1
|
Iwai S, Kaji K, Nishimura N, Kubo T, Tomooka F, Shibamoto A, Suzuki J, Tsuji Y, Fujinaga Y, Kitagawa K, Namisaki T, Akahane T, Yoshiji H. Glucagon-like peptide-1 receptor agonist, semaglutide attenuates chronic liver disease-induced skeletal muscle atrophy in diabetic mice. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166770. [PMID: 37276988 DOI: 10.1016/j.bbadis.2023.166770] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/15/2023] [Accepted: 05/28/2023] [Indexed: 06/07/2023]
Abstract
A glucagon-like peptide-1 receptor agonist (GLP-1RA) has recently been established as a pharmacological option for the treatment of type 2 diabetes. Recent studies have demonstrated the molecular role of GLP-1R in skeletal muscle homeostasis; however, the therapeutic efficacy of semaglutide, a GLP-1RA, on skeletal muscle atrophy in chronic liver disease (CLD) under diabetic conditions remains unclear. In the present study, semaglutide effectively inhibited psoas muscle atrophy and suppressed declines in grip strength in a diethoxycarbonyl-1,4-dihydrocollidine (DDC) diet-fed diabetic KK-Ay mouse model. Moreover, semaglutide inhibited ubiquitin-proteosome-mediated skeletal muscle proteolysis and promoted myogenesis in palmitic acid (PA)-stimulated C2C12 murine myocytes. Mechanistically, this effect of semaglutide on skeletal muscle atrophy was mediated by multiple functional pathways. First, semaglutide protected against hepatic injury in mice accompanied by increased production of insulin-like growth factor 1 and reduced accumulation of reactive oxygen species (ROS). These effects were associated with decreased proinflammatory cytokines and ROS accumulation, leading to the suppression of ubiquitin-proteosome muscle degradation. Moreover, semaglutide inhibited the amino acid starvation-related stress signaling that was activated under chronic liver injury, resulting in the recovery of the mammalian target of rapamycin activity in the skeletal muscle of DDC-diet fed KK-Ay mice. Second, semaglutide improved skeletal muscle atrophy by directly stimulating GLP-1R in myocytes. Semaglutide induced cAMP-mediated activation of PKA and AKT, enhanced mitochondrial biogenesis, and reduced ROS accumulation, thereby resulting in inhibition of NF-κB/myostatin-mediated ubiquitin-proteosome degradation and the augmentation of heat-shock factor-1-mediated myogenesis. Collectively, semaglutide may have potential as a new therapeutic strategy for CLD-related skeletal muscle wasting.
Collapse
Affiliation(s)
- Satoshi Iwai
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Kosuke Kaji
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan.
| | - Norihisa Nishimura
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Takahiro Kubo
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Fumimasa Tomooka
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Akihiko Shibamoto
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Junya Suzuki
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Yuki Tsuji
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Yukihisa Fujinaga
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Koh Kitagawa
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Tadashi Namisaki
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Takemi Akahane
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Hitoshi Yoshiji
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| |
Collapse
|
2
|
Jiménez-Loygorri JI, Benítez-Fernández R, Viedma-Poyatos Á, Zapata-Muñoz J, Villarejo-Zori B, Gómez-Sintes R, Boya P. Mitophagy in the retina: Viewing mitochondrial homeostasis through a new lens. Prog Retin Eye Res 2023; 96:101205. [PMID: 37454969 DOI: 10.1016/j.preteyeres.2023.101205] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 07/11/2023] [Accepted: 07/13/2023] [Indexed: 07/18/2023]
Abstract
Mitochondrial function is key to support metabolism and homeostasis in the retina, an organ that has one of the highest metabolic rates body-wide and is constantly exposed to photooxidative damage and external stressors. Mitophagy is the selective autophagic degradation of mitochondria within lysosomes, and can be triggered by distinct stimuli such as mitochondrial damage or hypoxia. Here, we review the importance of mitophagy in retinal physiology and pathology. In the developing retina, mitophagy is essential for metabolic reprogramming and differentiation of retina ganglion cells (RGCs). In basal conditions, mitophagy acts as a quality control mechanism, maintaining a healthy mitochondrial pool to meet cellular demands. We summarize the different autophagy- and mitophagy-deficient mouse models described in the literature, and discuss the potential role of mitophagy dysregulation in retinal diseases such as glaucoma, diabetic retinopathy, retinitis pigmentosa, and age-related macular degeneration. Finally, we provide an overview of methods used to monitor mitophagy in vitro, ex vivo, and in vivo. This review highlights the important role of mitophagy in sustaining visual function, and its potential as a putative therapeutic target for retinal and other diseases.
Collapse
Affiliation(s)
- Juan Ignacio Jiménez-Loygorri
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain.
| | - Rocío Benítez-Fernández
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain; Departament of Neuroscience and Movement Science, Faculty of Science and Medicine, University of Fribourg, 1700, Fribourg, Switzerland
| | - Álvaro Viedma-Poyatos
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Juan Zapata-Muñoz
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Beatriz Villarejo-Zori
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Raquel Gómez-Sintes
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Patricia Boya
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain; Departament of Neuroscience and Movement Science, Faculty of Science and Medicine, University of Fribourg, 1700, Fribourg, Switzerland.
| |
Collapse
|
3
|
Kim JM, Choi JS, Jung J, Yeo SG, Kim SH. Inhibitory effect of parthenolide on peripheral nerve degeneration. Anat Sci Int 2023; 98:529-539. [PMID: 37024641 DOI: 10.1007/s12565-023-00718-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/15/2023] [Indexed: 04/08/2023]
Abstract
Traumatic axonal damage disrupts connections between neurons, leading to the loss of motor and sensory functions. Although damaged peripheral nerves can regenerate, recovery depends on the variety and severity of nerve damage. Thus, many phytochemicals have been studied for their ability to reduce peripheral nerve degeneration, and among them, Parthenolide (PTL), which is extracted from Feverfew has effects against production of free radicals, inflammation, and apoptosis. Thus, we conducted a study to investigate whether PTL has an inhibitory effect on peripheral nerve degeneration during peripheral nerve damage. To verify the effect of PTL on peripheral nerve degeneration process, a morphological comparison of peripheral nerves with and without PTL was performed. PTL significantly reduced the quantity of fragmented ovoid formations at 3DIV (days in vitro). Immunostaining for MBP revealed that the ratio of intact myelin sheaths increased significantly in sciatic nerve with PTL compared with absence of PTL at 3DIV. Furthermore, nerve fibers in the presence of PTL maintained the continuity of Neurofilament (NF) compared to those without at 3DIV. Immunostaining for LAMP1 and p75 NTR showed that the expression of LAMP1 and p75 NTR decreased in the nerve after PTL addition at 3DIV. Lastly, immunostaining for anti-Ki67 revealed that PTL inhibited Ki67 expression at 3DIV compared to without PTL. These results confirm that PTL inhibits peripheral nerve degenerative processes. PTL may be a good applicant to inhibit peripheral nerve degeneration. Our study examined the effect of Parthenolide in preventing degeneration of peripheral nerves by inhibiting the breakdown of peripheral axons and myelin, also inhibiting Schwann cell trans-dedifferentiation and proliferation.
Collapse
Affiliation(s)
- Jung Min Kim
- Department of Otorhinolaryngology, Head and Neck Surgery, Kyung Hee University School of Medicine, Kyung Hee University Medical Center, Seoul, 02447, Korea
| | - Jae Sun Choi
- Clinical Research Institute, Kyung Hee Medical Center, Seou, 02447, Korea
| | - Junyang Jung
- Department of Anatomy and Neurobiology, College of Medicines, Kyung Hee University, Seoul, 02447, Korea
| | - Seung Geun Yeo
- Department of Otorhinolaryngology, Head and Neck Surgery, Kyung Hee University School of Medicine, Kyung Hee University Medical Center, Seoul, 02447, Korea
| | - Sang Hoon Kim
- Department of Otorhinolaryngology, Head and Neck Surgery, Kyung Hee University School of Medicine, Kyung Hee University Medical Center, Seoul, 02447, Korea.
- Department of Otohinolaryngology - H & N Surgery, School of Medicine, KyungHee University, #1 Hoegi-Dong, Dongdaemun-Gu, Seoul, 130-702, Korea.
| |
Collapse
|
4
|
Sheth S, Patel A, Foreman M, Mumtaz M, Reddy A, Sharaf R, Sheth S, Lucke-Wold B. The protective role of GLP-1 in neuro-ophthalmology. EXPLORATION OF DRUG SCIENCE 2023; 1:221-238. [PMID: 37711214 PMCID: PMC10501042 DOI: 10.37349/eds.2023.00015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/22/2023] [Indexed: 09/16/2023]
Abstract
Despite recent advancements in the field of neuro-ophthalmology, the rising rates of neurological and ophthalmological conditions, mismatches between supply and demand of clinicians, and an aging population underscore the urgent need to explore new therapeutic approaches within the field. Glucagon-like peptide 1 receptor agonists (GLP-1RAs), traditionally used in the treatment of type 2 diabetes, are becoming increasingly appreciated for their diverse applications. Recently, GLP-1RAs have been approved for the treatment of obesity and recognized for their cardioprotective effects. Emerging evidence indicates some GLP-1RAs can cross the blood-brain barrier and may have neuroprotective effects. Therefore, this article aims to review the literature on the neurologic and neuro-ophthalmic role of glucagon-like peptide 1 (GLP-1). This article describes GLP-1 peptide characteristics and the mechanisms mediating its known role in increasing insulin, decreasing glucagon, delaying gastric emptying, and promoting satiety. This article identifies the sources and targets of GLP-1 in the brain and review the mechanisms which mediate its neuroprotective effects, as well as implications for Alzheimer's disease (AD) and Parkinson's disease (PD). Furthermore, the preclinical works which unravel the effects of GLP-1 in ocular dynamics and the preclinical literature regarding GLP-1RA use in the management of several neuro-ophthalmic conditions, including diabetic retinopathy (DR), glaucoma, and idiopathic intracranial hypertension (IIH) are discussed.
Collapse
Affiliation(s)
- Sohum Sheth
- College of Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Aashay Patel
- College of Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Marco Foreman
- College of Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Mohammed Mumtaz
- College of Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Akshay Reddy
- College of Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Ramy Sharaf
- College of Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Siddharth Sheth
- College of Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA
| |
Collapse
|
5
|
Yang X, Huang Z, Xu M, Chen Y, Cao M, Yi G, Fu M. Autophagy in the retinal neurovascular unit: New perspectives into diabetic retinopathy. J Diabetes 2023; 15:382-396. [PMID: 36864557 PMCID: PMC10172025 DOI: 10.1111/1753-0407.13373] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/08/2023] [Accepted: 02/18/2023] [Indexed: 03/04/2023] Open
Abstract
Diabetic retinopathy (DR) is one of the most prevalent retinal disorders worldwide, and it is a major cause of vision impairment in individuals of productive age. Research has demonstrated the significance of autophagy in DR, which is a critical intracellular homeostasis mechanism required for the destruction and recovery of cytoplasmic components. Autophagy maintains the physiological function of senescent and impaired organelles under stress situations, thereby regulating cell fate via various signals. As the retina's functional and fundamental unit, the retinal neurovascular unit (NVU) is critical in keeping the retinal environment's stability and supporting the needs of retinal metabolism. However, autophagy is essential for the normal NVU structure and function. We discuss the strong association between DR and autophagy in this review, as well as the many kinds of autophagy and its crucial physiological activities in the retina. By evaluating the pathological changes of retinal NVU in DR and the latest advancements in the molecular mechanisms of autophagy that may be involved in the pathophysiology of DR in NVU, we seek to propose new ideas and methods for the prevention and treatment of DR.
Collapse
Affiliation(s)
- Xiongyi Yang
- Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
- The Second Clinical School, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Zexin Huang
- Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
- The Second Clinical School, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Mei Xu
- The Second People's Hospital of Jingmen, Jingmen, Hubei, People's Republic of China
| | - Yanxia Chen
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Mingzhe Cao
- Department of Ophthalmology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, P. R. China
| | - Guoguo Yi
- Department of Ophthalmology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P. R. China
| | - Min Fu
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| |
Collapse
|
6
|
Shih CC, Lee CY, Wong FF, Lin CH. Protective Effects of One 2,4-Dihydro-3H-Pyrazol-3-one Derivative against Posterior Capsular Opacification by Regulation of TGF-β2/SMADs and Non-SMAD Signaling, Collagen I, and Fibronectin Proteins. Curr Issues Mol Biol 2022; 44:5048-5066. [PMID: 36286058 PMCID: PMC9600192 DOI: 10.3390/cimb44100343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 11/16/2022] Open
Abstract
Many elderly individuals frequently experience cataracts that interfere with vision. After cataract surgery, the left lens epithelial cell (LEC) exhibited fibrosis and posterior capsule opacification (PCO). Sometimes, there is a need for a second surgery; nevertheless, people try other methods, such as a good pharmacological agent, to treat PCO to reduce transforming growth factor-β2 (TGF-β2) amounts to avoid secondary surgery. The aim of the present study was to explore the potential anti-PCO activity of five 2,4-dihydro-3H-pyrazol-3-one (DHPO) derivatives in a TGF-β2-induced fibrogenesis SRA01/04 cell model. The 2-phenyl-5-propyl-DHPO (TSE; no. 2: TSE-2) compound showed the best activity of reduced expression levels of TGF-β2 among five derivatives and therefore was chosen to evaluate the anti-PCO activity and molecular mechanisms on the Sma and mad protein (SMAD) signaling pathway (including TGF-β2, SMADs, and the inhibition of nuclear translocation of SMADs), non-SMAD pathway proteins, including p-extracellular, regulated protein kinases (ERK) 1/2, or p-c-Jun N-terminal kinase (JUN) by Western blotting, PCR, or confocal immunofluorescence analyses. Following treatment with 10 μg/mL of the five compounds, the cells displayed great viability by 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTT) assay. In this study, the result of lactate dehydrogenase (LDH) activity measurement did not affect the cytotoxicity of the five compounds. In TGF-β2-induced fibrogenesis in SRA01/04 cells, treatment with the TSE compound decreased the TGF-β2/SMAD signaling genes, including reduced mRNA or expression levels of TGF-β2, SMAD3, and SMAD4, leading to inhibition of TGF-β2-induced fibrogenesis. Our confocal immunofluorescence analyses demonstrated that TSE treatment displays a suppressive effect on SMAD2/3 or SMAD4 translocation to the nucleus. Furthermore, TSE treatment exhibits a reduction in the non-SMAD target gene expression levels of p- c-Jun N-terminal kinase (JUN), p- extracellular, regulated protein kinases (ERK)1/2, p- p38 mitogen-activated protein kinase (p38), p-phosphatidylinositol 3-kinase (PI3K), p-mammalian target of rapamycin complex (mTORC), p-Akt (Ser473), and p-Akt (Thr308). The overall effect of TSE is to reduce the expression levels of collagen I and fibrinogen (FN), thus contributing to antifibrotic effects in cell models mimicking PCO. Our findings reveal the benefits of TSE by regulating TGF-β/SMAD signaling and non-SMAD signaling-related gene proteins to display antifibrotic activity in cells for the possibility of preventing PCO after cataract surgery.
Collapse
Affiliation(s)
- Chun-Ching Shih
- Department of Nursing, College of Nursing, Central Taiwan University of Science and Technology, No.666 Buzih Road, Beitun District, Taichung City 40601, Taiwan
- Correspondence:
| | - Chia-Yi Lee
- Institute of Medicine, Chung Shan Medical University, Taichung City 40201, Taiwan
- Nobel Eye Institute, Taipei 100008, Taiwan
- Department of Ophthalmology, Jen-Ai Hospital Dali Branch, Taichung City 412224, Taiwan
| | - Fung-Fuh Wong
- School of Pharmacy, China Medical University, No. 91 Hsueh-Shih Road, Taichung City 40402, Taiwan
| | - Cheng-Hsiu Lin
- Department of Internal Medicine, Fengyuan Hospital, Ministry of Health and Welfare, Fengyuan District, Taichung City 42055, Taiwan
| |
Collapse
|
7
|
Reich N, Hölscher C. The neuroprotective effects of glucagon-like peptide 1 in Alzheimer's and Parkinson's disease: An in-depth review. Front Neurosci 2022; 16:970925. [PMID: 36117625 PMCID: PMC9475012 DOI: 10.3389/fnins.2022.970925] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/08/2022] [Indexed: 12/16/2022] Open
Abstract
Currently, there is no disease-modifying treatment available for Alzheimer's and Parkinson's disease (AD and PD) and that includes the highly controversial approval of the Aβ-targeting antibody aducanumab for the treatment of AD. Hence, there is still an unmet need for a neuroprotective drug treatment in both AD and PD. Type 2 diabetes is a risk factor for both AD and PD. Glucagon-like peptide 1 (GLP-1) is a peptide hormone and growth factor that has shown neuroprotective effects in preclinical studies, and the success of GLP-1 mimetics in phase II clinical trials in AD and PD has raised new hope. GLP-1 mimetics are currently on the market as treatments for type 2 diabetes. GLP-1 analogs are safe, well tolerated, resistant to desensitization and well characterized in the clinic. Herein, we review the existing evidence and illustrate the neuroprotective pathways that are induced following GLP-1R activation in neurons, microglia and astrocytes. The latter include synaptic protection, improvements in cognition, learning and motor function, amyloid pathology-ameliorating properties (Aβ, Tau, and α-synuclein), the suppression of Ca2+ deregulation and ER stress, potent anti-inflammatory effects, the blockage of oxidative stress, mitochondrial dysfunction and apoptosis pathways, enhancements in the neuronal insulin sensitivity and energy metabolism, functional improvements in autophagy and mitophagy, elevated BDNF and glial cell line-derived neurotrophic factor (GDNF) synthesis as well as neurogenesis. The many beneficial features of GLP-1R and GLP-1/GIPR dual agonists encourage the development of novel drug treatments for AD and PD.
Collapse
Affiliation(s)
- Niklas Reich
- Biomedical and Life Sciences Division, Faculty of Health and Medicine, Lancaster University, Lancaster, United Kingdom
| | - Christian Hölscher
- Neurology Department, Second Associated Hospital, Shanxi Medical University, Taiyuan, China
- Henan University of Chinese Medicine, Academy of Chinese Medical Science, Zhengzhou, China
| |
Collapse
|
8
|
Zhang Y, Wang S, Chen X, Wang Z, Wang X, Zhou Q, Fang W, Zheng C. Liraglutide prevents high glucose induced HUVECs dysfunction via inhibition of PINK1/Parkin-dependent mitophagy. Mol Cell Endocrinol 2022; 545:111560. [PMID: 35032624 DOI: 10.1016/j.mce.2022.111560] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/25/2021] [Accepted: 01/11/2022] [Indexed: 01/11/2023]
Abstract
Functional loss of endothelial cells will lead to development and progression of atherosclerosis in diabetic patients. However, dysfunction of endothelial cells in diabetes has yet to be fully understood. We aimed to characterize the potential effects of liraglutide, a glucagon-like peptide-1 (GLP-1) receptor agonist, on preventing high glucose-induced endothelial dysfunction and excessive mitophagic response. Pretreatment with liraglutide prevented downregulation of eNOS phosphorylation and NO secretion, and reduced apoptosis and oxidative stress of the human umbilical vein endothelial cells (HUVECs) exposed to high glucose. We further demonstrated that liraglutide likely mediated such protective effects by reducing PINK1/Parkin mediated mitophagy. Liraglutide markedly decreased high glucose-induced mitochondrial ROS, lessened PINK1 expression and mitochondrial accumulation of Parkin, but recovered SIRT1 expression. Seahorse analysis revealed that liraglutide mitigated high glucose-induced reduction of basal and maximum respiration rates as well as spare respiration capacity. Inhibition of Parkin by RNA silencing not only resulted in increased mitochondrial and cytosolic ROS and reduced mitochondrial mass and mitochondrial membrane potential, but also led to increased apoptotic responses in high glucose treated HUVECs which were not preventable by liraglutide. Together, our study reveals that liraglutide acts upstream of the PINK1/Parkin pathway to effectively counteract high glucose induced cell dysfunction by suppression of the PINK1/Parkin-dependent mitophagy. Therefore, its use as an adjunct therapy for type 2 diabetes mellitus is warranted to reduce the risk of atherosclerosis. Further research is required to examine the exact molecules, including SIRT1, upstream of the PINK1/parkin pathway that liraglutide targets to maintain the mitochondrial homeostasis.
Collapse
Affiliation(s)
- Yikai Zhang
- Department of Endocrinology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Shengyao Wang
- Department of Endocrinology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Xia Chen
- Department of Endocrinology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Zhe Wang
- Department of Endocrinology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Xinyi Wang
- Department of Endocrinology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Qiao Zhou
- Department of Endocrinology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Weihuan Fang
- Institute of Preventive Veterinary Medicine & Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Chao Zheng
- Department of Endocrinology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Department of Endocrinology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.
| |
Collapse
|
9
|
Arendt Nielsen T, Sega R, Uggerhøj Andersen C, Vorum H, Mohr Drewes A, Jakobsen PE, Brock B, Brock C. Liraglutide Treatment Does Not Induce Changes in the Peripapillary Retinal Nerve Fiber Layer Thickness in Patients with Diabetic Retinopathy. J Ocul Pharmacol Ther 2021; 38:114-121. [PMID: 34918951 DOI: 10.1089/jop.2021.0055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Purpose: Liraglutide treatment has shown promising anti-inflammatory and nerve regenerative results in preclinical and clinical trials. We sought to assess if liraglutide treatment would induce nerve regeneration through its anti-inflammatory and neurotrophic mechanisms by increasing peripapillary retinal nerve fiber layer (RNFL) thickness in individuals with long-term type 1 diabetes. Methods: Secondary analyses were performed on a prospective, double-blinded, randomized, placebo-controlled trial on adults with type 1 diabetes, distal symmetric polyneuropathy (DSPN), and confirmed diabetic retinopathy, who were randomized 1:1 to either 26 weeks placebo or liraglutide treatment. The primary endpoint was a change in peripapillary RNFL thickness between treatments, assessed by optical coherence tomography. Results: Thirty-seven participants were included in the secondary analysis. No differences in mean peripapillary RNFL thickness (overall ΔMean RNFL thickness; liraglutide -1 (±8) μm (-1%) vs. placebo -1 (±5) μm (-1%), P = 0.78, n = 37) or any of the quadrants. Peripapillary RNFL thicknesses were shown between treatments in either nonproliferative (ΔMean RNFL thickness; liraglutide -1 (±5) μm (-1%) vs. placebo 0 (±4) μm (0%), P = 0.80, N = 26) or proliferative diabetic retinopathy subgroup (ΔMean RNFL thickness; liraglutide -2 (±14) μm (-3%) vs. placebo -1 (±6) μm (-2%), P = 0.88, N = 11). Conclusions: In this study, 26 weeks of liraglutide treatment did not induce measurable changes in the assessed optic nerve thickness. Thus, this methodology does not support the induction of substantial nerve regeneration in this cohort with established retinopathy and DSPN. The trial was approved by the Danish Health and Medicines Authority. Informed consent was obtained from all participants. TODINELI study: EUDRA CT: 2013-004375-12, Ethics Ref: N-20130077 Clinical trial registration number: clinicaltrials.gov NCT02138045.
Collapse
Affiliation(s)
- Thomas Arendt Nielsen
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.,Department of Ophthalmology, Aalborg University Hospital, Aalborg University, Aalborg, Denmark
| | - Rok Sega
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.,Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital & Clinical Institute, Aalborg University, Aalborg, Denmark
| | - Carl Uggerhøj Andersen
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.,Department of Ophthalmology, Aalborg University Hospital, Aalborg University, Aalborg, Denmark
| | - Henrik Vorum
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.,Department of Ophthalmology, Aalborg University Hospital, Aalborg University, Aalborg, Denmark
| | - Asbjørn Mohr Drewes
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.,Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital & Clinical Institute, Aalborg University, Aalborg, Denmark.,Steno Diabetes Center North, Aalborg, Denmark
| | - Poul Erik Jakobsen
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.,Steno Diabetes Center North, Aalborg, Denmark.,Department of Endocrinology, Aalborg University Hospital, Aalborg, Denmark
| | - Birgitte Brock
- Steno Diabetes Center Copenhagen, Gentofte, Denmark.,Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Christina Brock
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.,Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital & Clinical Institute, Aalborg University, Aalborg, Denmark.,Steno Diabetes Center North, Aalborg, Denmark
| |
Collapse
|
10
|
Ye MJ, Meng N. Resveratrol acts via the mitogen-activated protein kinase (MAPK) pathway to protect retinal ganglion cells from apoptosis induced by hydrogen peroxide. Bioengineered 2021; 12:4878-4886. [PMID: 34374636 PMCID: PMC8806786 DOI: 10.1080/21655979.2021.1954742] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The current study investigated the ability of resveratrol to protect RGC-5 retinal ganglion cells in culture against H2O2-induced apoptosis and the underlying mechanism of protection. RGC-5 cells were pre-exposed to resveratrol (5, 10, or 20 μM), followed by 200 μM H2O2. Cell viability and apoptosis were detected to assess the cell growth, and expression levels of apoptosis-related and MAPK cascade-associated proteins were determined using western blotting. Levels of reactive oxygen species and mitochondrial membrane potential were also tested, as well as the activities of superoxide dismutase (SOD), catalase (CAT), and glutathione S-transferase (GSH). At a concentration-dependent way, resveratrol reversed H2O2-induced increases in expressions of cleaved caspase-3 and cleaved caspase-9, production of ROS, loss of mitochondrial membrane potential and the expressions of p-p38, p-ERK, and p-JNK. It also promoted the activities of SOD, CAT, and GSH. Furthermore, the agonists of p38, ERK, and JNK partially weakened the protective effects of resveratrol against H2O2-induced apoptosis in RGC-5 cells. Thus, resveratrol can protect retinal ganglion cells against H2O2-induced apoptosis by suppressing MAPK cascades. The drug therefore shows potential for preventing glaucoma.
Collapse
Affiliation(s)
- Ming-Jing Ye
- Department of Pharmacy, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Ni Meng
- Department of Pharmacy, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| |
Collapse
|
11
|
Gao Q, Guo X, Cao Y, Jia X, Xu S, Lu C, Zhu H. Melatonin Protects HT22 Hippocampal Cells from H 2O 2-induced Injury by Increasing Beclin1 and Atg Protein Levels to Activate Autophagy. Curr Pharm Des 2021; 27:446-454. [PMID: 32838711 DOI: 10.2174/1381612826666200824105835] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/30/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND The aging of hippocampal neurons leads to a substantial decline in memory formation, storage and processing. The neuroprotective effect of melatonin has been confirmed, however, its protective mechanism remains unclear. OBJECTIVE In this study, mouse hippocampus-derived neuronal HT22 cells were used to investigate whether melatonin protects the hippocampus from hydrogen peroxide (H2O2)-induced injury by regulating autophagy. METHODS Rapamycin (an activator of autophagy) and 3-methyladenine (3MA, an inhibitor of autophagy) were used to induce or inhibit autophagy, respectively. HT22 cells were treated with 200 μM H2O2 in the presence or absence of 50 μM melatonin. Cell counting kit 8 (CCK-8), β-galactosidase and Hoechst staining were used to measure the viability, aging and apoptosis of cells, respectively. Western blot analysis was used to detect the levels of autophagy-related proteins. RESULTS The activation of autophagy by rapamycin alleviated H2O2-induced oxidative injury, as evidenced by morphological changes and decreased viability, while the inhibition of autophagy by 3MA exacerbated H2O2- induced injury. The inhibitory effect of melatonin on H2O2-induced injury was similar to that of rapamycin. Melatonin also alleviated H2O2-induced aging and apoptosis. Melatonin activated autophagy in the presence or absence of H2O2, as evidenced by an increased Lc3b 14/16 kd ratio and a decreased P62 level. In addition, H2O2 decreased the levels of Beclin1 and Atg5/12/16, which were reversed by rapamycin or melatonin. The effects of melatonin on H2O2-induced injury, autophagy and protein expressions were effectively reversed by 3MA. CONCLUSION In conclusion, these results demonstrate that melatonin protects HT22 hippocampal neurons from H2O2-induced injury by increasing the levels of the Beclin1 and Atg proteins to activate autophagy.
Collapse
Affiliation(s)
- Qiang Gao
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Xiaocheng Guo
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Yang Cao
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Xiaotong Jia
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Shanshan Xu
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Chunmei Lu
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Hui Zhu
- Department of Physiology, Harbin Medical University, Harbin, China
| |
Collapse
|
12
|
Gong Q, Wang H, Yu P, Qian T, Xu X. Protective or Harmful: The Dual Roles of Autophagy in Diabetic Retinopathy. Front Med (Lausanne) 2021; 8:644121. [PMID: 33842506 PMCID: PMC8026897 DOI: 10.3389/fmed.2021.644121] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 02/26/2021] [Indexed: 12/19/2022] Open
Abstract
Autophagy is a self-degradative pathway involving intracellular substance degradation and recycling. Recently, this process has attracted a great deal of attention for its fundamental effect on physiological processes in cells, tissues, and the maintenance of organismal homeostasis. Dysregulation of autophagy occurs in some diseases, including immune disease, cancer, and neurodegenerative conditions. Diabetic retinopathy (DR), as a serious microvascular complication of diabetes, is the main cause of visual loss in working-age adults worldwide. The pathogenic mechanisms of DR are thought to be associated with accumulation of oxidative stress, retinal cell apoptosis, inflammatory response, endoplasmic reticulum (ER) stress, and nutrient starvation. These factors are closely related to the regulation of autophagy under pathological conditions. Increasing evidence has demonstrated the potential role of autophagy in the progression of DR through different pathways. However, to date this role is not understood, and whether the altered level of autophagy flux protects DR, or instead aggravates the progression, needs to be explored. In this review, we explore the alterations and functions of autophagy in different retinal cells and tissues under DR conditions, and explain the mechanisms involved in DR progression. We aim to provide a basis on which DR associated stress-modulated autophagy may be understood, and to suggest novel targets for future therapeutic intervention in DR.
Collapse
Affiliation(s)
- Qiaoyun Gong
- Shanghai Key Laboratory of Ocular Fundus Diseases, Department of Ophthalmology, National Clinical Research Center for Eye Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai General Hospital, Shanghai, China
| | - Haiyan Wang
- Shanghai Key Laboratory of Ocular Fundus Diseases, Department of Ophthalmology, National Clinical Research Center for Eye Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai General Hospital, Shanghai, China
| | - Ping Yu
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianwei Qian
- Shanghai Key Laboratory of Ocular Fundus Diseases, Department of Ophthalmology, National Clinical Research Center for Eye Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai General Hospital, Shanghai, China
| | - Xun Xu
- Shanghai Key Laboratory of Ocular Fundus Diseases, Department of Ophthalmology, National Clinical Research Center for Eye Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai General Hospital, Shanghai, China
| |
Collapse
|
13
|
Yang X, Wu S, Feng Z, Yi G, Zheng Y, Xia Z. Combination therapy with semaglutide and rosiglitazone as a synergistic treatment for diabetic retinopathy in rodent animals. Life Sci 2021; 269:119013. [PMID: 33417950 DOI: 10.1016/j.lfs.2020.119013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/22/2020] [Accepted: 12/27/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To investigate the protective efficacies and potent mechanisms of combination therapy with semaglutide and rosiglitazone (RSG) on the high-glucose incubated human ARPE-19 cells and diabetic retinopathy (DR) model rats. MAIN METHODS The CCK-8 methods were used to evaluate the protective effects of semaglutide and RSG alone or combination on the cell viability of high-glucose treated ARPE-19 cells. After the DR rat model was established, the effects of combined treatment on general indexes, retinal morphological changes, retinal Müller cells as well as PI3K/Akt/MTOR related factors of DR model rats were investigated. RESULTS The CCK-8 assay showed obviously enhanced protective efficacies of combination therapy with semaglutide and RSG on the ARPE-19 with oxidative stress induced by high-glucose with combination index all below 1.5 demonstrating obvious synergistic effects. Combined incubation could also effectively decrease the expression of inflammatory factors, including TNF-α, IL-1β, IL-6, and the increase of ROS content in ARPE cell culture supernatant induced by high-glucose. Combined use of the antioxidant, PI3K/Akt and mTOR inhibitors, we further demonstrated that combined incubation of semaglutide and RSG could effectively by reduce high glucose-induced inflammatory injury inhibiting ROS/PI3K/Akt/mTOR signaling. Furthermore, chronic combination treatment effectively improved the histopathological characteristics and down-regulated the GFAP expression in Müller cells as well as PI3K/Akt/MTOR signaling pathway-related factors in retina which was better than any monomer treatment group. CONCLUSIONS Combined semaglutide with RSG exhibited synergistically protective efficacies on retinal cells by decreasing the GFAP expression, inhibiting oxidative stress and PI3K/Akt/MTOR signaling-transduction in DR model rats.
Collapse
Affiliation(s)
- Xiaoxi Yang
- Department of Ophthalmology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 51000, PR China
| | - Shuduan Wu
- Department of Ophthalmology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 51000, PR China
| | - Zhizhen Feng
- Department of Ophthalmology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 51000, PR China
| | - Guoguo Yi
- Department of Ophthalmology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 51000, PR China
| | - Yuxing Zheng
- Department of Ophthalmology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 51000, PR China
| | - Zhaoxia Xia
- Department of Ophthalmology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 51000, PR China.
| |
Collapse
|
14
|
Yeung RO, Al Jundi M, Gubbi S, Bompu ME, Sirrs S, Tarnopolsky M, Hannah-Shmouni F. Management of mitochondrial diabetes in the era of novel therapies. J Diabetes Complications 2021; 35:107584. [PMID: 32331977 PMCID: PMC7554068 DOI: 10.1016/j.jdiacomp.2020.107584] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/12/2020] [Accepted: 03/10/2020] [Indexed: 12/19/2022]
Abstract
Mitochondrial disorders refer to the complex group of conditions affecting energy metabolism. A number of mitochondrial disorders can lead to the development of diabetes mellitus, and mitochondrial diabetes is thought to account for up to 3% of all diabetes mellitus cases. Depending on the degree of preservation of beta cell secretory capacity and peripheral muscle insulin sensitivity, the phenotype of mitochondrial diabetes may resemble that of type 1 or type 2 diabetes. Additionally, mitochondrial diabetes may rarely present with diabetic ketoacidosis, and can be distinguished from other forms of monogenic diabetes including maturity onset diabetes of the young by the presence of multi-organ involvement, particularly pre-senile sensorineural hearing loss, maternal transmission, and later-onset diagnosis, typically affecting adults over 35 years. Various guidelines on diabetes care do not address this important subset of cases, and this diagnosis is easily missed. Additionally, there is paucity of data on tailored diabetes therapies for mitochondrial diabetes, particularly in the era of novel therapies including glucagon-like peptide-1 receptor agonist and sodium glucose co-transporter-2 inhibitors. Here, we report three patients with mitochondrial diabetes who responded well to the addition of these novel agents and propose a new treatment algorithm for this condition.
Collapse
Affiliation(s)
- Roseanne O Yeung
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Edmonton, Canada.
| | - Mohammad Al Jundi
- Section on Endocrinology & Genetics (SEGEN), National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Sriram Gubbi
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Maria E Bompu
- 1st Department of Pediatrics, Aghia Sofia Children's Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Sandra Sirrs
- Division of Endocrinology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mark Tarnopolsky
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Fady Hannah-Shmouni
- Section on Endocrinology & Genetics (SEGEN), National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
15
|
Cataldi M, Cignarelli A, Giallauria F, Muscogiuri G, Barrea L, Savastano S, Colao A. Cardiovascular effects of antiobesity drugs: are the new medicines all the same? INTERNATIONAL JOURNAL OF OBESITY SUPPLEMENTS 2020; 10:14-26. [PMID: 32714509 DOI: 10.1038/s41367-020-0015-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Waiting for a definite answer from well-designed randomized prospective clinical trials, the impact of the new antiobesity drugs -liraglutide, bupropion/naltrexone, phentermine/topiramate and lorcaserin- on cardiovascular outcomes remains uncertain. What has been learned from previous experience with older medicines is that antiobesity drugs may influence cardiovascular health not only causing weight reduction but also through direct actions on the cardiovascular system. Therefore, in the present review, we examine what is known, mainly from preclinical investigations, about the cardiovascular pharmacology of the new antiobesity medicines with the aim of highlighting potential mechanistic differences. We will show that the two active substances of the bupropion/naltrexone combination both exert beneficial and unwanted cardiovascular effects. Indeed, bupropion exerts anti-inflammatory effects but at the same time it does increase heart rate and blood pressure by potentiating catecholaminergic neurotransmission, whereas naltrexone reduces TLR4-dependent inflammation and has potential protective effects in stroke but also impairs cardiac adaption to ischemia and the beneficial opioid protective effects mediated in the endothelium. On the contrary, with the only exception of a small increase in heat rate, liraglutide only exerts favorable cardiovascular effects by protecting myocardium and brain from ischemic damage, improving heart contractility, lowering blood pressure and reducing atherogenesis. As far as the phentermine/topiramate combination is concerned, no direct cardiovascular beneficial effect is expected for phentermine (as this drug is an amphetamine derivative), whereas topiramate may exert cardioprotective and neuroprotective effects in ischemia and anti-inflammatory and antiatherogenic actions. Finally, lorcaserin, a selective 5HT2C receptor agonist, does not seem to exert significant direct effects on the cardiovascular system though at very high concentrations this drug may also interact with other serotonin receptor subtypes and exert unwanted cardiovascular effects. In conclusion, the final effect of the new antiobesity drugs on cardiovascular outcomes will be a balance between possible (but still unproved) beneficial effects of weight loss and "mixed" weight-independent drug-specific effects. Therefore comparative studies will be required to establish which one of the new medicines is more appropriate in patients with specific cardiovascular diseases.
Collapse
Affiliation(s)
- Mauro Cataldi
- Department of Neuroscience, Reproductive Sciences and Dentistry, Division of Pharmacology, Federico II University of Naples, Naples, Italy
| | - Angelo Cignarelli
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Francesco Giallauria
- Department of Translational Medical Sciences, Internal Medicine (Metabolic and Cardiac Rehabilitation Unit), Federico II University of Naples, Naples, Italy
| | - Giovanna Muscogiuri
- Department of Clinical Medicine and Surgery, Unit of Endocrinology, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Luigi Barrea
- Department of Clinical Medicine and Surgery, Unit of Endocrinology, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Silvia Savastano
- Department of Clinical Medicine and Surgery, Unit of Endocrinology, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Annamaria Colao
- Department of Clinical Medicine and Surgery, Unit of Endocrinology, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Naples, Italy
| | | |
Collapse
|
16
|
Liraglutide Protects Against Brain Amyloid-β 1-42 Accumulation in Female Mice with Early Alzheimer's Disease-Like Pathology by Partially Rescuing Oxidative/Nitrosative Stress and Inflammation. Int J Mol Sci 2020; 21:ijms21051746. [PMID: 32143329 PMCID: PMC7084254 DOI: 10.3390/ijms21051746] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/23/2020] [Accepted: 02/28/2020] [Indexed: 12/14/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia worldwide, being characterized by the deposition of senile plaques, neurofibrillary tangles (enriched in the amyloid beta (Aβ) peptide and hyperphosphorylated tau (p-tau), respectively) and memory loss. Aging, type 2 diabetes (T2D) and female sex (especially after menopause) are risk factors for AD, but their crosslinking mechanisms remain unclear. Most clinical trials targeting AD neuropathology failed and it remains incurable. However, evidence suggests that effective anti-T2D drugs, such as the GLP-1 mimetic and neuroprotector liraglutide, can be also efficient against AD. Thus, we aimed to study the benefits of a peripheral liraglutide treatment in AD female mice. We used blood and brain cortical lysates from 10-month-old 3xTg-AD female mice, treated for 28 days with liraglutide (0.2 mg/kg, once/day) to evaluate parameters affected in AD (e.g., Aβ and p-tau, motor and cognitive function, glucose metabolism, inflammation and oxidative/nitrosative stress). Despite the limited signs of cognitive changes in mature female mice, liraglutide only reduced their cortical Aβ1–42 levels. Liraglutide partially attenuated brain estradiol and GLP-1 and activated PKA levels, oxidative/nitrosative stress and inflammation in these AD female mice. Our results support the earlier use of liraglutide as a potential preventive/therapeutic agent against the accumulation of the first neuropathological features of AD in females.
Collapse
|
17
|
Bethel MA, Mentz RJ, Merrill P, Buse JB, Chan JC, Goodman SG, Iqbal N, Jakuboniene N, Katona B, Lokhnygina Y, Lopes RD, Maggioni AP, Ohman P, Tankova T, Bakris GL, Hernandez AF, Holman RR. Microvascular and Cardiovascular Outcomes According to Renal Function in Patients Treated With Once-Weekly Exenatide: Insights From the EXSCEL Trial. Diabetes Care 2020; 43:446-452. [PMID: 31757838 PMCID: PMC7411285 DOI: 10.2337/dc19-1065] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 11/01/2019] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To evaluate the impact of once-weekly exenatide (EQW) on microvascular and cardiovascular (CV) outcomes by baseline renal function in the Exenatide Study of Cardiovascular Event Lowering (EXSCEL). RESEARCH DESIGN AND METHODS Least squares mean difference (LSMD) in estimated glomerular filtration rate (eGFR) from baseline between the EQW and placebo groups was calculated for 13,844 participants. Cox regression models were used to estimate effects by group on incident macroalbuminuria, retinopathy, and major adverse CV events (MACE). Interval-censored time-to-event models estimated effects on renal composite 1 (40% eGFR decline, renal replacement, or renal death) and renal composite 2 (composite 1 variables plus macroalbuminuria). RESULTS EQW did not change eGFR significantly (LSMD 0.21 mL/min/1.73 m2 [95% CI -0.27 to 0.70]). Macroalbuminuria occurred in 2.2% of patients in the EQW group and in 2.5% of those in the placebo group (hazard ratio [HR] 0.87 [95% CI 0.70-1.07]). Neither renal composite was reduced with EQW in unadjusted analyses, but renal composite 2 was reduced after adjustment (HR 0.85 [95% CI 0.74-0.98]). Retinopathy rates did not differ by treatment group or in the HbA1c-lowering or prior retinopathy subgroups. CV outcomes in those with eGFR <60 mL/min/1.73 m2 did not differ by group. Those with eGFR ≥60 mL/min/1.73 m2 had nominal risk reductions for MACE, all-cause mortality, and CV death, but interactions by renal function group were significant for only stroke (HR 0.74 [95% CI 0.58-0.93]; P for interaction = 0.035) and CV death (HR 1.08 [95% CI 0.85-1.38]; P for interaction = 0.031). CONCLUSIONS EQW had no impact on unadjusted retinopathy or renal outcomes. CV risk was modestly reduced only in those with eGFR ≥60 mL/min/1.73 m2 in analyses unadjusted for multiplicity.
Collapse
Affiliation(s)
- M Angelyn Bethel
- Diabetes Trials Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Oxford, U.K
| | - Robert J Mentz
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | - Peter Merrill
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | - John B Buse
- Division of Endocrinology, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Juliana C Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, The Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Shaun G Goodman
- St. Michael's Hospital, University of Toronto, Ontario, Canada.,Canadian VIGOUR Centre, University of Alberta, Edmonton, Canada
| | - Nayyar Iqbal
- AstraZeneca Research and Development, Gaithersburg, MD
| | - Neli Jakuboniene
- Department of Endocrinology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Brian Katona
- AstraZeneca Research and Development, Gaithersburg, MD
| | - Yuliya Lokhnygina
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | - Renato D Lopes
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | | | - Peter Ohman
- AstraZeneca Research and Development, Gaithersburg, MD
| | | | - George L Bakris
- Comprehensive Hypertension Center, The University of Chicago Medicine, Chicago, IL
| | - Adrian F Hernandez
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | - Rury R Holman
- Diabetes Trials Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Oxford, U.K.
| |
Collapse
|
18
|
Liraglutide provides neuroprotection by regulating autophagy through the AMPK-FOXO3 signaling pathway in a spinal contusion injury rat model. Neurosci Lett 2020; 720:134747. [DOI: 10.1016/j.neulet.2020.134747] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 11/23/2019] [Accepted: 01/06/2020] [Indexed: 12/25/2022]
|
19
|
Neuroprotective Peptides in Retinal Disease. J Clin Med 2019; 8:jcm8081146. [PMID: 31374938 PMCID: PMC6722704 DOI: 10.3390/jcm8081146] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 07/26/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023] Open
Abstract
In the pathogenesis of many disorders, neuronal death plays a key role. It is now assumed that neurodegeneration is caused by multiple and somewhat converging/overlapping death mechanisms, and that neurons are sensitive to unique death styles. In this respect, major advances in the knowledge of different types, mechanisms, and roles of neurodegeneration are crucial to restore the neuronal functions involved in neuroprotection. Several novel concepts have emerged recently, suggesting that the modulation of the neuropeptide system may provide an entirely new set of pharmacological approaches. Neuropeptides and their receptors are expressed widely in mammalian retinas, where they exert neuromodulatory functions including the processing of visual information. In multiple models of retinal diseases, different peptidergic substances play neuroprotective actions. Herein, we describe the novel advances on the protective roles of neuropeptides in the retina. In particular, we focus on the mechanisms by which peptides affect neuronal death/survival and the vascular lesions commonly associated with retinal neurodegenerative pathologies. The goal is to highlight the therapeutic potential of neuropeptide systems as neuroprotectants in retinal diseases.
Collapse
|
20
|
Ashrafizadeh M, Yaribeygi H, Atkin SL, Sahebkar A. Effects of newly introduced antidiabetic drugs on autophagy. Diabetes Metab Syndr 2019; 13:2445-2449. [PMID: 31405658 DOI: 10.1016/j.dsx.2019.06.028] [Citation(s) in RCA: 39] [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: 05/31/2019] [Accepted: 06/27/2019] [Indexed: 12/13/2022]
Abstract
Diabetes mellitus is a chronic metabolic disorder that has a complex molecular and cellular pathophysiology, resulting in its dynamic progression and that may show differing responses to therapy. The incidence of diabetes mellitus increases with age and requires additive therapeutic agents for its management. SGLT2i and DPP-4 inhibitors and GLP-1 receptor agonists (GLP-1RA) are newly introduced antidiabetic drugs that work through differing mechanisms; DPP-4 inhibitors maintain the endogenous level of GLP1; GLP-1RA result in pharmacological levels of GLP1, whilst SGLT2i act on the proximal tubules of the kidney. They have shown efficacy in the management of diabetes and in contrast to other antidiabetic drugs, do not inherently cause hypoglycemia in therapeutic doses. Autophagy as a highly conserved mechanism to maintain cell survival and homeostasis by degradation of damaged or aged organelles and components, and recognised to be increasingly important in diabetes. In the present review, we discuss the modulatory effects of these newly introduced antidiabetic drugs on the autophagy process.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran.
| | | | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
21
|
Zheng C, Zhou M, Sun J, Xiong H, Peng P, Gu Z, Deng Y. The protective effects of liraglutide on AD-like neurodegeneration induced by oxidative stress in human neuroblastoma SH-SY5Y cells. Chem Biol Interact 2019; 310:108688. [PMID: 31173752 DOI: 10.1016/j.cbi.2019.06.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 05/25/2019] [Accepted: 06/03/2019] [Indexed: 12/11/2022]
Abstract
Glucagon-like peptide 1 (GLP-1) has neuroprotective properties in Alzheimer's disease (AD). In this study, our aim is to explore the neuroprotective effects of liraglutide, a GLP-1 analogue, on AD-like neurodegeneration induced by H2O2 in human neuroblastoma SH-SY5Y cells. Cytotoxicity was determined by MTT assay and lactate dehydrogenase level was monitored by LDH assay. The level of lipid peroxidation and cell apoptosis rate were measured by malondialdehyde (MDA) assay and Annexin V-FITC/propidium iodide (PI) staining. Western blotting was used to assess the expression of Bcl-2, Bax, caspase-3, tau and the Akt/GSK-3β. Liraglutide pre-treatment enhanced cell viability with reduced cytotoxicity, lipid peroxidationand and apoptosis. In addition, pre-treatment of liraglutide displayed that increased the expression of the pro-survival Bcl-2 and reduced pro-apoptotic Bax with ameliorated the hyperphosphorylation of tau and Akt/GSK-3β signaling pathway in H2O2 stressed SH-SY5Y cells. These finding provided evidences that liraglutide protected the H2O2 induced AD-like neurodegeneration through improving Akt/GSK-3β signaling pathway. These results suggest that liraglutide may have potential values for the treatment for AD.
Collapse
Affiliation(s)
- Chen Zheng
- Pathophysiology Department, School of Basic Medical College, Tianjin Medical University, Tianjin, China; Department of Physiology, Zunyi Medical University, Zunyi, China
| | - Mei Zhou
- Pathophysiology Department, School of Basic Medical College, Tianjin Medical University, Tianjin, China
| | - Jie Sun
- Pathophysiology Department, School of Basic Medical College, Tianjin Medical University, Tianjin, China; Department of Pathology, Tianjin People's Hospital, Tianjin, China
| | - Hui Xiong
- Pathophysiology Department, School of Basic Medical College, Tianjin Medical University, Tianjin, China
| | - Peng Peng
- Pathophysiology Department, School of Basic Medical College, Tianjin Medical University, Tianjin, China
| | - Zhongya Gu
- Pathophysiology Department, School of Basic Medical College, Tianjin Medical University, Tianjin, China
| | - Yanqiu Deng
- Pathophysiology Department, School of Basic Medical College, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
22
|
The natural plant flavonoid apigenin is a strong antioxidant that effectively delays peripheral neurodegenerative processes. Anat Sci Int 2019; 94:285-294. [DOI: 10.1007/s12565-019-00486-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 03/21/2019] [Indexed: 12/22/2022]
|
23
|
Zhao W, Wang S, Qin T, Wang W. RETRACTED: Arbutin attenuates hydrogen peroxide-induced oxidative injury through regulation of microRNA-29a in retinal ganglion cells. Biomed Pharmacother 2019; 112:108729. [DOI: 10.1016/j.biopha.2019.108729] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/14/2019] [Accepted: 02/22/2019] [Indexed: 02/02/2023] Open
|
24
|
Yi EH, Xu F, Li P, Guo JQ. Transactive response DNA binding protein of 43/histone deacetylase 6 axis alleviates H 2 O 2 -induced retinal ganglion cells injury through inhibiting apoptosis and autophagy. J Cell Biochem 2018; 120:4312-4320. [PMID: 30320895 DOI: 10.1002/jcb.27717] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 08/29/2018] [Indexed: 12/12/2022]
Abstract
Oxidative damage is believed to contribute to the pathogenesis of diabetic retinopathy (DR). The current study aimed to detect the effects of transactive response DNA binding protein of 43 (TDP-43) on cell damage induced by hydrogen peroxide (H2 O2 ) in retinal ganglion cells (RGCs) and to investigate the molecular mechanisms involved in this process. We observed that TDP-43 was highly expressed in RGC-5 cells induced by H2 O2 , and that repression of TDP-43 obviously ameliorated H2 O2 -induced RGC-5 cell injury. In addition, loss of TDP-43 profoundly mitigated H2 O2 -triggered oxidative stress by decreasing the production of intracellular reactive oxygen species and the activity of oxidative stress indicator malondialdehyde, as well as enhancing the content of antioxidant enzymes superoxide dismutase, glutathione peroxidase and catalase to restore the antioxidant defense system. Moreover, suppression of TDP-43 obviously obstructed H2 O2 -induced apoptosis. Meanwhile, knockdown of TDP-43 attenuated the expression of the proapoptotic proteins Bax and Cytochrome c, elevated the anti-apoptotic protein Bcl-2, and suppressed the activation of caspase 3 in H2 O2 -induced RGC-5 cells. Moreover, elimination of TDP-43 inhibited H2 O2 -triggered autophagy, which appeared as decreased expression of LC3II/I and Beclin-1, along with p62 degradation. Importantly, silencing of TDP-43 diminished the expression of histone deacetylase 6 (HDAC6), and HDAC6 also abolished the inhibitory effect of TDP-43 inhibition on H2 O2 -induced apoptosis and autophagy. Collectively, our findings demonstrated that depletion of TDP-43 may protect RGC-5 cells against oxidative stress-mediated apoptosis and autophagy by suppressing its target HDAC6. Thus, the TDP-43/HDAC6 axis might be a promising strategy for the treatment of DR.
Collapse
Affiliation(s)
- En-Hui Yi
- Department of Ophthalmology, Xi'an Central Hospital, Xi'an, Shaanxi, China
| | - Feng Xu
- Department of Ophthalmology, Xi'an Children's Hospital, Xi'an, Shaanxi, China
| | - Peng Li
- Department of Ophthalmology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jian-Qiang Guo
- Department of Ophthalmology, Xi'an No. 1 Hospital, First Affiliated Hospital of Northwestern University; Shaanxi Institute of Ophthalmology; Shaanxi Key Laboratory of Ophthalmology; Clinical Research Center for Ophthalmology Diseases of Shaanxi Province, Xi'an, Shaanxi, China
| |
Collapse
|
25
|
Zhang D, Lv G. Therapeutic potential of spinal GLP-1 receptor signaling. Peptides 2018; 101:89-94. [PMID: 29329976 DOI: 10.1016/j.peptides.2018.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/24/2017] [Accepted: 01/08/2018] [Indexed: 12/16/2022]
Abstract
GLP-1 signaling pathway has been well studied for its role in regulating glucose homeostasis, as well as its beneficial effects in energy and nutrient metabolism. A number of drugs based on GLP-1 have been used to treat type 2 diabetes mellitus. GLP-1R is expressed in multiple organs and numerous experimental studies have demonstrated that GLP-1 signaling pathway exhibits pro-survival functions in various disorders. In the central nervous system, stimulation of GLP-1R produces neuroprotective effects in specific neurodegenerative disorders, such as Alzheimer's disease and Parkinson's disease. The preproglucagon neurons located in the brainstem can also produce GLP-1. GLP-1 analogs have a long-acting effect and are able to pass the blood-brain barrier, which probably extends the therapeutic efficacy of GLP-1R activation. Neurodegenerative or traumatic conditions can damage the spinal cord and result in motor and sensory dysfunction. Evidence supports that GLP-1R activation in the spinal cord possesses beneficial effects and significant therapeutic potential. Herein, we review studies that have focused on GLP-1 and the spinal cord, and summarize the expression of GLP-1R and the innervation of PPG neurons in the spinal cord, as well as the potential therapeutic benefits of GLP-1R activation.
Collapse
Affiliation(s)
- Dongao Zhang
- Department of Orthopaedics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Gang Lv
- Department of Orthopaedics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
26
|
Pang B, Zhou H, Kuang H. The potential benefits of glucagon-like peptide-1 receptor agonists for diabetic retinopathy. Peptides 2018; 100:123-126. [PMID: 28807775 DOI: 10.1016/j.peptides.2017.08.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/01/2017] [Accepted: 08/08/2017] [Indexed: 01/13/2023]
Abstract
For a long time, diabetic retinopathy (DR) has been one of the most severe complications of diabetes. The early treatment of DR is not clearly recognized. The additional benefit of hypoglycemic agents for DR has become a new research field. Glucagon-like peptide-1 receptor (GLP-1R) has been shown to be widely expressed in tissues including retina. Glucagon-like peptide-1 receptor agonists (GLP-1RA) have been generally used in the treatment of diabetic patients. Studies shows that GLP-1RA could inhibit nerve damage by decrease apoptosis of nerve cells and activation of glial cells. In addition, GLP-1RA plays a protective role for tight junction (TJ) and cells of blood retinal barrier (BRB). It also protects retina from BRB damage. In this review, we discuss the potential protective mechanisms of GLP-1RA for DR beyond the hypoglycemic effects.
Collapse
Affiliation(s)
- Baoying Pang
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Huanran Zhou
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongyu Kuang
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
27
|
Arden C. A role for Glucagon-Like Peptide-1 in the regulation of β-cell autophagy. Peptides 2018; 100:85-93. [PMID: 29412836 DOI: 10.1016/j.peptides.2017.12.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 12/04/2017] [Accepted: 12/04/2017] [Indexed: 12/11/2022]
Abstract
Autophagy is a highly conserved intracellular recycling pathway that serves to recycle damaged organelles/proteins or superfluous nutrients during times of nutritional stress to provide energy to maintain intracellular homeostasis and sustain core metabolic functions. Under these conditions, autophagy functions as a cell survival mechanism but impairment of this pathway can lead to pro-death stimuli. Due to their role in synthesising and secreting insulin, pancreatic β-cells have a high requirement for robust degradation pathways. Recent research suggests that functional autophagy is required to maintain β-cell survival and function in response to high fat diet suggesting a pro-survival role. However, a role for autophagy has also been implicated in the pathogenesis of type 2 diabetes. Thus, the pro-survival vs pro-death role of autophagy in regulating β-cell mass requires discussion. Emerging evidence suggests that Glucagon-Like Peptide-1 (GLP-1) may exert beneficial effects on glucose homeostasis via autophagy-dependent pathways both in pancreatic β-cells and in other cell types. The aim of the current review is to: i) summarise the literature surrounding β-cell autophagy and its pro-death vs pro-survival role in regulating β-cell mass; ii) review the literature describing the impact of GLP-1 on β-cell autophagy and in other cell types; iii) discuss the potential underlying mechanisms.
Collapse
Affiliation(s)
- Catherine Arden
- Institute of Cellular Medicine, The Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK.
| |
Collapse
|
28
|
Protective effects of agonists of growth hormone-releasing hormone (GHRH) in early experimental diabetic retinopathy. Proc Natl Acad Sci U S A 2017; 114:13248-13253. [PMID: 29180438 PMCID: PMC5740669 DOI: 10.1073/pnas.1718592114] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The studies described here are relevant to the cure of diabetic retinopathy, a leading cause of blindness with currently limited therapeutic options. Here we provided evidence showing that agonists of growth hormone-releasing hormone (GHRH) can significantly diminish retinal neurovascular injury characterizing the early stages of diabetic retinopathy through antioxidant and anti-inflammatory effects. The results of the presented studies provide information on the potential therapeutic effects of GHRH agonists and shed light on the role of hypothalamic hormones in retinal physiology and their effect on visual disorders. In addition, our findings suggest protective effects of GHRH analogs in other disease conditions affecting retinal neuronal cells and, possibly, other nonretinal neurons. The potential therapeutic effects of agonistic analogs of growth hormone-releasing hormone (GHRH) and their mechanism of action were investigated in diabetic retinopathy (DR). Streptozotocin-induced diabetic rats (STZ-rats) were treated with 15 μg/kg GHRH agonist, MR-409, or GHRH antagonist, MIA-602. At the end of treatment, morphological and biochemical analyses assessed the effects of these compounds on retinal neurovascular injury induced by hyperglycemia. The expression levels of GHRH and its receptor (GHRH-R) measured by qPCR and Western blotting were significantly down-regulated in retinas of STZ-rats and in human diabetic retinas (postmortem) compared with their respective controls. Treatment of STZ-rats with the GHRH agonist, MR-409, prevented retinal morphological alteration induced by hyperglycemia, particularly preserving survival of retinal ganglion cells. The reverse, using the GHRH antagonist, MIA-602, resulted in worsening of retinal morphology and a significant alteration of the outer retinal layer. Explaining these results, we have found that MR-409 exerted antioxidant and anti-inflammatory effects in retinas of the treated rats, as shown by up-regulation of NRF-2-dependent gene expression and down-regulation of proinflammatory cytokines and adhesion molecules. MR-409 also significantly down-regulated the expression of vascular endothelial growth factor while increasing that of pigment epithelium-derived factor in diabetic retinas. These effects correlated with decreased vascular permeability. In summary, our findings suggest a neurovascular protective effect of GHRH analogs during the early stage of diabetic retinopathy through their antioxidant and anti-inflammatory properties.
Collapse
|
29
|
Panagaki T, Michael M, Hölscher C. Liraglutide restores chronic ER stress, autophagy impairments and apoptotic signalling in SH-SY5Y cells. Sci Rep 2017; 7:16158. [PMID: 29170452 PMCID: PMC5700973 DOI: 10.1038/s41598-017-16488-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 11/13/2017] [Indexed: 12/23/2022] Open
Abstract
Growing evidence suggests that agonists of glucagon-like peptide (GLP-1) receptor exert neuroprotective and neurorestorative effects across a range of experimental models of neuronal degeneration, and, recently, a pilot clinical trial of Liraglutide in Alzheimer’s disease patients showed improvements in cerebral glucose consumption that signifies disease progression. However, the exact underlying mechanism of action remains unclear. Chronic endoplasmic reticulum (ER) stress has recently emerged as a mechanism for neuronal injury, rendering it a potent therapeutic target for acute and chronic neurodegenerative disorders. Here, we investigate the neuroprotective effects of Liraglutide along with the signalling network against prolong ER stress and autophagy impairments induced by the non-competitive inhibitor of sarco/ER Ca2+-ATPase, thapsigargin. We show that Liraglutide modulates the ER stress response and elicits ER proteostasis and autophagy machinery homeostasis in human SH-SY5Y neuroblastoma cell line. These effects correlate with resolution of hyper-activity of the antioxidant Nrf2 factor and restoration of the impaired cell viability and proliferation. Mechanistically, Liraglutide engages Akt and signal transducer and activator of transcription 3 (STAT3) signalling to favour adaptive responses and shift cell fate from apoptosis to survival under chronic stress conditions in SH-SY5Y cells.
Collapse
Affiliation(s)
- Theodora Panagaki
- Biomedical & Life Sciences Division, Lancaster University, Lancaster, LA1 4YG, UK
| | - Maria Michael
- Biomedical & Life Sciences Division, Lancaster University, Lancaster, LA1 4YG, UK
| | - Christian Hölscher
- Biomedical & Life Sciences Division, Lancaster University, Lancaster, LA1 4YG, UK.
| |
Collapse
|