1
|
Kannan G, Paul BM, Thangaraj P. Stimulation, regulation, and inflammaging interventions of natural compounds on nuclear factor kappa B (NF-kB) pathway: a comprehensive review. Inflammopharmacology 2025; 33:145-162. [PMID: 39776026 DOI: 10.1007/s10787-024-01635-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025]
Abstract
Nuclear factor kappa B (NF-kB) is a kind of transcription factor which resides in cytoplasm of each cell and on activation, it translocates to the nucleus. It is activated by a many inducible agents including endotoxins, inflammatory stimuli, carcinogens, pathogens, nicotine, and tumour promoters, etc. NF-kB is activated by canonical and non-canonical signalling pathways which has different signalling compounds and its biological functions. It controls the expression of 400 different genes including various enzymes, cytokines, viral proteins, regulatory molecules involved in the cell cycle etc. This pathway is linked with various ailments including respiratory diseases, inflammatory diseases, auto immune diseases, cancer and diabetes. NF-kB factor and signalling pathway are the mainstream of the innate and adaptive immune responses. Human subjects have been able to curb inflammation through inflammaging with the help of the phytomolecules interacting with the NF-κB pathway by adjusting the inflammation processes and alleviating aging stresses in cells. They successfully inhibit the activation of NF-κB, thereby curtailing chronic low-grade inflammation underlying both ageing and age-related disease processes. These phytocompounds discussed herewith not only down-regulate NF-κB-dependent pro-inflammatory pathways but also help build resilience at cellular levels, therefore, offering enhanced healthspan with late commencement of inflammaging pathogenesis. This review describes what stimulation and regulation of the Nuclear Factor kappa B (NF-kB) Pathway and its roles in the pathogenesis of human age related diseases. We also review the recent progress in attenuating the molecular mechanisms of the NF-kB Pathway by phytochemicals, which may open up novel therapeutic avenues.
Collapse
Affiliation(s)
- Gowtham Kannan
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | - Benedict Mathews Paul
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | - Parimelazhagan Thangaraj
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India.
| |
Collapse
|
2
|
Ma RX. A detective story of intermittent fasting effect on immunity. Immunology 2024; 173:227-247. [PMID: 38922825 DOI: 10.1111/imm.13829] [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: 02/03/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Intermittent fasting (IF) refers to periodic fasting routines, that caloric intake is minimized not by meal portion size reduction but by intermittently eliminating ingestion of one or several consecutive meals. IF can instigate comprehensive and multifaceted alterations in energy metabolism, these metabolic channels may aboundingly function as primordial mechanisms that interface with the immune system, instigating intricate immune transformations. This review delivers a comprehensive understanding of IF, paying particular attention to its influence on the immune system, thus seeking to bridge these two research domains. We explore how IF effects lipid metabolism, hormonal levels, circadian rhythm, autophagy, oxidative stress, gut microbiota, and intestinal barrier integrity, and conjecture about the mechanisms orchestrating the intersect between these factors and the immune system. Moreover, the review includes research findings on the implications of IF on the immune system and patients burdened with autoimmune diseases.
Collapse
Affiliation(s)
- Ru-Xue Ma
- School of Medical, Qinghai University, Xining, China
| |
Collapse
|
3
|
Alkandahri MY, Sadino A, Pamungkas BT, Oktoba Z, Arfania M, Yuniarsih N, Wahyuningsih ES, Dewi Y, Winarti SA, Dinita ST. Potential Nephroprotective Effect of Kaempferol: Biosynthesis, Mechanisms of Action, and Clinical Prospects. Adv Pharmacol Pharm Sci 2024; 2024:8907717. [PMID: 39377015 PMCID: PMC11458287 DOI: 10.1155/2024/8907717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 08/16/2024] [Accepted: 09/10/2024] [Indexed: 10/09/2024] Open
Abstract
Kidney is an essential organ that is highly susceptible to cellular injury caused by various toxic substances in the blood. Several studies have shown that untreated injuries to this organ can cause glomerulosclerosis, tubulointerstitial fibrosis, and tubular cell apoptosis, leading to kidney failure. Despite significant advancements in modern treatment, there is no fully effective drug for repairing its function, providing complete protection, and assisting in cell regeneration. Furthermore, some available medications have been reported to exacerbate injuries, showing the need to explore alternative treatments. Natural drugs are currently being explored as a new therapeutic strategy for managing kidney diseases. Kaempferol, a polyphenol found in plants, including vegetables, legumes, and fruits, has been extensively studied in various nephrotoxicity protocols. The compound has been reported to have potential as a nephroprotective agent with beneficial effects on various physiological pathways, such as CPL-induced kidney injury, DOX, LPO, ROS, RCC, and diabetic nephropathy. Therefore, this study aims to provide a brief overview of the current nephroprotective effects of kaempferol, as well as its molecular mechanisms of action, biosynthesis pathways, and clinical prospects.
Collapse
Affiliation(s)
- Maulana Yusuf Alkandahri
- Department of Pharmacology and Clinical PharmacyFaculty of PharmacyUniversitas Buana Perjuangan Karawang, Karawang, West Java, Indonesia
| | - Asman Sadino
- Department of PharmacyFaculty of Mathematics and Natural ScienceUniversitas Garut, Garut, West Java, Indonesia
| | - Barolym Tri Pamungkas
- Department of Pharmaceutical BiologyFaculty of PharmacyUniversitas Mulawarman, Samarinda, East Kalimantan, Indonesia
| | - Zulpakor Oktoba
- Department of PharmacyFaculty of MedicineUniversitas Lampung, Bandar Lampung, Indonesia
| | - Maya Arfania
- Department of Pharmacology and Clinical PharmacyFaculty of PharmacyUniversitas Buana Perjuangan Karawang, Karawang, West Java, Indonesia
| | - Nia Yuniarsih
- Department of Pharmaceutical TechnologyFaculty of PharmacyUniversitas Buana Perjuangan Karawang, Karawang, West Java, Indonesia
| | - Eko Sri Wahyuningsih
- Department of Pharmaceutical BiologyFaculty of PharmacyUniversitas Buana Perjuangan Karawang, Karawang, West Java, Indonesia
| | - Yuliani Dewi
- Faculty of PharmacyUniversitas Buana Perjuangan Karawang, Karawang, West Java, Indonesia
| | - Sri Ayu Winarti
- Faculty of PharmacyUniversitas Buana Perjuangan Karawang, Karawang, West Java, Indonesia
| | - Sri Tantia Dinita
- Faculty of PharmacyUniversitas Buana Perjuangan Karawang, Karawang, West Java, Indonesia
| |
Collapse
|
4
|
Amini N, Nejaddehbashi F, Badavi M, Bayati V, Zahra Basir. Combined effect of naringin and adipose tissue-derived mesenchymal stem cell on cisplatin nephrotoxicity through Sirtuin1/Nrf-2/HO-1 signaling pathway: a promising nephroprotective candidate. Cell Tissue Res 2024; 397:193-204. [PMID: 38953985 DOI: 10.1007/s00441-024-03902-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 06/19/2024] [Indexed: 07/04/2024]
Abstract
Cisplatin nephrotoxicity is a well-known emergency clinical condition caused by oxidative stress and inflammation. Naringin (NAR) is considered an antioxidant agent with renoprotective effects capable of removing reactive oxygen species. Adipose tissue-derived mesenchymal stem cells (AD-MSCs) are reported to have anti-inflammatory and antioxidant properties. The present research examined the renoprotective effect of the combination of NAR and AD-MSCs as opposed to each one alone on cisplatin-induced nephrotoxicity through SIRT-1/Nrf-2/HO-1 pathway. This study included five groups (n = 8 each) of male Sprague-Dawley rats (200 - 220 g): sham, cisplatin: rats receiving cisplatin (6.5 mg/kg, i.p.) on the 4th day; NAR+cisplatin: rats pretreated with NAR (1 week, i.p.) + cisplatin on the 4th day; AD-MSCs: rats receiving AD-MSCs (1 × 106) by injection through the tail vein on the 5th day + cisplatin on the 4th day; and NAR+AD-MSCs+cisplatin. On the 8th day, the animals were anesthetized to obtain tissue and blood samples. Biochemical factors, inflammation, oxidative stress, and gene expression were explored. Cisplatin increased blood urea nitrogen, creatinine, inflammation, and oxidative stress. Moreover, mRNA expression of Sirtuin1, nuclear factor erythroid 2-related factor 2 (Nrf-2), and heme oxygenase-1 (HO-1) remarkably reduced. Furthermore, cisplatin led to a disturbance in kidney structure (glomerular atrophy, cell infiltrations, and tubular dysfunction) as confirmed by histology findings. However, NAR pretreatment, AD-MSC administration, or a combination of both significantly reversed these changes. Overall, when used together, NAR and AD-MSCs had stronger cisplatin-induced effects on kidney dysfunction by inhibiting inflammation, reducing oxidative stress, and increasing the Sirtuin1/Nrf-2/HO-1 pathway.
Collapse
Affiliation(s)
- Negin Amini
- Department of Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Fereshteh Nejaddehbashi
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Badavi
- Department of Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Vahid Bayati
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zahra Basir
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| |
Collapse
|
5
|
Shu T, Li J, Gu J, Wu L, Xie P, Zhang D, Li W, Wan J, Zheng X. Long noncoding RNA UCA1 promotes the chondrogenic differentiation of human bone marrow mesenchymal stem cells via regulating PARP1 ubiquitination. Stem Cells 2024; 42:752-762. [PMID: 38829368 DOI: 10.1093/stmcls/sxae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 04/30/2024] [Indexed: 06/05/2024]
Abstract
Bone marrow mesenchymal stem cells (BMSCs) possess the potential to differentiate into cartilage cells. Long noncoding RNA (lncRNAs) urothelial carcinoma associated 1 (UCA1) has been confirmed to improve the chondrogenic differentiation of marrow mesenchymal stem cells (MSCs). Herein, we further investigated the effects and underlying mechanisms of these processes. The expression of UCA1 was positively associated with chondrogenic differentiation and the knockdown of UCA1 has been shown to attenuate the expression of chondrogenic markers. RNA pull-down assay and RNA immunoprecipitation showed that UCA1 could directly bind to PARP1 protein. UCA1 could improve PARP1 protein via facilitating USP9X-mediated PARP1 deubiquitination. Then these processes stimulated the NF-κB signaling pathway. In addition, PARP1 was declined in UCA1 knockdown cells, and silencing of PARP1 could diminish the increasing effects of UCA1 on the chondrogenic differentiation from MSCs and signaling pathway activation. Collectively, these outcomes suggest that UCA1 could act as a mediator of PARP1 protein ubiquitination and develop the chondrogenic differentiation of MSCs.
Collapse
Affiliation(s)
- Tao Shu
- Department of Orthopaedics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518000, People's Republic of China
- Department of Spine Surgery, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China
| | - Jiachun Li
- Department of Orthopaedics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518000, People's Republic of China
| | - Juyuan Gu
- Department of Orthopedics, Hebei Medical University Third Hospital, Shijiazhuang, Hebei 050051, People's Republic of China
| | - Liang Wu
- Department of Orthopedics, South China Hospital, Health Science Center, Shenzhen University, Shenzhen 518116, People's Republic of China
| | - Peng Xie
- Department of Nuclear Medicine, Hebei Medical University Third Hospital, Shijiazhuang, Hebei 050051, People's Republic of China
| | - Dongfeng Zhang
- Department of Orthopedics, South China Hospital, Health Science Center, Shenzhen University, Shenzhen 518116, People's Republic of China
| | - Wen Li
- Department of Spine Surgery, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China
| | - Junming Wan
- Department of Orthopaedics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518000, People's Republic of China
| | - Xiaozuo Zheng
- Department of Orthopedics, Hebei Medical University Third Hospital, Shijiazhuang, Hebei 050051, People's Republic of China
| |
Collapse
|
6
|
Shang T, Jiang T, Cui X, Pan Y, Feng X, Dong L, Wang H. Diverse functions of SOX9 in liver development and homeostasis and hepatobiliary diseases. Genes Dis 2024; 11:100996. [PMID: 38523677 PMCID: PMC10958229 DOI: 10.1016/j.gendis.2023.03.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 02/13/2023] [Accepted: 03/19/2023] [Indexed: 03/26/2024] Open
Abstract
The liver is the central organ for digestion and detoxification and has unique metabolic and regenerative capacities. The hepatobiliary system originates from the foregut endoderm, in which cells undergo multiple events of cell proliferation, migration, and differentiation to form the liver parenchyma and ductal system under the hierarchical regulation of transcription factors. Studies on liver development and diseases have revealed that SRY-related high-mobility group box 9 (SOX9) plays an important role in liver embryogenesis and the progression of hepatobiliary diseases. SOX9 is not only a master regulator of cell fate determination and tissue morphogenesis, but also regulates various biological features of cancer, including cancer stemness, invasion, and drug resistance, making SOX9 a potential biomarker for tumor prognosis and progression. This review systematically summarizes the latest findings of SOX9 in hepatobiliary development, homeostasis, and disease. We also highlight the value of SOX9 as a novel biomarker and potential target for the clinical treatment of major liver diseases.
Collapse
Affiliation(s)
- Taiyu Shang
- School of Life Sciences, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai 200438, China
| | - Tianyi Jiang
- National Center for Liver Cancer, The Naval Medical University, Shanghai 201805, China
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China
| | - Xiaowen Cui
- National Center for Liver Cancer, The Naval Medical University, Shanghai 201805, China
| | - Yufei Pan
- National Center for Liver Cancer, The Naval Medical University, Shanghai 201805, China
| | - Xiaofan Feng
- National Center for Liver Cancer, The Naval Medical University, Shanghai 201805, China
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China
| | - Liwei Dong
- National Center for Liver Cancer, The Naval Medical University, Shanghai 201805, China
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China
| | - Hongyang Wang
- School of Life Sciences, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai 200438, China
- National Center for Liver Cancer, The Naval Medical University, Shanghai 201805, China
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China
- Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer, Second Military Medical University & Ministry of Education, Shanghai 200438, China
| |
Collapse
|
7
|
Chen H, Ding Q, Li L, Wei P, Niu Z, Kong T, Fu P, Wang Y, Li J, Wang K, Zheng J. Extracellular Vesicle Spherical Nucleic Acids. JACS AU 2024; 4:2381-2392. [PMID: 38938802 PMCID: PMC11200237 DOI: 10.1021/jacsau.4c00338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/29/2024]
Abstract
Extracellular vesicles (EVs) are naturally occurring vesicles secreted by cells that can transport cargo between cells, making them promising bioactive nanomaterials. However, due to the complex and heterogeneous biological characteristics, a method for robust EV manipulation and efficient EV delivery is still lacking. Here, we developed a novel class of extracellular vesicle spherical nucleic acid (EV-SNA) nanostructures with scalability, programmability, and efficient cellular delivery. EV-SNA was constructed through the simple hydrophobic coassembly of natural EVs with cholesterol-modified oligonucleotides and can be stable for 1 month at room temperature. Based on programmable nucleic acid shells, EV-SNA can respond to AND logic gates to achieve vesicle assembly manipulation. Importantly, EV-SNA can be constructed from a wide range of biological sources EV, enhancing cellular delivery capability by nearly 10-20 times. Compared to artificial liposomal SNA, endogenous EV-SNA exhibited better biocompatibility and more effective delivery of antisense oligonucleotides in hard-to-transfect primary stem cells. Additionally, EV-SNA can deliver functional EVs for immune regulation. As a novel material form, EV-SNA may provide a modular and programmable framework paradigm for EV-based applications in drug delivery, disease treatment, nanovaccines, and other fields.
Collapse
Affiliation(s)
- Hao Chen
- Ningbo
Key Laboratory of Biomedical Imaging Probe Materials and Technology,
Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese
Academy of Sciences, Ningbo 315300, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiaojiao Ding
- Cixi
Biomedical Research Institute, Wenzhou Medical
University, Wenzhou 325035, China
| | - Lin Li
- Ningbo
Key Laboratory of Biomedical Imaging Probe Materials and Technology,
Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese
Academy of Sciences, Ningbo 315300, China
| | - Pengyao Wei
- Cixi
Biomedical Research Institute, Wenzhou Medical
University, Wenzhou 325035, China
| | - Zitong Niu
- Cixi
Biomedical Research Institute, Wenzhou Medical
University, Wenzhou 325035, China
| | - Tong Kong
- Ningbo
Key Laboratory of Biomedical Imaging Probe Materials and Technology,
Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese
Academy of Sciences, Ningbo 315300, China
| | - Pan Fu
- Ningbo
Key Laboratory of Biomedical Imaging Probe Materials and Technology,
Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese
Academy of Sciences, Ningbo 315300, China
| | - Yuhui Wang
- Ningbo
Key Laboratory of Biomedical Imaging Probe Materials and Technology,
Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese
Academy of Sciences, Ningbo 315300, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiang Li
- Institute
of Materiobiology, Department of Chemistry, College of Science, Shanghai University, Shanghai 200444, China
| | - Kaizhe Wang
- Ningbo
Key Laboratory of Biomedical Imaging Probe Materials and Technology,
Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese
Academy of Sciences, Ningbo 315300, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianping Zheng
- Ningbo
Key Laboratory of Biomedical Imaging Probe Materials and Technology,
Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese
Academy of Sciences, Ningbo 315300, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
8
|
Zhra M, Magableh AM, Samhan LM, Fatani LM, Qasem RJ, Aljada A. The Expression of a Subset of Aging and Antiaging Markers Following the Chondrogenic and Osteogenic Differentiation of Mesenchymal Stem Cells of Placental Origin. Cells 2024; 13:1022. [PMID: 38920652 PMCID: PMC11201886 DOI: 10.3390/cells13121022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/27/2024] Open
Abstract
Mesenchymal stem cells (MSCs) of placental origin hold great promise in tissue engineering and regenerative medicine for diseases affecting cartilage and bone. However, their utility has been limited by their tendency to undergo premature senescence and phenotypic drift into adipocytes. This study aimed to explore the potential involvement of a specific subset of aging and antiaging genes by measuring their expression prior to and following in vitro-induced differentiation of placental MSCs into chondrocytes and osteoblasts as opposed to adipocytes. The targeted genes of interest included the various LMNA/C transcript variants (lamin A, lamin C, and lamin A∆10), sirtuin 7 (SIRT7), and SM22α, along with the classic aging markers plasminogen activator inhibitor 1 (PAI-1), p53, and p16INK4a. MSCs were isolated from the decidua basalis of human term placentas, expanded, and then analyzed for phenotypic properties by flow cytometry and evaluated for colony-forming efficiency. The cells were then induced to differentiate in vitro into chondrocytes, osteocytes, and adipocytes following established protocols. The mRNA expression of the targeted genes was measured by RT-qPCR in the undifferentiated cells and those fully differentiated into the three cellular lineages. Compared to undifferentiated cells, the differentiated chondrocytes demonstrated decreased expression of SIRT7, along with decreased PAI-1, lamin A, and SM22α expression, but the expression of p16INK4a and p53 increased, suggesting their tendency to undergo premature senescence. Interestingly, the cells maintained the expression of lamin C, which indicates that it is the primary lamin variant influencing the mechanoelastic properties of the differentiated cells. Notably, the expression of all targeted genes did not differ from the undifferentiated cells following osteogenic differentiation. On the other hand, the differentiation of the cells into adipocytes was associated with decreased expression of lamin A and PAI-1. The distinct patterns of expression of aging and antiaging genes following in vitro-induced differentiation of MSCs into chondrocytes, osteocytes, and adipocytes potentially reflect specific roles for these genes during and following differentiation in the fully functional cells. Understanding these roles and the network of signaling molecules involved can open opportunities to improve the handling and utility of MSCs as cellular precursors for the treatment of cartilage and bone diseases.
Collapse
Affiliation(s)
- Mahmoud Zhra
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Ahmad M. Magableh
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Lara M. Samhan
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Lein M. Fatani
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Rani J. Qasem
- Department of Pharmacology and Pharmacy Practice, College of Pharmacy, Middle East University, Amman 11831, Jordan
| | - Ahmad Aljada
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
9
|
Elsaid FH, Hussein AM, Eid EA, Ammar OA, Khalil AA. Effect of intermittent fasting on adriamycin-induced nephropathy: Possible underlying mechanisms. Tissue Cell 2024; 88:102360. [PMID: 38489913 DOI: 10.1016/j.tice.2024.102360] [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: 10/28/2023] [Revised: 02/16/2024] [Accepted: 03/12/2024] [Indexed: 03/17/2024]
Abstract
PURPOSE Intermittent fasting (IF) has been shown to induce a well-organized adaptive defense against stress inside the cells, which increases the production of anti-oxidant defenses, repair of DNA, biogenesis of mitochondria, and genes that combat inflammation. So, the goal of the current investigation was to identify the effects of IF on rats with adriamycin (ADR)-induced nephropathy and any potential underlying mechanisms. METHODS Four groups of 40 mature Sprague-Dawley male rats were allocated as follow; control, fasting, ADR, and ADR plus fasting. After 8 weeks of ADR administration urine, blood samples and kidneys were taken for assessment of serum creatinine (Cr), BUN, urinary proteins, indicators of oxidative damage (malondialdehyde (MDA), reduced glutathione (GSH) and Catalase (CAT) levels), histopathological examinations, immunohistochemical examinations for caspase-3, Sirt1, aquaporin2 (AQP2) and real time PCR for antioxidant genes; Nrf2, HO-1 in kidney tissues. RESULTS IF significantly improved serum creatinine, BUN and urinary protein excretion, oxidative stress (low MDA with high CAT and GSH), in addition to morphological damage to the renal tubules and glomeruli as well as caspase-3 production during apoptosis. Moreover, IF stimulates significantly the expression of Sirt1 and Nrf2/HO-1 and AQP2. CONCLUSION AQP2, Sirt1, Nrf2/HO-1 signaling may be upregulated and activated by IF, which alleviates ADR nephropathy. Enhancing endogenous antioxidants, reducing apoptosis and tubulointerstitial damage, and maintaining the glomerular membrane's integrity are other goals.
Collapse
Affiliation(s)
- Fathy H Elsaid
- Department of Medical Physiology, Faculty of Medicine, Al-Azhar University, Assuit, Egypt
| | - Abdelaziz M Hussein
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Elsayed A Eid
- Department of Internal Medicine and Endocrinology, Faculty of Medicine, Delta University for Science and Technology, Gamasa, Egypt
| | - Omar A Ammar
- Department of Basic Science, Faculty of Applied Health Science and Technology, Delta University for Science and Technology, Gamasa, Egypt
| | - Ali Ali Khalil
- Department of Medical Physiology, Faculty of Medicine, Al-Azhar University, Assuit, Egypt
| |
Collapse
|
10
|
Brockmueller A, Sajeev A, Koklesova L, Samuel SM, Kubatka P, Büsselberg D, Kunnumakkara AB, Shakibaei M. Resveratrol as sensitizer in colorectal cancer plasticity. Cancer Metastasis Rev 2024; 43:55-85. [PMID: 37507626 PMCID: PMC11016130 DOI: 10.1007/s10555-023-10126-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023]
Abstract
Despite tremendous medical treatment successes, colorectal cancer (CRC) remains a leading cause of cancer deaths worldwide. Chemotherapy as monotherapy can lead to significant side effects and chemoresistance that can be linked to several resistance-activating biological processes, including an increase in inflammation, cellular plasticity, multidrug resistance (MDR), inhibition of the sentinel gene p53, and apoptosis. As a consequence, tumor cells can escape the effectiveness of chemotherapeutic agents. This underscores the need for cross-target therapeutic approaches that are not only pharmacologically safe but also modulate multiple potent signaling pathways and sensitize cancer cells to overcome resistance to standard drugs. In recent years, scientists have been searching for natural compounds that can be used as chemosensitizers in addition to conventional medications for the synergistic treatment of CRC. Resveratrol, a natural polyphenolic phytoalexin found in various fruits and vegetables such as peanuts, berries, and red grapes, is one of the most effective natural chemopreventive agents. Abundant in vitro and in vivo studies have shown that resveratrol, in interaction with standard drugs, is an effective chemosensitizer for CRC cells to chemotherapeutic agents and thus prevents drug resistance by modulating multiple pathways, including transcription factors, epithelial-to-mesenchymal transition-plasticity, proliferation, metastasis, angiogenesis, cell cycle, and apoptosis. The ability of resveratrol to modify multiple subcellular pathways that may suppress cancer cell plasticity and reversal of chemoresistance are critical parameters for understanding its anti-cancer effects. In this review, we focus on the chemosensitizing properties of resveratrol in CRC and, thus, its potential importance as an additive to ongoing treatments.
Collapse
Affiliation(s)
- Aranka Brockmueller
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Pettenkoferstr. 11, D-80336, Munich, Germany
| | - Anjana Sajeev
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Guwahati, Guwahati, Assam, 781039, India
| | - Lenka Koklesova
- Clinic of Gynecology and Obstetrics, Jessenius Faculty of Medicine, Comenius University in Bratislava, Kollarova 2, 03601, Martin, Slovakia
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar (Medbay), Education City, Qatar Foundation, 24144, Doha, Qatar
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 03601, Martin, Slovakia
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar (Medbay), Education City, Qatar Foundation, 24144, Doha, Qatar
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Guwahati, Guwahati, Assam, 781039, India
| | - Mehdi Shakibaei
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Pettenkoferstr. 11, D-80336, Munich, Germany.
| |
Collapse
|
11
|
Feng S, Qi Y, Xiao Z, Chen H, Liu S, Luo H, Wu H, Zhang W. CircHIPK3 relieves vascular calcification via mediating SIRT1/PGC-1α/MFN2 pathway by interacting with FUS. BMC Cardiovasc Disord 2023; 23:583. [PMID: 38012555 PMCID: PMC10683355 DOI: 10.1186/s12872-023-03602-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 11/07/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Circular RNAs (circRNAs) have been reported to regulate the biological processes of human diseases. CircHIPK3 has been implicated in vascular calcification, but the downstream regulatory mechanisms remain unclear. Our study aimed to understand the regulatory function of circHIPK3 in vascular calcification. METHODS CircHIPK3 expression in atherosclerosis (AS) serum samples and vascular smooth muscle cells (VSMCs) calcification model was assessed by quantitative real-time polymerase chain reaction (qRT-PCR). The binding relationships between fused in sarcoma (FUS) and circHIPK3 or sirtuin 1 (SIRT1) were verified by RNA immunoprecipitation (RIP) assay and RNA pull-down assays. Alkaline phosphatase (ALP) activity and alizarin red staining assays were performed to evaluate the biological effect of β-glycerophosphate (β-GP) and circHIPK3 on calcium deposition. qRT-PCR and western blot assays were used to examine the effect of β-GP, circHIPK3, SIRT1, mitofusin 2 (MFN2), and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) on VSMCs calcification and the expression of calcification-related proteins. RESULTS In AS serum samples and VSMCs calcification model, the expression of circHIPK3 was significantly reduced. CircHIPK3 overexpression inhibited ALP activity and calcium deposition in β-GP-induced VSMCs. Moreover, circHIPK3 could recruit FUS to further stabilize SIRT1 mRNA. CircHIPK3 promoted MFN2 expression to alleviate VSMCs calcification via activating SIRT1/PGC-1α signaling. CONCLUSION The positive regulation of circHIPK3/FUS/SIRT1/PGC-1α/MFN2 signaling pathway contributed to the alleviate VSMCs calcification, revealing a novel regulatory axis for vascular calcification.
Collapse
Affiliation(s)
- Siyi Feng
- Department of Ultrasound Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan Province, China
| | - Youfei Qi
- Department of Vascular Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19, Xiuhua Road, Haikou, 570311, Hainan Province, China
| | - Zhanxiang Xiao
- Department of Vascular Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19, Xiuhua Road, Haikou, 570311, Hainan Province, China
| | - Hao Chen
- Department of Vascular Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19, Xiuhua Road, Haikou, 570311, Hainan Province, China
| | - Sahua Liu
- Department of Vascular Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19, Xiuhua Road, Haikou, 570311, Hainan Province, China
| | - Haimei Luo
- Department of Vascular Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19, Xiuhua Road, Haikou, 570311, Hainan Province, China
| | - Hongfei Wu
- Department of Vascular Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19, Xiuhua Road, Haikou, 570311, Hainan Province, China
| | - Wenbo Zhang
- Department of Vascular Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19, Xiuhua Road, Haikou, 570311, Hainan Province, China.
| |
Collapse
|
12
|
Elsied MA, Sharawi ZW, Al-Amrah H, Hegazy RA, Mohamed AE, Saleh RM, El-kholy SS, Farrag FA, Fayed MH, El-Magd MA. Walnut Kernel Oil and Defatted Extracts Enhance Mesenchymal Stem Cell Stemness and Delay Senescence. Molecules 2023; 28:6281. [PMID: 37687109 PMCID: PMC10488345 DOI: 10.3390/molecules28176281] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/21/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Decreased stemness and increased cellular senescence impair the ability of mesenchymal stem cells (MSCs) to renew themselves, change into different cell types, and contribute to regenerative medicine. There is an urgent need to discover new compounds that can boost MSCs' stemness and delay senescence. Therefore, this study aimed to investigate the impact of walnut kernel oil (WKO) and defatted (WKD) extracts on bone marrow (BM)-MSC stemness and senescence. Premature senescence and inflammation were induced in BM-MSCs using H2O2 and LPS, respectively. Phytochemical constituents of WKO and WKD extracts were detected by HPLC. The stemness (proliferation and migration), senescence-related markers (p53, p21, SIRT1, and AMPK), oxidative stress/antioxidant markers, inflammatory cytokines, and cell cycle of BM-MSCs were measured by MTT assay, qPCR, ELISA, and flow cytometry. WKO and WKD extracts improved rat BM-MSC stemness, as evidenced by (1) increased cell viability, (2) decreased apoptosis (low levels of Bax and caspase3 and high levels of Bcl2), (3) upregulated MMP9 and downregulated TIMP1 expression, and (4) cell cycle arrest in the G0/G1 phase and declined cell number in the S and G2/M phases. Additionally, WKO and WKD extracts reduced rat BM-MSC senescence, as indicated by (1) decreased p53 and p21 expression, (2) upregulated expression and levels of SIRT1 and AMPK, (3) reduced levels of ROS and improved antioxidant activity (higher activity of CAT, SOD, and GPx and upregulated expression of NrF2 and HO-1), and (4) declined levels of TNFα, IL1β, and NF-κB. When compared to the WKO extract, the WKD extract had a greater impact on the induction of stemness and reduction of senescence of BM-MSCs due to its stronger antioxidant activity, which could be attributed to its higher levels of flavonoids and phenolic compounds, as detected by HPLC analysis. WKO and WKD extracts enhance rat BM-MSC stemness and protect them from senescence, suggesting their potential use as enhancers to increase MSCs' therapeutic efficacy.
Collapse
Affiliation(s)
- Marwa A. Elsied
- Anatomy and Embryology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh 33516, Egypt
| | - Zeina W. Sharawi
- Biological Sciences Department, Faculty of Sciences, King AbdulAziz University, Jeddah 21589, Saudi Arabia; (Z.W.S.); (H.A.-A.)
| | - Hadba Al-Amrah
- Biological Sciences Department, Faculty of Sciences, King AbdulAziz University, Jeddah 21589, Saudi Arabia; (Z.W.S.); (H.A.-A.)
| | - Rabab A. Hegazy
- Department of Biology, Al-Darb University College, Jazan University, Jazan 45142, Saudi Arabia;
| | - Amro E. Mohamed
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta 31527, Egypt;
| | - Rasha M. Saleh
- Department of Physiology, Faculty of Veterinary Medicine, Mansura University, Mansura 35516, Egypt;
| | - Sanad S. El-kholy
- Department of Physiology, Faculty of Medicine, Kafrelsheikh University, Kafr El-Shaikh 33516, Egypt;
| | - Foad A. Farrag
- Anatomy and Embryology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh 33516, Egypt
| | - Masoud H. Fayed
- Anatomy and Embryology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh 33516, Egypt
| | - Mohammed A. El-Magd
- Anatomy and Embryology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh 33516, Egypt
| |
Collapse
|
13
|
Brockmueller A, Girisa S, Motallebi M, Kunnumakkara AB, Shakibaei M. Calebin A targets the HIF-1α/NF-κB pathway to suppress colorectal cancer cell migration. Front Pharmacol 2023; 14:1203436. [PMID: 37583906 PMCID: PMC10423823 DOI: 10.3389/fphar.2023.1203436] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 07/14/2023] [Indexed: 08/17/2023] Open
Abstract
Background: Hypoxia-inducible factor-1α (HIF-1α) is one of the major tumor-associated transcription factors modulating numerous tumor properties such as tumor cell metabolism, survival, proliferation, angiogenesis, and metastasis. Calebin A (CA), a compound derived from turmeric, is known for its anti-cancer activity through modulation of the NF-κB pathway. However, its impact on HIF-1α in colorectal cancer (CRC) cell migration is unknown. Methods: Human CRC cells (HCT-116) in 3D alginate and monolayer multicellular TME (fibroblasts/T lymphocytes) were subjected to CA or the HIF-1α inhibitor to explore the efficacy of CA on TME-induced inflammation, migration, and tumor malignancy. Results: CA significantly inhibited TME-promoted proliferation and migration of HCT-116 cells, similar to the HIF-1α inhibitor. Colony formation, toluidine blue staining, and immunolabeling showed that CA inhibited the migration of HCT-116 cells partly by inhibiting HIF-1α, which is critical for CRC cell viability, and these observations were confirmed by electron microscopy. In addition, Western blot analysis confirmed that CA inhibited TME-initiated expression of HIF-1α and biomarkers of metastatic factors (such as NF-κB, β1-integrin, and VEGF), and promoted apoptosis (caspase-3), in a manner comparable to the HIF-1α inhibitor. Finally, TME induced a purposeful pairing between HIF-1α and NF-κB, suggesting that the synergistic interplay between the two tumor-associated transcription factors is essential for CRC cell malignancy and migration and that CA silences these factors in tandem. Conclusion: These results shed light on a novel regulatory modulation of CA signaling in CRC cell migration, partially via HIF-1α/NF-κB with potentially relevant implications for cancer therapy.
Collapse
Affiliation(s)
- Aranka Brockmueller
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Mahzad Motallebi
- Department of Biology, Yadegar-e-Imam Khomeini Shahr-e-Rey Branch, Islamic Azad University, Tehran, Iran
| | - Ajaikumar B. Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Mehdi Shakibaei
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
14
|
Fay LY, Kuo CH, Chang HK, Yeh MY, Chang CC, Ko CC, Tu TH, Kuo YH, Hsu WY, Hung CH, Chen CJ, Wu JC, Tsai MJ, Huang WC, Cheng H, Lee MJ. Comparative Study of the Cytokine Profiles of Serum and Tissues from Patients with the Ossification of the Posterior Longitudinal Ligament. Biomedicines 2023; 11:2021. [PMID: 37509659 PMCID: PMC10377187 DOI: 10.3390/biomedicines11072021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/12/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND The ossification of the posterior longitudinal ligament (OPLL) is one of the contributing factors leading to severe cervical spondylotic myelopathy (CSM). The mechanism causing ossification is still unclear. The current study was designed to analyze the specimens of patients with or without OPLL. METHODS The study collected 51 patients with cervical spondylosis. There were six serum samples in both the non-OPLL (NOPLL) and OPLL groups. For tissue analysis, there were seven samples in the NOPLL group and five samples in the OPLL group. The specimens of serum and tissue were analyzed by using Human Cytokine Antibody Arrays to differentiate biomarkers between the OPLL and NOPLL groups, as well as between serum and OPLL tissue. Immunohistochemical staining of the ligament tissue was undertaken for both groups. RESULTS For OPLL vs. NOPLL, the serum leptin levels are higher in the OPLL group, corroborating others' observations that it may serve as a disease marker. In the tissue, angiogenin (ANG), osteopontin (OPN), and osteopro-tegerin (OPG) are higher than they are in the OPLL group (p < 0.05). For serum vs. OPLL tissue, many chemotactic cytokines demonstrated elevated levels of MIP1 delta, MCP-1, and RANTES in the serum, while many cytokines promoting or regulating bone genesis were up-regulated in tissue (oncostatin M, FGF-9, LIF, osteopontin, osteoprotegerin, TGF-beta2), as well as the factor that inhibits osteoclastogenesis (IL-10), with very few cytokines responsible for osteoclastogenesis. Molecules promoting angiogenesis, including angiotensin, vEGF, and osteoprotegerin, are abundant in the OPLL tissue, which paves the way for robust bone growth.
Collapse
Affiliation(s)
- Li-Yu Fay
- Institute of Pharmacology, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Chao-Hung Kuo
- School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, and National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
| | - Hsuan-Kan Chang
- School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Mei-Yin Yeh
- Institute of Pharmacology, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Chih-Chang Chang
- School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Chin-Chu Ko
- Institute of Pharmacology, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Tsung-Hsi Tu
- School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Yi-Hsuan Kuo
- School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Wang-Yu Hsu
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Chien-Hui Hung
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Ching-Jung Chen
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Jau-Ching Wu
- Institute of Pharmacology, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - May-Jywan Tsai
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Wen-Cheng Huang
- School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Henrich Cheng
- Institute of Pharmacology, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Meng-Jen Lee
- Department of Applied Chemistry, Chaoyang University of Technology, 168, Jifeng E. Rd., Taichung 413310, Taiwan
| |
Collapse
|
15
|
Saito H, Yayama T, Mori K, Kumagai K, Fujikawa H, Chosei Y, Imai S. Increased Cellular Expression of Interleukin-6 in Patients With Ossification of the Posterior Longitudinal Ligament. Spine (Phila Pa 1976) 2023; 48:E78-E86. [PMID: 36729990 DOI: 10.1097/brs.0000000000004557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/24/2022] [Indexed: 02/03/2023]
Abstract
STUDY DESIGN We performed histologic, immunohistochemical, immunoblot examination and suspension array analyses of cytokine expression in cultured cells derived from human cervical ossification of the posterior longitudinal ligament (OPLL). OBJECTIVE To determine the roles of interleukin-6 (IL-6) during the maturation of osteoblasts and chondrocytes associated with the development of OPLL. SUMMARY OF BACKGROUND DATA Ectopic OPLL affects ~3% of the general population, with a higher incidence in Asian ethnic groups. Alterations in cytokine profiles may influence osteoblast differentiation, but the mechanisms and signaling pathways associated with the ossification process remain unclear. METHODS Samples were collected from 14 patients with OPLL who had undergone spinal surgery and seven with cervical spondylotic myelopathy without OPLL. Tissue sections were used for histologic and immunohistochemical studies, and primary cells from ligamentum samples were used for cytokine array and immunoblotting. A suspension array was used to measure the concentrations of 27 inflammatory cytokines or growth factors. RESULTS Suspension array and immunoblot analyses revealed significantly elevated levels of IL-6 in OPLL patients. Alterations in IL-6 concentrations were found to alter the expression of the genes Sox9 , Runx2 , and SIRT1 . In addition, immunohistochemical analysis revealed that these factors are present in mesenchymal cells within the degenerative portion of the ligament matrix that is adjacent to the ossification front. CONCLUSIONS IL-6 plays a profound role in the osteoblast differentiation process along with the induction of chondrocyte hypertrophy and cell apoptosis in the early stages of ossification in OPLL. These changes in cytokine profiles are essential factors for regulation of the ectopic ossified plaque in OPLL.
Collapse
Affiliation(s)
- Hideki Saito
- Department of Orthopaedic Surgery, Shiga University of Medical Science, Shiga, Japan
| | | | | | | | | | | | | |
Collapse
|
16
|
Brockmueller A, Girisa S, Kunnumakkara AB, Shakibaei M. Resveratrol Modulates Chemosensitisation to 5-FU via β1-Integrin/HIF-1α Axis in CRC Tumor Microenvironment. Int J Mol Sci 2023; 24:ijms24054988. [PMID: 36902421 PMCID: PMC10003050 DOI: 10.3390/ijms24054988] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/26/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Frequent development of resistance to chemotherapeutic agents such as 5-flourouracil (5-FU) complicates the treatment of advanced colorectal cancer (CRC). Resveratrol is able to utilize β1-integrin receptors, strongly expressed in CRC cells, to transmit and exert anti-carcinogenic signals, but whether it can also utilize these receptors to overcome 5-FU chemoresistance in CRC cells has not yet been investigated. Effects of β1-integrin knockdown on anti-cancer capabilities of resveratrol and 5-FU were investigated in HCT-116 and 5-FU-resistant HCT-116R CRC tumor microenvironment (TME) with 3D-alginate as well as monolayer cultures. Resveratrol increased CRC cell sensitivity to 5-FU by reducing TME-promoted vitality, proliferation, colony formation, invasion tendency and mesenchymal phenotype including pro-migration pseudopodia. Furthermore, resveratrol impaired CRC cells in favor of more effective utilization of 5-FU by down-regulating TME-induced inflammation (NF-kB), vascularisation (VEGF, HIF-1α) and cancer stem cell production (CD44, CD133, ALDH1), while up-regulating apoptosis (caspase-3) that was previously inhibited by TME. These anti-cancer mechanisms of resveratrol were largely abolished by antisense oligonucleotides against β1-integrin (β1-ASO) in both CRC cell lines, indicating the particular importance of β1-integrin receptors for the 5-FU-chemosensitising effect of resveratrol. Lastly, co-immunoprecipitation tests showed that resveratrol targets and modulates the TME-associated β1-integrin/HIF-1α signaling axis in CRC cells. Our results suggest for the first time the utility of the β1-integrin/HIF-1α signaling axis related to chemosensitization and overcoming chemoresistance to 5-FU in CRC cells by resveratrol, underlining its potential supportive applications in CRC treatment.
Collapse
Affiliation(s)
- Aranka Brockmueller
- Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-University Munich, Pettenkoferstr. 11, D-80336 Munich, Germany
| | - Sosmitha Girisa
- Cancer Biology Laboratory and DBT-AIST International Centre for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Guwahati, Guwahati 781039, India
| | - Ajaikumar B. Kunnumakkara
- Cancer Biology Laboratory and DBT-AIST International Centre for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Guwahati, Guwahati 781039, India
| | - Mehdi Shakibaei
- Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-University Munich, Pettenkoferstr. 11, D-80336 Munich, Germany
- Correspondence: ; Tel.: +49-89-2180-72624
| |
Collapse
|
17
|
Ciccone L, Piragine E, Brogi S, Camodeca C, Fucci R, Calderone V, Nencetti S, Martelli A, Orlandini E. Resveratrol-like Compounds as SIRT1 Activators. Int J Mol Sci 2022; 23:ijms232315105. [PMID: 36499460 PMCID: PMC9738298 DOI: 10.3390/ijms232315105] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 12/05/2022] Open
Abstract
The sirtuin 1 (SIRT1) activator resveratrol has emerged as a promising candidate for the prevention of vascular oxidative stress, which is a trigger for endothelial dysfunction. However, its clinical use is limited by low oral bioavailability. In this work, we have applied a previously developed computational protocol to identify the most promising derivatives from our in-house chemical library of resveratrol derivatives. The most promising compounds in terms of SIRT1 activation and oral bioavailability, predicted in silico, were evaluated for their ability to activate the isolated SIRT1 enzyme. Then, we assessed the antioxidant effects of the most effective derivative, compound 3d, in human umbilical vein endothelial cells (HUVECs) injured with H2O2 100 µM. The SIRT1 activator 3d significantly preserved cell viability and prevented an intracellular reactive oxygen species increase in HUVECs exposed to the oxidative stimulus. Such effects were partially reduced in the presence of a sirtuin inhibitor, sirtinol, confirming the potential role of sirtuins in the activity of resveratrol and its derivatives. Although 3d appeared less effective than resveratrol in activating the isolated enzyme, the effects exhibited by both compounds in HUVECs were almost superimposable, suggesting a higher ability of 3d to cross cell membranes and activate the intracellular target SIRT1.
Collapse
Affiliation(s)
- Lidia Ciccone
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
- Center for Instrument Sharing, University of Pisa (CISUP), Lungarno Pacinotti 43, 56126 Pisa, Italy
| | - Eugenia Piragine
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Simone Brogi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Caterina Camodeca
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Raffaele Fucci
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
- Center for Instrument Sharing, University of Pisa (CISUP), Lungarno Pacinotti 43, 56126 Pisa, Italy
| | - Susanna Nencetti
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
- Center for Instrument Sharing, University of Pisa (CISUP), Lungarno Pacinotti 43, 56126 Pisa, Italy
- Correspondence: (S.N.); (A.M.)
| | - Alma Martelli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
- Center for Instrument Sharing, University of Pisa (CISUP), Lungarno Pacinotti 43, 56126 Pisa, Italy
- Correspondence: (S.N.); (A.M.)
| | - Elisabetta Orlandini
- Department of Earth Science, University of Pisa, Via Santa Maria 53, 56126 Pisa, Italy
- Research Centre E. Piaggio, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
18
|
Phosphoproteomics reveals the BRAF-ERK1/2 axis as an important pathogenic signaling node in cartilage degeneration. Osteoarthritis Cartilage 2022; 30:1443-1454. [PMID: 36100125 DOI: 10.1016/j.joca.2022.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 08/14/2022] [Accepted: 08/15/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) causes gradual cellular alterations, structural anomalies and joint dysfunction. Progressive decline of chondrocyte function plays a vital role on OA pathogenesis. Although protein phosphorylation controls cartilage metabolism, its regulation mechanism in OA remains unclear. Thus, proteomic methods were used to investigate phosphorylation changes in preserved and OA articular cartilage samples, and to explore the intervention targets of phosphorylated kinase. METHODS Preserved (control) and lesioned (OA) cartilage samples from OA cases were assessed by phosphoproteomics. Immobilized metal affinity chromatography was performed for phosphopeptide enrichment. Quantitated phosphosites were comparatively assessed in the cartilage sample pair. Kinase-substrate enrichment analyses were carried out for identifying OA-related kinases. BRAF expression in cartilage tissues was assessed by immunohistochemical staining. The effects of BRAF inhibitor on cartilage degeneration were examined in mouse chondrocytes and OA mouse model. RESULTS High-sensitivity mass spectrometry-based proteomics revealed 7,471 peptides and 4,375 phosphorylated peptides differing between preserved and lesioned cartilage samples, which represented the significant alteration of kinase hubs and transduction pathways. Phosphoproteomics identified BRAF may be involved in developing OA. BRAF regulated the downstream ERK signaling pathway. In addition, BRAF was upregulated in human OA cartilage as shown by immunohistochemistry. Remarkably, BRAF inhibition alleviated cartilage degradation in a mouse model of OA through its downstream of ERK pathway activation. CONCLUSIONS Jointly, these findings provide an overview of phosphoproteomic alterations occurring during cartilage degeneration, identifying the BRAF-ERK1/2 Axis signaling as a potential signaling pathway involved in OA.
Collapse
|
19
|
Yu X, Xu X, Dong W, Yang C, Luo Y, He Y, Jiang C, Wu Y, Wang J. DDIT3/CHOP mediates the inhibitory effect of ER stress on chondrocyte differentiation by AMPKα-SIRT1 pathway. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119265. [PMID: 35381294 DOI: 10.1016/j.bbamcr.2022.119265] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/26/2022] [Accepted: 03/30/2022] [Indexed: 06/14/2023]
Abstract
Endoplasmic reticulum (ER) stress is an evolutionarily conserved cellular stress response related to multiple diseases, including temporomandibular joint (TMJ) cartilage-related diseases. Recent studies have indicated that DDIT3/CHOP (a downstream transcription factor of ER stress) is an important effector in mediating ER stress to inhibit chondrogenesis. However, the underlying mechanism by which DDIT3 regulates chondrogenesis remains unclear. In this study, tunicamycin (an ER stress agonist)-induced ER stress inhibited chondrocyte differentiation and matrix synthesis in vitro and led to an osteoarthritis-like phenotype in mouse TMJ cartilage. Meanwhile, DDIT3 expression in chondrocytes was robustly upregulated. Loss-of-function experiments validated the inhibiting effect of DDIT3 on chondrocyte differentiation and matrix synthesis. Mechanistically, the inhibiting effect was attributed to the direct and indirect regulatory effect of DDIT3 on SIRT1 (sirtuin1, silent mating type information regulation protein type 1, a member of NAD+ dependent class III histone deacetylases). On one hand, DDIT3 directly promoted the transcription of SIRT1. On the other hand, DDIT3 indirectly increased the expression of SIRT1 by promoting AMPKα phosphorylation and activation. Furthermore, activation of AMPKα or SIRT1 with the corresponding agonist AICAR or resveratrol in the DDIT3-knockdown cells partially restored the inhibiting effect of DDIT3 on chondrocyte differentiation and matrix synthesis. Collectively, these novel findings indicate that DDIT3 regulates the inhibitory effect of ER stress on chondrocyte differentiation and matrix synthesis partially via the AMPKα-SIRT1 pathway. A thorough understanding of ER stress in regulating chondrocyte homeostasis and its role in the onset of osteoarthritis may be promising to develop therapeutic targets and prevent condyle cartilage destruction.
Collapse
Affiliation(s)
- Xijie Yu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Xiaoxiao Xu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Wei Dong
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Chang Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Yao Luo
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Ying He
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Chenxi Jiang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China; Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Yanru Wu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Jiawei Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China.
| |
Collapse
|
20
|
Yan J, Wang J, He JC, Zhong Y. Sirtuin 1 in Chronic Kidney Disease and Therapeutic Potential of Targeting Sirtuin 1. Front Endocrinol (Lausanne) 2022; 13:917773. [PMID: 35795148 PMCID: PMC9251114 DOI: 10.3389/fendo.2022.917773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/12/2022] [Indexed: 11/13/2022] Open
Abstract
The incidence and prevalence of chronic kidney disease (CKD) continue to increase worldwide remaining as a major public health burden. CKD eventually progresses to end-stage kidney failure and patients with CKD have high morbidity and mortality. Sirtuin 1 (SIRT1), a NAD+-dependent deacetylases, has significant renal protective effects through its regulation of fibrosis, apoptosis, and senescence, oxidative stress, inflammation and aging process. The renal protective effects of Sirt1 have been described in many kidney diseases such as diabetic kidney disease and HIV-related kidney disease. SIRT1 also has protective effects against vascular calcification and therefore could be developed as a therapy for both CKD and CKD complications. In this narrative review, we will give an overview of the recent progress on the role of SIRT1 and its downstream pathways in CKD. We will also discuss potential therapeutic approach by activating SIRT1-related pathway in patients with CKD. The purpose is to hope to provide some insights on the future direction of the research in the field of SIRT1 for CKD.
Collapse
Affiliation(s)
- Jiayi Yan
- Division of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jue Wang
- Division of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - John Cijiang He
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Yifei Zhong
- Division of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
21
|
Zhang Y, Liu T, Yang H, He F, Zhu X. Melatonin: A novel candidate for the treatment of osteoarthritis. Ageing Res Rev 2022; 78:101635. [PMID: 35483626 DOI: 10.1016/j.arr.2022.101635] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 04/14/2022] [Accepted: 04/21/2022] [Indexed: 12/30/2022]
Abstract
Osteoarthritis (OA), characterized by cartilage erosion, synovium inflammation, and subchondral bone remodeling, is a common joint degenerative disease worldwide. OA pathogenesis is regulated by multiple predisposing factors, including imbalanced matrix metabolism, aberrant inflammatory response, and excessive oxidative stress. Moreover, melatonin has been implicated in development of several degenerative disorders owing to its potent biological functions. With regards to OA, melatonin reportedly promotes synthesis of cartilage matrix, inhibition of chondrocyte apoptosis, attenuation of inflammatory response, and suppression of matrix degradation by regulating the TGF-β, MAPK, or NF-κB signaling pathways. Notably, melatonin has been associated with amelioration of oxidative damage by restoring the OA-impaired intracellular antioxidant defense system in articular cartilage. Findings from preliminary application of melatonin or melatonin-loaded biomaterials in animal models have affirmed its potential anti-arthritic effects. Herein, we summarize the anti-arthritic effects of melatonin on OA cartilage and demonstrate that melatonin has potential therapeutic efficacy in treating OA.
Collapse
Affiliation(s)
- Yijian Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Orthopaedic Institute, Medical College, Soochow University, Suzhou 215007, China.
| | - Tao Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
| | - Huilin Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Orthopaedic Institute, Medical College, Soochow University, Suzhou 215007, China.
| | - Fan He
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Orthopaedic Institute, Medical College, Soochow University, Suzhou 215007, China.
| | - Xuesong Zhu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Orthopaedic Institute, Medical College, Soochow University, Suzhou 215007, China.
| |
Collapse
|
22
|
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: 7.3] [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
|
23
|
Smith CA, Humphreys PA, Bates N, Naven MA, Cain SA, Dvir‐Ginzberg M, Kimber SJ. SIRT1 activity orchestrates ECM expression during hESC-chondrogenic differentiation. FASEB J 2022; 36:e22314. [PMID: 35416346 PMCID: PMC9322318 DOI: 10.1096/fj.202200169r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 11/11/2022]
Abstract
Epigenetic modification is a key driver of differentiation, and the deacetylase Sirtuin1 (SIRT1) is an established regulator of cell function, ageing, and articular cartilage homeostasis. Here we investigate the role of SIRT1 during development of chondrocytes by using human embryonic stem cells (hESCs). HESC-chondroprogenitors were treated with SIRT1 activator; SRT1720, or inhibitor; EX527, during differentiation. Activation of SIRT1 early in 3D-pellet culture led to significant increases in the expression of ECM genes for type-II collagen (COL2A1) and aggrecan (ACAN), and chondrogenic transcription factors SOX5 and ARID5B, with SOX5 ChIP analysis demonstrating enrichment on the chondrocyte specific -10 (A1) enhancer of ACAN. Unexpectedly, when SIRT1 was activated, while ACAN was enhanced, glycosaminoglycans (GAGs) were reduced, paralleled by down regulation of gene expression for N-acetylgalactosaminyltransferase type 1 (GALNT1) responsible for GAG chain initiation/elongation. A positive correlation between ARID5B and COL2A1 was observed, and co-IP assays indicated association of ARID5B with SIRT1, further suggesting that COL2A1 expression is promoted by an ARID5B-SIRT1 interaction. In conclusion, SIRT1 activation positively impacts on the expression of the main ECM proteins, while altering ECM composition and suppressing GAG content during human cartilage development. These results suggest that SIRT1 activity has a differential effect on GAGs and proteins in developing hESC-chondrocytes and could only be beneficial to cartilage development and matrix protein synthesis if balanced by addition of positive GAG mediators.
Collapse
Affiliation(s)
- Christopher A. Smith
- Division of Cell Matrix Biology and Regenerative MedicineSchool of Biological SciencesUniversity of ManchesterManchesterUK
| | - Paul A. Humphreys
- Division of Cell Matrix Biology and Regenerative MedicineSchool of Biological SciencesUniversity of ManchesterManchesterUK
| | - Nicola Bates
- Division of Cell Matrix Biology and Regenerative MedicineSchool of Biological SciencesUniversity of ManchesterManchesterUK
| | - Mark A. Naven
- Division of Cell Matrix Biology and Regenerative MedicineSchool of Biological SciencesUniversity of ManchesterManchesterUK
| | - Stuart A. Cain
- Division of Cell Matrix Biology and Regenerative MedicineSchool of Biological SciencesUniversity of ManchesterManchesterUK
| | - Mona Dvir‐Ginzberg
- Laboratory of Cartilage BiologyFaculty of Dental MedicineHebrew University of JerusalemJerusalemIsrael
| | - Susan J. Kimber
- Division of Cell Matrix Biology and Regenerative MedicineSchool of Biological SciencesUniversity of ManchesterManchesterUK
| |
Collapse
|
24
|
Huang X, Chen C, Chen Y, Xu J, Liu L. Omentin-1 alleviate interleukin-1β(IL-1β)-induced nucleus pulposus cells senescence. Bioengineered 2022; 13:13849-13859. [PMID: 35707832 PMCID: PMC9275897 DOI: 10.1080/21655979.2022.2084495] [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] [Indexed: 11/25/2022] Open
Abstract
One of the main causes of low back pain (LBP) and degenerative musculoskeletal disorders is intervertebral disc degeneration (IVDD). Inflammation-associated senescence of Human nucleus pulposus cells (HNPCs) plays an essential function in the disease progression of IVDD. Omentin-1 is an adipokine that has been recently reported to have anti-inflammatory potential. In our research, IL-1β was used to simulate the inflammatory environment in the IVDD. We investigated in vitro the effects of Omentin-1 on HNPCs, including the components of senescence, cell cycle and extracellular matrix (ECM) synthesis. The results showed that the addition of Omentin-1 improved IL-1β-induced senescence in HNPCs. G1 phase cell cycle arrest and reduced ECM synthesis in HNPCs. Furthermore, we demonstrated that the effect of Omentin-1 in reducing senescence of HNPCs is dependent on SIRT1. These findings suggest that Omentin-1 plays an important function in protecting HNPCs against senescence and has the potential for IVDD gene target therapy.
Collapse
Affiliation(s)
- Xin Huang
- Department of Orthopaedic Surgery, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Changhong Chen
- Department of Orthopaedic Surgery, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Yaofei Chen
- Department of Orthopaedic Surgery, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Jun Xu
- Department of Orthopaedic Surgery, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Lin Liu
- Department of Orthopaedic Surgery, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| |
Collapse
|
25
|
Shen P, Deng X, Chen Z, Ba X, Qin K, Huang Y, Huang Y, Li T, Yan J, Tu S. SIRT1: A Potential Therapeutic Target in Autoimmune Diseases. Front Immunol 2021; 12:779177. [PMID: 34887866 PMCID: PMC8650132 DOI: 10.3389/fimmu.2021.779177] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/08/2021] [Indexed: 12/21/2022] Open
Abstract
The morbidity and mortality of autoimmune diseases (Ads) have been increasing worldwide, and the identification of novel therapeutic strategies for prevention and treatment is urgently needed. Sirtuin 1 (SIRT1), a member of the class III family of nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylases, has been reported to participate in the progression of several diseases. SIRT1 also regulates inflammation, oxidative stress, mitochondrial function, immune responses, cellular differentiation, proliferation and metabolism, and its altered functions are likely involved in Ads. Several inhibitors and activators have been shown to affect the development of Ads. SIRT1 may represent a novel therapeutic target in these diseases, and small molecules or natural products that modulate the functions of SIRT1 are potential therapeutic agents. In the present review, we summarize current studies of the biological functions of SIRT1 and its role in the pathogenesis and treatment of Ads.
Collapse
Affiliation(s)
- Pan Shen
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Xuan Deng
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhe Chen
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Xin Ba
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Kai Qin
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Ying Huang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Yao Huang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Li
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Jiahui Yan
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Shenghao Tu
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
26
|
Pharmaceutical therapeutics for articular regeneration and restoration: state-of-the-art technology for screening small molecular drugs. Cell Mol Life Sci 2021; 78:8127-8155. [PMID: 34783870 PMCID: PMC8593173 DOI: 10.1007/s00018-021-03983-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 09/20/2021] [Accepted: 10/14/2021] [Indexed: 02/07/2023]
Abstract
Articular cartilage damage caused by sports injury or osteoarthritis (OA) has gained increased attention as a worldwide health burden. Pharmaceutical treatments are considered cost-effective means of promoting cartilage regeneration, but are limited by their inability to generate sufficient functional chondrocytes and modify disease progression. Small molecular chemical compounds are an abundant source of new pharmaceutical therapeutics for cartilage regeneration, as they have advantages in design, fabrication, and application, and, when used in combination, act as powerful tools for manipulating cellular fate. In this review, we present current achievements in the development of small molecular drugs for cartilage regeneration, particularly in the fields of chondrocyte generation and reversion of chondrocyte degenerative phenotypes. Several clinically or preclinically available small molecules, which have been shown to facilitate chondrogenesis, chondrocyte dedifferentiation, and cellular reprogramming, and subsequently ameliorate cartilage degeneration by targeting inflammation, matrix degradation, metabolism, and epigenetics, are summarized. Notably, this review introduces essential parameters for high-throughput screening strategies, including models of different chondrogenic cell sources, phenotype readout methodologies, and transferable advanced systems from other fields. Overall, this review provides new insights into future pharmaceutical therapies for cartilage regeneration.
Collapse
|
27
|
Ren Z, Raut NA, Lawal TO, Patel SR, Lee SM, Mahady GB. Peonidin-3-O-glucoside and cyanidin increase osteoblast differentiation and reduce RANKL-induced bone resorption in transgenic medaka. Phytother Res 2021; 35:6255-6269. [PMID: 34704297 DOI: 10.1002/ptr.7271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/23/2020] [Accepted: 01/23/2021] [Indexed: 11/07/2022]
Abstract
Experimental and clinical studies suggest a positive impact of anthocyanins on bone health; however, the mechanisms of anthocyanins altering the differentiation and function of osteoblasts and osteoclasts are not fully understood. This work demonstrates that dietary anthocyanins and resveratrol increased proliferation of cultured human hFOB 1.19 osteoblasts. In addition, treatment of serum starvation of hFOB osteoblasts with anthocyanins and resveratrol at 1.0 μg/ml reduced apoptosis, the Bax/Bcl-2 ratio, p53, and HDAC1 expression, but increased SIRT1/3 and PGC1α mRNA expression, suggesting mitochondrial and epigenetic regulation. In Sp7/osterix:mCherry transgenic medaka, peonidin-3-O-glucoside and resveratrol increased osteoblast differentiation and increased the expression of Sp7/osterix. Cyanidin, peonidin-3-O-glucoside, and resveratrol also reduced RANKL-induced ectopic osteoclast formation and bone resorption in col10α1:nlGFP/rankl:HSE:CFP medaka in doses of 1-4 μg/ml. The results indicate that both cyanidin and peonidin-3-O-glucoside have anabolic effects on bone, increasing osteoblast proliferation and differentiation, mitochondrial biogenesis, and by altering the osteoblast epigenome. Cyanidin and peonidin-3-O-glucoside also reduced RANKL-induced bone resorption in a transgenic medaka model of bone resorption. Thus, peonidin-3-O-glucoside and cyanidin appear to both increase bone formation and reduce bone loss, suggesting that they be further investigated as potential treatments for osteoporosis and osteomalacia.
Collapse
Affiliation(s)
- Zhitao Ren
- Department of Pharmacy Practice, College of Pharmacy, WHO Collaborating Centre for Traditional Medicine, University of Illinois at Chicago, Chicago, Illinois, USA.,State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Nishikant A Raut
- Raman Fellow, Department of Pharmacy Practice, College of Pharmacy, WHO Collaborating Centre for Traditional Medicine, University of Illinois at Chicago, Chicago, Illinois, USA.,Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, India
| | - Temitope O Lawal
- Schlumberger Fellow, Department of Pharmacy Practice, College of Pharmacy, WHO Collaborating Centre for Traditional Medicine, University of Illinois at Chicago, Chicago, Illinois, USA.,Department of Pharmaceutical Microbiology, University of Ibadan, Ibadan, Nigeria
| | - Shital R Patel
- Department of Pharmacy Practice, College of Pharmacy, WHO Collaborating Centre for Traditional Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Simon M Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Gail B Mahady
- Department of Pharmacy Practice, College of Pharmacy, WHO Collaborating Centre for Traditional Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
28
|
Somemura S, Kumai T, Yatabe K, Sasaki C, Fujiya H, Niki H, Yudoh K. Physiologic Mechanical Stress Directly Induces Bone Formation by Activating Glucose Transporter 1 (Glut 1) in Osteoblasts, Inducing Signaling via NAD+-Dependent Deacetylase (Sirtuin 1) and Runt-Related Transcription Factor 2 (Runx2). Int J Mol Sci 2021; 22:9070. [PMID: 34445787 PMCID: PMC8396442 DOI: 10.3390/ijms22169070] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 12/23/2022] Open
Abstract
Mechanical stress is an important factor affecting bone tissue homeostasis. We focused on the interactions among mechanical stress, glucose uptake via glucose transporter 1 (Glut1), and the cellular energy sensor sirtuin 1 (SIRT1) in osteoblast energy metabolism, since it has been recognized that SIRT1, an NAD+-dependent deacetylase, may function as a master regulator of the mechanical stress response as well as of cellular energy metabolism (glucose metabolism). In addition, it has already been demonstrated that SIRT1 regulates the activity of the osteogenic transcription factor runt-related transcription factor 2 (Runx2). The effects of mechanical loading on cellular activities and the expressions of Glut1, SIRT1, and Runx2 were evaluated in osteoblasts and chondrocytes in a 3D cell-collagen sponge construct. Compressive mechanical loading increased osteoblast activity. Mechanical loading also significantly increased the expression of Glut1, significantly decreased the expression of SIRT1, and significantly increased the expression of Runx2 in osteoblasts in comparison with non-loaded osteoblasts. Incubation with a Glut1 inhibitor blocked mechanical stress-induced changes in SIRT1 and Runx2 in osteoblasts. In contrast with osteoblasts, the expressions of Glut1, SIRT1, and Runx2 in chondrocytes were not affected by loading. Our present study indicated that mechanical stress induced the upregulation of Glut1 following the downregulation of SIRT1 and the upregulation of Runx2 in osteoblasts but not in chondrocytes. Since SIRT1 is known to negatively regulate Runx2 activity, a mechanical stress-induced downregulation of SIRT1 may lead to the upregulation of Runx2, resulting in osteoblast differentiation. Incubation with a Glut1 inhibitor the blocked mechanical stress-induced downregulation of SIRT1 following the upregulation of Runx2, suggesting that Glut1 is necessary to mediate the responses of SIRT1 and Runx2 to mechanical loading in osteoblasts.
Collapse
Affiliation(s)
- Shu Somemura
- Department of Sports Medicine, St. Marianna University School of Medicine, Sugao 2-16-1, Miyamae-ku, Kawasaki 216-8511, Japan; (S.S.); (K.Y.); (H.F.)
- Department of Orthopaedic Surgery, St. Marianna University School of Medicine, Sugao 2-16-1, Miyamae-ku, Kawasaki 216-8512, Japan; (T.K.); (H.N.)
| | - Takanori Kumai
- Department of Orthopaedic Surgery, St. Marianna University School of Medicine, Sugao 2-16-1, Miyamae-ku, Kawasaki 216-8512, Japan; (T.K.); (H.N.)
| | - Kanaka Yatabe
- Department of Sports Medicine, St. Marianna University School of Medicine, Sugao 2-16-1, Miyamae-ku, Kawasaki 216-8511, Japan; (S.S.); (K.Y.); (H.F.)
| | - Chizuko Sasaki
- Institute for Ultrastructural Morphology, St. Marianna University Graduate School of Medicine, Sugao 2-16-1, Miyamae-ku, Kawasaki 216-8512, Japan;
| | - Hiroto Fujiya
- Department of Sports Medicine, St. Marianna University School of Medicine, Sugao 2-16-1, Miyamae-ku, Kawasaki 216-8511, Japan; (S.S.); (K.Y.); (H.F.)
| | - Hisateru Niki
- Department of Orthopaedic Surgery, St. Marianna University School of Medicine, Sugao 2-16-1, Miyamae-ku, Kawasaki 216-8512, Japan; (T.K.); (H.N.)
| | - Kazuo Yudoh
- Department of Frontier Medicine, Institute of Medical Science, St. Marianna University School of Medicine, Sugao 2-16-1, Miyamae-ku, Kawasaki 216-8512, Japan
| |
Collapse
|
29
|
Buhrmann C, Brockmueller A, Mueller AL, Shayan P, Shakibaei M. Curcumin Attenuates Environment-Derived Osteoarthritis by Sox9/NF-kB Signaling Axis. Int J Mol Sci 2021; 22:ijms22147645. [PMID: 34299264 PMCID: PMC8306025 DOI: 10.3390/ijms22147645] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 01/11/2023] Open
Abstract
Inflammation has a fundamental impact on the pathophysiology of osteoarthritis (OA), a common form of degenerative arthritis. It has previously been established that curcumin, a component of turmeric (Curcuma longa), has anti-inflammatory properties. This research evaluates the potentials of curcumin on the pathophysiology of OA in vitro. To explore the anti-inflammatory efficacy of curcumin in an inflamed joint, an osteoarthritic environment (OA-EN) model consisting of fibroblasts, T-lymphocytes, 3D-chondrocytes is constructed and co-incubated with TNF-α, antisense oligonucleotides targeting NF-kB (ASO-NF-kB), or an IkB-kinase (IKK) inhibitor (BMS-345541). Our results show that OA-EN, similar to TNF-α, suppresses chondrocyte viability, which is accompanied by a significant decrease in cartilage-specific proteins (collagen II, CSPG, Sox9) and an increase in NF-kB-driven gene proteins participating in inflammation, apoptosis, and breakdown (NF-kB, MMP-9, Cox-2, Caspase-3). Conversely, similar to knockdown of NF-kB at the mRNA level or at the IKK level, curcumin suppresses NF-kB activation, NF-kB-promotes gene proteins derived from the OA-EN, and stimulates collagen II, CSPG, and Sox9 expression. Furthermore, co-immunoprecipitation assay shows that curcumin reduces OA-EN-mediated inflammation and chondrocyte apoptosis, with concomitant chondroprotective effects, due to modulation of Sox-9/NF-kB signaling axis. Finally, curcumin selectively hinders the interaction of p-NF-kB-p65 directly with DNA—this association is disrupted through DTT. These results suggest that curcumin suppresses inflammation in OA-EN via modulating NF-kB-Sox9 coupling and is essential for maintaining homeostasis in OA by balancing chondrocyte survival and inflammatory responses. This may contribute to the alternative treatment of OA with respect to the efficacy of curcumin.
Collapse
Affiliation(s)
- Constanze Buhrmann
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, Pettenkoferstr. 11, D-80336 Munich, Germany; (C.B.); (A.B.); (A.-L.M.)
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Augsburg, Universitaetsstr. 2, D-86159 Augsburg, Germany
| | - Aranka Brockmueller
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, Pettenkoferstr. 11, D-80336 Munich, Germany; (C.B.); (A.B.); (A.-L.M.)
| | - Anna-Lena Mueller
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, Pettenkoferstr. 11, D-80336 Munich, Germany; (C.B.); (A.B.); (A.-L.M.)
| | - Parviz Shayan
- Department of Parasitology, Faculty of Veterinary Medicine, University of Tehran, Tehran 141556453, Iran;
| | - Mehdi Shakibaei
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, Pettenkoferstr. 11, D-80336 Munich, Germany; (C.B.); (A.B.); (A.-L.M.)
- Correspondence: ; Tel.: +49-89-2180-72624
| |
Collapse
|
30
|
Liu X, Chen A, Liang Q, Yang X, Dong Q, Fu M, Wang S, Li Y, Ye Y, Lan Z, Chen Y, Ou J, Yang P, Lu L, Yan J. Spermidine inhibits vascular calcification in chronic kidney disease through modulation of SIRT1 signaling pathway. Aging Cell 2021; 20:e13377. [PMID: 33969611 PMCID: PMC8208796 DOI: 10.1111/acel.13377] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 03/31/2021] [Accepted: 04/18/2021] [Indexed: 12/20/2022] Open
Abstract
Vascular calcification is a common pathologic condition in patients with chronic kidney disease (CKD) and aging individuals. It has been established that vascular calcification is a gene‐regulated biological process resembling osteogenesis involving osteogenic differentiation. However, there is no efficient treatment available for vascular calcification so far. The natural polyamine spermidine has been demonstrated to increase life span and protect against cardiovascular disease. It is unclear whether spermidine supplementation inhibits vascular calcification in CKD. Alizarin red staining and quantification of calcium content showed that spermidine treatment markedly reduced mineral deposition in both rat and human vascular smooth muscle cells (VSMCs) under osteogenic conditions. Additionally, western blot analysis revealed that spermidine treatment inhibited osteogenic differentiation of rat and human VSMCs. Moreover, spermidine treatment remarkably attenuated calcification of rat and human arterial rings ex vivo and aortic calcification in rats with CKD. Furthermore, treatment with spermidine induced the upregulation of Sirtuin 1 (SIRT1) in VSMCs and resulted in the downregulation of endoplasmic reticulum (ER) stress signaling components, such as activating transcription factor 4 (ATF4) and CCAAT/enhancer‐binding protein homologous protein (CHOP). Both pharmacological inhibition of SIRT1 by SIRT1 inhibitor EX527 and knockdown of SIRT1 by siRNA markedly blocked the inhibitory effect of spermidine on VSMC calcification. Consistently, EX527 abrogated the inhibitory effect of spermidine on aortic calcification in CKD rats. We for the first time demonstrate that spermidine alleviates vascular calcification in CKD by upregulating SIRT1 and inhibiting ER stress, and this may develop a promising therapeutic treatment to ameliorate vascular calcification in CKD.
Collapse
Affiliation(s)
- Xiaoyu Liu
- Department of Cardiology Laboratory of Heart Center Heart Center Zhujiang Hospital Southern Medical University Guangzhou China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease Guangzhou China
- Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure Guangzhou China
| | - An Chen
- Department of Cardiology Laboratory of Heart Center Heart Center Zhujiang Hospital Southern Medical University Guangzhou China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease Guangzhou China
- Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure Guangzhou China
| | - Qingchun Liang
- Department of Anesthesiology The Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Xiulin Yang
- Department of Cardiology Laboratory of Heart Center Heart Center Zhujiang Hospital Southern Medical University Guangzhou China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease Guangzhou China
- Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure Guangzhou China
| | - Qianqian Dong
- Department of Cardiology Laboratory of Heart Center Heart Center Zhujiang Hospital Southern Medical University Guangzhou China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease Guangzhou China
- Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure Guangzhou China
| | - Mingwei Fu
- Department of Cardiology Laboratory of Heart Center Heart Center Zhujiang Hospital Southern Medical University Guangzhou China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease Guangzhou China
- Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure Guangzhou China
| | - Siyi Wang
- Department of Cardiology Laboratory of Heart Center Heart Center Zhujiang Hospital Southern Medical University Guangzhou China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease Guangzhou China
- Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure Guangzhou China
| | - Yining Li
- Department of Cardiology Laboratory of Heart Center Heart Center Zhujiang Hospital Southern Medical University Guangzhou China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease Guangzhou China
- Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure Guangzhou China
| | - Yuanzhi Ye
- Department of Cardiology Laboratory of Heart Center Heart Center Zhujiang Hospital Southern Medical University Guangzhou China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease Guangzhou China
- Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure Guangzhou China
| | - Zirong Lan
- Department of Cardiology Laboratory of Heart Center Heart Center Zhujiang Hospital Southern Medical University Guangzhou China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease Guangzhou China
- Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure Guangzhou China
| | - Yanting Chen
- Department of Pathophysiolgy Zhongshan School of Medicine Sun Yat‐Sen University Guangzhou China
| | - Jing‐Song Ou
- Division of Cardiac Surgery The First Affiliated Hospital Sun Yat‐Sen University Guangzhou China
| | - Pingzhen Yang
- Department of Cardiology Laboratory of Heart Center Heart Center Zhujiang Hospital Southern Medical University Guangzhou China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease Guangzhou China
- Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure Guangzhou China
| | - Lihe Lu
- Department of Pathophysiolgy Zhongshan School of Medicine Sun Yat‐Sen University Guangzhou China
| | - Jianyun Yan
- Department of Cardiology Laboratory of Heart Center Heart Center Zhujiang Hospital Southern Medical University Guangzhou China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease Guangzhou China
- Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure Guangzhou China
| |
Collapse
|
31
|
Chae DS, Han S, Lee MK, Kim SW. Genome Edited Sirt1-Overexpressing Human Mesenchymal Stem Cells Exhibit Therapeutic Effects in Treating Collagen-Induced Arthritis. Mol Cells 2021; 44:245-253. [PMID: 33935044 PMCID: PMC8112166 DOI: 10.14348/molcells.2021.0037] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/18/2021] [Accepted: 04/01/2021] [Indexed: 12/15/2022] Open
Abstract
Even though mesenchymal stem cells (MSCs) are known for cartilage regeneration, their therapeutic efficacy needs to be enhanced. In the present study, we produced genome-edited silent information regulator 2 type 1 (Sirt1)-overexpressing MSCs, and evaluated their therapeutic potential in a damaged cartilage mouse liver fibrosis model. The Sirt1 gene was successfully inserted into a 'safe harbor' genomic locus in amniotic mesenchymal stem cells (AMMs), and the chondrogenic properties of the Sirt1 gene overexpressing AMMs (AMM/S) were characterized using quantitative PCR and histology. Therapeutic potentials were investigated in a collagen-induced arthritis (CIA) mouse model. Chondrocyte-differentiated AMM/S expressed cartilage-specific genes and were positive for Safranin O staining. Transplantation of AMM/S attenuated CIA progression and suppressed T helper (Th)-17 cell activation while increasing the Treg cell population in CIA mice. Pro-inflammatory factors, such as interleukin (IL)-1β, IL-6, monocyte chemoattractant protein (MCP)-1, and tumor necrosis factor (TNF)-α were significantly decreased in AMM/S-injected joint tissues. In conclusion, genome-edited AMM/S may represent a safe and alternative therapeutic option for the treatment and repair of damaged cartilage, or in inflammatory joint arthritis.
Collapse
Affiliation(s)
- Dong-Sik Chae
- Department of Orthopedic Surgery, International St. Mary’s Hospital, Catholic Kwandong University College of Medicine, Incheon 22711, Korea
| | - Seongho Han
- Department of Family Medicine, Dong-A University Medical Center, Dong-A University College of Medicine, Busan 49201, Korea
| | - Min-Kyung Lee
- Department of Dental Hygine, Dong-Eui Universtigy, Busan 47340, Korea
| | - Sung-Whan Kim
- Department Medicine, Catholic Kwandong University College of Medicine, Gangneung 25601, Korea
| |
Collapse
|
32
|
Zhou Z, Deng Z, Liu Y, Zheng Y, Yang S, Lu W, Xiao D, Zhu W. Protective Effect of SIRT1 Activator on the Knee With Osteoarthritis. Front Physiol 2021; 12:661852. [PMID: 33927645 PMCID: PMC8076744 DOI: 10.3389/fphys.2021.661852] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/24/2021] [Indexed: 11/16/2022] Open
Abstract
Osteoarthritis (OA), one of the most common chronic musculoskeletal disorders, is deemed to be correlated with aging. The SIRT1 activator, resveratrol, acts as a crucial regulator of aging and may have a potential therapeutic effect on OA. Rabbit OA models were established through destabilized medial meniscus surgery. A total of 40 healthy male New Zealand rabbits were divided into five groups: control group (sham operation), OA group, as well as low dose (LD), middle dose (MD), and high dose (HD) resveratrol-treated OA groups. 6 weeks after operation, 0.8 ml of normal saline was injected into the knee joints every other day in the control and OA groups, and 0.8 ml of 5, 10, and 15 μmol/L resveratrol was injected into the knee joints every other day in the LD, MD, and HD group, respectively. The rabbits were sacrificed 2 weeks after medication, and the articular cartilage of the knee joint was collected for Micro-CT, histology and Western blot analysis. Obvious articular cartilage lesion and joint space narrowing were detected in the OA group. Compared with the OA group, less osteoarthritic changes were observed in the MD and HD groups. The MD and HD groups had significantly lower bone volume fraction, trabecular number and Mankin scores than the LD and OA groups (p < 0.05). No significant difference was found between the OA and LD groups (p > 0.05). The expressions of SIRT1 and p53 detected by western blot were consistent with the aforementioned findings. Therefore, resveratrol can activate the SIRT1 gene to play a protective role in the OA process by inhibiting chondrocyte apoptosis, trabecular bone number increasing of the subchondral bone, as well as elevation of bone density. It demonstrated the importance of SIRT1 in maintaining articular cartilage health and provided a promising therapeutic intervention in the treatment of OA.
Collapse
Affiliation(s)
- Zhenquan Zhou
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China.,Department of Orthopaedics, Shenzhen Hospital of Southern Medical University, Shenzhen, China.,Clinical Medical College, Guangzhou Medical University, Guangzhou, China
| | - Zhenhan Deng
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China.,Clinical Medical College, Guangzhou Medical University, Guangzhou, China.,Clinical Medical College, Shenzhen University, Shenzhen, China.,Clinical Medical College, Guangxi University of Chinese Medicine, Nanning, China.,Clinical Medical College, Anhui Medical University, Hefei, China
| | - Yuwei Liu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Yizi Zheng
- Department of Thyroid and Breast Surgery, Shenzhen Breast Tumor Research Center for Diagnosis and Treatment, National Standardization Center for Breast Cancer Diagnosis and Treatment, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Shiwei Yang
- Clinical Medical College, Anhui Medical University, Hefei, China.,Teaching Office, Shenzhen Second People's Hospital, Shenzhen, China
| | - Wei Lu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Deming Xiao
- Clinical Medical College, Guangzhou Medical University, Guangzhou, China
| | - Weimin Zhu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China.,Clinical Medical College, Guangzhou Medical University, Guangzhou, China.,Clinical Medical College, Shenzhen University, Shenzhen, China.,Clinical Medical College, Guangxi University of Chinese Medicine, Nanning, China.,Clinical Medical College, Anhui Medical University, Hefei, China
| |
Collapse
|
33
|
Zhang M, Mou L, Liu S, Sun F, Gong M. Circ_0001103 alleviates IL-1β-induced chondrocyte cell injuries by upregulating SIRT1 via targeting miR-375. Clin Immunol 2021; 227:108718. [PMID: 33819576 DOI: 10.1016/j.clim.2021.108718] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Osteoarthritis (OA) is a common inflammatory disease characterized by articular cartilage degeneration and injury. Circular RNAs (circRNAs) are widely involved in the development of human diseases, including OA. The objective of this study was to investigate the function and functional mechanism of circ_0001103 in OA. METHODS Cell model of OA was established by treating chondrocytes with interleukin-1β (IL-1β). The expression of circ_0001103, miR-375 and sirtuin 1 (SIRT1) mRNA was measured using quantitative real-time polymerase chain reaction (qRT-PCR). Cell viability was assessed using cell counting kit-8 (CCK-8) assay. Cell apoptosis was determined using flow cytometry assay. The expression levels of inflammatory factors were quantified by qRT-PCR. The expression of extracellular matrix (ECM) metabolism-related markers, including Collagen Type II Alpha 1 Chain (COL2A1) and A disintegrin and metalloproteinase with thrombospondin motifs 4 (ADAMTS4), was detected by western blot. Predicted target relationship between miR-375 and circ_0001103 or SIRT1 by the bioinformatics tools was validated by dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay. RESULTS Circ_0001103 was downregulated in OA tissues and IL-1β-induced chondrocytes. Overexpression of circ_0001103 attenuated IL-1β-induced chondrocyte apoptosis, inflammatory responses and ECM degradation. MiR-375 was targeted by circ_0001103, and miR-375 could bind to SIRT1. Circ_0001103 overexpression increased the expression of SIRT1 by suppressing miR-375. Rescue experiments suggested that miR-375 restoration reversed the effects of circ_0001103 overexpression, and SIRT1 knockdown overturned the effects of miR-375 inhibition. CONCLUSION Circ_0001103 governed the miR-375/SIRT1 axis to ameliorate IL-1β-induced chondrocyte injuries, implying that circ_0001103 was a promising biomarker in OA pathogenesis.
Collapse
Affiliation(s)
- Miao Zhang
- Department of Trauma and Orthopaedics, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan City, China; Department of Joint Surgery, Weifang People's Hospital, Weifang City, China
| | - Leming Mou
- Department of Orthopedics, Weifang People's Hospital, Weifang City, China
| | - Shiwei Liu
- Department of Joint Surgery, Weifang People's Hospital, Weifang City, China
| | - Fengyi Sun
- Department of Gynaecology and Obstetrics, Weifang People's Hospital, Weifang City, China
| | - Mingzhi Gong
- Department of Trauma and Orthopaedics, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan City, China.
| |
Collapse
|
34
|
IL-38 restrains inflammatory response of collagen-induced arthritis in rats via SIRT1/HIF-1α signaling pathway. Biosci Rep 2021; 40:223089. [PMID: 32347300 PMCID: PMC7256678 DOI: 10.1042/bsr20182431] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 04/26/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE To observe the restraining effect of IL-38 on inflammatory response in collagen-induced arthritis rats (CIA), and to explore the regulatory mechanism of SIRT1/HIF-1α signaling pathway. METHODS 40 SD rats were randomly divided into Control group, CIA group, CLL group and CLH group, with 10 rats in each group; CIA rat model was established. The effects of IL-38 on arthritis index, inflammatory response, osteogenic factor and angiogenic factor were observed by methods including HE staining, ELISA, immunohistochemical and immunofluorescence. Human synoviocytes were cultured in vitro, and SIRT1 inhibitors were added to detect the expression for relating factors of SIRT1/HIF-1α signaling pathway by Western blot. RESULTS IL-38 could alleviate CIA joint damage and restrain inflammatory response, could up-regulate the expression of OPG in CIA rats and could down-regulate the expression of RANKL and RANK. IL-38 could restrain the expression of VEGF, VEGFR1, VEGFR2 and HIF. Moreover, we found that IL-38 could up-regulate the SIRT1 expression and down-regulate the HIF-1α, TLR4 and NF-KB p65 expression in CLL and CLH groups. From the treatment of synoviocytes to simulate the CIA model and the treatment of SIRT1 inhibitors, we demonstrated that the inhibitory effect of IL-38 on inflammatory factors and regulation of SIRT1/HIF-1α signaling pathway-related proteins were inhibited. CONCLUSION IL-38 can restrain the inflammatory response of CIA rats, can promote the expression of osteogenic factors, can inhibit neovascularization, and can alleviate joint damage in rats. The mechanism may be related to the regulation of SIRT1/HIF-1α signaling pathway.
Collapse
|
35
|
Govoni M, Muscari C, Bonafè F, Morselli PG, Cortesi M, Dallari D, Giordano E. A brief very-low oxygen tension regimen is sufficient for the early chondrogenic commitment of human adipose-derived mesenchymal stem cells. Adv Med Sci 2021; 66:98-104. [PMID: 33461101 DOI: 10.1016/j.advms.2020.12.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 10/19/2020] [Accepted: 12/23/2020] [Indexed: 12/30/2022]
Abstract
PURPOSE The aim of this study was to evaluate the effects exerted over chondrogenic commitment of human adipose-derived mesenchymal stem cells (ADSCs) by a very low oxygen tension (<1% pO2). MATERIALS/METHODS Cell morphology, mRNA levels of chondrocyte-specific marker genes and the involvement of p38 MAPK signalling were monitored in human ADSCs under a very low oxygen tension. RESULTS Cell morphology was significantly changed after two days of hypoxic preconditioning when they featured as elongated spindle-shaped cells. SRY-box containing gene 9, aggrecan and collagen type II mRNA levels were enhanced under severe hypoxic culture conditions. Moreover, the inhibition of p38 MAPK resulted in a substantial reduction in transcription of the above-mentioned specific genes, proving the pivotal role of this pathway in the transcriptional regulation of chondrogenesis. CONCLUSIONS Here, we propose a protocol showing the early commitment of stem cells towards the chondrogenic phenotype in only 2 days of culture via a very low hypoxic environment, in the absence of growth factors added in the culture medium.
Collapse
|
36
|
Rysz J, Franczyk B, Kujawski K, Sacewicz-Hofman I, Ciałkowska-Rysz A, Gluba-Brzózka A. Are Nutraceuticals Beneficial in Chronic Kidney Disease? Pharmaceutics 2021; 13:231. [PMID: 33562154 PMCID: PMC7915977 DOI: 10.3390/pharmaceutics13020231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/29/2021] [Accepted: 02/03/2021] [Indexed: 12/18/2022] Open
Abstract
Chronic kidney disease (CKD) is a worldwide health problem in which prevalence is constantly rising. The pathophysiology of CKD is complicated and has not been fully resolved. However, elevated oxidative stress is considered to play a vital role in the development of this disease. CKD is also thought to be an inflammatory disorder in which uremic toxins participate in the development of the inflammatory milieu. A healthy, balanced diet supports the maintenance of a good health status as it helps to reduce the risk of the development of chronic diseases, including chronic kidney disease, diabetes mellitus, and hypertension. Numerous studies have demonstrated that functional molecules and nutrients, including fatty acids and fiber as well as nutraceuticals such as curcumin, steviol glycosides, and resveratrol not only exert beneficial effects on pro-inflammatory and anti-inflammatory pathways but also on gut mucosa. Nutraceuticals have attracted great interest recently due to their potential favorable physiological effects on the human body and their safety. This review presents some nutraceuticals in which consumption could exert a beneficial impact on the development and progression of renal disease as well cardiovascular disease.
Collapse
Affiliation(s)
- Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.); (K.K.)
| | - Beata Franczyk
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.); (K.K.)
| | - Krzysztof Kujawski
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.); (K.K.)
| | | | | | - Anna Gluba-Brzózka
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.); (K.K.)
| |
Collapse
|
37
|
Komatsu DE, Duque E, Hadjiargyrou M. MicroRNAs and fracture healing: Pre-clinical studies. Bone 2021; 143:115758. [PMID: 33212318 PMCID: PMC7769985 DOI: 10.1016/j.bone.2020.115758] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/13/2020] [Accepted: 11/13/2020] [Indexed: 12/28/2022]
Abstract
During the past several years, pre-clinical experiments have established that microRNAs (miRNAs), small non-coding RNAs, serve as key regulatory molecules of fracture healing. Their easy modulation with agonists and antagonists make them highly desirable targets for future therapeutic strategies, especially for pathophysiologic fractures that either do not heal (nonunions) or are delayed. It is now well documented that these problematic fractures lead to human suffering and impairment of life quality. Additionally, financial difficulties are also encountered as work productivity decreases and income is reduced. Moreover, targeting miRNAs may also be an avenue to enhancing normal physiological fracture healing. Herein we present the most current knowledge of the involvement of miRNAs during fracture healing in pre-clinical studies. Following a brief description on the nature of miRNAs and of the fracture healing process, we present data from studies focusing specifically, on miRNA regulation of osteoblast differentiation and osteogenesis (within the context of known signaling pathways), chondrocytes, angiogenesis, and apoptosis, all critical to successful bone repair. Further, we also discuss miRNAs and exosomes. We hope that this manuscript serves as a comprehensive review that will facilitate basic/translational scientists in the orthopaedic arena to realize and further decipher the biological and future therapeutic impact of these small regulatory RNA molecules, especially as they relate to the molecular events of each of the major phases of fracture healing.
Collapse
Affiliation(s)
- David E Komatsu
- Department of Orthopaedics and Rehabilitation, Stony Brook University, United States of America
| | - Edie Duque
- Department of Orthopaedics and Rehabilitation, Stony Brook University, United States of America
| | - Michael Hadjiargyrou
- Department of Biological and Chemical Sciences, New York Institute of Technology, United States of America.
| |
Collapse
|
38
|
Chou YT, Liu TT, Yang UC, Huang CC, Liu CW, Huang SF, Li TH, Liu HM, Lin MW, Yang YY, Lee TY, Huang YH, Hou MC, Lin HC. Intestinal SIRT1 Deficiency-Related Intestinal Inflammation and Dysbiosis Aggravate TNFα-Mediated Renal Dysfunction in Cirrhotic Ascitic Mice. Int J Mol Sci 2021; 22:ijms22031233. [PMID: 33513830 PMCID: PMC7865325 DOI: 10.3390/ijms22031233] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 12/14/2022] Open
Abstract
In advanced cirrhosis, the TNFα-mediated intestinal inflammation and bacteria dysbiosis are involved in the development of inflammation and vasoconstriction-related renal dysfunction. In colitis and acute kidney injury models, activation of SIRT1 attenuates the TNFα-mediated intestinal and renal abnormalities. This study explores the impacts of intestinal SIRT1 deficiency and TNFα-mediated intestinal abnormalities on the development of cirrhosis-related renal dysfunction. Systemic and renal hemodynamics, intestinal dysbiosis [cirrhosis dysbiosis ratio (CDR) as marker of dysbiosis], and direct renal vasoconstrictive response (renal vascular resistance (RVR) and glomerular filtration rate (GFR)) to cumulative doses of TNFα were measured in bile duct ligated (BDL)-cirrhotic ascitic mice. In SIRT1IEC-KO-BDL-ascitic mice, the worsening of intestinal dysbiosis exacerbates intestinal inflammation/barrier dysfunction, the upregulation of the expressions of intestinal/renal TNFα-related pathogenic signals, higher TNFα-induced increase in RVR, and decrease in GFR in perfused kidney. In intestinal SIRT1 knockout groups, the positive correlations were identified between intestinal SIRT1 activity and CDR. Particularly, the negative correlations were identified between CDR and RVR, with the positive correlation between CDR and GFR. In mice with advanced cirrhosis, the expression of intestinal SIRT1 is involved in the linkage between intestinal dysbiosis and vasoconstriction/hypoperfusion-related renal dysfunction through the crosstalk between intestinal/renal TNFα-related pathogenic inflammatory signals.
Collapse
Affiliation(s)
- Yu-Te Chou
- Department of Medicine, Taipei Veterans General Hospital, Taipei 11267, Taiwan; (Y.-T.C.); (C.-W.L.); (Y.-H.H.); (M.-C.H.)
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 11267, Taiwan; (T.-T.L.); (U.-C.Y.); (C.-C.H.); (S.-F.H.); (T.-H.L.); (M.-W.L.)
| | - Tze-Tze Liu
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 11267, Taiwan; (T.-T.L.); (U.-C.Y.); (C.-C.H.); (S.-F.H.); (T.-H.L.); (M.-W.L.)
- Genomic Research Center, National Yang Ming Chiao Tung University, Taipei 11267, Taiwan
| | - Ueng-Cheng Yang
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 11267, Taiwan; (T.-T.L.); (U.-C.Y.); (C.-C.H.); (S.-F.H.); (T.-H.L.); (M.-W.L.)
- Institute of Biomedical Informatics, Taipei 11267, Taiwan
| | - Chia-Chang Huang
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 11267, Taiwan; (T.-T.L.); (U.-C.Y.); (C.-C.H.); (S.-F.H.); (T.-H.L.); (M.-W.L.)
- Division of Clinical Skills Training Center, Department of Medical Education, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 11267, Taiwan
| | - Chih-Wei Liu
- Department of Medicine, Taipei Veterans General Hospital, Taipei 11267, Taiwan; (Y.-T.C.); (C.-W.L.); (Y.-H.H.); (M.-C.H.)
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 11267, Taiwan; (T.-T.L.); (U.-C.Y.); (C.-C.H.); (S.-F.H.); (T.-H.L.); (M.-W.L.)
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 11267, Taiwan
| | - Shiang-Fen Huang
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 11267, Taiwan; (T.-T.L.); (U.-C.Y.); (C.-C.H.); (S.-F.H.); (T.-H.L.); (M.-W.L.)
- Division of Infection, Taipei Veterans General Hospital, Taipei 11267, Taiwan
| | - Tzu-Hao Li
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 11267, Taiwan; (T.-T.L.); (U.-C.Y.); (C.-C.H.); (S.-F.H.); (T.-H.L.); (M.-W.L.)
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 11267, Taiwan
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 11267, Taiwan
| | - Hsuan-Miao Liu
- Graduate Institute of Traditional Chinese Medicine, Chang Guang Memorial Hospital, Linkou 33371, Taiwan; (H.-M.L.); (T.-Y.L.)
| | - Ming-Wei Lin
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 11267, Taiwan; (T.-T.L.); (U.-C.Y.); (C.-C.H.); (S.-F.H.); (T.-H.L.); (M.-W.L.)
- Institute of Public Health, National Yang-Ming University, Taipei 11267, Taiwan
| | - Ying-Ying Yang
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 11267, Taiwan; (T.-T.L.); (U.-C.Y.); (C.-C.H.); (S.-F.H.); (T.-H.L.); (M.-W.L.)
- Division of Clinical Skills Training Center, Department of Medical Education, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 11267, Taiwan
- Division of Gastroenterology and Hepatology, Taipei Veterans General Hospital, Taipei 11267, Taiwan
- Correspondence: (Y.-Y.Y.); (H.-C.L.); Tel.: +886-2-2875-7725 (Y.-Y.Y.); +886-2-2875-2249 (H.-C.L.); Fax: +886-2-2875-7726 (Y.-Y.Y.); +886-2-2875-7809 (H.-C.L.)
| | - Tzung-Yan Lee
- Graduate Institute of Traditional Chinese Medicine, Chang Guang Memorial Hospital, Linkou 33371, Taiwan; (H.-M.L.); (T.-Y.L.)
| | - Yi-Hsiang Huang
- Department of Medicine, Taipei Veterans General Hospital, Taipei 11267, Taiwan; (Y.-T.C.); (C.-W.L.); (Y.-H.H.); (M.-C.H.)
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 11267, Taiwan; (T.-T.L.); (U.-C.Y.); (C.-C.H.); (S.-F.H.); (T.-H.L.); (M.-W.L.)
- Institute of Public Health, National Yang-Ming University, Taipei 11267, Taiwan
- Division of Gastroenterology and Hepatology, Taipei Veterans General Hospital, Taipei 11267, Taiwan
| | - Ming-Chih Hou
- Department of Medicine, Taipei Veterans General Hospital, Taipei 11267, Taiwan; (Y.-T.C.); (C.-W.L.); (Y.-H.H.); (M.-C.H.)
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 11267, Taiwan; (T.-T.L.); (U.-C.Y.); (C.-C.H.); (S.-F.H.); (T.-H.L.); (M.-W.L.)
| | - Han-Chieh Lin
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 11267, Taiwan; (T.-T.L.); (U.-C.Y.); (C.-C.H.); (S.-F.H.); (T.-H.L.); (M.-W.L.)
- Division of Gastroenterology and Hepatology, Taipei Veterans General Hospital, Taipei 11267, Taiwan
- Correspondence: (Y.-Y.Y.); (H.-C.L.); Tel.: +886-2-2875-7725 (Y.-Y.Y.); +886-2-2875-2249 (H.-C.L.); Fax: +886-2-2875-7726 (Y.-Y.Y.); +886-2-2875-7809 (H.-C.L.)
| |
Collapse
|
39
|
Yang Y, Li L. Depleting microRNA-146a-3p attenuates lipopolysaccharide-induced acute lung injury via up-regulating SIRT1 and mediating NF-κB pathway. J Drug Target 2021; 29:420-429. [PMID: 33185125 DOI: 10.1080/1061186x.2020.1850738] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The role of microRNAs (miRs) in acute lung injury (ALI) has been discussed. This study is to uncover the effects of miR-146a-3p/Sirtuin-1 (SIRT1)/Nuclear factor-kappa B (NF-κB) axis on ALI. METHODS Human normal lung epithelial cell line BEAS-2B was exposed to lipopolysaccharide (LPS) to establish an in vitro model of ALI. NF-κB expression, cell activity, apoptosis, inflammatory factors, oxidative stress indices were detected in LPS-induced BEAS-2B cells after miR-146a-3p was down-regulated or SIRT1 was up-regulated. ALI rat model was established and the NF-κB expression, wet/dry weight (W/D) ratio, pathological changes, pneumonocyte apoptosis, inflammatory factors, oxidative stress indices were detected in ALI rats after miR-146a-3p was down-regulated or SIRT1 was up-regulated. The target relationship between miR-146a-3p and SIRT1 was confirmed. RESULTS Reduced SIRT1 and raised miR-146a-3p were found in LPS-induced BEAS-2B cells and ALI rats. SIRT1-overexpressing or miR-146a-3p-underexpressing up-regulated NF-κB expression, promoted viability and inhibited apoptosis of LPS-induced BEAS-2B cells in vitro, and increased NF-κB expression, down-regulated the W/D ratio, attenuated pathological changes, suppressed apoptosis, and alleviated inflammatory response and oxidative stress in the lung of ALI rats. MiR-146a-3p directly binds to the 3'UTR of SIRT1 mRNA. CONCLUSION Depleting miR-146a-3p improves ALI through up-regulating SIRT1 and mediating NF-κB pathway.
Collapse
Affiliation(s)
- Yuxia Yang
- Department of Emergency Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Li Li
- Department of Emergency Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
40
|
Shtaif B, Bar-Maisels M, Gabet Y, Hiram-Bab S, Yackobovitch-Gavan M, Phillip M, Gat-Yablonski G. Cartilage -specific knockout of Sirt1 significantly reduces bone quality and catch-up growth efficiency. Bone 2020; 138:115468. [PMID: 32512163 DOI: 10.1016/j.bone.2020.115468] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/27/2020] [Accepted: 06/01/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Spontaneous catch-up (CU) growth occurs when a growth-restricting factor is resolved. However, its efficiency is sometimes inadequate and growth deficits remain permanent. The therapeutic toolbox for short stature is currently very limited, thus, finding new regulatory pathways is important for the development of novel means of treatment. Our previous studies using a nutrition-induced CU growth model showed that the level of sirtuin-1 (Sirt1) was significantly increased in food-restricted animals and decreased during CU growth. AIM This study sought to investigate the role of Sirt1 in modulating the response of the epiphyseal growth plate (EGP) to nutritional manipulation. METHOD Collagen type II-specific Sirt1 knockout (CKO) mice were tested for response to our CU growth model consisting of a period of food restriction followed by re-feeding. RESULTS The transgenic CKO mice weighed more than the control (CTL) mice, their EGP was higher and less organized, specifically at the resting and proliferative zones, leading to shorter bones. Ablation of Sirt1 in the chondrocytes was found to have a dramatic effect on bone mineralization on micro-CT analysis. The CKO mice were less responsive to the nutritional manipulation, and their CU growth was less efficient. They remained shorter than the CTL mice who corrected the food restriction-induced growth deficit during the re-feeding period. CONCLUSIONS Sirt1 appears to be important for normal regulation of the EGP. In its absence, the EGP is less organized and CU growth is less efficient. These results suggest that SIRT1 may serve as a novel therapeutic target for short stature.
Collapse
Affiliation(s)
- Biana Shtaif
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Felsenstein Medical Research Center, Petach Tikva, Israel.
| | - Meytal Bar-Maisels
- Felsenstein Medical Research Center, Petach Tikva, Israel; The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.
| | - Yankel Gabet
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Sahar Hiram-Bab
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Michal Yackobovitch-Gavan
- The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.
| | - Moshe Phillip
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Felsenstein Medical Research Center, Petach Tikva, Israel; The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.
| | - Galia Gat-Yablonski
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Felsenstein Medical Research Center, Petach Tikva, Israel; The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.
| |
Collapse
|
41
|
Buhrmann C, Honarvar A, Setayeshmehr M, Karbasi S, Shakibaei M, Valiani A. Herbal Remedies as Potential in Cartilage Tissue Engineering: An Overview of New Therapeutic Approaches and Strategies. Molecules 2020; 25:E3075. [PMID: 32640693 PMCID: PMC7411884 DOI: 10.3390/molecules25133075] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/01/2020] [Accepted: 07/03/2020] [Indexed: 01/01/2023] Open
Abstract
It is estimated that by 2023, approximately 20% of the population of Western Europe and North America will suffer from a degenerative joint disease commonly known as osteoarthritis (OA). During the development of OA, pro-inflammatory cytokines are one of the major causes that drive the production of inflammatory mediators and thus of matrix-degrading enzymes. OA is a challenging disease for doctors due to the limitation of the joint cartilage's capacity to repair itself. Though new treatment approaches, in particular with mesenchymal stem cells (MSCs) that integrate the tissue engineering (TE) of cartilage tissue, are promising, they are not only expensive but more often do not lead to the regeneration of joint cartilage. Therefore, there is an increasing need for novel, safe, and more effective alternatives to promote cartilage joint regeneration and TE. Indeed, naturally occurring phytochemical compounds (herbal remedies) have a great anti-inflammatory, anti-oxidant, and anabolic potential, and they have received much attention for the development of new therapeutic strategies for the treatment of inflammatory diseases, including the prevention of age-related OA and cartilage TE. This paper summarizes recent research on herbal remedies and their chondroinductive and chondroprotective effects on cartilage and progenitor cells, and it also emphasizes the possibilities that exist in this research area, especially with regard to the nutritional support of cartilage regeneration and TE, which may not benefit from non-steroidal anti-inflammatory drugs (NSAIDs).
Collapse
Affiliation(s)
- Constanze Buhrmann
- Musculoskeletal Research Group and Tumour Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, Pettenkoferstrasse 11, D-80336 Munich, Germany;
| | - Ali Honarvar
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan 73461-81746, Iran; (A.H.); (M.S.)
| | - Mohsen Setayeshmehr
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan 73461-81746, Iran; (A.H.); (M.S.)
- Biomaterials Nanotechnology and Tissue Engineering Group, Department of Advanced Medical Technology, Isfahan University of Medical Sciences, Isfahan 73461-81746, Iran;
| | - Saeed Karbasi
- Biomaterials Nanotechnology and Tissue Engineering Group, Department of Advanced Medical Technology, Isfahan University of Medical Sciences, Isfahan 73461-81746, Iran;
| | - Mehdi Shakibaei
- Musculoskeletal Research Group and Tumour Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, Pettenkoferstrasse 11, D-80336 Munich, Germany;
| | - Ali Valiani
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan 73461-81746, Iran; (A.H.); (M.S.)
| |
Collapse
|
42
|
Li W, Alahdal M, Deng Z, Liu J, Zhao Z, Cheng X, Chen X, Li J, Yin J, Li Y, Wang G, Wang D, Tang K, Zhang J. Molecular functions of FSTL1 in the osteoarthritis. Int Immunopharmacol 2020; 83:106465. [PMID: 32259701 DOI: 10.1016/j.intimp.2020.106465] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/11/2020] [Accepted: 03/29/2020] [Indexed: 12/18/2022]
|
43
|
The role of SIRT1 in BMP2-induced chondrogenic differentiation and cartilage maintenance under oxidative stress. Aging (Albany NY) 2020; 12:9000-9013. [PMID: 32445555 PMCID: PMC7288925 DOI: 10.18632/aging.103161] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 03/30/2020] [Indexed: 12/13/2022]
Abstract
Articular cartilage defects are common in the clinic but difficult to treat. Exploring the chondrogenic molecular mechanisms of mesenchymal stem cells (MSCs) is of great theoretical interest and industrial significance. Bone morphogenetic protein 2 (BMP2) is a key factor that induces cartilage differentiation and can induce stem cell chondrogenic differentiation. However, the oxidative stress in the microenvironment during cartilage injury and degeneration inhibits cartilage regeneration and homeostasis. Silent mating type information regulator 2 homolog-1 (SIRT1) is an important histone deacetylase that regulates proliferation, differentiation, aging, and inflammation processes; moreover, it is an essential factor for chondrogenesis. The specific mechanism of SIRT1 in cartilage differentiation and homeostasis is still unclear. First, we investigated whether SIRT1 could coordinate BMP2-induced chondrogenic differentiation. Second, we investigated the protective effect of SIRT1 on BMP2-induced MSCs under oxidative stress. The results showed that SIRT1 could promote BMP2-induced chondrogenic differentiation of MSCs, and reduce the apoptosis and decomposition of extracellular matrix under oxidative stress. In summary, these results suggested that SIRT1 plays an important coordination role in BMP2-induced chondrogenic differentiation of stem cells and cartilage maintenance under oxidative stress, establishing the experimental basis for exploring the use of SIRT1 in cartilage defect repair.
Collapse
|
44
|
Yessenkyzy A, Saliev T, Zhanaliyeva M, Masoud AR, Umbayev B, Sergazy S, Krivykh E, Gulyayev A, Nurgozhin T. Polyphenols as Caloric-Restriction Mimetics and Autophagy Inducers in Aging Research. Nutrients 2020; 12:E1344. [PMID: 32397145 PMCID: PMC7285205 DOI: 10.3390/nu12051344] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/16/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023] Open
Abstract
It has been thought that caloric restriction favors longevity and healthy aging where autophagy plays a vital role. However, autophagy decreases during aging and that can lead to the development of aging-associated diseases such as cancer, diabetes, neurodegeneration, etc. It was shown that autophagy can be induced by mechanical or chemical stress. In this regard, various pharmacological compounds were proposed, including natural polyphenols. Apart from the ability to induce autophagy, polyphenols, such as resveratrol, are capable of modulating the expression of pro- and anti-apoptotic factors, neutralizing free radical species, affecting mitochondrial functions, chelating redox-active transition metal ions, and preventing protein aggregation. Moreover, polyphenols have advantages compared to chemical inducers of autophagy due to their intrinsic natural bio-compatibility and safety. In this context, polyphenols can be considered as a potential therapeutic tool for healthy aging either as a part of a diet or as separate compounds (supplements). This review discusses the epigenetic aspect and the underlying molecular mechanism of polyphenols as an anti-aging remedy. In addition, the recent advances of studies on NAD-dependent deacetylase sirtuin-1 (SIRT1) regulation of autophagy, the role of senescence-associated secretory phenotype (SASP) in cells senescence and their regulation by polyphenols have been highlighted as well. Apart from that, the review also revised the latest information on how polyphenols can help to improve mitochondrial function and modulate apoptosis (programmed cell death).
Collapse
Affiliation(s)
- Assylzhan Yessenkyzy
- Research Institute of Fundamental and Applied Medicine named after B. Atchabarov, S.D. Asfendiyarov Kazakh National Medical University, Almaty 050000, Kazakhstan; (A.Y.); (T.N.)
| | - Timur Saliev
- Research Institute of Fundamental and Applied Medicine named after B. Atchabarov, S.D. Asfendiyarov Kazakh National Medical University, Almaty 050000, Kazakhstan; (A.Y.); (T.N.)
| | - Marina Zhanaliyeva
- Department of Human Anatomy, NSC “Medical University of Astana”, Nur-Sultan 010000, Kazakhstan;
| | - Abdul-Razak Masoud
- Department of Biological Sciences, Louisiana Tech University, Ruston, LA 71270, USA;
| | - Bauyrzhan Umbayev
- National Laboratory Astana, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (B.U.); (S.S.); (A.G.)
| | - Shynggys Sergazy
- National Laboratory Astana, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (B.U.); (S.S.); (A.G.)
| | - Elena Krivykh
- Khanty-Mansiysk State Medical Academy, Tyumen Region, Khanty-Mansiysk Autonomous Okrug—Ugra, Khanty-Mansiysk 125438, Russia;
| | - Alexander Gulyayev
- National Laboratory Astana, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (B.U.); (S.S.); (A.G.)
| | - Talgat Nurgozhin
- Research Institute of Fundamental and Applied Medicine named after B. Atchabarov, S.D. Asfendiyarov Kazakh National Medical University, Almaty 050000, Kazakhstan; (A.Y.); (T.N.)
| |
Collapse
|
45
|
Sirtuin 1: A Dilemma in Transplantation. J Transplant 2020; 2020:9012980. [PMID: 32373350 PMCID: PMC7196964 DOI: 10.1155/2020/9012980] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/15/2020] [Accepted: 03/20/2020] [Indexed: 12/30/2022] Open
Abstract
Sirtuin 1, a member of sirtuin family of histone deacetylase enzymes, has been implicated in a variety of physiologic and pathologic events, including energy metabolism, cell survival, and age-related alterations. In view of the anti-inflammatory properties of sirtuin 1 along with its protective role in ischemia reperfusion injury, it might be considered as contributing to the promotion of transplantation outcome. However, the potential ability of sirtuin 1 to induce malignancies raises some concerns about its overexpression in clinic. Moreover, despite the findings of sirtuin 1 implication in thymic tolerance induction and T regulatory (Treg) cells survival, there is also evidence for its involvement in Treg suppression and in T helper 17 cells differentiation. The identification of sirtuin 1 natural and synthetic activators leads to the proposal of sirtuin 1 as an eligible target for clinical interventions in transplantation. All positive and negative consequences of sirtuin 1 overactivation/overexpression in the allograft should therefore be studied thoroughly. Herein, we summarize previous findings concerning direct and indirect influences of sirtuin 1 manipulation on transplantation.
Collapse
|
46
|
Ali D, Chen L, Kowal JM, Okla M, Manikandan M, AlShehri M, AlMana Y, AlObaidan R, AlOtaibi N, Hamam R, Alajez NM, Aldahmash A, Kassem M, Alfayez M. Resveratrol inhibits adipocyte differentiation and cellular senescence of human bone marrow stromal stem cells. Bone 2020; 133:115252. [PMID: 31978617 DOI: 10.1016/j.bone.2020.115252] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 01/13/2020] [Accepted: 01/20/2020] [Indexed: 12/16/2022]
Abstract
Bone marrow adipose tissue (BMAT) is a unique adipose depot originating from bone marrow stromal stem cells (BMSCs) and regulates bone homeostasis and energy metabolism. An increased BMAT volume is observed in several conditions e.g. obesity, type 2 diabetes, osteoporosis and is known to be associated with bone fragility and increased risk for fracture. Therapeutic approaches to decrease the accumulation of BMAT are clinically relevant. In a screening experiment of natural compounds, we identified Resveratrol (RSV), a plant-derived antioxidant mediating biological effects via sirtuin- related mechanisms, to exert significant effects of BMAT formation. Thus, we examined in details the effects RSV on adipocytic and osteoblastic differentiation of tolermerized human BMSCs (hBMSC-TERT). RSV (1.0 μM) enhanced osteoblastic differentiation and inhibited adipocytic differentiation of hBMSC-TERT when compared with control and Sirtinol (Sirtuin inhibitor). Global gene expression profiling and western blot analysis revealed activation of a number of signaling pathways including focal adhesion kinase (FAK). Pharmacological inhibition of FAK using (PF-573228) and AKT inhibitor (LY-294002) (5μM), diminished RSV-induced osteoblast differentiation. In addition, RSV reduced the levels of senescence-associated secretory phenotype (SASP), gene markers associated with senescence (P53, P16, and P21), intracellular ROS levels and increased gene expression of enzymes protecting cells from oxidative damage (HMOX1 and SOD3). In vitro treatment of primary hBMSCs from aged patients characterized with high adipocytic and low osteoblastic differentiation ability with RSV, significantly enhanced osteoblast and decreased adipocyte formation when compared to hBMSCs from young donors. RSV targets hBMSCs and inhibits adipogenic differentiation and senescence-associated phenotype and thus a potential agent for treating conditions of increased BMAT formation.
Collapse
Affiliation(s)
- Dalia Ali
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology & Metabolism, University Hospital of Odense and University of Southern Denmark, Odense, Denmark.
| | - Li Chen
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology & Metabolism, University Hospital of Odense and University of Southern Denmark, Odense, Denmark.
| | - Justyna M Kowal
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology & Metabolism, University Hospital of Odense and University of Southern Denmark, Odense, Denmark.
| | - Meshail Okla
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia.
| | - Muthurangan Manikandan
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia.
| | - Moayad AlShehri
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia.
| | - Yousef AlMana
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia.
| | - Reham AlObaidan
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia.
| | - Najd AlOtaibi
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia.
| | - Rimi Hamam
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia.
| | - Nehad M Alajez
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia.
| | - Abdullah Aldahmash
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia; Prince Naif Health Research Center, King Saud University, Riyadh, Saudi Arabia.
| | - Moustapha Kassem
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia; Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology & Metabolism, University Hospital of Odense and University of Southern Denmark, Odense, Denmark; Department of Cellular and Molecular Medicine, Danish Stem Cell Center (DanStem), University of Copenhagen, 2200 Copenhagen, Denmark.
| | - Musaad Alfayez
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
47
|
Constanze B, Popper B, Aggarwal BB, Shakibaei M. Evidence that TNF-β suppresses osteoblast differentiation of mesenchymal stem cells and resveratrol reverses it through modulation of NF-κB, Sirt1 and Runx2. Cell Tissue Res 2020; 381:83-98. [PMID: 32140928 DOI: 10.1007/s00441-020-03188-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 02/13/2020] [Indexed: 12/16/2022]
Abstract
It has been established that inflammation plays an important role in bone formation and bone loss. Although a lot is known about the role of TNF-α in bone health, very little is understood about TNF-β, also called lymphotoxin. In this report, we examine the effect of TNF-β on osteogenic differentiation of mesenchymal stem cells (MSCs) and its modulation by resveratrol. Monolayer and high-density cultures of MSCs were treated with osteogenic induction medium with/without TNF-β, Sirt1 inhibitor nicotinamide (NAM), antisense oligonucleotides against Sirt1 (ASO) and/or Sirt1 stimulator resveratrol. We found that TNF-β inhibits, in a similar way to NAM or Sirt1-ASO, the early stage of osteogenic differentiation of MSCs and this was accompanied with downregulation of bone-specific matrix, β1-integrin, Runx2 and with upregulation of NF-κB phosphorylation and NF-κB-regulated gene products involved in the inflammatory, degradative processes and apoptosis. However, resveratrol reversed TNF-β- and NAM-suppressed MSCs osteogenesis by activation of Sirt1 and Runx2 that led to osteoblast differentiation. Furthermore, downregulation of Sirt1 by mRNA inhibited the effect of resveratrol, highlighting the important impact of this enzyme in the TNF-β signaling pathway. Finally, resveratrol was able to manifest its effect both by suppression of TNF-β-induced NF-κB and through direct activation of the Sirt1 and Runx2 pathway. Thus, through these studies, we present a mechanism by which a T cell-derived cytokine, TNF-β can affect bone formation through modulation of MSCs differentiation that involves NF-κB, Sirt1, Runx2 and resveratrol reversed TNF-β-promoted impairments in MSCs osteogenesis.
Collapse
Affiliation(s)
- Buhrmann Constanze
- Musculoskeletal Research Group and Tumour Biology, Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig-Maximilian-University Munich, Pettenkoferstrasse 11, 80336, Munich, Germany
| | - Bastian Popper
- Biomedical Center, Core facility animal models, Ludwig-Maximilian-University Munich, 82152, Martinsried, Germany.,Institute of Pathology, School of Medicine, Technical University of Munich, 81675, Munich, Germany
| | | | - Mehdi Shakibaei
- Musculoskeletal Research Group and Tumour Biology, Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig-Maximilian-University Munich, Pettenkoferstrasse 11, 80336, Munich, Germany.
| |
Collapse
|
48
|
Zhao X, Huang P, Li G, Feng Y, Zhendong L, Zhou C, Hu G, Xu Q. Overexpression of Pitx1 attenuates the senescence of chondrocytes from osteoarthritis degeneration cartilage-A self-controlled model for studying the etiology and treatment of osteoarthritis. Bone 2020; 131:115177. [PMID: 31783149 DOI: 10.1016/j.bone.2019.115177] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 11/24/2019] [Accepted: 11/25/2019] [Indexed: 12/13/2022]
Abstract
To explore the role of low expression of Pitx1 in degenerative cartilage tissue. A cartilage injury model was established by using the cartilage scratch method. The newly generated tissue by BrdU labeled in injured cartilage region expressed SOX-9 and Col2A1 in 5-week-old rats. Compared with that, the number of BrdU-positive cells was lower in 4-month-old cartilage injury model rats. Compared with that in lateral cartilage, the expression of Pitx1 was lower in medial cartilage. Compared with chondrocytes derived from the lateral cartilage, chondrocytes derived from the medial cartilage exhibited significantly increased cell aging, as determined by SA-β-GAL staining; downregulated Pitx1 expression; reduced autophagy levels; and decreased Col2A1 expression in a chondrogenic differentiation assay. Inhibition of Pitx1 expression in chondrocytes from the lateral cartilage significantly increased the ratio of cell senescence. Overexpression of Pitx1 in chondrocytes derived from the medial cartilage decreased the cell senescence ratio. In a luciferase assay, Pitx1 was found to promote Sirt1 gene transcription. Decreased Pitx1 expression is an essential cause of cartilage degeneration in the medial tibial plateau. The described self-controlled model is an excellent way to study OA etiology and screen therapeutic drugs for OA.
Collapse
Affiliation(s)
- Xiang Zhao
- Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Huang
- Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Gen Li
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yu Feng
- Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lv Zhendong
- Department of Spine Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chun Zhou
- Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guangyu Hu
- Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Qingrong Xu
- Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai JiaoTong University, 160 Pujian Road, Shanghai 200127, China; Department of Orthopaedics, South Campus, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 2000, Jiangyue Road, Shanghai 201112, China.
| |
Collapse
|
49
|
Concise Review: The Regulatory Mechanism of Lysine Acetylation in Mesenchymal Stem Cell Differentiation. Stem Cells Int 2020; 2020:7618506. [PMID: 32399051 PMCID: PMC7204305 DOI: 10.1155/2020/7618506] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 01/02/2020] [Indexed: 12/30/2022] Open
Abstract
Nowadays, the use of MSCs has attracted considerable attention in the global science and technology field, with the self-renewal and multidirectional differentiation potential for diabetes, obesity treatment, bone repair, nerve repair, myocardial repair, and so on. Epigenetics plays an important role in the regulation of mesenchymal stem cell differentiation, which has become a research hotspot in the medical field. This review focuses on the role of lysine acetylation modification on the determination of MSC differentiation direction. During this progress, the recruitment of lysine acetyltransferases (KATs) and lysine deacetylases (KDACs) is the crux of transcriptional mechanisms in the dynamic regulation of key genes controlling MSC multidirectional differentiation.
Collapse
|
50
|
Wagner DR, Karnik S, Gunderson ZJ, Nielsen JJ, Fennimore A, Promer HJ, Lowery JW, Loghmani MT, Low PS, McKinley TO, Kacena MA, Clauss M, Li J. Dysfunctional stem and progenitor cells impair fracture healing with age. World J Stem Cells 2019; 11:281-296. [PMID: 31293713 PMCID: PMC6600851 DOI: 10.4252/wjsc.v11.i6.281] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/26/2019] [Accepted: 06/13/2019] [Indexed: 02/06/2023] Open
Abstract
Successful fracture healing requires the simultaneous regeneration of both the bone and vasculature; mesenchymal stem cells (MSCs) are directed to replace the bone tissue, while endothelial progenitor cells (EPCs) form the new vasculature that supplies blood to the fracture site. In the elderly, the healing process is slowed, partly due to decreased regenerative function of these stem and progenitor cells. MSCs from older individuals are impaired with regard to cell number, proliferative capacity, ability to migrate, and osteochondrogenic differentiation potential. The proliferation, migration and function of EPCs are also compromised with advanced age. Although the reasons for cellular dysfunction with age are complex and multidimensional, reduced expression of growth factors, accumulation of oxidative damage from reactive oxygen species, and altered signaling of the Sirtuin-1 pathway are contributing factors to aging at the cellular level of both MSCs and EPCs. Because of these geriatric-specific issues, effective treatment for fracture repair may require new therapeutic techniques to restore cellular function. Some suggested directions for potential treatments include cellular therapies, pharmacological agents, treatments targeting age-related molecular mechanisms, and physical therapeutics. Advanced age is the primary risk factor for a fracture, due to the low bone mass and inferior bone quality associated with aging; a better understanding of the dysfunctional behavior of the aging cell will provide a foundation for new treatments to decrease healing time and reduce the development of complications during the extended recovery from fracture healing in the elderly.
Collapse
Affiliation(s)
- Diane R Wagner
- Department of Mechanical and Energy Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Sonali Karnik
- Department of Mechanical and Energy Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Zachary J Gunderson
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Jeffery J Nielsen
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, United States
| | - Alanna Fennimore
- Department of Physical Therapy, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Hunter J Promer
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN 46222, United States
| | - Jonathan W Lowery
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN 46222, United States
| | - M Terry Loghmani
- Department of Physical Therapy, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, IN 47907 United States
| | - Todd O McKinley
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, United States
| | - Matthias Clauss
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Jiliang Li
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| |
Collapse
|