1
|
Gezginci-Oktayoglu S, Sancar S, Karatug-Kacar A, Bolkent S. Glucotoxicity suppresses function of pancreatic beta and duct cells via miR-335-targeted Runx2 and insulin-mediated mechanism. PROTOPLASMA 2025; 262:341-352. [PMID: 39382633 DOI: 10.1007/s00709-024-01997-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 10/01/2024] [Indexed: 10/10/2024]
Abstract
Pancreatic cell dynamics have important contributions to the development of type 2 diabetes and related diseases such as nonalcoholic fatty pancreas disease. The aim of this study was to investigate the effects of prolonged excessive glucose exposure on the functions of pancreatic beta cells and duct cells in single and co-culture conditions. In this study, we focused on the effects of glucotoxicity on insulin secretion which is the main function of beta cells and on progenitor functions of duct cells. Rat primary INS1 beta cells and ARIP duct cells were exposed to glucose (25 mM) for 72 h under single or indirect co-culture conditions. Glucotoxicity stimuli increased insulin secretion and decreased insulin expression in single beta cells while stimulating beta-cell differentiation and adipogenesis in single duct cells. On the other hand, glucotoxicity caused functional loss and increased proliferation and apoptosis in beta cells while increasing proliferation but suppressed beta-cell differentiation and adipogenesis in duct cells under co-culture conditions. The expression level of miR-335, a microRNA known to be upregulated by leptin and target Runx2, was measured. As a result, unlike single-cell culture, glucotoxicity upregulated miR-335, downregulated Runx2, and decreased insulin signaling in beta cells while downregulating miR-335 and upregulating Runx2, and decreased insulin signaling in duct cells under co-culture conditions. When the results of single and co-culture experiments are compared, insulin and miR-335 may be seen as important mediators for setting up the relation between beta and duct cells. Our findings are important for preventing the development of type 2 diabetes and nonalcoholic fatty pancreas disease, even developing new diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Selda Gezginci-Oktayoglu
- Molecular Biology Section, Biology Department, Faculty of Science, Istanbul University, Vezneciler, 34134, Istanbul, Türkiye.
| | - Serap Sancar
- Molecular Biology Section, Biology Department, Faculty of Science, Istanbul University, Vezneciler, 34134, Istanbul, Türkiye
| | - Ayse Karatug-Kacar
- Molecular Biology Section, Biology Department, Faculty of Science, Istanbul University, Vezneciler, 34134, Istanbul, Türkiye
| | - Sehnaz Bolkent
- Molecular Biology Section, Biology Department, Faculty of Science, Istanbul University, Vezneciler, 34134, Istanbul, Türkiye
| |
Collapse
|
2
|
Kim EJ, Lee SH, Kim TH, Lee J, Choi CH, Lee SJ. Insect chitosan derived from Hermetia illucens larvae suppresses adipogenic signaling and promotes the restoration of gut microbiome balance. Int J Biol Macromol 2025; 284:138168. [PMID: 39613084 DOI: 10.1016/j.ijbiomac.2024.138168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/01/2024]
Abstract
Chitosan, the deacetylated form of chitin, is considered a valuable source of compounds in the feed and food industries. However, the impact of Hermetia illucens larvae chitosan (HCS) with specific physicochemical characteristics on obesity mediated by lipid accumulation and microbiome dysbiosis has not been fully elucidated. We purified HCS with a low molecular weight (84 kDa), low crystallinity, and a high deacetylation rate, characterizing it through several analytical techniques, including gel permeation chromatography, FT-IR, 1H NMR, FE-SEM, and XRD analysis. HCS effectively inhibited the differentiation of 3T3-L1 preadipocytes by suppressing the production of reactive oxygen species. The adipogenic signaling of preadipocytes, mediated by the phosphorylation of mTOR and PPARγ, which are essential for the expression of fatty acid synthase, was attenuated by HCS. In mouse models fed high-fat diets, the oral administration of HCS prevented changes in white adipose tissue and liver weight and reduced plasma levels of total cholesterol. Additionally, the analysis of the microbiota using 16S rRNA revealed that HCS improved dysbiosis by modulating the composition and abundance of specific bacterial genera, including F. rodentium, L. gasseri, L. reuteri, and L. murinus. These findings highlight the potential of HCS as a candidate for the treatment of obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Eun-Ju Kim
- Department of Pharmaceutical Engineering, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Seok-Hui Lee
- Major of Human Bio-convergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Tae Hoon Kim
- FoodyWorm, Co. Ltd., 76 Geumwangtekeuno 3-gil, Geumwang-eup, Eumseong-gun, Chungcheongbuk-do 27680, Republic of Korea
| | - Jin Lee
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Chang-Hyung Choi
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| | - Sei-Jung Lee
- Major of Human Bio-convergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
3
|
Li B, Zhang S, Yun X, Liu C, Xiao R, Lu M, Xu X, Lin F. NEDD4's effect on osteoblastogenesis potential of bone mesenchymal stem cells in rats concerned with PI3K/Akt pathway. Differentiation 2025; 141:100830. [PMID: 39674086 DOI: 10.1016/j.diff.2024.100830] [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: 09/18/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024]
Abstract
Neural precursor cell expressed developmentally down-regulated 4 (NEDD4) is an E3 ubiquitin ligase implicated in craniofacial development. Emerging evidence suggests that NEDD4 may down-regulates Akt signaling, a key element of the PI3K/Akt pathway involved in cell differentiation. This study aimed to investigate NEDD4's role in bone mesenchymal stem cells (BMSCs) differentiation and its interaction with the PI3K/Akt pathway. BMSCs were isolated from SD rats, and NEDD4 expression increased during osteogenic differentiation. Silencing NEDD4 with siRNA elevated alkaline phosphatase (ALP), osteocalcin (OCN), Akt, and mTORC1 expression during induction, while subsequent treatment with LY294002 (a broad spectrum PI3K inhibitor) reduced Akt, mTORC1, ALP, and OCN levels. These findings suggest that NEDD4 may inhibit BMSCs differentiation by suppressing the PI3K/Akt pathway during osteogenesis.
Collapse
Affiliation(s)
- Bo Li
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China; Department of Orthodontics, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China
| | - Shuang Zhang
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Xiaoxian Yun
- Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Chengyi Liu
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China; Department of Orthodontics, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China
| | - Rui Xiao
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Mingjie Lu
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China; Department of Orthodontics, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China
| | - Xiaomei Xu
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China; Department of Orthodontics, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China.
| | - Fuwei Lin
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China; Department of Orthodontics, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China.
| |
Collapse
|
4
|
Li P, Alenazi KKK, Dally J, Woods EL, Waddington RJ, Moseley R. Role of oxidative stress in impaired type II diabetic bone repair: scope for antioxidant therapy intervention? FRONTIERS IN DENTAL MEDICINE 2024; 5:1464009. [PMID: 39917650 PMCID: PMC11797775 DOI: 10.3389/fdmed.2024.1464009] [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: 07/12/2024] [Accepted: 10/02/2024] [Indexed: 02/09/2025] Open
Abstract
Impaired bone healing is a significant complication observed in individuals with type 2 diabetes mellitus (T2DM), leading to prolonged recovery, increased risk of complications, impaired quality of life, and increased risk of patient morbidity. Oxidative stress, resulting from an imbalance between the generation of reactive oxygen species (ROS) and cellular/tissue antioxidant defence mechanisms, has been identified as a critical contributor to the pathogenesis of impaired bone healing in T2DM. Antioxidants have shown promise in mitigating oxidative stress and promoting bone repair, particularly non-enzymic antioxidant entities. This comprehensive narrative review aims to explore the underlying mechanisms and intricate relationship between oxidative stress, impaired bone healing and T2DM, with a specific focus on the current preclinical and clinical evidence advocating the potential of antioxidant therapeutic interventions in improving bone healing outcomes in individuals with T2DM. From the ever-emerging evidence available, it is apparent that exogenously supplemented antioxidants, especially non-enzymic antioxidants, can ameliorate the detrimental effects of oxidative stress, inflammation, and impaired cellular function on bone healing processes during uncontrolled hyperglycaemia; and therefore, hold considerable promise as novel efficacious therapeutic entities. However, despite such conclusions, several important gaps in our knowledge remain to be addressed, including studies involving more sophisticated enzymic antioxidant-based delivery systems, further mechanistic studies into how these antioxidants exert their desirable reparative effects; and more extensive clinical trial studies into the optimisation of antioxidant therapy dosing, frequency, duration and their subsequent biodistribution and bioavailability. By enhancing our understanding of such crucial issues, we can fully exploit the oxidative stress-neutralising properties of these antioxidants to develop effective antioxidant interventions to mitigate impaired bone healing and reduce the associated complications in such T2DM patient populations.
Collapse
Affiliation(s)
- Pui Li
- Disease Mechanisms Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| | - Kuraym Khalid Kuraym Alenazi
- Disease Mechanisms Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| | - Jordanna Dally
- Disease Mechanisms Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| | - Emma Louise Woods
- Disease Mechanisms Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| | - Rachel Jane Waddington
- Biomaterials Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| | - Ryan Moseley
- Disease Mechanisms Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
5
|
Yu X, Xu R, Huang X, Chen H, Zhang Z, Wong I, Chen Z, Deng F. Size-Dependent Effect of Titania Nanotubes on Endoplasmic Reticulum Stress to Re-establish Diabetic Macrophages Homeostasis. ACS Biomater Sci Eng 2024; 10:4323-4335. [PMID: 38860558 DOI: 10.1021/acsbiomaterials.4c00549] [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] [Indexed: 06/12/2024]
Abstract
In patients with diabetes, endoplasmic reticulum stress (ERS) is a crucial disrupting factor of macrophage homeostasis surrounding implants, which remains an obstacle to oral implantation success. Notably, the ERS might be modulated by the implant surface morphology. Titania nanotubes (TNTs) may enhance diabetic osseointegration. However, a consensus has not been achieved regarding the tube-size-dependent effect and the underlying mechanism of TNTs on diabetic macrophage ERS. We manufactured TNTs with small (30 nm) and large diameters (100 nm). Next, we assessed how the different titanium surfaces affected diabetic macrophages and regulated ERS and Ca2+ homeostasis. TNTs alleviated the inflammatory response, oxidative stress, and ERS in diabetic macrophages. Furthermore, TNT30 was superior to TNT100. Inhibiting ERS abolished the positive effect of TNT30. Mechanistically, topography-induced extracellular Ca2+ influx might mitigate excessive ERS in macrophages by alleviating ER Ca2+ depletion and IP3R activation. Furthermore, TNT30 attenuated the peri-implant inflammatory response and promoted osseointegration in diabetic rats. TNTs with small nanodiameters attenuated ERS and re-established diabetic macrophage hemostasis by inhibiting IP3R-induced ER Ca2+ depletion.
Collapse
Affiliation(s)
- Xiaoran Yu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, PR China
| | - Ruogu Xu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, PR China
| | - Xiaoqiong Huang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, PR China
| | - Hongcheng Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, PR China
| | - Zhengchuan Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, PR China
| | - Iohong Wong
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, PR China
| | - Zetao Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, PR China
| | - Feilong Deng
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, PR China
| |
Collapse
|
6
|
Zhang J, Cao J, Liu Y, Zhao H. Advances in the Pathogenesis of Steroid-Associated Osteonecrosis of the Femoral Head. Biomolecules 2024; 14:667. [PMID: 38927070 PMCID: PMC11202272 DOI: 10.3390/biom14060667] [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: 04/18/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a refractory orthopedic condition characterized by bone cell ischemia, necrosis, bone trabecular fracture, and clinical symptoms such as pain, femoral head collapse, and joint dysfunction that can lead to disability. The disability rate of ONFH is very high, which imposes a significant economic burden on both families and society. Steroid-associated osteonecrosis of the femoral head (SANFH) is the most common type of ONFH. However, the pathogenesis of SANFH remains unclear, and it is an urgent challenge for orthopedic surgeons to explore it. In this paper, the pathogenesis of SANFH and its related signaling pathways were briefly reviewed to enhance comprehension of the pathogenesis and prevention of SANFH.
Collapse
Affiliation(s)
- Jie Zhang
- The First Clinical College of Medicine, Lanzhou University, Lanzhou 730000, China; (J.Z.); (J.C.); (Y.L.)
| | - Jianze Cao
- The First Clinical College of Medicine, Lanzhou University, Lanzhou 730000, China; (J.Z.); (J.C.); (Y.L.)
| | - Yongfei Liu
- The First Clinical College of Medicine, Lanzhou University, Lanzhou 730000, China; (J.Z.); (J.C.); (Y.L.)
| | - Haiyan Zhao
- Department of Orthopedics, The First Hospital of Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
7
|
Yang S, Zhao M, Lu M, Feng Y, Zhang X, Wang D, Jiang W. Network Pharmacology Analysis, Molecular Docking Integrated Experimental Verification Reveal the Mechanism of Gynostemma pentaphyllum in the Treatment of Type II Diabetes by Regulating the IRS1/PI3K/Akt Signaling Pathway. Curr Issues Mol Biol 2024; 46:5561-5581. [PMID: 38921004 PMCID: PMC11202160 DOI: 10.3390/cimb46060333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/25/2024] [Accepted: 05/28/2024] [Indexed: 06/27/2024] Open
Abstract
Gynostemma pentaphyllum (Thunb.) Makino (GP), a plant with homology of medicine and food, as a traditional Chinese medicine, possesses promising biological activities in the prevention and treatment of type 2 diabetes mellitus (T2DM). However, the material basis and the mechanism of action of GP in the treatment of T2DM have not been fully elucidated. This study aimed to clarify the active components, potential targets and signaling pathways of GP in treating T2DM. The chemical ingredients of GP were collected by combining UPLC-HRMS analysis and literature research. Network pharmacology revealed that GP had 32 components and 326 potential targets in treating T2DM. The results showed that GP affected T2DM by mediating the insulin resistance signaling pathway, PI3K/Akt signaling pathway and FoxO1 signaling pathway, which had a close relationship with T2DM. Molecular docking results showed that STAT3, PIK3CA, AKT1, EGFR, VEGFA and INSR had high affinity with the active compounds of GP. In vitro, GP extracts obviously increased the glucose uptake and glucose consumption in IR-HepG2 cells. GP extracts increased the levels of PI3K, p-AKT, p-GSK3β and p-FoxO1 and decreased the expression of p-IRS1, p-GS, PEPCK and G6Pase, which indicated that GP could promote glycogen synthesis and inhibit gluconeogenesis by regulating the IRS1/PI3K/Akt signaling pathway. The results demonstrated that GP could improve insulin resistance by promoting glucose uptake and glycogen synthesis and inhibiting gluconeogenesis through regulating the IRS1/PI3K/Akt signaling pathway, which might be a potential alternative therapy for T2DM.
Collapse
Affiliation(s)
- Songqin Yang
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China; (S.Y.); (M.Z.); (M.L.); (Y.F.); (X.Z.)
| | - Mao Zhao
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China; (S.Y.); (M.Z.); (M.L.); (Y.F.); (X.Z.)
| | - Mingxing Lu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China; (S.Y.); (M.Z.); (M.L.); (Y.F.); (X.Z.)
| | - Yuhan Feng
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China; (S.Y.); (M.Z.); (M.L.); (Y.F.); (X.Z.)
| | - Xia Zhang
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China; (S.Y.); (M.Z.); (M.L.); (Y.F.); (X.Z.)
| | - Daoping Wang
- Key Laboratory of Natural Products Chemistry, Guizhou Academy of Sciences, Guiyang 550014, China;
| | - Wenwen Jiang
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China; (S.Y.); (M.Z.); (M.L.); (Y.F.); (X.Z.)
| |
Collapse
|
8
|
Shen X, Zhang M, Cai H, Leslie WD, Lix LM, Jiang D, Feng L, Cheng H, Shi X, Gao Y, Yang S. Associations of global biomarkers of oxidative stress with osteoporosis, bone microstructure and bone turnover: Evidence from human and animal studies. Bone 2024; 183:117077. [PMID: 38521234 DOI: 10.1016/j.bone.2024.117077] [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: 09/25/2023] [Revised: 01/31/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
PURPOSE Human evidence on the association between oxidative stress and osteoporosis is inconsistent. Fluorescent Oxidation Products (FlOPs) are global biomarkers of oxidative stress. We examined the associations of FlOPs (excitation/emission wavelengths 320/420 nm for FlOP_320, 360/420 nm for FlOP_360, and 400/475 nm for FlOP_400) with osteoporosis, bone microstructure, and bone turnover markers in humans and rats. METHODS In humans, we conducted a 1:2 age, sex, hospital, and specimen-matched case-control study to test the association between FlOPs and osteoporosis diagnosed from dual-energy X-ray absorptiometry. In eight-week-old male Wistar rats, we administrated D-galactose and 0.9 % saline for 90 days in treatment and control groups (n = 8/group); micro-CT was used to determine bone microstructure. RESULTS In humans, higher levels of FlOP_320 (OR for per 1 SD increase = 1.49, 95 % CI: 1.01-2.20) and FlOP_360 (OR for per 1 SD increase = 1.59, 95 % CI: 1.07-2.37) were associated with increased odds of osteoporosis. FlOP_400 were not associated with osteoporosis. D-galactose treated rats, as compared with control rats, showed higher levels of FlOP_320 and MDA, and lower P1NP levels during 90 days of experiment (all P < 0.05). The D-galactose group had lower trabecular bone volume fraction (0.07 ± 0.03 vs. 0.13 ± 0.05; P = 0.008) and volumetric BMD (225.4 ± 13.8 vs. 279.1 ± 33.2 mg HA/cm3; P = 0.001) than the control group. CONCLUSION In conclusion, higher FlOP_320 levels were associated with increased odds of osteoporosis, impaired bone microstructure and decreased bone formation.
Collapse
Affiliation(s)
- Xue Shen
- Department of Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China; Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Mengmeng Zhang
- FAW General Hospital of Jilin Province, Changchun, Jilin, China
| | - Hanqing Cai
- Department of Endocrinology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - William D Leslie
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lisa M Lix
- Department of Community Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Depeng Jiang
- Department of Community Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lijie Feng
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Haitao Cheng
- FAW General Hospital of Jilin Province, Changchun, Jilin, China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yuzhong Gao
- Department of Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Shuman Yang
- Department of Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China; Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
9
|
Fakhrioliaei A, Tanhaei S, Pakmehr S, Noori Shakir M, Qasim MT, Hariri M, Nouhi Kararoudi A, Valilo M. Potential Role of Nrf2, HER2, and ALDH in Cancer Stem Cells: A Narrative Review. J Membr Biol 2024; 257:3-16. [PMID: 38356054 DOI: 10.1007/s00232-024-00307-2] [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: 12/08/2023] [Accepted: 01/16/2024] [Indexed: 02/16/2024]
Abstract
Cancer is one of the main causes of death among humans, second only to cardiovascular diseases. In recent years, numerous studies have been conducted on the pathophysiology of cancer, and it has been established that this disease is developed by a group of stem cells known as cancer stem cells (CSCs). Thus, cancer is considered a stem cell disease; however, there is no comprehensive consensus about the characteristics of these cells. Several different signaling pathways including Notch, Hedgehog, transforming growth factor-β (TGF-β), and WNT/β-catenin pathways cause the self-renewal of CSCs. CSCs change their metabolic pathways in order to access easy energy. Therefore, one of the key objectives of researchers in cancer treatment is to destroy CSCs. Nuclear factor erythroid 2-related factor 2 (Nrf2) plays an essential role in the protection of CSCs from reactive oxygen species (ROS) and chemotherapeutic agents by regulating antioxidants and detoxification enzymes. Human epidermal growth factor receptor 2 (HER2) is a member of the tyrosine kinase receptor family, which contributes to the protection of cancer cells against treatment and implicated in the invasion, epithelial-mesenchymal transition (EMT), and tumorigenesis. Aldehyde dehydrogenases (ALDHs) are highly active in CSCs and protect the cells against damage caused by active aldehydes through the regulation of aldehyde metabolism. On the other hand, ALDHs promote the formation and maintenance of tumor cells and lead to drug resistance in tumors through the activation of various signaling pathways, such as the ALDH1A1/HIF-1α/VEGF axis and Wnt/β-catenin, as well as changing the intracellular pH value. Given the growing body of information in this field, in the present narrative review, we attempted to shed light on the function of Nrf2, HER2, and ALDH in CSCs.
Collapse
Affiliation(s)
| | | | | | - Maha Noori Shakir
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | - Maytham T Qasim
- Department of Anesthesia, College of Health and Medical Technology, Al-Ayen University, Thi-Qar, Iraq
| | - Maryam Hariri
- Department of Pathobiology, Auburn University, Auburn, AL, 36832, USA
| | - Alireza Nouhi Kararoudi
- Department of Biology, Faculty of Sciences, Rasht Branch, Islamic Azad University, Rasht, Iran
| | - Mohammad Valilo
- Dpartment of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
10
|
Shamsan E, Almezgagi M, Gamah M, Khan N, Qasem A, Chuanchuan L, Haining F. The role of PI3k/AKT signaling pathway in attenuating liver fibrosis: a comprehensive review. Front Med (Lausanne) 2024; 11:1389329. [PMID: 38590313 PMCID: PMC10999701 DOI: 10.3389/fmed.2024.1389329] [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: 02/21/2024] [Accepted: 03/11/2024] [Indexed: 04/10/2024] Open
Abstract
Excessive accumulation of extracellular matrix (ECM) components within the liver leads to a pathological condition known as liver fibrosis. Alcohol abuse, non-alcoholic fatty liver disease (NAFLD), autoimmune issues, and viral hepatitis cause chronic liver injury. Exploring potential therapeutic targets and understanding the molecular mechanisms involved in liver fibrosis are essential for the development of effective interventions. The goal of this comprehensive review is to explain how the PI3K/AKT signaling pathway contributes to the reduction of liver fibrosis. The potential of this pathway as a therapeutic target is investigated through a summary of results from in vivo and in vitro studies. Studies focusing on PI3K/AKT activation have shown a significant decrease in fibrosis markers and a significant improvement in liver function. The review emphasizes how this pathway may prevent ECM synthesis and hepatic stellate cell (HSC) activation, ultimately reducing the fibrotic response. The specific mechanisms and downstream effectors of the PI3K/AKT pathway in liver fibrosis constitute a rapidly developing field of study. In conclusion, the PI3K/AKT signaling pathway plays a significant role in attenuating liver fibrosis. Its complex role in regulating HSC activation and ECM production, demonstrated both in vitro and in vivo, underscores its potential as a effective therapeutic approach for managing liver fibrosis and slowing disease progression. A comprehensive review of this field provides valuable insights into its future developments and implications for clinical applications.
Collapse
Affiliation(s)
- Emad Shamsan
- College of Clinical Medicine, Qinghai University, Xining, China
- College of Medical Science, Taiz University, Taiz, Yemen
| | - Maged Almezgagi
- College of Clinical Medicine, Qinghai University, Xining, China
| | - Mohammed Gamah
- College of Clinical Medicine, Qinghai University, Xining, China
| | - Naveed Khan
- College of Clinical Medicine, Qinghai University, Xining, China
| | | | - Liu Chuanchuan
- College of Clinical Medicine, Qinghai University, Xining, China
- Qinghai University Affiliated Hospital, Xining, China
| | - Fan Haining
- College of Clinical Medicine, Qinghai University, Xining, China
- Qinghai University Affiliated Hospital, Xining, China
| |
Collapse
|
11
|
Sun X, Xu X, Yue X, Wang T, Wang Z, Zhang C, Wang J. Nanozymes With Osteochondral Regenerative Effects: An Overview of Mechanisms and Recent Applications. Adv Healthc Mater 2024; 13:e2301924. [PMID: 37633309 DOI: 10.1002/adhm.202301924] [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: 06/17/2023] [Revised: 08/14/2023] [Indexed: 08/28/2023]
Abstract
With the discovery of the intrinsic enzyme-like activity of metal oxides, nanozymes garner significant attention due to their superior characteristics, such as low cost, high stability, multi-enzyme activity, and facile preparation. Notably, in the field of biomedicine, nanozymes primarily focus on disease detection, antibacterial properties, antitumor effects, and treatment of inflammatory conditions. However, the potential for application in regenerative medicine, which primarily addresses wound healing, nerve defect repair, bone regeneration, and cardiovascular disease treatment, is garnering interest as well. This review introduces nanozymes as an innovative strategy within the realm of bone regenerative medicine. The primary focus of this approach lies in the facilitation of osteochondral regeneration through the modulation of the pathological microenvironment. The catalytic mechanisms of four types of representative nanozymes are first discussed. The pathological microenvironment inhibiting osteochondral regeneration, followed by summarizing the therapy mechanism of nanozymes to osteochondral regeneration barriers is introduced. Further, the therapeutic potential of nanozymes for bone diseases is included. To improve the therapeutic efficiency of nanozymes and facilitate their clinical translation, future potential applications in osteochondral diseases are also discussed and some significant challenges addressed.
Collapse
Affiliation(s)
- Xueheng Sun
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, 200438, China
| | - Xiang Xu
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Rd, Shanghai, 200011, China
| | - Xiaokun Yue
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Rd, Shanghai, 200011, China
| | - Tianchang Wang
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Rd, Shanghai, 200011, China
| | - Zhaofei Wang
- Department of Orthopaedic Surgery, Shanghai ZhongYe Hospital, Genertec Universal Medical Group, Shanghai, 200941, China
| | - Changru Zhang
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Rd, Shanghai, 200011, China
- Institute of Translational Medicine, Shanghai Jiaotong University, No. 800 Dongchuan Road, Shanghai, 200240, China
| | - Jinwu Wang
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, 200438, China
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Rd, Shanghai, 200011, China
| |
Collapse
|
12
|
Sheng N, Xing F, Wang J, Zhang QY, Nie R, Li-Ling J, Duan X, Xie HQ. Recent progress in bone-repair strategies in diabetic conditions. Mater Today Bio 2023; 23:100835. [PMID: 37928253 PMCID: PMC10623372 DOI: 10.1016/j.mtbio.2023.100835] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 10/02/2023] [Accepted: 10/14/2023] [Indexed: 11/07/2023] Open
Abstract
Bone regeneration following trauma, tumor resection, infection, or congenital disease is challenging. Diabetes mellitus (DM) is a metabolic disease characterized by hyperglycemia. It can result in complications affecting multiple systems including the musculoskeletal system. The increased number of diabetes-related fractures poses a great challenge to clinical specialties, particularly orthopedics and dentistry. Various pathological factors underlying DM may directly impair the process of bone regeneration, leading to delayed or even non-union of fractures. This review summarizes the mechanisms by which DM hampers bone regeneration, including immune abnormalities, inflammation, reactive oxygen species (ROS) accumulation, vascular system damage, insulin/insulin-like growth factor (IGF) deficiency, hyperglycemia, and the production of advanced glycation end products (AGEs). Based on published data, it also summarizes bone repair strategies in diabetic conditions, which include immune regulation, inhibition of inflammation, reduction of oxidative stress, promotion of angiogenesis, restoration of stem cell mobilization, and promotion of osteogenic differentiation, in addition to the challenges and future prospects of such approaches.
Collapse
Affiliation(s)
- Ning Sheng
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Fei Xing
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Jie Wang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Qing-Yi Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Rong Nie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Jesse Li-Ling
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
- Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Xin Duan
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Hui-Qi Xie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
- Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China
| |
Collapse
|
13
|
Yun WJ, Zhang L, Yang N, Cui ZG, Jiang HM, Ha MW, Yu DY, Zhao MZ, Zheng HC. FAM64A aggravates proliferation, invasion, lipid droplet formation, and chemoresistance in gastric cancer: A biomarker for aggressiveness and a gene therapy target. Drug Dev Res 2023; 84:1537-1552. [PMID: 37571819 DOI: 10.1002/ddr.22105] [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: 02/21/2023] [Revised: 07/13/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023]
Abstract
FAM64A is a mitogen-induced regulator of the metaphase and anaphase transition. Here, we found that FAM64A messenger RNA (mRNA) and protein expression levels were higher in gastric cancer tissue than in normal mucosa (p < .05). FAM64A methylation was negatively correlated with FAM64A mRNA expression (p < .05). The differentially expressed genes of FAM64A were mainly involved in digestion, potassium transporting or exchanging ATPase, contractile fibers, endopeptidase, and pancreatic secretion (p < .05). The FAM64A-related genes were principally categorized into ubiquitin-mediated proteolysis, cell cycle, chromosome segregation and mitosis, microtubule binding and organization, metabolism of amino acids, cytokine receptors, lipid droplet, central nervous system, and collagen trimer (p < .05). FAM64A protein expression was lower in normal gastric mucosa than intestinal metaplasia, adenoma, and primary cancer (p < .05), negatively correlated with older age, T stage, lymphatic and venous invasion, tumor, node, metastasis stage, and dedifferentiation (p < .05), and associated with a favorable overall survival of gastric cancer patients. FAM64A overexpression promoted proliferation, antiapoptosis, migration, invasion, and epithelial-mesenchymal transition via the EGFR/Akt/mTOR/NF-κB, while the opposite effect was observed for FAM64A knockdown. FAM64A also induced chemoresistance directly or indirectly through lipid droplet formation via ING5. These results suggested that upregulation of FAM64A expression might induce aggressive phenotypes, leading to gastric carcinogenesis and its subsequent progression. Thus, FAM64A could be regarded as a prognosis biomarker and a target for gene therapy.
Collapse
Affiliation(s)
- Wen-Jing Yun
- Department of Oncology and Central Laboratory, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Li Zhang
- Department of Oncology and Central Laboratory, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Ning Yang
- Department of Oncology and Central Laboratory, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Zheng-Guo Cui
- Department of Environmental Health, University of Fukui School of Medical Sciences, Fukui, Japan
| | - Hua-Mao Jiang
- Cancer Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Min-Wen Ha
- Cancer Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Da-Yong Yu
- Department of Cell Biology, Basic Medical College of Chengde Medical University, Chengde, China
| | - Ming-Zhen Zhao
- Department of Oncology and Central Laboratory, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Hua-Chuan Zheng
- Department of Oncology and Central Laboratory, The Affiliated Hospital of Chengde Medical University, Chengde, China
| |
Collapse
|
14
|
Jiang H, Li D, Han Y, Li N, Tao X, Liu J, Zhang Z, Yu Y, Wang L, Yu S, Zhang N, Xiao H, Yang X, Zhang Y, Zhang G, Zhang BT. The role of sclerostin in lipid and glucose metabolism disorders. Biochem Pharmacol 2023; 215:115694. [PMID: 37481136 DOI: 10.1016/j.bcp.2023.115694] [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: 05/05/2023] [Revised: 07/01/2023] [Accepted: 07/11/2023] [Indexed: 07/24/2023]
Abstract
Lipid and glucose metabolism are critical for human activities, and their disorders can cause diabetes and obesity, two prevalent metabolic diseases. Studies suggest that the bone involved in lipid and glucose metabolism is emerging as an endocrine organ that regulates systemic metabolism through bone-derived molecules. Sclerostin, a protein mainly produced by osteocytes, has been therapeutically targeted by antibodies for treating osteoporosis owing to its ability to inhibit bone formation. Moreover, recent evidence indicates that sclerostin plays a role in lipid and glucose metabolism disorders. Although the effects of sclerostin on bone have been extensively examined and reviewed, its effects on systemic metabolism have not yet been well summarized. In this paper, we provide a systemic review of the effects of sclerostin on lipid and glucose metabolism based on in vitro and in vivo evidence, summarize the research progress on sclerostin, and prospect its potential manipulation for obesity and diabetes treatment.
Collapse
Affiliation(s)
- Hewen Jiang
- School of Chinese Medicine, Chinese University of Hong Kong, Hong Kong, China; Guangdong-Hong Kong Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, China
| | - Dijie Li
- Guangdong-Hong Kong Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, China; Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Ying Han
- Guangdong-Hong Kong Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, China; Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Nanxi Li
- Guangdong-Hong Kong Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, China; Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Xiaohui Tao
- Guangdong-Hong Kong Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, China; Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Jin Liu
- Guangdong-Hong Kong Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, China; Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Zongkang Zhang
- School of Chinese Medicine, Chinese University of Hong Kong, Hong Kong, China; Guangdong-Hong Kong Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, China
| | - Yuanyuan Yu
- Guangdong-Hong Kong Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, China; Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Luyao Wang
- Guangdong-Hong Kong Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, China; Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Sifan Yu
- School of Chinese Medicine, Chinese University of Hong Kong, Hong Kong, China; Guangdong-Hong Kong Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, China
| | - Ning Zhang
- School of Chinese Medicine, Chinese University of Hong Kong, Hong Kong, China; Guangdong-Hong Kong Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, China
| | - Huan Xiao
- School of Chinese Medicine, Chinese University of Hong Kong, Hong Kong, China; Guangdong-Hong Kong Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, China
| | - Xin Yang
- School of Chinese Medicine, Chinese University of Hong Kong, Hong Kong, China; Guangdong-Hong Kong Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, China
| | - Yihao Zhang
- School of Chinese Medicine, Chinese University of Hong Kong, Hong Kong, China; Guangdong-Hong Kong Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, China
| | - Ge Zhang
- Guangdong-Hong Kong Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, China; Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| | - Bao-Ting Zhang
- School of Chinese Medicine, Chinese University of Hong Kong, Hong Kong, China; Guangdong-Hong Kong Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, China.
| |
Collapse
|
15
|
Entz L, Falgayrac G, Chauveau C, Pasquier G, Lucas S. The extracellular matrix of human bone marrow adipocytes and glucose concentration differentially alter mineralization quality without impairing osteoblastogenesis. Bone Rep 2022; 17:101622. [PMID: 36187598 PMCID: PMC9519944 DOI: 10.1016/j.bonr.2022.101622] [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: 07/25/2022] [Revised: 09/09/2022] [Accepted: 09/19/2022] [Indexed: 11/18/2022] Open
Abstract
Bone marrow adipocytes (BMAds) accrue in various states of osteoporosis and interfere with bone remodeling through the secretion of various factors. However, involvement of the extracellular matrix (ECM) produced by BMAds in the impairment of bone marrow mesenchymal stromal cell (BM-MSC) osteoblastogenesis has received little attention. In type 2 diabetes (T2D), skeletal fragility is associated with several changes in bone quality that are incompletely understood, and BMAd quantity increases in relationship to poor glycemic control. Considering their altered phenotype in this pathophysiological context, we aimed to determine the contribution of the ECM of mature BMAds to osteoblastogenesis and mineralization quality in the context of chronic hyperglycemia. Human BM-MSCs were differentiated for 21 days in adipogenic medium containing either a normoglycemic (LG, 5.5 mM) or a high glucose concentration (HG, 25 mM). The ECM laid down by BMAds were devitalized through cell removal to examine their impact on the proliferation and differentiation of BM-MSCs toward osteoblastogenesis in LG and HG conditions. Compared to control plates, both adipocyte ECMs promoted cell adhesion and proliferation. As shown by the unmodified RUNX2 and osteocalcin mRNA levels, BM-MSC commitment in osteoblastogenesis was hampered by neither the hyperglycemic condition nor the adipocyte matrices. However, adipocyte ECMs or HG condition altered the mineralization phase with perturbed expression levels of type 1 collagen, MGP and osteopontin. Despite higher ALP activity, mineralization levels per cell were decreased for osteoblasts grown on adipocyte ECMs compared to controls. Raman spectrometry revealed that culturing on adipocyte matrices specifically prevents type-B carbonate substitution and favors collagen crosslinking, in contrast to exposure to HG concentration alone. Moreover, the mineral to organic ratio was disrupted according to the presence of adipocyte ECM and the glucose concentration used for adipocyte or osteoblast culture. HG concentration and adipocyte ECM lead to different defects in mineralization quality, recapitulating contradictory changes reported in T2D osteoporosis. Our study shows that ECMs from BMAds do not impair osteoblastogenesis but alter both the quantity and quality of mineralization partly in a glucose concentration-dependent manner. This finding sheds light on the involvement of BMAds, which should be considered in the compromised bone quality of T2D and osteoporosis patients more generally. Glucose level alters the Extracellular Matrix composition of Bone Marrow adipocytes. Osteoblastogenesis on adipocyte ECMs is unaltered but produced less mineral amount. The quality of the mineral is altered differently by adipocyte ECMs or glucose levels. The presence of BM adipocytes should be valued in damaged osteoporosis bone quality.
Collapse
Key Words
- AGEs, Advanced glycation end-products
- BM-MSC, Bone marrow mesenchymal stromal cell
- BMAd, Bone marrow adipocyte
- ECM, Extracellular matrix
- ECMBMAd HG, Extracellular matrix obtained from BMAds cultured in HG concentration
- ECMBMAd LG, Extracellular matrix obtained from BMAds cultured in LG concentration
- ECMBMAd, Extracellular matrix obtained from BMAds
- Extracellular matrix
- GAG, glycosaminoglycan
- HA, hydroxyapatite
- HG, High glucose
- Hyperglycemia
- LG, Low glucose
- LGM, Low glucose and mannitol
- Marrow adipocytes
- Osteoblast
- Osteoporosis
- Skeletal mesenchymal stromal cells
- T2D, Type 2 diabetes
Collapse
|
16
|
Ye J, Xiao J, Wang J, Ma Y, Zhang Y, Zhang Q, Zhang Z, Yin H. The Interaction Between Intracellular Energy Metabolism and Signaling Pathways During Osteogenesis. Front Mol Biosci 2022; 8:807487. [PMID: 35155568 PMCID: PMC8832142 DOI: 10.3389/fmolb.2021.807487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/20/2021] [Indexed: 11/28/2022] Open
Abstract
Osteoblasts primarily mediate bone formation, maintain bone structure, and regulate bone mineralization, which plays an important role in bone remodeling. In the past decades, the roles of cytokines, signaling proteins, and transcription factors in osteoblasts have been widely studied. However, whether the energy metabolism of cells can be regulated by these factors to affect the differentiation and functioning of osteoblasts has not been explored in depth. In addition, the signaling and energy metabolism pathways are not independent but closely connected. Although energy metabolism is mediated by signaling pathways, some intermediates of energy metabolism can participate in protein post-translational modification. The content of intermediates, such as acetyl coenzyme A (acetyl CoA) and uridine diphosphate N-acetylglucosamine (UDP-N-acetylglucosamine), determines the degree of acetylation and glycosylation in terms of the availability of energy-producing substrates. The utilization of intracellular metabolic resources and cell survival, proliferation, and differentiation are all related to the integration of metabolic and signaling pathways. In this paper, the interaction between the energy metabolism pathway and osteogenic signaling pathway in osteoblasts and bone marrow mesenchymal stem cells (BMSCs) will be discussed.
Collapse
Affiliation(s)
- Jiapeng Ye
- Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Jirimutu Xiao
- Mongolian Medicine College, Inner Mongolia Medical University, Hohhot, China
| | - Jianwei Wang
- Department of Orthopedics and Traumatology, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
- *Correspondence: Jianwei Wang, ; Heng Yin,
| | - Yong Ma
- Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Yafeng Zhang
- Department of Orthopedics and Traumatology, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Qiang Zhang
- Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Zongrui Zhang
- Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Heng Yin
- Department of Orthopedics and Traumatology, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
- *Correspondence: Jianwei Wang, ; Heng Yin,
| |
Collapse
|
17
|
Cifuentes‑Mendiola S, Moreno‑Fierros L, González‑Alva P, García‑Hernández A. Docosahexaenoic acid improves altered mineralization proteins, the decreased quality of hydroxyapatite crystals and suppresses oxidative stress induced by high glucose. Exp Ther Med 2022; 23:235. [PMID: 35222712 PMCID: PMC8815046 DOI: 10.3892/etm.2022.11160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/30/2021] [Indexed: 11/23/2022] Open
Abstract
Patients with type 2 diabetes mellitus (DM2) experience an increased risk of fractures and a variety of bone pathologies, such as osteoporosis. The suggested mechanisms of increased fracture risk in DM2 include chronic hyperglycaemia, which provokes oxidative stress, alters bone matrix, and decreases the quality of hydroxyapatite crystals. Docosahexaenoic acid (DHA), an omega-3 fatty acid, can increase bone formation, reduce bone loss, and it possesses antioxidant/anti-inflammatory properties. The present study aimed to determine the effect of DHA on altered osteoblast mineralisation and increased reactive oxygen species (ROS) induced by high glucose concentrations. A human osteoblast cell line was treated with 5.5 mM glucose (NG) or 24 mM glucose (HG), alone or in combination with 10 or 20 µM DHA. The collagen type 1 (Col1) scaffold, the expression of osteocalcin (OCN) and bone sialoprotein type-II (BSP-II), the alkaline phosphatase (ALP) specific activity, the mineral quality, the production of ROS and the mRNA expression of nuclear factor erythroid 2-related factor-2 (NRF2) were analysed. Osteoblasts cultured in HG and treated with either DHA concentration displayed an improved distribution of the Col1 scaffold, increased OCN and BSP-II expression, increased NRF2 mRNA, decreased ALP activity, carbonate substitution and reduced ROS production compared with osteoblasts cultured in HG alone. DHA counteracts the adverse effects of HG on bone mineral matrix quality and reduces oxidative stress, possibly by increasing the expression of NRF2.
Collapse
Affiliation(s)
- Saúl Cifuentes‑Mendiola
- Laboratory of Dental Research, Section of Osteoimmunology and Oral Immunology, FES Iztacala, National Autonomous University of Mexico, San Sebastián Xhala, Cuautitlán Izcalli 54714, Mexico
| | - Leticia Moreno‑Fierros
- Laboratory of Mucosal Immunity, FES Iztacala, National Autonomous University of Mexico, Los Reyes Iztacala, Tlalnepantla 54090, Mexico
| | - Patricia González‑Alva
- Laboratory of Tissue Bioengineering, Dentistry Faculty, National Autonomous University of Mexico, University City, Mexico City 04510, Mexico
| | - Ana García‑Hernández
- Laboratory of Dental Research, Section of Osteoimmunology and Oral Immunology, FES Iztacala, National Autonomous University of Mexico, San Sebastián Xhala, Cuautitlán Izcalli 54714, Mexico
| |
Collapse
|
18
|
Yan S, Zhang B, Feng J, Wu H, Duan N, Zhu Y, Zhao Y, Shen S, Zhang K, Wu W, Liu N. FGFC1 Selectively Inhibits Erlotinib-Resistant Non-Small Cell Lung Cancer via Elevation of ROS Mediated by the EGFR/PI3K/Akt/mTOR Pathway. Front Pharmacol 2022; 12:764699. [PMID: 35126111 PMCID: PMC8807551 DOI: 10.3389/fphar.2021.764699] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 12/15/2021] [Indexed: 12/27/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the most common malignancies in the world. Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) have been used as a first-line treatment for patients harboring with EGFR mutations in advanced NSCLC. Nevertheless, the drug resistance after continuous and long-term chemotherapies considerably limits its clinical efficacy. Therefore, it is of great importance to develop new chemotherapeutic agents and treatment strategies to conquer the drug resistance. FGFC1 (Fungi fibrinolytic compound 1), a type of bisindole alkaloid from a metabolite of the rare marine fungi Starchbotrys longispora. FG216, has exhibited excellent fibrinolytic and anti-inflammatory activity. However, the potent efficacy of FGFC1 in human cancer therapy requires further study. Herein, we demonstrated that FGFC1 selectively suppressed the growth of NSCLC cells with EGFR mutation. Mechanistically, FGFC1 treatment significantly induced the apoptosis of erlotinib-resistant NSCLC cells H1975 in a dose-dependent manner, which was proved to be mediated by mitochondrial dysfunction and elevated accumulation of intracellular reactive oxygen species (ROS). Scavenging ROS not only alleviated FGFC1-induced apoptosis but also relieved the decrease of phospho-Akt. We further confirmed that FGFC1 significantly decreased the phosphorylation of protein EGFR, phosphatidylinositol 3-kinase (PI3K), protein kinase B (Akt), and mammalian target of rapamycin (mTOR) in H1975 cells. Notably, PI3K inhibitor (LY294002) could promote the accumulation of ROS and the expression levels of apoptosis-related proteins induced by FGFC1. Molecular dynamics simulations indicated that FGFC1 can inhibit EGFR and its downstream PI3K/Akt/mTOR pathway through directly binding to EGFR, which displayed a much higher binding affinity to EGFRT790M/L858R than EGFRWT. Additionally, FGFC1 treatment also inhibited the migration and invasion of H1975 cells. Finally, FGFC1 effectively inhibited tumor growth in the nude mice xenograft model of NSCLC. Taken together, our results indicate that FGFC1 may be a potential candidate for erlotinib-resistant NSCLC therapy.
Collapse
Affiliation(s)
- Shike Yan
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Bing Zhang
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Jingwen Feng
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Haigang Wu
- School of Life Sciences, Henan University, Kaifeng, China
| | - Namin Duan
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Yamin Zhu
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Yueliang Zhao
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Shuang Shen
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Kai Zhang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wenhui Wu
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
- Engineering Research Center of Aquatic-Product Processing & Preservation, Shanghai, China
| | - Ning Liu
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
- Engineering Research Center of Aquatic-Product Processing & Preservation, Shanghai, China
| |
Collapse
|
19
|
Marin C, Tuts J, Luyten FP, Vandamme K, Kerckhofs G. Impaired soft and hard callus formation during fracture healing in diet-induced obese mice as revealed by 3D contrast-enhanced computed tomography imaging. Bone 2021; 150:116008. [PMID: 33992820 DOI: 10.1016/j.bone.2021.116008] [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: 01/14/2021] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 11/21/2022]
Abstract
The impact of diabetes mellitus on bone fracture healing is clinically relevant as the patients experience delayed fracture healing. Even though efforts have been made to understand the detrimental effects of type 2 diabetes mellitus (T2DM) on the fracture healing process, the exact mechanisms causing the pathophysiological outcomes remain unclear. The aim of this study was to assess alterations in bone fracture healing (tibial fracture surgery, intramedullary pinning) of diet-induced obese (DIO) mice, and to investigate the in vitro properties of osteochondroprogenitors derived from the diabetic micro-environment. High-resolution contrast-enhanced microfocus X-ray computed tomography (CE-CT) enabled a simultaneous 3D assessment of the amount and spatial distribution of the regenerated soft and hard tissues during fracture healing and revealed that osteogenesis as well as chondrogenesis are altered in DIO mice. Compared to age-matched lean controls, DIO mice presented a decreased bone volume fraction and increased callus volume and adiposity at day 14 post-fracture. Of note, bone turnover was found altered in DIO mice relative to controls, evidenced by decreased blood serum osteocalcin and increased serum CTX levels. The in vitro data revealed that not only the osteogenic and adipogenic differentiation of periosteum-derived cells (PDCs) were altered by hyperglycemic (HG) conditions, but also the chondrogenic differentiation. Elevated PPARγ expression in HG conditions confirmed the observed increase in differentiated adipocytes in vitro. Finally, chondrogenesis-related genes COL2 and COL10 were downregulated for PDCs treated with HG medium, confirming that chondrogenic differentiation is compromised in vitro and suggesting that this may affect callus formation and maturation during the fracture healing process in vivo. Altogether, these results provide novel insights into the alterations of long bone fracture repair and suggest a link between HG-induced dysfunctionality of osteochondroprogenitor differentiation and fracture healing impairment under T2DM conditions.
Collapse
Affiliation(s)
- Carlos Marin
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, Leuven, Belgium; Prometheus - Division of Skeletal Tissue Engineering Leuven, KU Leuven, Leuven, Belgium; Biomaterials - BIOMAT, Department of Oral Health Sciences, KU Leuven, Leuven, Belgium
| | - Jolien Tuts
- Biomaterials - BIOMAT, Department of Oral Health Sciences, KU Leuven, Leuven, Belgium
| | - Frank P Luyten
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, Leuven, Belgium; Prometheus - Division of Skeletal Tissue Engineering Leuven, KU Leuven, Leuven, Belgium
| | - Katleen Vandamme
- Prometheus - Division of Skeletal Tissue Engineering Leuven, KU Leuven, Leuven, Belgium; Biomaterials - BIOMAT, Department of Oral Health Sciences, KU Leuven, Leuven, Belgium
| | - Greet Kerckhofs
- Prometheus - Division of Skeletal Tissue Engineering Leuven, KU Leuven, Leuven, Belgium; Department of Material Science and Engineering, KU Leuven, Leuven, Belgium; Biomechanics lab, Institute of Mechanics, Materials, and Civil Engineering, UCLouvain, Louvain-la-Neuve, Belgium; Institute for Experimental and Clinical Research, UCLouvain, Woluwe, Belgium.
| |
Collapse
|
20
|
Funk MI, Conde MA, Piwien-Pilipuk G, Uranga RM. Novel antiadipogenic effect of menadione in 3T3-L1 cells. Chem Biol Interact 2021; 343:109491. [PMID: 33945810 DOI: 10.1016/j.cbi.2021.109491] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/31/2021] [Accepted: 04/19/2021] [Indexed: 12/30/2022]
Abstract
Inhibition of adipocyte differentiation can be used as a strategy for preventing adipose tissue expansion and, consequently, for obesity management. Since reactive oxygen species (ROS) have emerged as key modulators of adipogenesis, the effect of menadione (a synthetic form of vitamin K known to induce the increase of intracellular ROS) on 3T3-L1 preadipocyte differentiation was studied. Menadione (15 μM) increased ROS and lipid peroxidation, generating mild oxidative stress without affecting cell viability. Menadione drastically inhibited adipogenesis, accompanied by decreased intracellular lipid accumulation and diminished expression of the lipo/adipogenic markers peroxisome proliferator-activated receptor (PPAR)γ, fatty acid synthase (FAS), CCAAT/enhancer-binding protein (C/EBP) α, fatty acid binding protein (FABP) 4, and perilipin. Menadione treatment also increased lipolysis, as indicated by augmented glycerol release and reinforced by the increased expression of hormone-sensitive lipase (HSL). Additionally, menadione increased the inhibitory phosphorylation of acetyl-CoA-carboxylase (ACC), which results in the inhibition of fatty acid synthesis. As a consequence, triglyceride content was decreased. Menadione also inhibited the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. Further, treatment with increased concentration of insulin, a potent physiological activator of the PI3K/Akt pathway, rescued the normal level of expression of PPARγ, the master regulator of adipogenesis, and overcame the restraining effect of menadione on the differentiation capacity of 3T3-L1 preadipocytes. Our study reveals novel antiadipogenic action for menadione, which is, at least in part, mediated by the PI3K/Akt pathway signaling and raises its potential as a therapeutic agent in the treatment or prevention of adiposity.
Collapse
Affiliation(s)
- Melania Iara Funk
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Universidad Nacional Del Sur (UNS)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina. Departamento de Biología, Bioquímica y Farmacia, UNS, Bahía Blanca, Argentina
| | - Melisa Ailén Conde
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Universidad Nacional Del Sur (UNS)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina. Departamento de Biología, Bioquímica y Farmacia, UNS, Bahía Blanca, Argentina
| | | | - Romina María Uranga
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Universidad Nacional Del Sur (UNS)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina. Departamento de Biología, Bioquímica y Farmacia, UNS, Bahía Blanca, Argentina.
| |
Collapse
|
21
|
Gong F, Gao L, Ma L, Li G, Yang J. Uncarboxylated osteocalcin alleviates the inhibitory effect of high glucose on osteogenic differentiation of mouse bone marrow-derived mesenchymal stem cells by regulating TP63. BMC Mol Cell Biol 2021; 22:24. [PMID: 33906607 PMCID: PMC8080387 DOI: 10.1186/s12860-021-00365-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/12/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Progressive population aging has contributed to the increased global prevalence of diabetes and osteoporosis. Inhibition of osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) by hyperglycemia is a potential pathogenetic mechanism of osteoporosis in diabetic patients. Uncarboxylated osteocalcin (GluOC), a protein secreted by mature osteoblasts, regulates bone development as well as glucose and lipid metabolism. In our previous studies, GluOC was shown to promote osteoblastic differentiation of BMSCs; however, the underlying mechanisms are not well characterized. Tumor protein 63 (TP63), as a transcription factor, is closely related to bone development and glucose metabolism. RESULTS In this study, we verified that high glucose suppressed osteogenesis and upregulated adipogenesis in BMSCs, while GluOC alleviated this phenomenon. In addition, high glucose enhanced TP63 expression while GluOC diminished it. Knock-down of TP63 by siRNA transfection restored the inhibitory effect of high glucose on osteogenic differentiation. Furthermore, we detected the downstream signaling pathway PTEN/Akt/GSK3β. We found that diminishing TP63 decreased PTEN expression and promoted the phosphorylation of Akt and GSK3β. We then applied the activator and inhibitor of Akt, and concluded that PTEN/Akt/GSK3β participated in regulating the differentiation of BMSCs. CONCLUSIONS Our results indicate that GluOC reduces the inhibitory effect of high glucose on osteoblast differentiation by regulating the TP63/PTEN/Akt/GSK3β pathway. TP63 is a potential novel target for the prevention and treatment of diabetic osteoporosis.
Collapse
Affiliation(s)
- Fangzi Gong
- Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Le Gao
- Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Luyao Ma
- Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Guangxin Li
- College of sports medicine and physical therapy, Beijing Sport University, Beijing, China
| | - Jianhong Yang
- Medical School, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
22
|
Donat A, Knapstein PR, Jiang S, Baranowsky A, Ballhause TM, Frosch KH, Keller J. Glucose Metabolism in Osteoblasts in Healthy and Pathophysiological Conditions. Int J Mol Sci 2021; 22:ijms22084120. [PMID: 33923498 PMCID: PMC8073638 DOI: 10.3390/ijms22084120] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/11/2021] [Accepted: 04/14/2021] [Indexed: 01/01/2023] Open
Abstract
Bone tissue in vertebrates is essential to performing movements, to protecting internal organs and to regulating calcium homeostasis. Moreover, bone has also been suggested to contribute to whole-body physiology as an endocrine organ, affecting male fertility; brain development and cognition; and glucose metabolism. A main determinant of bone quality is the constant remodeling carried out by osteoblasts and osteoclasts, a process consuming vast amounts of energy. In turn, clinical conditions associated with impaired glucose metabolism, including type I and type II diabetes and anorexia nervosa, are associated with impaired bone turnover. As osteoblasts are required for collagen synthesis and matrix mineralization, they represent one of the most important targets for pharmacological augmentation of bone mass. To fulfill their function, osteoblasts primarily utilize glucose through aerobic glycolysis, a process which is regulated by various molecular switches and generates adenosine triphosphate rapidly. In this regard, researchers have been investigating the complex processes of energy utilization in osteoblasts in recent years, not only to improve bone turnover in metabolic disease, but also to identify novel treatment options for primary bone diseases. This review focuses on the metabolism of glucose in osteoblasts in physiological and pathophysiological conditions.
Collapse
|
23
|
Guo JM, Xing HJ, Cai JZ, Zhang HF, Xu SW. H 2S exposure-induced oxidative stress promotes LPS-mediated hepatocyte autophagy through the PI3K/AKT/TOR pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 209:111801. [PMID: 33383342 DOI: 10.1016/j.ecoenv.2020.111801] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 06/12/2023]
Abstract
Hydrogen sulfide (H2S), a common air pollutant and toxic gas, is detrimental to organisms and the environment. Exposure to highly concentrated H2S can induce oxidative stress and autophagy. However, the mechanism underlying the liver damage caused by H2S has not been identified. Lipopolysaccharide (LPS), the key component of endotoxin, can induce oxidative stress and autophagy. For this experiment, we used one-day-old chickens as model organisms to evaluate the effects of H2S combined with LPS on oxidative stress and autophagy. The four groups (control group, LPS group, H2S group and H2S-LPS group) were observed by electron microscopy, detected by oxidative stress kit, analyzed by quantitative real-time quantitative PCR, and analyzed by Western blot. We found that the activities of antioxidant enzymes (superoxide dismutase, antioxidant glutathione, catalase, and glutathione peroxidase) decreased in the H2S group compared to those in the control group; however, malondialdehyde levels in the H2S group increased. Molecular-level studies showed that the expression of genes associated with the PI3K/ AKT/ TOR pathways in the H2S group decreased, whereas the expression of other autophagy-related genes (Beclin1, ATG5 and the ratio of LC3-II/ LC3-I) increased compared to that in the control group. These findings suggest that H2S caused oxidative stress and induced autophagy through the PI3K/ AKT/ TOR pathway in chicken liver cells. Additionally, exposure to H2S aggravated LPS-induced oxidative stress and autophagy injury. Capsule: Aerial exposure to H2S can cause oxidative stress in chicken livers and induce autophagy through the PI3K/AKT/TOR pathway, and can aggravate LPS-induced oxidative stress and autophagy.
Collapse
Affiliation(s)
- Jin-Ming Guo
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, PR China
| | - Hou-Juan Xing
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Jing-Zeng Cai
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, PR China
| | - Hong-Fu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Shi-Wen Xu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
24
|
Fojtík P, Beckerová D, Holomková K, Šenfluk M, Rotrekl V. Both Hypoxia-Inducible Factor 1 and MAPK Signaling Pathway Attenuate PI3K/AKT via Suppression of Reactive Oxygen Species in Human Pluripotent Stem Cells. Front Cell Dev Biol 2021; 8:607444. [PMID: 33553145 PMCID: PMC7859355 DOI: 10.3389/fcell.2020.607444] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/15/2020] [Indexed: 12/15/2022] Open
Abstract
Mild hypoxia (5% O2) as well as FGFR1-induced activation of phosphatidylinositol-4,5-bisphosphate 3-kinase/protein kinase B (PI3K/AKT) and MAPK signaling pathways markedly support pluripotency in human pluripotent stem cells (hPSCs). This study demonstrates that the pluripotency-promoting PI3K/AKT signaling pathway is surprisingly attenuated in mild hypoxia compared to the 21% O2 environment. Hypoxia is known to be associated with lower levels of reactive oxygen species (ROS), which are recognized as intracellular second messengers capable of upregulating the PI3K/AKT signaling pathway. Our data denote that ROS downregulation results in pluripotency upregulation and PI3K/AKT attenuation in a hypoxia-inducible factor 1 (HIF-1)-dependent manner in hPSCs. Using specific MAPK inhibitors, we show that the MAPK pathway also downregulates ROS and therefore attenuates the PI3K/AKT signaling—this represents a novel interaction between these signaling pathways. This inhibition of ROS initiated by MEK1/2–ERK1/2 may serve as a negative feedback loop from the MAPK pathway toward FGFR1 and PI3K/AKT activation. We further describe the molecular mechanism resulting in PI3K/AKT upregulation in hPSCs—ROS inhibit the PI3K's primary antagonist PTEN and upregulate FGFR1 phosphorylation. These novel regulatory circuits utilizing ROS as second messengers may contribute to the development of enhanced cultivation and differentiation protocols for hPSCs. Since the PI3K/AKT pathway often undergoes an oncogenic transformation, our data could also provide new insights into the regulation of cancer stem cell signaling.
Collapse
Affiliation(s)
- Petr Fojtík
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czechia.,International Clinical Research Center (ICRC), St. Anne's University Hospital, Brno, Czechia
| | - Deborah Beckerová
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czechia.,International Clinical Research Center (ICRC), St. Anne's University Hospital, Brno, Czechia
| | - Katerina Holomková
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Martin Šenfluk
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czechia.,International Clinical Research Center (ICRC), St. Anne's University Hospital, Brno, Czechia
| | - Vladimir Rotrekl
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czechia.,International Clinical Research Center (ICRC), St. Anne's University Hospital, Brno, Czechia
| |
Collapse
|
25
|
Zhao R, Tao L, Qiu S, Shen L, Tian Y, Gong Z, Tao ZB, Zhu Y. Melatonin rescues glucocorticoid-induced inhibition of osteoblast differentiation in MC3T3-E1 cells via the PI3K/AKT and BMP/Smad signalling pathways. Life Sci 2020; 257:118044. [PMID: 32622944 DOI: 10.1016/j.lfs.2020.118044] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/20/2020] [Accepted: 06/29/2020] [Indexed: 12/20/2022]
Abstract
AIMS High-dose glucocorticoid (GC) administration causes osteoporosis. Many previous studies from our group and other groups have shown that melatonin participates in the regulation of osteoblast proliferation and differentiation, especially low concentrations of melatonin, which enhance osteoblast osteogenesis. However, the role of melatonin in glucocorticoid-induced osteoblast differentiation remains unknown. MATERIALS AND METHODS An examination of the expression of osteoblast differentiation markers (ALP, OCN, COLL-1), as well as alkaline phosphatase staining and alkaline phosphatase enzymatic activity assay to measure osteoblast differentiation and quantifying Alizarin red S staining to measure mineralization, were performed to determine the effects of dexamethasone (Dex) and melatonin on the differentiation of MC3T3-E1 cells. We used immunofluorescence staining to detect the expression of Runx2 in melatonin-treated MC3T3-E1 cells. The expression of mRNA was determined by qRT-PCR, and protein levels were measured by western blotting. KEY FINDINGS In the present study, we found that 100 μM Dex significantly reduced osteoblast differentiation and mineralization in MC3T3-E1 cells and that 1 μM melatonin attenuated these inhibitory effects. We found that only inhibition of PI3K/AKT (MK2206) and BMP/Smad (LDN193189) signalling abolished melatonin-induced differentiation and mineralization. Meanwhile, MK2206 decreased the expression of P-AKT and P-Smad1/5/9 and LDN193189 decreased the expression of P-Smad1/5/9 but had no obvious effect on P-AKT expression in melatonin-treated and Dex-induced MC3T3-E1 cells. SIGNIFICANCE These findings suggest that melatonin rescues Dex-induced inhibition of osteoblast differentiation in MC3T3-E1 cells via the PI3K/AKT and BMP/Smad signalling pathways and that PI3K/AKT signalling may be the upstream signal of BMP/Smad signalling.
Collapse
Affiliation(s)
- Rui Zhao
- Department of Orthopaedics, The First Hospital of China Medical University, Shenyang 110001, China
| | - Lin Tao
- Department of Orthopaedics, The First Hospital of China Medical University, Shenyang 110001, China
| | - Shui Qiu
- Department of Orthopaedics, The First Hospital of China Medical University, Shenyang 110001, China
| | - Lin Shen
- Department of Orthopaedics, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yihao Tian
- Department of Orthopaedics, The First Hospital of China Medical University, Shenyang 110001, China
| | - Zunlei Gong
- Department of Orthopaedics, The First Hospital of China Medical University, Shenyang 110001, China
| | - Zheng Bo Tao
- Department of Orthopaedics, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yue Zhu
- Department of Orthopaedics, The First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
26
|
Li Y, Shrestha A, Zhang H, Li L, Li D, Fu T, Song J, Ji P, Huang Y, Chen T. Impact of diabetes mellitus simulations on bone cell behavior through in vitro models. J Bone Miner Metab 2020; 38:607-619. [PMID: 32415376 DOI: 10.1007/s00774-020-01101-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 03/23/2020] [Indexed: 12/16/2022]
Abstract
Diabetes mellitus (DM) is related to impaired bone healing and an increased risk of bone fractures. While it is recognized that osteogenic differentiation and the function of osteoblasts are suppressed in DM, the influence of DM on osteoclasts is still unclear. Hyperglycemia and inflammatory environment are the hallmark of DM that causes dysregulation of various pro-inflammatory cytokines and alternated gene expression in periodontal ligament cells, osteoblasts, osteocytes, osteoclasts, and osteoclast precursors. A methodological review on conceptual and practical implications of in vitro study models is used for DM simulation on bone cells. Several major databases were screened to find literature related to the study objective. Published literature within last 20 years that used in vitro DM-simulated models to study how DM affects the cellular behavior of bone cells were selected for this review. Studies utilizing high glucose and serum acquired from diabetic animals are the mainly used methods to simulate the diabetic condition. The combination with various simulating factors such as lipopolysaccharide (LPS), hydrogen peroxide (H2O2), and advanced glycation end products (AGEs) have been reported in diabetic situations in vitro, as well. Through screening procedure, it was evident DM-simulated conditions exerted negative impact on bone-related cells. However, inconsistent results were found among different reported studies, which could be due to variation in culture conditions, concentrations of the stimulating factors and cell lineage, etc. This manuscript has concisely reviewed currently existing DM-simulated in vitro models and provides valuable insights of detailed components in simulating DM conditions in vitro. Studies using DM-simulated microenvironment revealed that in vitro simulation negatively impacted periodontal ligament cells, osteoblasts, osteocytes, osteoclasts, and osteoclast precursors. Contrarily, studies also indicated beneficial influence on bone-related cells when such conditions are reversed.
Collapse
Affiliation(s)
- Yihan Li
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, 426#Songshibei Road, Yubei District, Chongqing, 401147, P.R. China
| | - Annie Shrestha
- Faculty of Dentistry, University of Toronto, Toronto, ON, M5G 1G6, Canada
| | - Hongmei Zhang
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, 426#Songshibei Road, Yubei District, Chongqing, 401147, P.R. China
| | - Lingjie Li
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, 426#Songshibei Road, Yubei District, Chongqing, 401147, P.R. China
| | - Dize Li
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, 426#Songshibei Road, Yubei District, Chongqing, 401147, P.R. China
| | - Tiwei Fu
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, 426#Songshibei Road, Yubei District, Chongqing, 401147, P.R. China
| | - Jinlin Song
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, 426#Songshibei Road, Yubei District, Chongqing, 401147, P.R. China
| | - Ping Ji
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, 426#Songshibei Road, Yubei District, Chongqing, 401147, P.R. China
| | - Yuanding Huang
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, 426#Songshibei Road, Yubei District, Chongqing, 401147, P.R. China.
| | - Tao Chen
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, 426#Songshibei Road, Yubei District, Chongqing, 401147, P.R. China.
| |
Collapse
|
27
|
Rosa-Silva HTD, Panzenhagen AC, Schmidtt V, Alves Teixeira A, Espitia-Pérez P, de Oliveira Franco Á, Mingori M, Torres-Ávila JF, Schnorr CE, Hermann PRS, Moraes DP, Almeida RF, Moreira JCF. Hepatic and neurobiological effects of foetal and breastfeeding and adulthood exposure to methylmercury in Wistar rats. CHEMOSPHERE 2020; 244:125400. [PMID: 31809933 DOI: 10.1016/j.chemosphere.2019.125400] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/20/2019] [Accepted: 11/17/2019] [Indexed: 06/10/2023]
Abstract
Methylmercury (MeHg) is an organic bioaccumulated mercury derivative that strongly affects the environment and represents a public health problem primarily to riparian communities in South America. Our objective was to investigate the hepatic and neurological effects of MeHg exposure during the phases foetal and breast-feeding and adult in Wistar rats. Wistar rats (n = 10) were divided into 3 groups. Control group received mineral oil; The simple exposure (SE) group was exposed only in adulthood (0.5 mg/kg/day); and double exposure (DE) was pre-exposed to MeHg 0.5 mg/kg/day during pregnancy and breastfeeding (±40 days) and re-exposed to MeHg for 45 days from day 100. After, we evaluated possible abnormalities. Behavioral and biochemical parameters in liver and occipital cortex (CO), markers of liver injury, redox and AKT/GSK3β/mTOR signaling pathway. Our results showed that both groups treated with MeHg presented significant alterations, such as decreased locomotion and exploration and impaired visuospatial perception. The rats exposed to MeHg showed severe liver damage and increased hepatic glycogen concentration. The MeHg groups showed significant impairment in redox balance and oxidative damage to liver macromolecules and CO. MeHg upregulated the AKT/GSK3β/mTOR pathway and the phosphorylated form of the Tau protein. In addition, we found a reduction in NeuN and GFAP immunocontent. These results represent the first approach to the hepatotoxic and neural effects of foetal and adult MeHg exposure.
Collapse
Affiliation(s)
- Helen Tais da Rosa-Silva
- Centro de Estudos em Estresse Oxidativo, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.
| | - Alana Castro Panzenhagen
- Centro de Estudos em Estresse Oxidativo, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Victória Schmidtt
- Centro de Estudos em Estresse Oxidativo, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Alexsander Alves Teixeira
- Centro de Estudos em Estresse Oxidativo, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Pedro Espitia-Pérez
- Facultad de Ciencias de la Salud, Laboratorio de Investigación Biomédica y Biología Molecular, Universidad del Sinú, Córdoba, Colombia
| | - Álvaro de Oliveira Franco
- Centro de Estudos em Estresse Oxidativo, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Moara Mingori
- Centro de Estudos em Estresse Oxidativo, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - José F Torres-Ávila
- Unit for Development and Innovation in Genetics and Molecular Biology, Universidad Simón Bolívar, Barranquilla, Atlántico, Colombia
| | - Carlos Eduardo Schnorr
- Departamento de Civil y Ambiental, Programa de Ingeniería Ambiental, Universidad de la Costa, Barranquilla, Atlántico, Colombia
| | - Paolla Rissi Silva Hermann
- Instituto de Química, Sala 103, Campus do Vale, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Diogo Pompéu Moraes
- Instituto de Química, Sala 103, Campus do Vale, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Roberto Farina Almeida
- Departamento de Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - José Cláudio Fonseca Moreira
- Centro de Estudos em Estresse Oxidativo, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
28
|
Qi S, He J, Zheng H, Chen C, Jiang H, Lan S. Zinc Supplementation Increased Bone Mineral Density, Improves Bone Histomorphology, and Prevents Bone Loss in Diabetic Rat. Biol Trace Elem Res 2020; 194:493-501. [PMID: 31363990 DOI: 10.1007/s12011-019-01810-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/01/2019] [Indexed: 12/24/2022]
Abstract
Diabetic osteoporosis (DOP) is a complication of diabetes, with the characteristics of bone mineral density (BMD) reduction and bone structure destruction. Zinc was reported has a benefit effect on postmenopausal osteoporosise, it was also has hypoglycemic effect, whether zinc was beneficial on diabetes-induced osteoporosis has not been reported. So in the present study, we established a diabetic rat model by streptozotocin injection (60 mg/kg), and administered zinc sulfate by oral gavage to investigate the protective effects of zinc on DOP and the underline possible mechanism. Thirty six Sprague Dawley rats were divided into T1DM group (diabetic rats), control group (vehicle treatment), and T1DM-Zinc group (diabetic rats administered zinc sulfate 0.25 mg/kg by oral gavage). The bone histomorphological parameters, serum bone metabolism markers (including ALP, OPG, RUNX 2, and RANKL), BMD, and bone marrow adipocyte numbers were detected after eight weeks of zinc sulfate treatment. The results showed zinc sulfate administration (0.25 mg/kg/d) decreased blood glucose, increased the BMD, decreased serum ALP, and RANKL, increased serum OPG and RUNX 2 levels, as well as OPG/RANKL ratio of T1DM rats. Meanwhile, the bone histomorphological parameters, bone marrow adipocytes numbers were returned to be normal. The RUNX 2, and OPG mRNA expression levels in bone tissues of T1DM-Zinc group rats were increased after zinc sulfate treatment compared with the diabetic rats (P < 0.05). Those indicating that zinc sulfate can prevent DOP, the protective mechanism were mainly related to its hypoglycemic effect, bone marrow lipogenesis inhibition effect, OPG/RANKL ratio and RUNX 2 up-regulation effect.
Collapse
Affiliation(s)
- Shanshan Qi
- Vitamin D Research Institute, College of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, 723000, Shaanxi, China
| | - Jia He
- Vitamin D Research Institute, College of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, 723000, Shaanxi, China
| | - Hongxing Zheng
- Chinese-German Joint Laboratory for Natural Product Research, College of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, 723000, China.
| | - Chen Chen
- Chinese-German Joint Laboratory for Natural Product Research, College of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, 723000, China
| | - Hai Jiang
- Chinese-German Joint Laboratory for Natural Product Research, College of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, 723000, China
| | - Shiqiang Lan
- Vitamin D Research Institute, College of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, 723000, Shaanxi, China
| |
Collapse
|
29
|
Hyperglycemia compromises Rat Cortical Bone by Increasing Osteocyte Lacunar Density and Decreasing Vascular Canal Volume. Commun Biol 2020; 3:20. [PMID: 31925331 PMCID: PMC6952406 DOI: 10.1038/s42003-019-0747-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 12/06/2019] [Indexed: 12/13/2022] Open
Abstract
Uncontrolled diabetes is associated with increased risk of bony fractures. However, the mechanisms have yet to be understood. Using high-resolution synchrotron micro-CT, we calculated the changes in the microstructure of femoral cortices of streptozotocin-induced hyperglycemic (STZ) Wistar Albino rats and tested the mechanical properties of the mineralized matrix by nanoindentation. Total lacunar volume of femoral cortices increased in STZ group due to a 9% increase in lacunar density. However, total vascular canal volume decreased in STZ group due to a remarkable decrease in vascular canal diameter (7 ± 0.3 vs. 8.5 ± 0.4 µm). Osteocytic territorial matrix volume was less in the STZ group (14,908 ± 689 µm3) compared with healthy controls (16,367 ± 391 µm3). In conclusion, hyperglycemia increased cellularity and lacunar density, decreased osteocyte territorial matrix, and reduced vascular girth, in addition to decreasing matrix mechanical properties in the STZ group when compared with euglycemic controls. Birol Ay et al. use high-resolution synchrotron radiation micro-CT to calculate the changes in the microstructure of femoral cortices in STZ-induced hyperglycemic rats. They show that hyperglycemia increases lacunar density due to a reduction in osteocytic territorial matrix volume but decreases total vascular canal volume due to a decrease in canal diameter.
Collapse
|
30
|
IGF1 Knockdown Hinders Myocardial Development through Energy Metabolism Dysfunction Caused by ROS-Dependent FOXO Activation in the Chicken Heart. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7838754. [PMID: 31949883 PMCID: PMC6948330 DOI: 10.1155/2019/7838754] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 11/16/2019] [Accepted: 11/20/2019] [Indexed: 12/15/2022]
Abstract
Insulin-like growth factor 1 (IGF1) is a multifunctional cellular regulatory factor that can regulate cell growth and development by mediating growth hormone stimulation. However, the mechanism of IGF1 dysfunction in cardiomyocyte development is seldom reported. To study this, we employed the models of IGF1 knockdown in chicken embryo in vivo and in cardiomyocytes in vitro. We detected the antioxidant capacity, PI3K/Akt pathway, energy metabolism-related genes, and myocardial development-related genes. Our results revealed that the low expression of IGF1 can significantly suppress the antioxidant capacity and increase the ROS (P < 0.05) levels, activating the AMPK and PI3K pathway by inhibiting the expression of IRS1. We also found that myocardial energy metabolism is blocked through IGF1, GLUT, and IGFBP inhibition, further inducing myocardial developmental disorder by inhibiting Mesp1, GATA, Nkx2.5, and MyoD expression. Altogether, we conclude that low IGF1 expression can hinder myocardial development through the dysfunction of energy metabolism caused by ROS-dependent FOXO activation.
Collapse
|
31
|
Cordeiro ER, Filetti FM, Simões MR, Vassallo DV. Mercury induces nuclear estrogen receptors to act as vasoconstrictors promoting endothelial denudation via the PI3K/Akt signaling pathway. Toxicol Appl Pharmacol 2019; 381:114710. [PMID: 31415774 DOI: 10.1016/j.taap.2019.114710] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/29/2019] [Accepted: 08/10/2019] [Indexed: 10/26/2022]
Abstract
Cardiovascular diseases (CVD) are more frequent among postmenopausal women due to the decline of estrogen concentration in plasma. However, the role of the vascular modulator effect of estrogen is controversial, since it occurs both in physiological and pathological conditions, increasing or reducing vascular reactivity. As mercury is widely associated with the development of CVD, we investigated putative hazardous effects on the mechanisms that modulate vascular reactivity in aortic rings of female Wistar rats promoted by acute mercury exposure. Mercury increased vascular reactivity and oxidative stress possibly due to NADPH oxidase participation, increased production of cyclooxygenase-2 (COX-2) and thromboxane A2 (TXA2) formation. The metal also induced endothelial denudation in the aorta by reducing the bioavailability of nitric oxide (NO) and enhancing the activity of the PI3K/Akt signaling pathway. Mercury exposure also induced nuclear estrogen receptors (ERα, ERβ) to act as vasoconstrictors. Our findings suggest that mercury might increase the chances of developing cardiovascular diseases in females and should be considered an important environmental risk factor.
Collapse
Affiliation(s)
- Evellyn Rodrigues Cordeiro
- Dept. of Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, ES CEP 29043-900, Brazil
| | - Filipe Martinuzo Filetti
- Dept. of Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, ES CEP 29043-900, Brazil
| | - Maylla Ronacher Simões
- Dept. of Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, ES CEP 29043-900, Brazil
| | - Dalton Valentim Vassallo
- Dept. of Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, ES CEP 29043-900, Brazil; Health Science Center of Vitória-EMESCAM, Vitória, ES CEP 29045-402, Brazil.
| |
Collapse
|
32
|
Zhao J, Du X, Wang M, Yang P, Zhang J. Salidroside mitigates hydrogen peroxide-induced injury by enhancement of microRNA-27a in human trabecular meshwork cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:1758-1765. [PMID: 31062616 DOI: 10.1080/21691401.2019.1608222] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Salidroside (Sal) exerted widely pharmacological effects in multitudinous diseases had been certified. The actual study clarified the protective activity of Sal in H2O2-injured human trabecular meshwork (HTM) cells. HTM cells were disposed with H2O2 to construct an oxidative damage model in vitro. Then, Sal was utilized to administrate HTM cells, and cell viability, apoptosis, apoptosis-interrelated proteins and ROS production were appraised using CCK-8, flow cytometry, western blot and DCFH-DA staining. MiR-27a inhibitor and its control were transfected into HTM cells, and the influences of miR-27a inhibition in HTM cells stimulated with H2O2 and Sal were detected. PI3K/AKT and Wnt/β-catenin pathways were ultimately investigated to uncover the underlying mechanism. We found that H2O2 evoked HTM cells oxidative damage, as evidenced by repressing cell viability, inducing apoptosis, activating cleaved-caspase-3/-9 expression and increasing ROS production. Sal significantly lightened H2O2-evoked oxidative damage in HTM cells. Additionally, miR-27a was up-regulated by Sal, and miR-27a suppression significantly reversed the protective effect of Sal on H2O2-injured HTM cells. Finally, Sal activated PI3K/AKT and Wnt/β-catenin pathways through enhancement of miR-27a in H2O2-injured HTM cells. In conclusion, these discoveries suggested that Sal could protect HTM cells against H2O2-evoked oxidative damage by activating PI3K/AKT and Wnt/β-catenin pathways through enhancement of miR-27a. Highlights H2O2 evokes HTM cells oxidative damage; Sal relieves H2O2-induced oxidative damage in HTM cells; Sal enhances miR-27a expression in H2O2-injured HTM cells; Repressed miR-27a reverses the protective impacts of Sal on H2O2-injured HTM cells; Sal activates PI3K/AKT and Wnt/β-catenin pathways by increasing miR-27a.
Collapse
Affiliation(s)
- Jun Zhao
- a Department of Ophthalmology , Linyi People's Hospital , Linyi , China
| | - Xiujuan Du
- b Department of Ophthalmology, Eye Institute of Shandong University of Traditional Chinese Medicine , Affiliated Eye Hospital of Shandong University of TCM , Jinan , China
| | - Meng Wang
- a Department of Ophthalmology , Linyi People's Hospital , Linyi , China
| | | | - Juanmei Zhang
- a Department of Ophthalmology , Linyi People's Hospital , Linyi , China
| |
Collapse
|
33
|
Hyperglycemia Changes Expression of Key Adipogenesis Markers (C/EBPα and PPARᵞ)and Morphology of Differentiating Human Visceral Adipocytes. Nutrients 2019; 11:nu11081835. [PMID: 31398873 PMCID: PMC6723080 DOI: 10.3390/nu11081835] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 07/31/2019] [Accepted: 08/06/2019] [Indexed: 12/11/2022] Open
Abstract
Disturbances in adipose tissue significantly contribute to the development of metabolic disorders, which are connected with hyperglycemia (HG) and underlain by epigenetics-based mechanisms. Therefore, we aimed to evaluate the effect of hyperglycemia on proliferating, differentiating and maturating human visceral pre/adipocytes (HPA-v). Three stages of cell culture were conducted under constant or variable glycemic conditions. Adipogenesis progress was assessed using BODIPY 505/515 staining. Lipid content typical for normal and hyperglycemic conditions of adipocytes was analyzed using Raman spectroscopy and imaging. Expression of adipogenic markers, PPARγ and C/EBPα, was determined at the mRNA and protein levels. We also examined expression of miRNAs proven to target PPARγ (miR-34a-5p) and C/EBPα (miR-137-3p), employing TaqMan Low-Density Arrays (TLDA) cards. Hyperglycemia altered morphology of differentiating HPA-v in relation to normoglycemia by accelerating the formation of lipid droplets and making their numbers and volume increase. Raman results confirmed that the qualitative and quantitative lipid composition under normal and hyperglycemic conditions were different, and that the number of lipid droplets increased in (HG)-treated cells. Expression profiles of both examined genes markedly changed either during adipogenesis under physiological and hyperglycemic conditions, orat particular stages of adipogenesis upon chronic and/or variable glycemia. Expression levels of PPARγ seemed to correspond to some expression changes of miR-34a-5p. miR-137-3p, whose expression was rather stable throughout the culture, did not seem to affect C/EBPα. Our observations revealed that chronic and intermittent hyperglycemia change the morphology of visceral pre/adipocytes during adipogenesis. Moreover, hyperglycemia may utilize miR-34a-5p to induce some expression changes in PPARγ.
Collapse
|
34
|
p53 sensitizes chemoresistant non-small cell lung cancer via elevation of reactive oxygen species and suppression of EGFR/PI3K/AKT signaling. Cancer Cell Int 2019; 19:188. [PMID: 31360122 PMCID: PMC6642601 DOI: 10.1186/s12935-019-0910-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 07/15/2019] [Indexed: 01/21/2023] Open
Abstract
Background Non-small cell lung cancer (NSCLC) is the leading cause of cancer deaths primarily due to chemoresistance. Somatic mutation of TP53 (36%) and epidermal growth factor receptor (EGFR; > 30%) are major contributors to cisplatin (CDDP) resistance. Substantial evidence suggests the elevated levels of reactive oxygen species (ROS) is a key determinant in cancer. The elevated ROS can affect the cellular responses to chemotherapeutic treatments. Although the role of EGFR in PI3K/Akt signaling cascade in NSCLC is extensively studied, the molecular link between EGFR and p53 and the role of ROS in pathogenesis of NSCLC are limitedly addressed. In this study, we investigated the role of p53 in regulation of ROS production and EGFR signaling, and the chemosensitivity of NSCLC. Methods In multiple NSCLC cell lines with varied p53 and EGFR status, we compared and examined the protein contents involved in EGFR-Akt-P53 signaling loop (EGFR, P-EGFR, Akt, P-Akt, p53, P-p53) by Western blot. Apoptosis was determined based on nuclear morphological assessment using Hoechst 33258 staining. Cellular ROS levels were measured by dichlorofluorescin diacetate (DCFDA) staining followed by flow cytometry analysis. Results We have demonstrated for the first time that activation of p53 sensitizes chemoresistant NSCLC cells to CDDP by down-regulating EGFR signaling pathway and promoting intracellular ROS production. Likewise, blocking EGFR/PI3K/AKT signaling with PI3K inhibitor elicited a similar response. Our findings suggest that CDDP-induced apoptosis in chemosensitive NSCLC cells involves p53 activation, leading to suppressed EGFR signaling and ROS production. In contrast, in chemoresistant NSCLC, activated Akt promotes EGFR signaling by the positive feedback loop and suppresses CDDP-induced ROS production and apoptosis. Conclusion Collectively, our study reveals that the interaction of the p53 and Akt feedback loops determine the fate of NSCLC cells and their CDDP sensitivity. Electronic supplementary material The online version of this article (10.1186/s12935-019-0910-2) contains supplementary material, which is available to authorized users.
Collapse
|
35
|
Kalaitzoglou E, Fowlkes JL, Popescu I, Thrailkill KM. Diabetes pharmacotherapy and effects on the musculoskeletal system. Diabetes Metab Res Rev 2019; 35:e3100. [PMID: 30467957 PMCID: PMC6358500 DOI: 10.1002/dmrr.3100] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/14/2018] [Accepted: 11/19/2018] [Indexed: 12/13/2022]
Abstract
Persons with type 1 or type 2 diabetes have a significantly higher fracture risk than age-matched persons without diabetes, attributed to disease-specific deficits in the microarchitecture and material properties of bone tissue. Therefore, independent effects of diabetes drugs on skeletal integrity are vitally important. Studies of incretin-based therapies have shown divergent effects of different agents on fracture risk, including detrimental, beneficial, and neutral effects. The sulfonylurea class of drugs, owing to its hypoglycemic potential, is thought to amplify the risk of fall-related fractures, particularly in the elderly. Other agents such as the biguanides may, in fact, be osteo-anabolic. In contrast, despite similarly expected anabolic properties of insulin, data suggests that insulin pharmacotherapy itself, particularly in type 2 diabetes, may be a risk factor for fracture, negatively associated with determinants of bone quality and bone strength. Finally, sodium-dependent glucose co-transporter 2 inhibitors have been associated with an increased risk of atypical fractures in select populations, and possibly with an increase in lower extremity amputation with specific SGLT2I drugs. The role of skeletal muscle, as a potential mediator and determinant of bone quality, is also a relevant area of exploration. Currently, data regarding the impact of glucose lowering medications on diabetes-related muscle atrophy is more limited, although preclinical studies suggest that various hypoglycemic agents may have either aggravating (sulfonylureas, glinides) or repairing (thiazolidinediones, biguanides, incretins) effects on skeletal muscle atrophy, thereby influencing bone quality. Hence, the therapeutic efficacy of each hypoglycemic agent must also be evaluated in light of its impact, alone or in combination, on musculoskeletal health, when determining an individualized treatment approach. Moreover, the effect of newer medications (potentially seeking expanded clinical indication into the pediatric age range) on the growing skeleton is largely unknown. Herein, we review the available literature regarding effects of diabetes pharmacotherapy, by drug class and/or by clinical indication, on the musculoskeletal health of persons with diabetes.
Collapse
Affiliation(s)
- Evangelia Kalaitzoglou
- University of Kentucky Barnstable Brown Diabetes Center Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, USA
| | - John L Fowlkes
- University of Kentucky Barnstable Brown Diabetes Center Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Iuliana Popescu
- University of Kentucky Barnstable Brown Diabetes Center Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Kathryn M Thrailkill
- University of Kentucky Barnstable Brown Diabetes Center Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
36
|
Rharass T, Lucas S. High Glucose Level Impairs Human Mature Bone Marrow Adipocyte Function Through Increased ROS Production. Front Endocrinol (Lausanne) 2019; 10:607. [PMID: 31551934 PMCID: PMC6746912 DOI: 10.3389/fendo.2019.00607] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 08/21/2019] [Indexed: 12/20/2022] Open
Abstract
Bone marrow adipocytes (BMAds) accumulate in aging, menopause, and metabolic diseases such as Type 2 diabetes. These osteoporotic conditions are associated with oxidative stress and hyperglycemia which are both considered as critical factors underlying bone fragility. Glucose excess and reactive oxygen species (ROS) are known to favor adipogenesis over osteoblastogenesis. In this study, we investigated whether high glucose exposure could determine dysfunction of mature BMAds, specifically through ROS production. The effects of low (LG, 5 mM) or high glucose (HG, 25 mM) concentrations were examined using human bone mesenchymal stromal cells (hBMSCs) in the time course of differentiation, and, up to 21 days once adipocytes were mature. HG did not alter the adipocyte differentiation process of hBMSCs. Yet, after 21 days under HG exposure, PPARG, CEBPA, and adiponectin mRNA expressions were decreased. These alterations were also observed following adipogenic inducer withdrawal as well as in adipocytes fully differentiated in LG then cultured in HG for the last 11 days. Without inducers, HG condition also led to decreased leptin mRNA level. Importantly, intracellular and extracellular ROS concentrations measured using Amplex Red were significantly raised by 50% under HG exposure. This rise was observed once adipocytes ended differentiation and was reproduced within the different cell culture settings without any cytotoxicity. Among genes involved in ROS metabolism, the mRNA level of the H2O2 generating enzyme NOX4 was found upregulated in the presence of HG. Following cell separation, mature BMAds were shown to overproduce ROS and to display the gene alterations in contrast to non-lipid-laden cells. Finally, a non-lethal treatment with a pro-oxidant agent under LG condition reduces the mRNA levels of PPARG, adiponectin, and leptin as the HG condition does in the absence of inducers, and amplifies the effect of glucose excess on gene expression. HG concentration drives mature BMAds toward altered expression of the main adipokines and transcriptional factors. These perturbations are associated with a rise in ROS generation likely mediated through enhanced expression of NOX4. Mature BMAds are thus responsive to changes in glucose and ROS concentrations, which is relevant regarding with their phenotype and function in age- or metabolic disease-related osteoporosis.
Collapse
|
37
|
Role of Osteogenic Coatings on Implant Surfaces in Promoting Bone-To-Implant Contact in Experimental Osteoporosis: A Systematic Review and Meta-Analysis. IMPLANT DENT 2018; 26:770-777. [PMID: 28767464 DOI: 10.1097/id.0000000000000634] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE The aim of this systematic review and meta-analysis was to evaluate the role of osteogenic coatings (placement of a thin film of organic and inorganic osteoinductive and osteoproliferative materials) on implant surfaces in augmenting bone-to-implant contact (BIC) in osteoporotic bone. DATA SOURCES To answer the focused question "Do osteogenic coatings on implant surfaces increase BIC in osteoporotic bone?" PubMed/MEDLINE, EMBASE, ISI Web of Knowledge, Scopus, and Google-Scholar databases were searched till June 2017 using different combinations of the following key words: bone-to-implant contact, coating, implant surface, osseointegration, and osteoporosis. Letters to the Editor, review articles, case-reports/case-series, and commentaries were excluded. RESULTS Six animal studies were included, in which osteoporosis was induced by bilateral ovariectomy. In all studies, implant surface roughness was increased by various osteogenic surface coatings including alumina, hydroxyapatite, calcium phosphate, and zoledronic acid. Five studies showed that bone volume and BIC are significantly higher around implants with coated surfaces than noncoated implants. In 1 study, there was no difference in BIC around coated and noncoated implants. CONCLUSION Although experimental studies have shown that osteogenic coatings are effective in enhancing BIC, their clinical relevance requires further investigations.
Collapse
|
38
|
Tu C, Allen A, Deng W, Conroy O, Nambiar M, Zoldan J. Commonly used thiol-containing antioxidants reduce cardiac differentiation and alter gene expression ratios of sarcomeric isoforms. Exp Cell Res 2018; 370:150-159. [PMID: 29920245 DOI: 10.1016/j.yexcr.2018.06.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 06/07/2018] [Accepted: 06/15/2018] [Indexed: 01/31/2023]
Abstract
Reactive oxygen species (ROS) scavengers such as beta-mercaptoethanol (BME) and monothiol glycerol (MTG) are extensively used in stem cell research to prevent cellular oxidative stress. However, how these antioxidant supplements impact stem cell cardiac differentiation, a process regulated by redox-signaling remains unknown. In this study, we found that removal of BME from the conventional high-glucose, serum-based differentiation medium improved cardiac differentiation efficiency by 2-3 fold. BME and MTG treatments during differentiation significantly reduced mRNA expression of cardiac progenitor markers (NKX2.5 and ISL1) as well as sarcomeric markers (MLC2A, MLC2V, TNNI3, MYH6 and MYH7), suggesting reduced cardiomyogenesis by BME or MTG. Moreover, BME and MTG altered the expression ratios between the sarcomeric isoforms. In particular, TNNI3 to TNNI1 ratio and MLC2V to MLC2A ratio were significantly lower in BME or MTG treated cells than untreated cells, implying altered cardiomyocyte phenotype and maturity. Lastly, BME and MTG treatments resulted in less frequent beating, slower contraction and relaxation velocities than untreated cells. Interestingly, none of the above-mentioned effects was observed with Trolox, a non-thiol based antioxidant, despite its strong antioxidant activity. This work demonstrates that commonly used antioxidant supplements may cause considerable changes to cellular redox state and the outcome of differentiation.
Collapse
Affiliation(s)
- Chengyi Tu
- Department of Biomedical Engineering, University of Texas at Austin, United States
| | - Alicia Allen
- Department of Biomedical Engineering, University of Texas at Austin, United States
| | - Wei Deng
- Department of Biomedical Engineering, University of Texas at Austin, United States
| | - Olivia Conroy
- Department of Biomedical Engineering, University of Texas at Austin, United States
| | - Madhavi Nambiar
- Department of Biomedical Engineering, University of Texas at Austin, United States
| | - Janet Zoldan
- Department of Biomedical Engineering, University of Texas at Austin, United States.
| |
Collapse
|
39
|
Tsai CC, Lin YJ, Yu HR, Sheen JM, Tain YL, Huang LT, Tiao MM. Melatonin alleviates liver steatosis induced by prenatal dexamethasone exposure and postnatal high-fat diet. Exp Ther Med 2018; 16:917-924. [PMID: 30112044 DOI: 10.3892/etm.2018.6256] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 09/13/2017] [Indexed: 12/12/2022] Open
Abstract
Prenatal exposure to glucocorticoids is associated with negative health consequences for the offspring that persist into adulthood, including liver steatosis. Melatonin has previously been demonstrated to suppress liver steatosis and oxidative stress in humans with non-alcoholic fatty liver disease and in animal models of obesity. The present study aimed to determine whether melatonin protects against liver steatosis induced by prenatal dexamethasone exposure followed by postnatal high-fat diet. Pregnant Sprague-Dawley rats at gestational days 14-21 were administered dexamethasone (0.1 mg/kg/day) or saline via intraperitoneal injection. The offspring were then divided into five groups, as follows: Vehicle, postnatal high-fat diet (VHF), prenatal dexamethasone exposure (DEX), prenatal dexamethasone exposure + postnatal high-fat diet (DHF), and prenatal dexamethasone exposure + postnatal high-fat diet + melatonin (DHFM) group. Following vehicle or dexamethasone exposure of the maternal rats, the offspring rats in the VHF, DHF and DHFM groups received a high-fat diet (58% fat) between weaning and 6 months of age. In the DHFM group, melatonin was administered to the mothers from gestational days 14-21 until weaning. The offspring continued to receive melatonin until they were sacrificed at 6 months old. Oil Red O staining demonstrated stronger intensity in the DHF group compared with that in the other four groups. Western blot analysis also revealed higher levels of cleaved caspase-3, tumor necrosis factor-α (TNF-α), suppressor of cytokine signaling 3 (SOCS3) and malondialdehyde (MDA), as well as reduced expression of manganese superoxide dismutase (MnSOD) and phosphoinositide 3-kinase (PI3K) in the DHF group compared with the vehicle and DHFM groups. In addition, melatonin reduced the Oil Red O staining intensity and the levels of cleaved caspase-3, TNF-α, SOCS3 and MDA, while it increased the MnSOD and PI3K levels, in the DHFM group compared with the DHF group. In conclusion, postnatal high-fat diet aggravated the prenatal dexamethasone-induced liver steatosis in adult rat offspring via inflammation, oxidative stress and cellular apoptosis, which may be ameliorated by prenatal melatonin therapy.
Collapse
Affiliation(s)
- Ching-Chou Tsai
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Kaohsiung 83301, Taiwan, R.O.C.,Department of Obstetrics and Gynecology, Chiayi Chang Gung Memorial Hospital, Chiayi 61363, Taiwan, R.O.C
| | - Yu-Ju Lin
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Kaohsiung 83301, Taiwan, R.O.C
| | - Hong-Ren Yu
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Kaohsiung 83301, Taiwan, R.O.C
| | - Jiunn-Ming Sheen
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Kaohsiung 83301, Taiwan, R.O.C
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Kaohsiung 83301, Taiwan, R.O.C.,Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Kaohsiung 83301, Taiwan, R.O.C
| | - Li-Tung Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Kaohsiung 83301, Taiwan, R.O.C
| | - Mao-Meng Tiao
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Kaohsiung 83301, Taiwan, R.O.C
| |
Collapse
|
40
|
Samanta T, Sharma P, Kukri D, Kar S. Decoding the regulatory mechanism of glucose and insulin induced phosphatidylinositol 3,4,5-trisphosphate dynamics in β-cells. MOLECULAR BIOSYSTEMS 2018. [PMID: 28636047 DOI: 10.1039/c7mb00227k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In MIN6 pancreatic β-cells, glucose and insulin act in a synergistic manner to regulate the dynamics of Phosphatidylinositol (3,4,5)-trisphosphate (PIP3). However, the precise regulatory mechanism behind such an experimentally observed synergy is poorly understood. In this article, we propose a phenomenological mathematical model for studying the glucose and insulin driven PIP3 activation dynamics under various stimulatory conditions to unfold the mechanism responsible for the observed synergy. The modeling study reveals that the experimentally observed oscillation in PIP3 dynamics with disparate time scales for different external glucose doses is mainly orchestrated by the complex dynamic regulation of cytosolic Ca2+ in β-cells. The model accounts for the dose-dependent activation of PIP3 as a function of externally added insulin, and further shows that even in the absence of Ca2+ signaling, externally added glucose can still maintain a basal level of endogenous insulin secretion via the fatty acid metabolism pathway. Importantly, the model analysis suggests that the glucose mediated ROS (reactive oxygen species) activation often contributes considerably to the synergistic activation of PIP3 by glucose and insulin in a context dependent manner. Under the physiological conditions that keep β-cells in an insulin responsive state, the effect of glucose induced ROS signaling plays a moderate role in PIP3 activation. As β-cells approach an insulin resistant state, the glucose induced ROS signaling significantly affects the PIP3 dynamics. Our findings provide a plausible mechanistic insight into the experimentally observed synergy, and can lead to novel therapeutic strategies.
Collapse
Affiliation(s)
- Tagari Samanta
- Department of Chemistry, IIT Bombay, Powai, Mumbai - 400076, India.
| | - Peeyush Sharma
- Department of Chemistry, IIT Bombay, Powai, Mumbai - 400076, India.
| | - Dwijendra Kukri
- Department of Chemistry, IIT Bombay, Powai, Mumbai - 400076, India.
| | - Sandip Kar
- Department of Chemistry, IIT Bombay, Powai, Mumbai - 400076, India.
| |
Collapse
|
41
|
Sun M, Chi G, Xu J, Tan Y, Xu J, Lv S, Xu Z, Xia Y, Li L, Li Y. Extracellular matrix stiffness controls osteogenic differentiation of mesenchymal stem cells mediated by integrin α5. Stem Cell Res Ther 2018; 9:52. [PMID: 29490668 PMCID: PMC5831741 DOI: 10.1186/s13287-018-0798-0] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 01/18/2018] [Accepted: 02/05/2018] [Indexed: 01/12/2023] Open
Abstract
Background Human mesenchymal stem cell (hMSC) differentiation into osteoblasts has important clinical significance in treating bone injury, and the stiffness of the extracellular matrix (ECM) has been shown to be an important regulatory factor for hMSC differentiation. The aim of this study was to further delineate how matrix stiffness affects intracellular signaling through integrin α5/β1, FAK, and Wnt signaling, subsequently regulating the osteogenic phenotype of hMSCs. Methods hMSCs were cultured on tunable polyacrylamide hydrogels coated with fibronectin with stiffness corresponding to a Young’s modulus of 13–16 kPa and 62–68 kPa. After hMSCs were cultured on gels for 1 week, gene expression of alpha-1typeIcollagen, BGLAP, and RUNX2 were evaluated by real-time PCR. After hMSCs were cultured on gels for 24 h, signaling molecules relating to integrin α5 (FAK, ERK, p-ERK, Akt, p-Akt, GSK-3β, p-GSK-3β, and β-catenin) were evaluated by western blot analysis. Results Osteogenic differentiation was increased on 62–68 kPa ECM, as evidenced by alpha-1 type I collagen, BGLAP, and RUNX2 gene expression, calcium deposition, and ALP staining. In the process of differentiation, gene and protein expression of integrin α5/β1 increased, together with protein expression of the downstream signaling molecules FAK, p-ERK, p-Akt, GSK-3β, p-GSK-3β, and β-catenin, indicating that these molecules can affect the osteogenic differentiation of hMSCs. An antibody blocking integrin α5 suppressed the stiffness-induced expression of all osteoblast markers examined. In particular, alpha-1 type I collagen, RUNX2, and BGLAP were significantly downregulated, indicating that integrin α5 regulates hMSC osteogenic differentiation. Downstream expression of FAK, ERK, p-ERK, and β-catenin protein was unchanged, whereas Akt, p-Akt, GSK-3β, and p-GSK-3β were upregulated. Moreover, expression of Akt and p-Akt was upregulated with anti-integrin α5 antibody, but no difference was observed for FAK, ERK, and p-ERK between the with or without anti-integrin α5 antibody groups. At the same time, expression of GSK-3β and p-GSK-3β was upregulated and β-catenin levels showed no difference between the groups with or without anti-integrin α5 antibody. Since Akt, p-Akt, GSK-3β, and p-GSK-3β displayed the same changes between the groups with or without anti-integrin α5 antibody, we then detected the links among them. Expression of p-Akt and p-GSK-3β was reduced effectively in the presence of the Akt inhibitor Triciribine. However, Akt, GSK-3β, and β-catenin were unchanged. These results suggested that expression of p-GSK-3β was regulated by p-Akt on 62–68 kPa ECM. Conclusions Taken together, our results provide evidence that matrix stiffness (62–68 kPa) affects the osteogenic outcome of hMSCs through mechanotransduction events that are mediated by integrin α5. Electronic supplementary material The online version of this article (10.1186/s13287-018-0798-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Meiyu Sun
- The Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, 130021, People's Republic of China
| | - Guangfan Chi
- The Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, 130021, People's Republic of China
| | - Juanjuan Xu
- The Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, 130021, People's Republic of China
| | - Ye Tan
- The Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, 130021, People's Republic of China
| | - Jiayi Xu
- The Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, 130021, People's Republic of China
| | - Shuang Lv
- The Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, 130021, People's Republic of China
| | - Ziran Xu
- The Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, 130021, People's Republic of China
| | - Yuhan Xia
- The Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, 130021, People's Republic of China
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, 130021, People's Republic of China.
| | - Yulin Li
- The Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, 130021, People's Republic of China.
| |
Collapse
|
42
|
Ma XY, Feng YF, Wang TS, Lei W, Li X, Zhou DP, Wen XX, Yu HL, Xiang LB, Wang L. Involvement of FAK-mediated BMP-2/Smad pathway in mediating osteoblast adhesion and differentiation on nano-HA/chitosan composite coated titanium implant under diabetic conditions. Biomater Sci 2018; 6:225-238. [PMID: 29231215 DOI: 10.1039/c7bm00652g] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nanophase HA/CS composite coated porous titanium implant exhibited superior biological performance under diabetic conditions compared to pure Ti.
Collapse
Affiliation(s)
- Xiang-Yu Ma
- Department of Orthopedics
- General Hospital of Shenyang Military Area Command of Chinese PLA
- Shenyang
- China
- Department of Orthopedics of the 463 Hospital of PLA
| | - Ya-Fei Feng
- Department of Orthopedics
- Xijing Hospital
- Fourth Military Medical University
- Xi'an
- China
| | - Tian-Sheng Wang
- Department of Orthopedics of the 463 Hospital of PLA
- Shenyang
- China
| | - Wei Lei
- Department of Orthopedics
- Xijing Hospital
- Fourth Military Medical University
- Xi'an
- China
| | - Xiang Li
- School of Mechanical Engineering
- Shanghai Jiao Tong University
- State Key Laboratory of Mechanical System and Vibration
- Shanghai
- China
| | - Da-Peng Zhou
- Department of Orthopedics
- General Hospital of Shenyang Military Area Command of Chinese PLA
- Shenyang
- China
| | - Xin-Xin Wen
- Department of Orthopedics of the 463 Hospital of PLA
- Shenyang
- China
- Department of Orthopedics
- Xijing Hospital
| | - Hai-Long Yu
- Department of Orthopedics
- General Hospital of Shenyang Military Area Command of Chinese PLA
- Shenyang
- China
| | - Liang-Bi Xiang
- Department of Orthopedics
- General Hospital of Shenyang Military Area Command of Chinese PLA
- Shenyang
- China
| | - Lin Wang
- Department of Orthopedics
- Xijing Hospital
- Fourth Military Medical University
- Xi'an
- China
| |
Collapse
|
43
|
Hyperglycemia Augments the Adipogenic Transdifferentiation Potential of Tenocytes and Is Alleviated by Cyclic Mechanical Stretch. Int J Mol Sci 2017; 19:ijms19010090. [PMID: 29283422 PMCID: PMC5796040 DOI: 10.3390/ijms19010090] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 12/25/2017] [Accepted: 12/26/2017] [Indexed: 12/19/2022] Open
Abstract
Diabetes mellitus is associated with damage to tendons, which may result from cellular dysfunction in response to a hyperglycemic environment. Tenocytes express diminished levels of tendon-associated genes under hyperglycemic conditions. In contrast, mechanical stretch enhances tenogenic differentiation. However, whether hyperglycemia increases the non-tenogenic differentiation potential of tenocytes and whether this can be mitigated by mechanical stretch remains elusive. We explored the in vitro effects of high glucose and mechanical stretch on rat primary tenocytes. Specifically, non-tenogenic gene expression, adipogenic potential, cell migration rate, filamentous actin expression, and the activation of signaling pathways were analyzed in tenocytes treated with high glucose, followed by the presence or absence of mechanical stretch. We analyzed tenocyte phenotype in vivo by immunohistochemistry using an STZ (streptozotocin)-induced long-term diabetic mouse model. High glucose-treated tenocytes expressed higher levels of the adipogenic transcription factors PPARγ and C/EBPs. PPARγ was also highly expressed in diabetic tendons. In addition, increased adipogenic differentiation and decreased cell migration induced by high glucose implicated a fibroblast-to-adipocyte phenotypic change. By applying mechanical stretch to tenocytes in high-glucose conditions, adipogenic differentiation was repressed, while cell motility was enhanced, and fibroblastic morphology and gene expression profiles were strengthened. In part, these effects resulted from a stretch-induced activation of ERK (extracellular signal-regulated kinases) and a concomitant inactivation of Akt. Our results show that mechanical stretch alleviates the augmented adipogenic transdifferentiation potential of high glucose-treated tenocytes and helps maintain their fibroblastic characteristics. The alterations induced by high glucose highlight possible pathological mechanisms for diabetic tendinopathy. Furthermore, the beneficial effects of mechanical stretch on tenocytes suggest that an appropriate physical load possesses therapeutic potential for diabetic tendinopathy.
Collapse
|
44
|
Wang R, Gao D, Zhou Y, Chen L, Luo B, Yu Y, Li H, Hu J, Huang Q, He M, Peng W, Luo D. High glucose impaired estrogen receptor alpha signaling via β-catenin in osteoblastic MC3T3-E1. J Steroid Biochem Mol Biol 2017; 174:276-283. [PMID: 29030155 DOI: 10.1016/j.jsbmb.2017.10.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 10/07/2017] [Accepted: 10/09/2017] [Indexed: 01/09/2023]
Abstract
Diabetic Mellitus is a risk factor for osteoporosis. It has been suggested that altered estrogen or estrogen receptor α/β (ERα/β) signaling may be involved in diabetic osteoporosis. The present study is to investigate the effects of high glucose on ERα/β signaling in osteoblastic MC3T3-E1 and how the altered signaling of ERα/β affect osteoblastic bone formation. ERα/β signaling was demonstrated as ERα/β protein expression (Western Blotting) and ER transcription activity (Luciferase Reporter assays). Proliferation (WSK-1 assaying), differentiation (ALP staining) and mineralization (Alizalard Red staining) of MC3T3-E1 were examined to evaluate bone formation function. It has been found that high glucose increased ERα/β expression dose-dependently and time-dependently, but high glucose (33mM) decreased ERα transcription activity. 17β-estradiol increased the ERα/β expression dose-dependently in normal medium, but decreased the ERα/β expression dose-dependently in medium with high glucose (33mM). High glucose decreased bone formation and also decreased the osteogenic effects of 17β-estradiol (10-8M). High glucose decreased β-catenin expression dose-dependently and time-dependently. LiCl, an inhibitor of β-catenin degradation, decreased ERα expression but increased ERα transcription activity. When compared with high glucose treatment, LiCl (5mM) increased ALP activity and calcified nodes. Besides, high glucose also decreased the protein expression PI-3K, pAKT/AKT, GSK-3β. In conclusion, the present study suggested that high glucose may impair ERα transcription activity by inhibiting β-catenin signaling in osteoblastic MC3T3-E1, leading decreased bone formation ligand-dependently or ligand-independently.
Collapse
Affiliation(s)
- Rui Wang
- School of Pharmaceutics, Jiangxi Medical College, Nanchang University, Bayi Road 461, Nanchang, Jiangxi Province, 330006, PR China
| | - Dong Gao
- School of Pharmaceutics, Jiangxi Medical College, Nanchang University, Bayi Road 461, Nanchang, Jiangxi Province, 330006, PR China
| | - Yin Zhou
- School of Pharmaceutics, Jiangxi Medical College, Nanchang University, Bayi Road 461, Nanchang, Jiangxi Province, 330006, PR China
| | - Lu Chen
- School of Pharmaceutics, Jiangxi Medical College, Nanchang University, Bayi Road 461, Nanchang, Jiangxi Province, 330006, PR China
| | - Bin Luo
- School of Pharmaceutics, Jiangxi Medical College, Nanchang University, Bayi Road 461, Nanchang, Jiangxi Province, 330006, PR China
| | - Yanrong Yu
- School of Pharmaceutics, Jiangxi Medical College, Nanchang University, Bayi Road 461, Nanchang, Jiangxi Province, 330006, PR China
| | - Hao Li
- School of Pharmaceutics, Jiangxi Medical College, Nanchang University, Bayi Road 461, Nanchang, Jiangxi Province, 330006, PR China
| | - Jiawei Hu
- School of Pharmaceutics, Jiangxi Medical College, Nanchang University, Bayi Road 461, Nanchang, Jiangxi Province, 330006, PR China
| | - Qiren Huang
- School of Pharmaceutics, Jiangxi Medical College, Nanchang University, Bayi Road 461, Nanchang, Jiangxi Province, 330006, PR China
| | - Ming He
- School of Pharmaceutics, Jiangxi Medical College, Nanchang University, Bayi Road 461, Nanchang, Jiangxi Province, 330006, PR China
| | - Weijie Peng
- School of Pharmaceutics, Jiangxi Medical College, Nanchang University, Bayi Road 461, Nanchang, Jiangxi Province, 330006, PR China; Jiangxi Academy of Medical Science, Bayi Road 461, Nanchang, Jiangxi Province, 330006, PR China.
| | - Dan Luo
- Department of Physiology, School of Basic Medicine, Jiangxi Medical College, Nanchang University, Bayi Road 461, Nanchang, Jiangxi Province, 330006, PR China.
| |
Collapse
|
45
|
Simon F, Tapia P, Armisen R, Echeverria C, Gatica S, Vallejos A, Pacheco A, Sanhueza ME, Alvo M, Segovia E, Torres R. Human Peritoneal Mesothelial Cell Death Induced by High-Glucose Hypertonic Solution Involves Ca 2+ and Na + Ions and Oxidative Stress with the Participation of PKC/NOX2 and PI3K/Akt Pathways. Front Physiol 2017; 8:379. [PMID: 28659813 PMCID: PMC5468383 DOI: 10.3389/fphys.2017.00379] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 05/22/2017] [Indexed: 01/07/2023] Open
Abstract
Chronic peritoneal dialysis (PD) therapy is equally efficient as hemodialysis while providing greater patient comfort and mobility. Therefore, PD is the treatment of choice for several types of renal patients. During PD, a high-glucose hyperosmotic (HGH) solution is administered into the peritoneal cavity to generate an osmotic gradient that promotes water and solutes transport from peritoneal blood to the dialysis solution. Unfortunately, PD has been associated with a loss of peritoneal viability and function through the generation of a severe inflammatory state that induces human peritoneal mesothelial cell (HPMC) death. Despite this deleterious effect, the precise molecular mechanism of HPMC death as induced by HGH solutions is far from being understood. Therefore, the aim of this study was to explore the pathways involved in HGH solution-induced HPMC death. HGH-induced HPMC death included influxes of intracellular Ca2+ and Na+. Furthermore, HGH-induced HPMC death was inhibited by antioxidant and reducing agents. In line with this, HPMC death was induced solely by increased oxidative stress. In addition to this, the cPKC/NOX2 and PI3K/Akt intracellular signaling pathways also participated in HGH-induced HPMC death. The participation of PI3K/Akt intracellular is in agreement with previously shown in rat PMC apoptosis. These findings contribute toward fully elucidating the underlying molecular mechanism mediating peritoneal mesothelial cell death induced by high-glucose solutions during peritoneal dialysis.
Collapse
Affiliation(s)
- Felipe Simon
- Departamento de Ciencias Biologicas, Facultad de Ciencias Biologicas and Facultad de Medicina, Universidad Andres BelloSantiago, Chile.,Millennium Institute on Immunology and ImmunotherapySantiago, Chile
| | - Pablo Tapia
- Unidad de Paciente Critico, Hospital Clínico Metropolitano de La FloridaSantiago, Chile
| | - Ricardo Armisen
- Centro de Investigación y Tratamiento del Cancer, Facultad de Medicina, Universidad de ChileSantiago, Chile.,Center for Excellence in Precision Medicine Pfizer, Pfizer ChileSantiago, Chile
| | - Cesar Echeverria
- Centro Integrativo de Biología y Química Aplicada, Universidad Bernardo OHigginsSantiago, Chile
| | - Sebastian Gatica
- Departamento de Ciencias Biologicas, Facultad de Ciencias Biologicas and Facultad de Medicina, Universidad Andres BelloSantiago, Chile
| | - Alejandro Vallejos
- Departamento de Ciencias Biologicas, Facultad de Ciencias Biologicas and Facultad de Medicina, Universidad Andres BelloSantiago, Chile
| | - Alejandro Pacheco
- Sección de Nefrología, Departamento de Medicina, Hospital Clínico Universidad de ChileSantiago, Chile
| | - Maria E Sanhueza
- Sección de Nefrología, Departamento de Medicina, Hospital Clínico Universidad de ChileSantiago, Chile
| | - Miriam Alvo
- Sección de Nefrología, Departamento de Medicina, Hospital Clínico Universidad de ChileSantiago, Chile
| | - Erico Segovia
- Centro Integrativo de Biología y Química Aplicada, Universidad Bernardo OHigginsSantiago, Chile
| | - Rubén Torres
- Sección de Nefrología, Departamento de Medicina, Hospital Clínico Universidad de ChileSantiago, Chile.,Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de ChileSantiago, Chile
| |
Collapse
|
46
|
Li G, Ning C, Ma Y, Jin L, Tang Q, Li X, Li M, Liu H. miR-26b Promotes 3T3-L1 Adipocyte Differentiation Through Targeting PTEN. DNA Cell Biol 2017; 36:672-681. [PMID: 28570839 DOI: 10.1089/dna.2017.3712] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
microRNAs (miRNAs) play important roles in adipogenesis that is closely linked to obesity and energy homeostasis. Thus far, only a few miRNAs have been identified to regulate adipocyte development, arousing interest in the detailed function of miRNAs during adipogenesis. In this study, we found that the miR-26b expression showed an increasing trend during 3T3-L1 cells differentiation. To investigate the role of miR-26b in adipogenesis, the synthetic miR-26b agomirs and antagomirs were used to perform overexpression and knockdown experiment, respectively. Our data revealed that overexpression of miR-26b significantly accelerated the mRNA expression of the adipogenic markers, peroxisome proliferator-activated receptor gamma (PPARγ), fatty acid synthase (FAS), CCAAT/enhancer binding protein alpha (C/EBPα), and lipoprotein lipase, and the protein level of PPARγ and FAS. miR-26b overexpression also resulted in a significant increase in lipid accumulation. In contrast, inhibition of miR-26b expression decreased differentiation of 3T3-L1 cells. By target gene prediction and luciferase reporter assay, we demonstrated that miR-26b may directly bind to the 3' UTR of phosphatase and tensin homolog (PTEN). Taken together, these results demonstrate that miR-26b might participate in regulating adipogenic differentiation in 3T3-L1 cells by inhibiting the PTEN expression, further highlighting the importance of miRNA in adipogenesis.
Collapse
Affiliation(s)
- Guilin Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University , Chengdu, People's Republic of China
| | - Chunyou Ning
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University , Chengdu, People's Republic of China
| | - Yao Ma
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University , Chengdu, People's Republic of China
| | - Long Jin
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University , Chengdu, People's Republic of China
| | - Qianzi Tang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University , Chengdu, People's Republic of China
| | - Xuewei Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University , Chengdu, People's Republic of China
| | - Mingzhou Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University , Chengdu, People's Republic of China
| | - Haifeng Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University , Chengdu, People's Republic of China
| |
Collapse
|
47
|
Silva E, Greene AF, Strauss K, Herrick JR, Schoolcraft WB, Krisher RL. Antioxidant supplementation during in vitro culture improves mitochondrial function and development of embryos from aged female mice. Reprod Fertil Dev 2017; 27:975-83. [PMID: 25739837 DOI: 10.1071/rd14474] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 01/20/2015] [Indexed: 12/31/2022] Open
Abstract
Maternal aging results in reduced oocyte and blastocyst quality, thought to be due, in part, to mitochondrial dysfunction and accumulation of reactive oxygen species. To reduce oxidative stress, the antioxidants α-lipoic acid (ALA; 10µM), α-tocopherol (250µM), hypotaurine (1mM) and N-acetylcysteine (NAC; 1mM), and sirtuin (100ngmL(-1)) were added to embryo culture medium (AntiOX) and compared with a control (CON) without antioxidants to assess blastocyst development after in vitro maturation and fertilisation of oocytes from aged B6D2F1 female mice (13.5 months). Development to the blastocyst stage increased in the AntiOX compared with CON group (87.6% vs 72.7%, respectively; P<0.01), in addition to higher mitochondrial membrane potential and ATP levels in the AntiOX group. Expression of genes associated with oxidative stress (PI3K, FOXO3A and GLRX2) was upregulated in the CON compared with AntiOX group. In addition to AntiOX, a medium containing only NAC and ALA (rAntiOX) was used to culture embryos from young CF1 females (6-8 weeks). More blastocysts developed in the rAntiOX compared with CON group (64.1% vs 43.3%, respectively; P<0.01), although AntiOX (48.0% blastocysts) did not result in improved development in young mice. Antioxidants improved mitochondrial activity, gene expression and development in embryos of older female mice, whereas a reduced level of antioxidants during culture was beneficial to embryos from young mice.
Collapse
Affiliation(s)
- Elena Silva
- National Foundation for Fertility Research, 10290 RidgeGate Cr., Lone Tree, CO 80124, USA
| | - Alison F Greene
- National Foundation for Fertility Research, 10290 RidgeGate Cr., Lone Tree, CO 80124, USA
| | - Kevin Strauss
- National Foundation for Fertility Research, 10290 RidgeGate Cr., Lone Tree, CO 80124, USA
| | - Jason R Herrick
- National Foundation for Fertility Research, 10290 RidgeGate Cr., Lone Tree, CO 80124, USA
| | - William B Schoolcraft
- Colorado Center for Reproductive Medicine, 10290 RidgeGate Cr., Lone Tree, CO 80124, USA
| | - Rebecca L Krisher
- National Foundation for Fertility Research, 10290 RidgeGate Cr., Lone Tree, CO 80124, USA
| |
Collapse
|
48
|
Zhang C, Li C, Chen S, Li Z, Jia X, Wang K, Bao J, Liang Y, Wang X, Chen M, Li P, Su H, Wan JB, Lee SMY, Liu K, He C. Berberine protects against 6-OHDA-induced neurotoxicity in PC12 cells and zebrafish through hormetic mechanisms involving PI3K/AKT/Bcl-2 and Nrf2/HO-1 pathways. Redox Biol 2017; 11:1-11. [PMID: 27835779 PMCID: PMC5107737 DOI: 10.1016/j.redox.2016.10.019] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 10/29/2016] [Accepted: 10/31/2016] [Indexed: 11/10/2022] Open
Abstract
Berberine (BBR) is a renowned natural compound that exhibits potent neuroprotective activities. However, the cellular and molecular mechanisms are still unclear. Hormesis is an adaptive mechanism generally activated by mild oxidative stress to protect the cells from further damage. Many phytochemicals have been shown to induce hormesis. This study aims to investigate whether the neuroprotective activity of BBR is mediated by hormesis and the related signaling pathways in 6-OHDA-induced PC12 cells and zebrafish neurotoxic models. Our results demonstrated that BBR induced a typical hormetic response in PC12 cells, i.e. low dose BBR significantly increased the cell viability, while high dose BBR inhibited the cell viability. Moreover, low dose BBR protected the PC12 cells from 6-OHDA-induced cytotoxicity and apoptosis, whereas relatively high dose BBR did not show neuroprotective activity. The hormetic and neuroprotective effects of BBR were confirmed to be mediated by up-regulated PI3K/AKT/Bcl-2 cell survival and Nrf2/HO-1 antioxidative signaling pathways. In addition, low dose BBR markedly mitigated the 6-OHDA-induced dopaminergic neuron loss and behavior movement deficiency in zebrafish, while high dose BBR only slightly exhibited neuroprotective activities. These results strongly suggested that the neuroprotection of BBR were attributable to the hormetic mechanisms via activating cell survival and antioxidative signaling pathways.
Collapse
Affiliation(s)
- Chao Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Chuwen Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Shenghui Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; Lee's Pharmaceutical (Hong Kong) Ltd., Shatin, Hong Kong 999077, China
| | - Zhiping Li
- Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, Shandong Provincial Key Laboratory for Biosensor, Biology Institute of Shandong Academy of Sciences, Jinan 250014, China
| | - Xuejing Jia
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Kai Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Jiaolin Bao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Yeer Liang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Xiaotong Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Peng Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Jian-Bo Wan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Simon Ming Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Kechun Liu
- Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, Shandong Provincial Key Laboratory for Biosensor, Biology Institute of Shandong Academy of Sciences, Jinan 250014, China.
| | - Chengwei He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China.
| |
Collapse
|
49
|
Morris G, Walder K, McGee SL, Dean OM, Tye SJ, Maes M, Berk M. A model of the mitochondrial basis of bipolar disorder. Neurosci Biobehav Rev 2017; 74:1-20. [PMID: 28093238 DOI: 10.1016/j.neubiorev.2017.01.014] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 01/08/2017] [Accepted: 01/10/2017] [Indexed: 12/11/2022]
|
50
|
Pandey A, Raj P, Goru SK, Kadakol A, Malek V, Sharma N, Gaikwad AB. Esculetin ameliorates hepatic fibrosis in high fat diet induced non-alcoholic fatty liver disease by regulation of FoxO1 mediated pathway. Pharmacol Rep 2017; 69:666-672. [PMID: 28527877 DOI: 10.1016/j.pharep.2017.02.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 01/20/2017] [Accepted: 02/03/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD), a chronic metabolic disorder is associated with oxidative stress, inflammation and fibrotic cascades. In this study, we aimed to examine the effects of Esculetin, a well-known anti-oxidant on TGF-β1 mediated liver fibrosis and FoxO1 activity. METHODS A non-genetic murine model for NAFLD was developed by chronic high fat diet (HFD) (58% calories from fats) feeding in Wistar rats. The plasma biochemical parameters, liver function tests, oxidative stress, and histopathological alterations were assessed. The alterations in extracellular matrix (ECM) deposition and FoxO1 activity were assessed by immunohistochemistry. RESULTS The aberrations in plasma parameters, liver functioning, morphometric and microscopic changes in liver structure of HFD fed rats were significantly improved by treatment with Esculetin. Liver fibrosis, identified in the form of collagen deposition and expression of fibrotic proteins like TGF-β1 and fibronectin was also markedly controlled by Esculetin. The expression of phospho-FoxO1 was found to be reduced in HFD fed rats' liver, showing an increase in activation of FoxO1 under insulin resistant and hyperglycemic states. Esculetin treatment could improve phospho-FoxO1 expression, thus showing its ability to act on Akt/PI3K/FoxO1 pathway. CONCLUSIONS As per the previous studies, a potential therapy for NAFLD may be the one with multi-faceted actions on insulin resistance, oxidative stress, inflammation and fibrosis. This study demonstrates the efficiency of Esculetin in improving liver fibrosis in HFD induced NAFLD.
Collapse
Affiliation(s)
- Anuradha Pandey
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, India
| | - Priyank Raj
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, India
| | - Santosh Kumar Goru
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, India
| | - Almesh Kadakol
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, India
| | - Vajir Malek
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, India
| | - Nisha Sharma
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, India
| | - Anil Bhanudas Gaikwad
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, India.
| |
Collapse
|