1
|
Cui Y, Tan C, Zhang W, Jiang P, Sun J, Mei F. Establishment of Mouse Models of Abdominal Aortic Aneurysm. Angiology 2024:33197241284848. [PMID: 39268808 DOI: 10.1177/00033197241284848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Abdominal aortic aneurysm (AAA) is a chronic vascular disease that commonly affects elderly individuals but has recently increased in younger populations. As the aneurysm grows, it can cause compression symptoms such as abdominal pain, rupture, and bleeding, which are absent in the early stages. Once an AAA ruptures and causes bleeding, the mortality rate is alarmingly high. Currently, the pathogenesis for AAA is unknown, and therapeutic options are limited, necessitating improvement in treatment efficacy. An essential research method for studying the processes and potential treatment of AAA is establishing animal models using mice. The present study provides a detailed overview of the widely used AAA mouse animal models and their construction strategies, advantages, disadvantages, scope of applications, and prospects.
Collapse
Affiliation(s)
- Yongpan Cui
- Department of Vascular Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Hubei, China
| | - Chengpeng Tan
- Department of Vascular Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Hubei, China
| | - Wuming Zhang
- Department of Vascular Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Hubei, China
| | - Peng Jiang
- Department of Vascular Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Hubei, China
| | - Jianfeng Sun
- Department of Vascular Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Hubei, China
| | - Fei Mei
- Department of Vascular Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Hubei, China
| |
Collapse
|
2
|
Zhang G, Huang C, Wang R, Guo J, Qin Y, Lv S. Chondroprotective effects of Apolipoprotein D in knee osteoarthritis mice through the PI3K/AKT/mTOR signaling pathway. Int Immunopharmacol 2024; 133:112005. [PMID: 38626543 DOI: 10.1016/j.intimp.2024.112005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/18/2024]
Abstract
BACKGROUND Because the pathophysiology of osteoarthritis (OA) has not been fully elucidated, targeted treatments are lacking. In this study, we assessed the role and underlying mechanism apolipoprotein D (APOD) on the development of OA. METHODS To establish an in vitro OA model, we extracted primary chondrocytes from the cartilage of C57BL/6 mice and stimulated the chondrocytes with IL-1β. After APOD intervention or incubation with an overexpressing plasmid, we detected inflammatory-related markers using RT-qPCR, Western blotting, and ELISA. To detect apoptosis and autophagy-related markers, we used flow cytometry, immunofluorescence, and transmission electron microscopy (TEM). Finally, we measured the level of oxidative stress. We also used RNA-seq to identify the APOD-regulated downstream signaling pathways. We used an in vivo mice OA model of the anterior cruciate ligament transection (ACLT) and administered intra-articular adenovirus overexpressing APOD. To examine cartilage damage severity, we used immunohistochemical analysis (IHC), micro-CT, scanning electron microscopy (SEM), and Safranin O-fast green staining. RESULTS Our results showed that APOD inhibited chondrocyte inflammation, degeneration, and apoptosis induced by IL-1β. Additionally, APOD reversed autophagy inhibition and oxidative stress and also blocked activation of the PI3K/AKT/mTOR signaling pathway induced by IL-1β. Finally, overexpression of the APOD gene through adenovirus was sufficient to mitigate OA progression. CONCLUSIONS Our findings revealed that APOD had a chondroprotective role in OA progression by the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Gang Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; Department of Orthopedics, Harbin First Hospital, Harbin, Heilongjiang Province, China; Future Medical Laboratory of the Second Affiliated Hospital of Harbin Medical University, China
| | - Chao Huang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Ren Wang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jiangrong Guo
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yong Qin
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.
| | - Songcen Lv
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.
| |
Collapse
|
3
|
Xu R, Zheng L, Huang M, Zhao M. High gastrointestinal digestive stability endows chondroitin sulfate-soluble undenatured type II collagen complex with high activity: Improvement of osteoarthritis in rats. Int J Biol Macromol 2024; 257:128630. [PMID: 38070808 DOI: 10.1016/j.ijbiomac.2023.128630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 11/08/2023] [Accepted: 12/02/2023] [Indexed: 12/17/2023]
Abstract
Previously, we prepared a chondroitin sulfate-soluble undenatured type II collagen complex (CS-SC II) with low salt content. This paper further explored the differences between CS-SC II and SC II in terms of gastrointestinal digestive characteristics and osteoarthritis (OA) improvement. In vitro and in vivo experiments showed that the gastric digestive stability of CS-SC II was high under both pH 2.0 and pH 3.0, the α1 chain and triple helix structure of type II collagen retained >60 %. However, SC II had high gastric digestive stability only under pH 3.0. Furthermore, intestinal digestion had little effect on α1 chains of CS-SC II and SC II, and distribution experiments showed that they might exert their biological activities in the intestine. CS-SC II had obvious improvement in OA rats at 1.0 mg/kg/d, that is, the joint swelling was significantly reduced and the weight-bearing ratio of the right hind limb was increased to 49 %, which was close to that of 4.0 mg/kg/d SC II. The wear of articular cartilage, Mankin and OARSI scores of rats in CS-SC II group were significantly reduced. The effects of low-dose CS-SC II on the proportion of regulatory T cells (Treg), mRNA expression of OA key biomarkers (Il6, Ccl7, MMP-3 and MMP13) and signaling pathway genes (NF-κB, AKT or AMPKα) were comparable to those of high-dose SC II. These results showed that CS-SC II might have greater potential to improve OA at a lower dose than SC II due to its high gastrointestinal digestive stability at a wide range of pH conditions.
Collapse
Affiliation(s)
- Rong Xu
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China; Guangdong Food Green Processing and Nutrition Regulation Technologies Research Center, Guangzhou 510650, China
| | - Lin Zheng
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China; Guangdong Food Green Processing and Nutrition Regulation Technologies Research Center, Guangzhou 510650, China.
| | - Mingtao Huang
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China; Guangdong Food Green Processing and Nutrition Regulation Technologies Research Center, Guangzhou 510650, China
| | - Mouming Zhao
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China; Guangdong Food Green Processing and Nutrition Regulation Technologies Research Center, Guangzhou 510650, China; Food Laboratory of Zhongyuan, Luohe 462300, Henan, China.
| |
Collapse
|
4
|
Chagan-Yasutan H, He N, Arlud S, Fang J, Hattori T. The elevation of plasma galectin-9 levels in patients with psoriasis and its associations with inflammatory and immune checkpoint molecules in skin tissues. Hum Immunol 2024; 85:110741. [PMID: 38092632 DOI: 10.1016/j.humimm.2023.110741] [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: 10/08/2023] [Revised: 11/29/2023] [Accepted: 12/04/2023] [Indexed: 01/24/2024]
Abstract
Psoriasis is a chronic, immune-mediated disorder that mainly affects the skin, with an estimated global prevalence of 2-3%. Galectin-9 (Gal-9) is a β-galactoside-binding lectin capable of promoting or suppressing the progression of infectious and immune-mediated diseases. Here, we determined if the expression of Gal-9 is observed in psoriasis. Gal-9 levels were measured in plasma of psoriasis (n = 62) and healthy control (HC) (n = 31) using an enzyme-linked immunosorbent assay. In addition, skin samples from seven patients were screened for RNA transcriptomes and the expression of Gal-9 was compared with inflammatory, immune checkpoint molecules (ICMs) and Foxp3. The plasma Gal-9 levels in patients with psoriasis were significantly higher (841 pg/mL) than in HCs (617 pg/mL) (P < 0.0001) and were associated with white blood cell numbers, eosinophils (%) and alanine transaminase. The levels of inflammatory molecules IL-36B, IL-17RA, IL-6R, IL-10, IRF8, TGFb1, and IL-37, and those of ICMs of Tim-3, CTLA-4, CD86, CD80, PD-1LG2, CLEC4G, and Foxp3 were significantly correlated with Gal-9 (LGALS9) in skin. However, HMGB1, CD44, CEACAM1 and PDL1-known to be associated with a variety of Gal-9 biological functions were not correlated with LGALS9. Thus, it is likely that Gal-9 expression affects the disease state of PS.
Collapse
Affiliation(s)
- Haorile Chagan-Yasutan
- Mongolian Psychosomatic Medicine Department, Inner Mongolia International Mongolian Medicine Hospital, Hohhot 010065, China; Research Institute of Health and Welfare, Kibi International University, 8-Iga-machi, Takahashi, Okayama 716-8508, Japan.
| | - Nagongbilige He
- Mongolian Psychosomatic Medicine Department, Inner Mongolia International Mongolian Medicine Hospital, Hohhot 010065, China; The Inner Mongolia Institute of Chinese and Mongolian Medicine, Hohhot 010010, China.
| | - Sarnai Arlud
- Mongolian Psychosomatic Medicine Department, Inner Mongolia International Mongolian Medicine Hospital, Hohhot 010065, China
| | - Jun Fang
- Mongolian Psychosomatic Medicine Department, Inner Mongolia International Mongolian Medicine Hospital, Hohhot 010065, China; The Inner Mongolia Institute of Chinese and Mongolian Medicine, Hohhot 010010, China
| | - Toshio Hattori
- Research Institute of Health and Welfare, Kibi International University, 8-Iga-machi, Takahashi, Okayama 716-8508, Japan; Shizuoka Graduate University of Public Health, 4-27-2 Kita Ando Aoi-ku, Shizuoka City 420-0881, Japan.
| |
Collapse
|
5
|
Chen H, Li Z, Li X, Lu J, Chen B, Wang Q, Wu G. Biomaterial-Based Gene Delivery: Advanced Tools for Enhanced Cartilage Regeneration. Drug Des Devel Ther 2023; 17:3605-3624. [PMID: 38076630 PMCID: PMC10706074 DOI: 10.2147/dddt.s432056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/09/2023] [Indexed: 12/18/2023] Open
Abstract
Gene therapy has emerged as a promising and innovative approach in cartilage regeneration. Integrating biomaterials into gene therapy offers a unique opportunity to enhance gene delivery efficiency, optimize gene expression dynamics, modulate immune responses, and promote tissue regeneration. Despite the rapid progress in biomaterial-based gene delivery, there remains a deficiency of comprehensive discussions on recent advances and their specific application in cartilage regeneration. Therefore, this review aims to provide a thorough overview of various categories of biomaterials employed in gene delivery, including both viral and non-viral vectors, with discussing their distinct advantages and limitations. Furthermore, the diverse strategies employed in gene therapy are discussed and summarized, such as the utilization of growth factors, anti-inflammatory cytokines, and chondrogenic genes. Additionally, we highlights the significant challenges that hinder biomaterial-based gene delivery in cartilage regeneration, including immune response modulation, gene delivery efficiency, and the sustainability of long-term gene expression. By elucidating the functional properties of biomaterials-based gene therapy and their pivotal roles in cartilage regeneration, this review aims to enhance further advances in the design of sophisticated gene delivery systems for improved cartilage regeneration outcomes.
Collapse
Affiliation(s)
- Hongfeng Chen
- Department of Foot and Ankle Surgery, The Second Affiliated Hospital of Luohe Medical College, Luohe, Henan, 462300, People’s Republic of China
| | - Zhen Li
- Department of Foot and Ankle Surgery, The Second Affiliated Hospital of Luohe Medical College, Luohe, Henan, 462300, People’s Republic of China
| | - Xiaoqi Li
- Department of Foot and Ankle Surgery, The Second Affiliated Hospital of Luohe Medical College, Luohe, Henan, 462300, People’s Republic of China
| | - Jiongjiong Lu
- Department of Foot and Ankle Surgery, The Second Affiliated Hospital of Luohe Medical College, Luohe, Henan, 462300, People’s Republic of China
| | - Beibei Chen
- Department of Foot and Ankle Surgery, The Second Affiliated Hospital of Luohe Medical College, Luohe, Henan, 462300, People’s Republic of China
| | - Qiongchao Wang
- Department of Foot and Ankle Surgery, The Second Affiliated Hospital of Luohe Medical College, Luohe, Henan, 462300, People’s Republic of China
| | - Guangliang Wu
- Department of Orthopaedics, The Second Affiliated Hospital of Luohe Medical College, Luohe, Henan, 462300, People’s Republic of China
| |
Collapse
|
6
|
Xu R, Du Y, Li X, Mao X, Zheng L, Zhao M. Differences between soluble and insoluble undenatured type II collagen in improving osteoarthritis in rats and their potential mechanisms. Food Funct 2023; 14:10240-10251. [PMID: 37921641 DOI: 10.1039/d3fo02954a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Our previous research showed that soluble (SC II) and insoluble (IC II) undenatured type II collagen had significant differences during gastrointestinal digestion in vitro, and SC II exposed more type II collagen with triple helix structure. However, the differences in their in vivo digestive characteristics, improvement on osteoarthritis (OA), and possible mechanisms have not been elucidated. The aim of this study was to explore these issues. After oral administration of SC II and IC II, the joint swelling of OA rats significantly reduced, and the weight bearing ratio of right hind limb significantly increased, especially in SC II group (raised to 48%). The Mankin and OARSI scores decreased by 35% and 48% in SC II group, respectively. SC II and IC II increased the mRNA expression of anti-inflammatory factors and the proportion of regulatory T cells (Treg). Importantly, type II collagen released by IC II during in vivo gastrointestinal digestion was far less than SC II, which explained the higher ability of SC II to induce immune tolerance in small intestine than IC II. Bioinformatics analysis showed that the differential genes between model and control were significantly enriched in PI3K/AKT, PPAR and AMPK signalling pathways, and 24 hub genes were analyzed. SC II significantly down-regulated the mRNA expression of Il6, Ccl7, NF-κB, AKT and up-regulated the mRNA expression of Scd1. These results showed that SC II was superior to IC II in improving OA by inducing immune tolerance and could regulate key biomarkers and signalling pathways in OA rats.
Collapse
Affiliation(s)
- Rong Xu
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China.
| | - Yulan Du
- Perfect (China) Co., Ltd, Zhongshan, 528400, China
- Perfect Life and Health Sciences Co., Ltd, 528451, China
| | - Xiaomin Li
- Perfect (China) Co., Ltd, Zhongshan, 528400, China
- Perfect Life and Health Sciences Co., Ltd, 528451, China
| | - Xinliang Mao
- Perfect (China) Co., Ltd, Zhongshan, 528400, China
- Perfect Life and Health Sciences Co., Ltd, 528451, China
| | - Lin Zheng
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China.
| | - Mouming Zhao
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China.
| |
Collapse
|
7
|
Khumukcham SS, Penugurti V, Bugide S, Dwivedi A, Kumari A, Kesavan PS, Kalali S, Mishra YG, Ramesh VA, Nagarajaram HA, Mazumder A, Manavathi B. HPIP and RUFY3 are noncanonical guanine nucleotide exchange factors of Rab5 to regulate endocytosis-coupled focal adhesion turnover. J Biol Chem 2023; 299:105311. [PMID: 37797694 PMCID: PMC10641178 DOI: 10.1016/j.jbc.2023.105311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 09/01/2023] [Accepted: 09/15/2023] [Indexed: 10/07/2023] Open
Abstract
While the role of endocytosis in focal adhesion turnover-coupled cell migration has been established in addition to its conventional role in cellular functions, the molecular regulators and precise molecular mechanisms that underlie this process remain largely unknown. In this study, we report that proto-oncoprotein hematopoietic PBX-interacting protein (HPIP) localizes to focal adhesions as well as endosomal compartments along with RUN FYVE domain-containing protein 3 (RUFY3) and Rab5, an early endosomal protein. HPIP contains two coiled-coil domains (CC1 and CC2) that are necessary for its association with Rab5 and RUFY3 as CC domain double mutant, that is, mtHPIPΔCC1-2 failed to support it. Furthermore, we show that HPIP and RUFY3 activate Rab5 by serving as noncanonical guanine nucleotide exchange factors of Rab5. In support of this, either deletion of coiled-coil domains or silencing of HPIP or RUFY3 impairs Rab5 activation and Rab5-dependent cell migration. Mechanistic studies further revealed that loss of HPIP or RUFY3 expression severely impairs Rab5-mediated focal adhesion disassembly, FAK activation, fibronectin-associated-β1 integrin trafficking, and thus cell migration. Together, this study underscores the importance of HPIP and RUFY3 as noncanonical guanine nucleotide exchange factors of Rab5 and in integrin trafficking and focal adhesion turnover, which implicates in cell migration.
Collapse
Affiliation(s)
| | - Vasudevarao Penugurti
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | - Suresh Bugide
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | - Anju Dwivedi
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | - Anita Kumari
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | - P S Kesavan
- Department of Biological Sciences, Tata Institute of Fundamental Research (TIFR), Hyderabad, Telangana, India
| | - Sruchytha Kalali
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | - Yasaswi Gayatri Mishra
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | - Vakkalagadda A Ramesh
- Laboratory of Computational Biology, Centre for DNA Finger Printing and Diagnostics (CDFD), Hyderabad, Telangana, India; Laboratory of Computational Biology, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | | | - Aprotim Mazumder
- Department of Biological Sciences, Tata Institute of Fundamental Research (TIFR), Hyderabad, Telangana, India
| | - Bramanandam Manavathi
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India.
| |
Collapse
|
8
|
Xu R, Wu J, Zheng L, Zhao M. Undenatured type II collagen and its role in improving osteoarthritis. Ageing Res Rev 2023; 91:102080. [PMID: 37774932 DOI: 10.1016/j.arr.2023.102080] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/13/2023] [Accepted: 09/25/2023] [Indexed: 10/01/2023]
Abstract
Osteoarthritis (OA) is a degenerative joint disease, affecting 32.5 million US adults or 242 million people worldwide. There is no cure for OA. Many animal and clinical trials showed that oral administration of undenatured type II collagen could significantly reduce the incidence of OA or alleviate the symptoms of articular cartilage. Type II collagen is an important component of cartilage matrix. This article reviewed research progress of undenatured type II collagen including its methods of extraction and preparation, structure and characterization, solubility, thermal stability, gastrointestinal digestive stability, its role in improving OA, and the mechanism of its action in improving OA. Type II collagen has been extensively explored for its potential in improving arthritis. Methods of extraction of type II collagen are inefficient and tedious. The method of limited enzymatic hydrolysis is mainly used to prepare soluble undenatured type II collagen (SC II). The solubility, thermal and gastrointestinal digestive stability of SC II are affected by the sources of raw material, pH, salt ions, and temperature. Oral administration of undenatured type II collagen improves OA, whereas its activity is affected by the sources, degree of denaturalization, intervention methods and doses. However, the influence of the structure of undenatured type II collagen on its activity and the mechanism are unclear. The findings in this review support that undenatured type II collagen can be used in the intervention or auxiliary intervention of patients with OA.
Collapse
Affiliation(s)
- Rong Xu
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China; Guangdong Food Green Processing and Nutrition Regulation Technologies Research Center, Guangzhou 510650, China
| | - Jianping Wu
- Department of Agricultural, Food and Nutritional Science, 4-10 Ag/For Building, University of Alberta, Edmonton, Alberta T6G 2P5, Canada
| | - Lin Zheng
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China; Guangdong Food Green Processing and Nutrition Regulation Technologies Research Center, Guangzhou 510650, China.
| | - Mouming Zhao
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China; Guangdong Food Green Processing and Nutrition Regulation Technologies Research Center, Guangzhou 510650, China.
| |
Collapse
|
9
|
Martin-Vega A, Cobb MH. Navigating the ERK1/2 MAPK Cascade. Biomolecules 2023; 13:1555. [PMID: 37892237 PMCID: PMC10605237 DOI: 10.3390/biom13101555] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
The RAS-ERK pathway is a fundamental signaling cascade crucial for many biological processes including proliferation, cell cycle control, growth, and survival; common across all cell types. Notably, ERK1/2 are implicated in specific processes in a context-dependent manner as in stem cells and pancreatic β-cells. Alterations in the different components of this cascade result in dysregulation of the effector kinases ERK1/2 which communicate with hundreds of substrates. Aberrant activation of the pathway contributes to a range of disorders, including cancer. This review provides an overview of the structure, activation, regulation, and mutational frequency of the different tiers of the cascade; with a particular focus on ERK1/2. We highlight the importance of scaffold proteins that contribute to kinase localization and coordinate interaction dynamics of the kinases with substrates, activators, and inhibitors. Additionally, we explore innovative therapeutic approaches emphasizing promising avenues in this field.
Collapse
Affiliation(s)
- Ana Martin-Vega
- Department of Pharmacology, UT Southwestern Medical Center, 6001 Forest Park Rd., Dallas, TX 75390, USA;
| | - Melanie H. Cobb
- Department of Pharmacology, UT Southwestern Medical Center, 6001 Forest Park Rd., Dallas, TX 75390, USA;
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, 6001 Forest Park Rd., Dallas, TX 75390, USA
| |
Collapse
|
10
|
Liu L, Tang H, Wang Y. Nanotechnology-Boosted Biomaterials for Osteoarthritis Treatment: Current Status and Future Perspectives. Int J Nanomedicine 2023; 18:4969-4983. [PMID: 37693887 PMCID: PMC10487746 DOI: 10.2147/ijn.s423737] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/14/2023] [Indexed: 09/12/2023] Open
Abstract
Osteoarthritis (OA) is a prevalent global health concern, posing a significant and increasing public health challenge worldwide. Recently, nanotechnology-boosted biomaterials have emerged as a highly promising strategy for OA therapy due to their exceptional physicochemical properties and capacity to regulate pathological processes. However, there is an urgent need for a deeper understanding of the potential therapeutic applications of these biomaterials in the clinical management of diseases, particularly in the treatment of OA. In this comprehensive review, we present an extensive discussion of the current status and future prospects concerning nanotechnology-boosted biomaterials for OA therapy. Initially, we discuss the pathophysiology of OA and the constraints associated with existing treatment modalities. Subsequently, various types of nanomaterials utilized for OA therapy, including nanoparticles, nanofibers, and nanocomposites, are thoroughly discussed and summarized, elucidating their respective advantages and challenges. Furthermore, we analyze recent preclinical and clinical studies that highlight the potential of nanotechnology-boosted biomaterials in OA therapy. Additionally, future research directions in this evolving field are highlighted. By establishing a link between the structural properties of nanotechnology-boosted biomaterials and their therapeutic functions in OA treatment, we aim to foster advances in designing sophisticated nanomaterials for OA, ultimately resulting in improved therapeutic efficacy of OA therapy through translation into clinical setting in the near future.
Collapse
Affiliation(s)
- Lin Liu
- Department of Emergency, Honghui Hospital of Xi’an Jiaotong University, Xi’an, 710054, People’s Republic of China
| | - Haifeng Tang
- Department of Emergency, Honghui Hospital of Xi’an Jiaotong University, Xi’an, 710054, People’s Republic of China
| | - Yanjun Wang
- Department of Emergency, Honghui Hospital of Xi’an Jiaotong University, Xi’an, 710054, People’s Republic of China
| |
Collapse
|
11
|
Jiang Q, Zhang D, Liu J, Liang C, Yang R, Zhang C, Wu J, Lin J, Ye T, Ding L, Li J, Gao S, Li B, Ye Q. HPIP is an essential scaffolding protein running through the EGFR-RAS-ERK pathway and drives tumorigenesis. SCIENCE ADVANCES 2023; 9:eade1155. [PMID: 37294756 PMCID: PMC10256163 DOI: 10.1126/sciadv.ade1155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 05/04/2023] [Indexed: 06/11/2023]
Abstract
The EGFR-RAS-ERK pathway plays a key role in cancer development and progression. However, the integral assembly of EGFR-RAS-ERK signaling complexes from the upstream component EGFR to the downstream component ERK is largely unknown. Here, we show that hematopoietic PBX-interacting protein (HPIP) interacts with all classical components of the EGFR-RAS-ERK pathway and forms at least two complexes with overlapping components. Experiments of HPIP knockout or knockdown and chemical inhibition of HPIP expression showed that HPIP is required for EGFR-RAS-ERK signaling complex formation, EGFR-RAS-ERK signaling activation, and EGFR-RAS-ERK signaling-mediated promotion of aerobic glycolysis as well as cancer cell growth in vitro and in vivo. HPIP expression is correlated with EGFR-RAS-ERK signaling activation and predicts worse clinical outcomes in patients with lung cancer. These results provide insights into EGFR-RAS-ERK signaling complex formation and EGFR-RAS-ERK signaling regulation and suggest that HPIP may be a promising therapeutic target for cancer with dysregulated EGFR-RAS-ERK signaling.
Collapse
Affiliation(s)
- Qiwei Jiang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Bejing 100850, China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China
| | - Deyu Zhang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Bejing 100850, China
| | - Juan Liu
- Department of Cell Engineering, Beijing Institute of Biotechnology, Bejing 100850, China
- Department of Hematology, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China
| | - Chaoyang Liang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Bejing 100850, China
- Department of Thoracic Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Ronghui Yang
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Cheng Zhang
- Outpatient Department, Jingnan Medical Area, Chinese PLA General Hospital, Beijing 100850, China
| | - Jun Wu
- Department of Microorganism Engineering, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Jing Lin
- Department of Cell Engineering, Beijing Institute of Biotechnology, Bejing 100850, China
- Department of Clinical Laboratory, The Fourth Medical Center, Chinese PLA General Hospital, Beijing 100048, China
| | - Tianxing Ye
- Department of Cell Engineering, Beijing Institute of Biotechnology, Bejing 100850, China
| | - Lihua Ding
- Department of Cell Engineering, Beijing Institute of Biotechnology, Bejing 100850, China
| | - Jianbin Li
- Department of Cell Engineering, Beijing Institute of Biotechnology, Bejing 100850, China
| | - Shan Gao
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing 210096, China
| | - Binghui Li
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Qinong Ye
- Department of Cell Engineering, Beijing Institute of Biotechnology, Bejing 100850, China
| |
Collapse
|
12
|
Xie D, Song L, Xiang D, Gao X, Zhao W. Salvianolic acid A alleviates atherosclerosis by inhibiting inflammation through Trc8-mediated 3-hydroxy-3-methylglutaryl-coenzyme A reductase degradation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 112:154694. [PMID: 36804757 DOI: 10.1016/j.phymed.2023.154694] [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: 05/31/2022] [Revised: 01/12/2023] [Accepted: 02/02/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Atherosclerosis is the most prevalent cardiovascular disease and remains the major contributor to death and mortality globally. Salvianolic acid A (SalA) is a water-soluble phenolic acid that benefits atherosclerosis. However, the mechanisms of SalA protecting against atherosclerosis remain unclear. PURPOSE We aimed to determine whether SalA prevents atherosclerosis by modulating 3-Hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) degradation via the ubiquitin-proteasomal pathway. METHODS The animal and cellular models of atherosclerosis were established by subjecting apolipoprotein E (ApoE) knockout mice to a high-fat diet (HFD) and exposing human umbilical vein endothelial cells (HUVECs) to oxidized low-density lipoprotein (ox-LDL), respectively. RESULTS Our results showed that similar to atorvastatin, SalA suppressed atherosclerotic plaque formation, improved serum lipid accumulation, and reduced cholesterol levels in HFD-fed ApoE-/- mice. Moreover, SalA protected HUVECs from ox-LDL-caused cell viability reduction and lipid accumulation. The mechanism study revealed that SalA reduced the production of proinflammatory cytokines tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, and IL-6, and augmented the generation of the anti-inflammatory cytokine IL-10 in ApoE-/- mice and HUVECs, accompanied by increased HMGCR ubiquitination and degradation via translocation in renal carcinoma on chromosome 8 (Trc8), insulin-induced gene (Insig)1 and Insig2. Furthermore, the knockdown of Trc8 abolished the SalA-induced HMGCR degradation and anti-atherosclerosis activity. CONCLUSION SalA rescues atherosclerosis by inhibiting inflammation through the Trc8-regulated degradation of HMGCR. These findings underscore Trc8 as a potential target of atherosclerosis.
Collapse
Affiliation(s)
- Dan Xie
- Emergency Department, Kunshan Hospital Affiliated to Nanjing University of Chinese Medicine, Kunshan, Jiangsu 215300, China
| | - Lijun Song
- School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, China
| | - Dongyang Xiang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550000, China
| | - Xiangyu Gao
- Emergency Department, Kunshan Hospital Affiliated to Nanjing University of Chinese Medicine, Kunshan, Jiangsu 215300, China.
| | - Wenchang Zhao
- School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, Guangdong 524023, China.
| |
Collapse
|
13
|
Jin J, Liu Y, Jiang C, Shen Y, Chu G, Liu C, Jiang L, Huang G, Qin Y, Zhang Y, Zhang C, Wang Y. Arbutin-modified microspheres prevent osteoarthritis progression by mobilizing local anti-inflammatory and antioxidant responses. Mater Today Bio 2022; 16:100370. [PMID: 35937573 PMCID: PMC9352975 DOI: 10.1016/j.mtbio.2022.100370] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/29/2022] [Accepted: 07/14/2022] [Indexed: 12/03/2022] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease worldwide and currently there is no effective strategy to stop its progression. It is known that oxidative stress and inflammation can promote the development of OA, and therapeutic strategies against these conditions may alleviate OA. Arbutin (ARB), a major ingredient of the Chinese medicinal herb cowberry leaf, exerts good antioxidant and anti-inflammatory activities yet has not been studied in OA. Here we developed ARB-loaded gelatine methacryloyl-Liposome (GM-Lipo@ARB) microspheres which showed long-term release of ARB and excellent cartilage-targeting effects. The ARB-loaded microspheres effectively reduced the inflammatory response in interleukin (IL)-1β-treated arthritic chondrocytes. Moreover, the synthesized GM-Lipo@ARB microspheres regulated cartilage extracellular matrix (ECM) homeostasis through anti-inflammation effect via inhibiting NF-κB signaling and anti-oxidative stress effect via activating Nrf2 pathway. Intra-articular use of GM-Lipo@ARB can effectively reduce inflammation and oxidative stress in the articular cartilage and thus, attenuating OA progression in a mouse model. The study proposed a novel ARB-laden functional microsphere, GM-Lipo@ARB, and demonstrated that this compound may be used as an alternative therapeutics for treating OA.
Collapse
|
14
|
Chen J, Chen Z, Yuan P, Huang H, Wang J, Shi P, Sun X. ERK1 loss accelerates the progression of osteoarthritis in aged mice via NRF2/BACH1 signaling. Biochem Biophys Res Commun 2022; 622:129-135. [PMID: 35849954 DOI: 10.1016/j.bbrc.2022.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/02/2022] [Accepted: 07/04/2022] [Indexed: 12/14/2022]
Abstract
Osteoarthritis (OA) is now a common degenerative joint related disease. However, the clinical efficacy of drugs associated with cartilage regeneration remains limited. In our study, we firstly explored the role of ERK1 in the progression of OA. We clarified that ERK1-deficient mice were susceptible to age-related OA. The higher OARSI scores and more severe cartilage degeneration was observed in the ERK1-deficient mice. ERK1 deficiency decreased the nuclear transportation of Nrf2 in the chondrocytes and accelerated chondrocyte aging in vitro. Moreover, chondrocytes with ERK1 deficiency elevated the nuclear expression of BACH1, resulting in lowered expression of antioxidant enzymes in ERK1-deficient chondrocytes. The Nrf2 activator dimethyl fumarate (DMF) was used. Our experiments demonstrated the protective function of DMF against OA in ERK1 knockout mice. Above all, we confirmed the effects of ERK1 on the progression of OA and clarified the mechanisms underlying these effects. DMF might has significant use in the development of novel drugs for the therapy of OA in the future.
Collapse
Affiliation(s)
- Jian Chen
- Department of Orthopaedic, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China; Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Zhijun Chen
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China; Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Putao Yuan
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China; Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Hai Huang
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Jiying Wang
- Department of Orthopaedic, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Peihua Shi
- Department of Orthopaedic, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China; Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China.
| | - Xuewu Sun
- Department of Orthopaedic, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China; Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
15
|
Bai RJ, Liu D, Li YS, Tian J, Yu DJ, Li HZ, Zhang FJ. OPN inhibits autophagy through CD44, integrin and the MAPK pathway in osteoarthritic chondrocytes. Front Endocrinol (Lausanne) 2022; 13:919366. [PMID: 36034459 PMCID: PMC9411521 DOI: 10.3389/fendo.2022.919366] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Background To investigate whether osteopontin (OPN) affects autophagy in human osteoarthritic chondrocytes and determine the roles of CD44, αvβ3 integrin and the Mitogen-activated protein kinase (MAPK) pathway in this progress. Methods First, we compared the autophagy levels in the human osteoarthritis (OA) and normal cartilage, then, we cultured human OA chondrocytes in vitro and treated cells with recombinant human OPN (rhOPN) to determine autophagy changes. Next, the anti-CD44 and anti-CD51/61 monoclonal antibodies (Abs) or isotype IgG were used to determine the possible role of CD44 and αvβ3 integrin; subsequently, an inhibitor of the ERK MAPK pathway was used to investigate the role of ERK MAPK. Western blotting was used to measure the Beclin1, LC3 II and MAPK proteins expressions, mRFP-GFP-LC3 confocal imaging and transmission electron microscopy were also used to detect the autophagy levels. Cell Counting Kit-8 (CCK-8) was used to assay the proliferation and activity of chondrocytes. Results The LC3 protein was greatly decreased in OA cartilage compared to normal cartilage, and OPN suppressed the autophagy activity in chondrocytes in vitro. Blocking experiments with anti-CD44 and anti-CD51/61 Abs indicated that OPN could suppress the expression of LC3II and Beclin1 through αvβ3 integrin and CD44. Our results also indicated that the ratio of p-ERK/ERK but not p-P38/P38 and p-JNK/JNK was increased after the rhOPN treatment. The ERK inhibitor inhibited the activity of OPN in the suppression of autophagy, and the CCK-8 results showed that rhOPN could promote chondrocyte proliferation. Conclusion OPN inhibited chondrocyte autophagy through CD44 and αvβ3 integrin receptors and via the ERK MAPK signaling pathway.
Collapse
Affiliation(s)
- Rui-Jun Bai
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Di Liu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Yu-Sheng Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jian Tian
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Deng-Jie Yu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Heng-Zhen Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Fang-Jie Zhang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Emergency Medicine, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
16
|
Ran Q, Xu D, Wang Q, Wang D. Hypermethylation of the Promoter Region of miR-23 Enhances the Metastasis and Proliferation of Multiple Myeloma Cells via the Aberrant Expression of uPA. Front Oncol 2022; 12:835299. [PMID: 35707350 PMCID: PMC9189361 DOI: 10.3389/fonc.2022.835299] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 05/02/2022] [Indexed: 11/24/2022] Open
Abstract
Multiple myeloma has a long course, with no obvious symptoms in the early stages. However, advanced stages are characterized by injury to the bone system and represent a severe threat to human health. The results of the present work indicate that the hypermethylation of miR-23 promoter mediates the aberrant expression of uPA/PLAU (urokinase plasminogen activator, uPA) in multiple myeloma cells. miR-23, a microRNA that potentially targets uPA’s 3’UTR, was predicted by the online tool miRDB. The endogenous expressions of uPA and miR-23 are related to disease severity in human patients, and the expression of miR-23 is negatively related to uPA expression. The hypermethylation of the promoter region of miR-23 is a promising mechanism to explain the low level of miR-23 or aberrant uPA expression associated with disease severity. Overexpression of miR-23 inhibited the expression of uPA by targeting the 3’UTR of uPA, not only in MM cell lines, but also in patient-derived cell lines. Overexpression of miR-23 also inhibited in vitro and in vivo invasion of MM cells in a nude mouse model. The results therefore extend our knowledge about uPA in MM and may assist in the development of more effective therapeutic strategies for MM treatment.
Collapse
Affiliation(s)
- Qijie Ran
- Department of Hematology, General Hospital of Central Theater Command, Wuhan, China
- *Correspondence: Qijie Ran, ; Dongsheng Wang,
| | - Dehong Xu
- Department of Hematology, General Hospital of Central Theater Command, Wuhan, China
| | - Qi Wang
- Department of Hematology, General Hospital of Central Theater Command, Wuhan, China
| | - Dongsheng Wang
- Department of Neurosurgery, The Fifth People’s Hospital of Dalian, Dalian, China
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian City, China
- *Correspondence: Qijie Ran, ; Dongsheng Wang,
| |
Collapse
|
17
|
Yang L, Ren Z, Yan S, Zhao L, Liu J, Zhao L, Li Z, Ye S, Liu A, Li X, Guo J, Zhao W, Kuang W, Liu H, Chen D. Nsun4 and Mettl3 mediated translational reprogramming of Sox9 promotes BMSC chondrogenic differentiation. Commun Biol 2022; 5:495. [PMID: 35614315 PMCID: PMC9133052 DOI: 10.1038/s42003-022-03420-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/27/2022] [Indexed: 11/09/2022] Open
Abstract
The chondrogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) has been used in the treatment and repair of cartilage defects; however, the in-depth regulatory mechanisms by which RNA modifications are involved in this process are still poorly understood. Here, we found that Sox9, a critical transcription factor that mediates chondrogenic differentiation, exhibited enhanced translation by ribosome sequencing in chondrogenic pellets, which was accompanied by increased 5-methylcytosine (m5C) and N6-methyladenosine (m6A) levels. Nsun4-mediated m5C and Mettl3-mediated m6A modifications were required for Sox9-regulated chondrogenic differentiation. Interestingly, we showed that in the 3’UTR of Sox9 mRNA, Nsun4 catalyzed the m5C modification and Mettl3 catalyzed the m6A modification. Furthermore, we found that Nsun4 and Mettl3 co-regulated the translational reprogramming of Sox9 via the formation of a complex. Surface plasmon resonance (SPR) assays showed that this complex was assembled along with the recruitment of Ythdf2 and eEF1α-1. Moreover, BMSCs overexpressing Mettl3 and Nsun4 can promote the repair of cartilage defects in vivo. Taken together, our study demonstrates that m5C and m6A co-regulate the translation of Sox9 during the chondrogenic differentiation of BMSCs, which provides a therapeutic target for clinical implications. Nsun4-mediated m5C and Mettl3-mediated m6A are found to be required for Sox9-regulated chondrogenic differentiation, whereby Nsun4 and Mettl3 interact with each other and recruit Ythdf2 and eEF1a-1 to form a complex at the 3’UTR of Sox9.
Collapse
Affiliation(s)
- Lin Yang
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, 518101, Guangdong, China
| | - Zhenxing Ren
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Shenyu Yan
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, 61001-89999, China
| | - Ling Zhao
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jie Liu
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, 518101, Guangdong, China
| | - Lijun Zhao
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, 518101, Guangdong, China
| | - Zhen Li
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, 518101, Guangdong, China
| | - Shanyu Ye
- Department of Anatomy, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Aijun Liu
- Department of Anatomy, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xichan Li
- School of Chinese Herbal Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jiasong Guo
- Department of Histology and Embryology, Southern Medical University, Guangzhou, 510515, China
| | - Wei Zhao
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Weihong Kuang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Key Laboratory of Research and Development of New Medical Materials of Guangdong Medical University, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Helu Liu
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, 518101, Guangdong, China.
| | - Dongfeng Chen
- Department of Anatomy, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
18
|
Li Y, Xie W, Xiao W, Dou D. Progress in osteoarthritis research by the National Natural Science Foundation of China. Bone Res 2022; 10:41. [PMID: 35610209 PMCID: PMC9130253 DOI: 10.1038/s41413-022-00207-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 12/24/2021] [Accepted: 02/17/2022] [Indexed: 01/10/2023] Open
Abstract
Osteoarthritis (OA) in China is gradually becoming an important scientific research area that has had a significant impact on research and development (R&D) activities in the OA field worldwide. This article summarizes the R&D progress related to OA in China in recent years. The National Natural Science Foundation of China (NSFC) is a national funding institution for basic research and plays a critical role in promoting and supporting Chinese scholars' R&D activities. We collected and analyzed information on NSFC funding in the field of OA from 2010 to 2019, including the amount, the level and the program categories of the funded projects. The data fully demonstrate the important and positive role of the NSFC in supporting free exploration, cultivating research teams and young talent, and boosting OA R&D. In this article, we outline and discuss hot topics in focused areas, key advances in this field and the prospects for progress in OA research in China.
Collapse
Affiliation(s)
- Yusheng Li
- Deparment of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Wenqing Xie
- Deparment of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Wenfeng Xiao
- Deparment of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Dou Dou
- Department of Health Sciences, National Natural Science Foundation of China, Beijing, 100085, China.
| |
Collapse
|
19
|
Jia Z, Kang B, Cai Y, Chen C, Yu Z, Li W, Zhang W. Cell-free fat extract attenuates osteoarthritis via chondrocytes regeneration and macrophages immunomodulation. Stem Cell Res Ther 2022; 13:133. [PMID: 35365233 PMCID: PMC8973552 DOI: 10.1186/s13287-022-02813-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/02/2022] [Indexed: 01/15/2023] Open
Abstract
Background The prevalence of osteoarthritis (OA) is increasing, yet clinically effective and economical treatments are unavailable. We have previously proposed a cell-free fat extract (CEFFE) containing multiple cytokines, which possessed antiapoptotic, anti-oxidative, and proliferation promotion functions, as a “cell-free” strategy. In this study, we aimed to evaluate the therapeutic effect of CEFFE in vivo and in vitro. Methods In vivo study, sodium iodoacetate-induced OA rats were treated with CEFFE by intra-articular injections for 8 weeks. Behavioral experiments were performed every two weeks. Histological analyses, anti-type II collagen, and toluidine staining provided structural evaluation. Macrophage infiltration was assessed by anti-CD68 and anti-CD206 staining. In vitro study, the effect of CEFFE on macrophage polarization and secretory factors was evaluated by flow cytometry, immunofluorescence, and quantitative reverse-transcription polymerase chain reaction (qRT-PCR). The effect of CEFFE on cartilage regeneration was accessed by cell counting kit-8 assay and qRT-PCR. The generation of reactive oxygen species (ROS) and levels of ROS-related enzymes were investigated by qRT-PCR and western blotting. Results In rat models with sodium iodoacetate (MIA)-induced OA, CEFFE increased claw retraction pressure while decreasing bipedal pressure in a dose-dependent manner. Moreover, CEFFE promoted cartilage structure restoration and increased the proportion of CD206+ macrophages in the synovium. In vitro, CEFFE decreased the proportion of CD86+ cells and reduced the expression of pro-inflammatory factors in LPS + IFN-γ induced Raw 264.7. In addition, CEFFE decreased the expression of interleukin-6 and ADAMTs-5 and promoted the expression of SOX-9 in mouse primary chondrocytes. Besides, CEFFE reduced the intracellular levels of reactive oxygen species in both in vitro models through regulating ROS-related enzymes. Conclusions CEFFE inhibits the progression of OA by promoting cartilage regeneration and limiting low-grade joint inflammation. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02813-3.
Collapse
Affiliation(s)
- Zhuoxuan Jia
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, National Tissue Engineering Center of China, 639 ZhiZaoJu Road, Shanghai, 200011, China
| | - Bijun Kang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, National Tissue Engineering Center of China, 639 ZhiZaoJu Road, Shanghai, 200011, China
| | - Yizuo Cai
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, National Tissue Engineering Center of China, 639 ZhiZaoJu Road, Shanghai, 200011, China
| | - Chingyu Chen
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, National Tissue Engineering Center of China, 639 ZhiZaoJu Road, Shanghai, 200011, China
| | - Zheyuan Yu
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, National Tissue Engineering Center of China, 639 ZhiZaoJu Road, Shanghai, 200011, China.
| | - Wei Li
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, National Tissue Engineering Center of China, 639 ZhiZaoJu Road, Shanghai, 200011, China.
| | - Wenjie Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, National Tissue Engineering Center of China, 639 ZhiZaoJu Road, Shanghai, 200011, China.
| |
Collapse
|
20
|
Effects of the Leptin-Mediated MAPK/ERK Signaling Pathway on Collagen II Expression in Knee Cartilage of Newborn Male Mice from Obese Maternal Offspring. Biomolecules 2022; 12:biom12030477. [PMID: 35327669 PMCID: PMC8946789 DOI: 10.3390/biom12030477] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/15/2022] [Accepted: 03/19/2022] [Indexed: 12/19/2022] Open
Abstract
Epidemiological data suggest that various noncommunicable diseases develop as a result of altered maternal metabolic and physiological status due to exposure to several adverse factors during pregnancy. However, evidence for intrauterine exposure factors and mechanisms underlying the origin of early cartilage disease in chronic osteoarthritic disease is still lacking. In this study, we found that persistent overnutrition during pregnancy in obese mothers led to cartilage damage in neonatal male mice. This was mainly characterized by increased apoptosis with decreased expression of chondrocyte collagen II and low expression of Runx family transcription factor 2 (RUNX2) and SRY-box transcription factor 9 (SOX9). This reduction was also found to be associated with high leptin expression in newborn male mice of obese maternal offspring. Furthermore, the administration of leptin and mitogen-activated protein kinase (MAPK)-extracellular signal-regulated kinase (ERK) inhibitors in primary chondrocytes showed that leptin mediated MAPK/ERK signaling activation and thus affected the key regulators of cartilage matrix metallopeptidase 1 (MMP1) and tissue inhibitor of metalloproteinase 1 (TIMP1), thereby altering the expression of collagen II in mouse cartilage. Altogether, this study provided insights into the molecular mechanisms of cartilage-related disease development and also new clues and evidence for the fetogenetic origin of cartilage diseases.
Collapse
|
21
|
Kang D, Lee J, Jung J, Carlson BA, Chang MJ, Chang CB, Kang SB, Lee BC, Gladyshev VN, Hatfield DL, Lee BJ, Kim JH. Selenophosphate synthetase 1 deficiency exacerbates osteoarthritis by dysregulating redox homeostasis. Nat Commun 2022; 13:779. [PMID: 35140209 PMCID: PMC8828855 DOI: 10.1038/s41467-022-28385-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 01/24/2022] [Indexed: 12/13/2022] Open
Abstract
Aging and mechanical overload are prominent risk factors for osteoarthritis (OA), which lead to an imbalance in redox homeostasis. The resulting state of oxidative stress drives the pathological transition of chondrocytes during OA development. However, the specific molecular pathways involved in disrupting chondrocyte redox homeostasis remain unclear. Here, we show that selenophosphate synthetase 1 (SEPHS1) expression is downregulated in human and mouse OA cartilage. SEPHS1 downregulation impairs the cellular capacity to synthesize a class of selenoproteins with oxidoreductase functions in chondrocytes, thereby elevating the level of reactive oxygen species (ROS) and facilitating chondrocyte senescence. Cartilage-specific Sephs1 knockout in adult mice causes aging-associated OA, and augments post-traumatic OA, which is rescued by supplementation of N-acetylcysteine (NAC). Selenium-deficient feeding and Sephs1 knockout have synergistic effects in exacerbating OA pathogenesis in mice. Therefore, we propose that SEPHS1 is an essential regulator of selenium metabolism and redox homeostasis, and its dysregulation governs the progression of OA. Osteoarthritis is caused by the gradual accumulation of oxidative stress in cartilage. Here, the authors show that dysregulation of the selenium metabolic pathway underlies a shift in redox homeostasis in chondrocytes, leading to chronic osteoarthritic changes in joints.
Collapse
Affiliation(s)
- Donghyun Kang
- Center for RNA Research, Institute for Basic Science, Seoul, 08826, South Korea.,Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Jeeyeon Lee
- Center for RNA Research, Institute for Basic Science, Seoul, 08826, South Korea.,Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Jisu Jung
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Bradley A Carlson
- Mouse Cancer Genetics Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Moon Jong Chang
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, Boramae Hospital, Seoul, 07061, South Korea
| | - Chong Bum Chang
- Department of Orthopaedic Surgery, Seoul National University Bundang Hospital, Seongnam, 13620, South Korea
| | - Seung-Baik Kang
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, Boramae Hospital, Seoul, 07061, South Korea
| | - Byung Cheon Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Dolph L Hatfield
- Mouse Cancer Genetics Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Byeong Jae Lee
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea. .,Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, South Korea.
| | - Jin-Hong Kim
- Center for RNA Research, Institute for Basic Science, Seoul, 08826, South Korea. .,Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea. .,Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
22
|
Contribution of ferroptosis and GPX4's dual functions to osteoarthritis progression. EBioMedicine 2022; 76:103847. [PMID: 35101656 PMCID: PMC8822178 DOI: 10.1016/j.ebiom.2022.103847] [Citation(s) in RCA: 192] [Impact Index Per Article: 96.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 01/13/2022] [Accepted: 01/13/2022] [Indexed: 12/19/2022] Open
Abstract
Background Osteoarthritis (OA) is the most common joint disease and is the leading cause of chronic disability among older people. Chondrocyte death and extracellular matrix (ECM) degradation was involved in OA pathogenesis. Ferroptosis was an iron-dependent cell death associated with peroxidation of lipids. Here, we proved that ferroptosis exists in OA and identified glutathione peroxidase 4 (GPX4) as an important regulator of OA. Methods Ferroptosis-related alterations were analyzed in human OA and undamaged cartilage. Expression of GPX4 was examined in 55 paired human OA samples. Ferrostatin-1 (Fer-1) and Deferoxamine (DFO) were used to treat OA, in vitro and in vivo. Alterations of GPX4-mediated signaling pathway were identified by RNA-seq analysis. AAV-Gpx4-shRNA were used to downregulate GPX4 expression in vivo. Findings Transcriptomic, biochemical, and microscopical analyses indicated that ferroptosis was closely associated with OA. Expression of GPX4 in the OA cartilage from 55 OA patients were significantly lower than undamaged cartilage. Fer-1 and DFO could protect OA in a necroptosis-independent manner, suggesting that ferroptosis exists in OA prog. Importantly, GPX4 downregulation could increase the sensitivity of chondrocytes to oxidative stress and aggravate ECM degradation through the MAPK/NFκB pathway. Furthermore, downregulation of GPX4 expression by AAV-Gpx4 shRNA aggravated OA in vivo. Interpretation Ferroptosis contributes to OA pathogenesis and GPX4 was the intersection of two mechanisms in regulating OA progression: ferroptosis and ECM degradation.
Collapse
|
23
|
KLF4, negatively regulated by miR-7, suppresses osteoarthritis development via activating TGF-β1 signaling. Int Immunopharmacol 2021; 102:108416. [PMID: 34891002 DOI: 10.1016/j.intimp.2021.108416] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 11/12/2021] [Accepted: 11/25/2021] [Indexed: 12/20/2022]
Abstract
Osteoarthritis (OA) is a chronic degenerative disease which seriously affects the patients' daily activities and quality of life. In our previous findings, we demonstrated that overexpression of miR-7 was found in OA and promoted OA development. Its exact mechanism remains unclear. Herein, we confirmed that KLF4 was the target gene of miR-7 and KLF4 was down-regulated in human OA tissues and OA chondrocyte. KLF4 was negatively modulated by miR-7 via dual luciferase reporter assay. Cartilage-specific genes (SOX9, COL2A1, RUNX2, MMP13) are crucial regulators in cartilage degeneration. Through qRT-PCR and western blot, we observed that KLF4 overexpression could increase the expression of SOX9 and COL2A1, decrease RUNX2 and MMP13. In the meanwhile, miR-7 was proven to regulate the expression of the above cartilage-specific genes by targeting KLF4, which demonstrated KLF4 could prevent OA development. Subsequently, KLF4 also activated TGF-β1 signaling pathway, thereby affecting OA progression. Excessive KLF4 could up-regulate TGF-β1 and p-Smad2/3 level, and Smad4 level was prevented in OA chondrocytes, while adding TGF-β1 inhibitor SB525334 could rescue this impact, along with reduced TGF-β1 and p-Smad2/3 level, enriched Smad4 level. KLF4 could also reverse the effect of miR-7 on TGF-β1 signaling. Besides, it was confirmed that KLF4 could improve OA in rat OA models by HE and Safranin O-Fast green staining, and immunohistochemistry. Collectively, our findings will give more detailed evidence about miR-7 and KLF4 in OA diagnosis and treatment.
Collapse
|
24
|
Liu J, Yang C, Huang XM, Lv PP, Yang YK, Zhao JN, Zhao SY, Sun WJ. Knockdown of FBI-1 Inhibits the Warburg Effect and Enhances the Sensitivity of Hepatocellular Carcinoma Cells to Molecular Targeted Agents via miR-3692/HIF-1α. Front Oncol 2021; 11:796839. [PMID: 34869045 PMCID: PMC8633402 DOI: 10.3389/fonc.2021.796839] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 10/25/2021] [Indexed: 01/20/2023] Open
Abstract
The transcription suppressor factor FBI-1 (the factor that binds to inducer of short transcripts-1) is an important regulator of hepatocellular carcinoma (HCC). In this work, the results showed that FBI-1 promoted the Warburg effect and enhances the resistance of hepatocellular carcinoma cells to molecular targeted agents. Knockdown of FBI-1 via its small-interfering RNA (siRNA) inhibited the ATP level, lactate productions, glucose uptake or lactate dehydrogenase (LDH) activation of HCC cells. Transfection of siFBI-1 also decreased the expression of the Warburg-effect-related factors: hypoxia-inducible factor-1 alpha (HIF-1α), lactate dehydrogenase A (LDHA), or GLUT1, and the epithelial-mesenchymal transition-related factors, Vimentin or N-cadherin. The positive correlation between the expression of FBI-1 with HIF-1α, LDHA, or GLUT1 was confirmed in HCC tissues. Mechanistically, the miR-30c repressed the expression of HIF-1α by binding to the 3'-untranslated region (3'-UTR) of HIF-1α in a sequence-specific manner, and FBI-1 enhanced the expression of HIF-1α and HIF-1α pathway's activation by repressing the expression of miR. By modulating the miR-30c/HIF-1α, FBI-1 promoted the Warburg effect or the epithelial-mesenchymal transition of HCC cells and promoted the resistance of HCC cells to molecular targeted agents.
Collapse
Affiliation(s)
- Juan Liu
- Department of Hematology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Chao Yang
- Department of Hematology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Xiao-Mei Huang
- Department of Hematology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Pan-Pan Lv
- Department of Hematology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Ya-Kun Yang
- Department of Hematology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Jin-Na Zhao
- Department of Hematology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Si-Yuan Zhao
- Department of Neurosurgery, Beijing Huicheng Medical Research Institute, Beijing, China
| | - Wan-Jun Sun
- Department of Hematology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| |
Collapse
|
25
|
Yan Q, Xiao Q, Ge J, Wu C, Wang Y, Yu H, Yang H, Zou J. Bioinformatics-Based Research on Key Genes and Pathways of Intervertebral Disc Degeneration. Cartilage 2021; 13:582S-591S. [PMID: 33233925 PMCID: PMC8804785 DOI: 10.1177/1947603520973247] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE To find out the pathways and key genes and to reveal disc degeneration pathogenesis based on bioinformatic analyses. DESIGN The GSE70362 dataset was downloaded from the GEO (Gene Expression Omnibus) database. Differentially expressed genes (DEGs) between the patients having disc degeneration and healthy controls were screened by Limma package in R language. Critical genes were identified by adopting gene ontologies (GOs), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, and protein-protein interaction (PPI) networks. RESULTS We identified 112 DEGs, including 60 genes which were upregulated and 52 that were downregulated. Analyses, such as GO and KEGG demonstrated that the DEGs got enriched in 4 biological processes and 2 signaling pathways, mainly related to disc degeneration. The PPI network analyses identified 5 key proteins, CCND1 (cyclin D1), GATA3, TNFSF11, LEF1, and DKK1 (Dickkopf related protein 1). CONCLUSION In this study, the DEGs and pathways determined promoted us understand the disc degeneration mechanisms. Also, the study may contribute novel biomarkers for the diagnosis and prevention of disc degeneration, and seek new treatment methods to repair and even regenerate degenerative intervertebral disc.
Collapse
Affiliation(s)
- Qi Yan
- Department of Orthopaedic Surgery, The
First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Quan Xiao
- Department of Orthopaedic Surgery, The
First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Orthopaedic Surgery, The
Affiliated Lianshui People’s Hospital of Kangda College of Nan Jing Medical
Universty, Lianshui, Jiangsu, China
| | - Jun Ge
- Department of Orthopaedic Surgery, The
First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Cenhao Wu
- Department of Orthopaedic Surgery, The
First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yingjie Wang
- Department of Orthopaedic Surgery, The
First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hao Yu
- Department of Orthopaedic Surgery, The
First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Huilin Yang
- Department of Orthopaedic Surgery, The
First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jun Zou
- Department of Orthopaedic Surgery, The
First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
26
|
Wang Y, Li T, Yang Q, Feng B, Xiang Y, Lv Z, Weng X. LncRNA THUMPD3-AS1 enhances the proliferation and inflammatory response of chondrocytes in osteoarthritis. Int Immunopharmacol 2021; 100:108138. [PMID: 34509934 DOI: 10.1016/j.intimp.2021.108138] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/29/2021] [Accepted: 09/05/2021] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Long noncoding RNAs (lncRNAs) regulate the occurrence and development of osteoarthritis (OA), whereas the biological roles and mechanisms of the lncRNA THUMPD3-AS1 (THUMPD3 antisense RNA 1) in OA remain still unclear. This study described the role and molecular mechanism of lncRNA THUMPD3-AS1 in regulating OA biology. METHOD The knee normal and OA cartilage tissues from ten participants were sequenced to reveal the differentially expressed lncRNAs. The interleukin (IL)-1β-stimulated C28/I2 cell served as OA cells. Flow cytometry assays, Western blot, enzyme-linked immunosorbent assays were used for our experiments. RESULTS The results revealed that lncRNA THUMPD3-AS1 was downregulated in OA cartilage tissues and IL-1β-stimulated chondrocyte cell line. Overexpression of lncRNA THUMPD3-AS1 alleviated cell apoptosis and facilitated inflammatory responses, whereas knockdown had opposite effects. LncRNA THUMPD3-AS1 markedly increased the cyclin E2, cyclin-dependent kinase 4, B-cell lymphoma 2, tumor necrosis factor-α, nitric oxide, and IL-6 levels, and decreased the caspase-3 level. Furthermore, the target proteins of phosphorylation were identified as nuclear factor-κB p65 and mitogen-activated protein kinase p38, which could be indirectly suppressed by lncRNA THUMPD3-AS1 knockdown. CONCLUSION Our findings highlight the different effects of lncRNA THUMPD3-AS1 on cell apoptosis and inflammatory response, which extend the multiple functions of lncRNA epigenetics in OA biology.
Collapse
Affiliation(s)
- Yingjie Wang
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China.
| | - Qi Yang
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Bin Feng
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Yongbo Xiang
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Zehui Lv
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Xisheng Weng
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
27
|
A novel chemical inhibitor suppresses breast cancer cell growth and metastasis through inhibiting HPIP oncoprotein. Cell Death Discov 2021; 7:198. [PMID: 34326318 PMCID: PMC8322322 DOI: 10.1038/s41420-021-00580-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/07/2021] [Accepted: 07/03/2021] [Indexed: 11/09/2022] Open
Abstract
Increasing evidence suggests the pivotal role of hematopoietic pre-B-cell leukemia transcription factor (PBX)-interacting protein (HPIP/PBXIP1) in cancer development and progression, indicating that HPIP inhibition may be a promising target for cancer therapy. Here, we screened compounds inhibiting breast cancer cell proliferation with HPIP fused with green fluorescent protein as a reporter. A novel agent named TXX-1-10 derived from rimonabant, an antagonist of cannabinoid receptor 1 with anticancer effects, has been discovered to reduce HPIP expression and has greater inhibitory effects on breast cancer cell growth and metastasis in vitro and in vivo than rimonabant. TXX-1-10 regulates HPIP downstream targets, including several important kinases involved in cancer development and progression (e.g., AKT, ERK1/2, and FAK) as well as cell cycle-, apoptosis-, migration-, and epithelial-to-mesenchymal transition (EMT)-related genes. Consistent with the results of anticancer effects, genome-wide RNA sequencing indicated that TXX-1-10 has more significant effects on regulation of the expression of genes related to DNA replication, cell cycle, apoptosis, cell adhesion, cell migration, and invasion than rimonabant. In addition, TXX-1-10 significantly regulated genes associated with the cell growth and extracellular matrix organization, many of which were shown to be regulated by HPIP. Moreover, compared with rimonabant, TXX-1-10 greatly reduces blood-brain barrier penetrability to avoid adverse central depressive effects. These findings suggest that HPIP inhibition may be a useful strategy for cancer treatment and TXX-1-10 is a promising candidate drug for cancer therapy.
Collapse
|
28
|
Penugurti V, Khumukcham SS, Padala C, Dwivedi A, Kamireddy KR, Mukta S, Bhopal T, Manavathi B. HPIP protooncogene differentially regulates metabolic adaptation and cell fate in breast cancer cells under glucose stress via AMPK and RNF2 dependent pathways. Cancer Lett 2021; 518:243-255. [PMID: 34302919 DOI: 10.1016/j.canlet.2021.07.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/27/2021] [Accepted: 07/19/2021] [Indexed: 12/13/2022]
Abstract
While cancer cells rewire metabolic pathways to sustain growth and survival under metabolic stress in solid tumors, the molecular mechanisms underlying these processes remain largely unknown. In this study, cancer cells switched from survival to death during the early to late phases of metabolic stress by employing a novel signaling switch from AMP activated protein kinase (AMPK)-Forkhead box O3 (FOXO3a)-hematopoietic PBX1-interacting protein (HPIP) to the ring finger protein 2 (RNF2)-HPIP-ubiquitin (Ub) pathway. Acute metabolic stress induced proto-oncogene HPIP expression in an AMPK-FOXO3a-dependent manner in breast cancer (BC) cells. HPIP depletion reduced cell survival and tumor formation in mouse xenografts, which was accompanied by diminished intracellular ATP levels and increased apoptosis in BC cells in response to metabolic (glucose) stress. Glutamine flux (13C-labeled) analysis further suggested that HPIP rewired glutamine metabolism by controlling the expression of the solute carrier family 1 member 5 (SLC1A5) and glutaminase (GLS) genes by acting as a coactivator of MYC to ensure cell survival upon glucose deprivation. However, in response to chronic glucose stress, HPIP was ubiquitinated by the E3-Ub ligase, RNF2, and was concomitantly degraded by the proteasome-mediated pathway, ensuring apoptosis. In support of these data, clinical analyses further indicated that elevated levels of HPIP correlated with AMPK activation in BC. Taken together, these data suggest that HPIP is a signal coordinator during metabolic stress and thus serves as a potential therapeutic target in BC.
Collapse
Affiliation(s)
- Vasudevarao Penugurti
- Molecular and Cellular Oncology Laboratory, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, Telangana, India
| | - Saratchandra Singh Khumukcham
- Molecular and Cellular Oncology Laboratory, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, Telangana, India
| | - Chiranjeevi Padala
- Molecular and Cellular Oncology Laboratory, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, Telangana, India
| | - Anju Dwivedi
- Molecular and Cellular Oncology Laboratory, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, Telangana, India
| | - Karthik Reddy Kamireddy
- Molecular and Cellular Biology Laboratory, Baylor College of Medicine, Houston, TX, United States
| | - Srinivasulu Mukta
- MNJ Institute of Oncology and Regional Cancer Center, Hyderabad, 500004, Telangana, India
| | - Triveni Bhopal
- MNJ Institute of Oncology and Regional Cancer Center, Hyderabad, 500004, Telangana, India
| | - Bramanandam Manavathi
- Molecular and Cellular Oncology Laboratory, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, Telangana, India.
| |
Collapse
|
29
|
Khumukcham SS, Manavathi B. Two decades of a protooncogene HPIP/PBXIP1: Uncovering the tale from germ cell to cancer. Biochim Biophys Acta Rev Cancer 2021; 1876:188576. [PMID: 34090932 DOI: 10.1016/j.bbcan.2021.188576] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/25/2021] [Accepted: 05/28/2021] [Indexed: 01/17/2023]
Abstract
Hematopoietic PBX interacting protein (HPIP or pre-B-cell leukemia transcription factor interacting protein (PBXIP1) was discovered two decades ago as a corepressor of pre-B-cell leukemia homeobox (PBX) 1 with a vital functional role in hematopoiesis. Later it emerged as a potential biomarker of poor prognosis and tumorigenesis for more than a dozen different cancers. It regulates aggressive cancer phenotypes, cell proliferation, metastasis, EMT, etc. The anomaly in the regulation of HPIP is linked with physiological disorders like renal fibrosis, chronic kidney disease and osteoarthritis. Scientists have unraveled more than twenty interacting proteins of HPIP and its functional role in various physiological and cellular processes that involves normal neuronal development, embryogenesis, endometrium decidualization, and germ cell proliferation. Over the past 20 years, we have witnessed the emerging role of HPIP and its association with a myriad of cellular activities ranging from germ cell proliferation to cancer aggressiveness, modulating multitude of signaling cascades like TGF-β1, PI3K/AKT, Wnt, mTOR, and Sonic hedgehog signaling pathways. This review will give the current understanding of HPIP, in terms of its diverse functions, theoretical ideas, and further explore cellular links and promising areas that need to be investigated. We also provide a comprehensive overview of the transcript variants of HPIP and distinct sets of transcription factors regulating their expression, which may help to understand the role of HPIP in various cellular or physiological conditions.
Collapse
Affiliation(s)
- Saratchandra Singh Khumukcham
- Molecular and Cellular Oncology Laboratory, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | - Bramanandam Manavathi
- Molecular and Cellular Oncology Laboratory, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India.
| |
Collapse
|
30
|
Zhang C, Wang Y, Sun K, Yu D, Tian S. Effects of Melatonin on Osteogenic Differentiation of Bone Marrow Mesenchymal Stem Cells in Inflammatory Environment by Regulating Mammalian Target of Rapamycin/Phosphatidylinositol 3-Kinase/Protein Kinase B Signaling. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Human bone marrow mesenchymal stem cells (BMSCs) differentiation into special cell types is affected by inflammation. Melatonin has various effects such as anti-oxidation and immune regulation. However, melatonin’s effect on BMSCs osteogenic differentiation during inflammation
has not been elucidated. Rat BMSCs were isolated and assigned into control group, inflammation group (1 μg/ml lipopolysaccharide, LPS) and melatonin group (100 μM melatonin was added to LPSstimulated BMSCs cells) followed by analysis of BMSCs proliferation by MTT assay, Caspase 3 and
ALP activity, expression of Runx2 and OP by Real time PCR, ROS content and SOD activity, TNF-α and IL-1β secretion by ELISA and mTOR/PI3K/AKT signaling protein level by Western blot. LPS action on BMSCs significantly inhibits BMSCs proliferation, promotes Caspase 3 activity, inhibits
ALP activity, decreases Runx2 and OP expression and SOD activity, increases ROS content and TNF-α and IL-1β secretion as well as reduced mTOR and p-PI3K level (P <0.05). Melatonin addition significantly reversed the above changes (P <0.05). Melatonin can regulate oxidative
stress, inhibit inflammation, and promote BMSCs proliferation and osteogenic differentiation in inflammatory environment by activating mTOR/PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266555, China
| | - Yuanhe Wang
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266555, China
| | - Kang Sun
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266555, China
| | - Dingzhu Yu
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266555, China
| | - Shaoqi Tian
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266555, China
| |
Collapse
|
31
|
Candidates for Intra-Articular Administration Therapeutics and Therapies of Osteoarthritis. Int J Mol Sci 2021; 22:ijms22073594. [PMID: 33808364 PMCID: PMC8036705 DOI: 10.3390/ijms22073594] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/26/2021] [Accepted: 03/26/2021] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis (OA) of the knee is a disease that significantly decreases the quality of life due to joint deformation and pain caused by degeneration of articular cartilage. Since the degeneration of cartilage is irreversible, intervention from an early stage and control throughout life is important for OA treatment. For the treatment of early OA, the development of a disease-modifying osteoarthritis drug (DMOAD) for intra-articular (IA) injection, which is attracting attention as a point-of-care therapy, is desired. In recent years, the molecular mechanisms involved in OA progression have been clarified while new types of drug development methods based on gene sequences have been established. In addition to conventional chemical compounds and protein therapeutics, the development of DMOAD from the new modalities such as gene therapy and oligonucleotide therapeutics is accelerating. In this review, we have summarized the current status and challenges of DMOAD for IA injection, especially for protein therapeutics, gene therapy, and oligonucleotide therapeutics.
Collapse
|
32
|
Lin Z, Miao J, Zhang T, He M, Zhou X, Zhang H, Gao Y, Bai L. d-Mannose suppresses osteoarthritis development in vivo and delays IL-1β-induced degeneration in vitro by enhancing autophagy activated via the AMPK pathway. Biomed Pharmacother 2021; 135:111199. [PMID: 33401221 DOI: 10.1016/j.biopha.2020.111199] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/13/2020] [Accepted: 12/26/2020] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis (OA) is a heterogeneous disease that is consistently difficult to treat due to the complexity of the regulatory network involved in OA pathogenesis, especially in terms of cartilage degeneration. As a C-2 epimer of glucose, d-mannose can alleviate bone loss and repress immunopathology by upregulating regulatory T cells; however, the role of d-mannose in OA-related cartilage degeneration remains unknown. In this study, we investigated the chondroprotective effect of d-mannose in vitro and in vivo on OA. We found that incubating interleukin (IL)-1β-treated rat chondrocytes with d-mannose restrained OA degeneration by elevating cell proliferation, strongly activating autophagy, reducing apoptosis, and downregulating catabolism. Additionally, oral gavage administration of d-mannose to monosodium iodoacetate (MIA)-treated rats revealed that a median (1.25 g/kg/day) rather than high or low dose of d-mannose suppressed OA progression and attenuated OA development based on lower macroscopic scores for cartilage, decreased histological scores for cartilage and synovium, strongly activated autophagy, and downregulated catabolism. In terms of a downstream mechanism, we showed that d-mannose might attenuate OA degeneration by activating autophagy in IL-1β-treated rat chondrocytes by promoting the phosphorylation of 5' AMP-activated protein kinase (AMPK). Our in vitro findings revealed that d-mannose delayed IL-1β-induced OA degeneration in rat chondrocytes by enhancing autophagy activation through the AMPK pathway. Furthermore, the in vivo results indicated that a median dose of d-mannose suppressed MIA-induced OA development. These results suggested that d-mannose exhibits chondroprotective effects and represents a potential disease-modifying drug and novel therapeutic agent for OA.
Collapse
Affiliation(s)
- Zhiming Lin
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110000, China
| | - Jianing Miao
- Medical Research Center/Liaoning Key Laboratory of Research and Application of Animal Models for Environmental and Metabolic Diseases, Shenyang, 110000, China
| | - Tao Zhang
- Department of Thoracic Surgery, Xiamen Branch of Zhongshan Hospital of Fudan University, Xiamen, 361000, China
| | - Ming He
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110000, China
| | - Xiaonan Zhou
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110000, China
| | - He Zhang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110000, China
| | - Yue Gao
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110000, China
| | - Lunhao Bai
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110000, China.
| |
Collapse
|
33
|
Zhang Y, Weng Q, Chen J, Li M, Han J. Oroxylin A attenuates IL-1β-induced inflammatory reaction via inhibiting the activation of the ERK and PI3K/AKT signaling pathways in osteoarthritis chondrocytes. Exp Ther Med 2021; 21:388. [PMID: 33680110 PMCID: PMC7918508 DOI: 10.3892/etm.2021.9819] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 10/27/2020] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA) is characterized by degradation of the articular cartilage, synovium inflammation, subchondral bone sclerosis and osteophyte formation. OA is the most common degenerative joint disorder among the elderly population. In particular, currently available therapeutic strategies, such as non-steroidal anti-inflammatory drugs (NSAIDs) may cause severe side-effects. Therefore, novel candidate targets for OA therapy are urgently needed. Oroxylin A (OrA) is a natural mono-flavonoid that can be extracted from Scutellariae radix. The present study aimed to investigate the potential effects of OrA on interleukin (IL)-1β-induced chondrocytes inflammatory reactions. The current study performed quantitative PCR, western blotting and cell immunofluorescence to evaluate the effect of Oroxylin A in chondrocyte inflammation. The results demonstrated that OrA significantly attenuated the upregulation of inducible nitric oxide synthase and cyclooxygenase 2 by IL-1β at both protein and mRNA levels. IL-1β-stimulated upregulation of matrix metalloproteinase (MMP)-3 and MMP-13 expression, in addition to disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)-4 and ADAMTS-5 expression, were all inhibited by OrA. Treatment with OrA significantly reversed the degradation of type II collagen and aggrecan by IL-1β. Mechanistically, OrA suppressed the IL-1β induced activation of ERK1/2 and PI3K/AKT signaling pathways. In conclusion, these findings suggest that OrA can serve as a potential therapeutic agent for the treatment of OA.
Collapse
Affiliation(s)
- Yong Zhang
- Department of Trauma Orthopedics, Ningbo No. 6 Hospital, Ningbo, Zhejiang 315000, P.R. China
| | - Qiuyan Weng
- Department of Neurology, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, Zhejiang 315000, P.R. China
| | - Jianming Chen
- Department of Trauma Orthopedics, Ningbo No. 6 Hospital, Ningbo, Zhejiang 315000, P.R. China
| | - Ming Li
- Department of Trauma Orthopedics, Ningbo No. 6 Hospital, Ningbo, Zhejiang 315000, P.R. China
| | - Jinming Han
- Department of Spine, Ningbo No. 6 Hospital, Ningbo, Zhejiang 315000, P.R. China
| |
Collapse
|
34
|
Meng F, Shuai J, Li G, Weng J, Zeng H. Effect of Down-Regulation of Long-Chain Non-Coding RNAs Myocardial Infarction Associated Transcript 2 Expression on Osteoarthritis Chondrocytes. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Osteoarthritis (OA) is featured as articular cartilage degradation. LncRNA Mirt2 involves in inflammation, but its role in osteoarthritis is unclear. Our study intends to assess LncRNA Mirt2’s role in OA chondrocytes. The chondrocytes of OA patients (OA group) and healthy controls
(control group) were isolated to measure LncRNA Mirt2 expression by Real time PCR. Chondrocytes were assigned into control group, LPS group, LPS + si-NC group, LPS + Mirt2 siRNA group followed by analysis of LncRNA Mirt2 level by real time PCR, cell proliferation by MTT assay, cell apoptosis
by flow cytometry, expression of COL2A1, MMP13, ADAMTS-5, MEK1/2, Erk1/2 and phosphorylated Erk1/2 by western blot. LncRNA Mirt2 level was increased in OA chondrocytes. Under LPS stim-ulation, Mirt2 expression was significantly increased in chondrocytes and chondrocyte proliferation was decreased,
along with significantly increased apoptosis and upregulated COL2A1, MMP13, ADAMTS-5, MEK1/2 and Erk1/2 and phosphorylated Erk1/2 (P < 0.05). Transfection of Mirt2 siRNA down-regulated its expression in chondrocytes stimulated by LPS, which significantly reversed the above changes
(P < 0.05). LncRNA Mirt2 expression is increased in OA chondrocytes. Downregulation of LncRNA Mirt2 can regulate COL2A1, MMP13 and ADAMTS-5 level via MAPK/ERK signaling pathway, promote OA chondrocytes proliferation and inhibit apoptosis.
Collapse
Affiliation(s)
- Fanbin Meng
- Hand and Microsurgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Jun Shuai
- Department of Dermatology, Shenzhen Futian Center for Chronic Disease Control, Shenzhen, Guangdong, 518046, China
| | - Guoqing Li
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Jian Weng
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Hui Zeng
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| |
Collapse
|
35
|
Chen L, Hu J, Zhou B, Li Y, Wei K, Wang J, Lv H, Zeng F. Effect of Integrin-Linked Kinase on Osteogenesis of Bone Marrow Mesenchymal Stem Cells in Inflammatory Environment via Regulating Mitogen Activated Protein Kinase/Protein Kinase B Signaling Pathway. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Osteoarthritis (OA) pathogenesis involves inflammation, age, weight and other factors. Integrin-linked kinase (ILK) regulates cell apoptosis, metastasis, and growth. However, whether ILK affects bone formation of bone marrow mesenchymal stem cells in an inflammatory environment has
not been elucidated. Rat BMSCs were isolated and assigned into control group, inflammation group (lipopolysaccharide was added to cells); and si-ILK group (ILK siRNA was transfected into the inflammation group BMSCs) followed by analysis of cell proliferation by MTT assay, expression of ILK,
Runx2 and OP by real time PCR, ALp activity, TNF-α and IL-6 secretion by ELISA and MAPK/AKT signaling protein expression by western blot. Compared to control, ILK in BMSCs cells in inflammatory environment was significantly upregulated, resulting in inhibition of cell proliferation,
decreased ALP activity, reduced expression of osteogenic genes Runx2 and OP, increased secretion of TNF-α and IL-6, and downregulated p-AKT (P < 0.05); transfection of ILK siRNA down-regulated ILK in inflammatory environment BMSCs, which significantly increased BMSCs
cell proliferation, increased ALP activity and expression of Runx2 and OP, decreased TNF-α and IL-6 secretion and increased p-AKT expression (P < 0.05). ILK expression is increased in BMSCs in an inflammatory environment. Down-regulation of ILK in BMSCs cells in an inflammatory
environment can regulate MAPK/AKT signaling, inhibit inflammatory factors secretion, thereby promoting BMSCs proliferation and osteogenesis differentiation.
Collapse
Affiliation(s)
- Liyuan Chen
- Second Department of Orthopedics, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine (First People’s Hospital of Baiyin City), Baiyin, Gansu, 730900, China
| | - Jieliang Hu
- Second Department of Orthopedics, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine (First People’s Hospital of Baiyin City), Baiyin, Gansu, 730900, China
| | - Baojun Zhou
- Second Department of Orthopedics, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine (First People’s Hospital of Baiyin City), Baiyin, Gansu, 730900, China
| | - Yan Li
- Department of Electrophysiology, People’s Hospital of Xingguo County, Ganzhou, Jiangxi, 342400, China
| | - Kongxing Wei
- Second Department of Orthopedics, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine (First People’s Hospital of Baiyin City), Baiyin, Gansu, 730900, China
| | - Jinglei Wang
- Second Department of Orthopedics, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine (First People’s Hospital of Baiyin City), Baiyin, Gansu, 730900, China
| | - Hongyan Lv
- Second Department of Orthopedics, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine (First People’s Hospital of Baiyin City), Baiyin, Gansu, 730900, China
| | - Fanyun Zeng
- Emergency Traumatic Surgery, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou People’s Hospital), Ganzhou, Jiangxi, 341000, China
| |
Collapse
|
36
|
Lin Z, Miao J, Zhang T, He M, Wang Z, Feng X, Bai L. JUNB-FBXO21-ERK axis promotes cartilage degeneration in osteoarthritis by inhibiting autophagy. Aging Cell 2021; 20:e13306. [PMID: 33450132 PMCID: PMC7884044 DOI: 10.1111/acel.13306] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 12/05/2020] [Accepted: 12/23/2020] [Indexed: 01/22/2023] Open
Abstract
Osteoarthritis (OA) is a heterogeneous disease that is extremely hard to cure owing to its complex regulation network of pathogenesis, especially cartilage degeneration. FBXO21 is a subunit of ubiquitin E3 ligases that degrades P-glycoprotein and EID1 by ubiquitination and activates the JNK and p38 pathways; however, its role in OA remains unknown. Here, the main objective of this study was to evaluate the potential effects and mechanism of FBXO21 in OA degeneration, we revealed that FBXO21 is upregulated in the cartilage of patients with OA, aging, and monosodium iodoacetate-induced OA rats, and chondrocytes treated with interleukin-1β, tumor necrosis factor-α, and lipopolysaccharide. Moreover, the in vivo and in vitro knockdown of FBXO21 suppressed OA-related cartilage degeneration, as evidenced by activated autophagy, upregulated anabolism, alleviated apoptosis, and downregulated catabolism. In contrast, its overexpression promoted OA-related cartilage degeneration. In addition, using mass spectrometry and co-immunoprecipitation assay, we demonstrated that the downstream mechanism of FBXO21 inhibits autophagy by interacting with and phosphorylating ERK. Furthermore, FBXO21 alleviated anabolism and enhanced apoptosis and catabolism by inhibiting autophagy in rat chondrocytes. Interestingly, for its upstream mechanism, JUNB promoted FBXO21 expression by directly targeting the FBXO21 promoter, thus further accelerating cartilage degeneration in SW1353 cells and rat chondrocytes. Overall, our findings reveal that the JUNB-FBXO21-ERK axis regulates OA apoptosis and cartilage matrix metabolism by inhibiting autophagy. Therefore, FBXO21 is an attractive target for regulating OA pathogenesis, and its knockdown may provide a novel targeted therapy for OA.
Collapse
Affiliation(s)
- Zhiming Lin
- Department of Orthopedics Shengjing Hospital of China Medical University Shenyang China
| | - Jianing Miao
- Medical Research Center/Liaoning Key Laboratory of Research and Application of Animal Models for Environmental and Metabolic Diseases Shenyang China
| | - Tao Zhang
- Department of Thoracic Surgery Xiamen Branch of Zhongshan Hospital of Fudan University Xiamen China
| | - Ming He
- Department of Orthopedics Shengjing Hospital of China Medical University Shenyang China
| | - Ziyuan Wang
- Department of Orthopedics Shengjing Hospital of China Medical University Shenyang China
| | - Xinyuan Feng
- Department of Orthopedics Shengjing Hospital of China Medical University Shenyang China
| | - Lunhao Bai
- Department of Orthopedics Shengjing Hospital of China Medical University Shenyang China
| |
Collapse
|
37
|
Cai X, Wei B, Li L, Chen X, Yang J, Li X, Jiang X, Lv M, Li M, Lin Y, Xu Q, Guo W, Gu Y. Therapeutic Potential of Apatinib Against Colorectal Cancer by Inhibiting VEGFR2-Mediated Angiogenesis and β-Catenin Signaling. Onco Targets Ther 2020; 13:11031-11044. [PMID: 33154652 PMCID: PMC7606303 DOI: 10.2147/ott.s266549] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 10/05/2020] [Indexed: 01/05/2023] Open
Abstract
Purpose Apatinib is an inhibitor of VEGFR2 (vascular endothelial growth factor receptor 2) that has attracted a great deal of attention due to its promotion of anticancer activity. In the present study, we investigated the therapeutic effects of apatinib against colorectal cancer (CRC) and examined the underlying mechanism. Materials and Methods Both in vivo and in vitro assays were conducted to study the effect of apatinib on CRC. To elucidate the associated mechanism, RNA-seq (transcriptome) analysis was conducted on apatinib-treated HCT116 cells. Results Apatinib showed antiproliferative and proapoptotic effects, induced G0/G1 arrest and blocked cell migration and invasion in CRC. An analysis of the mechanism associated with apatinib activity demonstrated that by interacting with VEGFR2, apatinib decreased p-Src, p-Akt, and p-GSK3β levels, which further increased β-catenin ubiquitination and reduced the nuclear translocation of β-catenin. Furthermore, apatinib strongly suppressed CT26 cell growth in mouse xenograft models by inhibiting β-catenin signaling and angiogenesis. Conclusion Overall, the results of the present study here indicated that by inhibiting the VEGFR2-β-catenin-mediated malignant phenotype, apatinib significantly suppresses the growth of CRC, suggesting that the use of apatinib is a promising therapeutic strategy for CRC.
Collapse
Affiliation(s)
- Xiaomin Cai
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Bin Wei
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China.,Department of Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an 223300, People's Republic of China
| | - Lele Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Xiaofeng Chen
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Jing Yang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Xiaofei Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Xiaozheng Jiang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Mu Lv
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Mingyang Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Yumeng Lin
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, People's Republic of China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, People's Republic of China
| | - Wenjie Guo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, People's Republic of China
| | - Yanhong Gu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
38
|
Therapeutic Manipulation of Macrophages Using Nanotechnological Approaches for the Treatment of Osteoarthritis. NANOMATERIALS 2020; 10:nano10081562. [PMID: 32784839 PMCID: PMC7466380 DOI: 10.3390/nano10081562] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/31/2020] [Accepted: 08/07/2020] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) is the most common joint pathology causing severe pain and disability. Macrophages play a central role in the pathogenesis of OA. In the joint microenvironment, macrophages with an M1-like pro-inflammatory phenotype induce chronic inflammation and joint destruction, and they have been correlated with the development and progression of the disease, while the M2-like anti-inflammatory macrophages support the recovery of the disease, promoting tissue repair and the resolution of inflammation. Nowadays, the treatment of OA in the clinic relies on systemic and/or intra-articular administration of anti-inflammatory and pain relief drugs, as well as surgical interventions for the severe cases (i.e., meniscectomy). The disadvantages of the pharmacological therapy are related to the chronic nature of the disease, requiring prolonged treatments, and to the particular location of the pathology in joint tissues, which are separated anatomical compartments with difficult access for the drugs. To overcome these challenges, nanotechnological approaches have been investigated to improve the delivery of drugs toward macrophages into the diseased joint. This strategy may offer advantages by reducing off-target toxicities and improving long-term therapeutic efficacy. In this review, we describe the nanomaterial-based approaches designed so far to directly or indirectly manipulate macrophages for the treatment of osteoarthritis.
Collapse
|
39
|
Chen S, Luo Z, Chen X. Andrographolide mitigates cartilage damage via miR-27-3p-modulated matrix metalloproteinase13 repression. J Gene Med 2020; 22:e3187. [PMID: 32196852 DOI: 10.1002/jgm.3187] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/09/2020] [Accepted: 03/12/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND As a potential anti-arthritic agent, Andrographolide (And) is capable of promoting chondrocyte proliferation and preventing apoptosis in pathologic condition. The present study aimed to explore the roles of And in in vivo and in vitro models of osteoarthritis (OA), as well as its underlying molecular mechanisms. METHODS An OA mouse model was established using anterior cruciate ligament transection operation on the left knee joint. The pathological changes of articular cartilage were assessed using safranin O staining. Chondrocyte proliferation and apoptosis were measured using cell a counting kit-8 assay and flow cytometry. Bioinformatics algorithms and a luciferase reporter assay were used to evaluate matrix metalloproteinase13 (MMP13) as a direct target of miR-27-3p. RESULTS And had the ability to prevent catabolism and facilitate anabolism of articular cartilage in an experimental OA model in mice. In addition, And alleviated chondrocyte apoptosis in in vitro and in vivo models of OA. We also found that both up-regulation of MMP13 and down-regulation of miR-27-3p in the proximal tibia of OA mice and interleukin (IL)-1β-stimulated chondrocytes were reversed by And administration simultaneously. MMP13 was validated as direct target of miR-27-3p and could be suppressed by overexpression of miR-27-3p in mouse chondrocyte. Furthermore, overexpression of miR-27-3p or MMP13 loss-of-function in chondrocytes could alleviate IL-1β-induced apoptosis. CONCLUSIONS These results indicated that miR-27-3p/MMP13 signaling axis might be a potential therapeutic target of And for preventing the progression of OA.
Collapse
Affiliation(s)
- Shaojian Chen
- Department of Sports Medical, Ganzhou People's Hospital & the Affiliated Ganzhou Hospital of Nanchang University, Jiangxi Province, China
| | - Zhihuan Luo
- Department of Sports Medical, Ganzhou People's Hospital & the Affiliated Ganzhou Hospital of Nanchang University, Jiangxi Province, China
| | - Xiaguang Chen
- Department of Sports Medical, Ganzhou People's Hospital & the Affiliated Ganzhou Hospital of Nanchang University, Jiangxi Province, China
| |
Collapse
|
40
|
Fan Z, Liu Y, Shi Z, Deng K, Zhang H, Li Q, Cao S, Li S, Zhang H. MiR-155 promotes interleukin-1β-induced chondrocyte apoptosis and catabolic activity by targeting PIK3R1-mediated PI3K/Akt pathway. J Cell Mol Med 2020; 24:8441-8451. [PMID: 32562373 PMCID: PMC7412691 DOI: 10.1111/jcmm.15388] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/01/2020] [Accepted: 04/27/2020] [Indexed: 12/23/2022] Open
Abstract
Osteoarthritis (OA) is a common joint disease characterized by progressive cartilage degradation, in which elevated chondrocyte apoptosis and catabolic activity play an important role. MicroRNA-155 (miR-155) has recently been shown to regulate apoptosis and catabolic activity in some pathological circumstances, yet, whether and how miR-155 is associated with OA pathology remain unexplored. We report here that miR-155 level is significantly up-regulated in human OA cartilage biopsies and also in primary chondrocytes stimulated by interleukin-1β (IL-1β), a pivotal pro-catabolic factor promoting cartilage degradation. Moreover, miR-155 inhibition attenuates and its overexpression promotes IL-1β-induced apoptosis and catabolic activity in chondrocytes in vitro. We also demonstrate that the PIK3R1 (p85α regulatory subunit of phosphoinositide 3-kinase (PI3K)) is a target of miR-155 in chondrocytes, and more importantly, PIK3R1 restoration abrogates miR-155 effects on chondrocyte apoptosis and catabolic activity. Mechanistically, PIK3R1 positively regulates the transduction of PI3K/Akt pathway, and a specific Akt inhibitor reverses miR-155 effects on promoting chondrocyte apoptosis and catabolic activity, phenocopying the results obtained via PIK3R1 knockdown, hence establishing that miR-155 promotes chondrocyte apoptosis and catabolic activity through targeting PIK3R1-mediated PI3K/Akt pathway activation. Altogether, our study discovers novel roles and mechanisms of miR-155 in regulating chondrocyte apoptosis and catabolic activity, providing an implication for therapeutically intervening cartilage degradation and OA progression.
Collapse
Affiliation(s)
- Zhiyong Fan
- Department of Orthopaedic SurgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Yinghui Liu
- Department of Infectious DiseaseThe Third Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Zhengliang Shi
- Department of Orthopaedic SurgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Kai Deng
- Department of Orthopaedic SurgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Hua Zhang
- Department of Orthopaedic SurgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Qiutong Li
- Department of Orthopaedic SurgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Shuxing Cao
- Department of Orthopaedic SurgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Shentai Li
- Department of Orthopaedic SurgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Hongliang Zhang
- Department of Orthopaedic SurgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| |
Collapse
|
41
|
Lin J, Chen L, Dou D. Progress of orthopaedic research in China over the last decade. J Orthop Translat 2020; 24:131-137. [PMID: 32913711 PMCID: PMC7452214 DOI: 10.1016/j.jot.2020.04.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/26/2020] [Accepted: 04/28/2020] [Indexed: 02/07/2023] Open
Abstract
Objective To summarize the representative scientific achievements in the past decade, and discuss the future challenges and directions for orthopaedic research in China. Methods In this review, we used the data provided by National Natural Science Foundation of China (NSFC) for analysis. Results Over the last decade, NSFC has initiated various research programs with a total funding of over 1149 million RMB to support orthopaedic exploration. Under the strong support of NSFC, great progresses have been made in basic research, talent training, platform construction and the clinical translation in the field of orthopaedics in China. Conclusion In general, since the establishment of the Department of Health Sciences of NSFC 10 years ago, both the amount of funding and the scale of researchers in the field of orthopaedic research have increased substantially. Despite of several shortcomings in orthopaedic research, with continuous support from NSFC both in funding and in policy, we believe that the orthopaedic research in China will surely make steady and significant progress. The translational potential of this article This article summarizes the representative scientific achievements in the past decade and puts forward the future challenges and directions for orthopaedic research in China.
Collapse
Affiliation(s)
- Jun Lin
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lin Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Dou Dou
- Department of Health Sciences, National Natural Science Foundation of China, Beijing, China
| |
Collapse
|
42
|
Wang JN, Yang Q, Yang C, Cai YT, Xing T, Gao L, Wang F, Chen X, Liu XQ, He XY, Wei B, Jiang L, Li C, Jin J, Wen JG, Ma TT, Chen HY, Li J, Meng XM. Smad3 promotes AKI sensitivity in diabetic mice via interaction with p53 and induction of NOX4-dependent ROS production. Redox Biol 2020; 32:101479. [PMID: 32143149 PMCID: PMC7058410 DOI: 10.1016/j.redox.2020.101479] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/03/2020] [Accepted: 02/23/2020] [Indexed: 12/13/2022] Open
Abstract
The incidence and severity of acute kidney injury (AKI) is increased yearly in diabetic patients. Although the mechanisms for this remain unclear, the prevention of AKI in diabetic nephropathy is feasible and of value. As we detected highly activation of TGF-β/Smad3 signaling in both human biopsy and mouse model of diabetic nephropathy, we hypothesized that Smad3 activation in diabetic kidneys may increase AKI sensitivity. We tested our hypothesis in vitro using TGF-β type II receptor (TGF-βRII) disrupted tubular epithelial cells (TECs) and in vivo in mice with streptozotocin (STZ)-induced diabetic nephropathy before the induction of ischemia/reperfusion (I/R) injury. We found that high glucose (HG)-cultured TECs showed increased inflammation, apoptosis and oxidative stress following hypoxia/reoxygenation (H/R) injury. Disruption of TGF-βRII attenuated cell injury induced by H/R in HG-treated TECs. Consistently, Smad3 knockdown in diabetic kidney attenuated I/R-induced AKI. Mechanistically, Smad3 binds to p53 and enhances p53 activity in cells treated with HG and H/R, which may lead to TECs apoptosis. Additionally, ChIP assay showed that Smad3 bound with the promoter region of NOX4 and induced ROS production and inflammation. In conclusion, our results demonstrate that Smad3 promotes AKI susceptibility in diabetic mice by interacting with p53 and NOX4. Smad3 activation in diabetic kidneys may increase AKI sensitivity. Blockade of Smad3 in diabetic kidney may both prevent AKI and CKD progression. Smad3 interacts with p53 to enhance TECs apoptosis. Smad3 binds with promoter region of NOX4 to induce ROS production and inflammation.
Collapse
Affiliation(s)
- Jia-Nan Wang
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Qin Yang
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Chen Yang
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China
| | - Yu-Ting Cai
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China; Department of Nephrology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Tian Xing
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| | - Li Gao
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Fang Wang
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Xin Chen
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Xue-Qi Liu
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China; Department of Nephrology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Xiao-Yan He
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Biao Wei
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Ling Jiang
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China; Department of Nephrology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Chao Li
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Juan Jin
- Department of Pharmacology, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Jia-Gen Wen
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Tao-Tao Ma
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Hai-Yong Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
| | - Jun Li
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Xiao-Ming Meng
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China.
| |
Collapse
|
43
|
Effects of rAAV-Mediated sox9 Overexpression on the Biological Activities of Human Osteoarthritic Articular Chondrocytes in Their Intrinsic Three-Dimensional Environment. J Clin Med 2019; 8:jcm8101637. [PMID: 31591319 PMCID: PMC6832991 DOI: 10.3390/jcm8101637] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/01/2019] [Accepted: 10/02/2019] [Indexed: 12/11/2022] Open
Abstract
Gene therapy for osteoarthritis offers powerful, long-lasting tools that are well adapted to treat such a slow, progressive disorder, especially those therapies based on the clinically adapted recombinant adeno-associated viral (rAAV) vectors. Here, we examined the ability of an rAAV construct carrying a therapeutic sequence for the cartilage-specific SOX9 transcription factor to modulate the phenotype of human osteoarthritic articular chondrocytes compared with normal chondrocytes in a three-dimensional environment where the cells are embedded in their extracellular matrix. Successful sox9 overexpression via rAAV was noted for at least 21 days, leading to the significant production of major matrix components (proteoglycans, type-II collagen) without affecting the proliferation of the cells, while the cells contained premature hypertrophic processes relative to control conditions (reporter rAAV-lacZ application, absence of vector treatment). These findings show the value of using rAAV to adjust the osteoarthritic phenotype when the chondrocytes are confined in their inherently altered environment and the possibility of impacting key cellular processes via gene therapy to remodel human osteoarthritic cartilage lesions.
Collapse
|
44
|
Huo Y, Wang Q, Liu Y, Wang J, Li Q, Li Z, Dong Y, Huang Y, Wang L. A temperature-sensitive phase-change hydrogel of topotecan achieves a long-term sustained antitumor effect on retinoblastoma cells. Onco Targets Ther 2019; 12:6069-6082. [PMID: 31534347 PMCID: PMC6681573 DOI: 10.2147/ott.s214024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/08/2019] [Indexed: 12/24/2022] Open
Abstract
Background Retinoblastoma (Rb) is one of the most common malignancies among children. Following early diagnosis and prompt treatment, the clinical outcome or prognosis of Rb is promising. However, the prognosis or survival rates of patients with late-stage Rb remain poor. Current therapeutic strategies for advanced Rb mainly involve the use of advanced chemotherapeutic options. However, the efficacy of these strategies is not satisfactory. Therefore, the development of novel strategies to achieve a more effective antitumor effect on late-stage Rb is of crucial importance. Methods and materials Topotecan was dissolved in phosphate-buffered saline and prepared into a temperature-sensitive phase-change hydrogel (termed Topo-Gel). Moreover, Topo-Gel was injected into tumor tissues formed by Y79 cells (an Rb cell line) in nude mice to examine the long-term release and long-acting antitumor effect of Topo-Gel on Rb tumors. Results Topo-Gel transforms from liquid to a hydrogel at near body temperatures (phase-change temperature [T1/2] was 37.23±0.473 °C), and maintains the slow release of topotecan in Rb tumor tissues. Following the subcutaneous injection of Topo-Gel, the treatment induced long-acting inhibition of tumor growth and relieved the adverse effects associated with topotecan. Topo-Gel, a temperature-sensitive phase-change hydrogel, is a slow-release system that prolongs the presence of topotecan in Rb tissues, and preserves the efficacy of topotecan in the long term. Conclusion Preparation of topotecan into a temperature-sensitive phase-change hydrogel achieves a long-term sustained antitumor effect on Rb cells, and may be a useful strategy for the treatment of intraocular Rb.
Collapse
Affiliation(s)
- Yan Huo
- Department of Ophthalmology, The 1st Medical Center Of Chinese PLA General Hospital , Beijing 100853, People's Republic of China.,The Institutional Animal Care and Use Committee of National Beijing Center for Drug Safety Assessment, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing 100850, People's Republic of China
| | - Qun Wang
- Department of Ophthalmology, The 1st Medical Center Of Chinese PLA General Hospital , Beijing 100853, People's Republic of China
| | - Ying Liu
- Department of Ophthalmology, The 1st Medical Center Of Chinese PLA General Hospital , Beijing 100853, People's Republic of China
| | - Junyi Wang
- Department of Ophthalmology, The 1st Medical Center Of Chinese PLA General Hospital , Beijing 100853, People's Republic of China
| | - Qian Li
- Department of Ophthalmology, The 1st Medical Center Of Chinese PLA General Hospital , Beijing 100853, People's Republic of China
| | - Zongyuan Li
- Department of Ophthalmology, The 1st Medical Center Of Chinese PLA General Hospital , Beijing 100853, People's Republic of China
| | - Yan Dong
- Department of Ophthalmology, The 1st Medical Center Of Chinese PLA General Hospital , Beijing 100853, People's Republic of China
| | - Yifei Huang
- Department of Ophthalmology, The 1st Medical Center Of Chinese PLA General Hospital , Beijing 100853, People's Republic of China
| | - Liqiang Wang
- Department of Ophthalmology, The 1st Medical Center Of Chinese PLA General Hospital , Beijing 100853, People's Republic of China
| |
Collapse
|