1
|
Li Y, Zhang L, Wu D, Zhang Z, Zhou Y, Li J. Kynurenic Acid, a Small Foodborne Molecule with the Potential to Affect Human Health. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:8729-8739. [PMID: 40176362 DOI: 10.1021/acs.jafc.4c12355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Kynurenic acid (KYNA) is one of the metabolites of tryptophan (Trp), which can be supplied by endogenous synthesis or diet and involves various aspects of nutrition and metabolism. KYNA has been proven to be an antagonist of α7-nicotinic acetylcholine and NMDA receptors, which can regulate the neurotransmission of cholinergic and glutaminergic acid, so KYNA is mainly associated with neurodegenerative diseases. Other metabolites in the Trp and kynurenine metabolic pathways, such as 3-hydroxy-l-kynurenine (3-HK), 5-hydroxytryptophan (5-HT), and quinolinic acid (QA), are also involved in immune regulation and have pro-stimulatory or anti-excitatory toxicity. KYNA is present in both the human body and the daily diet, so it has the potential as a new dietary supplement. It may be of great significance in treating psychiatric disorders.
Collapse
Affiliation(s)
- Yuqi Li
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Lingyu Zhang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
- Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen 361021, China
| | - Daren Wu
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
- Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen 361021, China
| | - Zhengxiao Zhang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
- Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen 361021, China
| | - Yongbo Zhou
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
- Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen 361021, China
| | - Jian Li
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
- Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen 361021, China
| |
Collapse
|
2
|
Luo B, Liang Z, Lin W, Li Y, Zhong W, Bai D, Hu X, Xie J, Li X, Wang P, Zhu X, Zhang R, Yang L. Aqueous extract of Rehmanniae Radix Praeparata improves bone health in ovariectomized rats by modulating the miR-29a-3p/NFIA/Wnt signaling pathway axis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119549. [PMID: 40024453 DOI: 10.1016/j.jep.2025.119549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/23/2025] [Accepted: 02/22/2025] [Indexed: 03/04/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Rehmanniae Radix Praeparata (RRP), a widely used traditional Chinese medicine and a processed form of Rehmannia glutinosa, is primarily utilized to supplement kidney function and promote bone health. Clinical evidence suggests that RRP exhibits significant efficacy in the treatment of osteoporosis (OP). However, the precise mechanisms underlying its therapeutic effects remain incompletely understood. AIM OF THE STUDY OP is a systemic skeletal disorder characterized by reduced bone density and quality, leading to an increased risk of fractures. The aim of this study is to evaluate the effectiveness and underlying mechanisms of RRP in treating OP. MATERIALS AND METHODS Ovariectomized (OVX) rats were administered RRP aqueous extract via gavage for three months. After the treatment period, femoral microstructure and osteogenic protein levels were assessed to evaluate the efficacy of RRP. Serum exosomes (Exos) derived from different groups of rats were isolated and characterized. The levels of miR-29a-3p in serum-derived Exos and femoral tissue were quantified. Subsequently, Exos were co-cultured with rat bone marrow mesenchymal stem cells (rBMSCs) to investigate their role in promoting osteogenic differentiation and explore the molecular mechanisms underlying this process, particularly through the miR-29a-3p/NFIA/Wnt signaling pathway axis. RESULTS OVX rats exhibited significant bone microdamage. In contrast, the RRP-treated OVX rats showed marked improvements in femoral bone microstructure and increased osteogenic protein expression. MiR-29a-3p levels were elevated in serum-derived Exos from the RRP-treated rats. Furthermore, rBMSCs treated with these Exos displayed an increase in miR-29a-3p expression. Further investigations revealed that miR-29a-3p promoted osteogenesis by inhibiting NFIA expression in both bone tissue and rBMSCs. Overexpression of NFIA reversed the osteogenic effects of miR-29a-3p, confirming NFIA as its direct target and suggesting that miR-29a-3p enhances osteogenesis by inhibiting NFIA. Additionally, NFIA was found to promote the transcription of SFRP1, an inhibitor of the Wnt signaling pathway. Our findings suggest that the RRP aqueous extract increases miR-29a-3p levels in serum Exos, which in turn inhibits NFIA and activates the Wnt signaling pathway, thereby promoting osteogenesis. CONCLUSION These findings suggest that the RRP aqueous extract improves bone health and mitigates bone microstructural damage caused by OP through the regulation of the miR-29a-3p/NFIA/Wnt signaling pathway axis.
Collapse
Affiliation(s)
- Bingjie Luo
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510630, PR China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Ziwen Liang
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510630, PR China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Weiwen Lin
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510630, PR China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Yan Li
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China; College of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Wenqiang Zhong
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510630, PR China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Donghui Bai
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China; College of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Xueling Hu
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510630, PR China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Ji Xie
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510630, PR China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Xiaoyun Li
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510630, PR China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Panpan Wang
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China; College of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Xiaofeng Zhu
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China; College of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Ronghua Zhang
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510630, PR China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China.
| | - Li Yang
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510630, PR China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China.
| |
Collapse
|
3
|
Lin W, Chow SKH, Cui C, Liu C, Wang Q, Chai S, Wong RMY, Zhang N, Cheung WH. Wnt/β-catenin signaling pathway as an important mediator in muscle and bone crosstalk: A systematic review. J Orthop Translat 2024; 47:63-73. [PMID: 39007034 PMCID: PMC11245956 DOI: 10.1016/j.jot.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 03/21/2024] [Accepted: 06/02/2024] [Indexed: 07/16/2024] Open
Abstract
Background The interaction between muscle and bone is shown to be clinically important but the underlying mechanisms are largely unknown. The canonical Wnt/β-catenin signaling pathway is reported to be involved in muscle-bone crosstalk, but its detailed function remains unclear. This systematic review aims to investigate and elucidate the role of the Wnt/β-catenin signaling pathways in muscle-bone crosstalk. Methods We conducted a literature search on the Web of Science, PubMed, EBSCO and Embase with keywords "Wnt*", "bone*" and "muscle*". A systematic review was completed according to the guideline of preferred reporting items of systematic reviews and meta-analyses (PRISMA). Data synthesis included species (human, animal or cell type used), treatments involved, outcome measures and key findings with respect to Wnts. Results Seventeen papers were published from 2007 to 2021 and were extracted from a total of 1529 search results in the databases of Web of Science (468 papers), PubMed (457 papers), EBSCO (371) and Embase (233). 12 Wnt family members were investigated in the papers, including Wnt1, Wnt2, Wnt2b, Wnt3a, Wnt4, Wnt5a, Wnt8a, Wnt8b, Wnt9a, Wnt10a, Wnt10b and Wnt16. Many studies showed that muscles were able to increase or decrease osteogenesis of bone, while bone increased myogenesis of muscle through Wnt/β-catenin signaling pathways. Wnt3a, Wnt4 and Wnt10b were shown to play important roles in the crosstalk between muscle and bone. Conclusions Wnt3a, Wnt4 and Wnt10b are found to play important mediatory roles in muscle-bone crosstalk. The role of Wnt4 was mostly found to regulate muscle from the bone side. Whilst the role of Wnt10b during muscle ageing was proposed, current evidence is insufficient to clarify the specific role of Wnt/β-catenin signaling in the interplay between sarcopenia and osteoporosis. More future studies are required to investigate the exact regulatory roles of Wnts in muscle-bone crosstalk in musculoskeletal disease models such as sarcopenia and osteoporosis. Translational potential of this article The systematic review provides an extensive overview to reveal the roles of Wnt/β-catenin signaling pathways in muscle-bone crosstalk. These results provide novel research directions to further understand the underlying mechanism of sarcopenia, osteoporosis, and their crosstalk, finally helping the future development of new therapeutic interventions.
Collapse
Affiliation(s)
- Wujian Lin
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
- Department of Rehabilitation Medicine, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Simon Kwoon Ho Chow
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
| | - Can Cui
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
| | - Chaoran Liu
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
| | - Qianjin Wang
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
| | - Senlin Chai
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
| | - Ronald Man Yeung Wong
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
| | - Ning Zhang
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
| | - Wing Hoi Cheung
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
| |
Collapse
|
4
|
Li SY, Xue ST, Li ZR. Osteoporosis: Emerging targets on the classical signaling pathways of bone formation. Eur J Pharmacol 2024; 973:176574. [PMID: 38642670 DOI: 10.1016/j.ejphar.2024.176574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/30/2024] [Accepted: 04/10/2024] [Indexed: 04/22/2024]
Abstract
Osteoporosis is a multifaceted skeletal disorder characterized by reduced bone mass and structural deterioration, posing a significant public health challenge, particularly in the elderly population. Treatment strategies for osteoporosis primarily focus on inhibiting bone resorption and promoting bone formation. However, the effectiveness and limitations of current therapeutic approaches underscore the need for innovative methods. This review explores emerging molecular targets within crucial signaling pathways, including wingless/integrated (WNT), bone morphogenetic protein (BMP), hedgehog (HH), and Notch signaling pathway, to understand their roles in osteogenesis regulation. The identification of crosstalk targets between these pathways further enhances our comprehension of the intricate bone metabolism cycle. In summary, unraveling the molecular complexity of osteoporosis provides insights into potential therapeutic targets beyond conventional methods, offering a promising avenue for the development of new anabolic drugs.
Collapse
Affiliation(s)
- Si-Yan Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Si-Tu Xue
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Zhuo-Rong Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
5
|
Gao G, Li L, Li C, Liu D, Wang Y, Li C. Mesenchymal stem cells: Guardians of women's health. Regen Ther 2024; 26:1087-1098. [PMID: 39582803 PMCID: PMC11585475 DOI: 10.1016/j.reth.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/13/2024] [Accepted: 10/23/2024] [Indexed: 11/26/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have attracted more and more attention because of their multidirectional differentiation potential, immune regulatory abilities and self-renewal capacity. In recent years, their use has become prominent in the domains of regenerative medicine and tissue engineering. MSCs have shown promise in therapeutic studies for a variety of diseases and have become a new source of innovative solutions for the treatment of some obstetric and gynecological diseases. This review systematically presents the latest research on the use of MSCs in the treatment of obstetrics- and gynecology-related diseases. Specifically, this review encompasses the latest findings related to the role of MSCs in premature ovarian failure, polycystic ovary syndrome, ovarian cancer, fallopian tube-related diseases, uterine adhesions, endometriosis, cesarean scar defects, postmenopausal osteoporosis, and pelvic floor dysfunction. The shortcomings and challenges of the future use of MSCs in disease treatment are also discussed, with the intent to motivate improvements in MSC applications in clinical therapy. It is believed that with further research, MSCs will play a more important role in the treatment of obstetrics- and gynecology-related diseases.
Collapse
Affiliation(s)
- Guanwen Gao
- Peking University Shenzhen Clinical Institute of Shantou University Medical College, Shenzhen, 518036, China
- Center of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Shenzhen Key Laboratory on Technology for Early Diagnosis of Major Gynecologic Diseases, Shenzhen, 518036, China
| | - Li Li
- Department of Internal Medicine, Jinan Central Hospital Affiliated to Shandong University, Ji Nan, 250000, China
| | - Changling Li
- Department of Obstetrics and Gynecology, Pingyi People's Hospital, Linyi City, Shandong Province, 276000, China
| | - Degao Liu
- Center of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Shenzhen Key Laboratory on Technology for Early Diagnosis of Major Gynecologic Diseases, Shenzhen, 518036, China
| | - Yunfei Wang
- Center of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Shenzhen Key Laboratory on Technology for Early Diagnosis of Major Gynecologic Diseases, Shenzhen, 518036, China
| | - Changzhong Li
- Center of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Shenzhen Key Laboratory on Technology for Early Diagnosis of Major Gynecologic Diseases, Shenzhen, 518036, China
| |
Collapse
|
6
|
Takkar S, Sharma G, Kaushal JB, Abdullah KM, Batra SK, Siddiqui JA. From orphan to oncogene: The role of GPR35 in cancer and immune modulation. Cytokine Growth Factor Rev 2024; 77:56-66. [PMID: 38514303 PMCID: PMC11793123 DOI: 10.1016/j.cytogfr.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/15/2024] [Accepted: 03/15/2024] [Indexed: 03/23/2024]
Abstract
G protein-coupled receptors (GPCRs) are well-studied and the most traceable cell surface receptors for drug discovery. One of the intriguing members of this family is G protein-coupled receptors 35 (GPR35), which belongs to the class A rhodopsin-like family of GPCRs identified over two decades ago. GPR35 presents interesting features such as ubiquitous expression and distinct isoforms. Moreover, functional and genome-wide association studies on its widespread expression have linked GPR35 with pathophysiological disease progression. Various pieces of evidence have been accumulated regarding the independent or endogenous ligand-dependent role of GPR35 in cancer progression and metastasis. In the current scenario, the relationship of this versatile receptor and its putative endogenous ligands for the activation of oncogenic signal transduction pathways at the cellular level is an active area of research. These intriguing features offered by GPR35 make it an oncological target, justifying its uniqueness at the physiological and pathophysiological levels concerning other GPCRs. For pharmacologically targeting receptor-induced signaling, few potential competitive antagonists have been discovered that offer high selectivity at a human level. In addition to its fascinating features, targeting GPR35 at rodent and human orthologue levels is distinct, thus contributing to the sub-species selectivity. Strategies to modulate these issues will help us understand and truly target GPR35 at the therapeutic level. In this article, we have provided prospects on each topic mentioned above and suggestions to overcome the challenges. This review discusses the molecular mechanism and signal transduction pathways activated by endogenous ligands or spontaneous auto-activation of GPR35 that contributes towards disease progression. Furthermore, we have highlighted the GPR35 structure, ubiquitous expression, its role in immunomodulation, and at the pathophysiological level, especially in cancer, indicating its status as a versatile receptor. Subsequently, we discussed the various proposed ligands and their mechanism of interaction with GPR35. Additionally, we have summarized the GPR35 antagonist that provides insights into the opportunities for therapeutically targeting this receptor.
Collapse
Affiliation(s)
- Simran Takkar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Gunjan Sharma
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jyoti B Kaushal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - K M Abdullah
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Jawed A Siddiqui
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
7
|
Yang X, Zhang W, Wang L, Zhao Y, Wei W. Metabolite-sensing GPCRs in rheumatoid arthritis. Trends Pharmacol Sci 2024; 45:118-133. [PMID: 38182481 DOI: 10.1016/j.tips.2023.12.001] [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: 11/13/2023] [Revised: 12/05/2023] [Accepted: 12/12/2023] [Indexed: 01/07/2024]
Abstract
Persistent inflammation in damaged joints results in metabolic dysregulation of the synovial microenvironment, causing pathogenic alteration of cell activity in rheumatoid arthritis (RA). Recently, the role of metabolite and metabolite-sensing G protein-coupled receptors (GPCRs) in the RA-related inflammatory immune response (IIR) has become a focus of research attention. These GPCRs participate in the progression of RA by modulating immune cell activation, migration, and inflammatory responses. Here, we discuss recent evidence implicating metabolic dysregulation in RA pathogenesis, focusing on the connection between RA-related IIR and GPCR signals originating from the synovial joint and gut. Furthermore, we discuss future directions for targeting metabolite-sensing GPCRs for therapeutic benefit, emphasizing the importance of identifying endogenous ligands and investigating the various transduction mechanisms involved.
Collapse
Affiliation(s)
- Xuezhi Yang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Wankang Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Luping Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Yingjie Zhao
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China.
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| |
Collapse
|
8
|
Pham TH, Kim EN, Trang NM, Jeong GS. Gallic acid induces osteoblast differentiation and alleviates inflammatory response through GPR35/GSK3β/β-catenin signaling pathway in human periodontal ligament cells. J Periodontal Res 2024; 59:204-219. [PMID: 37957813 DOI: 10.1111/jre.13208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 10/16/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND AND OBJECTIVE Gallic acid (GA) possesses various beneficial functions including antioxidant, anticancer, anti-inflammatory as well as inhibiting osteoclastogeneis. However, effects on osteogenic differentiation, especially in human ligament periodontal (hPDL) cells, remain unclear. Thus, the aim of this study was to evaluate the function of GA on osteogenesis and anti-inflammation in hPDL cells and to explore the involved underlying mechanism. METHODS Porphyromonas gingivalis lipopolysaccharide (Pg-LPS) treatment was used as a model for periodontitis. ROS production was determined by H2DCFDA staining. Trans-well and wound healing assays were performed for checking the migration effect of GA. Alizarin red and alkaline phosphatase activity (ALP) assays were performed to evaluate osteogenic differentiation. Osteogenesis and inflammatory-related genes and proteins were measured by real-time PCR and western blot. RESULTS Our results showed that GA-treated hPDL cells had higher proliferation and migration effect. GA inhibited ROS production-induced by Pg-LPS. Besides, GA abolished Pg-LPS-induced inflammation cytokines (il-6, il-1β) and inflammasome targets (Caspase-1, NLRP3). In addition, GA promoted ALP activity and mineralization in hPDL cells, lead to enhance osteoblast differentiation process. The effect of GA is related to G-protein-coupled receptor 35 (GPR35)/GSK3β/β-catenin signaling pathway. CONCLUSION GA attenuated Pg-LPS-induced inflammatory responses and periodontitis in hPDL cells. Taken together, GA may be targeted for therapeutic interventions in periodontal diseases.
Collapse
Affiliation(s)
- Thi Hoa Pham
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Eun-Nam Kim
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Nguyen Minh Trang
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Gil-Saeng Jeong
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
9
|
Ma L, Zhang S, Liang Q, Huang W, Wang H, Pan E, Xu P, Zhang S, Tao F, Tang J, Qing R. CrMP-Sol database: classification, bioinformatic analyses and comparison of cancer-related membrane proteins and their water-soluble variant designs. BMC Bioinformatics 2023; 24:360. [PMID: 37743473 PMCID: PMC10518928 DOI: 10.1186/s12859-023-05477-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 09/12/2023] [Indexed: 09/26/2023] Open
Abstract
Membrane proteins are critical mediators for tumor progression and present enormous therapeutic potentials. Although gene profiling can identify their cancer-specific signatures, systematic correlations between protein functions and tumor-related mechanisms are still unclear. We present here the CrMP-Sol database ( https://bio-gateway.aigene.org.cn/g/CrMP ), which aims to breach the gap between the two. Machine learning was used to extract key functional descriptions for protein visualization in the 3D-space, where spatial distributions provide function-based predictive connections between proteins and cancer types. CrMP-Sol also presents QTY-enabled water-soluble designs to facilitate native membrane protein studies despite natural hydrophobicity. Five examples with varying transmembrane helices in different categories were used to demonstrate the feasibility. Native and redesigned proteins exhibited highly similar characteristics, predicted structures and binding pockets, and slightly different docking poses against known ligands, although task-specific designs are still required for proteins more susceptible to internal hydrogen bond formations. The database can accelerate therapeutic developments and biotechnological applications of cancer-related membrane proteins.
Collapse
Affiliation(s)
- Lina Ma
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Sitao Zhang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Qi Liang
- Zhejiang Lab, Research Center for Intelligent Computing Platforms, Hangzhou, 311121, Zhejiang, China
| | - Wenting Huang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hui Wang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Emily Pan
- The Lawrenceville School, 2500 Main Street, Lawrenceville, NJ, 08648, USA
| | - Ping Xu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shuguang Zhang
- Media Lab, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Fei Tao
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Jin Tang
- Zhejiang Lab, Research Center for Intelligent Computing Platforms, Hangzhou, 311121, Zhejiang, China.
| | - Rui Qing
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
10
|
Im DS. Recent advances in GPR35 pharmacology; 5-HIAA serotonin metabolite becomes a ligand. Arch Pharm Res 2023:10.1007/s12272-023-01449-y. [PMID: 37227682 DOI: 10.1007/s12272-023-01449-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/15/2023] [Indexed: 05/26/2023]
Abstract
GPR35, an orphan receptor, has been waiting for its ligand since its cloning in 1998. Many endogenous and exogenous molecules have been suggested to act as agonists of GPR35 including kynurenic acid, zaprinast, lysophosphatidic acid, and CXCL17. However, complex and controversial responses to ligands among species have become a huge hurdle in the development of therapeutics in addition to the orphan state. Recently, a serotonin metabolite, 5-hydroxyindoleacetic acid (5-HIAA), is reported to be a high potency ligand for GPR35 by investigating the increased expression of GPR35 in neutrophils. In addition, a transgenic knock-in mouse line is developed, in which GPR35 was replaced with a human ortholog, making it possible not only to overcome the different selectivity of agonists among species but also to conduct therapeutic experiments on human GPR35 in mouse models. In the present article, I review the recent advances and prospective therapeutic directions in GPR35 research. Especially, I'd like to draw attention of readers to the finding of 5-HIAA as a ligand of GPR35 and lead to apply the 5-HIAA and human GPR35 knock-in mice to their research fields in a variety of pathophysiological conditions.
Collapse
Affiliation(s)
- Dong-Soon Im
- Department of Biomedical and Pharmaceutical Sciences and Department of Fundamental Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, 02446, Republic of Korea.
- Laboratory of Pharmacology, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| |
Collapse
|
11
|
Ma J, Chen P, Deng B, Wang R. Kynurenic acid promotes osteogenesis via the Wnt/β-catenin signaling. In Vitro Cell Dev Biol Anim 2023; 59:356-365. [PMID: 37291335 DOI: 10.1007/s11626-023-00774-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 05/09/2023] [Indexed: 06/10/2023]
Abstract
The role of kynurenic acid (KynA) in neurological and mental diseases has been widely studied. Emerging studies disclosed that KynA has a protective effect on tissues including heart, kidney, and retina. However, the role of KynA in osteoporosis has not been reported so far. To elucidate the role of KynA in age-related osteoporosis, both control and osteoporosis mice were administrated KynA for three consecutive months, and micro-computed tomography (μCT) analysis was then performed. In addition, primary bone marrow mesenchymal stem cells (BMSCs) were isolated for osteogenic differentiation induction and treated with KynA in vitro. Our data suggested that KynA administration rescued age-related bone loss in vivo, and KynA treatment promotes BMSC osteogenic differentiation in vitro. Moreover, KynA activated the Wnt/β-catenin signaling during BMSC osteogenic differentiation. Wnt inhibitor MSAB inhibited KynA-induced osteogenic differentiation. Further data demonstrated that KynA exerted its effect on BMSC osteogenic differentiation and Wnt/β-catenin signaling activation via G protein-coupled receptor 35 (GPR35). In conclusion, the protective effect of KynA on age-related osteoporosis was disclosed. Additionally, the promoting effect of KynA on osteoblastic differentiation via Wnt/β-catenin signaling was verified and the effect dependent on GPR35. These data suggest that KynA administration potentially contributes to the treatment of age-related osteoporosis.
Collapse
Affiliation(s)
- Jiangwei Ma
- Department of Orthopedics, The First Hospital of Yulin, No. 93, Yu Xi Street, Yulin, 719000, Shaanxi, People's Republic of China
| | - Pu Chen
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Baojuan Deng
- Department of General Practice, The First Hospital of Yulin, No. 93, Yu Xi Street, Yulin, 719000, Shaanxi, People's Republic of China
| | - Rong Wang
- Department of General Practice, The First Hospital of Yulin, No. 93, Yu Xi Street, Yulin, 719000, Shaanxi, People's Republic of China.
| |
Collapse
|
12
|
Noonan ML, Ni P, Solis E, Marambio YG, Agoro R, Chu X, Wang Y, Gao H, Xuei X, Clinkenbeard EL, Jiang G, Liu S, Stegen S, Carmeliet G, Thompson WR, Liu Y, Wan J, White KE. Osteocyte Egln1/Phd2 links oxygen sensing and biomineralization via FGF23. Bone Res 2023; 11:7. [PMID: 36650133 PMCID: PMC9845350 DOI: 10.1038/s41413-022-00241-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 09/29/2022] [Accepted: 11/03/2022] [Indexed: 01/19/2023] Open
Abstract
Osteocytes act within a hypoxic environment to control key steps in bone formation. FGF23, a critical phosphate-regulating hormone, is stimulated by low oxygen/iron in acute and chronic diseases, however the molecular mechanisms directing this process remain unclear. Our goal was to identify the osteocyte factors responsible for FGF23 production driven by changes in oxygen/iron utilization. Hypoxia-inducible factor-prolyl hydroxylase inhibitors (HIF-PHI) which stabilize HIF transcription factors, increased Fgf23 in normal mice, as well as in osteocyte-like cells; in mice with conditional osteocyte Fgf23 deletion, circulating iFGF23 was suppressed. An inducible MSC cell line ('MPC2') underwent FG-4592 treatment and ATACseq/RNAseq, and demonstrated that differentiated osteocytes significantly increased HIF genomic accessibility versus progenitor cells. Integrative genomics also revealed increased prolyl hydroxylase Egln1 (Phd2) chromatin accessibility and expression, which was positively associated with osteocyte differentiation. In mice with chronic kidney disease (CKD), Phd1-3 enzymes were suppressed, consistent with FGF23 upregulation in this model. Conditional loss of Phd2 from osteocytes in vivo resulted in upregulated Fgf23, in line with our findings that the MPC2 cell line lacking Phd2 (CRISPR Phd2-KO cells) constitutively activated Fgf23 that was abolished by HIF1α blockade. In vitro, Phd2-KO cells lost iron-mediated suppression of Fgf23 and this activity was not compensated for by Phd1 or -3. In sum, osteocytes become adapted to oxygen/iron sensing during differentiation and are directly sensitive to bioavailable iron. Further, Phd2 is a critical mediator of osteocyte FGF23 production, thus our collective studies may provide new therapeutic targets for skeletal diseases involving disturbed oxygen/iron sensing.
Collapse
Affiliation(s)
- Megan L Noonan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Pu Ni
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Emmanuel Solis
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yamil G Marambio
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Rafiou Agoro
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Xiaona Chu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yue Wang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Hongyu Gao
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Xiaoling Xuei
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Erica L Clinkenbeard
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Guanglong Jiang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Steve Stegen
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, 3000, Leuven, Belgium
| | - Geert Carmeliet
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, 3000, Leuven, Belgium
| | - William R Thompson
- Department of Physical Therapy, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Kenneth E White
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Departments of Medicine/Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
13
|
Chen S, Zeng J, Huang L, Peng Y, Yan Z, Zhang A, Zhao X, Li J, Zhou Z, Wang S, Jing S, Hu M, Li Y, Wang D, Wang W, Yu H, Miao J, Li J, Deng Y, Li Y, Liu T, Xu D. RNA adenosine modifications related to prognosis and immune infiltration in osteosarcoma. J Transl Med 2022; 20:228. [PMID: 35568866 PMCID: PMC9107650 DOI: 10.1186/s12967-022-03415-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 04/27/2022] [Indexed: 11/14/2022] Open
Abstract
Background RNA adenosine modifications, which are primarily mediated by “writer” enzymes (RMWs), play a key role in epigenetic regulation in various biological processes, including tumorigenesis. However, the expression and prognostic role of these genes in osteosarcoma (OS) remain unclear. Methods Univariate and multivariate Cox analyses were used to construct the RMW signature for OS using Target datasets. RMW expression in OS tissue was detected by qPCR analysis. Xcell and GSVA were used to determine the relationship between RMWs and immune infiltration. The DGIdb and CMap databases were used for drug prediction. In vivo and in vitro experiments showed that strophanthidin elicited antitumor activity against OS. Results A 3-RMW (CSTF2, ADAR and WTAP) prognostic signature in OS was constructed using the Target dataset and verified using GEO datasets and 63 independent OS tissues via qPCR analysis. High-risk OS patients had poor overall survival, and the prognostic signature was an independent prognostic factor for OS. Functional studies showed that tumour-, metabolism-, cell cycle- and immune-related pathways were related to high risk. Next, we found that RMW-derived high-risk patients exhibited increased infiltration of M2 macrophages and cDCs. Furthermore, we predicted the potential drugs for OS using the DGIdb and CMap databases. In vivo and in vitro experiments showed that strophanthidin elicited antitumor activity against OS by repressing cell growth and inducing cell cycle arrest at the G1 phase. Conclusion The 3-RWM-based prognostic signature established in this study is a novel gene signature associated with immune infiltration, and strophanthidin was identified as a candidate therapy for OS by repressing OS cell growth and the cell cycle. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03415-6.
Collapse
Affiliation(s)
- Shijie Chen
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China.,Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Rd, Shanghai, 200241, China
| | - Jin Zeng
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Liping Huang
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Yi Peng
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Zuyun Yan
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Aiqian Zhang
- Department of Obstetrics and Gynecology, The Third Xiangya Hospital of Central South University, 138, Tongzipo Road, Changsha, 410013, China
| | - Xingping Zhao
- Department of Obstetrics and Gynecology, The Third Xiangya Hospital of Central South University, 138, Tongzipo Road, Changsha, 410013, China
| | - Jun Li
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Rd, Hefei, 230601, Anhui, China
| | - Ziting Zhou
- The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Sidan Wang
- The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Shengyu Jing
- The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Minghua Hu
- Department of Anatomy, Histology, and Embryology, Changsha Medical University, 1501 Leifeng Avenue, Changsha, 410219, Hunan, China
| | - Yuezhan Li
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Dong Wang
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Weiguo Wang
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Haiyang Yu
- School of Basic Medical Science, Central South University, 172 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Jinglei Miao
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Jinsong Li
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Youwen Deng
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Yusheng Li
- Department of Orthopeadics, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, 410008, Hunan, China.
| | - Tang Liu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, 139 Renmin Middle Rd, Changsha, 410011, Hunan, China.
| | - Dabao Xu
- Department of Obstetrics and Gynecology, The Third Xiangya Hospital of Central South University, 138, Tongzipo Road, Changsha, 410013, China.
| |
Collapse
|
14
|
Exercised accelerated the production of muscle-derived kynurenic acid in skeletal muscle and alleviated the postmenopausal osteoporosis through the Gpr35/NFκB p65 pathway. J Orthop Translat 2022; 35:1-12. [PMID: 35846727 PMCID: PMC9260440 DOI: 10.1016/j.jot.2022.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/22/2022] [Accepted: 03/09/2022] [Indexed: 01/06/2023] Open
Abstract
Background Reduced serum estrogen levels in postmenopausal patients not only aggravate bone loss but also impact myokine secretion. Emerging evidence has revealed the importance of myokines in bone metabolism, and exercise can interfere with the secretion of myokines. However, few studies have explored the impact of exercise on myokine secretion in the postmenopausal osteoporosis (PMOP) process. Methods Ten-weeks-old C57B/L6 female mice were used for constructing the postmenopausal osteoporosis model. The expression levels of kynurenine aminotransferases (Kats) were detected by RT-PCR and Western Blot. The concentration of serum kynurenic acid (Kyna) was detected by HPLC-MS. Micro-CT analysis was used for determine the changes of bone mineral density and the microstructure. The primary osteoblast and osteoclast were isolated from mice to determine the effect and mechanism of Kyna on the bone formation and resorption. Results In our research, we found a lower serum level of muscle-derived kynurenic acid (Kyna) in PMOP model mice, accompanied by a decreased level of kynurenine aminotransferases (Kats) in the gastrocnemius muscle. Moreover, treadmill-running exercise upregulated the muscle levels of KATs and increased the serum concentration of Kyna, which was positively correlated with the alleviation of bone loss. Furthermore, we found that exogenous Kyna treatment alleviated bone mineral loss and microstructure destruction in PMOP mice by inhibiting osteoclast maturation and increasing osteoblast viability. Mechanistically, we observed that Kyna reduced the NFκB p65 phosphorylation level by activating the Gpr35 receptor, which inhibited NFATc1 expression in osteoclasts and upregulated Runx2 expression in osteoblasts. Conclusion Our results revealed that the muscle levels of Kats and serum level of Kyna were negatively correlated with the severity of PMOP. Exercise intervention and exogenous Kyna treatment alleviated the impairment of bone microstructure through the Gpr35 receptor, paving the way for a novel therapeutic intervention in PMOP. The Translational potential of this article This study provides evidences that Kyna could increase the osteoblastgenesis and inhibit the osteoclastgenesis, which could be a novel therapeutic approach for osteoporosis treatment.
Collapse
|
15
|
G-Protein Coupled Receptor 35 Induces Intervertebral Disc Degeneration by Mediating the Influx of Calcium Ions and Upregulating Reactive Oxygen Species. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5469220. [PMID: 35087615 PMCID: PMC8789411 DOI: 10.1155/2022/5469220] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/22/2022]
Abstract
Intervertebral disc degeneration (IDD) is a chronic disease affecting millions of patients; however, its specific etiology is unknown. G protein-coupled receptors (GPRs) are a superfamily of integral membrane receptors in cells, and the receptors respond to a diverse range of stimuli and participate in multiple cellular activities. Here, using RNA-sequencing (RNA-seq) methods and immunohistochemistry, we revealed that G protein-coupled receptor 35 (GPR35) may have a relationship with IDD. Then, we demonstrated that the deletion of GPR35 in nucleus pulposus cells (NPCs) with siRNA or in Gpr35−/− mice significantly alleviated IDD caused by senescence or mechanical stress, further validating the pathological role of GPR35 in IDD. In addition, GPR35 induced the influx of Ca2+ and upregulation of reactive oxygen species (ROS) under mechanical stress in NPCs, which we believe to be the mechanism of GPR35-induced IDD. Finally, GPR35 caused upregulation of ROS in NPCs under mechanical stress, while excessive ROS stimulated the NPCs to express more GPR35 with a significant dose or time response. The u-regulated GPR35 could sense mechanical stress to produce more ROS and perpetuate this harmful cycle. In summary, our study shows that GPR35 plays a critical role in mediating IDD via mediating the influx of calcium ions and upregulating ROS, which implies a strong potential advantage of GPR35 as a prevention and treatment target in IDD.
Collapse
|
16
|
Sanaei R, Kularathna P, Taghavi N, Hooper J, Pagel C, Mackie E. Protease-activated receptor-2 promotes osteogenesis in skeletal mesenchymal stem cells at the expense of adipogenesis: Involvement of interleukin-6. Bone Rep 2021; 15:101113. [PMID: 34430676 PMCID: PMC8365448 DOI: 10.1016/j.bonr.2021.101113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/26/2021] [Accepted: 07/30/2021] [Indexed: 10/27/2022] Open
Abstract
Bone marrow mesenchymal stem cells (MSCs) give rise to osteoblasts and adipocytes, with an inverse relationship between the two. The MSCs from protease-activated receptor-2 knockout (PAR2 KO) mice have a reduced capacity to generate osteoblasts. Here we describe the observation that PAR2 KO osteoblastic cultures generate more adipocytes than wildtype (WT) cultures. Osteoblasts from PAR2 KO mice expressed lower levels of osteoblastic genes (Runx2, Col1a1 and Bglap), and higher levels of the adipocytic gene Pparg than WT osteoblasts. Bone marrow stromal cells from PAR2 KO mice generated fewer osteoblastic colonies (assessed by staining for alkaline phosphatase activity and mineral deposition) and more adipocytic (Oil Red-O positive) colonies than cultures from WT mice. Similarly, cultures of the bone marrow stromal cell line (Kusa 4b10) in which PAR2 was knocked down (F2rl1 KD), were less osteoblastic and more adipocytic than vector control cells. Putative regulators of PAR2-mediated osteogenesis and suppression of adipogenesis were identified in an RNA-sequencing (RNA-seq) investigation; these include C1qtnf3, Gpr35, Grem1, Snorc and Tcea3, which were more highly expressed, and Cnr1, Enpep, Hmgn5, Il6 and Ramp3 which were expressed at lower levels, in control than in F2rl1 KD cells. Interleukin-6 (IL-6) levels were higher in medium harvested from F2rl1 KD cells than from control cells, and a neutralising anti-IL-6 antibody reduced the number of adipocytes in F2rl1 KD cultures to that of control cultures. Thus, PAR2 appears to be a mediator of the reciprocal relationship between osteogenesis and adipogenesis, with IL-6 having a regulatory role in these PAR2-mediated effects.
Collapse
Affiliation(s)
- R. Sanaei
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - P.K. Kularathna
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - N. Taghavi
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - J.D. Hooper
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - C.N. Pagel
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - E.J. Mackie
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|