1
|
Chen Y, Jiang Q, Xing X, Yuan T, Li P. Clinical research progress on β-cell dysfunction in T2DM development in the Chinese population. Rev Endocr Metab Disord 2024:10.1007/s11154-024-09914-9. [PMID: 39382753 DOI: 10.1007/s11154-024-09914-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/06/2024] [Indexed: 10/10/2024]
Abstract
The prevalence of type-2 diabetes mellitus (T2DM) has increased over 10-fold in the past 40 years in China, which now has the largest T2DM population in the world. Insulin resistance and β-cell dysfunction are the typical features of T2DM. Although both factors play a role, decreased β-cell function and β-cell mass are the predominant factors for progression to T2DM. Considering the differences between Chinese T2DM patients and those of other ethnicities, it is important to characterize β-cell dysfunction in Chinese patients during T2DM progression. Herein, we reviewed the studies on the relationships between β-cell function and T2DM progression in the Chinese population and discussed the differences among individuals of varying ethnicities. Meanwhile, we summarized the risk factors and current treatments of T2DM in Chinese individuals and discussed their impacts on β-cell function with the hope of identifying a better T2DM therapy.
Collapse
Affiliation(s)
- Yibing Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing, 100050, China
| | - Qian Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing, 100050, China
| | - Xiaowei Xing
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing, 100050, China
| | - Tao Yuan
- Department of Endocrinology, Key Laboratory of Endocrinology of Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Pingping Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
- Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, 100050, China.
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing, 100050, China.
| |
Collapse
|
2
|
Pourhassan H, Murphy L, Aldoss I. Glucocorticoid Therapy in Acute Lymphoblastic Leukemia: Navigating Short-Term and Long-Term Effects and Optimal Regimen Selection. Curr Hematol Malig Rep 2024; 19:175-185. [PMID: 38867099 DOI: 10.1007/s11899-024-00735-w] [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] [Accepted: 05/16/2024] [Indexed: 06/14/2024]
Abstract
PURPOSE OF REVIEW Glucocorticoids are a mainstay in acute lymphoblastic leukemia treatment and lack of early response is predictive for overall disease prognosis. Given the vital position of glucocorticoids and well known long and short-term side effects associated with differing glucocorticoids, we aim to highlight the wide breadth of historical and more contemporary data to describe the current landscape of glucocorticoid use in this arena. RECENT FINDINGS Emerging studies aim to overcome issues such as steroid resistance and to optimize the antileukemic effects of glucocorticoids while aiming to mitigate the risks and side effects associated with their exposure. Glucocorticoids have and likely always will be a fundamental component of acute lymphoblastic leukemia treatment and understanding how to navigate short- and long-term effects and how to optimize regimens is at the heart of continued treatment success.
Collapse
Affiliation(s)
- Hoda Pourhassan
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| | - Lindsey Murphy
- Department of Pediatrics, City of Hope National Medical Center, Duarte, California, USA
| | - Ibrahim Aldoss
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA.
| |
Collapse
|
3
|
Binayi F, Saeidi B, Farahani F, Sadat Izadi M, Eskandari F, Azarkish F, Sahraei M, Ghasemi R, Khodagholi F, Zardooz H. Sustained feeding of a diet high in fat resulted in a decline in the liver's insulin-degrading enzyme levels in association with the induction of oxidative and endoplasmic reticulum stress in adult male rats: Evaluation of 4-phenylbutyric acid. Heliyon 2024; 10:e32804. [PMID: 38975085 PMCID: PMC11226834 DOI: 10.1016/j.heliyon.2024.e32804] [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: 01/17/2024] [Revised: 06/06/2024] [Accepted: 06/10/2024] [Indexed: 07/09/2024] Open
Abstract
The current study explored the impact of high fat diet (HFD) on hepatic oxidative and endoplasmic reticulum (ER) stress and its insulin degrading enzyme (IDE) content with the injection of 4-phenyl butyric acid (4-PBA) in adult male rats. Following the weaning period, male offspring were distributed among six distinct groups. The corresponding diet was used for 20 weeks, subsequently 4-PBA was administered for three consecutive days. Plasma glucose and insulin levels, HOMA-β (homeostasis model assessment of β-cell), hepatic ER and oxidative stress biomarkers and IDE protein content were assessed. Long-term ingestion of HFD (31 % cow butter) induced oxidative and ER stress in the liver tissue. Accordingly, a rise in the malondialdehyde (MDA) content and catalase enzyme activity and a decrease in the glutathione (GSH) content were detected within the liver of the HFD and HFD + DMSO groups. Consumption of this diet elevated the liver expression of binding immunoglobulin protein (BIP) and C/enhancer-binding protein homologous protein (CHOP) levels while reduced its IDE content. The HOMA-β decreased significantly. The injection of the 4-PBA moderated all the induced changes. Findings from this study indicated that prolonged HFD consumption led to a reduction in plasma insulin levels, likely attributed to pancreatic β cell malfunction, as evidenced by a decline in the HOMA-β index. Also, the HFD appears to have triggered oxidative and ER stress in the liver, along with a decrease in its IDE content.
Collapse
Affiliation(s)
- Fateme Binayi
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behnam Saeidi
- Protein Research Center, Shahid Beheshti University, Tehran, Iran
| | - Fatemeh Farahani
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mina Sadat Izadi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Farzaneh Eskandari
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Azarkish
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Sahraei
- School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rasoul Ghasemi
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Homeira Zardooz
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Ngema M, Xulu ND, Ngubane PS, Khathi A. Pregestational Prediabetes Induces Maternal Hypothalamic-Pituitary-Adrenal (HPA) Axis Dysregulation and Results in Adverse Foetal Outcomes. Int J Mol Sci 2024; 25:5431. [PMID: 38791468 PMCID: PMC11122116 DOI: 10.3390/ijms25105431] [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: 03/26/2024] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
Maternal type 2 diabetes mellitus (T2DM) has been shown to result in foetal programming of the hypothalamic-pituitary-adrenal (HPA) axis, leading to adverse foetal outcomes. T2DM is preceded by prediabetes and shares similar pathophysiological complications. However, no studies have investigated the effects of maternal prediabetes on foetal HPA axis function and postnatal offspring development. Hence, this study investigated the effects of pregestational prediabetes on maternal HPA axis function and postnatal offspring development. Pre-diabetic (PD) and non-pre-diabetic (NPD) female Sprague Dawley rats were mated with non-prediabetic males. After gestation, male pups born from the PD and NPD groups were collected. Markers of HPA axis function, adrenocorticotropin hormone (ACTH) and corticosterone, were measured in all dams and pups. Glucose tolerance, insulin and gene expressions of mineralocorticoid (MR) and glucocorticoid (GR) receptors were further measured in all pups at birth and their developmental milestones. The results demonstrated increased basal concentrations of ACTH and corticosterone in the dams from the PD group by comparison to NPD. Furthermore, the results show an increase basal ACTH and corticosterone concentrations, disturbed MR and GR gene expression, glucose intolerance and insulin resistance assessed via the Homeostasis Model Assessment (HOMA) indices in the pups born from the PD group compared to NPD group at all developmental milestones. These observations reveal that pregestational prediabetes is associated with maternal dysregulation of the HPA axis, impacting offspring HPA axis development along with impaired glucose handling.
Collapse
Affiliation(s)
| | | | | | - Andile Khathi
- School of Laboratory Medicine & Medical Sciences, University of KwaZulu-Natal, Westville, Private Bag X54001, Durban 4041, KwaZulu Natal, South Africa; (M.N.); (N.D.X.); (P.S.N.)
| |
Collapse
|
5
|
Karbaschi R, Zardooz H. Pancreatic GLUT2 protein expression and isolated islets insulin secretion decreased in high-fat fed rat dams. J Diabetes Metab Disord 2023; 22:1511-1518. [PMID: 37975089 PMCID: PMC10638334 DOI: 10.1007/s40200-023-01274-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 07/30/2023] [Indexed: 11/19/2023]
Abstract
Purpose Chronic consumption of high-fat foods during the reproductive period may endanger the dams' metabolic homeostasis and might adversely affect pregnancy outcome. In this regard the present study aimed to investigate the effect of long-term high-fat feeding on pancreatic glucose transporter-2 (GLUT2) protein expression and isolated islets glucose-stimulated insulin secretion in Wistar rat dams. Materials and methods Female rats were randomly divided into normal (N) and high-fat (HF; containing cow butter) diet groups and consumed their respective diets for 10 weeks (from prepregnancy to the end of lactation). After lactation, fasting plasma concentrations of glucose and insulin were measured to calculate HOMA-IR index, then intraperitoneal glucose tolerance test (IPGTT) was performed. Moreover, the pancreatic GLUT2 protein expression and insulin secretion from isolated islets at basal (5.6 mM) and stimulated (16.7 mM) glucose concentrations were assessed. Results In HF group compared to N group, the plasma insulin level increased, whereas the plasma glucose level did not change in fasting state. Accordingly, the HOMA-IR index increased in HF fed animals. Furthermore, the IPGTT revealed glucose intolerance based on the plasma glucose and insulin results. Also, the pancreatic GLUT2 expression and isolated islets insulin secretion, in response to high glucose concentration, were decreased. Conclusion The chronic consumption of high-fat foods during prepregnancy, pregnancy, and lactation periods can lead to glucose intolerance, insulin resistance, and inhibition of pancreatic GLUT2 expression, which impairs glucose homeostasis. Therefore, it is crucial to carefully monitor the diet composition of dams during this critical period. Supplementary Information The online version contains supplementary material available at 10.1007/s40200-023-01274-6.
Collapse
Affiliation(s)
- Roxana Karbaschi
- Faculty of Nursing and Midwifery, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Homeira Zardooz
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Pofi R, Caratti G, Ray DW, Tomlinson JW. Treating the Side Effects of Exogenous Glucocorticoids; Can We Separate the Good From the Bad? Endocr Rev 2023; 44:975-1011. [PMID: 37253115 PMCID: PMC10638606 DOI: 10.1210/endrev/bnad016] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/25/2023] [Accepted: 05/26/2023] [Indexed: 06/01/2023]
Abstract
It is estimated that 2% to 3% of the population are currently prescribed systemic or topical glucocorticoid treatment. The potent anti-inflammatory action of glucocorticoids to deliver therapeutic benefit is not in doubt. However, the side effects associated with their use, including central weight gain, hypertension, insulin resistance, type 2 diabetes (T2D), and osteoporosis, often collectively termed iatrogenic Cushing's syndrome, are associated with a significant health and economic burden. The precise cellular mechanisms underpinning the differential action of glucocorticoids to drive the desirable and undesirable effects are still not completely understood. Faced with the unmet clinical need to limit glucocorticoid-induced adverse effects alongside ensuring the preservation of anti-inflammatory actions, several strategies have been pursued. The coprescription of existing licensed drugs to treat incident adverse effects can be effective, but data examining the prevention of adverse effects are limited. Novel selective glucocorticoid receptor agonists and selective glucocorticoid receptor modulators have been designed that aim to specifically and selectively activate anti-inflammatory responses based upon their interaction with the glucocorticoid receptor. Several of these compounds are currently in clinical trials to evaluate their efficacy. More recently, strategies exploiting tissue-specific glucocorticoid metabolism through the isoforms of 11β-hydroxysteroid dehydrogenase has shown early potential, although data from clinical trials are limited. The aim of any treatment is to maximize benefit while minimizing risk, and within this review we define the adverse effect profile associated with glucocorticoid use and evaluate current and developing strategies that aim to limit side effects but preserve desirable therapeutic efficacy.
Collapse
Affiliation(s)
- Riccardo Pofi
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | - Giorgio Caratti
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | - David W Ray
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
- NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Oxford Kavli Centre for Nanoscience Discovery, University of Oxford, Oxford OX37LE, UK
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| |
Collapse
|
7
|
Mkhize BC, Mosili P, Ngubane PS, Sibiya NH, Khathi A. The Relationship between Renin-Angiotensin-Aldosterone System (RAAS) Activity, Osteoporosis and Estrogen Deficiency in Type 2 Diabetes. Int J Mol Sci 2023; 24:11963. [PMID: 37569338 PMCID: PMC10419188 DOI: 10.3390/ijms241511963] [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: 06/29/2023] [Revised: 07/15/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Type 2 diabetes (T2D) is associated with a plethora of comorbidities, including osteoporosis, which occurs due to an imbalance between bone resorption and formation. Numerous mechanisms have been explored to understand this association, including the renin-angiotensin-aldosterone system (RAAS). An upregulated RAAS has been positively correlated with T2D and estrogen deficiency in comorbidities such as osteoporosis in humans and experimental studies. Therefore, research has focused on these associations in order to find ways to improve glucose handling, osteoporosis and the downstream effects of estrogen deficiency. Upregulation of RAAS may alter the bone microenvironment by altering the bone marrow inflammatory status by shifting the osteoprotegerin (OPG)/nuclear factor kappa-Β ligand (RANKL) ratio. The angiotensin-converting-enzyme/angiotensin II/Angiotensin II type 1 receptor (ACE/Ang II/AT1R) has been evidenced to promote osteoclastogenesis and decrease osteoblast formation and differentiation. ACE/Ang II/AT1R inhibits the wingless-related integration site (Wnt)/β-catenin pathway, which is integral in bone formation. While a lot of literature exists on the effects of RAAS and osteoporosis on T2D, the work is yet to be consolidated. Therefore, this review looks at RAAS activity in relation to osteoporosis and T2D. This review also highlights the relationship between RAAS activity, osteoporosis and estrogen deficiency in T2D.
Collapse
Affiliation(s)
- Bongeka Cassandra Mkhize
- Human Physiology, Health Science, Westville Campus, University of KwaZulu-Natal, Westville 4041, South Africa; (B.C.M.); (P.M.); (P.S.N.)
| | - Palesa Mosili
- Human Physiology, Health Science, Westville Campus, University of KwaZulu-Natal, Westville 4041, South Africa; (B.C.M.); (P.M.); (P.S.N.)
| | - Phikelelani Sethu Ngubane
- Human Physiology, Health Science, Westville Campus, University of KwaZulu-Natal, Westville 4041, South Africa; (B.C.M.); (P.M.); (P.S.N.)
| | | | - Andile Khathi
- Human Physiology, Health Science, Westville Campus, University of KwaZulu-Natal, Westville 4041, South Africa; (B.C.M.); (P.M.); (P.S.N.)
| |
Collapse
|
8
|
Hall LG, Thyfault JP, Johnson JD. Exercise and inactivity as modifiers of β cell function and type 2 diabetes risk. J Appl Physiol (1985) 2023; 134:823-839. [PMID: 36759159 PMCID: PMC10042613 DOI: 10.1152/japplphysiol.00472.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/11/2023] Open
Abstract
Exercise and regular physical activity are beneficial for the prevention and management of metabolic diseases such as obesity and type 2 diabetes, whereas exercise cessation, defined as deconditioning from regular exercise or physical activity that has lasted for a period of months to years, can lead to metabolic derangements that drive disease. Adaptations to the insulin-secreting pancreatic β-cells are an important benefit of exercise, whereas less is known about how exercise cessation affects these cells. Our aim is to review the impact that exercise and exercise cessation have on β-cell function, with a focus on the evidence from studies examining glucose-stimulated insulin secretion (GSIS) using gold-standard techniques. Potential mechanisms by which the β-cell adapts to exercise, including exerkine and incretin signaling, autonomic nervous system signaling, and changes in insulin clearance, will also be explored. We will highlight areas for future research.
Collapse
Affiliation(s)
- Liam G Hall
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| | - John P Thyfault
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States
- KU Diabetes Institute, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - James D Johnson
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
9
|
Mao X, Zhou J, Kong L, Zhu L, Yang D, Zhang Z. A peptide encoded by lncRNA MIR7-3 host gene (MIR7-3HG) alleviates dexamethasone-induced dysfunction in pancreatic β-cells through the PI3K/AKT signaling pathway. Biochem Biophys Res Commun 2023; 647:62-71. [PMID: 36731335 DOI: 10.1016/j.bbrc.2023.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 11/26/2022] [Accepted: 01/02/2023] [Indexed: 01/05/2023]
Abstract
BACKGROUND Dysfunction of pancreatic β-cells induced by glucocorticoids contributes to diabetes mellitus development. Long noncoding RNAs (lncRNAs) have been recognized to contain short open reading frames (ORFs) that can be translated into functional small peptides. Here, we investigated whether the short peptide encoded by the lncRNA MIR7-3 host gene (MIR7-3HG) can affect dexamethasone (DEX)-induced β-cell dysfunction. METHODS Bioinformatics analysis was used for selection of MIR7-3HG and prediction of its protein encoding potential. The small peptide was identified by a western blot method. The cell-permeable TAT was fused into MIR7-3HG ORF to produce the cell-permeable fusion peptide (TAT-MIR7-3HG-ORF). The effects of TAT-MIR7-3HG-ORF on DEX-induced β-cell dysfunction were evaluated by examining cell viability, apoptosis, insulin secretion, and reactive oxygen species (ROS) generation. RESULTS DEX induced β-TC6 cell dysfunction by impairing cell viability, insulin secretion and promoting cell apoptosis and ROS generation. The MIR7-3HG ORF could encode a 125-amino-acid-long short peptide. TAT-MIR7-3HG-ORF effectively transduced into β-TC6 cells and attenuated DEX-induced dysfunction in β-TC6 cells. Moreover, transduced TAT-MIR7-3HG-ORF reversed DEX-mediated inhibition of the activation of the PI3K/AKT signaling pathway. The inhibitor of the PI3K/AKT pathway partially abolished the alleviative effect of transduced TAT-MIR7-3HG-ORF on DEX-induced β-TC6 cell dysfunction. CONCLUSION The lncRNA MIR7-3HG encodes a short peptide, which can protect pancreatic β-cells from DEX-induced dysfunction by activating the PI3K/AKT pathway. Our study broadens the diversity and breadth of lncRNAs in human disorders.
Collapse
Affiliation(s)
- Xiaoming Mao
- Department of Geriatrics, Henan Key Laboratory for Geriatrics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, 450003, China
| | - Jinliang Zhou
- Department of Hip Surgery, Luoyang Orthopedic Hospital of Henan Province, Luoyang, Henan, 471000, China
| | - Limin Kong
- Xinxiang Medical University, Xinxiang, Henan, 453003, China; The Sixth People's Hospital of Zhengzhou, Zhengzhou, Henan, 450000, China
| | - Li Zhu
- Department of Thoracic Surgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China
| | - Desheng Yang
- Department of Geriatrics, Henan Key Laboratory for Geriatrics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, 450003, China.
| | - Zhiyu Zhang
- Department of Geriatrics, Henan Key Laboratory for Geriatrics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, 450003, China.
| |
Collapse
|
10
|
Ingrosso DMF, Primavera M, Samvelyan S, Tagi VM, Chiarelli F. Stress and Diabetes Mellitus: Pathogenetic Mechanisms and Clinical Outcome. Horm Res Paediatr 2023; 96:34-43. [PMID: 35124671 DOI: 10.1159/000522431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 02/01/2022] [Indexed: 11/19/2022] Open
Abstract
Evidence suggests that psychological and physical stress are relevant triggering factors for the onset of type 1 diabetes (T1D) and type 2 diabetes (T2D). The underlying mechanisms involve a complex neuroendocrine structure, involving the central nervous system and the periphery. Psychological stress leads to an increase of serum glucocorticoid concentrations and catecholamines release increasing the insulin need and the insulin resistance. According to the β-cell stress hypothesis, also causes of increased insulin demand, such as rapid growth, overweight, puberty, low physical activity, trauma, infections, and glucose overload, are potentially relevant factors in development of T1D. It has also been demonstrated that chronic stress and obesity form a vicious circle which leads to a definitive metabolic failure, increasing the risk of developing T2D. In this review, we will provide the most recent data concerning the role of stress in the outcomes of T1D and T2D, with a focus on the role of physical and psychological stress on the onset of T1D.
Collapse
Affiliation(s)
| | | | - Sona Samvelyan
- Paediatric Outpatient Department No. 122, Moscow, Russian Federation
| | | | | |
Collapse
|
11
|
Endoplasmic reticulum stress inhibition ameliorated WFS1 expression alterations and reduced pancreatic islets' insulin secretion induced by high-fat diet in rats. Sci Rep 2023; 13:1860. [PMID: 36725880 PMCID: PMC9892558 DOI: 10.1038/s41598-023-28329-1] [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: 08/14/2022] [Accepted: 01/17/2023] [Indexed: 02/03/2023] Open
Abstract
Endoplasmic reticulum (ER) stress is involved in the development of glucose homeostasis impairment. When ER stress occurs, the unfolded protein response (UPR) is activated to cope with it. One of the UPR components is WFS1 (Wolfram syndrome 1), which plays important roles in ER homeostasis and pancreatic islets glucose-stimulated insulin secretion (GSIS). Accordingly and considering that feeding high-fat food has a major contribution in metabolic disorders, this study aimed to investigate the possible involvement of pancreatic ER stress in glucose metabolism impairment induced by feeding high-fat diet (HFD) in male rats. After weaning, the rats were divided into six groups, and fed on normal diet and HFD for 20 weeks, then 4-phenyl butyric acid (4-PBA, an ER stress inhibitor) was administered. Subsequently, in all groups, after performing glucose tolerance test, the animals were dissected and their pancreases were removed to extract ER, islets isolation and assessment of GSIS. Moreover, the pancreatic ER stress [binding of immunoglobulin protein (BIP) and enhancer-binding protein homologous protein (CHOP)] and oxidative stress [malondialdehyde (MDA), glutathione (GSH) and catalase] biomarkers as well as WFS1 expression level were evaluated. HFD decreased pancreatic WFS1 protein and GSH levels, and enhanced pancreatic catalase activity, MDA content, BIP and CHOP protein and mRNA levels as well as Wfs1 mRNA amount. Accordingly, it increased BIP, CHOP and WFS1 protein levels in the extracted ER of pancreas. In addition, the HFD caused glucose intolerance, and decreased the islets' GSIS and insulin content. However, 4-PBA administration restored the alterations. It seems that, HFD consumption through inducing pancreatic ER stress, altered WFS1 expression levels, reduced the islets' GSIS and insulin content and finally impaired glucose homeostasis.
Collapse
|
12
|
Oh HY, Yoo JE. The Association between Duration of Noise Exposure in the Workplace and Glucose Metabolism Status: Evidence from the Korea National Health and Nutrition Examination Survey. Korean J Fam Med 2022; 43:396-402. [PMID: 36444125 PMCID: PMC9708854 DOI: 10.4082/kjfm.21.0180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/28/2021] [Accepted: 10/11/2021] [Indexed: 08/22/2023] Open
Abstract
BACKGROUND This study aimed to evaluate the association between the duration of workplace noise exposure and glucose metabolism status in a nationally representative Korean sample. METHODS This cross-sectional study included 3,534 participants aged ≥40 years without tinnitus or hearing loss from the Korea National Health and Nutrition Examination Survey (2018). The primary exposure was noise in the workplace and its duration. We divided the noise exposure group into four groups according to the duration of noise exposure (<3 years, 3-10 years, 10-20 years, and ≥20 years). The primary outcomes were fasting blood sugar (FBS), hemoglobin A1c (HbA1c), and pre-diabetes and diabetes diagnosed using FBS. Logistic and linear regression analyses were used to test the association between noise exposure and glycemic status. RESULTS After adjustment, HbA1c levels were significantly higher in the noise exposure than in the non-noise exposure group. HbA1c levels were significantly higher in those exposed to occupational noise for more than 20 years than in others. In the subgroup analysis among those who had been exposed to noise for >20 years, the non-aerobic physical activity group had significantly higher HbA1c levels than the physical activity group. Furthermore, among those who had been exposed to noise for >20 years, the without hearing protection group had significantly higher HbA1c levels than those using hearing protection. CONCLUSION The association between noise exposure and the prevalence of diabetes is unclear. However, our study clearly suggests that there is a relationship between elevated HbA1c levels and workplace noise exposure and that a long period of workplace noise exposure, no physical activity, and not wearing a hearing protection device could increase the risk of diabetes.
Collapse
Affiliation(s)
- Hye Yeong Oh
- Department of Family Medicine, Seoul National University Hospital, Seoul, Korea
| | - Jung Eun Yoo
- Department of Family Medicine, Seoul National University Hospital Healthcare System Gangnam Center, Seoul, Korea
| |
Collapse
|
13
|
Tajbakhsh A, Gheibihayat SM, Karami N, Savardashtaki A, Butler AE, Rizzo M, Sahebkar A. The regulation of efferocytosis signaling pathways and adipose tissue homeostasis in physiological conditions and obesity: Current understanding and treatment options. Obes Rev 2022; 23:e13487. [PMID: 35765849 DOI: 10.1111/obr.13487] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 12/14/2022]
Abstract
Obesity is associated with changes in the resolution of acute inflammation that contribute to the clinical complications. The exact mechanisms underlying unresolved inflammation in obesity are not fully understood. Adipocyte death leads to pro-inflammatory adipose tissue macrophages, stimulating additional adipocyte apoptosis. Thus, a complex and tightly regulated process to inhibit inflammation and maintain homeostasis after adipocyte apoptosis is needed to maintain health. In normal condition, a specialized phagocytic process (efferocytosis) performs this function, clearing necrotic and apoptotic cells (ACs) and controlling inflammation. For efficient and continued efferocytosis, phagocytes must internalize multiple ACs in physiological conditions and handle the excess metabolic burden in adipose tissue. In obesity, this control is lost and can be an important hallmark of the disease. In this regard, the deficiency of efferocytosis leads to delayed resolution of acute inflammation and can result in ongoing inflammation, immune system dysfunction, and insulin resistance in obesity. Hence, efficient clearance of ACs by M2 macrophages could limit long-term inflammation and ensue clinical complications, such as cardiovascular disease and diabetes. This review elaborates upon the molecular mechanisms to identify efferocytosis regulators in obesity, and the mechanisms that can improve efferocytosis and reduce obesity-related complications, such as the use of pharmacological agents and regular exercise.
Collapse
Affiliation(s)
- Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Neda Karami
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.,Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland Bahrain, Adliya, 15503, Bahrain
| | - Manfredi Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, School of Medicine, University of Palermo, Palermo, Italy
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Medicine, The University of Western Australia, Perth, Western Australia, Australia.,Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
14
|
Eskandari F, Salimi M, Binayi F, Abdollahifar MA, Eftekhary M, Hedayati M, Ghanbarian H, Zardooz H. Investigating the Effects of Maternal Separation on Hypothalamic-Pituitary-Adrenal Axis and Glucose Homeostasis under Chronic Social Defeat Stress in Young Adult Male Rat Offspring. Neuroendocrinology 2022; 113:361-380. [PMID: 36088912 DOI: 10.1159/000526989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 09/05/2022] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Given the suggested metabolic regulatory effects of stress-responsive genes and based on the impacts of early-life stress on HPA axis development, this study aimed to characterize the maternal separation (MS) impact on the communication between glucose metabolism and HPA axis dysregulations under chronic social defeat stress (CSDS). METHODS During the first 2 weeks of life, male Wistar rats were either exposed to MS or left undisturbed with their mothers (Std). Starting on postnatal day 50, the animals of each group were either left undisturbed in the standard group housing (Con) or underwent CSDS for 3 weeks. There were four groups (n = 10/group): Std-Con, MS-Con, Std-CSDS, and MS-CSDS. RESULTS Early and/or adult life adversity reduced β-cell number, muscular FK506-binding protein 51 (FKBP51) content, and BMI in adulthood. The reduction of β-cell number and BMI in the MS-CSDS rats were more profound than MS-Con group. CSDS either alone or in combination with MS reduced locomotor activity and increased and decreased corticotropin-releasing factor type 1 receptor (CRFR1) content, respectively, in hypothalamus and pancreas. Although, under CSDS, MS intensified HPA axis overactivity and reduced isolated islets' insulin secretion, it could promote resilience to depression symptoms. No differences were observed in hypothalamic Fkbp5 gene DNA methylation and glucose tolerance among groups. CONCLUSION MS exacerbated HPA axis overactivity and the endocrine pancreas dysfunctions under CSDS. The intensified corticosterone secretion and the diminished content of pancreatic CRFR1 protein could be involved in the reduced β-cell number and islets' insulin secretion under CSDS. The decreased muscular FKBP51 content might be a homeostatic response to slow down insulin resistance development under chronic stress.
Collapse
Affiliation(s)
- Farzaneh Eskandari
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mina Salimi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fateme Binayi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad-Amin Abdollahifar
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohamad Eftekhary
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Ghanbarian
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Homeira Zardooz
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Handattu K, Sharma LK, Vijayasekharan K, Bhat K V, Aroor S, Sudhanshu S. Drug Induced Diabetes Mellitus in Pediatric Acute Lymphoblastic Leukemia: Approach to Diagnosis and Management. J Pediatr Hematol Oncol 2022; 44:273-279. [PMID: 35700365 DOI: 10.1097/mph.0000000000002494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/08/2022] [Indexed: 11/26/2022]
Abstract
Corticosteroids and l -asparaginase used in the treatment of pediatric acute lymphoblastic leukemia (ALL) can cause drug-induced diabetes mellitus (DIDM). DIDM can lead to dyselectrolytemia, a higher risk of infections including cellulitis, bacteremia, fungemia, and a higher incidence of febrile neutropenia and may have an impact on the outcome of ALL. Literature on the management of DIDM among children with ALL is sparse and the diagnostic criteria for pediatric diabetes should be carefully applied considering the acute and transient nature of DIDM during ALL therapy. Insulin remains the standard of care for DIDM management and the choice of Insulin regimen (stand-alone Neutral Protamine Hagedorn or basal bolus) should be based on the type and dose of steroids used for ALL and the pattern of hyperglycemia. A modest glycemic control (postmeal 140 to 180 mg/dL, premeal <140 mg/dL) to prevent complications of hyperglycemia, as well as hypoglycemia, would be the general approach. This review is intended to suggest evidence-based practical guidance in the diagnosis and management of DIDM during pediatric ALL therapy.
Collapse
Affiliation(s)
| | - Lokesh K Sharma
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh
| | | | - Vasudeva Bhat K
- Division of Pediatric Hematology and Oncology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka
| | | | - Siddhnath Sudhanshu
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh
| |
Collapse
|
16
|
Li X, He Q, Zhao N, Chen X, Li T, Cheng B. High intensity interval training ameliorates cognitive impairment in T2DM mice possibly by improving PI3K/Akt/mTOR Signaling-regulated autophagy in the hippocampus. Brain Res 2021; 1773:147703. [PMID: 34743961 DOI: 10.1016/j.brainres.2021.147703] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 10/11/2021] [Accepted: 10/22/2021] [Indexed: 10/19/2022]
Abstract
Exercise can improve cognitive impairment in type 2 diabetes mellitus (T2DM). However, the underlying mechanisms are not clear, and the optimal exercise modes for cognitive benefits are controversial. The aim of this study was to investigate the effects of high-intensity interval training (HIIT) and moderate-intensity interval training (MICT) on cognitive function and the PI3K/Akt/mTOR pathway as well as autophagy in T2DM mice. The results showed that 8 weeks of HIIT and MICT intervention could improve the spatial learning and memory ability of T2DM mice, as determined by the Morris water maze (MWM) test. Both HIIT and MICT similarly improved autophagy, as evidenced by increased Beclin1 and LC3 II/I ratios and decreased p62. Meanwhile, HIIT and MICT inhibited excessive activation of the PI3K/Akt/mTOR pathway in the hippocampus. HIIT induced a larger reduction in mTOR activity than MICT. This study suggests that both HIIT and MICT can alleviate cognitive decline induced by T2DM, improve autophagy in the hippocampus, and downregulate the excessive activation of the PI3K/Akt/mTOR signaling pathway, with similar effects.
Collapse
Affiliation(s)
- Xuejiao Li
- School of Physical Education of Shandong University, Jinan, China
| | - Qiang He
- School of Physical Education of Shandong University, Jinan, China
| | - Na Zhao
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
| | - Xianghe Chen
- College of Physical Education, Yangzhou University, Yangzhou, China
| | - Tuojian Li
- School of Physical Education of Shandong University, Jinan, China
| | - Bin Cheng
- School of Physical Education of Shandong University, Jinan, China.
| |
Collapse
|
17
|
Sadeghimahalli F, Karbaschi R, Salimi M, Khodagholi F, Zardooz H. Pancreatic HB9 protein level is affected by early life stress in young adult rats: possible involvement of TNF-α and corticosterone. Arch Physiol Biochem 2021; 127:406-413. [PMID: 31368362 DOI: 10.1080/13813455.2019.1645699] [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: 02/27/2019] [Revised: 06/25/2019] [Accepted: 07/15/2019] [Indexed: 01/06/2023]
Abstract
This study examined foot shock stress effects, during weaning, on pancreatic HB9 protein expression in young adult male rats in the presence or absence of adulthood stress. The pups were divided into Control, Early life stress, Young adult stress, and Early + young adult stress groups. Plasma corticosterone, insulin, glucose, and TNF-α concentrations, and pancreatic HB9 protein expression were assessed. At 2 weeks of age, stress increased plasma corticosterone level. During young adulthood, plasma TNF-α and glucose concentrations increased, whereas plasma insulin and pancreatic HB9 protein levels decreased in Early life stress group. Whereas, Early + young adulthood stress group showed no change in the study parameters, except for plasma corticosterone and insulin concentrations. Overall, early life stress reduced pancreatic HB9 protein expression possibly by elevating plasma corticosterone and TNF-α levels in early life and adulthood, respectively. However, combined with adulthood stress, HB9 protein expression increased to the level of Control.
Collapse
Affiliation(s)
- Forouzan Sadeghimahalli
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Education Development Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Roxana Karbaschi
- Faculty of Nursing and Midwifery, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mina Salimi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- NeuroBiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Homeira Zardooz
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Binayi F, Zardooz H, Ghasemi R, Hedayati M, Askari S, Pouriran R, Sahraei M. The chemical chaperon 4-phenyl butyric acid restored high-fat diet- induced hippocampal insulin content and insulin receptor level reduction along with spatial learning and memory deficits in male rats. Physiol Behav 2021; 231:113312. [PMID: 33412188 DOI: 10.1016/j.physbeh.2021.113312] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/01/2021] [Accepted: 01/02/2021] [Indexed: 12/18/2022]
Abstract
This study assessed the effect of a chronic high-fat diet (HFD) on plasma and hippocampal insulin and corticosterone levels, the hippocampus insulin receptor amount, and spatial learning and memory with or without receiving 4-phenyl butyric acid (4-PBA) in male rats. Rats were divided into high-fat and normal diet groups, then each group was subdivided into dimethyl sulfoxide (DMSO) and 4-PBA groups. After weaning, the rats were fed with HFD for 20 weeks. Then, 4-PBA or DMSO were injected for 3 days. Subsequently, oral glucose tolerance test was done. On the following day, spatial memory tests were performed. Then the hippocampus Bip, Chop, insulin, corticosterone, and insulin receptor levels were determined. HFD increased plasma glucose, leptin and corticosterone concentrations, hippocampus Bip, Chop and corticosterone levels, food intake, abdominal fat weight and body weight along with impaired glucose tolerance. It decreased plasma insulin, and insulin content, and its receptor amount in hippocampus. HFD lengthened escape latency and shortened the duration spent in target zone. 4-PBA administration improved the HFD- induced adverse changes. Chronic HFD possibly through the induction of endoplasmic reticulum (ER) stress and subsequent changes in the levels of hippocampal corticosterone, insulin and insulin receptor along with possible leptin resistance caused spatial learning and memory deficits.
Collapse
Affiliation(s)
- Fateme Binayi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Homeira Zardooz
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Rasoul Ghasemi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Askari
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ramin Pouriran
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Sahraei
- School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Wei D, Liu X, Huo W, Yu S, Li L, Wang C, Mao Z. Serum cortisone and glucocorticoid receptor gene (NR3C1) polymorphism in human dysglycemia. Hormones (Athens) 2020; 19:385-393. [PMID: 32304041 DOI: 10.1007/s42000-020-00196-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 04/06/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE We aimed to explore the associations of serum cortisone and glucocorticoid receptor (GR) polymorphism with glucose metabolism and type 2 diabetes mellitus (T2DM) among Chinese adults. METHODS A total of 2315 participants were included in the present study. Serum cortisone was measured by liquid chromatography-tandem mass spectrometry. Multivariable logistic regression and linear regression were employed to assess the associations between serum cortisone and different glucose metabolism status. RESULTS Serum cortisone was positively associated with impaired fasting glucose (IFG) and T2DM ((Quartile 4 vs Quartile 1, odds ratio (OR) = 1.36, 95% confidence interval (CI) 1.01, 1.84, and OR = 2.08, 95% CI 1.50, 2.89, respectively)). A 100% increase in cortisone was associated with a 0.015 (95% CI 0.005, 0.025) mg/dl higher fasting plasma glucose (FPG), a 0.007 (95% CI 0.001, 0.013) higher glycosylated hemoglobin (HbA1c), a 0.4% (95% CI - 0.007, 0.000) lower HOMA2-IR, and a 58.1% (95% CI - 0.788, - 0.373) lower HOMA2-β. After stratification by genotype, the association between serum cortisone and T2DM was not significant in TT genotype carriers. In addition, at the higher concentrations of cortisone, TT genotype carriers had a lower FPG, HbA1c, and HOMA2-IR and a higher HOMA2-β than GG and GT carriers. CONCLUSIONS Elevated serum cortisone was associated with an increased risk of IFG and T2DM, and the associations may be modified by rs9324924 polymorphism.
Collapse
Affiliation(s)
- Dandan Wei
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, People's Republic of China
| | - Xue Liu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, People's Republic of China
| | - Wenqian Huo
- Department of Occupational and Environmental Health Sciences, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Songcheng Yu
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Linlin Li
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, People's Republic of China
| | - Chongjian Wang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, People's Republic of China
| | - Zhenxing Mao
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, People's Republic of China.
| |
Collapse
|
20
|
Zhou J, Zhang Z, Zhou H, Qian G. Diabetic Cognitive Dysfunction: From Bench to Clinic. Curr Med Chem 2020; 27:3151-3167. [PMID: 30727866 DOI: 10.2174/1871530319666190206225635] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 12/30/2018] [Accepted: 01/30/2019] [Indexed: 02/07/2023]
Abstract
Type 2 diabetes increases the risk of developing cognitive dysfunction in the elderly in the form of short-term memory and executive function impairment. Genetic and diet-induced models of type 2 diabetes further support this link, displaying deficits in working memory, learning, and memory performance. The risk factors for diabetic cognitive dysfunction include vascular disease, hypoglycaemia, hyperlipidaemia, adiposity, insulin resistance, lifestyle factors, and genetic factors. Using neuronal imaging technologies, diabetic patients with cognitive dysfunction show atrophy of the whole brain, particularly the grey matter, hippocampus and amygdala; increased volume of the ventricular and white matter; brain infarcts; impaired network integrity; abnormal microstructure; and reduced cerebral blood flow and amplitude of low-frequency fluctuations. The pathogenesis of type 2 diabetes with cognitive dysfunction involves hyperglycaemia, macrovascular and microvascular diseases, insulin resistance, inflammation, apoptosis, and disorders of neurotransmitters. Large clinical trials may offer further proof of biomarkers and risk factors for diabetic cognitive dysfunction. Advanced neuronal imaging technologies and novel disease animal models will assist in elucidating the precise pathogenesis and to provide better therapeutic interventions and treatment.
Collapse
Affiliation(s)
- Jiyin Zhou
- National Drug Clinical Trial Institution, the Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| | - Zuo Zhang
- National Drug Clinical Trial Institution, the Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| | - Hongli Zhou
- National Drug Clinical Trial Institution, the Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| | - Guisheng Qian
- Institute of Respiratory Diseases, the Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| |
Collapse
|
21
|
Kaki A, Nikbakht M, Habibi A, Moghadam H. Effect of aerobic exercise on innate immune responses and inflammatory mediators in the spinal cord of diabetic rats. COMPARATIVE EXERCISE PHYSIOLOGY 2020. [DOI: 10.3920/cep190050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Neuronal inflammation is one of the pathophysiological causes of diabetes neuropathic pain. The purpose of this research was to determine the effect of aerobic exercise on innate immune responses and inflammatory mediators in the spinal dorsal horn in rats with diabetic neuropathic pain. 40 eight-week-old male Wistar rats (weight range 220±10.2 g) were randomly divided into four groups of (1) sedentary diabetic neuropathy (SDN), (2) training diabetic neuropathy (TDN), (3) training control (TC), and (4) sedentary control (SC). Diabetes was induced by injection of streptozocin (50 mg/kg). Following confirmation of behavioural tests for diabetes neuropathy, the training groups performed 6 weeks of moderate-intensity aerobic exercise on the treadmill. The expression of Toll like receptor (TLR)4, TLR2, tumour necrosis factor (TNF)-α, interleukin (IL)-1β and IL-10 genes in L4-L6 spinal cord sensory neurons was measured by Real Time PCR. Two-way ANOVA and Bonferroni’s post hoc tests were used for statistical analysis. After performing aerobic exercise protocol, the TDN compared to the SDN showed a significant decrease in the mean score of pain in the formalin test and a significant increase in the latency in Tail-Flick test was observed. The expression of TLR4, TLR2, TNF-α and IL-1β genes was significantly higher in the SDN than in the SC group (P<0.05). The expression of the above genes in the TDN was significantly lower than the SDN group (P<0.05). Also, the expression level of IL-10 gene was significantly higher in the TDN than the SDN group (P<0.05). Aerobic exercise improved sensitivity of nociceptors to pain-inducing agents in diabetic neuropathy due to inhibition of inflammatory receptors and increased levels of anti-inflammatory agents in the nervous system. Thus, aerobic exercise should be used as a non-pharmacological intervention for diabetic patients to reduce neuropathic pain.
Collapse
Affiliation(s)
- A. Kaki
- Department of Sport Physiology, Faculty of Sports Science, Shahid Chamran University of Ahvaz, 6135783151 Ahvaz, Iran
| | - M. Nikbakht
- Department of Sport Physiology, Faculty of Sports Science, Shahid Chamran University of Ahvaz, 6135783151 Ahvaz, Iran
| | - A.H. Habibi
- Department of Sport Physiology, Faculty of Sports Science, Shahid Chamran University of Ahvaz, 6135783151 Ahvaz, Iran
| | - H.F. Moghadam
- Department of Medical Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
22
|
Shah M, Adel MM, Tahsin B, Guerra Y, Fogelfeld L. Effect of short-term prednisone on beta-cell function in subjects with type 2 diabetes mellitus and healthy subjects. PLoS One 2020; 15:e0231190. [PMID: 32369480 PMCID: PMC7199958 DOI: 10.1371/journal.pone.0231190] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 03/10/2020] [Indexed: 12/05/2022] Open
Abstract
Objective For those with type 2 diabetes mellitus (T2DM), impact of short-term high-dose glucocorticoid exposure on beta-cell function is unknown. This study aims to compare the impact on beta-cell function and insulin resistance of prednisone 40 mg between adults with newly diagnosed T2DM and healthy adults. Methods Five adults with T2DM and five healthy adults, all between 18–50 years, were enrolled. T2DM diagnosis was less than one year prior, HbA1c<75 mmol/mol (9.0%), with metformin treatment only. Pre- and post-therapy testing included 75-g oral glucose tolerance, plasma glucose, C-peptide, and insulin. Intervention therapy was prednisone 40mg daily for 3 days. Results Upon therapy completion, HOMA-IR did not increase or differ between groups. Percentile difference for HOMA-%B and insulinogenic index in those with T2DM was significantly lower statistically (50.4% and 69.2% respectively) compared to healthy subjects (19% and 32.2%). Conclusions Contrary to the assumption that insulin resistance is the main driver of glucocorticoid-induced hyperglycemia, results indicate that decreased beta-cell insulin secretion is the more likely cause in those with T2DM. This is evidenced by significant drops in C-peptide AUC and HOMA-%B and increased glucose AUC in T2DM group only. These results may be caused by increased beta-cell fragility along with reduced recovery ability after glucocorticoid exposure. ClinicalTrials.gov NCT03661684.
Collapse
Affiliation(s)
- Monica Shah
- John H. Stroger Jr. Hospital of Cook County, Chicago, Illinois, United States of America
| | - May M. Adel
- John H. Stroger Jr. Hospital of Cook County, Chicago, Illinois, United States of America
| | - Bettina Tahsin
- John H. Stroger Jr. Hospital of Cook County, Chicago, Illinois, United States of America
| | - Yannis Guerra
- John H. Stroger Jr. Hospital of Cook County, Chicago, Illinois, United States of America
- * E-mail:
| | - Leon Fogelfeld
- John H. Stroger Jr. Hospital of Cook County, Chicago, Illinois, United States of America
| |
Collapse
|
23
|
Gregoriou K, Craigie I, Gibson B, Mason A, Shaikh MG. Risk factors and management of corticosteroid-induced hyperglycaemia in paediatric acute lymphoblastic leukaemia. Pediatr Blood Cancer 2020; 67:e28085. [PMID: 31736211 DOI: 10.1002/pbc.28085] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/23/2019] [Accepted: 10/30/2019] [Indexed: 02/02/2023]
Abstract
Corticosteroids are incorporated into protocols for the treatment of acute lymphoblastic leukaemia, and hyperglycaemia is a recognised side effect. Corticosteroids exert their hyperglycaemic effect with a multifactorial mechanism. Complications of hyperglycaemia include an increased risk of infection - bacterial, viral and fungal. Approximately half of the children who develop corticosteroid-associated hyperglycaemia are predicted to require insulin treatment, with age and obesity having found to be predictive factors. Fasting and random glucose values can be used to define hyperglycaemia. This review focuses on the published evidence for significant predictive factors for the development of corticosteroid-induced hyperglycaemia and provides guidance on management.
Collapse
Affiliation(s)
- Kyriacos Gregoriou
- Departments of Endocrinology, Royal Hospital for Children, Glasgow, UK.,Department of Diabetes, Royal Hospital for Children, Glasgow, UK
| | - Ian Craigie
- Department of Diabetes, Royal Hospital for Children, Glasgow, UK
| | - Brenda Gibson
- Department of Haematology, Royal Hospital for Children, Glasgow, UK
| | - Avril Mason
- Departments of Endocrinology, Royal Hospital for Children, Glasgow, UK.,Department of Diabetes, Royal Hospital for Children, Glasgow, UK
| | - Mohamad Guftar Shaikh
- Departments of Endocrinology, Royal Hospital for Children, Glasgow, UK.,Department of Diabetes, Royal Hospital for Children, Glasgow, UK
| |
Collapse
|
24
|
Sharma VK, Singh TG. Chronic Stress and Diabetes Mellitus: Interwoven Pathologies. Curr Diabetes Rev 2020; 16:546-556. [PMID: 31713487 DOI: 10.2174/1573399815666191111152248] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 09/25/2019] [Accepted: 10/27/2019] [Indexed: 12/15/2022]
Abstract
Stress threatens the homeostasis and mobilizes a plethora of adaptive physiological and behavioral changes via the hypothalamic-pituitary-adrenal (HPA) axis and the sympathetic nervous system. The HPA axis influences the pituitary gland, hypothalamus and adrenal gland via a complex set of positive and negative feedback system. The feedback system operates in a well regulated neuroendocrine manner to reestablish the threatened body equilibrium. The HPA axis secreted major product is a glucocorticoid (cortisol) which is kept within a physiologically optimal range and serves to accomplish the various physiological functions crucial for survival. In chronically stressed individuals dishabituation of HPA axis is followed by increased release of glucocorticoids and catecholamines. Higher secretion of glucocorticoids influences glucose metabolism by promoting gluconeogenesis in the liver, suppressing glucose uptake (adipocytes and skeletal muscles), promoting lipolysis in adipocytes, suppressing insulin secretion, inflicting insulin resistance and inflammation. These biological changes alter neuroendocrine mechanisms and lead to maladaptive congregation of events that form the underlying cause of development of Type 2 diabetes (T2D). The currently reviewed evidences advocate that targeting stress mediated hypersecretion of glucocorticoids may be a viable approach for the treatment of T2D and to reinstate glucose homeostasis.
Collapse
Affiliation(s)
- Vivek Kumar Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
- Department of Pharmacology, Government College of Pharmacy, Rohru, Distt. Shimla-171207, Himachal Pradesh, India
| | | |
Collapse
|
25
|
Akbib S, Stichelmans J, Stangé G, Ling Z, Assefa Z, Hellemans KH. Glucocorticoids and checkpoint tyrosine kinase inhibitors stimulate rat pancreatic beta cell proliferation differentially. PLoS One 2019; 14:e0212210. [PMID: 30779812 PMCID: PMC6380609 DOI: 10.1371/journal.pone.0212210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/29/2019] [Indexed: 12/27/2022] Open
Abstract
Cell therapy for diabetes could benefit from the identification of small-molecule compounds that increase the number of functional pancreatic beta cells. Using a newly developed screening assay, we previously identified glucocorticoids as potent stimulators of human and rat beta cell proliferation. We now compare the stimulatory action of these steroid hormones to a selection of checkpoint tyrosine kinase inhibitors that were also found to activate the cell cycle-in beta cells and analyzed their respective effects on DNA-synthesis, beta cell numbers and expression of cell cycle regulators. Our data using glucocorticoids in combination with a receptor antagonist, mifepristone, show that 48h exposure is sufficient to allow beta cells to pass the cell cycle restriction point and to become committed to cell division regardless of sustained glucocorticoid-signaling. To reach the end-point of mitosis another 40h is required. Within 14 days glucocorticoids stimulate up to 75% of the cells to undergo mitosis, which indicates that these steroid hormones act as proliferation competence-inducing factors. In contrast, by correlating thymidine-analogue incorporation to changes in absolute cell numbers, we show that the checkpoint kinase inhibitors, as compared to glucocorticoids, stimulate DNA-synthesis only during a short time-window in a minority of cells, insufficient to give a measurable increase of beta cell numbers. Glucocorticoids, but not the kinase inhibitors, were also found to induce changes in the expression of checkpoint regulators. Our data, using checkpoint kinase-specific inhibitors further point to a role for Chk1 and Cdk1 in G1/S transition and progression of beta cells through the cell cycle upon stimulation with glucocorticoids.
Collapse
Affiliation(s)
- Sarah Akbib
- Unit Diabetes Pathology and Therapy, Diabetes Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jordy Stichelmans
- Unit Diabetes Pathology and Therapy, Diabetes Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
| | - Geert Stangé
- Unit Diabetes Pathology and Therapy, Diabetes Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
| | - Zhidong Ling
- Unit Diabetes Pathology and Therapy, Diabetes Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
- Beta Cell Bank, University Hospital Brussels, Brussels, Belgium
| | - Zerihun Assefa
- Unit Diabetes Pathology and Therapy, Diabetes Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine H. Hellemans
- Unit Diabetes Pathology and Therapy, Diabetes Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
- Center for Beta Cell Therapy in Diabetes, Brussels, Belgium
| |
Collapse
|
26
|
Ježková J, Ďurovcová V, Wenchich L, Hansíková H, Zeman J, Hána V, Marek J, Lacinová Z, Haluzík M, Kršek M. The relationship of mitochondrial dysfunction and the development of insulin resistance in Cushing's syndrome. Diabetes Metab Syndr Obes 2019; 12:1459-1471. [PMID: 31695455 PMCID: PMC6707348 DOI: 10.2147/dmso.s209095] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 06/13/2019] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Cushing's syndrome is characterized by metabolic disturbances including insulin resistance. Mitochondrial dysfunction is one pathogenic factor in the development of insulin resistance in patients with obesity. We explored whether mitochondrial dysfunction correlates with insulin resistance and other metabolic complications. PATIENTS AND METHODS We investigated the changes of mRNA expression of genes encoding selected subunits of oxidative phosphorylation system (OXPHOS), pyruvate dehydrogenase (PDH) and citrate synthase (CS) in subcutaneous adipose tissue (SCAT) and peripheral monocytes (PM) and mitochondrial enzyme activity in platelets of 24 patients with active Cushing's syndrome and in 9 of them after successful treatment and 22 healthy control subjects. RESULTS Patients with active Cushing's syndrome had significantly increased body mass index (BMI), homeostasis model assessment of insulin resistance (HOMA-IR) and serum lipids relative to the control group. The expression of all investigated genes for selected mitochondrial proteins was decreased in SCAT in patients with active Cushing's syndrome and remained decreased after successful treatment. The expression of most tested genes in SCAT correlated inversely with BMI and HOMA-IR. The expression of genes encoding selected OXPHOS subunits and CS was increased in PM in patients with active Cushing's syndrome with a tendency to decrease toward normal levels after cure. Patients with active Cushing's syndrome showed increased enzyme activity of complex I (NQR) in platelets. CONCLUSION Mitochondrial function in SCAT in patients with Cushing's syndrome is impaired and only slightly affected by its treatment which may reflect ongoing metabolic disturbances even after successful treatment of Cushing's syndrome.
Collapse
Affiliation(s)
- Jana Ježková
- Third Department of Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
- Correspondence: Jana JežkováThird Department of Medicine, First Faculty of Medicine, Charles University and General University Hospital, U Nemocnice 1128 02 Praha 2, Prague, Czech RepublicTel +420 60 641 2613Fax +420 22 491 9780Email
| | - Viktória Ďurovcová
- Third Department of Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Laszlo Wenchich
- Institute of Rheumatology, Prague, Czech Republic
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Hana Hansíková
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Jiří Zeman
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Václav Hána
- Third Department of Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Josef Marek
- Third Department of Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Zdeňka Lacinová
- Institute of Medical Biochemistry and Laboratory Diagnostic, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Martin Haluzík
- Institute of Medical Biochemistry and Laboratory Diagnostic, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Michal Kršek
- Third Department of Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| |
Collapse
|
27
|
Maghami S, Zardooz H, Khodagholi F, Binayi F, Ranjbar Saber R, Hedayati M, Sahraei H, Ansari MA. Maternal separation blunted spatial memory formation independent of peripheral and hippocampal insulin content in young adult male rats. PLoS One 2018; 13:e0204731. [PMID: 30332425 PMCID: PMC6192583 DOI: 10.1371/journal.pone.0204731] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 09/13/2018] [Indexed: 01/22/2023] Open
Abstract
This study explores the effects of maternal separation as a chronic early life stress (ELS) on pancreatic islets insulin content and secretion, and their potential relationship with the hippocampus insulin content and spatial memory in young adulthood. Male rat offspring were divided into two groups: stress (STR) and non-stress (non-STR) groups. The animals of the STR group were separated from their mothers during postnatal days (PND) 1 to 21. During the weaning time, that is, PND-0 to PND-21, the body weight and length of the pups were measured. Blood samples were collected on PND-1, 21, 29 and 34 and during young adulthood (53±2 days) to determine plasma corticosterone and insulin levels. The young adult animals were also tested for spatial memory. One day after the memory test, the animals were decapitated and their pancreases were removed to measure the islets insulin content and secretion. Finally, the animals' hippocampi were isolated to determine their insulin content and insulin receptor protein amounts. During the period of weaning, the body weight and length of pups belonging to the STR group were significantly lower as compared to those in the non-STR group. Maternal separation did not change the plasma levels of insulin but increased plasma corticosterone levels from PND-21 to young adulthood and also reduced the islets insulin content but did not affect insulin secretion and the hippocampus insulin content and insulin receptor protein amount. Although, at the end of the memory tests, rats of the STR group reached the escape box at almost the same time and distance and with the same errors as rats of the non-STR group, the distance traveled to reach the escape box showed a steep reduction in the non-STR group as compared to the STR group after the first trial. Moreover, as compared to the STR group, the non-STR group showed an increasing trend for direct strategy to find the escape box. The islets insulin content and secretion, and the plasma insulin concentration were not significantly correlated with the hippocampus insulin content. From the results of the present study, it appears that the main behavioral effect of the maternal separation stress in the spatial memory task was to impair the strategy used by the animals to reach the escape box. This may indicate that maternal separation stress affects brain regions other than the hippocampus. Moreover, due to the reduction of the body weight and length of offspring belonging to the STR group, it should be further considered that both maternal separation and early life malnutrition are directly (and mechanistically) linked to cognitive alterations later in life in ways that are not dependent on peripheral and hippocampal insulin content.
Collapse
Affiliation(s)
- Soheila Maghami
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Homeira Zardooz
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- * E-mail: ,
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Binayi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roya Ranjbar Saber
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hedayat Sahraei
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Ansari
- Laser and Plasma Research Institute, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
28
|
Effects of moderate exercise on biochemical, morphological, and physiological parameters of the pancreas of female mice with estrogen deprivation and dyslipidemia. Med Mol Morphol 2018; 51:118-127. [PMID: 29335884 DOI: 10.1007/s00795-018-0179-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/04/2018] [Indexed: 12/17/2022]
Abstract
Menopausal women are at high risk of developing heart disease. However, physical exercise practice can reverse this scenario. We evaluated the biochemical, morphological, and physiological effects of moderate aerobic physical exercise on the pancreas of knockout mice for LDL receptor with estrogen deprivation by ovariectomy. Animals were divided into six groups (n = 5): sedentary non-ovariectomized control; sedentary ovariectomized control; trained ovariectomized control; sedentary non-ovariectomized LDL-R knockout; sedentary ovariectomized LDL-R knockout; and trained ovariectomized LDL-R knockout. Physical exercise practice promoted improvement in biometric and biochemical parameters analyzed, with reduction of visceral adipose tissue and VLDL, triglycerides, total cholesterol, and blood glucose levels. In addition, physical exercise practice altered the morphology of pancreatic islets and improved their response to the effects of menopause. Thus, physical exercise practice was fundamental to minimize the effects of dyslipidemia associated with ovariectomy in the pancreatic tissue of LDL-R knockout animals, contributing to reduce the risk of developing cardiac diseases in the menopause period.
Collapse
|
29
|
Galaviz KI, Narayan KMV, Lobelo F, Weber MB. Lifestyle and the Prevention of Type 2 Diabetes: A Status Report. Am J Lifestyle Med 2018; 12:4-20. [PMID: 30202378 PMCID: PMC6125024 DOI: 10.1177/1559827615619159] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 11/03/2015] [Accepted: 11/04/2015] [Indexed: 12/11/2022] Open
Abstract
Diabetes is a costly disease affecting 387 million individuals globally and 28 million in the United States. Its precursor, prediabetes, affects 316 and 86 million individuals globally and in the United States, respectively. People living with elevated blood glucose levels are at high risk for all-cause mortality and numerous cardiometabolic ailments. Fortunately, diabetes can be prevented or delayed by maintaining a healthy lifestyle and a healthy body weight. In this review, we summarize the literature around lifestyle diabetes prevention programs and provide recommendations for introducing prevention strategies in clinical practice. Overall, evidence supports the efficacy and effectiveness of lifestyle diabetes prevention interventions across clinical and community settings, delivery formats (eg, individual-, group-, or technology-based), and implementers (eg, clinicians, community members). Evidence-based diabetes prevention strategies that can be implemented in clinical practice include brief behavior change counseling, group-based education, community referrals, and health information technologies. These strategies represent opportunities where practitioners, communities, and health care systems can work together to provide individuals with education, support and opportunities to maintain healthy, diabetes-free lifestyles.
Collapse
Affiliation(s)
- Karla I. Galaviz
- Karla I. Galaviz, MSc, PhD, Emory Global Diabetes Research Center, Hubert Department of Global Health, Emory University, No. 1518 Clifton Rd, Atlanta, GA 30322; e-mail:
| | | | | | | |
Collapse
|
30
|
Nemati M, Zardooz H, Rostamkhani F, Abadi A, Foroughi F. High-fat diet effects on metabolic responses to chronic stress. Arch Physiol Biochem 2017; 123:182-191. [PMID: 28276709 DOI: 10.1080/13813455.2017.1295083] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
CONTEXT High-fat diets and chronic stress are prevalent risk factors for various chronic diseases in modern societies. OBJECTIVE This study investigated the effect of high-fat diet on glucose-related metabolic responses to chronic foot-shock stress. MATERIALS AND METHODS Male rats were divided into high-fat diet (containing 54.21% saturated and 44.89% unsaturated fatty acids) and normal diet groups and then into stress and non-stress subgroups. The diets were applied for 5 weeks, and stress was induced during the last week of the diet course. Plasma levels of metabolic parameters, HOMA-IR index, intra-abdominal fat weight, and islets' insulin secretion were assessed. RESULTS High-fat diet increased abdominal fat weight and plasma leptin, and insulin levels in response to stress without affecting HOMA-IR index and islets' insulin secretion. CONCLUSIONS High proportion of unsaturated fat may not lead to deleterious metabolic responses; however combined with chronic stress has a synergistic and adverse effect on visceral adiposity and results in elevated plasma leptin.
Collapse
Affiliation(s)
- Marzieh Nemati
- a Neurophysiology Research Center and Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Homeira Zardooz
- a Neurophysiology Research Center and Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences , Tehran , Iran
- b Neuroscience Research Center, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Fatemeh Rostamkhani
- c Department of Biology , College of Basic Sciences, Yadegar-e-Imam Khomeini (RAH) Shahre Rey Branch, Islamic Azad University , Tehran , Iran
| | - Alireza Abadi
- d Department of Community Medicine , School of Medicine, Shahid Beheshti University of Medical Sciences , Tehran , Iran , and
| | - Forough Foroughi
- e Department of Pathology , School of Medicine, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| |
Collapse
|
31
|
Karbaschi R, Zardooz H, Khodagholi F, Dargahi L, Salimi M, Rashidi F. Maternal high-fat diet intensifies the metabolic response to stress in male rat offspring. Nutr Metab (Lond) 2017; 14:20. [PMID: 28261314 PMCID: PMC5329934 DOI: 10.1186/s12986-017-0177-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 02/17/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The mother's consumption of high-fat food can affect glucose metabolism and the hypothalamic-pituitary-adrenal axis responsiveness in the offspring and potentially affect the metabolic responses to stress as well. This study examines the effect of maternal high-fat diet on the expression of pancreatic glucose transporter 2 and the secretion of insulin in response to stress in offspring. METHODS Female rats were randomly divided into normal and high-fat diet groups and were fed in accordance with their given diets from pre-pregnancy to the end of lactation. The offspring were divided into control (NC and HFC) and stress (NS and HFS) groups based on their mothers' diet and exposure to stress in adulthood. After the two-week stress induction period was over, an intraperitoneal glucose tolerance test (IPGTT) was performed and plasma glucose and insulin levels were assessed. The pancreas was then removed for measuring insulin secretion from the isolated islets as well as glucose transporter 2 mRNA expression and protein levels. RESULTS According to the results obtained, plasma corticosterone concentrations increased significantly on days 1 and 14 of the stress induction period and were lower on the last day compared to on the first day. In both the NS and HFS groups, stress reduced plasma insulin concentration in the IPGTT without changing the plasma glucose concentration, suggesting an increased insulin sensitivity in the NS and HFS groups, although more markedly in the latter. Stress reduced insulin secretion (at high glucose concentrations) and increased glucose transporter 2 mRNA and protein expression, especially in the HFS group. CONCLUSION Mothers' high-fat diet appears to intensify the stress response by changing the programming of the neuroendocrine system in the offspring.
Collapse
Affiliation(s)
- Roxana Karbaschi
- Neurophysiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, 19615-1178 Tehran, Iran
| | - Homeira Zardooz
- Neurophysiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, 19615-1178 Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, 19615-1178 Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- NeuroBiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mina Salimi
- Neurophysiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, 19615-1178 Tehran, Iran
| | - FatemehSadat Rashidi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
32
|
Jiménez-Maldonado A, Virgen-Ortiz A, Melnikov V, Rodríguez-Hernández A, Gamboa-Domínguez A, Montero S, Muñiz-Murguía J, Lemus M, Roces de Álvarez-Buylla E. Effect of moderate and high intensity chronic exercise on the pancreatic islet morphometry in healthy rats: BDNF receptor participation. Islets 2017; 9:1-10. [PMID: 27922332 PMCID: PMC5270655 DOI: 10.1080/19382014.2016.1260796] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The function and morphology of β-cells is largely dependent on insulin demand. As β-cells cover a bigger cell proportion in pancreas islets, changes of insulin producer cells affect the whole pancreatic islet morphology. Growth factors as the neurotrophins regulate the pancreas physiology, besides; physical exercise increases insulin sensitivity, and further modifies brain derived neurotrophic factor (BDNF) concentration in plasma. The aim of this study was to investigate the effects of chronic exercise (running in a treadmill for 8 weeks) intensity on pancreatic islet morphometry in healthy state. The BDNF receptor effect on the pancreatic islet morphometry was also evaluated. Adult male Wistar rats were divided in 6 groups: Control (C); moderate intensity training (MIT); high intensity training (HIT) did not treat with BDNF receptor inhibitor (K252a), and C, MIT and HIT treated with K252a. The results shown that chronic exercise induces β-cells hypertrophy without BDNF receptor participation. On the other hand, the moderate exercise increases the number of β cells per islet; the last effect does not require TrkB participation. In sedentary conditions, the K252a treatment reduced the β-cell density. Exercise intensity has differential effects on pancreas islet morphometry in healthy model; furthermore, BDNF receptor plays a role to maintain the amount of β-cells in sedentary state.
Collapse
Affiliation(s)
| | - Adolfo Virgen-Ortiz
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, México
| | | | | | | | - Sergio Montero
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, México
- Facultad de Medicina, Universidad de Colima, Colima, México
| | - Jesús Muñiz-Murguía
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, México
| | - Mónica Lemus
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, México
| | - Elena Roces de Álvarez-Buylla
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, México
- CONTACT Elena Roces de Álvarez-Buylla Jefe del Laboratorio de Neuroendocrinología, CUIB, Ave. 25 de Julio 965, Colima, Col. 298045 México
| |
Collapse
|
33
|
Mandel ER, Dunford EC, Trifonova A, Abdifarkosh G, Teich T, Riddell MC, Haas TL. Prazosin Can Prevent Glucocorticoid Mediated Capillary Rarefaction. PLoS One 2016; 11:e0166899. [PMID: 27861620 PMCID: PMC5115834 DOI: 10.1371/journal.pone.0166899] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 11/04/2016] [Indexed: 01/11/2023] Open
Abstract
Glucocorticoids (GC) elicit skeletal muscle capillary rarefaction, which can subsequently impair blood distribution and muscle function; however, the mechanisms have not been established. We hypothesized that CORT would inhibit endothelial cell survival signals but that treatment with the alpha-1 adrenergic receptor inhibitor prazosin, which leads to angiogenesis in skeletal muscle of healthy rats, would reverse these effects and induce angiogenesis within the skeletal muscle of corticosterone (CORT)-treated rats. Male Sprague Dawley rats were implanted subcutaneously with CORT pellets (400 mg/rat), with or without concurrent prazosin treatment (50mg/L in drinking water), for 1 or 2 weeks. Skeletal muscle capillary rarefaction, as indicated by a significant reduction in capillary-to-fiber ratio (C:F), occurred after 2 weeks of CORT treatment. Concurrent prazosin administration prevented this capillary rarefaction in CORT-treated animals but did not induce angiogenesis or arteriogenesis as was observed with prazosin treatment in control rats. CORT treatment reduced the mRNA level of Angiopoietin-1 (Ang-1), which was partially offset in the muscles of rats that received 2 weeks of co-treatment with prazosin. In 2W CORT animals, prazosin treatment elicited a significant increase in vascular endothelial growth factor-A (VEGF-A) mRNA and protein. Conversely prazosin did not rescue CORT-induced reductions in transforming growth factor beta-1 (TGFβ1 and matrix metalloproteinase-2 (MMP-2) mRNA. To determine if CORT impaired shear stress dependent signaling, cultured rat skeletal muscle endothelial cells were pre-treated with CORT (600nM) for 48 hours, then exposed to 15 dynes/cm2 shear stress or maintained with no flow. CORT blunted the shear stress-induced increase in pSer473 Akt, while pThr308 Akt, ERK1/2 and p38 phosphorylation and nitric oxide (NO) production were unaffected. This study demonstrates that GC-mediated capillary rarefaction is associated with a reduction in Ang-1 mRNA within the skeletal muscle microenvironment and that concurrent prazosin treatment effectively increases VEGF-A levels and prevents capillary loss.
Collapse
Affiliation(s)
- Erin R. Mandel
- School of Kinesiology and Health Science and Muscle Health Research Centre, York University, Toronto, Canada
| | - Emily C. Dunford
- School of Kinesiology and Health Science and Muscle Health Research Centre, York University, Toronto, Canada
| | | | - Ghoncheh Abdifarkosh
- School of Kinesiology and Health Science and Muscle Health Research Centre, York University, Toronto, Canada
| | - Trevor Teich
- School of Kinesiology and Health Science and Muscle Health Research Centre, York University, Toronto, Canada
| | - Michael C. Riddell
- School of Kinesiology and Health Science and Muscle Health Research Centre, York University, Toronto, Canada
| | - Tara L. Haas
- School of Kinesiology and Health Science and Muscle Health Research Centre, York University, Toronto, Canada
- * E-mail:
| |
Collapse
|
34
|
Liu L, Wang F, Lu H, Cao S, Du Z, Wang Y, Feng X, Gao Y, Zha M, Guo M, Sun Z, Wang J. Effects of Noise Exposure on Systemic and Tissue-Level Markers of Glucose Homeostasis and Insulin Resistance in Male Mice. ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:1390-1398. [PMID: 27128844 PMCID: PMC5010391 DOI: 10.1289/ehp162] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 01/24/2016] [Accepted: 04/12/2016] [Indexed: 06/05/2023]
Abstract
BACKGROUND Epidemiological studies have indicated that noise exposure is associated with an increased risk of type 2 diabetes mellitus (T2DM). However, the nature of the connection between noise exposure and T2DM remains to be explored. OBJECTIVES We explored whether and how noise exposure affects glucose homeostasis in mice as the initial step toward T2DM development. METHODS Male ICR mice were randomly assigned to one of four groups: the control group and three noise groups (N20D, N10D, and N1D), in which the animals were exposed to white noise at 95 decibel sound pressure level (dB SPL) for 4 hr per day for 20 successive days, 10 successive days, or 1 day, respectively. Glucose tolerance and insulin sensitivity were evaluated 1 day, 1 week, and 1 month after the final noise exposure (1DPN, 1WPN, and 1MPN). Standard immunoblots, immunohistochemical methods, and enzyme-linked immunosorbent assays (ELISA) were performed to assess insulin signaling in skeletal muscle, the morphology of β cells, and plasma corticosterone levels. RESULTS Noise exposure for 1 day caused transient glucose intolerance and insulin resistance, whereas noise exposure for 10 and 20 days had no effect on glucose tolerance but did cause prolonged insulin resistance and an increased insulin response to glucose challenge. Akt phosphorylation and GLUT4 translocation in response to exogenous insulin were decreased in the skeletal muscle of noise-exposed animals. CONCLUSIONS Noise exposure at 95 dB SPL caused insulin resistance in male ICR mice, which was prolonged with longer noise exposure and was likely related to the observed blunted insulin signaling in skeletal muscle. CITATION Liu L, Wang F, Lu H, Cao S, Du Z, Wang Y, Feng X, Gao Y, Zha M, Guo M, Sun Z, Wang J. 2016. Effects of noise exposure on systemic and tissue-level markers of glucose homeostasis and insulin resistance in male mice. Environ Health Perspect 124:1390-1398; http://dx.doi.org/10.1289/EHP162.
Collapse
Affiliation(s)
- Lijie Liu
- Department of Physiology, Medical College, Southeast University, Nanjing, China
| | - Fanfan Wang
- Institute of Life Sciences, Southeast University, Nanjing, China
| | - Haiying Lu
- Institute of Life Sciences, Southeast University, Nanjing, China
| | - Shuangfeng Cao
- Institute of Life Sciences, Southeast University, Nanjing, China
| | - Ziwei Du
- Medical College, Southeast University, Nanjing, China
| | - Yongfang Wang
- Medical College, Southeast University, Nanjing, China
| | - Xian Feng
- Medical College, Southeast University, Nanjing, China
| | - Ye Gao
- Medical College, Southeast University, Nanjing, China
| | - Mingming Zha
- Medical College, Southeast University, Nanjing, China
| | - Min Guo
- Medical College, Southeast University, Nanjing, China
| | - Zilin Sun
- Department of Endocrinology, Medical College, Affiliated ZhongDa Hospital of Southeast University, Nanjing, China
| | - Jian Wang
- Department of Physiology, Medical College, Southeast University, Nanjing, China
- School of Human Communication Disorders, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
35
|
Diz-Chaves Y, Gil-Lozano M, Toba L, Fandiño J, Ogando H, González-Matías LC, Mallo F. Stressing diabetes? The hidden links between insulinotropic peptides and the HPA axis. J Endocrinol 2016; 230:R77-94. [PMID: 27325244 DOI: 10.1530/joe-16-0118] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 06/20/2016] [Indexed: 12/25/2022]
Abstract
Diabetes mellitus exerts metabolic stress on cells and it provokes a chronic increase in the long-term activity of the hypothalamus-pituitary-adrenocortical (HPA) axis, perhaps thereby contributing to insulin resistance. GLP-1 receptor (GLP-1R) agonists are pleiotropic hormones that not only affect glycaemic and metabolic control, but they also produce many other effects including activation of the HPA axis. In fact, several of the most relevant effects of GLP-1 might involve, at least in part, the modulation of the HPA axis. Thus, the anorectic activity of GLP-1 could be mediated by increasing CRF at the hypothalamic level, while its lipolytic effects could imply a local increase in glucocorticoids and glucocorticoid receptor (GC-R) expression in adipose tissue. Indeed, the potent activation of the HPA axis by GLP-1R agonists occurs within the range of therapeutic doses and with a short latency. Interestingly, the interactions of GLP-1 with the HPA axis may underlie most of the effects of GLP-1 on food intake control, glycaemic metabolism, adipose tissue biology and the responses to stress. Moreover, such activity has been observed in animal models (mice and rats), as well as in normal humans and in type I or type II diabetic patients. Accordingly, better understanding of how GLP-1R agonists modulate the activity of the HPA axis in diabetic subjects, especially obese individuals, will be crucial to design new and more efficient therapies for these patients.
Collapse
Affiliation(s)
- Yolanda Diz-Chaves
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| | - Manuel Gil-Lozano
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| | - Laura Toba
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| | - Juan Fandiño
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| | - Hugo Ogando
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| | - Lucas C González-Matías
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| | - Federico Mallo
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| |
Collapse
|
36
|
Zheng H, Wu J, Jin Z, Yan LJ. Protein Modifications as Manifestations of Hyperglycemic Glucotoxicity in Diabetes and Its Complications. BIOCHEMISTRY INSIGHTS 2016; 9:1-9. [PMID: 27042090 PMCID: PMC4807886 DOI: 10.4137/bci.s36141] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 02/25/2016] [Accepted: 02/27/2016] [Indexed: 02/07/2023]
Abstract
Diabetes and its complications are hyperglycemic toxicity diseases. Many metabolic pathways in this array of diseases become aberrant, which is accompanied with a variety of posttranslational protein modifications that in turn reflect diabetic glucotoxicity. In this review, we summarize some of the most widely studied protein modifications in diabetes and its complications. These modifications include glycation, carbonylation, nitration, cysteine S-nitrosylation, acetylation, sumoylation, ADP-ribosylation, O-GlcNAcylation, and succination. All these posttranslational modifications can be significantly attributed to oxidative stress and/or carbon stress induced by diabetic redox imbalance that is driven by activation of pathways, such as the polyol pathway and the ADP-ribosylation pathway. Exploring the nature of these modifications should facilitate our understanding of the pathological mechanisms of diabetes and its associated complications.
Collapse
Affiliation(s)
- Hong Zheng
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, UNT Health Science Center, Fort Worth, TX, USA.; Department of Basic Theory of Traditional Chinese Medicine, College of Basic Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jinzi Wu
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, UNT Health Science Center, Fort Worth, TX, USA
| | - Zhen Jin
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, UNT Health Science Center, Fort Worth, TX, USA
| | - Liang-Jun Yan
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, UNT Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
37
|
Regulation of Glucose Homeostasis by Glucocorticoids. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015. [PMID: 26215992 DOI: 10.1007/978-1-4939-2895-8_5] [Citation(s) in RCA: 389] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Glucocorticoids are steroid hormones that regulate multiple aspects of glucose homeostasis. Glucocorticoids promote gluconeogenesis in liver, whereas in skeletal muscle and white adipose tissue they decrease glucose uptake and utilization by antagonizing insulin response. Therefore, excess glucocorticoid exposure causes hyperglycemia and insulin resistance. Glucocorticoids also regulate glycogen metabolism. In liver, glucocorticoids increase glycogen storage, whereas in skeletal muscle they play a permissive role for catecholamine-induced glycogenolysis and/or inhibit insulin-stimulated glycogen synthesis. Moreover, glucocorticoids modulate the function of pancreatic α and β cells to regulate the secretion of glucagon and insulin, two hormones that play a pivotal role in the regulation of blood glucose levels. Overall, the major glucocorticoid effect on glucose homeostasis is to preserve plasma glucose for brain during stress, as transiently raising blood glucose is important to promote maximal brain function. In this chapter we will discuss the current understanding of the mechanisms underlying different aspects of glucocorticoid-regulated mammalian glucose homeostasis.
Collapse
|
38
|
Lee SF, Pei D, Chi MJ, Jeng C. An investigation and comparison of the effectiveness of different exercise programmes in improving glucose metabolism and pancreatic β cell function of type 2 diabetes patients. Int J Clin Pract 2015; 69:1159-70. [PMID: 26119968 DOI: 10.1111/ijcp.12679] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Moderately intensive aerobic exercise can improve glucose metabolism and pancreatic β cell function in diabetic patients. To date, there is no evidence to support the long-term effectiveness of home-based exercise interventions on these outcomes. OBJECTIVE This study investigated the effectiveness of two moderately intense exercise programmes on glucose metabolism and pancreatic β cell function in type 2 diabetes mellitus (T2DM) patients. METHODS A randomised controlled trial of 120 T2DM patients (with a mean age of 55.54 ± 9.09 years) was conducted. Patients were assigned by block randomisation to either an aerobic exercise group (AEG), an accumulated million steps group (AMSG), or a control group (CG); each consisting of 40 patients. Glucose metabolism and pancreatic β cell function of patients were measured at three time intervals for 1 year. RESULTS There was no difference in baseline scores, and respective compliance rates for the AEG and AMSG were 94.4% and 99.2%. After generalised estimating equation analysis, the AMSG results for glycated haemoglobin (HbA1c) were significantly lower than those of the CG. The insulinogenic index-acute insulin response (BIGTT-AIR ) of both exercise groups was significantly higher than that of the CG. The AMSG group improved their overall HbA1c and BIGTT-AIR results compared with the AEG group after 3 months exercise programme (T1 ) and 12 months of implementation (T2 ). CONCLUSION This study demonstrates that regardless of the type of exercise intervention, it is potentially beneficially effective for glucose metabolism and pancreatic β cell function in T2DM patients. The AMSG had better glucose metabolism and pancreatic β cell function compared with those in the AEG. Nurses can easily integrate exercise interventions into T2DM patient care plans.
Collapse
Affiliation(s)
- S F Lee
- Cardinal Tien College of Healthcare & Management, New Taipei City, Taiwan
- Graduate Institute of Nursing, College of Nursing, Taipei Medical University, Taipei, Taiwan
| | - D Pei
- Department of Internal Medicine, Cardinal Tien Hospital, New Taipei City, Taiwan
- Medical School, Fu Jen Catholic University, New Taipei City, Taiwan
| | - M J Chi
- Graduate Institute of Nursing, College of Nursing, Taipei Medical University, Taipei, Taiwan
| | - C Jeng
- Graduate Institute of Nursing, College of Nursing, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
39
|
Beaudry JL, Dunford EC, Leclair E, Mandel ER, Peckett AJ, Haas TL, Riddell MC. Voluntary exercise improves metabolic profile in high-fat fed glucocorticoid-treated rats. J Appl Physiol (1985) 2015; 118:1331-43. [PMID: 25792713 DOI: 10.1152/japplphysiol.00467.2014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 03/15/2015] [Indexed: 01/12/2023] Open
Abstract
Diabetes is rapidly induced in young male Sprague-Dawley rats following treatment with exogenous corticosterone (CORT) and a high-fat diet (HFD). Regular exercise alleviates insulin insensitivity and improves pancreatic β-cell function in insulin-resistant/diabetic rodents, but its effect in an animal model of elevated glucocorticoids is unknown. We examined the effect of voluntary exercise (EX) on diabetes development in CORT-HFD-treated male Sprague-Dawley rats (∼6 wk old). Animals were acclimatized to running wheels for 2 wk, then given a HFD, either wax (placebo) or CORT pellets, and split into 4 groups: placebo-sedentary (SED) or -EX and CORT-SED or -EX. After 2 wk of running combined with treatment, CORT-EX animals had reduced visceral adiposity, and increased skeletal muscle type IIb/x fiber area, oxidative capacity, capillary-to-fiber ratio and insulin sensitivity compared with CORT-SED animals (all P < 0.05). Although CORT-EX animals still had fasting hyperglycemia, these values were significantly improved compared with CORT-SED animals (14.3 ± 1.6 vs. 18.8 ± 0.9 mM). In addition, acute in vivo insulin response to an oral glucose challenge was enhanced ∼2-fold in CORT-EX vs. CORT-SED (P < 0.05) which was further demonstrated ex vivo in isolated islets. We conclude that voluntary wheel running in rats improves, but does not fully normalize, the metabolic profile and skeletal muscle composition of animals administered CORT and HFD.
Collapse
Affiliation(s)
- Jacqueline L Beaudry
- School of Kinesiology and Health Science, Faculty of Health, Muscle Health Research Center and Physical Activity and Chronic Disease Unit, York University, Toronto, Ontario, Canada
| | - Emily C Dunford
- School of Kinesiology and Health Science, Faculty of Health, Muscle Health Research Center and Physical Activity and Chronic Disease Unit, York University, Toronto, Ontario, Canada
| | - Erwan Leclair
- School of Kinesiology and Health Science, Faculty of Health, Muscle Health Research Center and Physical Activity and Chronic Disease Unit, York University, Toronto, Ontario, Canada
| | - Erin R Mandel
- School of Kinesiology and Health Science, Faculty of Health, Muscle Health Research Center and Physical Activity and Chronic Disease Unit, York University, Toronto, Ontario, Canada
| | - Ashley J Peckett
- School of Kinesiology and Health Science, Faculty of Health, Muscle Health Research Center and Physical Activity and Chronic Disease Unit, York University, Toronto, Ontario, Canada
| | - Tara L Haas
- School of Kinesiology and Health Science, Faculty of Health, Muscle Health Research Center and Physical Activity and Chronic Disease Unit, York University, Toronto, Ontario, Canada
| | - Michael C Riddell
- School of Kinesiology and Health Science, Faculty of Health, Muscle Health Research Center and Physical Activity and Chronic Disease Unit, York University, Toronto, Ontario, Canada
| |
Collapse
|
40
|
Guthrie GE, Bogue RJ. Impact of a Shared Medical Appointment Lifestyle Intervention on Weight and Lipid Parameters in Individuals with Type 2 Diabetes: A Clinical Pilot. J Am Coll Nutr 2015; 34:300-9. [DOI: 10.1080/07315724.2014.933454] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
41
|
Sadeghimahalli F, Karbaschi R, Zardooz H, Khodagholi F, Rostamkhani F. Effect of early life stress on pancreatic isolated islets' insulin secretion in young adult male rats subjected to chronic stress. Endocrine 2015; 48:493-503. [PMID: 25030548 DOI: 10.1007/s12020-014-0337-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 06/10/2014] [Indexed: 01/12/2023]
Abstract
Early stressful experiences may predispose organisms to certain disorders, including those of metabolic defects. This study aimed to explore the effects of early life stress on pancreatic insulin secretion and glucose transporter 2 (GLUT2) protein levels in stressed young adult male rats. Foot shock stress was induced in early life (at 2 weeks of age) and/or in young adulthood (at 8-10 weeks of age) for five consecutive days. Blood samples were taken before and after stress exposure in young adult rats. At the end of the experiment, glucose tolerance, isolated islets' insulin secretion, and pancreatic amounts of GLUT2 protein were measured. Our results show that early life stress has no effect on basal plasma corticosterone levels and adrenal weight, either alone or combined with young adulthood stress, but that early life + young adulthood stress could prevent weight gain, and cause an increase in basal plasma glucose and insulin. The homeostasis model assessment of insulin resistance index did not increase, when the rats were subjected to early life stress alone, but increased when combined with young adulthood stress. Moreover, glucose tolerance was impaired by the combination of early life + young adult stress. There was a decrease in islet's insulin secretion in rats subjected to early life stress in response to 5.6 mM glucose concentration, but an increase with a concentration of 16.7 mM glucose. However, in rats subjected to early life + young adulthood stress, islet's insulin secretion increased in response to both the levels of glucose concentrations. GLUT2 protein levels decreased in response to early life stress and early life + young adulthood stress, but there was a greater decrease in the early life stress group. In conclusion, perhaps early life stress sensitizes the body to stressors later in life, making it more susceptible to metabolic syndrome only when the two are in combination.
Collapse
Affiliation(s)
- Forouzan Sadeghimahalli
- Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | | | | |
Collapse
|
42
|
Gallo LC, Fortmann AL, McCurley JL, Isasi CR, Penedo FJ, Daviglus ML, Roesch SC, Talavera GA, Gouskova N, Gonzalez F, Schneiderman N, Carnethon MR. Associations of structural and functional social support with diabetes prevalence in U.S. Hispanics/Latinos: results from the HCHS/SOL Sociocultural Ancillary Study. J Behav Med 2015; 38:160-70. [PMID: 25107504 PMCID: PMC4349398 DOI: 10.1007/s10865-014-9588-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 07/25/2014] [Indexed: 12/25/2022]
Abstract
Little research has examined associations of social support with diabetes (or other physical health outcomes) in Hispanics, who are at elevated risk. We examined associations between social support and diabetes prevalence in the Hispanic Community Health Study/Study of Latinos Sociocultural Ancillary Study. Participants were 5,181 adults, 18-74 years old, representing diverse Hispanic backgrounds, who underwent baseline exam with fasting blood draw, oral glucose tolerance test, medication review, sociodemographic assessment, and sociocultural exam with functional and structural social support measures. In adjusted analyses, one standard deviation higher structural and functional social support related to 16 and 15% lower odds, respectively, of having diabetes. Structural and functional support were related to both previously diagnosed diabetes (OR = .84 and .88, respectively) and newly recognized diabetes prevalence (OR = .84 and .83, respectively). Higher functional and structural social support are associated with lower diabetes prevalence in Hispanics/Latinos.
Collapse
Affiliation(s)
- Linda C Gallo
- Department of Psychology, San Diego State University, 9245 Sky Park Court, Suite 115, San Diego, CA, 92123, USA,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Jiménez-Maldonado A, de Álvarez-Buylla ER, Montero S, Melnikov V, Castro-Rodríguez E, Gamboa-Domínguez A, Rodríguez-Hernández A, Lemus M, Murguía JM. Chronic exercise increases plasma brain-derived neurotrophic factor levels, pancreatic islet size, and insulin tolerance in a TrkB-dependent manner. PLoS One 2014; 9:e115177. [PMID: 25531651 PMCID: PMC4274083 DOI: 10.1371/journal.pone.0115177] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 11/19/2014] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Physical exercise improves glucose metabolism and insulin sensitivity. Brain-derived neurotrophic factor (BDNF) enhances insulin activity in diabetic rodents. Because physical exercise modifies BDNF production, this study aimed to investigate the effects of chronic exercise on plasma BDNF levels and the possible effects on insulin tolerance modification in healthy rats. METHODS Wistar rats were divided into five groups: control (sedentary, C); moderate- intensity training (MIT); MIT plus K252A TrkB blocker (MITK); high-intensity training (HIT); and HIT plus K252a (HITK). Training comprised 8 weeks of treadmill running. Plasma BDNF levels (ELISA assay), glucose tolerance, insulin tolerance, and immunohistochemistry for insulin and the pancreatic islet area were evaluated in all groups. In addition, Bdnf mRNA expression in the skeletal muscle was measured. PRINCIPAL FINDINGS Chronic treadmill exercise significantly increased plasma BDNF levels and insulin tolerance, and both effects were attenuated by TrkB blocking. In the MIT and HIT groups, a significant TrkB-dependent pancreatic islet enlargement was observed. MIT rats exhibited increased liver glycogen levels following insulin administration in a TrkB-independent manner. CONCLUSIONS/SIGNIFICANCE Chronic physical exercise exerted remarkable effects on insulin regulation by inducing significant increases in the pancreatic islet size and insulin sensitivity in a TrkB-dependent manner. A threshold for the induction of BNDF in response to physical exercise exists in certain muscle groups. To the best of our knowledge, these are the first results to reveal a role for TrkB in the chronic exercise-mediated insulin regulation in healthy rats.
Collapse
Affiliation(s)
| | | | - Sergio Montero
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, México
| | | | - Elena Castro-Rodríguez
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, México
| | - Armando Gamboa-Domínguez
- Depto de Patología, Instituto Nacional de Nutrición y Ciencias Médicas "Salvador Zubirán,” México City, México D.F.
| | | | - Mónica Lemus
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, México
| | - Jesús Muñiz Murguía
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, México
| |
Collapse
|
44
|
Tripathi YB, Pandey N. Carbimazole inhibits TNF-α expression in Fat-induced hypothyroidism. J Diabetes Metab Disord 2014; 13:83. [PMID: 25258706 PMCID: PMC4174658 DOI: 10.1186/s40200-014-0083-4] [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: 10/21/2013] [Accepted: 07/22/2014] [Indexed: 11/26/2022]
Abstract
The effect of the carbimazole on expression of tumor necrosis factor (TNF-α) in liver, was investigated in an experimental model of high fat diet (HFD) induced obesity. The HFD (orally given for 4 months) induced TNF-α in liver tissue along with raised serum triglyceride (TG), cholesterol and high TSH (62%). In carbimazole (1 mg/100 gbw) treatment, the induction of TNF-α was significantly inhibited, without affecting other parameters. It also improved the liver function, which was raised due to HFD in experimental control rats.
Collapse
Affiliation(s)
- Yamani Bhusan Tripathi
- Department of Medicinal Chemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005 India
| | - Nidhi Pandey
- Department of Medicinal Chemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005 India
| |
Collapse
|
45
|
Fardet L, Fève B. Systemic Glucocorticoid Therapy: a Review of its Metabolic and Cardiovascular Adverse Events. Drugs 2014; 74:1731-45. [DOI: 10.1007/s40265-014-0282-9] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
46
|
Panagiotakopoulos L, Neigh GN. Development of the HPA axis: where and when do sex differences manifest? Front Neuroendocrinol 2014; 35:285-302. [PMID: 24631756 DOI: 10.1016/j.yfrne.2014.03.002] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 02/22/2014] [Accepted: 03/04/2014] [Indexed: 01/21/2023]
Abstract
Sex differences in the response to stress contribute to sex differences in somatic, neurological, and psychiatric diseases. Despite a growing literature on the mechanisms that mediate sex differences in the stress response, the ontogeny of these differences has not been comprehensively reviewed. This review focuses on the development of the hypothalamic-pituitary-adrenal (HPA) axis, a key component of the body's response to stress, and examines the critical points of divergence during development between males and females. Insight gained from animal models and clinical studies are presented to fully illustrate the current state of knowledge regarding sex differences in response to stress over development. An appreciation for the developmental timelines of the components of the HPA axis will provide a foundation for future areas of study by highlighting both what is known and calling attention to areas in which sex differences in the development of the HPA axis have been understudied.
Collapse
Affiliation(s)
| | - Gretchen N Neigh
- Emory University, Department of Physiology, United States; Emory University, Department of Psychiatry & Behavioral Sciences, United States.
| |
Collapse
|
47
|
Biessels GJ, Strachan MWJ, Visseren FLJ, Kappelle LJ, Whitmer RA. Dementia and cognitive decline in type 2 diabetes and prediabetic stages: towards targeted interventions. Lancet Diabetes Endocrinol 2014; 2:246-55. [PMID: 24622755 DOI: 10.1016/s2213-8587(13)70088-3] [Citation(s) in RCA: 382] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Type 2 diabetes is associated with dementia, and also with more slight cognitive decrements. In this Review we discuss trajectories from normal cognition to dementia in people with type 2 diabetes, and explore opportunities for treatment. Slight diabetes-associated cognitive decrements and dementia affect different age groups and show a different evolution. These cognitive entities should therefore not be regarded as a continuum, although their effects might be additive. Vascular damage is a key underlying process in both entities. Glucose-mediated processes and other metabolic disturbances might also have a role. No treatment has been established, but management of vascular risk factors and optimisation of glycaemic control could have therapeutic benefit. We identify possible opportunities for intervention to improve cognitive outcomes in people with type 2 diabetes, and suggest how treatment can be tailored to individual risk profiles and comorbidities.
Collapse
Affiliation(s)
- Geert Jan Biessels
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands.
| | | | - Frank L J Visseren
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht, Netherlands
| | - L Jaap Kappelle
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
| | | |
Collapse
|
48
|
Beaudry JL, Riddell MC. Response to 'letter to the editor' by Dr Rafacho. Diabetes Metab Res Rev 2014; 30:122-3. [PMID: 24500991 DOI: 10.1002/dmrr.2472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 09/10/2013] [Indexed: 11/09/2022]
|
49
|
Rafacho A. Effects of glucocorticoids and exercise on pancreatic β-cell function and diabetes development: comments on Beaudry and Riddel. Diabetes Metab Res Rev 2014; 30:120-1. [PMID: 24500990 DOI: 10.1002/dmrr.2473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 09/10/2013] [Indexed: 11/11/2022]
Affiliation(s)
- Alex Rafacho
- Department of Physiological Sciences, Centre of Biological Sciences, Federal University of Santa Catarina (UFSC), 88040-900 Florianópolis, SC, Brazil
| |
Collapse
|
50
|
Henderson M, Gray-Donald K, Rabasa-Lhoret R, Bastard JP, Barnett TA, Benedetti A, Chaput JP, Tremblay A, Lambert M. Insulin secretion and its association with physical activity, fitness and screen time in children. Obesity (Silver Spring) 2014; 22:504-11. [PMID: 24030901 DOI: 10.1002/oby.20619] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 08/21/2013] [Accepted: 08/27/2013] [Indexed: 01/22/2023]
Abstract
OBJECTIVES To determine the independent associations of moderate to vigorous physical activity (MVPA), fitness, screen time, and adiposity with insulin secretion in children. DESIGN AND METHODS Caucasian youth (n = 423/630), 8-10 years old, with at least one obese biological parent, were studied (QUALITY cohort). Insulin secretion was measured using HOMA2-%B, area under the curve (AUC) of insulin to glucose over the first 30 minutes (AUC I/G(t30min)) of the OGTT and AUC I/G(t120min) over 2 hours. Fitness was measured by VO₂peak ; percent fat mass (PFM) by DXA; 7-day MVPA by accelerometry; self-reported screen time included television, video game, or computer use. Models were adjusted for age, sex, season, puberty, PFM, and insulin sensitivity [IS] (HOMA2-IS, Matsuda-ISI). RESULTS PFM was strongly associated with insulin secretion, even after adjustment for IS: for every 1% increase in PFM, insulin secretion increased from 0.3% to 0.8% across indices. MVPA was negatively associated with HOMA2-%B (P < 0.05), but not with OGTT-derived measures. Fitness was negatively associated with AUC I/G(t120min) (P < 0.05). Screen time showed a trend toward higher HOMA2-%B in girls (P = 0.060). CONCLUSIONS In children with an obese parent, lower insulin secretion is associated with lower adiposity, higher MVPA, better fitness, and possibly reduced screen time.
Collapse
Affiliation(s)
- M Henderson
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montréal, Quebec, Canada; Division of Endocrinology, Department of Pediatrics, CHU Sainte-Justine and Université de Montréal, Montreal, Quebec, Canada; Centre de Recherche du CHU Sainte Justine, Montréal, Quebec, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|