1
|
Amroodi MN, Maghsoudloo M, Amiri S, Mokhtari K, Mohseni P, Pourmarjani A, Jamali B, Khosroshahi EM, Asadi S, Tabrizian P, Entezari M, Hashemi M, Wan R. Unraveling the molecular and immunological landscape: Exploring signaling pathways in osteoporosis. Biomed Pharmacother 2024; 177:116954. [PMID: 38906027 DOI: 10.1016/j.biopha.2024.116954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/05/2024] [Accepted: 06/15/2024] [Indexed: 06/23/2024] Open
Abstract
Osteoporosis, characterized by compromised bone density and microarchitecture, represents a significant global health challenge, particularly in aging populations. This comprehensive review delves into the intricate signaling pathways implicated in the pathogenesis of osteoporosis, providing valuable insights into the pivotal role of signal transduction in maintaining bone homeostasis. The exploration encompasses cellular signaling pathways such as Wnt, Notch, JAK/STAT, NF-κB, and TGF-β, all of which play crucial roles in bone remodeling. The dysregulation of these pathways is a contributing factor to osteoporosis, necessitating a profound understanding of their complexities to unveil the molecular mechanisms underlying bone loss. The review highlights the pathological significance of disrupted signaling in osteoporosis, emphasizing how these deviations impact the functionality of osteoblasts and osteoclasts, ultimately resulting in heightened bone resorption and compromised bone formation. A nuanced analysis of the intricate crosstalk between these pathways is provided to underscore their relevance in the pathophysiology of osteoporosis. Furthermore, the study addresses some of the most crucial long non-coding RNAs (lncRNAs) associated with osteoporosis, adding an additional layer of academic depth to the exploration of immune system involvement in various types of osteoporosis. Finally, we propose that SKP1 can serve as a potential biomarker in osteoporosis.
Collapse
Affiliation(s)
- Morteza Nakhaei Amroodi
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, department of orthopedic, school of medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Shayan Amiri
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, department of orthopedic, school of medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Khatere Mokhtari
- Department of Cellular and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Parnaz Mohseni
- Department of Pediatrics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Azadeh Pourmarjani
- Department of Pediatrics, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Behdokht Jamali
- Department of microbiology and genetics, kherad Institute of higher education, Busheher, lran
| | - Elaheh Mohandesi Khosroshahi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saba Asadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Pouria Tabrizian
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, department of orthopedic, school of medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Runlan Wan
- Department of Oncology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
2
|
Lunesu MF, Battacone G, Mellino MR, Carta S, Pulina G, Nudda A. The heavy suckling lamb of Sarda dairy sheep and its crossbreed with Dorper rams: Performance, meat quality and consumer perceptions. Meat Sci 2023; 204:109234. [PMID: 37295034 DOI: 10.1016/j.meatsci.2023.109234] [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: 03/09/2023] [Revised: 05/12/2023] [Accepted: 05/28/2023] [Indexed: 06/11/2023]
Abstract
The increase of meat production in dairy sheep farms, has been evaluated by the extension of the suckling period from the traditional 28 days to 75 days to obtain a new product, the heavy suckling lamb. Nineteen single-born Sarda (S) lambs (10 male, 9 female) and 20 single-born Dorper x Sarda (DS) lambs (9 male, 11 female), randomly selected from autumn lambing season, were fed exclusively with maternal milk until slaughtering at 20 ± 0.28 kg of body weight (BW, mean ± standard deviation, SD) and approximately 11 weeks of age. Body weight were recorded at birth and every 15 days until slaughter to estimate average daily gain (ADG). At slaughter, carcass measurements, pH and colour traits were recorded from the left side of the carcass. Proximate composition, fatty acid (FA) profile, cooking and drip losses were evaluated using the Longissimus thoracis et lumborum (LTL) muscle. In addition, Visual Panel Test (VPT) and Taste Panel Test (TPT) were performed. Experimental results evidenced that ADG did not differ between pure and crossbreed lambs and between sexes. The S lamb carcasses showed a higher fat content and rib fat thickness compared to that of crossbreed. No significant differences were found between genetic types and sex for colour and pH determinations, cooking and drip losses, whereas LTL fat of DS displayed a higher nutritional FA profile with higher content of 22:5n-3, 22:6n-3, branched-chain FA, and odd- and branched chain FA. No differences emerged during VPT and TPT, demonstrating that both DS and S lamb meats present no distinguishable visual and eating quality characteristics. The production of heavy suckling lambs from Sarda and Dorper crossbreed by the extension of suckling period appeared a promising strategy for producing meat of high quality, well appreciated by the consumers.
Collapse
Affiliation(s)
| | - Gianni Battacone
- Dipartimento di Agraria, University of Sassari, Viale Italia 39a, 07100 Sassari, Italy.
| | - Maria Rita Mellino
- Dipartimento di Agraria, University of Sassari, Viale Italia 39a, 07100 Sassari, Italy
| | - Silvia Carta
- Dipartimento di Agraria, University of Sassari, Viale Italia 39a, 07100 Sassari, Italy
| | - Giuseppe Pulina
- Dipartimento di Agraria, University of Sassari, Viale Italia 39a, 07100 Sassari, Italy
| | - Anna Nudda
- Dipartimento di Agraria, University of Sassari, Viale Italia 39a, 07100 Sassari, Italy.
| |
Collapse
|
3
|
Wang J, Xiao S, Cai Q, Miao J, Li J. Antioxidant Capacity and Protective Effects on H 2O 2-Induced Oxidative Damage in PC12 Cells of the Active Fraction of Brassica rapa L. Foods 2023; 12:2075. [PMID: 37238893 PMCID: PMC10217163 DOI: 10.3390/foods12102075] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/13/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023] Open
Abstract
Brassica rapa L. (BR), a traditional biennial herb belonging to the Brassica species of Brassicaceae, has been widely used for functions of anti-inflammatory, antitumor, antioxidation, antiaging, and regulation of immunity. In this study, antioxidant activity and protective effects on H2O2-induced oxidative damage in PC12 cells of the active fractions of BR were investigated in vitro. Among all active fractions, the ethyl acetate fraction of ethanol extract from BR (BREE-Ea) showed the strongest antioxidant activity. Additionally, it was noted that BREE-Ea and n-butyl alcohol fraction of ethanol extract from BR (BREE-Ba) both have protective effects in oxidatively damaged PC12 cells, while BREE-Ea displayed the best protective effect in all determined experimental doses. Furthermore, flow cytometry (DCFH-DA staining) analysis indicated that BREE-Ea could reduce the H2O2-induced apoptosis in PC12 cells by reducing the production of intracellular reactive oxygen species (ROS) and increasing enzymatic activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px). Moreover, BREE-Ea could decrease the malondialdehyde (MDA) content and reduce the release of extracellular lactic dehydrogenase (LDH) from H2O2-induced PC12 cells. All these results demonstrate that BREE-Ea has a good antioxidant capacity and protective effect on PC12 cells against apoptosis induced by H2O2 and that it can be used as a good edible antioxidant to improve the body's endogenous antioxidant defense.
Collapse
Affiliation(s)
- Jin Wang
- College of Life Science and Technology, Xinjiang University, Urumqi 830000, China; (J.W.); (S.X.); (Q.C.)
| | - Shuang Xiao
- College of Life Science and Technology, Xinjiang University, Urumqi 830000, China; (J.W.); (S.X.); (Q.C.)
| | - Qi Cai
- College of Life Science and Technology, Xinjiang University, Urumqi 830000, China; (J.W.); (S.X.); (Q.C.)
| | - Jing Miao
- Pharmaceutical Institute, Xinjiang University, Urumqi 830000, China
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Xinjiang University, Urumqi 830000, China
| | - Jinyao Li
- College of Life Science and Technology, Xinjiang University, Urumqi 830000, China; (J.W.); (S.X.); (Q.C.)
- Pharmaceutical Institute, Xinjiang University, Urumqi 830000, China
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Xinjiang University, Urumqi 830000, China
| |
Collapse
|
4
|
Wang S, Tang C, Chen J, Tang H, Zhang L, Tang G. Changes in Bone Marrow Fatty Acids Early after Ovariectomy-Induced Osteoporosis in Rats and Potential Functions. Metabolites 2022; 13:metabo13010036. [PMID: 36676961 PMCID: PMC9863616 DOI: 10.3390/metabo13010036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/11/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
The aim of this study was to investigate the changes in bone marrow fatty acids early after ovariectomy-induced osteoporosis in rats, and explore the potential function of the bone marrow fatty acids. Ninety-six female Sprague Dawley rats (12 weeks) were randomly divided into an ovariectomized (OVX) group and Sham group (N = 48/group) and received ovariectomy or Sham surgery, respectively. After 3, 5, 7,14, 21 and 28 days, eight rats in each group were sacrificed to detect the composition of bone marrow fatty acids by means of gas chromatography-mass spectrometry and evaluate the trabecular bone microarchitecture by means of microCT. Bone marrow rinsing fluid and serum were collected for the detection of nitric oxide synthase/nitric oxide (NOS/NO) and bone metabolism related parameters, respectively. Our results demonstrated that the bone microstructure was damaged significantly from 14 days after OVX surgery onwards. Sample clustering and group separation were observed between the OVX group and Sham group 3 and 14 days after surgery, which suggested the role of bone marrow fatty acids in the early stage of postmenopausal osteoporosis. Palmitoleate, myristate and arachidonate were found to play an important role in classification between the OVX group and Sham group on the 3rd day after surgery (VIP > 1, p < 0.05). Palmitoleate, myristate, alpha linolenate, stearate and eicosenoate were found to play an important role in classification between the OVX group and Sham group on the 14th day after surgery (VIP > 1, p < 0.05). The levels of myristate, palmitoleate, alpha linolenate and eicosenoate were significantly decreased in the OVX group, while the levels of arachidonate and stearate were significantly increased in OVX group (p < 0.05). Additionally, myristate, palmitoleate, alpha linoleate and eicosenoate were negatively correlated with C-terminal telopeptide of type 1 collagen (CTX-1, a bone resorption marker), while arachidonate was negative correlated with osteocalcin (OCN, a bone formation marker) (p < 0.05). A significant correlation was also found between eicosenoate and NOS (p < 0.05). Profound bone marrow fatty acids changes have taken place in the early stage of post-menopausal osteoporosis. They may affect bone formation though affecting the differentiation and function of osteoclasts or osteoblasts, respectively. The NOS/NO system may mediate the influence of eicosenoate on bone formation.
Collapse
Affiliation(s)
- Sizhu Wang
- Department of Radiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Cuisong Tang
- Department of Radiology, Clinical Medical College of Shanghai Tenth People’s Hospital of Nanjing Medical University, Shanghai 200072, China
| | - Jieying Chen
- Department of Radiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Huan Tang
- Department of Radiology, Huadong Hospital of Fudan University, Shanghai 200040, China
| | - Lin Zhang
- Department of Radiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
- Correspondence: (L.Z.); (G.T.)
| | - Guangyu Tang
- Department of Radiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
- Department of Radiology, Clinical Medical College of Shanghai Tenth People’s Hospital of Nanjing Medical University, Shanghai 200072, China
- Correspondence: (L.Z.); (G.T.)
| |
Collapse
|
5
|
Srivastava RK, Sapra L, Mishra PK. Osteometabolism: Metabolic Alterations in Bone Pathologies. Cells 2022; 11:3943. [PMID: 36497201 PMCID: PMC9735555 DOI: 10.3390/cells11233943] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/20/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
Renewing interest in the study of intermediate metabolism and cellular bioenergetics is brought on by the global increase in the prevalence of metabolic illnesses. Understanding of the mechanisms that integrate energy metabolism in the entire organism has significantly improved with the application of contemporary biochemical tools for quantifying the fuel substrate metabolism with cutting-edge mouse genetic procedures. Several unexpected findings in genetically altered mice have prompted research into the direction of intermediate metabolism of skeletal cells. These findings point to the possibility of novel endocrine connections through which bone cells can convey their energy status to other metabolic control centers. Understanding the expanded function of skeleton system has in turn inspired new lines of research aimed at characterizing the energy needs and bioenergetic characteristics of these bone cells. Bone-forming osteoblast and bone-resorbing osteoclast cells require a constant and large supply of energy substrates such as glucose, fatty acids, glutamine, etc., for their differentiation and functional activity. According to latest research, important developmental signaling pathways in bone cells are connected to bioenergetic programs, which may accommodate variations in energy requirements during their life cycle. The present review article provides a unique perspective of the past and present research in the metabolic characteristics of bone cells along with mechanisms governing energy substrate utilization and bioenergetics. In addition, we discussed the therapeutic inventions which are currently being utilized for the treatment and management of bone-related diseases such as osteoporosis, rheumatoid arthritis (RA), osteogenesis imperfecta (OIM), etc., by modulating the energetics of bone cells. We further emphasized on the role of GUT-associated metabolites (GAMs) such as short-chain fatty acids (SCFAs), medium-chain fatty acids (MCFAs), indole derivates, bile acids, etc., in regulating the energetics of bone cells and their plausible role in maintaining bone health. Emphasis is importantly placed on highlighting knowledge gaps in this novel field of skeletal biology, i.e., "Osteometabolism" (proposed by our group) that need to be further explored to characterize the physiological importance of skeletal cell bioenergetics in the context of human health and bone related metabolic diseases.
Collapse
Affiliation(s)
- Rupesh K. Srivastava
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Leena Sapra
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | | |
Collapse
|
6
|
Shang Q, Chen G, Zhang P, Zhao W, Chen H, Yu D, Yu F, Liu H, Zhang X, He J, Yu X, Zhang Z, Tan R, Wu Z, Tang J, Liang D, Shen G, Jiang X, Ren H. Myristic acid alleviates hippocampal aging correlated with GABAergic signaling. Front Nutr 2022; 9:907526. [PMID: 36159502 PMCID: PMC9493098 DOI: 10.3389/fnut.2022.907526] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
Previous studies have shown that myristic acid (MA), a saturated fatty acid, could promote the proliferation and differentiation of neural stem cells in vitro. However, the effect of MA on hippocampal neurons aging has not been reported in vivo. Here we employed 22-month-old naturally aged C57BL/6 mice to evaluate the effect and mechanism of MA on hippocampal aging. First, we examined a decreased exploration and spatial memory ability in aging mice using the open field test and Morris water maze. Consistently, aging mice showed degenerative hippocampal histomorphology by H&E and Nissl staining. In terms of mechanism, imbalance of GABRB2 and GABRA2 expression in aging mice might be involved in hippocampus aging by mRNA high throughput sequencing (mRNA-seq) and immunohistochemistry (IHC) validation. Then, we revealed that MA alleviated the damage of exploration and spatial memory ability and ameliorated degeneration and aging of hippocampal neurons. Meanwhile, MA downregulated GABRB2 and upregulated GABRA2 expression, indicating MA might alleviate hippocampal aging correlated with GABAergic signaling. In conclusion, our findings revealed MA alleviated hippocampal aging correlated with GABAergic signaling, which might provide insight into the treatment of aging-associated diseases.
Collapse
Affiliation(s)
- Qi Shang
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guifeng Chen
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peng Zhang
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenhua Zhao
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Honglin Chen
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Die Yu
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fuyong Yu
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huiwen Liu
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuelai Zhang
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiahui He
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiang Yu
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhida Zhang
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Riwei Tan
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zixian Wu
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingjing Tang
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - De Liang
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Gengyang Shen
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Nanshan Hospital, The First Affiliated Hospital of Guangzhou University of Chinese Medicine (Shenzhen Nanshan Hospital of Chinese Medicine), Guangzhou, China
- Gengyang Shen
| | - Xiaobing Jiang
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Xiaobing Jiang
| | - Hui Ren
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Hui Ren
| |
Collapse
|
7
|
Dogan S, Kimyon G, Ozkan H, Kacmaz F, Camdeviren B, Karaaslan I. TNF-alpha, IL-6, IL-10 and fatty acids in rheumatoid arthritis patients receiving cDMARD and bDMARD therapy. Clin Rheumatol 2022; 41:2341-2349. [PMID: 35467309 DOI: 10.1007/s10067-022-06180-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 04/07/2022] [Accepted: 04/13/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVE The present study aimed to examine the effects of cDMARD and bDMARD therapy on both gene expressions and protein levels of TNF-α, IL-6, IL-10 and fatty acid levels in patients with RA. METHOD Plasma TNF-α, IL-6, and IL-10 levels were examined by the ELISA method, while TNF-α, IL-6, and IL-10 gene expression levels were examined by RT-qPCR, and fatty acid levels were examined by GC/MS. RESULTS IL-10 gene expression levels significantly increased in RA patients receiving cDMARD treatment compared to those of the control group. Also, eicosadienoic acid, myristoleic acid and capric acid levels were significantly lower in the patient groups compared to those in the control group. CONCLUSION The drugs used in the treatment of RA had no effect on the fatty acid levels whereas had effects on the mRNA and protein levels of the target cytokines.
Collapse
Affiliation(s)
- Serdar Dogan
- Department of Biochemistry, Faculty of Medicine, Hatay Mustafa Kemal University, Antakya, Hatay, 31060, Turkey.
| | - Gezmis Kimyon
- Department of Rheumatology, Faculty of Medicine, Hatay Mustafa Kemal University, Antakya, Hatay, 31060, Turkey
| | - Huseyin Ozkan
- Department of Genetics, Faculty of Veterinary Medicine, Hatay Mustafa Kemal University, Antakya, Hatay, 31060, Turkey
| | - Filiz Kacmaz
- Department of Molecular Biochemistry and Genetics, Health Science Institute, Hatay Mustafa Kemal University, Antakya, Hatay, 31060, Turkey
| | - Baran Camdeviren
- Department of Molecular Biochemistry and Genetics, Health Science Institute, Hatay Mustafa Kemal University, Antakya, Hatay, 31060, Turkey
| | - Irem Karaaslan
- Research and Application Center for Technology and Research and Development, Hatay Mustafa Kemal University, Antakya, Hatay, 31060, Turkey
| |
Collapse
|
8
|
Gong R, Xiao HM, Zhang YH, Zhao Q, Su KJ, Lin X, Mo CL, Zhang Q, Du YT, Lyu FY, Chen YC, Peng C, Liu HM, Hu SD, Pan DY, Chen Z, Li ZF, Zhou R, Wang XF, Lu JM, Ao ZX, Song YQ, Weng CY, Tian Q, Schiller MR, Papasian CJ, Brotto M, Shen H, Shen J, Deng HW. Identification and Functional Characterization of Metabolites for Bone Mass in Peri- and Postmenopausal Chinese Women. J Clin Endocrinol Metab 2021; 106:e3159-e3177. [PMID: 33693744 PMCID: PMC8277206 DOI: 10.1210/clinem/dgab146] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Indexed: 12/14/2022]
Abstract
CONTEXT Although metabolic profiles appear to play an important role in menopausal bone loss, the functional mechanisms by which metabolites influence bone mineral density (BMD) during menopause are largely unknown. OBJECTIVE We aimed to systematically identify metabolites associated with BMD variation and their potential functional mechanisms in peri- and postmenopausal women. DESIGN AND METHODS We performed serum metabolomic profiling and whole-genome sequencing for 517 perimenopausal (16%) and early postmenopausal (84%) women aged 41 to 64 years in this cross-sectional study. Partial least squares regression and general linear regression analysis were applied to identify BMD-associated metabolites, and weighted gene co-expression network analysis was performed to construct co-functional metabolite modules. Furthermore, we performed Mendelian randomization analysis to identify causal relationships between BMD-associated metabolites and BMD variation. Finally, we explored the effects of a novel prominent BMD-associated metabolite on bone metabolism through both in vivo/in vitro experiments. RESULTS Twenty metabolites and a co-functional metabolite module (consisting of fatty acids) were significantly associated with BMD variation. We found dodecanoic acid (DA), within the identified module causally decreased total hip BMD. Subsequently, the in vivo experiments might support that dietary supplementation with DA could promote bone loss, as well as increase the osteoblast and osteoclast numbers in normal/ovariectomized mice. Dodecanoic acid treatment differentially promoted osteoblast and osteoclast differentiation, especially for osteoclast differentiation at higher concentrations in vitro (eg,10, 100 μM). CONCLUSIONS This study sheds light on metabolomic profiles associated with postmenopausal osteoporosis risk, highlighting the potential importance of fatty acids, as exemplified by DA, in regulating BMD.
Collapse
Affiliation(s)
- Rui Gong
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, USA
- Cadre Ward Endocrinology Department, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Hong-Mei Xiao
- Center of System Biology, Data Information and Reproductive Health, School of Basic Medical Science, Central South University, Changsha, China
| | - Yin-Hua Zhang
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Qi Zhao
- Department of Preventive Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Kuan-Jui Su
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Xu Lin
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Cheng-Lin Mo
- Bone-Muscle Research Center, College of Nursing and Health Innovation, The University of Texas-Arlington, Arlington, TX, USA
| | - Qiang Zhang
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, USA
- School of Nursing and Health, Zhengzhou University, Zhengzhou, China
| | - Ya-Ting Du
- Bone-Muscle Research Center, College of Nursing and Health Innovation, The University of Texas-Arlington, Arlington, TX, USA
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Feng-Ye Lyu
- LC-Bio Technologies (Hangzhou) CO.LTD, Hangzhou, China
| | - Yuan-Cheng Chen
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Cheng Peng
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Hui-Min Liu
- Center of System Biology, Data Information and Reproductive Health, School of Basic Medical Science, Central South University, Changsha, China
| | - Shi-Di Hu
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Dao-Yan Pan
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zhi Chen
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zhang-Fang Li
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Rou Zhou
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xia-Fang Wang
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Jun-Min Lu
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zeng-Xin Ao
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Yu-Qian Song
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Chan-Yan Weng
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Qing Tian
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Martin R Schiller
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Christopher J Papasian
- Department of Biomedical Sciences, University of Missouri-Kansas City, School of Medicine, Kansas City, MO, USA
| | - Marco Brotto
- Bone-Muscle Research Center, College of Nursing and Health Innovation, The University of Texas-Arlington, Arlington, TX, USA
| | - Hui Shen
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Jie Shen
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Shunde Hospital of Southern Medical University (The First People’s Hospital of Shunde), Foshan, China
- Jie Shen, No.1 of Jiazi Road, Lunjiao, Shunde District, Foshan 528000, Guangdong, China.
| | - Hong-Wen Deng
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, USA
- Center of System Biology, Data Information and Reproductive Health, School of Basic Medical Science, Central South University, Changsha, China
- Correspondence: Hong-Wen Deng, 1440 Canal St, Suite 2001, New Orleans, LA 70112, USA.
| |
Collapse
|
9
|
Choi YK, Kang JI, Hyun JW, Koh YS, Kang JH, Hyun CG, Yoon KS, Lee KS, Lee CM, Kim TY, Yoo ES, Kang HK. Myristoleic Acid Promotes Anagen Signaling by Autophagy through Activating Wnt/β-Catenin and ERK Pathways in Dermal Papilla Cells. Biomol Ther (Seoul) 2021; 29:211-219. [PMID: 33518533 PMCID: PMC7921852 DOI: 10.4062/biomolther.2020.169] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/11/2020] [Accepted: 01/06/2021] [Indexed: 02/06/2023] Open
Abstract
Alopecia is a distressing condition caused by the dysregulation of anagen, catagen, and telogen in the hair cycle. Dermal papilla cells (DPCs) regulate the hair cycle and play important roles in hair growth and regeneration. Myristoleic acid (MA) increases Wnt reporter activity in DPCs. However, the action mechanisms of MA on the stimulation of anagen signaling in DPCs is not known. In this study, we evaluated the effects of MA on anagen-activating signaling pathways in DPCs. MA significantly increased DPC proliferation and stimulated the G2/M phase, accompanied by increasing cyclin A, Cdc2, and cyclin B1. To elucidate the mechanism by which MA promotes DPC proliferation, we evaluated the effect of MA on autophagy and intracellular pathways. MA induced autophagosome formation by decreasing the levels of the phospho-mammalian target of rapamycin (phospho-mTOR) and increasing autophagy-related 7 (Atg7) and microtubule-associated protein 1A/1B-light chain 3II (LC3II). MA also increased the phosphorylation levels of Wnt/β-catenin proteins, such as GSK3β (Ser9) and β-catenin (Ser552 and Ser675). Treatment with XAV939, an inhibitor of the Wnt/β-catenin pathway, attenuated the MA-induced increase in β-catenin nuclear translocation. Moreover, XAV939 reduced MA-induced effects on cell cycle progression, autophagy, and DPC proliferation. On the other hand, MA increased the levels of phospho (Thr202/Tyr204)-extracellular signal regulated kinases (ERK). MA-induced ERK phosphorylation led to changes in the expression levels of Cdc2, Atg7 and LC3II, as well as DPC proliferation. Our results suggest that MA promotes anagen signaling via autophagy and cell cycle progression by activating the Wnt/β-catenin and ERK pathways in DPCs.
Collapse
Affiliation(s)
- Youn Kyung Choi
- Department of Medicine, School of Medicine, Jeju National University, Jeju 63243, Republic of Korea.,Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Jung-Il Kang
- Department of Medicine, School of Medicine, Jeju National University, Jeju 63243, Republic of Korea.,Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Jin Won Hyun
- Department of Medicine, School of Medicine, Jeju National University, Jeju 63243, Republic of Korea.,Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Young Sang Koh
- Department of Medicine, School of Medicine, Jeju National University, Jeju 63243, Republic of Korea.,Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Ji-Hoon Kang
- Department of Medicine, School of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Chang-Gu Hyun
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea.,Department of Chemistry & Cosmetics, Jeju National University, Jeju 63243, Republic of Korea
| | - Kyung-Sup Yoon
- Department of Chemistry & Cosmetics, Jeju National University, Jeju 63243, Republic of Korea
| | - Kwang Sik Lee
- DSongpa R&D Center, Coreana Cosmetic Co., Ltd, Cheonan 31041, Republic of Korea
| | - Chun Mong Lee
- DSongpa R&D Center, Coreana Cosmetic Co., Ltd, Cheonan 31041, Republic of Korea
| | - Tae Yang Kim
- DSongpa R&D Center, Coreana Cosmetic Co., Ltd, Cheonan 31041, Republic of Korea
| | - Eun-Sook Yoo
- Department of Medicine, School of Medicine, Jeju National University, Jeju 63243, Republic of Korea.,Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Hee-Kyoung Kang
- Department of Medicine, School of Medicine, Jeju National University, Jeju 63243, Republic of Korea.,Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| |
Collapse
|
10
|
Lee JW, Lee IH, Iimura T, Kong SW. Two macrophages, osteoclasts and microglia: from development to pleiotropy. Bone Res 2021; 9:11. [PMID: 33568650 PMCID: PMC7875961 DOI: 10.1038/s41413-020-00134-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 12/11/2022] Open
Abstract
Tissue-resident macrophages are highly specialized to their tissue-specific microenvironments, activated by various inflammatory signals and modulated by genetic and environmental factors. Osteoclasts and microglia are distinct tissue-resident cells of the macrophage lineage in bone and brain that are responsible for pathological changes in osteoporosis and Alzheimer’s disease (AD), respectively. Osteoporosis is more frequently observed in individuals with AD compared to the prevalence in general population. Diagnosis of AD is often delayed until underlying pathophysiological changes progress and cause irreversible damages in structure and function of brain. As such earlier diagnosis and intervention of individuals at higher risk would be indispensable to modify clinical courses. Pleiotropy is the phenomenon that a genetic variant affects multiple traits and the genetic correlation between two traits could suggest a shared molecular mechanism. In this review, we discuss that the Pyk2-mediated actin polymerization pathway in osteoclasts and microglia in bone and brain, respectively, is the horizontal pleiotropic mediator of shared risk factors for osteoporosis and AD.
Collapse
Affiliation(s)
- Ji-Won Lee
- Department of Nephrology, Transplant Research Program, Boston Children's Hospital, Boston, MA, 02115, USA.,Department of Pharmacology, Graduate School of Dental Medicine, Hokkaido University, Sapporo, 060-8586, Japan
| | - In-Hee Lee
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Tadahiro Iimura
- Department of Pharmacology, Graduate School of Dental Medicine, Hokkaido University, Sapporo, 060-8586, Japan
| | - Sek Won Kong
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, 02115, USA. .,Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
11
|
Regulation of Osteoclast Differentiation and Activity by Lipid Metabolism. Cells 2021; 10:cells10010089. [PMID: 33430327 PMCID: PMC7825801 DOI: 10.3390/cells10010089] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/02/2021] [Accepted: 01/05/2021] [Indexed: 02/07/2023] Open
Abstract
Bone is a dynamic tissue and is constantly being remodeled by bone cells. Metabolic reprogramming plays a critical role in the activation of these bone cells and skeletal metabolism, which fulfills the energy demand for bone remodeling. Among various metabolic pathways, the importance of lipid metabolism in bone cells has long been appreciated. More recent studies also establish the link between bone loss and lipid-altering conditions—such as atherosclerotic vascular disease, hyperlipidemia, and obesity—and uncover the detrimental effect of fat accumulation on skeletal homeostasis and increased risk of fracture. Targeting lipid metabolism with statin, a lipid-lowering drug, has been shown to improve bone density and quality in metabolic bone diseases. However, the molecular mechanisms of lipid-mediated regulation in osteoclasts are not completely understood. Thus, a better understanding of lipid metabolism in osteoclasts can be used to harness bone cell activity to treat pathological bone disorders. This review summarizes the recent developments of the contribution of lipid metabolism to the function and phenotype of osteoclasts.
Collapse
|
12
|
Kong L, Wang B, Yang X, He B, Hao D, Yan L. Integrin-associated molecules and signalling cross talking in osteoclast cytoskeleton regulation. J Cell Mol Med 2020; 24:3271-3281. [PMID: 32045092 PMCID: PMC7131929 DOI: 10.1111/jcmm.15052] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 01/22/2020] [Accepted: 01/27/2020] [Indexed: 12/30/2022] Open
Abstract
In the ageing skeleton, the balance of bone reconstruction could commonly be broken by the increasing of bone resorption and decreasing of bone formation. Consequently, the bone resorption gradually occupies a dominant status. During this imbalance process, osteoclast is unique cell linage act the bone resorptive biological activity, which is a highly differentiated ultimate cell derived from monocyte/macrophage. The erosive function of osteoclasts is that they have to adhere the bone matrix and migrate along it, in which adhesive cytoskeleton recombination of osteoclast is essential. In that, the podosome is a membrane binding microdomain organelle, based on dynamic actin, which forms a cytoskeleton superstructure connected with the plasma membrane. Otherwise, as the main adhesive protein, integrin regulates the formation of podosome and cytoskeleton, which collaborates with the various molecules including: c-Cbl, p130Cas , c-Src and Pyk2, through several signalling cascades cross talking, including: M-CSF and RANKL. In our current study, we discuss the role of integrin and associated molecules in osteoclastogenesis cytoskeletal, especially podosomes, regulation and relevant signalling cascades cross talking.
Collapse
Affiliation(s)
- Lingbo Kong
- Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, China
| | - Biao Wang
- Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, China
| | - Xiaobin Yang
- Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, China
| | - Baorong He
- Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, China
| | - Dingjun Hao
- Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, China
| | - Liang Yan
- Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, China
| |
Collapse
|
13
|
Bao M, Zhang K, Wei Y, Hua W, Gao Y, Li X, Ye L. Therapeutic potentials and modulatory mechanisms of fatty acids in bone. Cell Prolif 2020; 53:e12735. [PMID: 31797479 PMCID: PMC7046483 DOI: 10.1111/cpr.12735] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 11/02/2019] [Accepted: 11/05/2019] [Indexed: 02/05/2023] Open
Abstract
Bone metabolism is a lifelong process that includes bone formation and resorption. Osteoblasts and osteoclasts are the predominant cell types associated with bone metabolism, which is facilitated by other cells such as bone marrow mesenchymal stem cells (BMMSCs), osteocytes and chondrocytes. As an important component in our daily diet, fatty acids are mainly categorized as long-chain fatty acids including polyunsaturated fatty acids (LCPUFAs), monounsaturated fatty acids (LCMUFAs), saturated fatty acids (LCSFAs), medium-/short-chain fatty acids (MCFAs/SCFAs) as well as their metabolites. Fatty acids are closely associated with bone metabolism and associated bone disorders. In this review, we summarized the important roles and potential therapeutic implications of fatty acids in multiple bone disorders, reviewed the diverse range of critical effects displayed by fatty acids on bone metabolism, and elucidated their modulatory roles and mechanisms on specific bone cell types. The evidence supporting close implications of fatty acids in bone metabolism and disorders suggests fatty acids as potential therapeutic and nutritional agents for the treatment and prevention of metabolic bone diseases.
Collapse
Affiliation(s)
- Minyue Bao
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Kaiwen Zhang
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Yangyini Wei
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Weihan Hua
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Yanzi Gao
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Xin Li
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Ling Ye
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesDepartment of Cariology and EndodonticsWest China Hospital of StomatologySichuan UniversityChengduChina
| |
Collapse
|
14
|
Quach D, Parameswaran N, McCabe L, Britton RA. Characterizing how probiotic Lactobacillus reuteri 6475 and lactobacillic acid mediate suppression of osteoclast differentiation. Bone Rep 2019; 11:100227. [PMID: 31763377 PMCID: PMC6864341 DOI: 10.1016/j.bonr.2019.100227] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 10/07/2019] [Accepted: 10/16/2019] [Indexed: 12/18/2022] Open
Abstract
Osteoporosis is a disease that impacts over 200 million people worldwide. The probiotic bacterium Lactobacillus reuteri (L. reuteri) has been shown to prevent bone loss during estrogen deficiency. Lactobacillic acid is important for L. reuteri-induced suppression of in vitro osteoclastogenesis. Osteoclastogenesis was inhibited by L. reuteri and lactobacillic acid via GPR120 signaling.
Osteoporosis is a disease that impacts over 200 million people worldwide. Taking into consideration the side effects stemming from medications used to treat this illness, investigators have increased their efforts to develop novel therapeutics for osteoporosis. In a previous study, we demonstrated that ovariectomy-induced bone loss in mice was prevented by treatment with the probiotic bacterium Lactobacillus reuteri 6475 (L. reuteri), an effect that correlated with reduced osteoclastogenesis in the bone marrow of L. reuteri treated mice. We also demonstrated that L. reuteri directly inhibited osteoclastogenesis in vitro. To better understand how L. reuteri impacts osteoclast formation, we used additional in vitro analyses to identify that conditioned supernatant from L. reuteri inhibited osteoclastogenesis at the intermediate stage of fused polykaryons. To elucidate the effect of L. reuteri treatment on host cell physiology, we performed RNAseq at multiple time points during in vitro osteoclastogenesis and established that L. reuteri downregulated several KEGG pathways including osteoclast differentiation as well as TNF-α, NF-κB, and MAP kinase signaling. These results were consistent with Western Blot data demonstrating that NF-κB and p38 activation were decreased by L. reuteri treatment. We further identified that lactobacillic acid (LA), a cyclopropane fatty acid produced by L. reuteri, contributed significantly to the suppression of osteoclastogenesis. Additionally, we demonstrated that L. reuteri is signaling through the long chain fatty acid receptor, GPR120, to impact osteoclastogenesis. Overall, these studies provide both bacterial and host mechanisms by which L. reuteri impacts osteoclastogenesis and suggest that long chain fatty acid receptors could be targets for preventing osteoclastogenesis.
Collapse
Affiliation(s)
- Darin Quach
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | | | - Laura McCabe
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Robert A. Britton
- Baylor College of Medicine, Department of Molecular Virology and Microbiology, Alkek Center for Metagenomics and Microbiome Research, Houston, TX, USA
- Corresponding author.
| |
Collapse
|
15
|
Liu C, Wu H, Liu S, Chai S, Meng Q, Zhou Z. Dynamic Alterations in Yak Rumen Bacteria Community and Metabolome Characteristics in Response to Feed Type. Front Microbiol 2019; 10:1116. [PMID: 31191470 PMCID: PMC6538947 DOI: 10.3389/fmicb.2019.01116] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 05/02/2019] [Indexed: 01/07/2023] Open
Abstract
Current knowledge about the relationships between ruminal bacterial communities and metabolite profiles in the yak rumen is limited. This is due to differences in the nutritional and metabolic features between yak and other ordinary cattle combined with difficulties associated with farm-based research and a lack of technical guidance. A comprehensive analysis of the composition and alterations in ruminal metabolites is required to advance the development of modern yak husbandry. In the current study, we characterized the effect of feed type on the ruminal fluid microbiota and metabolites in yak using 16S rRNA gene sequencing and liquid chromatography-mass spectrometry (LC-MS). Bacteroidetes and Firmicutes were the predominant bacterial phyla in the yak rumen. At the genus level, the relative abundance of Bacteroidales BS11 gut group, Prevotellaceae UCG-003, Ruminococcaceae UCG-011, Bacteroidales RF16 group and Ruminococcaceae UCG-010 was significantly (P < 0.01) higher in the forage group compared to that in the concentrate group, while the concentrate group harbored higher proportions of Bacteroidales S24-7 group, Ruminococcaceae NK4A214, Succiniclasticum and Ruminococcus 2. Yak rumen metabolomics analysis combined with enrichment analysis revealed that feed type altered the concentrations of ruminal metabolites as well as the metabolic pattern, and significantly (P < 0.01) affected the concentrations of ruminal metabolites involved in protein digestion and absorption (e.g., L-arginine, ornithine, L-threonine, L-proline and β-alanine), purine metabolism (e.g., xanthine, hypoxanthine, deoxyadenosine and deoxyadenosine monophosphate) and fatty acid biosynthesis (e.g., stearic acid, myristic acid and arachidonic acid). Correlation analysis of the association of microorganisms with metabolite features provides us with a comprehensive understanding of the composition and function of microbial communities. Associations between utilization or production were widely identified between affected microbiota and certain metabolites, and these findings will contribute to the direction of future research in yak.
Collapse
Affiliation(s)
- Chang Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Hao Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shujie Liu
- Qinghai Academy of Animal and Veterinary Sciences, Qinghai University, Xining, China
| | - Shatuo Chai
- Qinghai Academy of Animal and Veterinary Sciences, Qinghai University, Xining, China
| | - Qingxiang Meng
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhenming Zhou
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China,*Correspondence: Zhenming Zhou
| |
Collapse
|
16
|
Zhao L, Li M, Sun H. Effects of dietary calcium to available phosphorus ratios on bone metabolism and osteoclast activity of the OPG /RANK/RANKL signalling pathway in piglets. J Anim Physiol Anim Nutr (Berl) 2019; 103:1224-1232. [PMID: 31062421 DOI: 10.1111/jpn.13115] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/19/2019] [Accepted: 04/07/2019] [Indexed: 12/12/2022]
Abstract
Hydroxyapatite, a mineral form of calcium (Ca) and phosphorus (P) that gives bones their rigidity, is the major and essential component of bones and teeth in the human and animal body. A suitable ratio of Ca and P is vital for bone growth. The aim of this study was to explore the effects of dietary calcium to available phosphorus ratios (Ca/AP) on bone metabolism and osteoclast activity of the osteoprotegerin (OPG)/receptor activator of nuclear factor kappa B ligand (RANKL) signalling pathway in piglets. At days 15 and 29, the piglets were assessed for growth performance, blood indicators, cytokines and the OPG/RANK/RANKL signalling pathway. Our results showed that piglets fed a dietary Ca/AP ratio of 2:1 increases growth performance and regulates blood indicators and cytokines (parathyroid hormone (PTH), calcitonin (CT), vitamin D3 (VD3 ), insulin-like growth factor-1 (IGF-1), transforming growth factor-β (TGF-β), interleukin-1 (IL-1), interleukin-6 (IL-6), carboxyterminal propeptide of type I procollagen (PICP), tartrate-resistant acid phosphatase (TRACP), alkaline phosphatase (ALP) and osteocalcin (OCN) content). We also demonstrated that this ratio affects hormone secretion and further bone metabolism through the OPG/RANK/RANKL signalling pathway of osteoclasts. These results indicate that a suitable dietary Ca/AP ratio is vital for bone growth and reduce the incidence of bone diseases such as osteoporosis, providing a practical basis for the raising of piglets.
Collapse
Affiliation(s)
- Lei Zhao
- College of Animal Science and Technology, Jilin Agriculture University, Changchun, China.,Ministry of education laboratory of animal production and quality security, Jilin Agricultural University, Changchun, China.,Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Muyang Li
- College of Animal Science and Technology, Jilin Agriculture University, Changchun, China.,Ministry of education laboratory of animal production and quality security, Jilin Agricultural University, Changchun, China.,Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Hui Sun
- College of Animal Science and Technology, Jilin Agriculture University, Changchun, China.,Ministry of education laboratory of animal production and quality security, Jilin Agricultural University, Changchun, China.,Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| |
Collapse
|
17
|
Shi L, Han B, Liu L, Lv X, Ma Z, Li C, Xu L, Li Y, Zhao F, Yang Y, Sun D. Determination of Genetic Effects of LIPK and LIPJ Genes on Milk Fatty Acids in Dairy Cattle. Genes (Basel) 2019; 10:genes10020086. [PMID: 30696079 PMCID: PMC6409763 DOI: 10.3390/genes10020086] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 01/13/2019] [Accepted: 01/14/2019] [Indexed: 12/30/2022] Open
Abstract
In our previous genome-wide association study (GWAS) on milk fatty acids (FAs) in Chinese Holstein, we discovered 83 genome-wide significant single nucleotide polymorphisms (SNPs) associated with milk FAs. Two of them were close to lipase family member K (LIPK) and lipase family member J (LIPJ), respectively. Hence, this study is a follow-up to verify whether the LIPK and LIPJ have significant genetic effects on milk FAs in dairy cattle. By re-sequencing the entire exons, and 3 kb of 5′ and 3′ flanking regions, two and seven SNPs were identified in LIPK and LIPJ, respectively, including a novel SNP, ss158213049726. With the Haploview 4.1 software, we found that five of the SNPs in LIPJ formed a haplotype block (D′ = 0.96 ~ 1.00). Single-locus association analyses revealed that each SNP in LIPK and LIPJ was significantly associated with at least one milk FA (p = < 1.00 × 10−4 ~ 4.88 × 10−2), and the haplotype-based association analyses showed significant genetic effects on nine milk FAs (p = < 1.00 × 10−4 ~ 3.98 × 10−2). Out of these SNPs, the missense mutation in LIPK gene, rs42774527, could change the protein secondary structure and function predicted by SOPMA, SIFT, and PROVEAN softwares. With the Genomatix software, we predicted that two SNPs, rs110322221 in LIPK and rs211373799 in LIPJ, altered the transcription factors binding sites (TFBSs), indicating their potential regulation on promoter activity of the genes. Furthermore, we found that both LIPK and LIPJ had relatively high expressions in the mammary gland. In conclusion, our research is the first to demonstrate that LIPK and LIPJ genes have significant associations with milk FAs, and the identified SNPs might be served as genetic markers to optimize breeding programs for milk FAs in dairy cattle. This research deserves in-depth verification.
Collapse
Affiliation(s)
- Lijun Shi
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing 100193, China.
| | - Bo Han
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing 100193, China.
| | - Lin Liu
- Beijing Dairy Cattle Center, Qinghe'nanzhen Deshengmenwai Street, Chaoyang District, Beijing 100192, China.
| | - Xiaoqing Lv
- Beijing Dairy Cattle Center, Qinghe'nanzhen Deshengmenwai Street, Chaoyang District, Beijing 100192, China.
| | - Zhu Ma
- Beijing Dairy Cattle Center, Qinghe'nanzhen Deshengmenwai Street, Chaoyang District, Beijing 100192, China.
| | - Cong Li
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing 100193, China.
| | - Lingna Xu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing 100193, China.
| | - Yanhua Li
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing 100193, China.
- Beijing Dairy Cattle Center, Qinghe'nanzhen Deshengmenwai Street, Chaoyang District, Beijing 100192, China.
| | - Feng Zhao
- Beijing Dairy Cattle Center, Qinghe'nanzhen Deshengmenwai Street, Chaoyang District, Beijing 100192, China.
| | - Yuze Yang
- Beijing General Station of Animal Husbandry, N0.96 Huizhongsi, Yayun Village, Chaoyang District, Beijing, 100101, China.
| | - Dongxiao Sun
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
18
|
An J, Hao D, Zhang Q, Chen B, Zhang R, Wang Y, Yang H. Natural products for treatment of bone erosive diseases: The effects and mechanisms on inhibiting osteoclastogenesis and bone resorption. Int Immunopharmacol 2016; 36:118-131. [PMID: 27131574 DOI: 10.1016/j.intimp.2016.04.024] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 03/28/2016] [Accepted: 04/18/2016] [Indexed: 01/13/2023]
Abstract
Excessive bone resorption plays a central role on the development of bone erosive diseases, including osteoporosis, rheumatoid arthritis, and periodontitis. Osteoclasts, bone-resorbing multinucleated cells, are differentiated from hemopoietic progenitors of the monocyte/macrophage lineage. Regulation of osteoclast differentiation is considered an effective therapeutic target to the treatment of pathological bone loss. Natural plant-derived products, with potential therapeutic and preventive activities against bone-lytic diseases, have received increasing attention in recent years because of their whole regulative effects and specific pharmacological activities, which are more suitable for long-term use than chemically synthesized medicines. In this review, we summarized the detailed research progress on the active compounds derived from medical plants with potential anti-resorptive effects and their molecular mechanisms on inhibiting osteoclast formation and function. The active ingredients derived from natural plants that are efficacious in suppressing osteoclastogenesis and bone resorption include flavonoids, terpenoids (sesquiterpenoids, diterpenoids, triterpenoids), glycosides, lignans, coumarins, alkaloids, polyphenols, limonoids, quinones and others (steroid, oxoxishhone, fatty acid). Studies have shown that above natural products exert the inhibitory effects via regulating many factors involved in the process of osteoclast differentiation and bone resorption, including the essential cytokines (RANKL, M-CSF), transcription factors (NFATc1, c-Fos), signaling pathways (NF-κB, MAPKs, Src/PI3K/Akt, the calcium ion signaling), osteoclast-specific genes (TRAP, CTSK, MMP-9, integrin β3, OSCAR, DC-STAMP, Atp6v0d2) and local factors (ROS, LPS, NO). The development of osteoclast-targeting natural products is of great value for the prevention or treatment of bone diseases and for bone regenerative medicine.
Collapse
Affiliation(s)
- Jing An
- Translational Medicine Centre, Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710054, China
| | - Dingjun Hao
- Translational Medicine Centre, Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710054, China
| | - Qian Zhang
- Translational Medicine Centre, Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710054, China
| | - Bo Chen
- Translational Medicine Centre, Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710054, China
| | - Rui Zhang
- Translational Medicine Centre, Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710054, China
| | - Yi Wang
- Translational Medicine Centre, Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710054, China
| | - Hao Yang
- Translational Medicine Centre, Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710054, China.
| |
Collapse
|