and miR-152 were examined using bioinformatics, luciferase assays, and RNA immunoprecipitation. Of note, we found that circINSR was involved in adipogenic differentiation via the miR-152/mesenchyme homeobox 2 (MEOX2) pathway. MEOX2 inhibited adipogenic differentiation of ovine SVFs and miR-152 inhibited the expression of MEOX2. In other words, circINSR directly isolates miR-152 in the cytoplasm and inhibits its ability to promote adipogenic differentiation of ovine SVFs. In summary, this study revealed the role of circINSR in the adipogenic differentiation of ovine SVFs and its regulatory mechanisms, providing a reference for further interpretation of the development of ovine fat and its regulatory mechanisms.
Collapse
|
49
|
Ayaki S, Mii T, Matsuno K, Tokuyama T, Tokuyama T, Tokuyama T, Uyama T, Ueda N. β-1,4-Galactan suppresses lipid synthesis in sebaceous gland cells via TLR4. J Biochem 2023; 173:85-94. [PMID: 36288613 DOI: 10.1093/jb/mvac085] [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: 06/17/2022] [Revised: 09/28/2022] [Accepted: 10/15/2022] [Indexed: 02/07/2023] Open
Abstract
Sebum is a lipid mixture secreted from sebaceous glands of the skin. The excessive secretion of sebum causes acne vulgaris and seborrheic dermatitis, while its deficiency causes xerosis. Therefore, the appropriate control of sebum secretion is crucially important to keep the skin healthy. In the present study, we evaluated the effects of naturally occurring polysaccharides on lipid biosynthesis in hamster sebaceous gland cells. Among the tested polysaccharides, β-1,4-galactan, the main chain of type I arabinogalactan, most potently suppressed lipid synthesis in the sebaceous gland cells as analysed by oil red O staining. Toll-like receptor (TLR)4 inhibitors counteracted this suppressive effect and lipopolysaccharide, a TLR4 ligand, mimicked this effect, suggesting the involvement of the TLR4 signalling pathway. In the cells β-1,4-galactan significantly decreased mRNA levels of lipogenesis-related transcription factors (peroxisomeGraphical Abstract$\includegraphics{\bwartpath }$ proliferator-activated receptor γ and sterol regulatory element-binding protein 1) and enzymes (acetyl-CoA carboxylase and fatty acid synthase) as well as the glucose transporter GLUT4. Furthermore, β-1,4-galactan increased the production of lactic acid serving as a natural moisturizing factor and enhanced the proliferation of sebaceous gland cells. These results suggest potential of β-1,4-galactan as a material with therapeutic and cosmetic values for the skin.
Collapse
Affiliation(s)
- Satomi Ayaki
- Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa 761-0793, Japan.,Yushin Brewer Co., Ltd., Ayagawa, Kagawa 761-2307, Japan
| | - Tomohiro Mii
- Yushin Brewer Co., Ltd., Ayagawa, Kagawa 761-2307, Japan
| | - Kosuke Matsuno
- Yushin Brewer Co., Ltd., Ayagawa, Kagawa 761-2307, Japan
| | | | | | | | - Toru Uyama
- Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa 761-0793, Japan
| | - Natsuo Ueda
- Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa 761-0793, Japan
| |
Collapse
|
50
|
Castillo P, Pomar CA, Palou A, Palou M, Picó C. Influence of Maternal Metabolic Status and Diet during the Perinatal Period on the Metabolic Programming by Leptin Ingested during the Suckling Period in Rats. Nutrients 2023; 15:nu15030570. [PMID: 36771278 PMCID: PMC9921535 DOI: 10.3390/nu15030570] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
We aimed to analyze the long-term metabolic effects of leptin supplementation at physiological doses during suckling in the offspring of diet-induced obese rats, together with the potential benefits of improving maternal diet during lactation. Thus, the offspring of: dams fed standard-diet (SD) (CON-dams), dams fed western-diet (WD) before and during gestation and lactation (WD-dams), and dams fed as WD-dams but moved to SD during lactation (REV-dams) were supplemented throughout suckling with leptin or vehicle, and fed SD or WD from weaning to four months. Under SD, leptin treatment significantly improved metabolic profile and body fat accumulation, with stronger effects in the male offspring of CON-dams and REV-dams. Under WD, the offspring of WD-dams presented metabolic alterations that were not evident in the offspring of REV-dams. Moreover, leptin supplementation improved glucose homeostasis in the male offspring of REV-dams. Conversely, leptin supplementation in females born to WD-dams and fed WD from weaning resulted in impaired insulin sensitivity and increased hepatic lipid content. These results highlight the importance of a balanced maternal diet during the perinatal period, especially lactation, for the subsequent metabolic health of the offspring and for the beneficial effects of leptin supplementation during suckling, more evident in the male offspring.
Collapse
Affiliation(s)
- Pedro Castillo
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain
- Health Research Institute of the Balearic Islands IdISBa, 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
| | - Catalina Amadora Pomar
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain
- Health Research Institute of the Balearic Islands IdISBa, 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
| | - Andreu Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain
- Health Research Institute of the Balearic Islands IdISBa, 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
| | - Mariona Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain
- Health Research Institute of the Balearic Islands IdISBa, 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-971172373
| | - Catalina Picó
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain
- Health Research Institute of the Balearic Islands IdISBa, 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
| |
Collapse
|