1
|
Cheng EY, Mirzaei A. Potential molecular targets for the pharmacologic management of non-traumatic osteonecrosis. Expert Opin Ther Targets 2024; 28:991-1000. [PMID: 39469902 DOI: 10.1080/14728222.2024.2421755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 10/23/2024] [Indexed: 10/30/2024]
Abstract
INTRODUCTION Non-traumatic osteonecrosis is a debilitating condition marked by bone death, primarily due to reduced blood supply. Currently, no effective pharmacologic intervention is available to manage this condition effectively. AREAS COVERED Lipid metabolic disorders, chronic inflammation, vascular dysfunction, coagulopathy, and impaired bone homeostasis are suggested as the key pathogenic mechanisms involved in the development of non-traumatic osteonecrosis. Targeting any of these dysfunctions offers a potential avenue for pharmacologic intervention. However, the potential molecular targets for pharmacologic treatment of non-traumatic osteonecrosis remain underexplored. In this study, we reviewed available databases to compile a comprehensive set of pathogenic mechanisms and corresponding therapeutic targets for non-traumatic osteonecrosis. EXPERT OPINION Evidence suggests that a single pathogenic mechanism cannot fully explain the development of osteonecrosis, supporting the adoption of a multi-pathogenic theory. This theory implies that effective management of non-traumatic osteonecrosis requires targeting multiple pathogenic mechanisms simultaneously. Moreover, the same pathogenic mechanisms are unlikely to explain osteonecrosis development in patients with different etiologies. Consequently, a one-size-fits-all approach to medication is unlikely to be effective across all types of non-traumatic osteonecrosis. Future research should, therefore, focus on developing multi-target pharmacologic treatments tailored to the specific etiology of non-traumatic osteonecrosis.
Collapse
Affiliation(s)
- Edward Y Cheng
- Department of Orthopedic Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Alireza Mirzaei
- Department of Orthopedic Surgery, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
2
|
Wu T, Zhou Y, Shi W, Guo S, Tian H, Li W, Wang Y, Li T. Translational horizons in stem cell therapy for osteonecrosis of the femoral head: a journey from basic research to clinical practice through bibliometric insights. J Transl Med 2024; 22:982. [PMID: 39478610 PMCID: PMC11523765 DOI: 10.1186/s12967-024-05784-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 10/19/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Osteonecrosis of the femoral head (ONFH) significantly impacts young and middle-aged adults, with steroid use implicated in many cases. Traditional treatments have limited efficacy, prompting a shift towards innovative approaches, such as stem cell therapy, offering less invasive regenerative solutions. METHODS Using bibliometric analysis from 1997 to 2023, we identified 392 articles on stem cell therapy for ONFH from the Web of Science Core Collection and analysed them using VOSviewer and CiteSpace to identify key trends and research directions. RESULTS From 1997 to 2023, stem cell therapy for ONFH research expanded significantly, with 392 articles evidencing global collaboration, particularly from China, the United States and South Korea. The field is characterised by 158 core authors across 26 clusters and contributions from 417 institutions in 104 research clusters, with Shanghai Jiao Tong University as a notable leader. This research is disseminated through 23 journal clusters, emphasising interdisciplinary work, with Clinical Orthopaedics and Related Research among the most influential journals. Key findings include the identification of the most influential papers, highlighting advances, such as use of autologous mesenchymal stem cells (MSCs) and innovative delivery mechanisms. High-frequency keyword analysis further mapped the evolution of the field, from basic mechanisms to advanced therapies, underscoring a trend towards more targeted stem cell treatments for ONFH. CONCLUSION Stem cell therapy for ONFH has advanced significantly, showcasing a successful transition from basic research to clinical practice, particularly highlighted by developments in use of autologous MSCs and delivery methods. Future research will focus on refining therapies through exosome technology, targeted modulation of stress and inflammation and integration with surgical techniques, with the aim of tailored patient care and improved ONFH outcomes.
Collapse
Affiliation(s)
- Tingyu Wu
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China
| | - Yinxue Zhou
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Weipeng Shi
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China
| | - Sijia Guo
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China
| | - Hua Tian
- Department of Neurological Rehabilitation, Qingdao Special Servicemen Recuperation Center of PLA Navy, Qingdao, 266000, China
| | - Weiyan Li
- Department of Emergency Surgery and Joint Surgery, Qingdao Third People's Hospital, Qingdao, 266000, China
| | - Yingzhen Wang
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China
| | - Tao Li
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China.
| |
Collapse
|
3
|
Peyravian N, Milan PB, Kebria MM, Mashayekhan S, Ghasemian M, Amiri S, Hamidi M, Shavandi A, Moghtadaei M. Designing and synthesis of injectable hydrogel based on carboxymethyl cellulose/carboxymethyl chitosan containing QK peptide for femoral head osteonecrosis healing. Int J Biol Macromol 2024; 270:132127. [PMID: 38718991 DOI: 10.1016/j.ijbiomac.2024.132127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 05/03/2024] [Accepted: 05/04/2024] [Indexed: 05/18/2024]
Abstract
Femoral head necrosis is a debilitating disorder that typically caused by impaired blood supply to the hip joint. In this study, a novel injectable hydrogel based on Oxidized Carboxymethyl Cellulose (OCMC)-Carboxymethyl Chitosan (CMCS) polymers containing an angiogenesis stimulator peptide (QK) with a non-toxic crosslinking interaction (Schiff based reaction) was synthesized to enhance angiogenesis following femoral head necrosis in an animal model. The physicochemical features of fabricated injectable hydrogel were analyzed by FTIR, swelling and degradation rate, rheometry, and peptide release. Also, the safety and efficacy were evaluated following an in vitro hydrogel injection study and an avascular necrosis (AVN) animal model. According to the results, the hydrogel exhibited an appropriate swelling ratio and water uptake (>90 %, 24 h) as well as a suitable degradation rate over 21 days accompanied by a continuous peptide release. Also, data showed that hydrogels containing QK peptide boosted the proliferation, differentiation, angiogenesis, and osteogenic potential of both Bone Marrow mesenchymal Stem Cells (BM-MSCs) and human umbilical vein endothelial cells (HUVECs) (****p < 0.0001 and ***p < 0.001, respectively). Furthermore, molecular and histological evaluations significantly demonstrated the overexpression of Runx2, Osteocalcin, Collagen I, VEGF and CD34 genes (**p < 0.01 and ***p < 0.001, respectively), and also femoral head necrosis was effectively prohibited, and more blood vessels were detected in defect area by OCMC-CMCS hydrogel containing QK peptide (bone trabeculae >9000, ***p < 0.001). In conclusion, the findings demonstrate that OCMC-CMCS-QK injectable hydrogel could be considered as an impressive therapeutic construct for femoral head AVN healing.
Collapse
Affiliation(s)
- Noshad Peyravian
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Peiman Brouki Milan
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Maziar Malekzadeh Kebria
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shohreh Mashayekhan
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Azadi Ave., Tehran, Iran
| | - Melina Ghasemian
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shayan Amiri
- Shohadaye Haftom-e-tir Hospital, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Hamidi
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles - 3BIO-BioMatter unit, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050 Brussels, Belgium
| | - Amin Shavandi
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles - 3BIO-BioMatter unit, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050 Brussels, Belgium
| | - Mehdi Moghtadaei
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Orthopaedic Department, Hazrat-Rasul Hospital, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Wang Q, Yang Z, Li Q, Zhang W, Kang P. Lithium prevents glucocorticoid-induced osteonecrosis of the femoral head by regulating autophagy. J Cell Mol Med 2024; 28:e18385. [PMID: 38801405 PMCID: PMC11129728 DOI: 10.1111/jcmm.18385] [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: 10/19/2023] [Revised: 04/17/2024] [Accepted: 04/27/2024] [Indexed: 05/29/2024] Open
Abstract
Autophagy may play an important role in the occurrence and development of glucocorticoid-induced osteonecrosis of the femoral head (GC-ONFH). Lithium is a classical autophagy regulator, and lithium can also activate osteogenic pathways, making it a highly promising therapeutic agent for GC-ONFH. We aimed to evaluate the potential therapeutic effect of lithium on GC-ONFH. For in vitro experiments, primary osteoblasts of rats were used for investigating the underlying mechanism of lithium's protective effect on GC-induced autophagy levels and osteogenic activity dysfunction. For in vivo experiments, a rat model of GC-ONFH was used for evaluating the therapeutic effect of oral lithium on GC-ONFH and underlying mechanism. Findings demonstrated that GC over-activated the autophagy of osteoblasts and reduced their osteogenic activity. Lithium reduced the over-activated autophagy of GC-treated osteoblasts through PI3K/AKT/mTOR signalling pathway and increased their osteogenic activity. Oral lithium reduced the osteonecrosis rates in a rat model of GC-ONFH, and restrained the increased expression of autophagy related proteins in bone tissues through PI3K/AKT/mTOR signalling pathway. In conclusion, lithium can restrain over-activated autophagy by activating PI3K/AKT/mTOR signalling pathway and up-regulate the expression of genes for bone formation both in GC induced osteoblasts and in a rat model of GC-ONFH. Lithium may be a promising therapeutic agent for GC-ONFH. However, the role of autophagy in the pathogenesis of GC-ONFH remains controversial. Studies are still needed to further explore the role of autophagy in the pathogenesis of GC-ONFH, and the efficacy of lithium in the treatment of GC-ONFH and its underlying mechanisms.
Collapse
Affiliation(s)
- Qiuru Wang
- Department of Orthopedic Surgery, West China HospitalSichuan UniversityChengduChina
| | - Zhouyuan Yang
- Department of Orthopedic Surgery, West China HospitalSichuan UniversityChengduChina
| | - Qianhao Li
- Department of Orthopedic Surgery, West China HospitalSichuan UniversityChengduChina
| | - Wanli Zhang
- Public Laboratory Technology Center, West China HospitalSichuan UniversityChengduChina
| | - Pengde Kang
- Department of Orthopedic Surgery, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
5
|
Chen J, Jin W, Zhong C, Cai W, Huang L, Zhou J, Peng H. Human umbilical cord mesenchymal stem cells promote steroid-induced osteonecrosis of the femoral head repair by improving microvascular endothelial cell function. Aging (Albany NY) 2024; 16:7928-7945. [PMID: 38696318 PMCID: PMC11132024 DOI: 10.18632/aging.205794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/10/2024] [Indexed: 05/04/2024]
Abstract
Recently, there has been growing interest in using cell therapy through core decompression (CD) to treat osteonecrosis of the femoral head (ONFH). Our study aimed to investigate the effectiveness and mechanism of human umbilical cord mesenchymal stem cells (hUCMSCs) in treating steroid-induced ONFH. We constructed a steroid-induced ONFH rabbit model as well as dexamethasone (Dex)-treated bone microvascular endothelial cells (BMECs) model of human femoral head. We injected hUCMSCs into the rabbit femoral head via CD. The effects of hUCMSCs on steroid-induced ONFH rabbit model and Dex-treated BMECs were evaluated via micro-CT, microangiography, histology, immunohistochemistry, wound healing, tube formation, and western blotting assay. Furthermore, we conducted single-cell RNA sequencing (scRNA-seq) to examine the characteristics of endothelial cells, the activation of signaling pathways, and inter-cellular communication in ONFH. Our data reveal that hUCMSCs improved the femoral head microstructure and bone repair and promoted angiogenesis in the steroid-induced ONFH rabbit model. Importantly, hUCMSCs improved the migration ability and angioplasty of Dex-treated BMECs by secreting COL6A2 to activate FAK/PI3K/AKT signaling pathway via integrin α1β1.
Collapse
Affiliation(s)
- Junwen Chen
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430062, China
| | - Wenyi Jin
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430062, China
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong 999077, Hong Kong Special Administrative Region
| | - Changheng Zhong
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430062, China
| | - Wenxiang Cai
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430062, China
| | - Liangkun Huang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430062, China
| | - Jianlin Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430062, China
| | - Hao Peng
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430062, China
| |
Collapse
|
6
|
Muramoto M, Mineoka N, Fukuda K, Kuriyama S, Masatani T, Fujita A. Coordinated regulation of phosphatidylinositol 4-phosphate and phosphatidylserine levels by Osh4p and Osh5p is an essential regulatory mechanism in autophagy. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2024; 1866:184308. [PMID: 38437942 DOI: 10.1016/j.bbamem.2024.184308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 01/26/2024] [Accepted: 02/26/2024] [Indexed: 03/06/2024]
Abstract
Macroautophagy (hereafter autophagy) is an intracellular degradative pathway in budding yeast cells. Certain lipid types play essential roles in autophagy; yet the precise mechanisms regulating lipid composition during autophagy remain unknown. Here, we explored the role of the Osh family proteins in the modulating lipid composition during autophagy in budding yeast. Our results showed that osh1-osh7∆ deletions lead to autophagic dysfunction, with impaired GFP-Atg8 processing and the absence of autophagosomes and autophagic bodies in the cytosol and vacuole, respectively. Freeze-fracture electron microscopy (EM) revealed elevated phosphatidylinositol 4-phosphate (PtdIns(4)P) levels in cytoplasmic and luminal leaflets of autophagic bodies and vacuolar membranes in all deletion mutants. Phosphatidylserine (PtdSer) levels were significantly decreased in the autophagic bodies and vacuolar membranes in osh4∆ and osh5∆ mutants, whereas no significant changes were observed in other osh deletion mutants. Furthermore, we identified defects in autophagic processes in the osh4∆ and osh5∆ mutants, including rare autophagosome formation in the osh5∆ mutant and accumulation of autophagic bodies in the vacuole in the osh4∆ mutant, even in the absence of the proteinase inhibitor PMSF. These findings suggest that Osh4p and Osh5p play crucial roles in the transport of PtdSer to autophagic bodies and autophagosome membranes, respectively. The precise control of lipid composition in the membranes of autophagosomes and autophagic bodies by Osh4p and Osh5p represents an important regulatory mechanism in autophagy.
Collapse
Affiliation(s)
- Moe Muramoto
- Department of Molecular and Cell Biology and Biochemistry, Basic Veterinary Science, Faculty of Veterinary Medicine, Kagoshima University, Korimoto 1-21-24, Kagoshima 890-0065, Japan
| | - Nanaru Mineoka
- Department of Molecular and Cell Biology and Biochemistry, Basic Veterinary Science, Faculty of Veterinary Medicine, Kagoshima University, Korimoto 1-21-24, Kagoshima 890-0065, Japan
| | - Kayoko Fukuda
- Department of Molecular and Cell Biology and Biochemistry, Basic Veterinary Science, Faculty of Veterinary Medicine, Kagoshima University, Korimoto 1-21-24, Kagoshima 890-0065, Japan
| | - Sayuri Kuriyama
- Department of Molecular and Cell Biology and Biochemistry, Basic Veterinary Science, Faculty of Veterinary Medicine, Kagoshima University, Korimoto 1-21-24, Kagoshima 890-0065, Japan
| | - Tatsunori Masatani
- Laboratory of Zoonotic Diseases, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; Center for One Medicine Innovative Translational Research (COMIT), Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Akikazu Fujita
- Department of Molecular and Cell Biology and Biochemistry, Basic Veterinary Science, Faculty of Veterinary Medicine, Kagoshima University, Korimoto 1-21-24, Kagoshima 890-0065, Japan.
| |
Collapse
|
7
|
Lin Z, Long F, Kang R, Klionsky DJ, Yang M, Tang D. The lipid basis of cell death and autophagy. Autophagy 2024; 20:469-488. [PMID: 37768124 PMCID: PMC10936693 DOI: 10.1080/15548627.2023.2259732] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/25/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
ABBREVIATIONS ACSL: acyl-CoA synthetase long chain family; DISC: death-inducing signaling complex; DAMPs: danger/damage-associated molecular patterns; Dtgn: dispersed trans-Golgi network; FAR1: fatty acyl-CoA reductase 1; GPX4: glutathione peroxidase 4; LPCAT3: lysophosphatidylcholine acyltransferase 3; LPS: lipopolysaccharide; MUFAs: monounsaturated fatty acids; MOMP: mitochondrial outer membrane permeabilization; MLKL, mixed lineage kinase domain like pseudokinase; oxPAPC: oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine; OxPCs: oxidized phosphatidylcholines; PUFAs: polyunsaturated fatty acids; POR: cytochrome p450 oxidoreductase; PUFAs: polyunsaturated fatty acids; RCD: regulated cell death; RIPK1: receptor interacting serine/threonine kinase 1; SPHK1: sphingosine kinase 1; SOAT1: sterol O-acyltransferase 1; SCP2: sterol carrier protein 2; SFAs: saturated fatty acids; SLC47A1: solute carrier family 47 member 1; SCD: stearoyl-CoA desaturase; VLCFA: very long chain fatty acids.
Collapse
Affiliation(s)
- Zhi Lin
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Pediatric Cancer, Changsha, Hunan, China
| | - Fei Long
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Postdoctoral Research Station of Basic Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Daniel J. Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Minghua Yang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Pediatric Cancer, Changsha, Hunan, China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
8
|
Lashgari NA, Roudsari NM, Zadeh SST, Momtaz S, Abbasifard M, Reiner Ž, Abdolghaffari AH, Sahebkar A. Statins block mammalian target of rapamycin pathway: a possible novel therapeutic strategy for inflammatory, malignant and neurodegenerative diseases. Inflammopharmacology 2023; 31:57-75. [PMID: 36574095 PMCID: PMC9792946 DOI: 10.1007/s10787-022-01077-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 07/12/2022] [Indexed: 12/28/2022]
Abstract
Inflammation plays a critical role in several diseases such as cancer, gastric, heart and nervous system diseases. Data suggest that the activation of mammalian target of rapamycin (mTOR) pathway in epithelial cells leads to inflammation. Statins, the inhibitors of the 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA), seem to be able to inhibit the mTOR. Statins are considered to have favorable effects on inflammatory diseases by reducing the complications caused by inflammation and by regulating the inflammatory process and cytokines secretion. This critical review collected data on this topic from clinical, in vivo and in vitro studies published between 1998 and June 2022 in English from databases including PubMed, Google Scholar, Scopus, and Cochrane libraries.
Collapse
Affiliation(s)
- Naser-Aldin Lashgari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nazanin Momeni Roudsari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Saeideh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Tehran, Iran
- Toxicology and Diseases Group (TDG), The Institute of Pharmaceutical Sciences (TIPS), and Faculty of Pharmacy, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mitra Abbasifard
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Internal Medicine, Ali-Ibn Abi-Talib Hospital, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Željko Reiner
- Department of Internal Medicine, School of Medicine, University Hospital Center Zagreb, University of Zagreb, Zagreb, Croatia
| | - Amir Hossein Abdolghaffari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Tehran, Iran.
- Toxicology and Diseases Group (TDG), The Institute of Pharmaceutical Sciences (TIPS), and Faculty of Pharmacy, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences, Tehran, Iran.
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- School of Medicine, The University of Western Australia, Perth, Australia.
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
9
|
Feng G, Zhang P, Huang J, Yu Y, Yang F, Zhao X, Wang W, Li D, Sun S, Niu X, Chai L, Li J. Sequential Release of Panax Notoginseng Saponins and Osteopractic Total Flavone from Poly ( L-Lactic Acid) Scaffold for Treating Glucocorticoid-Associated Osteonecrosis of Femoral Head. J Funct Biomater 2023; 14:jfb14010031. [PMID: 36662078 PMCID: PMC9863477 DOI: 10.3390/jfb14010031] [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: 11/23/2022] [Revised: 12/23/2022] [Accepted: 12/23/2022] [Indexed: 01/06/2023] Open
Abstract
Glucocorticoids inhibit angiogenesis in the femoral head, which fails to nourish the bone tissue and leads to osteonecrosis. Restoring angiogenesis is not only essential for vessel formation, but also crucial for osteogenesis. Poly (L-lactic acid) (PLLA) is commonly used in the bone tissue engineering field. Panax notoginseng saponins (PNS) and osteopractic total flavone (OTF) promote angiogenesis and osteogenesis, respectively. We designed a sequentially releasing PLLA scaffold including PLLA loaded with OTF (inner layer) and PLLA loaded with PNS (outer layer). We assessed the osteogenic effect of angiogenesis in this scaffold by comparing it with the one-layered scaffold (PLLA embedded with OTF and PNS) in vivo. Results from the micro-CT showed that the data of bone mineral density (BMD), bone volume (BV), and percent bone volume (BV/TV) in the PO-PP group were significantly higher than those in the POP group (p < 0.01). Histological analyses show that the PO-PP scaffold exhibits better angiogenic and osteogenic effects compared with the one-layered scaffold. These might result from the different structures between them, where the sequential release of a bi-layer scaffold achieves the osteogenic effect of vascularization by initially releasing PNS in the outer layer. We further explored the possible mechanism by an immunohistochemistry analysis and an immunofluorescence assay. The results showed that the protein expressions of vascular endothelial growth factor (VEGF) and platelet endothelial cell adhesion molecule-1(CD31) in the PO-PP scaffold were significantly higher than those in the POP scaffold (p < 0.01); the protein expressions of osteocalcin (OCN), osteopontin (OPN), and alkaline phosphatase (ALP) in the PO-PP scaffold were significantly higher than those in the POP scaffold (p < 0.05). Upregulating the expressions of angiogenic and osteogenic proteins might be the possible mechanism.
Collapse
Affiliation(s)
- Guiyu Feng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Pingxin Zhang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Jian Huang
- Department of Orthopedic, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yao Yu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Fenghe Yang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Xueqian Zhao
- Yuquan Hospital Affiliated to Tsinghua University, Beijing 100040, China
| | - Wei Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Dongyang Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Song Sun
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Xufeng Niu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
- Correspondence: (X.N.); (L.C.); (J.L.)
| | - Limin Chai
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
- Correspondence: (X.N.); (L.C.); (J.L.)
| | - Jinyu Li
- Department of Orthopedic, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
- Correspondence: (X.N.); (L.C.); (J.L.)
| |
Collapse
|
10
|
Ng MYW, Charsou C, Lapao A, Singh S, Trachsel-Moncho L, Schultz SW, Nakken S, Munson MJ, Simonsen A. The cholesterol transport protein GRAMD1C regulates autophagy initiation and mitochondrial bioenergetics. Nat Commun 2022; 13:6283. [PMID: 36270994 PMCID: PMC9586981 DOI: 10.1038/s41467-022-33933-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 10/05/2022] [Indexed: 12/25/2022] Open
Abstract
During autophagy, cytosolic cargo is sequestered into double-membrane vesicles called autophagosomes. The contributions of specific lipids, such as cholesterol, to the membranes that form the autophagosome, remain to be fully characterized. Here, we demonstrate that short term cholesterol depletion leads to a rapid induction of autophagy and a corresponding increase in autophagy initiation events. We further show that the ER-localized cholesterol transport protein GRAMD1C functions as a negative regulator of starvation-induced autophagy and that both its cholesterol transport VASt domain and membrane binding GRAM domain are required for GRAMD1C-mediated suppression of autophagy initiation. Similar to its yeast orthologue, GRAMD1C associates with mitochondria through its GRAM domain. Cells lacking GRAMD1C or its VASt domain show increased mitochondrial cholesterol levels and mitochondrial oxidative phosphorylation, suggesting that GRAMD1C may facilitate cholesterol transfer at ER-mitochondria contact sites. Finally, we demonstrate that expression of GRAMD family proteins is linked to clear cell renal carcinoma survival, highlighting the pathophysiological relevance of cholesterol transport proteins.
Collapse
Affiliation(s)
- Matthew Yoke Wui Ng
- grid.5510.10000 0004 1936 8921Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway ,grid.5510.10000 0004 1936 8921Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, 0450 Oslo, Norway
| | - Chara Charsou
- grid.5510.10000 0004 1936 8921Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway ,grid.5510.10000 0004 1936 8921Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, 0450 Oslo, Norway
| | - Ana Lapao
- grid.5510.10000 0004 1936 8921Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway ,grid.5510.10000 0004 1936 8921Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, 0450 Oslo, Norway
| | - Sakshi Singh
- grid.5510.10000 0004 1936 8921Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway ,grid.5510.10000 0004 1936 8921Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, 0450 Oslo, Norway
| | - Laura Trachsel-Moncho
- grid.5510.10000 0004 1936 8921Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway ,grid.5510.10000 0004 1936 8921Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, 0450 Oslo, Norway
| | - Sebastian W. Schultz
- grid.5510.10000 0004 1936 8921Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, 0450 Oslo, Norway ,grid.55325.340000 0004 0389 8485Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital Montebello, 0379 Oslo, Norway
| | - Sigve Nakken
- grid.5510.10000 0004 1936 8921Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, 0450 Oslo, Norway ,grid.55325.340000 0004 0389 8485Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Montebello, 0379 Oslo, Norway
| | - Michael J. Munson
- grid.5510.10000 0004 1936 8921Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway ,grid.5510.10000 0004 1936 8921Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, 0450 Oslo, Norway ,grid.418151.80000 0001 1519 6403Present Address: Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anne Simonsen
- grid.5510.10000 0004 1936 8921Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway ,grid.5510.10000 0004 1936 8921Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, 0450 Oslo, Norway ,grid.55325.340000 0004 0389 8485Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital Montebello, 0379 Oslo, Norway
| |
Collapse
|
11
|
Jang BY, Guo SB, Bai R, Liu WL, Gong YL, Zhao ZQ. Methylprednisolone Inhibits Autophagy of Vascular Endothelial Cells in Rat Femoral Head Via PI3K/Akt/mTOR Pathway. Orthop Surg 2022; 14:2669-2681. [PMID: 36052745 PMCID: PMC9531065 DOI: 10.1111/os.13369] [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: 12/29/2020] [Revised: 05/23/2022] [Accepted: 05/23/2022] [Indexed: 11/29/2022] Open
Abstract
Objective To study the relationship between vascular endothelial cells (VEC) and autophagy, and its regulatory mechanism in steroid‐induced avascular necrosis of the femoral head (SANFH). Methods In cell experiment, VEC were isolated and cultured from the femoral head of Sprague–Dawley rats and divided into three groups: blank control group (Ctrl), methylprednisolone group (MP), and methylprednisolone+mTOR‐shRNA group (MP + shmTOR). The autophagy formation was observed by transmission electron microscope. The mRNA expression of PI3K, Akt, mTOR, Beclin1 and MAP1LC3 was detected by RT‐PCR and the protein expression was detected by Western blot and immunofluorescence. Expression of the damage marker 6‐keto‐PGF1α was detected by the ELISA method. In vivo experiment, after establishing the model, the grouping method was the same as cell experiment. Autophagosomes were observed by same method, and the expression of related factors was detected by the same method in cell experiment. Results In the cell experiment, autophagosomes in the MP group were significantly lower than in the Ctrl group, and the autophagosomes in the MP + shmTOR group were intermediate between two groups (P < 0.05). The mRNA expression levels of PI3K, Akt and mTOR in the MP group were significantly higher than in the Ctrl group, while the MP+ shmTOR group presented intermediate levels between these groups (average gray value were 3837.90, 2996.30, 3005.60, F = 428.64, P < 0.05). MRNA expression levels of Beclin1 and MAP1LC3 in the MP group were significantly lower than that in Ctrl group (P < 0.05). The content of 6‐keto‐PGF1α in the MP + shmTOR group was higher than in the Ctrl group and lower than in the MP group at the evaluated time intervals (average absorbance value were 104.98, 206.83, 145.91, F = 352.83, P < 0.01). In vivo experiment, the content of 6‐Keto‐PGF1α in the hormone group increased as time went on; the mTOR‐si group was higher than that in control group, but lower than that in the hormone group (P < 0.01). The mRNA expressions of Beclin1 and MAP1LC3 in the control group were higher than those in the hormone group, while the mRNA expressions of PI3K, Akt and mTOR were lower than those in the mTOR‐si group (P < 0.05). Conclusion The steroid inhibited the physiological protective effect of autophagy on SANFH by increasing the expression of PI3K/Akt/mTOR signaling pathway related factors and decreasing the expression of Beclin1 and MAP1LC3 in the femoral head VEC.
Collapse
Affiliation(s)
- Bo-Yong Jang
- Graduate School of Inner Mongolia Medical University, Hohhot, China
| | - Shi-Bing Guo
- Department of Bone Tumor, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Rui Bai
- Department of Pediatric Orthopedics, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Wan-Lin Liu
- Department of Pediatric Orthopedics, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Yu-Lin Gong
- Department of Rheumatology and Immunology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Zhen-Qun Zhao
- Department of Pediatric Orthopedics, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
12
|
Ma J, Shen M, Yue D, Wang W, Gao F, Wang B. Extracellular Vesicles from BMSCs Prevent Glucocorticoid-Induced BMECs Injury by Regulating Autophagy via the PI3K/Akt/mTOR Pathway. Cells 2022; 11:2104. [PMID: 35805188 PMCID: PMC9265732 DOI: 10.3390/cells11132104] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/22/2022] [Accepted: 05/26/2022] [Indexed: 12/17/2022] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a common clinical disease with a high disability rate. Injury of bone microvascular endothelial cells (BMECs) caused by glucocorticoid administration is one of the important causes of ONFH, and there is currently a lack of effective clinical treatments. Extracellular vesicles derived from bone stem cells (BMSC-EVs) can prevent ONFH by promoting angiogenesis and can inhibit cell apoptosis by regulating autophagy via the PI3K/Akt/mTOR signaling pathway. The present study aimed to investigate the effect of extracellular vesicles derived from bone marrow stem cells (BMSC) on a glucocorticoid-induced injury of BMECs and possible mechanisms. We found that BMSC-EVs attenuated glucocorticoid-induced viability, angiogenesis capacity injury, and the apoptosis of BMECs. BMSC-EVs increased the LC3 level, but decreased p62 (an autophagy protein receptor) expression, suggesting that BMSC-Exos activated autophagy in glucocorticoid-treated BMECs. The protective effects of BMSC-EVs on the glucocorticoid-induced injury of BMECs was mimicked by a known stimulator of autophagy (rapamycin) and could be enhanced by co-treatment with an autophagy inhibitor (LY294002). BMSC-EVs also suppressed the PI3K/Akt/mTOR signaling pathway, which regulates cell autophagy, in glucocorticoid-treated BMECs. In conclusion, the results indicate that BMSC-EVs prevent the glucocorticoid-induced injury of BMECs by regulating autophagy via the PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Jinhui Ma
- Department of Orthopaedic Surgery, Center for Osteonecrosis and Joint Preserving & Reconstruction, China-Japan Friendship Hospital, Beijing 100029, China; (J.M.); (D.Y.); (W.W.)
| | - Mengran Shen
- Department of Orthopaedic Surgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China;
| | - Debo Yue
- Department of Orthopaedic Surgery, Center for Osteonecrosis and Joint Preserving & Reconstruction, China-Japan Friendship Hospital, Beijing 100029, China; (J.M.); (D.Y.); (W.W.)
| | - Weiguo Wang
- Department of Orthopaedic Surgery, Center for Osteonecrosis and Joint Preserving & Reconstruction, China-Japan Friendship Hospital, Beijing 100029, China; (J.M.); (D.Y.); (W.W.)
| | - Fuqiang Gao
- Department of Orthopaedic Surgery, Center for Osteonecrosis and Joint Preserving & Reconstruction, China-Japan Friendship Hospital, Beijing 100029, China; (J.M.); (D.Y.); (W.W.)
| | - Bailiang Wang
- Department of Orthopaedic Surgery, Center for Osteonecrosis and Joint Preserving & Reconstruction, China-Japan Friendship Hospital, Beijing 100029, China; (J.M.); (D.Y.); (W.W.)
| |
Collapse
|
13
|
Novel Effects of Statins on Cancer via Autophagy. Pharmaceuticals (Basel) 2022; 15:ph15060648. [PMID: 35745567 PMCID: PMC9228383 DOI: 10.3390/ph15060648] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 02/05/2023] Open
Abstract
Cancer is one of the main causes of death globally. Most of the molecular mechanisms underlying cancer are marked by complex aberrations that activate the critical cell-signaling pathways that play a pivotal role in cell metabolism, tumor development, cytoskeletal reorganization, and metastasis. The phosphatidylinositol 3-kinase/protein kinase-B/mammalian target of the rapamycin (PI3K/AKT/mTOR) pathway is one of the main signaling pathways involved in carcinogenesis and metastasis. Autophagy, a cellular pathway that delivers cytoplasmic components to lysosomes for degradation, plays a dual role in cancer, as either a tumor promoter or a tumor suppressor, depending on the stage of the carcinogenesis. Statins are the group of drugs of choice to lower the level of low-density lipoprotein (LDL) cholesterol in the blood. Experimental and clinical data suggest the potential of statins in the treatment of cancer. In vitro and in vivo studies have demonstrated the molecular mechanisms through which statins inhibit the proliferation and metastasis of cancer cells in different types of cancer. The anticancer properties of statins have been shown to result in the suppression of tumor growth, the induction of apoptosis, and autophagy. This literature review shows the dual role of the autophagic process in cancer and the latest scientific evidence related to the inducing effect exerted by statins on autophagy, which could explain their anticancer potential.
Collapse
|
14
|
Osteonecrosis of the Femoral Head. J Am Acad Orthop Surg Glob Res Rev 2022; 6:01979360-202205000-00002. [PMID: 35511598 PMCID: PMC9076447 DOI: 10.5435/jaaosglobal-d-21-00176] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 01/23/2022] [Indexed: 11/18/2022]
Abstract
Osteonecrosis of the femoral head is a progressive and debilitating condition with a wide variety of etiologies including trauma, steroid use, and alcohol intake. Diagnosis and staging are based on imaging including MRI at any stage and plain radiography in more advanced lesions. The only definitive treatment is total hip arthroplasty, although numerous treatments including disphosphonates and core decompression are used to delay the progression. Lack of satisfactory conservative measures suggests the need for additional research of osteonecrosis including large patient registries to further understand this condition.
Collapse
|
15
|
Pravastatin Administration Alleviates Kanamycin-Induced Cochlear Injury and Hearing Loss. Int J Mol Sci 2022; 23:ijms23094524. [PMID: 35562915 PMCID: PMC9105065 DOI: 10.3390/ijms23094524] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/13/2022] [Accepted: 04/18/2022] [Indexed: 11/23/2022] Open
Abstract
The effect of statins on aminoglycoside-induced ototoxicity is controversial. This study aimed to explore the role of pravastatin (PV) in kanamycin-induced hearing loss in rats. Adult rats were intraperitoneally treated with 20 mg/kg/day of kanamycin (KM) for 10 days. In the PV- and PV + KM-treated rats, 25 mg/kg/day of PV was intraperitoneally administered for 5 days. The auditory brainstem response (ABR) thresholds were measured before and after drug treatment using a smartEP system at 4, 8, 16, and 32 kHz. Cochlear changes in poly ADP-ribose (PAR) polymerase (PARP), PAR, and caspase 3 were estimated using Western blotting. PV administration did not increase the ABR thresholds. The KM-treated rats showed elevated ABR thresholds at 4, 8, 16, and 32 kHz. The PV + KM-treated rats demonstrated lower ABR thresholds than the KM-treated rats at 4, 8, and 16 kHz. The cochlear outer hair cells and spiral ganglion cells were relatively preserved in the PV + KM-treated rats when compared with that in the KM-treated rats. The cochlear expression levels of PARP, PAR, and caspase 3 were higher in the KM-treated rats. The PV + KM-treated rats showed lower levels of PARP, PAR, and caspase 3 than the KM-treated rats. PV protected cochleae from KM-induced hearing loss in rats. The regulation of autophagy and apoptosis mediated the otoprotective effects of PV.
Collapse
|
16
|
Sconza C, Coletta F, Magarelli N, D'Agostino MC, Egan CG, Di Matteo B, Respizzi S, Mazziotti G. Multimodal conservative treatment of migrating bone marrow edema associated with early osteonecrosis of the hip. SAGE Open Med Case Rep 2022; 10:2050313X211067617. [PMID: 35186294 PMCID: PMC8855375 DOI: 10.1177/2050313x211067617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 12/01/2021] [Indexed: 11/16/2022] Open
Abstract
Bone marrow edema syndrome is a severely disabling painful condition without a defined treatment and related to pathogenetic mechanisms not yet clearly recognized. We report the case of a 59-year-old post-menopausal woman, affected by bone marrow edema associated with early osteonecrosis of the femoral head with secondary appearance of a rare migrant bone edema of the hip acetabulum. Clinical evaluation and magnetic resonance imaging were used to monitor the outcome of the patient. Pre-treatment clinical evaluation revealed pain upon stepping with the left limb, reduced range of motion of spine and hip, and hip pain during passive rotation. Magnetic resonance imaging showed diffuse signal alteration of the head and neck of the left femur in relation to bone edema, associated with an unclear small cephalic area of the femoral head suggestive of initial osteonecrosis. A further computed tomography scan was performed that did not reveal any alterations in bone profile, interruption of the cortex, or trabecular bone collapse. We immediately started a multimodal conservative treatment administering neridronate (100 mg, intravenously) combined with calcium and vitamin D supplementation and biophysical therapies (magnetotherapy and extracorporeal shockwave therapy). We also instructed the patient not to bear the load on the affected lower limb during standing and walking, using crutches. After 2 months, a notable regression of pain with improvement in mobility was observed. Magnetic resonance imaging revealed complete regression of edema at the head and neck of the femur; however, the new appearance of acetabular bone edema of the ipsilateral acetabular roof was detected. After 4 months, a third magnetic resonance imaging showed the disappearance of the femoral head and acetabular roof defects as well as the complete clinical recovery of the patient. An early diagnosis and intervention are essential to conservatively treat cases of bone marrow edema syndrome.
Collapse
Affiliation(s)
- Cristiano Sconza
- Department of Rehabilitation, IRCCS Humanitas Research Hospital, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Francesco Coletta
- Department of Rehabilitation, IRCCS Humanitas Research Hospital, Milan, Italy.,Physical Medicine and Rehabilitation School, University of Milan, Milan, Italy
| | - Nicola Magarelli
- Department of Radiology, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Maria Cristina D'Agostino
- Department of Rehabilitation, IRCCS Humanitas Research Hospital, Milan, Italy.,Ortho-Rehabilitation Department, IRCCS Humanitas Research Hospital, Shock Waves Center, Milan, Italy
| | | | - Berardo Di Matteo
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,Department of Traumatology, Orthopaedics and Disaster Surgery, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Stefano Respizzi
- Department of Rehabilitation, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Gherardo Mazziotti
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,Endocrinology, Diabetology and Andrology Unit, Metabolic Bone Diseases and Osteoporosis Section, IRCCS Humanitas Research Hospital, Milan, Italy
| |
Collapse
|
17
|
Huang C, Wen Z, Niu J, Lin S, Wang W. Steroid-Induced Osteonecrosis of the Femoral Head: Novel Insight Into the Roles of Bone Endothelial Cells in Pathogenesis and Treatment. Front Cell Dev Biol 2021; 9:777697. [PMID: 34917616 PMCID: PMC8670327 DOI: 10.3389/fcell.2021.777697] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/16/2021] [Indexed: 01/18/2023] Open
Abstract
Steroid-induced osteonecrosis of the femoral head (SONFH) is a disease characterized by the collapse of the femoral head. SONFH occurs due to the overuse of glucocorticoids (GCs) in patients with immune-related diseases. Among various pathogenesis proposed, the mechanism related to impaired blood vessels is gradually becoming the most convincing hypothesis. Bone endothelial cells including bone microvascular endothelial cells (BMECs) and endothelial progenitor cells (EPCs) play a crucial role in the maintenance of vascular homeostasis. Therefore, bone endothelial cells are key regulators in the occurrence and progression of SONFH. Impaired angiogenesis, abnormal apoptosis, thrombosis and fat embolism caused by the dysfunctions of bone endothelial cells are considered to be the pathogenesis of SONFH. In addition, even with high disability rates, SONFH lacks effective therapeutic approach. Icariin (ICA, a flavonoid extracted from Epimedii Herba), pravastatin, and VO-OHpic (a potent inhibitor of PTEN) are candidate reagents to prevent and treat SONFH through improving above pathological processes. However, these reagents are still in the preclinical stage and will not be widely used temporarily. In this case, bone tissue engineering represented by co-transplantation of bone endothelial cells and bone marrow mesenchymal stem cells (BMSCs) may be another feasible therapeutic strategy.
Collapse
Affiliation(s)
- Cheng Huang
- Department of Orthopedics, China-Japan Friendship Hospital, Beijing, China
| | - Zeqin Wen
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Junjie Niu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Subin Lin
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Weiguo Wang
- Department of Orthopedics, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
18
|
Gujiansan Ameliorates Avascular Necrosis of the Femoral Head by Regulating Autophagy via the HIF-1 α/BNIP3 Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6683007. [PMID: 34512780 PMCID: PMC8426065 DOI: 10.1155/2021/6683007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 06/21/2021] [Accepted: 08/12/2021] [Indexed: 11/18/2022]
Abstract
Background Clinically, the traditional Chinese medicine compound Gujiansan has been widely used in the treatment of steroid-induced avascular necrosis of the femoral head (SANFH). The present study aimed to investigate the mechanisms underlying the therapeutic effect of Gujiansan. Methods A rat model of SANFH was established by the injection of dexamethasone (DEX) at a high dosage of 25 mg/kg/d. Then, Gujiansan was intragastrically administered for 2 weeks, 4 weeks, and 8 weeks, and histological examination of the femoral head was performed. The expression levels of related mRNAs and proteins were analyzed by qRT-PCR, Western blotting, and immunohistochemistry, and the levels of bone biochemical markers and cytokines were detected with ELISA kits. Results Gujiansan administration ameliorated SANFH and induced the expression of hypoxia-inducible factor-1α (HIF-1α), Bcl-2/adenovirus E1B 19 kDa interacting protein 3 (BNIP3), LC3, and Beclin-1 in the rat model in a dose- and time-dependent manner, and Gujiansan promoted osteocalcin secretion at the femoral head. In addition, Gujiansan increased the levels of bone formation- and bone resorption-specific markers (osteocalcin (OC), bone-specific alkaline phosphatase (BAP), tartrate resistant acid phosphatase-5b (TRACP-5b), N-terminal telopeptides of type I collagen (NTX-1), and C-terminal telopeptide of type I collagen (CTX-1)) and decreased the levels of proinflammatory cytokines (TNF-α, IL-6, and CRP) in a dose- and time-dependent manner. Conclusions Gujiansan accelerates the formation of a new bone, promotes the absorption of the damaged bone, inhibits the inflammatory response, induces autophagy of the femoral head via the HIF-1α/BNIP3 pathway, and ultimately ameliorates SANFH.
Collapse
|
19
|
Implications on the Therapeutic Potential of Statins via Modulation of Autophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9599608. [PMID: 34373771 PMCID: PMC8349293 DOI: 10.1155/2021/9599608] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 07/20/2021] [Indexed: 01/05/2023]
Abstract
Statins, which are functionally known as 3-hydroxy-3-methyl-glutaryl-CoA (HMG-CoA) inhibitors, are lipid-lowering compounds widely prescribed in patients with cardiovascular diseases (CVD). Several biological and therapeutic functions have been attributed to statins, including neuroprotection, antioxidation, anti-inflammation, and anticancer effects. Pharmacological characteristics of statins have been attributed to their involvement in the modulation of several cellular signaling pathways. Over the past few years, the therapeutic role of statins has partially been attributed to the induction of autophagy, which is critical in maintaining cellular homeostasis and accounts for the removal of unfavorable cells or specific organelles within cells. Dysregulated mechanisms of the autophagy pathway have been attributed to the etiopathogenesis of various disorders, including neurodegenerative disorders, malignancies, infections, and even aging. Autophagy functions as a double-edged sword during tumor metastasis. On the one hand, it plays a role in inhibiting metastasis through restricting necrosis of tumor cells, suppressing the infiltration of the inflammatory cell to the tumor niche, and generating the release of mediators that induce potent immune responses against tumor cells. On the other hand, autophagy has also been associated with promoting tumor metastasis. Several anticancer medications which are aimed at inducing autophagy in the tumor cells are related to statins. This review article discusses the implications of statins in the induction of autophagy and, hence, the treatment of various disorders.
Collapse
|
20
|
Abstract
Objectives
This study aims to explore the mechanism by which osteoblast autophagy participated in glucocorticoid-induced femoral head necrosis (FHN). Materials and methods
Thirty male specific-pathogen-free C57 mice (age, one month; weighing 20-25 g) were randomly divided into blank control, dexamethasone and rapamycin-dexamethasone groups (n=10). After six weeks of intervention, right femoral head was obtained to observe morphology and to calculate percentage of empty lacunae. MC3T3-E1 cells were randomly divided into normal, dexamethasone, rapamycin and dexamethasone-rapamycin groups, and cultured for 24 h. Microtubule-associated protein 1 light chain 3 (LC3)-I, LC3-II, mammalian target of rapamycin (mTOR) and Beclin-1 protein expressions were detected by Western blot. Results
In rapamycin-dexamethasone group, some bone trabeculae in medullary cavity ruptured and atrophied, and subchondral bone underwent local necrosis. The total apoptosis rates of dexamethasone and rapamycin-dexamethasone groups surpassed that of blank control group, and the former two groups had significantly different rates (p<0.001). LC3-II/LC3-I of dexamethasone group was lower than those of rapamycin and dexamethasone-rapamycin groups (p<0.001), and the ratio of rapamycin group surpassed that of dexamethasone-rapamycin group (p<0.001). Dexamethasone group had higher mTOR protein expression than those of rapamycin and dexamethasone- rapamycin groups (p<0.001), and the expression of rapamycin group was lower than that of dexamethasone-rapamycin group (p<0.001). The Beclin-1 protein expression of dexamethasone group was lower than those of rapamycin and dexamethasone- rapamycin groups (p<0.001), and the expression of rapamycin group exceeded that of dexamethasone-rapamycin group (p<0.05). Conclusion Osteoblast autophagy may play a crucial protective role in dexamethasone-induced FHN. The attenuation of autophagy may be related to the affected expressions of key autophagy regulators mTOR and Beclin-1.
Collapse
|
21
|
Sheng Z, Xiaoping H, Lu D, XiZhe W, Jie Z, Qing L, Yong C. Identification of key non-coding RNAs and transcription factors regulators and their potential drugs for steroid-induced femoral head necrosis. Genomics 2020; 113:490-496. [PMID: 33385494 DOI: 10.1016/j.ygeno.2020.12.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 09/20/2020] [Accepted: 12/22/2020] [Indexed: 12/09/2022]
Abstract
Steroid-induced necrosis of femoral head (SINFH) is a femoral head necrotic disease caused by prolonged use of hormones. The detailed pathogenesis has not been fully demonstrated. In this study, we employed the bioinformatics approach to probe the roles of SINFH inhibitors. Core dysfunction modules related to SINFH was obtained. Meanwhile, GO and KEGG analysis of genes in dysfunction modules are carried out. Furthermore, the pivot prediction analysis of dysfunction modules related to ncRNA and transcription factor (TF) has been performed. The functions of the enriched modules were focused on multiple perspectives, including circulation, gland development, bone development and reconstruction, calcium production, and fatty acid metabolism regulation. The ncRNAs and TFs analysis showed that miR-322-5p, miR-124-3p, miR-125a-3p, and Ctnnb1 were important members of SINFH dysfunction. Drug targets suggested that Zinc and adenosine monophosphate may have an impact on SINFH dysfunction. SINFH was closely related to bone development and reconstruction.
Collapse
Affiliation(s)
- Zhai Sheng
- The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Han Xiaoping
- The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Ding Lu
- The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Wang XiZhe
- The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Zhang Jie
- The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Lv Qing
- The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Cui Yong
- The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China.
| |
Collapse
|
22
|
Bao SB. Professor Cristobal G dos Remedios mentorship. Biophys Rev 2020; 12:757-759. [PMID: 32661901 DOI: 10.1007/s12551-020-00741-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 07/09/2020] [Indexed: 01/20/2023] Open
Abstract
As a non-English-speaking PhD student without previous scientific skills, I have been so lucky to be supervised and mentored by Professor Cristobal dos Remedios. In this commentary, I have commented my experience in dos Remedios laboratory. Finally, I would like to express my greatest appreciation to Professor dos Remedios for his kindness and mentorship over the last 31 years. His continuous support, which continues to the present, has been instrumental for the achievement of my current position.
Collapse
Affiliation(s)
- Shisan Bob Bao
- Discipline of Pathology, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
23
|
Zheng LW, Wang WC, Mao XZ, Luo YH, Tong ZY, Li D. TNF-α regulates the early development of avascular necrosis of the femoral head by mediating osteoblast autophagy and apoptosis via the p38 MAPK/NF-κB signaling pathway. Cell Biol Int 2020; 44:1881-1889. [PMID: 32437045 DOI: 10.1002/cbin.11394] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 04/23/2020] [Accepted: 05/18/2020] [Indexed: 01/16/2023]
Abstract
Previous studies have shown that the tumor necrosis factor-α (TNF-α) levels in serum and bone tissues formed in avascular necrosis of femoral head (ANFH) patients were higher than those of normal individuals, indicating TNF-α might play a role in the pathogenesis of ANFH. However, the underlying mechanisms remain unclear. Hematoxylin and eosin staining was performed to show the pathological changes of ANFH bone tissues. TNF-α expression in normal and ANFH tissues was examined by quantitative real-time polymerase chain reaction and western blot analyses. Osteoblast autophagy and apoptosis, as well as signaling pathways activation, were measured by their corresponding marker proteins. Osteoblast proliferation, autophagy, and apoptosis were evaluated using cell counting kit-8, transmission electron microscopy, and flow cytometry. The structures of bone tissues of ANFH were obviously damaged. TNF-α expression was significantly upregulated in ANFH bone tissues compared to normal tissues. Autophagy and apoptosis were remarkably promoted, and p38 mitogen-activated protein kinase (MAPK)/nuclear factor-κB (NF-κB) signaling pathways were markedly activated in ANFH. Suppression of the p38 MAPK/NF-κB pathway significantly attenuated the TNF-α-induced autophagy, however, enhanced the TNF-α-induced apoptosis in osteoblasts. Increased TNF-α in ANFH regulated osteoblast autophagy and apoptosis by p38 MAPK/NF-κB signaling pathways, blocking the pathway by inhibitors exacerbated TNF-α-induced apoptosis through impairing autophagy flux.
Collapse
Affiliation(s)
- Li-Wen Zheng
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wan-Chun Wang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin-Zhan Mao
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yong-Heng Luo
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhong-Yi Tong
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ding Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
24
|
Jing X, Du T, Yang X, Zhang W, Wang G, Liu X, Li T, Jiang Z. Desferoxamine protects against glucocorticoid-induced osteonecrosis of the femoral head via activating HIF-1α expression. J Cell Physiol 2020; 235:9864-9875. [PMID: 32437020 DOI: 10.1002/jcp.29799] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/07/2020] [Indexed: 12/21/2022]
Abstract
Glucocorticoid-induced osteonecrosis of the femoral head (GIOFH) is one of the most common complications of glucocorticoid administration. By chelating Fe2+ , desferoxamine (DFO) was reported to be able to activate the HIF-1α/VEGF pathway and promote angiogenesis. In the present study, we examined whether DFO administration could promote angiogenesis and bone repair in GIOFH. GIOFH was induced in rats by methylprednisolone in combination with lipopolysaccharide. Bone repair was assessed by histologic analysis and microcomputed tomography (micro-CT). Vascularization was assessed by Microfil perfusion and micro-CT analysis. Immunohistochemical staining was performed to analyze the expression of HIF-1α, VEGF, and CD31. Our in vivo study revealed that DFO increased HIF-1α/VEGF expression and promoted angiogenesis and osteogenesis in GIOFH. Moreover, our in vitro study revealed that DFO restored dexamethone-induced HIF-1α downregulation and angiogenesis inhibition. Besides, our in vitro study also demonstrated that DFO could protect bone marrow-derived stem cells from dexamethone-induced apoptosis and mitochondrial dysfunction by promoting mitophagy and mitochondrial fission. In summary, our data provided useful information for the development of novel therapeutics for management of GIOFH.
Collapse
Affiliation(s)
- Xingzhi Jing
- Department of Spine Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Ting Du
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Xiaoxia Yang
- Department of Spine Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Weimin Zhang
- Department of Spine Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Guodong Wang
- Department of Spine Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xiaoyang Liu
- Department of Spine Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Tao Li
- Department of Spine Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhensong Jiang
- Department of Spine Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
25
|
De Munck DG, De Meyer GR, Martinet W. Autophagy as an emerging therapeutic target for age-related vascular pathologies. Expert Opin Ther Targets 2020; 24:131-145. [PMID: 31985292 DOI: 10.1080/14728222.2020.1723079] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: The incidence of age-related vascular diseases such as arterial stiffness, hypertension and atherosclerosis, is rising dramatically and is substantially impacting healthcare systems. Mounting evidence suggests that there is an important role for autophagy in maintaining (cardio)vascular health. Impaired vascular autophagy has been linked to arterial aging and the initiation of vascular disease.Areas covered: The function and implications of autophagy in vascular smooth muscle cells and endothelial cells are discussed in healthy blood vessels and arterial disease. Furthermore, we discuss current treatment options for vascular disease and their links with autophagy. A literature search was conducted in PubMed up to October 2019.Expert opinion: Although the therapeutic potential of inducing autophagy in age-related vascular pathologies is considerable, several issues should be addressed before autophagy induction can be clinically used to treat vascular disease. These issues include uncertainty regarding the most effective drug target as well as the lack of potency and selectivity of autophagy inducing drugs. Moreover, drug tolerance or autophagy mediated cell death have been reported as possible adverse effects. Special attention is required for determining the cause of autophagy deficiency to optimize the treatment strategy.
Collapse
Affiliation(s)
- Dorien G De Munck
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Guido Ry De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
26
|
Xu H, Xu X, Wang H, Qimuge A, Liu S, Chen Y, Zhang C, Hu M, Song L. LKB1/p53/TIGAR/autophagy-dependent VEGF expression contributes to PM2.5-induced pulmonary inflammatory responses. Sci Rep 2019; 9:16600. [PMID: 31719630 PMCID: PMC6851103 DOI: 10.1038/s41598-019-53247-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 09/02/2019] [Indexed: 12/19/2022] Open
Abstract
One of the health hazards of PM2.5 exposure is to induce pulmonary inflammatory responses. In our previous study, we demonstrated that exposing both the immortalized and primary human bronchial epithelial cells to PM2.5 results in a significant upregulation of VEGF production, a typical signaling event to trigger chronic airway inflammation. Further investigations showed that PM2.5 exposure strongly induces ATR/CHK1/p53 cascade activation, leading to the induction of DRAM1-dependent autophagy to mediate VEGF expression by activating Src/STAT3 pathway. In the current study, we further revealed that TIGAR was another transcriptional target of p53 to trigger autophagy and VEGF upregulation in Beas-2B cells after PM2.5 exposure. Furthermore, LKB1, but not ATR and CHK1, played a critical role in mediating p53/TIGAR/autophagy/VEGF pathway activation also by linking to Src/STAT3 signaling cascade. Therefore, on combination of the previous report, we have identified both ATR/CHK1/p53/DRAM1- and LKB1/p53/TIGAR- dependent autophagy in mediating VEGF production in the bronchial epithelial cells under PM2.5 exposure. Moreover, the in vivo study further confirmed VEGF induction in the airway potentially contributed to the inflammatory responses in the pulmonary vascular endothelium of PM2.5-treated rats. Therefore, blocking VEGF expression or autophagy induction might be the valuable strategies to alleviating PM2.5-induced respiratory injuries.
Collapse
Affiliation(s)
- Huan Xu
- Institute of Military Cognitive and Brain Sciences, 27 Taiping Road, Beijing, 100850, People's Republic of China.,Anhui Medical University, 81 Meishan Road, Hefei, 230032, People's Republic of China
| | - Xiuduan Xu
- Institute of Military Cognitive and Brain Sciences, 27 Taiping Road, Beijing, 100850, People's Republic of China.,Anhui Medical University, 81 Meishan Road, Hefei, 230032, People's Republic of China.,School of Life Sciences, Tsinghua University, Beijing, China
| | - Hongli Wang
- Institute of Military Cognitive and Brain Sciences, 27 Taiping Road, Beijing, 100850, People's Republic of China.,Laboratory of Cellular and Molecular Immunology, School of Medicine, Henan University, 357 Ximen Road, Kaifeng, 475004, People's Republic of China
| | - Aodeng Qimuge
- Institute of Military Cognitive and Brain Sciences, 27 Taiping Road, Beijing, 100850, People's Republic of China.,Department of New Drug Screening Center, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Shasha Liu
- Institute of Military Cognitive and Brain Sciences, 27 Taiping Road, Beijing, 100850, People's Republic of China.,Department of Pathology, School of Basic Medical Sciences, Lanzhou University, Tianshui South Road, Lanzhou, 730000, People's Republic of China
| | - Yuanlian Chen
- Institute of Military Cognitive and Brain Sciences, 27 Taiping Road, Beijing, 100850, People's Republic of China.,Guiin Medical University, 1 Zhiyuan Road, Guilin, 541100, P.R. China
| | - Chongchong Zhang
- Institute of Military Cognitive and Brain Sciences, 27 Taiping Road, Beijing, 100850, People's Republic of China.,Laboratory of Cellular and Molecular Immunology, School of Medicine, Henan University, 357 Ximen Road, Kaifeng, 475004, People's Republic of China
| | - Meiru Hu
- Institute of Military Cognitive and Brain Sciences, 27 Taiping Road, Beijing, 100850, People's Republic of China
| | - Lun Song
- Institute of Military Cognitive and Brain Sciences, 27 Taiping Road, Beijing, 100850, People's Republic of China. .,Anhui Medical University, 81 Meishan Road, Hefei, 230032, People's Republic of China.
| |
Collapse
|
27
|
Ashrafizadeh M, Ahmadi Z, Farkhondeh T, Samarghandian S. Modulatory effects of statins on the autophagy: A therapeutic perspective. J Cell Physiol 2019; 235:3157-3168. [DOI: 10.1002/jcp.29227] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 09/03/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine University of Tabriz Tabriz Iran
| | - Zahra Ahmadi
- Department of Basic Science, Faculty of Veterinary Medicine University of Tabriz Tabriz Iran
| | - Tahereh Farkhondeh
- Cardiovascular Diseases Research Center Birjand University of Medical Sciences Birjand Iran
| | - Saeed Samarghandian
- Department of Basic Medical Science Neyshabur University of Medical Sciences Neyshabur Iran
| |
Collapse
|
28
|
Zeng W, Wang W, Wu S, Zhu X, Zheng T, Chen X, Ren J, Gong Y, Ke M. Mitochondria and Autophagy Dysfunction in Glucocorticoid-Induced Ocular Hypertension/Glaucoma Mice Model. Curr Eye Res 2019; 45:190-198. [PMID: 31425668 DOI: 10.1080/02713683.2019.1657462] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Purpose: This study is aimed to investigate the effects of periocular steroids induction on intraocular pressure (IOP), retinal ganglion cells (RGCs) and trabecular meshwork (TM) ultrastructure in glucocorticoid-induced ocular hypertension mice model.Materials and Methods: Dexamethasone-21-acetate (Dex-Ace) was administered through periocular conjunctival fornix injection every 3 days in C57BL/6J mice. Intraocular pressure was measured weekly by rebound tonometry. RGCs were examined with immunofluorescent staining of BRN3a at week 1, 4, and 8. TM morphology was visualized with electron microscopy. Autophagy was evaluated with immunoblotting in TM tissues.Results: Dex-Ace rapidly and significantly induced IOP, which peaked at week 4. The absolute increase in IOP in the Dex-Ace-treated mice was 8.1 ± 1.4 mmHg, a 60% induction (p < .0001) compared with that in the vehicle-treated mice. The IOP sustained a higher level in the Dex-Ace group from week 4 to week 8. Dex-Ace treatment decreased the number of RGCs in a time-dependent manner, suggesting that high IOP resulted in optic neuropathy. In addition, Dex-Ace thickened trabecular beams and decreased intertrabecular spaces, with marked accumulation of fibrillar and amorphous granular extracellular material. Moreover, Dex-Ace induced swollen and elongated mitochondria in TM cells. The average mitochondria area was 0.090 ± 0.044 µm2 in the vehicle-treated mice, and increased to 0.161 ± 0.094 µm2 (p < .0001), 0.121 ± 0.029 µm2 (p = .0223) and 0.171 ± 0.076 µm2 (p < .0001) in the Dex-Ace-treated mice at weeks 1, 4 and 8, respectively. Autophagy was also increased by Dex-Ace treatment, indicating by the upregulation of LC3-I, LC3-II and beclin-1, and downregulation of p62.Conclusion: Dex-Ace administration decreased RGCs and changed TM ultrastructure, mimicking hallmarks of human glucocorticoid-induced glaucoma (GIG). In addition, mitochondria and autophagy dysfunction suggested abnormal energy metabolism in TM cells, which warranted further study to fully elucidate the pathogenesis of GIG.
Collapse
Affiliation(s)
- Wen Zeng
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wenhuan Wang
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shengyu Wu
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiaolu Zhu
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Tian Zheng
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiaomin Chen
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiangbo Ren
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yan Gong
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Min Ke
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
29
|
Zhang Q, Gao F, Cheng L, Liu L, Sun W, Li Z. [Effects of icariin on autophagy and exosome production of bone microvascular endothelial cells]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2019; 33:568-577. [PMID: 31090350 DOI: 10.7507/1002-1892.201811009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Objective To evaluate the effects of icariin on autophagy induced by low-concentration of glucocorticoid and exosome production in bone microvascular endothelial cells (BMECs). Methods BMECs were isolated from femoral heads resected in total hip arthroplasty and then intervened with hydrocortisone of low concentration (0, 0.03, 0.06, 0.10 mg/mL), which were set as groups A, B, C, and D, respectively. On the basis of hydrocortisone intervention, 5×10 -5 mol/L of icariin was added to each group (set as groups A1, B1, C1 and D1, respectively). Western blot was used to detect the expressions of microtubule-associated protein 1 light chain 3B (LC3B) and dead bone slice 1 (p62) after 24 hours. Exosomes were extracted from BMECs treated with icariin (intervention group) and without icariin (non-intervention group), and the diameter and concentration of exosomes were evaluated by nanoparticle tracking analysis technique. The total protein content of exosomes was detected by BCA method, and the expressions of proteins carried by exosomes including CD9, CD81, transforming growth factor β 1 (TGF-β 1), and vascular endothelial growth factor A (VEGFA) were assessed by Western blot. The BMECs were further divided into three groups: BMECs in the experimental group and the control group were co-cultured with exosomes secreted by BMECs treated with or without icariin, respectively; the blank control group was BMECs without exosome intervention. The three groups were treated with hydrocortisone and Western blot was used to detect the expressions of LC3B and p62. The scratching assay was used to detect cell migration ability; angiogenic ability of BMECs was also assessed. Results With the increase of hydrocortisone concentration, the protein expression of LC3B-Ⅱ increased gradually, and the protein expression of p62 decreased gradually ( P<0.01). Compared with group with same concentration of hydrocortisone, the protein expression of LC3B-Ⅱ decreased and the protein expression of p62 increased after the administration of icariin ( P<0.01). The concentration of exosomes in the intervention group was significantly higher than that in the non-intervention group ( t=-10.191, P=0.001); and there was no significant difference in exosome diameter and total protein content between the two groups ( P>0.05). CD9 and CD81 proteins were highly expressed in the non-intervention group and the intervention group, and the relative expression ratios of VEGFA/CD9 and TGF-β 1/CD9 proteins in the intervention group were significantly higher than those in the non-intervention group ( P<0.01). After co-culture of exosomes, the protein expression of p62 increased in blank control group, control group, and experimental group, while the protein expression of LC3B-Ⅱ decreased. There were significant differences among groups ( P<0.05). When treated with hydrocortisone for 12 and 24 hours, the scratch closure rate of the control group and experimental group was significantly higher than that of the blank control group ( P<0.05), and the scratch closure rate of the experimental group was significantly higher than that of the control group ( P<0.05). When treated with hydrocortisone for 4 and 8 hours, the number of lumens, number of sprouting vessels, and length of tubule branches in the experimental group and the control group were significantly greater than those in the blank control group ( P<0.05); the length of tubule branches and the number of lumens in the experimental group were significantly greater than those in the control group ( P<0.05). Conclusion Icariin and BMECs-derived exosomes can improve the autophagy of BMECs induced by low concentration of glucocorticoid.
Collapse
Affiliation(s)
- Qingyu Zhang
- Graduate School of Peking Union Medical College, Beijing, 100730, P.R.China;Department of Orthopedics, China-Japan Friendship Hospital, Beijing, 100029, P.R.China
| | - Fuqiang Gao
- Department of Orthopedics, China-Japan Friendship Hospital, Beijing, 100029, P.R.China
| | - Liming Cheng
- Department of Orthopedics, China-Japan Friendship Hospital, Beijing, 100029, P.R.China
| | - Lihua Liu
- Graduate School of Peking Union Medical College, Beijing, 100730, P.R.China;Department of Orthopedics, China-Japan Friendship Hospital, Beijing, 100029, P.R.China
| | - Wei Sun
- Graduate School of Peking Union Medical College, Beijing, 100730, P.R.China;Department of Orthopedics, China-Japan Friendship Hospital, Beijing, 100029, P.R.China;Center for Osteonecrosis and Joint Preserving & Reconstruction, China-Japan Friendship Hospital, Beijing, 100029,
| | - Zirong Li
- Department of Orthopedics, China-Japan Friendship Hospital, Beijing, 100029, P.R.China
| |
Collapse
|
30
|
Lin J, Jia C, Wang Y, Jiang S, Jia Z, Chen N, Sheng S, Li S, Jiang L, Xu H, Zhou K, Chen Y. Therapeutic potential of pravastatin for random skin flaps necrosis: involvement of promoting angiogenesis and inhibiting apoptosis and oxidative stress. Drug Des Devel Ther 2019; 13:1461-1472. [PMID: 31118580 PMCID: PMC6505465 DOI: 10.2147/dddt.s195479] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 03/23/2019] [Indexed: 12/23/2022] Open
Abstract
Background: Random skin flap is frequently used in plastic and reconstructive surgery, but its distal part often occurs ischemia and necrosis. Pravastatin (Prava) with bioactivities of pro-angiogenesis, anti-apoptosis and anti-oxidative stress, may be beneficial for flap survival. Materials and methods: A modified McFarlane flap model was performed in Sprague-Dawley rats. The animals were divided into the Control and Prava groups and treated as follows: the Prava group was injected intraperitoneally with 2 mg/kg Prava for consecutive 7 days, and the Control group received an equal volume of vehicle daily. On day 7, the necrosis skin flaps were observed, while visualization of blood flow below the tissue surface was performed by Laser Doppler blood flow imaging (LDBFI). Then animals were euthanized, and levels of angiogenesis, apoptosis and oxidative stress were analyzed. Results: Prava decreased necrosis and edema of skin flaps compared with the Control group, with more blood flow in the flap under LDBFI. Prava treatment increased the mean vessels density, elevated the expression levels of angiogenic proteins (matrix metallopeptidase 9, vascular endothelial growth factor, Cadherin5) and antioxidant proteins (superoxide dismutase 1 (SOD1), endothelial nitric oxide synthase, heme oxygenase), and decreased the expression of apoptotic factors (BAX, CYC, Caspase3). In addition, malondialdehyde content was reduced, and glutathione level and SOD activity were increased in the skin flaps after treatment with Prava. Conclusion: Prava promotes survival of random skin flap through induction of angiogenesis, and inhibition of apoptosis and oxidative stress.
Collapse
Affiliation(s)
- Jinti Lin
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou325027, People’s Republic of China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou325027, People’s Republic of China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou325027, People’s Republic of China
| | - Chang Jia
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou325027, People’s Republic of China
- Pediatric Research Institute, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou325027, People’s Republic of China
| | - Yongli Wang
- Department of Orthopaedics, Huzhou Central Hospital, Huzhou313300, People’s Republic of China
| | - Shanghong Jiang
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou325027, People’s Republic of China
| | - Zhenyu Jia
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325027, People’s Republic of China
| | - Nan Chen
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou325027, People’s Republic of China
| | - Shimin Sheng
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou325027, People’s Republic of China
| | - Shihen Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou325027, People’s Republic of China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou325027, People’s Republic of China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou325027, People’s Republic of China
| | - Liangfu Jiang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou325027, People’s Republic of China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou325027, People’s Republic of China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou325027, People’s Republic of China
| | - Huazi Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou325027, People’s Republic of China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou325027, People’s Republic of China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou325027, People’s Republic of China
| | - Kailiang Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou325027, People’s Republic of China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou325027, People’s Republic of China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou325027, People’s Republic of China
| | - Yijie Chen
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou325027, People’s Republic of China
- Department of Obstetrics and Gynecology, The Second Affliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou325027, People’s Republic of China
| |
Collapse
|
31
|
Liao Y, Ouyang L, Ci L, Chen B, Lv D, Li Q, Sun Y, Fei J, Bao S, Liu X, Li L. Pravastatin regulates host foreign-body reaction to polyetheretherketone implants via miR-29ab1-mediated SLIT3 upregulation. Biomaterials 2019; 203:12-22. [PMID: 30851489 DOI: 10.1016/j.biomaterials.2019.02.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/27/2019] [Accepted: 02/28/2019] [Indexed: 12/20/2022]
Abstract
Host rejection to biomaterials can induce uncontrolled foreign-body reactions (FBR), resulting in a dense fibrous encapsulation that blocks mass transport and/or communication between the host and the implant. Adequate angiogenesis between the body and the implant has been implicated as a key regulator for overcoming FBR. Thus, approaches for stimulating neovascularization and/or suppressing FBR are under investigation. In this study, pravastatin (Pra) was loaded onto a 3D network surface of sulfonated polyetheretherketone (SP) to achieve superior local drug effects. The SP loaded with Pra (SP-Pra) promoted angiogenesis and mitigated FBR via miR-29 dependent SLIT3 upregulation in wild-type (WT) mice. miR-29a and miR-29b1 were significantly downregulated in the SP-Pra capsule compared to levels in the SP capsule, while SLIT3 and neovascularization were substantially upregulated in WT mice. However, the above effects presented in the WT mice were not detected in miR-29ab1 knockout mice which was generated by the CRISPR/Cas9 approach. Overall, the results suggest that miR-29 plays a critical role in reducing FBR to these implants by targeting SLIT3. Suppression of FBR by SP-Pra implants offers the potential to improve the performance of current medical devices.
Collapse
Affiliation(s)
- Yun Liao
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Liping Ouyang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lei Ci
- Shanghai Engineering Research Center for Model Organisms, Shanghai Model Organisms Center, INC., Shanghai 201203, China
| | - Baohui Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dan Lv
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Qin Li
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Yingxiao Sun
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Jian Fei
- School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Shisan Bao
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China; Discipline of Pathology, Charles Perkin Centre, Bosch Institute and School of Medical Sciences, The University of Sydney, Australia.
| | - Xuanyong Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China.
| | - Ling Li
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China.
| |
Collapse
|
32
|
Tong W, Tower RJ, Chen C, Wang L, Zhong L, Wei Y, Sun H, Cao G, Jia H, Pacifici M, Koyama E, Enomoto-Iwamoto M, Qin L. Periarticular Mesenchymal Progenitors Initiate and Contribute to Secondary Ossification Center Formation During Mouse Long Bone Development. Stem Cells 2019; 37:677-689. [PMID: 30681752 DOI: 10.1002/stem.2975] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 12/19/2018] [Accepted: 12/27/2018] [Indexed: 12/16/2022]
Abstract
Long bone development involves the embryonic formation of a primary ossification center (POC) in the incipient diaphysis followed by postnatal development of a secondary ossification center (SOC) at each epiphysis. Studies have elucidated major basic mechanisms of POC development, but relatively little is known about SOC development. To gain insights into SOC formation, we used Col2-Cre Rosa-tdTomato (Col2/Tomato) reporter mice and found that their periarticular region contained numerous Tomato-positive lineage cells expressing much higher Tomato fluorescence (termed TomatoH ) than underlying epiphyseal chondrocytes (termed TomatoL ). With time, the TomatoH cells became evident at the SOC invagination site and cartilage canal, increased in number in the expanding SOC, and were present as mesenchymal lineage cells in the subchondral bone. These data were verified in two mouse lineage tracing models, Col2-CreER Rosa-tdTomato and Gli1-CreER Rosa-tdTomato. In vitro tests showed that the periarticular TomatoH cells from Col2/Tomato mice contained mesenchymal progenitors with multidifferentiation abilities. During canal initiation, the cells expressed vascular endothelial growth factor (VEGF) and migrated into epiphyseal cartilage ahead of individual or clusters of endothelial cells, suggesting a unique role in promoting vasculogenesis. Later during SOC expansion, chondrocytes in epiphyseal cartilage expressed VEGF, and angiogenic blood vessels preceded TomatoH cells. Gene expression analyses of microdissected samples revealed upregulation of MMPs in periarticular cells at the invagination site and suggested potential roles for novel kinase and growth factor signaling pathways in regulating SOC canal initiation. In summary, our data indicate that the periarticular region surrounding epiphyseal cartilage contains mesenchymal progenitors that initiate SOC development and form subchondral bone. Stem Cells 2019;37:677-689.
Collapse
Affiliation(s)
- Wei Tong
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Robert J Tower
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Chider Chen
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Luqiang Wang
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Leilei Zhong
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yulong Wei
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Hao Sun
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gaoyuan Cao
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, New Jersey, USA
| | - Haoruo Jia
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Maurizio Pacifici
- Translational Research Program in Pediatric Orthopaedics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Eiki Koyama
- Translational Research Program in Pediatric Orthopaedics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Motomi Enomoto-Iwamoto
- Department of Orthopaedic Surgery, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
33
|
Godoy JC, Niesman IR, Busija AR, Kassan A, Schilling JM, Schwarz A, Alvarez EA, Dalton ND, Drummond JC, Roth DM, Kararigas G, Patel HH, Zemljic-Harpf AE. Atorvastatin, but not pravastatin, inhibits cardiac Akt/mTOR signaling and disturbs mitochondrial ultrastructure in cardiac myocytes. FASEB J 2018; 33:1209-1225. [PMID: 30169110 DOI: 10.1096/fj.201800876r] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Statins, which reduce LDL-cholesterol by inhibition of 3-hydroxy-3-methylglutaryl-coenzyme A reductase, are among the most widely prescribed drugs. Skeletal myopathy is a known statin-induced adverse effect associated with mitochondrial changes. We hypothesized that similar effects would occur in cardiac myocytes in a lipophilicity-dependent manner between 2 common statins: atorvastatin (lipophilic) and pravastatin (hydrophilic). Neonatal cardiac ventricular myocytes were treated with atorvastatin and pravastatin for 48 h. Both statins induced endoplasmic reticular (ER) stress, but only atorvastatin inhibited ERK1/2T202/Y204, AktSer473, and mammalian target of rapamycin signaling; reduced protein abundance of caveolin-1, dystrophin, epidermal growth factor receptor, and insulin receptor-β; decreased Ras homolog gene family member A activation; and induced apoptosis. In cardiomyocyte-equivalent HL-1 cells, atorvastatin, but not pravastatin, reduced mitochondrial oxygen consumption. When male mice underwent atorvastatin and pravastatin administration per os for up to 7 mo, only long-term atorvastatin, but not pravastatin, induced elevated serum creatine kinase; swollen, misaligned, size-variable, and disconnected cardiac mitochondria; alteration of ER structure; repression of mitochondria- and endoplasmic reticulum-related genes; and a 21% increase in mortality in cardiac-specific vinculin-knockout mice during the first 2 months of administration. To our knowledge, we are the first to demonstrate in vivo that long-term atorvastatin administration alters cardiac ultrastructure, a finding with important clinical implications.-Godoy, J. C., Niesman, I. R., Busija, A. R., Kassan, A., Schilling, J. M., Schwarz, A., Alvarez, E. A., Dalton, N. D., Drummond, J. C., Roth, D. M., Kararigas, G., Patel, H. H., Zemljic-Harpf, A. E. Atorvastatin, but not pravastatin, inhibits cardiac Akt/mTOR signaling and disturbs mitochondrial ultrastructure in cardiac myocytes.
Collapse
Affiliation(s)
- Joseph C Godoy
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA
| | - Ingrid R Niesman
- Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| | - Anna R Busija
- Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| | - Adam Kassan
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, University of California, San Diego, San Diego, California, USA.,Department of Pharmaceutical Sciences, School of Pharmacy, West Coast University, North Hollywood, California, USA
| | - Jan M Schilling
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| | - Anna Schwarz
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA
| | - Erika A Alvarez
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Nancy D Dalton
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - John C Drummond
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| | - David M Roth
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| | - Georgios Kararigas
- Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Hemal H Patel
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| | - Alice E Zemljic-Harpf
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| |
Collapse
|