1
|
Bauer C, Smith C, Vogrin S, Palmer AS, Woessner M, Landen S, Jacques M, Byrnes E, Eynon N, Sim M, Lewis JR, Levinger I. Circulating lipocalin-2 across the adult lifespan. JBMR Plus 2025; 9:ziae162. [PMID: 39830147 PMCID: PMC11739805 DOI: 10.1093/jbmrpl/ziae162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/27/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
Lipocalin-2 (LCN2), a hormone produced by adipocytes, osteoblasts, and renal tubular cells, is implicated in age-related diseases, including cardio-metabolic disease. To understand the role LCN2 may play in pathological states, we first need to elucidate the relationship between circulating LCN2 with indices of cardio-metabolic health during "normal" aging. This study examined the relationship between serum levels of LCN2, age, and cardio-metabolic measures across the adult lifespan in males and females. We conducted a pooled cohort analysis including 124 community-dwelling males (n = 52) and females (n = 72) (age 20-87 yr, median BMI 25.92 [23.04, 29.81] kg/m2). Serum LCN2 was analyzed using a two-step chemiluminescent microparticle monoclonal immunoassay. The relationship between LCN2 and age was evaluated by linear regression and cubic spline. Simple linear regressions were performed to investigate the relationship between LCN2 and the following variables: BMI, VO2peak, serum glucose, and body composition (DXA). For every 1 yr increase in age, LCN2 levels were 0.26 mg/L higher (P = .007, 95% CI [0.07, 0.45]). Each 1 unit increase in BMI (kg/m2) was associated with 0.88 mg/L higher LCN2 levels (P = .027, [0.10, 1.66]) and each 1 unit increase in VO2peak (mL/kg/min) was associated with 0.38 mg/L lower LCN2 (p = .003, [-0.63, -0.13]).There was no significant relationship between LCN2 and sex, glucose levels or body composition (all p > .05). LCN2 increased linearly across the adult lifespan while it decreased as fitness level increased. Future research should build on these findings to determine whether LCN2 can be used as a biomarker for chronic disease and if exercise can mitigate age-related disease associated with LCN2 changes.
Collapse
Affiliation(s)
- Carlie Bauer
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3011, Australia
- Australian Institute for Musculoskeletal Science, Victoria University, University of Melbourne, St Albans, VIC 3021, Australia
| | - Cassandra Smith
- Nutrition & Health Innovation Research Institute, Edith Cowan University, Joondalup, WA 6027, Australia
- Medical School, The University of Western Australia, Perth, WA 6009, Australia
| | - Sara Vogrin
- Australian Institute for Musculoskeletal Science, Victoria University, University of Melbourne, St Albans, VIC 3021, Australia
- Department of Medicine, Western Health, The University of Melbourne, St Albans, VIC 3021, Australia
- Department of Medicine, St Vincent’s Hospital Melbourne, The University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Andrew S Palmer
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3011, Australia
| | - Mary Woessner
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3011, Australia
| | - Shanie Landen
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
| | - Macsue Jacques
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3011, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3168, Australia
| | | | - Nir Eynon
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3168, Australia
| | - Marc Sim
- Nutrition & Health Innovation Research Institute, Edith Cowan University, Joondalup, WA 6027, Australia
- Medical School, The University of Western Australia, Perth, WA 6009, Australia
| | - Joshua R Lewis
- Nutrition & Health Innovation Research Institute, Edith Cowan University, Joondalup, WA 6027, Australia
- Medical School, The University of Western Australia, Perth, WA 6009, Australia
- Centre for Kidney Research, Children’s Hospital at Westmead School of Public Health, Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - Itamar Levinger
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3011, Australia
- Australian Institute for Musculoskeletal Science, Victoria University, University of Melbourne, St Albans, VIC 3021, Australia
| |
Collapse
|
2
|
Dave S, Patel B. The lipocalin saga: Insights into its role in cancer-associated cachexia. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167684. [PMID: 39837432 DOI: 10.1016/j.bbadis.2025.167684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 01/23/2025]
Abstract
Cancer-associated cachexia (CAC) is a debilitating condition, observed in patients with advanced stages of cancer. It is marked by ongoing weight loss, weakness, and nutritional impairment. Lower tolerance of chemotherapeutic agents and radiation therapy makes it difficult to treat CAC. Anorexia is a significant contributor to worsening CAC. Anorexia can be found in the early or advanced stages of cancer. Anorexia in cancer patients arises from a confluence of factors. Tumor-related inflammatory cytokines can directly impact the gastrointestinal tract, leading to dysphagia and compromised gut function. Additionally, increased serotonin and hormonal disruptions lead to early satiety, suppressing appetite. Due to the complexities in the pathogenesis of the disease, identifying druggable targets is a challenge. Research is ongoing to identify novel targets for the treatment of this condition. Recent research suggests a potential link between elevated levels of Lipocalin 2 (LCN2) and cachexia in cancer patients. LCN2, a glycoprotein primarily released by neutrophils, is implicated in numerous illnesses, including skin disorders, cancer, atherosclerosis, and type 2 diabetes. LCN2 suppresses hunger by binding to the melanocortin-4 receptors. Several in vitro, in vivo, and clinical studies indicate the association between LCN2 levels and appetite suppression. Further research should be explored emphasizing the significance of well-crafted clinical trials to confirm LCN2's usefulness as a therapeutic target and its ability to help cancer patients who are suffering from the fatal hallmark of cachexia. This review explores LCN2's function in the multifaceted dynamics of CAC and anorexia.
Collapse
Affiliation(s)
- Srusti Dave
- National Forensic Sciences University, Gandhinagar 382007, Gujarat, India
| | - Bhoomika Patel
- National Forensic Sciences University, Gandhinagar 382007, Gujarat, India.
| |
Collapse
|
3
|
Xiang Q, Wu Z, Zhao Y, Tian S, Lin J, Wang L, Jiang S, Sun Z, Li W. Cellular and molecular mechanisms underlying obesity in degenerative spine and joint diseases. Bone Res 2024; 12:71. [PMID: 39658574 PMCID: PMC11632072 DOI: 10.1038/s41413-024-00388-8] [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: 06/24/2024] [Revised: 10/22/2024] [Accepted: 11/07/2024] [Indexed: 12/12/2024] Open
Abstract
Degenerative spine and joint diseases, including intervertebral disc degeneration (IDD), ossification of the spinal ligaments (OSL), and osteoarthritis (OA), are common musculoskeletal diseases that cause pain or disability to the patients. However, the pathogenesis of these musculoskeletal disorders is complex and has not been elucidated clearly to date. As a matter of fact, the spine and joints are not independent of other organs and tissues. Recently, accumulating evidence demonstrates the association between obesity and degenerative musculoskeletal diseases. Obesity is a common metabolic disease characterized by excessive adipose tissue or abnormal adipose distribution in the body. Excessive mechanical stress is regarded as a critical risk factor for obesity-related pathology. Additionally, obesity-related factors, mainly including lipid metabolism disorder, dysregulated pro-inflammatory adipokines and cytokines, are reported as plausible links between obesity and various human diseases. Importantly, these obesity-related factors are deeply involved in the regulation of cell phenotypes and cell fates, extracellular matrix (ECM) metabolism, and inflammation in the pathophysiological processes of degenerative spine and joint diseases. In this study, we systematically discuss the potential cellular and molecular mechanisms underlying obesity in these degenerative musculoskeletal diseases, and hope to provide novel insights for developing targeted therapeutic strategies.
Collapse
Affiliation(s)
- Qian Xiang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
| | - Zhenquan Wu
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
| | - Yongzhao Zhao
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
| | - Shuo Tian
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
| | - Jialiang Lin
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
| | - Longjie Wang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
| | - Shuai Jiang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
| | - Zhuoran Sun
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
| | - Weishi Li
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China.
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China.
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China.
| |
Collapse
|
4
|
Cho JY, Woo HE, Yeom J, An M, Ma S, Yim DJ, Kim SH, Lim YH. Microbial chaperonin 60 inhibits osteoclast differentiation by interfering with RANK/RANKL binding and overexpression of lipocalin2. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119850. [PMID: 39303785 DOI: 10.1016/j.bbamcr.2024.119850] [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: 04/09/2024] [Revised: 08/31/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
Osteoclasts play a crucial role in bone destruction in rheumatoid arthritis (RA). This study aimed to investigate the inhibitory effects of chaperonin 60 (CPN60), identified in the surface proteins of Propionibacterium freudenreichii MJ2, on receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclast differentiation, and elucidate the underlying mechanisms. Treatment with CPN60 inhibited RANKL-induced osteoclast differentiation by decreasing the expression of osteoclast differentiation-related genes and proteins. CPN60 interfered with the binding of RANKL to RANK, as elucidated using surface plasmon resonance (SPR) and immunofluorescence. In silico molecular docking analysis further supported the interference of CPN60 with the binding of RANKL and RANK. CPN60 suppressed the expression of molecules linked to the calcium-dependent pathway in RANKL-induced osteoclast differentiation at both mRNA and protein levels. Microarray analysis showed elevated expression of lipocalin 2 (Lcn2), which was closely linked to the inhibition of osteoclast differentiation in CPN60-treated RAW 264.7 cells. Inhibition of Lcn2 decreased the inhibitory effect of CPN60 on osteoclast differentiation, indicating that increased expression of Lcn2 by CPN60 contributes to the inhibition of osteoclastogenesis. In addition, CPN60 treatment alleviated arthritis symptoms in collagen-induced arthritis mice by reducing the generation of collagen-specific antibodies and inhibiting osteoclast differentiation. In conclusion, CPN60 of P. freudenreichii MJ2 interfered with RANKL-RANK binding, reduced the expression of genes and proteins related to osteoclast differentiation and upregulated Lcn2 expression, thereby inhibiting RANKL-induced osteoclast differentiation, which might contribute to ameliorate collagen-induced arthritis.
Collapse
Affiliation(s)
- Joo-Young Cho
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Hee-Eun Woo
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Jiah Yeom
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Mirae An
- Department of Public Health Science, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Seongho Ma
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Dong Joon Yim
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Sang-Hun Kim
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Young-Hee Lim
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea; School of Biosystems and Biomedical Sciences, Korea University, Seoul 02841, Republic of Korea; Department of Laboratory Medicine, Korea University Guro Hospital, Seoul 08308, Republic of Korea.
| |
Collapse
|
5
|
Wang Y, Zhang Y, Gao M, Chen Z, Lu J, Li Y, Di Y, Zhao Y, Liu B, Tang R. Lipocalin-2 promotes CKD vascular calcification by aggravating VSMCs ferroptosis through NCOA4/FTH1-mediated ferritinophagy. Cell Death Dis 2024; 15:865. [PMID: 39613734 DOI: 10.1038/s41419-024-07260-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 11/17/2024] [Accepted: 11/22/2024] [Indexed: 12/01/2024]
Abstract
Vascular calcification (VC) is a common complication of chronic kidney disease (CKD), for which no effective therapies are available. Hyperphosphatemia, a feature of CKD, is a well-known inducer of VC. High phosphate (HP)-induced ferroptosis plays a crucial role in CKD-related VC (CKD-VC), but the mechanisms remain unclear. Lipocalin-2 (LCN2), an iron-trafficking protein, has been implicated in ferroptosis regulation. In the present study, the role of LCN2 as a potential mediator of CKD-VC was investigated. HP-induced LCN2 expression in the arteries of CKD-VC patients, animal models and vascular smooth muscle cells (VSMCs). LCN2 knockout (LCN2KO) mice and wild-type (WT) mice fed with a high adenine and phosphate (AP) diet were studied to explore CKD-VC. Compared with WT mice, LCN2KO mice showed an amelioration of the CKD-VC induced by the AP diet. The inhibition of LCN2 also alleviated HP-induced calcium deposition and phenotypic transition in VSMCs. Conversely, VSMCs-targeted LCN2 overexpression or recombinant LCN2 treatment exacerbated CKD-VC in vivo and in vitro. Mechanistically, nuclear receptor coactivator 4 (NCOA4)/ferritin heavy chain 1 (FTH1)-mediated ferritinophagy-dependent ferroptosis was involved in LCN2-mediated CKD-VC. Under HP conditions, LCN2 interacted with NCOA4, potentially accelerating the degradation of FTH1 and inducing ferroptosis. The inhibition of LCN2 may rescue the degradation of FTH1 and thus ameliorate ferroptosis, significantly suppressing VSMCs calcification. In summary, our study revealed a novel role for LCN2 induced ferritinophagy-dependent ferroptosis in CKD-VC, and targeting LCN2 might be a promising treatment for CKD-VC.
Collapse
MESH Headings
- Animals
- Lipocalin-2/metabolism
- Lipocalin-2/genetics
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Ferroptosis
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Mice
- Humans
- Nuclear Receptor Coactivators/metabolism
- Nuclear Receptor Coactivators/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Mice, Knockout
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Mice, Inbred C57BL
- Ferritins/metabolism
- Male
- Disease Models, Animal
- Oxidoreductases
Collapse
Affiliation(s)
- Yujia Wang
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yuxia Zhang
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Min Gao
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zhiqing Chen
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Jing Lu
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yongqi Li
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yan Di
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yinan Zhao
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Bicheng Liu
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Rining Tang
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.
| |
Collapse
|
6
|
Solís-Suárez DL, Cifuentes-Mendiola SE, García-Hernández AL. A systematic review of the implications of lipocalin-2 expression in periodontal disease. Evid Based Dent 2024:10.1038/s41432-024-01070-y. [PMID: 39516276 DOI: 10.1038/s41432-024-01070-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 09/29/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVE Evidence suggests that lipocalin-2 (LCN-2), a bone-derived protein, is upregulated in periodontal diseases. This systematic review aimed to evaluate LCN-2 concentrations in individuals with periodontal diseases, identifying the most suitable body fluids for its detection, the type of periodontal disease with the highest LCN-2 expression, its association with other inflammatory markers and systemic diseases, and whether its expression can be modified by periodontal treatment. METHODS A systematic search of Google Scholar, PubMed, and ProQuest up to August 2024 was conducted. The studies were screened and selected by the authors according to specific eligibility criteria. Quality assessment of the included studies was performed according to the study type using STROBE statement for observational studies or the modified Jadad scale for experimental studies. The review was registered in PROSPERO (CRD42023458565). RESULTS In total, three thousand six hundred and thirty-eight reports were identified, of which twenty-seven were full-text assessed for eligibility, including eleven articles. Seven articles were observational, and four were experimental. Significantly elevated LCN-2 levels were reported in patients with periodontal disease across 9 studies, being higher in periodontitis rather than gingivitis. LCN-2 was mainly detected in gingival crevicular fluid (GCF) and saliva. LCN-2 expression is related to the increment of inflammatory markers, and periodontal therapy decreases LCN-2 concentrations. LCN-2 levels were aggravated when periodontitis was accompanied by obesity and type 2 diabetes. CONCLUSION LCN-2 is implicated in periodontal diseases, probably through the inflammation process.
Collapse
Affiliation(s)
- Diana L Solís-Suárez
- Laboratory of Dental Research, Section of Osteoimmunology and Oral Immunology, FES Iztacala, National Autonomous University of Mexico, 54714, Mexico, Mexico State, Mexico
- Postgraduate Course in Dental Sciences. National Autonomous University of Mexico, Mexico City, Mexico
| | - Saúl E Cifuentes-Mendiola
- Laboratory of Dental Research, Section of Osteoimmunology and Oral Immunology, FES Iztacala, National Autonomous University of Mexico, 54714, Mexico, Mexico State, Mexico
| | - Ana L García-Hernández
- Laboratory of Dental Research, Section of Osteoimmunology and Oral Immunology, FES Iztacala, National Autonomous University of Mexico, 54714, Mexico, Mexico State, Mexico.
| |
Collapse
|
7
|
Smith C, Lin X, Parker L, Yeap BB, Hayes A, Levinger I. The role of bone in energy metabolism: A focus on osteocalcin. Bone 2024; 188:117238. [PMID: 39153587 DOI: 10.1016/j.bone.2024.117238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 08/06/2024] [Accepted: 08/14/2024] [Indexed: 08/19/2024]
Abstract
Understanding the mechanisms involved in whole body glucose regulation is key for the discovery of new treatments for type 2 diabetes (T2D). Historically, glucose regulation was largely focused on responses to insulin and glucagon. Impacts of incretin-based therapies, and importance of muscle mass, are also highly relevant. Recently, bone was recognized as an endocrine organ, with several bone proteins, known as osteokines, implicated in glucose metabolism through their effects on the liver, skeletal muscle, and adipose tissue. Research efforts mostly focused on osteocalcin (OC) as a leading example. This review will provide an overview on this role of bone by discussing bone turnover markers (BTMs), the receptor activator of nuclear factor kB ligand (RANKL), osteoprotegerin (OPG), sclerostin (SCL) and lipocalin 2 (LCN2), with a focus on OC. Since 2007, some, but not all, research using mostly OC genetically modified animal models suggested undercarboxylated (uc) OC acts as a hormone involved in energy metabolism. Most data generated from in vivo, ex vivo and in vitro models, indicate that exogenous ucOC administration improves whole-body and skeletal muscle glucose metabolism. Although data in humans are generally supportive, findings are often discordant likely due to methodological differences and observational nature of that research. Overall, evidence supports the concept that bone-derived factors are involved in energy metabolism, some having beneficial effects (ucOC, OPG) others negative (RANKL, SCL), with the role of some (LCN2, other BTMs) remaining unclear. Whether the effect of osteokines on glucose regulation is clinically significant and of therapeutic value for people with insulin resistance and T2D remains to be confirmed.
Collapse
Affiliation(s)
- Cassandra Smith
- Nutrition & Health Innovation Research Institute, School of Health and Medical Sciences, Edith Cowan University, Perth, Western Australia, Australia; Medical School, The University of Western Australia, Perth, Western Australia, Australia; Institute for Health and Sport (IHES), Victoria University, Melbourne, VIC, Australia; Australian Institute for Musculoskeletal Science (AIMSS), Victoria University and Western Health, St Albans, VIC, Australia
| | - Xuzhu Lin
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Lewan Parker
- Institute for Physical Activity and Nutrition (IPAN), Deakin University, Geelong, VIC, Australia
| | - Bu B Yeap
- Medical School, The University of Western Australia, Perth, Western Australia, Australia; Department of Endocrinology and Diabetes, Fiona Stanley Hospital, Perth, Australia
| | - Alan Hayes
- Institute for Health and Sport (IHES), Victoria University, Melbourne, VIC, Australia; Australian Institute for Musculoskeletal Science (AIMSS), Victoria University and Western Health, St Albans, VIC, Australia; Department of Medicine - Western Health, The University of Melbourne, Footscray, VIC, Australia
| | - Itamar Levinger
- Institute for Health and Sport (IHES), Victoria University, Melbourne, VIC, Australia; Australian Institute for Musculoskeletal Science (AIMSS), Victoria University and Western Health, St Albans, VIC, Australia; Department of Medicine - Western Health, The University of Melbourne, Footscray, VIC, Australia.
| |
Collapse
|
8
|
Hemat Jouy S, Mohan S, Scichilone G, Mostafa A, Mahmoud AM. Adipokines in the Crosstalk between Adipose Tissues and Other Organs: Implications in Cardiometabolic Diseases. Biomedicines 2024; 12:2129. [PMID: 39335642 PMCID: PMC11428859 DOI: 10.3390/biomedicines12092129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Adipose tissue was previously regarded as a dormant organ for lipid storage until the identification of adiponectin and leptin in the early 1990s. This revelation unveiled the dynamic endocrine function of adipose tissue, which has expanded further. Adipose tissue has emerged in recent decades as a multifunctional organ that plays a significant role in energy metabolism and homeostasis. Currently, it is evident that adipose tissue primarily performs its function by secreting a diverse array of signaling molecules known as adipokines. Apart from their pivotal function in energy expenditure and metabolism regulation, these adipokines exert significant influence over a multitude of biological processes, including but not limited to inflammation, thermoregulation, immune response, vascular function, and insulin sensitivity. Adipokines are pivotal in regulating numerous biological processes within adipose tissue and facilitating communication between adipose tissue and various organs, including the brain, gut, pancreas, endothelial cells, liver, muscle, and more. Dysregulated adipokines have been implicated in several metabolic diseases, like obesity and diabetes, as well as cardiovascular diseases. In this article, we attempted to describe the significance of adipokines in developing metabolic and cardiovascular diseases and highlight their role in the crosstalk between adipose tissues and other tissues and organs.
Collapse
Affiliation(s)
- Shaghayegh Hemat Jouy
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Central Tehran Branch, Islamic Azad University, Tehran 14778-93855, Iran;
| | - Sukrutha Mohan
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (S.M.); (G.S.)
| | - Giorgia Scichilone
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (S.M.); (G.S.)
| | - Amro Mostafa
- Department of Pharmacology, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA;
| | - Abeer M. Mahmoud
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (S.M.); (G.S.)
- Department of Kinesiology and Nutrition, College of Applied Health Sciences, University of Illinois Chicago, Chicago, IL 60612, USA
| |
Collapse
|
9
|
Martiniakova M, Mondockova V, Kovacova V, Babikova M, Zemanova N, Biro R, Penzes N, Omelka R. Interrelationships among metabolic syndrome, bone-derived cytokines, and the most common metabolic syndrome-related diseases negatively affecting bone quality. Diabetol Metab Syndr 2024; 16:217. [PMID: 39238022 PMCID: PMC11378428 DOI: 10.1186/s13098-024-01440-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 08/05/2024] [Indexed: 09/07/2024] Open
Abstract
Metabolic syndrome (MetS), as a set of medical conditions including hyperglycemia, hypertension, abdominal obesity, and dyslipidemia, represents a highly prevalent disease cluster worldwide. The individual components of MetS together increase the risk of MetS-related disorders. Recent research has demonstrated that bone, as an endocrine organ, releases several systemic cytokines (osteokines), including fibroblast growth factor 23 (FGF23), lipocalin 2 (LCN2), and sclerostin (SCL). This review not only summarizes current knowledge about MetS, osteokines and the most common MetS-related diseases with a detrimental impact on bone quality (type 2 diabetes mellitus: T2DM; cardiovascular diseases: CVDs; osteoporosis: OP), but also provides new interpretations of the relationships between osteokines and individual components of MetS, as well as between osteokines and MetS-related diseases mentioned above. In this context, particular emphasis was given on available clinical studies. According to the latest knowledge, FGF23 may become a useful biomarker for obesity, T2DM, and CVDs, as FGF23 levels were increased in patients suffering from these diseases. LCN2 could serve as an indicator of obesity, dyslipidemia, T2DM, and CVDs. The levels of LCN2 positively correlated with obesity indicators, triglycerides, and negatively correlated with high-density lipoprotein (HDL) cholesterol. Furthermore, subjects with T2DM and CVDs had higher LCN2 levels. SCL may act as a potential biomarker predicting the incidence of MetS including all its components, T2DM, CVDs, and OP. Elevated SCL levels were noted in individuals with T2DM, CVDs and reduced in patients with OP. The aforementioned bone-derived cytokines have the potential to serve as promising predictors and prospective treatment targets for MetS and MetS-related diseases negatively affecting bone quality.
Collapse
Affiliation(s)
- Monika Martiniakova
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 01, Nitra, Slovakia
| | - Vladimira Mondockova
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Tr. A. Hlinku 1, 949 01, Nitra, Slovakia
| | - Veronika Kovacova
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 01, Nitra, Slovakia
| | - Martina Babikova
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Tr. A. Hlinku 1, 949 01, Nitra, Slovakia
| | - Nina Zemanova
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 01, Nitra, Slovakia
| | - Roman Biro
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 01, Nitra, Slovakia
| | - Noemi Penzes
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Tr. A. Hlinku 1, 949 01, Nitra, Slovakia
| | - Radoslav Omelka
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Tr. A. Hlinku 1, 949 01, Nitra, Slovakia.
| |
Collapse
|
10
|
Sólis-Suarez DL, Cifuentes-Mendiola SE, González-Alva P, Rodríguez-Hernández AP, Martínez-Dávalos A, Llamosas-Hernandez FE, Godínez-Victoria M, García-Hernández AL. Lipocalin-2 as a fundamental protein in type 2 diabetes and periodontitis in mice. J Periodontol 2024. [PMID: 39189666 DOI: 10.1002/jper.24-0215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND Lipocalin-2 (LCN-2) is an osteokine that suppresses appetite, stimulates insulin secretion, regulates bone remodeling, and is induced by proinflammatory cytokines. The aim of this work was to investigate the participation of LCN-2 in periodontitis associated with type 2 diabetes (T2D) by evaluating alveolar bone loss, glycemic control, inflammation, and femur fragility. METHODS A murine model of periodontitis with T2D and elevated LCN-2 concentration was used. Functional LCN-2 inhibition was achieved using an anti-LCN-2 polyclonal antibody, and isotype immunoglobulin G was used as a control. The alveolar bone and femur were evaluated by micro-CT. Glucose metabolism was determined. Tumor necrosis factor (TNF-α) and receptor activator of nuclear factor kappa-B ligand (RANKL) levels in alveolar bone lysates were quantified using ELISA, and serum cytokines were quantified using flow cytometry. A three-point bending test was performed in the femur, and RANKL levels were measured in femur lysates using ELISA. RESULTS Functional inhibition of LCN-2 in T2D-periodontitis mice decreased alveolar bone loss in buccal and palatal surfaces and preserved the microarchitecture of the remaining bone, decreased TNF-α and RANKL in alveolar bone, reduced hyperglycemia, glucose intolerance, and insulin resistance, and increased insulin production through improving the functionality of pancreatic β cells. Furthermore, this inhibition increased serum free-glycerol levels, decreased serum interleukin (IL)-6, increased serum IL-4, and reduced femur fragility and RANKL expression in the femur. CONCLUSIONS LCN-2 participates in periodontitis associated with T2D. Inhibiting its function in mice with T2D and periodontitis improves pancreatic β-cell function, and glucose metabolism and decreases inflammatory cytokines and bone-RANKL levels, which results in the preservation of femoral and alveolar bone microarchitecture. PLAIN LANGUAGE SUMMARY In this study, we explored the role of a bone protein known as lipocalin-2 (LCN-2) in the connection between periodontitis and type 2 diabetes (T2D). Periodontitis is a destructive gum and alveolar bone disease. LCN-2 levels are increased in both T2D and periodontitis. Using a mouse model of T2D with periodontitis, we examined how blocking LCN-2 function affected various aspects of these two diseases. We found that this inhibition led to significant improvements. First, it reduced alveolar bone loss and preserved bone structure by decreasing local inflammation and bone resorption. Second, it improved glucose and lipid metabolism, leading to better blood-sugar control and decreased insulin resistance. Blocking the functions of LCN-2 also decreased systemic inflammation throughout the body and strengthened bone integrity. Overall, our results suggest that LCN-2 plays a crucial role in the periodontitis associated with T2D. By inhibiting LCN-2 function, we were able to improve pancreatic function, improve glucose metabolism, reduce inflammation, and enhance bone health. Targeting LCN-2 could be a promising strategy for the harmful effects of T2D and periodontitis.
Collapse
Affiliation(s)
- Diana Laura Sólis-Suarez
- Laboratory of Dental Research, Section of Osteoimmunology and Oral Immunology, FES Iztacala, National Autonomous University of Mexico (UNAM), State of Mexico, Mexico, Mexico
- Postgraduate Course in Dental Sciences, National Autonomous University of Mexico, Mexico City, Mexico
| | - Saúl Ernesto Cifuentes-Mendiola
- Laboratory of Dental Research, Section of Osteoimmunology and Oral Immunology, FES Iztacala, National Autonomous University of Mexico (UNAM), State of Mexico, Mexico, Mexico
| | - Patricia González-Alva
- Laboratory of Tissue Bioengineering, Faculty of Dentistry, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | | | - Arnulfo Martínez-Dávalos
- Endo-periodontology Department, Physics Institute, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | | | - Marycarmen Godínez-Victoria
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Ana Lilia García-Hernández
- Laboratory of Dental Research, Section of Osteoimmunology and Oral Immunology, FES Iztacala, National Autonomous University of Mexico (UNAM), State of Mexico, Mexico, Mexico
| |
Collapse
|
11
|
Pompili S, Cappariello A, Vetuschi A, Sferra R. G-Protein-Coupled Receptor 120 Agonist Mitigates Steatotic and Fibrotic Features Triggered in Obese Mice by the Administration of a High-Fat and High-Carbohydrate Diet. ACS OMEGA 2024; 9:31899-31909. [PMID: 39072106 PMCID: PMC11270546 DOI: 10.1021/acsomega.4c03507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/30/2024]
Abstract
Nonalcoholic fatty liver disease (NALFD) represents a complex condition ranging from simple steatosis (nonalcoholic fatty liver, NAFL) to inflammation, and fibrosis is one of the main features of nonalcoholic steatohepatitis (NASH). The pathogenesis of NAFLD is not well established but involves several factors (i.e., predisposition of genetic variants, obesity, and unhealthy lifestyle as unbalanced diets) that lead to an alteration of lipid homeostasis and consequently to an abnormal accumulation of triglycerides and other lipids in the liver parenchyma. Currently, no resolutive pharmacological treatment for NAFLD is available, and the only therapeutic approach is a healthy diet and physical exercise. In this study, we investigated the potential beneficial effect of GprA, a new synthetic agonist of G-protein-coupled receptor 120/free fatty acid receptor 4 (GPR120/FFAR4), in the progression of NAFL/NASH in mice fed for different periods (26 weeks and 30 weeks), with a high-fat (40% kcal) and high-carbohydrate diet, also called a Western-style diet (WSD). In our experimental model, the histological, protein, and transcriptomic analyses highlighted that the GprA can reduce signs of steatosis in WSD-fed mice. Furthermore, in 30 week-treated mice, GprA is also effective in the reduction of collagen deposition and fibrosis development. Altogether, our data validate the central role of FFAR4 in the context of NAFL/NASH onset and progression and reveal that GprA could represent an interesting candidate for the development of a new therapeutic approach in NAFLD treatment.
Collapse
Affiliation(s)
- Simona Pompili
- Department
of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Alfredo Cappariello
- Department
of Life, Health and Experimental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Antonella Vetuschi
- Department
of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Roberta Sferra
- Department
of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| |
Collapse
|
12
|
Wang S, Zhu J, Liu W, Liu A. A Machine Learning Framework for Screening Plasma Cell-Associated Feature Genes to Estimate Osteoporosis Risk and Treatment Vulnerability. Biochem Genet 2024:10.1007/s10528-024-10861-y. [PMID: 38898268 DOI: 10.1007/s10528-024-10861-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 06/06/2024] [Indexed: 06/21/2024]
Abstract
Osteoporosis, in which bones become fragile owing to low bone density and impaired bone mass, is a global public health concern. Bone mineral density (BMD) has been extensively evaluated for the diagnosis of low bone mass and osteoporosis. Circulating monocytes play an indispensable role in bone destruction and remodeling. This work proposed a machine learning-based framework to investigate the impact of circulating monocyte-associated genes on bone loss in osteoporosis patients. Females with discordant BMD levels were included in the GSE56815, GSE7158, GSE7429, and GSE62402 datasets. Circulating monocyte types were quantified via CIBERSORT, with subsequent selection of plasma cell-associated DEGs. Generalized linear models, random forests, extreme gradient boosting (XGB), and support vector machines were adopted for feature selection. Artificial neural networks and nomograms were subsequently constructed for osteoporosis diagnosis, and the molecular machinery underlying the identified genes was explored. SVM outperformed the other tuned models; thus, the expression of several genes (DEFA4, HLA-DPB1, LCN2, HP, and GAS7) associated with osteoporosis were determined. ANNs and nomograms were proposed to robustly distinguish low and high BMDs and estimate the risk of osteoporosis. Clozapine, aspirin, pyridoxine, etc. were identified as possible treatment agents. The expression of these genes is extensively posttranscriptionally regulated by miRNAs and m6A modifications. Additionally, they participate in modulating key signaling pathways, e.g., autophagy. The machine learning framework based on plasma cell-associated feature genes has the potential for estimating personalized risk stratification and treatment vulnerability in osteoporosis patients.
Collapse
Affiliation(s)
- Shoubao Wang
- Department of Orthopedics, Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huai'an, 223300, China
| | - Jiafu Zhu
- Department of Orthopaedics, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, China
| | - Weinan Liu
- Department of Orthopedics, The Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, 350004, China
- Key Laboratory of Orthopedics & Traumatology of Traditional Chinese Medicine and Rehabilitation Ministry of Education, Fujian University of TCM, Fuzhou, 350004, China
| | - Aihua Liu
- Department of Orthopaedics, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, 445000, China.
| |
Collapse
|
13
|
Puri C, Dannenberg C, Ucci A, Ponzetti M, Pucci E, Silvestri L, Lau P, Frings-Meuthen P, Heer M, Rucci N, Teti A, Maurizi A. Pre-proenkephalin 1 is Downregulated Under Unloading and is Involved in Osteoblast Biology. Calcif Tissue Int 2024; 114:524-534. [PMID: 38506955 PMCID: PMC11061007 DOI: 10.1007/s00223-024-01199-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 02/20/2024] [Indexed: 03/22/2024]
Abstract
Pre-proenkephalin 1 (Penk1) is a pro-neuropeptide that belongs to the typical opioid peptide's family, having analgesic properties. We previously found Penk1 to be the most downregulated gene in a whole gene profiling analysis performed in osteoblasts subjected to microgravity as a model of mechanical unloading. In this work, Penk1 downregulation was confirmed in the bones of two in vivo models of mechanical unloading: tail-suspended and botulinum toxin A (botox)-injected mice. Consistently, in the sera from healthy volunteers subjected to bed rest, we observed an inverse correlation between PENK1 and bed rest duration. These results prompted us to investigate a role for this factor in bone. Penk1 was highly expressed in mouse bone, but its global deletion failed to impact bone metabolism in vivo. Indeed, Penk1 knock out (Penk1-/-) mice did not show an overt bone phenotype compared to the WT littermates. Conversely, in vitro Penk1 gene expression progressively increased during osteoblast differentiation and its transient silencing in mature osteoblasts by siRNAs upregulated the transcription of the Sost1 gene encoding sclerostin, and decreased Wnt3a and Col1a1 mRNAs, suggesting an altered osteoblast activity due to an impairment of the Wnt pathway. In line with this, osteoblasts treated with the Penk1 encoded peptide, Met-enkephalin, showed an increase of Osx and Col1a1 mRNAs and enhanced nodule mineralization. Interestingly, primary osteoblasts isolated from Penk1-/- mice showed lower metabolic activity, ALP activity, and nodule mineralization, as well as a lower number of CFU-F compared to osteoblasts isolated from WT mice, suggesting that, unlike the transient inhibition, the chronic Penk1 deletion affects both osteoblast differentiation and activity. Taken together, these results highlight a role for Penk1 in the regulation of the response of the bone to mechanical unloading, potentially acting on osteoblast differentiation and activity in a cell-autonomous manner.
Collapse
Affiliation(s)
- Chiara Puri
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio - Coppito 2, 67100, L'Aquila, Italy
| | - Charlotte Dannenberg
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio - Coppito 2, 67100, L'Aquila, Italy
| | - Argia Ucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio - Coppito 2, 67100, L'Aquila, Italy
| | - Marco Ponzetti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio - Coppito 2, 67100, L'Aquila, Italy
| | - Elisa Pucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio - Coppito 2, 67100, L'Aquila, Italy
| | - Luciana Silvestri
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio - Coppito 2, 67100, L'Aquila, Italy
| | - Patrick Lau
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Cologne, Germany
| | - Petra Frings-Meuthen
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Cologne, Germany
| | - Martina Heer
- Institute of Nutritional and Food Sciences, University of Bonn, Bonn, Germany
| | - Nadia Rucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio - Coppito 2, 67100, L'Aquila, Italy
| | - Anna Teti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio - Coppito 2, 67100, L'Aquila, Italy
| | - Antonio Maurizi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio - Coppito 2, 67100, L'Aquila, Italy.
| |
Collapse
|
14
|
Zhang Z, Jing Y, Zhang A, Liu J, Yang H, Lou X, Xu L, Liu M, Zhang Y, Gu J. Long non-coding RNA-NONMMMUT004552.2 regulates the unloading-induced bone loss through the miRNA-15b-5p/Syne1 in mice. NPJ Microgravity 2024; 10:37. [PMID: 38521778 PMCID: PMC10960867 DOI: 10.1038/s41526-024-00382-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 03/08/2024] [Indexed: 03/25/2024] Open
Abstract
Exercise-induced mechanical loading can increase bone strength whilst mechanical unloading enhances bone-loss. Here, we investigated the role of lncRNA NONMMUT004552.2 in unloading-induced bone-loss. Knockout of lncRNA NONMMUT004552.2 in hindlimb-unloaded mice caused an increase in the bone formation and osteoblast activity. The silencing of lncRNA NONMMUT004552.2 also decreased the osteoblast apoptosis and expression of Bax and cleaved caspase-3, increased Bcl-2 protein expression in MC3T3-E1 cells. Mechanistic investigations demonstrated that NONMMUT004552.2 functions as a competing endogenous RNA (ceRNA) to facilitate the protein expression of spectrin repeat containing, nuclear envelope 1 (Syne1) by competitively binding miR-15b-5p and subsequently inhibits the osteoblast differentiation and bone formation in the microgravity unloading environment. These data highlight the importance of the lncRNA NONMMUT004552.2/miR-15b-5p/Syne1 axis for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Medical Engineering, PLA Strategic Support Force Characteristic Medical Center, Beijing, 100101, China
| | - Yu Jing
- Department of Haematology, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, 100071, China
| | - Ang Zhang
- Department of Hematology, PLA Strategic Support Force Characteristic Medical Center, Beijing, 100101, China
| | - JiShan Liu
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Heming Yang
- Department of General Surgery, PLA Strategic Support Force Characteristic Medical Center, Beijing, 100101, China
| | - Xiaotong Lou
- Department of Research, PLA Strategic Support Force Characteristic Medical Center, Beijing, 100101, China
| | - Liyan Xu
- Department of Blood Transfusion, PLA Strategic Support Force Characteristic Medical Center, Beijing, 100101, China
| | - Min Liu
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Yikun Zhang
- Department of Hematology, PLA Strategic Support Force Characteristic Medical Center, Beijing, 100101, China.
| | - Jianwen Gu
- Department of Neurosurgery, PLA Strategic Support Force Characteristic Medical Center, Beijing, 100101, China.
| |
Collapse
|
15
|
Patil JD, Fredericks S. The role of adipokines in osteoporosis management: a mini review. Front Endocrinol (Lausanne) 2024; 15:1336543. [PMID: 38516409 PMCID: PMC10956128 DOI: 10.3389/fendo.2024.1336543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/22/2024] [Indexed: 03/23/2024] Open
Abstract
The prevalence of osteoporosis has been on the rise globally. With ageing populations, research has sought therapeutic solutions in novel areas. One such area is that of the adipokines. Current literature points to an important role for these chemical mediators in relation to bone metabolism. Well-established adipokines have been broadly reported upon. These include adiponectin and leptin. However, other novel adipokines such as visfatin, nesfatin-1, meteorin-like protein (Metrnl), apelin and lipocalin-2 are starting to be addressed pre-clinically and clinically. Adipokines hold pro-inflammatory and anti-inflammatory properties that influence the pathophysiology of various bone diseases. Omentin-1 and vaspin, two novel adipokines, share cardioprotective effects and play essential roles in bone metabolism. Studies have reported bone-protective effects of omentin-1, whilst others report negative associations between omentin-1 and bone mineral density. Lipocalin-2 is linked to poor bone microarchitecture in mice and is even suggested to mediate osteoporosis development from prolonged disuse. Nesfatin-1, an anorexigenic adipokine, has been known to preserve bone density. Animal studies have demonstrated that nesfatin-1 treatment limits bone loss and increases bone strength, suggesting exogenous use as a potential treatment for osteopenic disorders. Pre-clinical studies have shown adipokine apelin to have a role in bone metabolism, mediated by the enhancement of osteoblast genesis and the inhibition of programmed cell death. Although many investigations have reported conflicting findings, sufficient literature supports the notion that adipokines have a significant influence on the metabolism of bone. This review aims at highlighting the role of novel adipokines in osteoporosis while also discussing their potential for treating osteoporosis.
Collapse
Affiliation(s)
| | - Salim Fredericks
- The Royal College of Surgeons in Ireland – Medical University of Bahrain, Al Sayh, Bahrain
| |
Collapse
|
16
|
Yang Y, Liu J, Kousteni S. Lipocalin 2-A bone-derived anorexigenic and β-cell promoting signal: From mice to humans. J Diabetes 2024; 16:e13504. [PMID: 38035773 PMCID: PMC10940901 DOI: 10.1111/1753-0407.13504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/16/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
The skeleton is traditionally known for its structural support, organ protection, movement, and maintenance of mineral homeostasis. Over the last 10 years, bone has emerged as an endocrine organ with diverse physiological functions. The two key molecules in this context are fibroblast growth factor 23 (FGF23), secreted by osteocytes, and osteocalcin, a hormone produced by osteoblasts. FGF23 affects mineral homeostasis through its actions on the kidneys, and osteocalcin has beneficial effects in improving glucose homeostasis, muscle function, brain development, cognition, and male fertility. In addition, another osteoblast-derived hormone, lipocalin 2 (LCN2) has emerged into the researchers' field of vision. In this review, we mainly focus on LCN2's role in appetite regulation and glucose metabolism and also briefly introduce its effects in other pathophysiological conditions, such as nonalcoholic fatty liver disease, sarcopenic obesity, and cancer-induced cachexia.
Collapse
Affiliation(s)
- Yuying Yang
- Department of Endocrine and Metabolic Diseases, Rui‐jin Hospital, Shanghai Jiao Tong University School of MedicineShanghai Institute of Endocrine and Metabolic Diseases, and Shanghai Clinical Center for Endocrine and Metabolic DiseasesShanghaiChina
- Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Clinical Research Center for Metabolic Diseases, Shanghai National Center for Translational Medicine, Rui‐jin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jianmin Liu
- Department of Endocrine and Metabolic Diseases, Rui‐jin Hospital, Shanghai Jiao Tong University School of MedicineShanghai Institute of Endocrine and Metabolic Diseases, and Shanghai Clinical Center for Endocrine and Metabolic DiseasesShanghaiChina
- Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Clinical Research Center for Metabolic Diseases, Shanghai National Center for Translational Medicine, Rui‐jin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Stavroula Kousteni
- Department of Physiology and Cellular BiophysicsColumbia University Medical CenterNew YorkNew YorkUSA
| |
Collapse
|
17
|
Al-Daghestani H, Qaisar R, Al Kawas S, Ghani N, Rani KGA, Azeem M, Hasnan HK, Kassim NK, Samsudin AR. Pharmacological inhibition of endoplasmic reticulum stress mitigates osteoporosis in a mouse model of hindlimb suspension. Sci Rep 2024; 14:4719. [PMID: 38413677 PMCID: PMC10899598 DOI: 10.1038/s41598-024-54944-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/19/2024] [Indexed: 02/29/2024] Open
Abstract
Hindlimb suspension (HLS) mice exhibit osteoporosis of the hindlimb bones and may be an excellent model to test pharmacological interventions. We investigated the effects of inhibiting endoplasmic reticulum (ER) stress with 4-phenyl butyrate (4-PBA) on the morphology, physicochemical properties, and bone turnover markers of hindlimbs in HLS mice. We randomly divided 21 male C57BL/6J mice into three groups, ground-based controls, untreated HLS group and 4-PBA treated group (HLS+4PBA) (100mg/kg/day, intraperitoneal) for 21 days. We investigated histopathology, micro-CT imaging, Raman spectroscopic analysis, and gene expression. Untreated HLS mice exhibited reduced osteocyte density, multinucleated osteoclast-like cells, adipocyte infiltration, and reduced trabecular striations on micro-CT than the control group. Raman spectroscopy revealed higher levels of ER stress, hydroxyproline, non-collagenous proteins, phenylalanine, tyrosine, and CH2Wag as well as a reduction in proteoglycans and adenine. Furthermore, bone alkaline phosphatase and osteocalcin were downregulated, while Cathepsin K, TRAP, and sclerostin were upregulated. Treatment with 4-PBA partially restored normal bone histology, increased collagen crosslinking, and mineralization, promoted anti-inflammatory markers, and downregulated bone resorption markers. Our findings suggest that mitigating ER stress with 4-PBA could be a therapeutic intervention to offset osteoporosis in conditions mimicking hindlimb suspension.
Collapse
Affiliation(s)
- Hiba Al-Daghestani
- Department of Oral and Craniofacial Health Sciences, College of Dental Medicine, University of Sharjah, Sharjah, 27272, UAE
| | - Rizwan Qaisar
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, UAE
- Space Medicine Research Group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, UAE
| | - Sausan Al Kawas
- Department of Oral and Craniofacial Health Sciences, College of Dental Medicine, University of Sharjah, Sharjah, 27272, UAE
| | - Nurhafizah Ghani
- School of Dental Sciences, Health Campus, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - K G Aghila Rani
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, UAE
| | - Muhammad Azeem
- Department of Mathematical and Physical Sciences, University of Nizwa, Nizwa 33, Sultanate of Oman
| | - Hijaz Kamal Hasnan
- Department of Geology, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Nur Karyatee Kassim
- School of Dental Sciences, Health Campus, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia.
| | - A R Samsudin
- Department of Oral and Craniofacial Health Sciences, College of Dental Medicine, University of Sharjah, Sharjah, 27272, UAE.
| |
Collapse
|
18
|
Bauer C, Sim M, Prince RL, Zhu K, Lim EM, Byrnes E, Pavlos N, Lim WH, Wong G, Lewis JR, Levinger I. Circulating lipocalin-2 and features of metabolic syndrome in community-dwelling older women: A cross-sectional study. Bone 2023; 176:116861. [PMID: 37524293 DOI: 10.1016/j.bone.2023.116861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/21/2023] [Accepted: 07/28/2023] [Indexed: 08/02/2023]
Abstract
Lipocalin-2 (LCN2) is released by several cell types including osteoblasts and adipocytes and has been suggested as a marker of renal dysfunction, metabolic syndrome (MetS) and type 2 diabetes (T2D). Whether LCN2 is linked to these diseases in older women remains unknown. This study investigated whether LCN2 is related to features of MetS and T2D in older women. This cross-sectional study included 705 non-diabetic women (mean age 75.1 ± 2.6 years) for MetS analysis and 76 women (mean age 75.4 ± 2.8 years) with T2D. Total circulating LCN2 levels were analysed using a two-step chemiluminescent microparticle monoclonal immunoassay. MetS was determined by a modified National Cholesterol Education Program Adult Treatment Panel III classification. Multivariable-adjusted logistic regression analysis was used to assess odds ratios between LCN2 quartiles and MetS. Women in the highest LCN2 quartile had approximately 3 times greater risk for MetS compared to women in the lowest quartile (OR 3.05; 95%CI 1.86-5.02). Women with T2D or MetS scores of ≥ 3 had higher LCN2 levels compared to women with a MetS score of 0 (p < 0.05). Higher LCN2 correlated with higher body mass index, fat mass, triglycerides and glycated haemoglobin and lower high-density lipoprotein cholesterol and estimated glomerular filtration rate (p < 0.05). Higher circulating levels of LCN2 are associated with worsened cardio-metabolic risk factors and increased odds of MetS and T2D in older women. Whether it can be used as a biomarker for identifying those at risk for MetS and T2D should be explored further.
Collapse
Affiliation(s)
- Carlie Bauer
- Institute for Health and Sport, Victoria University, Melbourne, Australia
| | - Marc Sim
- Nutrition & Health Innovation Research Institute, Edith Cowan University, Joondalup, Australia; Medical School, University of Western Australia, Perth, Australia
| | - Richard L Prince
- Medical School, University of Western Australia, Perth, Australia
| | - Kun Zhu
- Medical School, University of Western Australia, Perth, Australia; Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Perth, Australia
| | - Ee M Lim
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Perth, Australia; PathWest, QEII Medical Centre, Perth, Australia
| | | | - Nathan Pavlos
- School of Biomedical Sciences, University of Western Australia, Perth, Australia
| | - Wai H Lim
- Medical School, University of Western Australia, Perth, Australia; Department of Renal Medicine, Sir Charles Gairdner Hospital, Perth, Australia
| | - Germaine Wong
- Centre for Kidney Research, Children's Hospital at Westmead School of Public Health, Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Joshua R Lewis
- Nutrition & Health Innovation Research Institute, Edith Cowan University, Joondalup, Australia; Medical School, University of Western Australia, Perth, Australia; Centre for Kidney Research, Children's Hospital at Westmead School of Public Health, Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Itamar Levinger
- Institute for Health and Sport, Victoria University, Melbourne, Australia; Australian Institute for Musculoskeletal Science, Victoria University, University of Melbourne, Western Health, St Albans, VIC, Australia.
| |
Collapse
|
19
|
Ito K, Yamamoto T, Hayashi Y, Sato S, Nakayama J, Urabe F, Shimasaki T, Nakamura E, Matui Y, Fujimoto H, Kimura T, Egawa S, Ochiya T, Yamamoto Y. Osteoblast-derived extracellular vesicles exert osteoblastic and tumor-suppressive functions via SERPINA3 and LCN2 in prostate cancer. Mol Oncol 2023; 17:2147-2167. [PMID: 37408474 PMCID: PMC10552899 DOI: 10.1002/1878-0261.13484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 04/25/2023] [Accepted: 07/04/2023] [Indexed: 07/07/2023] Open
Abstract
Clinically, the osteolytic phenotype is rare in prostate cancer (PCa), and the prognosis is generally worse than that of the osteoblastic phenotype. Osteoblastic prostate cancer (BPCa) is a major type of bone metastasis. Several factors responsible for osteogenesis have been identified, but the molecular mechanism of osteoblastic bone metastasis in PCa is not fully understood. Here, we show the osteogenic and tumor-suppressive roles of SERPINA3 and LCN2 in BPCa. In a co-culture of osteoblasts (OBs) and BPCa cells, SERPINA3 and LCN2 were remarkably upregulated in BPCa via OB-derived extracellular vesicles, while they were not in the co-culture of OBs and osteolytic prostate cancer (LPCa) cells. In both the co-culture system and mouse xenograft experiments with intracaudal injection, enhanced expression of SERPINA3 and LCN2 in PCa led to osteogenesis. Additionally, the addition of SERPINA3 and LCN2 to BPCa cells significantly suppressed the proliferative potential. Retrospective analysis also confirmed that high expression levels of SERPINA3 and LCN2 were significantly correlated with a better prognosis. Our results may partially explain how osteoblastic bone metastasis develops and why the prognosis for BPCa is relatively better than that for LPCa.
Collapse
Affiliation(s)
- Kagenori Ito
- Laboratory of Integrative OncologyNational Cancer Center Research InstituteChuo‐kuJapan
- Department of UrologyJikei University School of MedicineMinato‐kuJapan
| | - Tomofumi Yamamoto
- Laboratory of Integrative OncologyNational Cancer Center Research InstituteChuo‐kuJapan
- Department of Molecular and Cellular MedicineTokyo Medical UniversityShinjuku‐kuJapan
| | - Yusuke Hayashi
- Laboratory of Integrative OncologyNational Cancer Center Research InstituteChuo‐kuJapan
| | - Shun Sato
- Department of PathologyJikei University School of MedicineMinato‐kuJapan
| | - Jun Nakayama
- Laboratory of Integrative OncologyNational Cancer Center Research InstituteChuo‐kuJapan
| | - Fumihiko Urabe
- Department of UrologyJikei University School of MedicineMinato‐kuJapan
| | - Takeo Shimasaki
- Medical Research InstituteKanazawa Medical UniversityKahoku‐gunJapan
| | - Eijiro Nakamura
- Department of Urology and Retroperitoneal SurgeryNational Cancer Center HospitalChuo‐kuJapan
| | - Yoshiyuki Matui
- Department of Urology and Retroperitoneal SurgeryNational Cancer Center HospitalChuo‐kuJapan
| | - Hiroyuki Fujimoto
- Department of Urology and Retroperitoneal SurgeryNational Cancer Center HospitalChuo‐kuJapan
| | - Takahiro Kimura
- Department of UrologyJikei University School of MedicineMinato‐kuJapan
| | - Shin Egawa
- Department of UrologyJikei University School of MedicineMinato‐kuJapan
| | - Takahiro Ochiya
- Department of Molecular and Cellular MedicineTokyo Medical UniversityShinjuku‐kuJapan
| | - Yusuke Yamamoto
- Laboratory of Integrative OncologyNational Cancer Center Research InstituteChuo‐kuJapan
| |
Collapse
|
20
|
Xia Y, Ge G, Xiao H, Wu M, Wang T, Gu C, Yang H, Geng D. REPIN1 regulates iron metabolism and osteoblast apoptosis in osteoporosis. Cell Death Dis 2023; 14:631. [PMID: 37749079 PMCID: PMC10519990 DOI: 10.1038/s41419-023-06160-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 09/06/2023] [Accepted: 09/15/2023] [Indexed: 09/27/2023]
Abstract
Osteoporosis is not well treated due to the difficulty of finding commonalities between the various types of it. Iron homeostasis is a vital component in supporting biochemical functions, and iron overload is recognized as a common risk factor for osteoporosis. In this research, we found that there is indeed evidence of iron accumulation in the bone tissue of patients with osteoporosis and REPIN1, as an origin specific DNA binding protein, may play a key role in this process. We revealed that sh-Repin1 therapy can rescue bone loss in an iron-overload-induced osteoporosis mouse model. Knockdown of Repin1 can inhibit apoptosis and enhance the resistance of osteoblasts to iron overload toxicity. REPIN1 promoted apoptosis by regulating iron metabolism in osteoblasts. Mechanistically, knockdown of Repin1 decreased the expression of Lcn2, which ameliorated the toxic effects of intracellular iron overload. The anti-iron effect of lentivirus sh-Repin1 was partially reversed or replicated by changing LCN2 expression level via si-RNA or plasmid, which indirectly verified the key regulatory role of LCN2 as a downstream target. Furthermore, the levels of BCL2 and BAX, which play a key role in the mitochondrial apoptosis pathway, were affected. In summary, based on the results of clinical specimens, animal models and in vitro experiments, for the first time, we proved the key role of REPIN1 in iron metabolism-related osteoporosis.
Collapse
Affiliation(s)
- Yu Xia
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Gaoran Ge
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Haixiang Xiao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Mingzhou Wu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Taicang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Taicang, China
| | - Tianhao Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Chengyong Gu
- Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital (North District), Suzhou, China.
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
21
|
Yeom J, Ma S, Yim DJ, Lim YH. Surface proteins of Propionibacterium freudenreichii MJ2 inhibit RANKL-induced osteoclast differentiation by lipocalin-2 upregulation and lipocalin-2-mediated NFATc1 inhibition. Sci Rep 2023; 13:15644. [PMID: 37730858 PMCID: PMC10511438 DOI: 10.1038/s41598-023-42944-y] [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: 01/31/2023] [Accepted: 09/16/2023] [Indexed: 09/22/2023] Open
Abstract
Osteoclasts degrade bone and osteoclast differentiation has been implicated in bone destruction in rheumatoid arthritis. The dairy bacterium Propionibacterium freudenreichii MJ2 (MJ2) isolated from raw milk inhibits osteoclast differentiation and ameliorates collagen-induced arthritis. This study aimed to investigate the inhibitory effect of the surface proteins of MJ2 on receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast differentiation and explain the underlying mechanism. The murine macrophage cell line RAW 264.7 was used to study the inhibition of osteoclast differentiation. The surface proteins significantly inhibited RANKL-induced osteoclast differentiation in a protein concentration-dependent manner by inhibiting the expression of genes and proteins related to osteoclast differentiation. RNA microarray analysis showed that the surface proteins significantly upregulated lipocalin-2 (lcn2) expression. In addition, they downregulated c-fos and NFATc1 and inhibited the expression of NFATc1-downstream genes Atp6v0d2, Calcr, and Ctsk. siRNA silencing of lcn2 decreased the extent of surface protein inhibition on osteoclast differentiation, suggesting that lcn2 plays an important role in the inhibition of RANKL-induced osteoclast differentiation. In conclusion, surface proteins of MJ2 show inhibitory effects on RANKL-induced osteoclast differentiation by upregulating lcn2 expression, in turn downregulating NFATc1, leading to the inhibition of NFATc1-downstream osteoclastogenesis-related gene expression.
Collapse
Affiliation(s)
- Jiah Yeom
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul, 02841, Republic of Korea
| | - Seongho Ma
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul, 02841, Republic of Korea
| | - Dong Joon Yim
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul, 02841, Republic of Korea
| | - Young-Hee Lim
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul, 02841, Republic of Korea.
- School of Biosystems and Biomedical Sciences, Korea University, Seoul, 02841, Republic of Korea.
- Department of Laboratory Medicine, Korea University Guro Hospital, Seoul, 08308, Republic of Korea.
| |
Collapse
|
22
|
Zheng XQ, Lin JL, Huang J, Wu T, Song CL. Targeting aging with the healthy skeletal system: The endocrine role of bone. Rev Endocr Metab Disord 2023; 24:695-711. [PMID: 37402956 DOI: 10.1007/s11154-023-09812-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/29/2023] [Indexed: 07/06/2023]
Abstract
Aging is an inevitable biological process, and longevity may be related to bone health. Maintaining strong bone health can extend one's lifespan, but the exact mechanism is unclear. Bone and extraosseous organs, including the heart and brain, have complex and precise communication mechanisms. In addition to its load bearing capacity, the skeletal system secretes cytokines, which play a role in bone regulation of extraosseous organs. FGF23, OCN, and LCN2 are three representative bone-derived cytokines involved in energy metabolism, endocrine homeostasis and systemic chronic inflammation levels. Today, advanced research methods provide new understandings of bone as a crucial endocrine organ. For example, gene editing technology enables bone-specific conditional gene knockout models, which allows the study of bone-derived cytokines to be more precise. We systematically evaluated the various effects of bone-derived cytokines on extraosseous organs and their possible antiaging mechanism. Targeting aging with the current knowledge of the healthy skeletal system is a potential therapeutic strategy. Therefore, we present a comprehensive review that summarizes the current knowledge and provides insights for futures studies.
Collapse
Affiliation(s)
- Xuan-Qi Zheng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Jia-Liang Lin
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Jie Huang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Tong Wu
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Chun-Li Song
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China.
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China.
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China.
| |
Collapse
|
23
|
Deepika F, Bathina S, Armamento-Villareal R. Novel Adipokines and Their Role in Bone Metabolism: A Narrative Review. Biomedicines 2023; 11:biomedicines11020644. [PMID: 36831180 PMCID: PMC9953715 DOI: 10.3390/biomedicines11020644] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/02/2023] [Accepted: 02/14/2023] [Indexed: 02/23/2023] Open
Abstract
The growing burden of obesity and osteoporosis is a major public health concern. Emerging evidence of the role of adipokines on bone metabolism has led to the discovery of novel adipokines over the last decade. Obesity is recognized as a state of adipose tissue inflammation that adversely affects bone health. Adipokines secreted from white adipose tissue (WAT) and bone marrow adipose tissue (BMAT) exerts endocrine and paracrine effects on the survival and function of osteoblasts and osteoclasts. An increase in marrow fat is implicated in osteoporosis and, hence, it is crucial to understand the complex interplay between adipocytes and bone. The objective of this review is to summarize recent advances in our understanding of the role of different adipokines on bone metabolism. METHODS This is a comprehensive review of the literature available in PubMED and Cochrane databases, with an emphasis on the last five years using the keywords. RESULTS Leptin has shown some positive effects on bone metabolism; in contrast, both adiponectin and chemerin have consistently shown a negative association with BMD. No significant association was found between resistin and BMD. Novel adipokines such as visfatin, LCN-2, Nesfatin-1, RBP-4, apelin, and vaspin have shown bone-protective and osteoanabolic properties that could be translated into therapeutic targets. CONCLUSION New evidence suggests the potential role of novel adipokines as biomarkers to predict osteoporosis risk, and as therapeutic targets for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Fnu Deepika
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Translational Research on Inflammatory Disease, Michael E DeBakey Veterans Affairs (VA) Medical Center, Houston, TX 77030, USA
- Correspondence: (F.D.); (R.A.-V.); Tel.: +1-713-794-1414 (R.A.-V.)
| | - Siresha Bathina
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Translational Research on Inflammatory Disease, Michael E DeBakey Veterans Affairs (VA) Medical Center, Houston, TX 77030, USA
| | - Reina Armamento-Villareal
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Translational Research on Inflammatory Disease, Michael E DeBakey Veterans Affairs (VA) Medical Center, Houston, TX 77030, USA
- Correspondence: (F.D.); (R.A.-V.); Tel.: +1-713-794-1414 (R.A.-V.)
| |
Collapse
|
24
|
Cellular and Molecular Mechanisms Associating Obesity to Bone Loss. Cells 2023; 12:cells12040521. [PMID: 36831188 PMCID: PMC9954309 DOI: 10.3390/cells12040521] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
Obesity is an alarming disease that favors the upset of other illnesses and enhances mortality. It is spreading fast worldwide may affect more than 1 billion people by 2030. The imbalance between excessive food ingestion and less energy expenditure leads to pathological adipose tissue expansion, characterized by increased production of proinflammatory mediators with harmful interferences in the whole organism. Bone tissue is one of those target tissues in obesity. Bone is a mineralized connective tissue that is constantly renewed to maintain its mechanical properties. Osteoblasts are responsible for extracellular matrix synthesis, while osteoclasts resorb damaged bone, and the osteocytes have a regulatory role in this process, releasing growth factors and other proteins. A balanced activity among these actors is necessary for healthy bone remodeling. In obesity, several mechanisms may trigger incorrect remodeling, increasing bone resorption to the detriment of bone formation rates. Thus, excessive weight gain may represent higher bone fragility and fracture risk. This review highlights recent insights on the central mechanisms related to obesity-associated abnormal bone. Publications from the last ten years have shown that the main molecular mechanisms associated with obesity and bone loss involve: proinflammatory adipokines and osteokines production, oxidative stress, non-coding RNA interference, insulin resistance, and changes in gut microbiota. The data collection unveils new targets for prevention and putative therapeutic tools against unbalancing bone metabolism during obesity.
Collapse
|
25
|
Martiniakova M, Mondockova V, Biro R, Kovacova V, Babikova M, Zemanova N, Ciernikova S, Omelka R. The link between bone-derived factors osteocalcin, fibroblast growth factor 23, sclerostin, lipocalin 2 and tumor bone metastasis. Front Endocrinol (Lausanne) 2023; 14:1113547. [PMID: 36926025 PMCID: PMC10012867 DOI: 10.3389/fendo.2023.1113547] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/13/2023] [Indexed: 03/04/2023] Open
Abstract
The skeleton is the third most common site of metastatic disease, which causes serious bone complications and short-term prognosis in cancer patients. Prostate and breast cancers are responsible for the majority of bone metastasis, resulting in osteolytic or osteoblastic lesions. The crosstalk between bone cells and their interactions with tumor cells are important in the development of lesions. Recently, both preclinical and clinical studies documented the clinical relevance of bone-derived factors, including osteocalcin (OC) and its undercarboxylated form (ucOC), fibroblast growth factor 23 (FGF23), sclerostin (SCL), and lipocalin 2 (LCN2) as prognostic tumor biomarkers and potential therapeutic targets in bone metastasis. Both OC and ucOC could be useful targets for the prevention of bone metastasis in breast cancer. Moreover, elevated OC level may be a metastatic marker of prostate cancer. FGF23 is particularly important for those forms of cancer that primarily affect bone and/or are characterized by bone metastasis. In other tumor entities, increased FGF23 level is enigmatic. SCL plays a significant role in the pathogenesis of both osteolytic and osteoblastic lesions, as its levels are high in metastatic breast and prostate cancers. Elevated expression levels of LCN2 have been found in aggressive subtypes of cancer. However, its role in anti-metastasis varies significantly between different cancer types. Anyway, all aforementioned bone-derived factors can be used as promising tumor biomarkers. As metastatic bone disease is generally not curable, targeting bone factors represents a new trend in the prevention of bone metastasis and patient care.
Collapse
Affiliation(s)
- Monika Martiniakova
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra, Slovakia
- *Correspondence: Monika Martiniakova, ; Radoslav Omelka,
| | - Vladimira Mondockova
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra, Slovakia
| | - Roman Biro
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra, Slovakia
| | - Veronika Kovacova
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra, Slovakia
| | - Martina Babikova
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra, Slovakia
| | - Nina Zemanova
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra, Slovakia
| | - Sona Ciernikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, Bratislava, Slovakia
| | - Radoslav Omelka
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra, Slovakia
- *Correspondence: Monika Martiniakova, ; Radoslav Omelka,
| |
Collapse
|
26
|
Jover E, Matilla L, Martín-Núñez E, Garaikoetxea M, Navarro A, Fernández-Celis A, Gainza A, Arrieta V, García-Peña A, Álvarez V, Sádaba R, Jaisser F, López-Andrés N. Sex-dependent expression of neutrophil gelatinase-associated lipocalin in aortic stenosis. Biol Sex Differ 2022; 13:71. [PMID: 36510294 PMCID: PMC9743642 DOI: 10.1186/s13293-022-00480-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 11/20/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Accumulating evidence suggest the existence of sex-related differences in the pathogenesis of aortic stenosis (AS) with inflammation, oxidative stress, fibrosis and calcification being over-represented in men. Neutrophil gelatinase-associated lipocalin (NGAL) is expressed in a myriad of tissues and cell types, and it is associated with acute and chronic pathological processes comprising inflammation, fibrosis or calcification. Sex-dependent signatures have been evidenced for NGAL which expression has been associated predominantly in males to metabolic and cardiovascular disorders. We aimed to analyse sex-related differences of NGAL in AS and its role in the inflammatory and fibrocalcific progression of AS. METHODS AND RESULTS 220 (60.45% men) patients with severe AS elective for surgical aortic valve (AV) replacement were recruited. Immunohistochemistry revealed higher expression of NGAL in calcific areas of AVs and that was validated by qPCR in in 65 (60% men) donors. Valve interstitial cells (VICs) were a source of NGAL in these samples. Proteome profiler analyses evidenced higher expression of NGAL in men compared to women, and that was further validated by ELISA. NGAL expression in the AV was correlated with inflammation, oxidative stress, and osteogenic markers, as well as calcium score. The expression of NGAL, both intracellular and secreted (sNGAL), was significantly deregulated only in calcifying male-derived VICs. Depletion of intracellular NGAL in calcifying male-derived VICs was associated with pro-inflammatory profiles, dysbalanced matrix remodelling and pro-osteogenic profiles. Conversely, exogenous NGAL mediated inflammatory and dysbalanced matrix remodelling in calcifying VICs, and all that was prevented by the pharmacological blockade of NGAL. CONCLUSIONS Owing to the over-expression of NGAL, the AV from men may be endowed with higher expression of inflammatory, oxidative stress, matrix remodelling and osteogenic markers supporting the progression of calcific AS phenotypes. The expression of NGAL in the VIC emerges as a potential therapeutic checkpoint, with its effects being potentially reverted by the pharmacological blockade of extracellular NGAL.
Collapse
Affiliation(s)
- Eva Jover
- grid.411730.00000 0001 2191 685XCardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Lara Matilla
- grid.411730.00000 0001 2191 685XCardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Ernesto Martín-Núñez
- grid.411730.00000 0001 2191 685XCardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Mattie Garaikoetxea
- grid.411730.00000 0001 2191 685XCardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Adela Navarro
- grid.411730.00000 0001 2191 685XCardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Amaya Fernández-Celis
- grid.411730.00000 0001 2191 685XCardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Alicia Gainza
- grid.411730.00000 0001 2191 685XCardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Vanessa Arrieta
- grid.411730.00000 0001 2191 685XCardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Amaia García-Peña
- grid.411730.00000 0001 2191 685XCardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Virginia Álvarez
- grid.411730.00000 0001 2191 685XCardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Rafael Sádaba
- grid.411730.00000 0001 2191 685XCardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Frederic Jaisser
- grid.508487.60000 0004 7885 7602Centre de Recherche des Cordeliers, INSERM, UMRS 1138, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris Cité, 15 rue de l’Ecole de Médecine, 75006 Paris, France ,grid.410527.50000 0004 1765 1301Université de Lorraine, INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116, CHRU de Nancy, French-Clinical Research Infrastructure Network (F-CRIN) INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France
| | - Natalia López-Andrés
- grid.411730.00000 0001 2191 685XCardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW Fibroblast growth factor 23 (FGF23) excess is associated with left ventricular hypertrophy (LVH) and early mortality in patients with chronic kidney disease (CKD) and in animal models. Elevated Lipocalin-2 (LCN2), produced by the injured kidneys, contributes to CKD progression and might aggravate cardiovascular outcomes. The current review aims to highlight the role of LCN2 in CKD, particularly its interactions with FGF23. RECENT FINDINGS Inflammation, disordered iron homeostasis and altered metabolic activity are common complications of CKD, and are associated with elevated levels of kidney-produced LCN2 and bone-secreted FGF23. A recent study shows that elevated LCN2 increases FGF23 production, and contributes to cardiac injury in patients and animals with CKD, whereas LCN2 reduction in mice with CKD reduces FGF23, improves cardiovascular outcomes and prolongs lifespan. SUMMARY In this manuscript, we discuss the potential pathophysiological functions of LCN2 as a major kidney-bone crosstalk molecule, linking the progressive decline in kidney function to excessive bone FGF23 production. We also review associations of LCN2 with kidney, cardiovascular and bone and mineral alterations. We conclude that the presented data support the design of novel therapeutic approaches to improve outcomes in CKD.
Collapse
Affiliation(s)
- Guillaume Courbon
- Division of Nephrology and Hypertension, Center for Translational Metabolism and Health, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Valentin David
- Division of Nephrology and Hypertension, Center for Translational Metabolism and Health, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
28
|
Saalbach A, Kunz M. Impact of Chronic Inflammation in Psoriasis on Bone Metabolism. Front Immunol 2022; 13:925503. [PMID: 35812457 PMCID: PMC9259794 DOI: 10.3389/fimmu.2022.925503] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/25/2022] [Indexed: 11/30/2022] Open
Abstract
Psoriasis is a chronic inflammatory disease of the skin and joints associated with several comorbidities such as arthritis, diabetes mellitus and metabolic syndrome, including obesity, hypertension and dyslipidaemia, Crohn's disease, uveitis and psychiatric and psychological diseases. Psoriasis has been described as an independent risk factor for cardiovascular diseases and thus patients with psoriasis should be monitored for the development of cardiovascular disease or metabolic syndrome. However, there is mounting evidence that psoriasis also affects the development of osteoporosis, an important metabolic disease with enormous clinical and socioeconomic impact. At present, there are still controversial opinions about the role of psoriasis in osteoporosis. A more in depth analysis of this phenomenon is of great importance for affected patients since, until now, bone metabolism is not routinely examined in psoriatic patients, which might have important long-term consequences for patients and the health system. In the present review, we summarize current knowledge on the impact of psoriatic inflammation on bone metabolism and osteoporosis.
Collapse
Affiliation(s)
- Anja Saalbach
- Department of Dermatology, Venereology and Allergology, University of Leipzig Medical Center, Leipzig, Germany
| | | |
Collapse
|
29
|
Zhou Y, Zhang C, Zhou Z, Zhang C, Wang J. Identification of Key Genes and Pathways Associated with PIEZO1 in Bone-Related Disease Based on Bioinformatics. Int J Mol Sci 2022; 23:5250. [PMID: 35563641 PMCID: PMC9104149 DOI: 10.3390/ijms23095250] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 04/21/2022] [Accepted: 05/05/2022] [Indexed: 12/20/2022] Open
Abstract
PIEZO1 is a mechano-sensitive ion channel that can sense various forms of mechanical stimuli and convert them into biological signals, affecting bone-related diseases. The present study aimed to identify key genes and signaling pathways in Piezo1-regulated bone-related diseases and to explain the potential mechanisms using bioinformatic analysis. The differentially expressed genes (DEGs) in tendon, femur, and humerus bone tissue; cortical bone; and bone-marrow-derived macrophages were identified with the criteria of |log2FC| > 1 and adjusted p-value < 0.05 analysis based on a dataset from GSE169261, GSE139121, GSE135282, and GSE133069, respectively, and visualized in a volcano plot. Venn diagram analyses were performed to identify the overlapping DEGs expressed in the above-mentioned tissues. Gene Ontology (GO) enrichment analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, protein−protein interaction (PPI) analysis, and module analysis were also conducted. Furthermore, qRT-PCR was performed to validate the above results using primary chondrocytes. As a result, a total of 222 overlapping DEGs and 12 mostly overlapping DEGs were identified. Key Piezo1-related genes, such as Lcn2, Dkk3, Obscn, and Tnnt1, were identified, and pathways, such as Wnt/β-catenin and PI3k-Akt, were also identified. The present informatic study provides insight, for the first time, into the potential therapeutic targets of Piezo1-regulated bone-related diseases
Collapse
Affiliation(s)
- Yuanyuan Zhou
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510006, China; (C.Z.); (C.Z.)
| | - Chen Zhang
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510006, China; (C.Z.); (C.Z.)
| | - Zhongguo Zhou
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane City 4072, Australia;
| | - Chao Zhang
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510006, China; (C.Z.); (C.Z.)
| | - Jiali Wang
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510006, China; (C.Z.); (C.Z.)
| |
Collapse
|
30
|
Plebeian Sage (Salvia plebeia R. Br) Extract Ameliorates Inflammation and Cartilage Degradation in Surgically Induced Osteoarthritis Rats. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12042030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Osteoarthritis (OA), the most prevalent articular disease with the clinical syndrome of joint pain accompanied by varying degrees of functional limitation, reduces the quality of elderly life. In this study, the effects of Plebeian sage extract (PS) on anti-inflammatory and anti-articular cartilage degradation activities were evaluated in rats with surgically induced OA. PS supplement for 12 weeks significantly decreased Mankin scores, including inflammatory cell numbers, and improved surface cartilage damage and mean femur and tibia articular cartilage (AC) thicknesses in OA rats. PS diminished IL-1β, IL-6, TNF-α, MMP-2, MMP-3, and MMP-9, as well as lipocalin-2 levels in serum or cartilage, which were increased due to OA. The results suggested that PS decreased joint inflammation and loss of articular cartilage by suppressing provocative responses and synovial tissue decimation in the OA model. Thus, PS may be used as a novel potential therapeutic regime for OA in the elderly.
Collapse
|
31
|
Dissociation of Bone Resorption and Formation in Spaceflight and Simulated Microgravity: Potential Role of Myokines and Osteokines? Biomedicines 2022; 10:biomedicines10020342. [PMID: 35203551 PMCID: PMC8961781 DOI: 10.3390/biomedicines10020342] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 11/16/2022] Open
Abstract
The dissociation of bone formation and resorption is an important physiological process during spaceflight. It also occurs during local skeletal unloading or immobilization, such as in people with neuromuscular disorders or those who are on bed rest. Under these conditions, the physiological systems of the human body are perturbed down to the cellular level. Through the absence of mechanical stimuli, the musculoskeletal system and, predominantly, the postural skeletal muscles are largely affected. Despite in-flight exercise countermeasures, muscle wasting and bone loss occur, which are associated with spaceflight duration. Nevertheless, countermeasures can be effective, especially by preventing muscle wasting to rescue both postural and dynamic as well as muscle performance. Thus far, it is largely unknown how changes in bone microarchitecture evolve over the long term in the absence of a gravity vector and whether bone loss incurred in space or following the return to the Earth fully recovers or partly persists. In this review, we highlight the different mechanisms and factors that regulate the humoral crosstalk between the muscle and the bone. Further we focus on the interplay between currently known myokines and osteokines and their mutual regulation.
Collapse
|
32
|
Ponzetti M, Ucci A, Maurizi A, Giacchi L, Teti A, Rucci N. Lipocalin 2 Influences Bone and Muscle Phenotype in the MDX Mouse Model of Duchenne Muscular Dystrophy. Int J Mol Sci 2022; 23:ijms23020958. [PMID: 35055145 PMCID: PMC8780970 DOI: 10.3390/ijms23020958] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 01/07/2023] Open
Abstract
Lipocalin 2 (Lcn2) is an adipokine involved in bone and energy metabolism. Its serum levels correlate with bone mechanical unloading and inflammation, two conditions representing hallmarks of Duchenne Muscular Dystrophy (DMD). Therefore, we investigated the role of Lcn2 in bone loss induced by muscle failure in the MDX mouse model of DMD. We found increased Lcn2 serum levels in MDX mice at 1, 3, 6, and 12 months of age. Consistently, Lcn2 mRNA was higher in MDX versus WT muscles. Immunohistochemistry showed Lcn2 expression in mononuclear cells between muscle fibres and in muscle fibres, thus confirming the gene expression results. We then ablated Lcn2 in MDX mice, breeding them with Lcn2−/− mice (MDXxLcn2−/−), resulting in a higher percentage of trabecular volume/total tissue volume compared to MDX mice, likely due to reduced bone resorption. Moreover, MDXxLcn2−/− mice presented with higher grip strength, increased intact muscle fibres, and reduced serum creatine kinase levels compared to MDX. Consistently, blocking Lcn2 by treating 2-month-old MDX mice with an anti-Lcn2 monoclonal antibody (Lcn2Ab) increased trabecular volume, while reducing osteoclast surface/bone surface compared to MDX mice treated with irrelevant IgG. Grip force was also increased, and diaphragm fibrosis was reduced by the Lcn2Ab. These results suggest that Lcn2 could be a possible therapeutic target to treat DMD-induced bone loss.
Collapse
|
33
|
Stunes AK, Brobakken CL, Sujan MAJ, Aagård N, Brevig MS, Wang E, Syversen U, Mosti MP. Acute Effects of Strength and Endurance Training on Bone Turnover Markers in Young Adults and Elderly Men. Front Endocrinol (Lausanne) 2022; 13:915241. [PMID: 35846315 PMCID: PMC9279869 DOI: 10.3389/fendo.2022.915241] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
CONTEXT Exercise is recognized as an important strategy to prevent bone loss, but its acute effects on bone turnover markers (BTMs) and related markers remain uncertain. OBJECTIVE To assess the acute effects of two different exercise modes on BTMs and related markers in young adults of both sexes and elderly men. DESIGN SETTING PARTICIPANTS This was a three-group crossover within-subjects design study with a total of 53 participants-19 young women (aged 22-30), 20 young men (aged 21-30 years), and 14 elderly men (aged 63-74 years)-performing two different exercise sessions [strength training (ST) and high-intensity interval training (HIIT)] separated by 2 weeks, in a supervised laboratory setting. MAIN OUTCOME MEASURES Plasma volume-corrected serum measurements of the BTMs C-terminal telopeptide of type 1 collagen (CTX-I) and procollagen of type 1 N-terminal propeptide (P1NP), total osteocalcin (OC), sclerostin, and lipocalin-2 (LCN2) at baseline, immediately after, and 3 and 24 h after each of the two exercise modes were performed. RESULTS AND CONCLUSION Analyses revealed sex- and age-dependent differences in BTMs and related bone markers at baseline and time-, sex-, and age-dependent differences in response to exercise. No differences between exercise modes were observed for BTM response except for sclerostin in young men and LCN2 in elderly men. An acute, transient, and uniform increase in P1NP/CTX-1 ratio was found in young participants, demonstrating that beneficial skeletal effects on bone metabolism can be attained through both aerobic endurance and resistance exercise, although this effect seems to be attenuated with age. The acute effects of exercise on bone-related biomarkers were generally blunted after 24 h, suggesting that persistent alterations following prolonged exercise interventions should be assessed at later time points.
Collapse
Affiliation(s)
- Astrid Kamilla Stunes
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Medical Clinic, St. Olavs University Hospital, Trondheim, Norway
- *Correspondence: Astrid Kamilla Stunes, orcid.org/0000-0003-1074-5199
| | - Cathrine Langlie Brobakken
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Myworkout, Medical Rehabilitation Clinic, Trondheim, Norway
| | - Md Abu Jafar Sujan
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Norun Aagård
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Myworkout, Medical Rehabilitation Clinic, Trondheim, Norway
| | - Martin Siksjø Brevig
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Eivind Wang
- Faculty of Health and Social Sciences, Molde University College, Molde, Norway
- Department of Psychosis and Rehabilitation, Psychiatry Clinic, St. Olavs University Hospital, Trondheim, Norway
| | - Unni Syversen
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Endocrinology, St. Olavs University Hospital, Trondheim, Norway
| | - Mats Peder Mosti
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Medical Clinic, St. Olavs University Hospital, Trondheim, Norway
- Department of Research and Development, Clinic of Substance Use and Addiction Medicine, St. Olavs University Hospital, Trondheim, Norway
| |
Collapse
|
34
|
Loid P, Hauta-alus H, Mäkitie O, Magnusson P, Mäkitie RE. Lipocalin-2 is associated with FGF23 in WNT1 and PLS3 osteoporosis. Front Endocrinol (Lausanne) 2022; 13:954730. [PMID: 36157448 PMCID: PMC9493469 DOI: 10.3389/fendo.2022.954730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The pathogenic mechanisms of early-onset osteoporosis caused by WNT1 and PLS3 mutations are incompletely understood and diagnostic biomarkers of these disorders are limited. Recently, lipocalin-2 has been recognized as an osteokine involved in bone development and homeostasis. However, the role of lipocalin-2 in WNT1 and PLS3 osteoporosis is unknown. OBJECTIVE We aimed to investigate if plasma lipocalin-2 could be utilized as a biomarker for WNT1 and PLS3 osteoporosis and to evaluate the association between lipocalin-2 and other parameters of bone metabolism. METHODS We measured plasma lipocalin-2 in 17 WNT1 and 14 PLS3 mutation-positive patients and compared them to those of 34 mutation-negative (MN) healthy subjects. We investigated possible associations between lipocalin-2 and several bone biomarkers including collagen type I cross-linked C-telopeptide (CTX), alkaline phosphatase (ALP), type I procollagen intact N-terminal propeptide (PINP), intact and C-terminal fibroblast growth factor 23 (FGF23), dickkopf-1 (DKK1) and sclerostin as well as parameters of iron metabolism (iron, transferrin, transferrin saturation, soluble transferrin receptor and ferritin). RESULTS We found no differences in plasma lipocalin-2 levels in WNT1 or PLS3 patients compared with MN subjects. However, lipocalin-2 was associated with C-terminal FGF23 in WNT1 patients (r=0.62; p=0.008) and PLS3 patients (r=0.63, p=0.017), and with intact FGF23 in PLS3 patients (r=0.80; p<0.001). In addition, lipocalin-2 correlated with serum transferrin in WNT1 patients (r=0.72; p=0.001). CONCLUSION We conclude that plasma lipocalin-2 is not altered in WNT1 or PLS3 mutation-positive subjects but is associated with FGF23 in abnormal WNT1 or PLS3 signaling and with iron status in abnormal WNT1 signaling.
Collapse
Affiliation(s)
- Petra Loid
- Folkhälsan Research Center, Genetics Research Program, Helsinki, Finland
- Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- University of Helsinki, Helsinki, Finland
- *Correspondence: Petra Loid,
| | - Helena Hauta-alus
- Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- University of Helsinki, Helsinki, Finland
- Population Health Unit, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
- Research Unit for Pediatrics, Pediatric Neurology, Pediatric Surgery, Child Psychiatry, Dermatology, Clinical Genetics, Obstetrics and Gynecology, Otorhinolaryngology and Ophthalmology, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Outi Mäkitie
- Folkhälsan Research Center, Genetics Research Program, Helsinki, Finland
- Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- University of Helsinki, Helsinki, Finland
- Department of Molecular Medicine and Surgery, Karolinska Institutet, and Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Per Magnusson
- Department of Clinical Chemistry, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Riikka E. Mäkitie
- Folkhälsan Research Center, Genetics Research Program, Helsinki, Finland
- University of Helsinki, Helsinki, Finland
- Department of Otorhinolaryngology–Head and Neck Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
35
|
Ucci A, Cappariello A, Ponzetti M, Tennant F, Loftus AEP, Shefferd K, Maurizi A, Delle Monache S, Teti A, Rucci N. Anti-osteoblastogenic, pro-inflammatory and pro-angiogenic effect of extracellular vesicles isolated from the human osteosarcoma cell line MNNG/HOS. Bone 2021; 153:116130. [PMID: 34329816 DOI: 10.1016/j.bone.2021.116130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 07/15/2021] [Accepted: 07/21/2021] [Indexed: 01/17/2023]
Abstract
Extracellular Vesicles (EVs) are becoming increasingly recognized as integral signaling vehicles in several types of cancers, including bone malignancies. However, the specific mechanisms by which EVs influence osteosarcoma progression have not been fully determined. We evaluated the effects of EVs derived from the human osteosarcoma cell line MNNG/HOS (MNNG/HOS-EVs) on bone resident cells. We found that MNNG/HOS-EVs are internalized by osteoblasts and osteoclasts in vitro, with potent inhibitory effects on osteoblast metabolic activity, cell density and alkaline phosphatase activity. Consistently, MNNG/HOS-EVs reduced the expression of cell cycle and pro-osteoblastogenic genes, whilst increasing transcriptional expression and protein release of pro-osteoclastogenic/inflammatory cytokines (RankL, Il1b, Il6 and Lcn2), pro-tumoral cytokines (CCL2,5,6,12 and CXCL1,2,5) and the metalloproteinase MMP3. MNNG/HOS-EVs did not induce osteoclast differentiation, while promoting in vitro and in vivo angiogenesis. Intriguingly, EVs derived from another osteosarcoma cell line (U2OS) reduced ALP activity but had no other effect on osteoblast phenotype. MNNG/HOS-EVs were also found to dramatically increase Serpin b2 expression in osteoblasts. To evaluate the significance of this finding, osteoblasts were forced to overexpress Serpin b2, which however did not affect osteoblast differentiation, while Il6 and Lcn2 mRNAs were up regulated. Overall, we shed light on the interactions of osteosarcoma EVs with the cells of the bone microenvironment, identifying key anti-osteoblastogenic, pro-inflammatory and pro-angiogenic factors that could contribute to osteosarcoma expansion.
Collapse
Affiliation(s)
- Argia Ucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Alfredo Cappariello
- Oncohematology Department, IRCCS Bambino Gesù Children's Hospital Research Laboratories, Rome, Italy
| | - Marco Ponzetti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Fabianna Tennant
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | | | - Kirsty Shefferd
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Antonio Maurizi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Simona Delle Monache
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Anna Teti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Nadia Rucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| |
Collapse
|
36
|
Garg P, Strigini M, Peurière L, Vico L, Iandolo D. The Skeletal Cellular and Molecular Underpinning of the Murine Hindlimb Unloading Model. Front Physiol 2021; 12:749464. [PMID: 34737712 PMCID: PMC8562483 DOI: 10.3389/fphys.2021.749464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/23/2021] [Indexed: 01/08/2023] Open
Abstract
Bone adaptation to spaceflight results in bone loss at weight bearing sites following the absence of the stimulus represented by ground force. The rodent hindlimb unloading model was designed to mimic the loss of mechanical loading experienced by astronauts in spaceflight to better understand the mechanisms causing this disuse-induced bone loss. The model has also been largely adopted to study disuse osteopenia and therefore to test drugs for its treatment. Loss of trabecular and cortical bone is observed in long bones of hindlimbs in tail-suspended rodents. Over the years, osteocytes have been shown to play a key role in sensing mechanical stress/stimulus via the ECM-integrin-cytoskeletal axis and to respond to it by regulating different cytokines such as SOST and RANKL. Colder experimental environments (~20-22°C) below thermoneutral temperatures (~28-32°C) exacerbate bone loss. Hence, it is important to consider the role of environmental temperatures on the experimental outcomes. We provide insights into the cellular and molecular pathways that have been shown to play a role in the hindlimb unloading and recommendations to minimize the effects of conditions that we refer to as confounding factors.
Collapse
Affiliation(s)
- Priyanka Garg
- INSERM, U1059 Sainbiose, Université Jean Monnet, Mines Saint-Étienne, Université de Lyon, Saint-Étienne, France
| | - Maura Strigini
- INSERM, U1059 Sainbiose, Université Jean Monnet, Mines Saint-Étienne, Université de Lyon, Saint-Étienne, France
| | - Laura Peurière
- INSERM, U1059 Sainbiose, Université Jean Monnet, Mines Saint-Étienne, Université de Lyon, Saint-Étienne, France
| | - Laurence Vico
- INSERM, U1059 Sainbiose, Université Jean Monnet, Mines Saint-Étienne, Université de Lyon, Saint-Étienne, France
| | - Donata Iandolo
- INSERM, U1059 Sainbiose, Université Jean Monnet, Mines Saint-Étienne, Université de Lyon, Saint-Étienne, France
| |
Collapse
|
37
|
Jia XY, Wei K, Chen J, Xi LH, Kong XL, Wei Y, Wang L, Wang ZS, Liu YP, Liang LM, Xu DM. Association of plasma neutrophil gelatinase-associated lipocalin with parameters of CKD-MBD in maintenance hemodialysis patients. J Bone Miner Metab 2021; 39:1058-1065. [PMID: 34392464 DOI: 10.1007/s00774-021-01248-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 06/30/2021] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Neutrophil gelatinase-associated lipocalin (NGAL) is not only a biomarker of kidney injury but also a bone-derived factor involved in metabolism. We aimed to explore relationships between plasma NGAL and chronic kidney disease-mineral bone disorder (CKD-MBD) parameters in maintenance hemodialysis (MHD) patients. MATERIALS AND METHODS First, a cross sectional observational study, including 105 MHD patients, was conducted to explore relationships between plasma NGAL levels and CKD-MBD parameters. Second, impact of parathyroidectomy and auto-transplantation (PTX + AT) on plasma NGAL was investigated in 12 MHD patients with severe secondary hyperparathyroidism (SHPT). RESULTS According to Spearman correlation analysis, plasma NGAL levels were positively correlated with female (r = 0.243, P = 0.012), vintage (r = 0.290, P = 0.003), Klotho (r = 0.234, P = 0.016), calcium(Ca) (r = 0.332, P = 0.001), alkaline phosphatase (ALP) (r = 0.401, P < 0.001) and intact parathyroid hormone (iPTH) (r = 0.256, P = 0.008); while inversely correlated with albumin(Alb) (r = - 0.201, P = 0.039). After adjusting for age, sex, vintage, Alb and all parameters of CKD-MBD(Ca, P, lg(ALP), lg(iPTH), Klotho and fibroblast growth factor 23(FGF23)), lg(NGAL) were positively correlated with Ca (r = 0.481, P < 0.001), P (r = 0.336, P = 0.037), lg(ALP) (r = 0.646, P < 0.001) in Partial correlation analysis; further multiple linear regression analysis showed same positive associations between lg(NGAL) and Ca (β = 0.330, P = 0.002), P (β = 0.218, P = 0.037), lg(ALP) (β = 0.671, P < 0.001). During the 4-7 days after PTX + AT, plasma NGAL decreased from 715.84 (578.73, 988.14) to 688.42 (660.00, 760.26) ng/mL (P = 0.071), Klotho increased from 496.45 (341.73, 848.30) to 1138.25 (593.87, 2009.27) pg/mL (P = 0.099). CONCLUSION Plasma NGAL levels were positively associated with ALP in MHD patients; and downtrends were shown after PTX + AT in patients with severe SHPT. These findings suggest that NGAL is a participant in CKD-MBD under MHD condition.
Collapse
Affiliation(s)
- Xiao-Yan Jia
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, No.16766, Jingshi Road, Jinan, 250014, China
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Provincial Insititute of Nephrology, Jinan, China
| | - Kai Wei
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, No.16766, Jingshi Road, Jinan, 250014, China
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Provincial Insititute of Nephrology, Jinan, China
| | - Juan Chen
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, No.16766, Jingshi Road, Jinan, 250014, China
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Provincial Insititute of Nephrology, Jinan, China
| | - Lin-He Xi
- Department of Plastic and Reconstructive Surgery, Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Xiang-Lei Kong
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, No.16766, Jingshi Road, Jinan, 250014, China
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Provincial Insititute of Nephrology, Jinan, China
| | - Yong Wei
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, No.16766, Jingshi Road, Jinan, 250014, China
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Provincial Insititute of Nephrology, Jinan, China
| | - Li Wang
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, No.16766, Jingshi Road, Jinan, 250014, China
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Provincial Insititute of Nephrology, Jinan, China
| | - Zun-Song Wang
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, No.16766, Jingshi Road, Jinan, 250014, China
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Provincial Insititute of Nephrology, Jinan, China
| | - Yi-Peng Liu
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, No.16766, Jingshi Road, Jinan, 250014, China
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Provincial Insititute of Nephrology, Jinan, China
| | - Li-Ming Liang
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, No.16766, Jingshi Road, Jinan, 250014, China
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Provincial Insititute of Nephrology, Jinan, China
| | - Dong-Mei Xu
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, No.16766, Jingshi Road, Jinan, 250014, China.
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, China.
- Shandong Provincial Insititute of Nephrology, Jinan, China.
| |
Collapse
|
38
|
Zhao Y, Suo Y, Yang Z, Hao Y, Li W, Su Y, Shi Y, Gao Y, Song L, Yin X, Shi H. Inspiration for the prevention and treatment of neuropsychiatric disorders: New insight from the bone-brain-axis. Brain Res Bull 2021; 177:263-272. [PMID: 34678443 DOI: 10.1016/j.brainresbull.2021.10.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 10/09/2021] [Accepted: 10/15/2021] [Indexed: 11/30/2022]
Abstract
Bone is the main supporting structure of the body and the main organ involved in body movement and calcium and phosphorus metabolism. Recent studies have shown that bone is also a potential new endocrine organ that participates in the physiological and pathophysiological processes of the cardiovascular, digestive, and endocrine systems through various bioactive cytokines secreted by bone cells and bone marrow. Bone-derived active cytokines can also directly act on the central nervous system and regulate brain function and individual behavior. The bidirectional regulation of the bone-brain axis has gradually attracted attention in the field of neuroscience. This paper reviews the regulatory effects of bone-derived active cytokines and bone-derived cells on individual brain function and brain diseases, as well as the occurrence and development of related neuropsychiatric diseases. The central regulatory mechanism function is briefly introduced, which will broaden the scope for mechanistic research and help establish prevention and treatment strategies for neuropsychiatric diseases based on the bone-brain axis.
Collapse
Affiliation(s)
- Ye Zhao
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key laboratory of Neurophysiology, Hebei Medicinal University, 050017, China
| | - Yining Suo
- Child Health Department, Hebei Children's Hospital, Shijiazhuang 050031, China
| | - Zhenbang Yang
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key laboratory of Neurophysiology, Hebei Medicinal University, 050017, China
| | - Ying Hao
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key laboratory of Neurophysiology, Hebei Medicinal University, 050017, China
| | - Wenshuya Li
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key laboratory of Neurophysiology, Hebei Medicinal University, 050017, China
| | - Yujiao Su
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key laboratory of Neurophysiology, Hebei Medicinal University, 050017, China
| | - Yun Shi
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang 050017, China; Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medicinal University, Shijiazhuang 050017, China
| | - Yuan Gao
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key laboratory of Neurophysiology, Hebei Medicinal University, 050017, China; Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medicinal University, Shijiazhuang 050017, China
| | - Li Song
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key laboratory of Neurophysiology, Hebei Medicinal University, 050017, China; Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medicinal University, Shijiazhuang 050017, China
| | - Xi Yin
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang 050017, China; Department of Functional Region of Diagnosis, Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China.
| | - Haishui Shi
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key laboratory of Neurophysiology, Hebei Medicinal University, 050017, China; Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medicinal University, Shijiazhuang 050017, China.
| |
Collapse
|
39
|
Spatz JM, Ko FC, Ayturk UM, Warman ML, Bouxsein ML. RNAseq and RNA molecular barcoding reveal differential gene expression in cortical bone following hindlimb unloading in female mice. PLoS One 2021; 16:e0250715. [PMID: 34637435 PMCID: PMC8509868 DOI: 10.1371/journal.pone.0250715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/12/2021] [Indexed: 11/24/2022] Open
Abstract
Disuse-induced bone loss is seen following spinal cord injury, prolonged bed rest, and exposure to microgravity. We performed whole transcriptomic profiling of cortical bone using RNA sequencing (RNAseq) and RNA molecular barcoding (NanoString) on a hindlimb unloading (HLU) mouse model to identify genes whose mRNA transcript abundances change in response to disuse. Eleven-week old female C57BL/6 mice were exposed to ambulatory loading or HLU for 7 days (n = 8/group). Total RNA from marrow-flushed femoral cortical bone was analyzed on HiSeq and NanoString platforms. The expression of several previously reported genes associated with Wnt signaling and metabolism was altered by HLU. Furthermore, the increased abundance of transcripts, such as Pfkfb3 and Mss51, after HLU imply these genes also have roles in the cortical bone’s response to altered mechanical loading. Our study demonstrates that an unbiased approach to assess the whole transcriptomic profile of cortical bone can reveal previously unidentified mechanosensitive genes and may eventually lead to novel targets to prevent disuse-induced osteoporosis.
Collapse
Affiliation(s)
- Jordan M Spatz
- Center for Advanced Orthopedic Studies, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America.,University of California San Francisco School of Medicine, San Francisco, California, United States of America.,Harvard Medical School, Boston, Massachusetts, United States of America
| | - Frank C Ko
- Center for Advanced Orthopedic Studies, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America.,Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ugur M Ayturk
- Harvard Medical School, Boston, Massachusetts, United States of America.,Boston Children's Hospital, Boston, Massachusetts, United States of America
| | - Matthew L Warman
- Harvard Medical School, Boston, Massachusetts, United States of America.,Boston Children's Hospital, Boston, Massachusetts, United States of America
| | - Mary L Bouxsein
- Center for Advanced Orthopedic Studies, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America.,Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
40
|
Cappariello A, Rucci N. Extracellular Vesicles in Bone Tumors: How to Seed in the Surroundings Molecular Information for Malignant Transformation and Progression. Front Oncol 2021; 11:722922. [PMID: 34616676 PMCID: PMC8488258 DOI: 10.3389/fonc.2021.722922] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/26/2021] [Indexed: 12/19/2022] Open
Abstract
Bone is a very dynamic tissue hosting different cell types whose functions are regulated by a plethora of membrane-bound and soluble molecules. Intercellular communication was recently demonstrated to be also sustained by the exchange of extracellular vesicles (EVs). These are cell-derived nanosized structures shuttling biologically active molecules, such as nucleic acids and proteins. The bone microenvironment is a preferential site of primary and metastatic tumors, in which cancer cells find a fertile soil to “seed and blossom”. Nowadays, many oncogenic processes are recognized to be sustained by EVs. For example, EVs can directly fuel the vicious cycle in the bone/bone marrow microenvironment. EVs create a favourable environment for tumor growth by affecting osteoblasts, osteoclasts, osteocytes, adipocytes, leukocytes, and endothelial cells. At the same time other crucial tumor-mediated events, such as the premetastatic niche formation, tumor cell dormancy, as well as drug resistance, have been described to be fostered by tumor-derived EVs. In this review, we will discuss the main body of literature describing how the cancer cells use the EVs for their growth into the bone and for educating the bone microenvironment to host metastases.
Collapse
Affiliation(s)
- Alfredo Cappariello
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Nadia Rucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
41
|
Dekens DW, Eisel ULM, Gouweleeuw L, Schoemaker RG, De Deyn PP, Naudé PJW. Lipocalin 2 as a link between ageing, risk factor conditions and age-related brain diseases. Ageing Res Rev 2021; 70:101414. [PMID: 34325073 DOI: 10.1016/j.arr.2021.101414] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/12/2022]
Abstract
Chronic (neuro)inflammation plays an important role in many age-related central nervous system (CNS) diseases, including Alzheimer's disease, Parkinson's disease and vascular dementia. Inflammation also characterizes many conditions that form a risk factor for these CNS disorders, such as physical inactivity, obesity and cardiovascular disease. Lipocalin 2 (Lcn2) is an inflammatory protein shown to be involved in different age-related CNS diseases, as well as risk factor conditions thereof. Lcn2 expression is increased in the periphery and the brain in different age-related CNS diseases and also their risk factor conditions. Experimental studies indicate that Lcn2 contributes to various neuropathophysiological processes of age-related CNS diseases, including exacerbated neuroinflammation, cell death and iron dysregulation, which may negatively impact cognitive function. We hypothesize that increased Lcn2 levels as a result of age-related risk factor conditions may sensitize the brain and increase the risk to develop age-related CNS diseases. In this review we first provide a comprehensive overview of the known functions of Lcn2, and its effects in the CNS. Subsequently, this review explores Lcn2 as a potential (neuro)inflammatory link between different risk factor conditions and the development of age-related CNS disorders. Altogether, evidence convincingly indicates Lcn2 as a key constituent in ageing and age-related brain diseases.
Collapse
Affiliation(s)
- Doortje W Dekens
- Department of Neurology and Alzheimer Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Ulrich L M Eisel
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Leonie Gouweleeuw
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Regien G Schoemaker
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Peter P De Deyn
- Department of Neurology and Alzheimer Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Laboratory of Neurochemistry and Behaviour, Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Petrus J W Naudé
- Department of Neurology and Alzheimer Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands; Department of Psychiatry and Mental Health and Neuroscience Institute, Brain Behaviour Unit, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
42
|
Ponzetti M, Aielli F, Ucci A, Cappariello A, Lombardi G, Teti A, Rucci N. Lipocalin 2 increases after high-intensity exercise in humans and influences muscle gene expression and differentiation in mice. J Cell Physiol 2021; 237:551-565. [PMID: 34224151 PMCID: PMC9291458 DOI: 10.1002/jcp.30501] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 06/04/2021] [Accepted: 06/25/2021] [Indexed: 12/22/2022]
Abstract
Lipocalin 2 (LCN2) is an adipokine that accomplishes several functions in diverse organs. However, its importance in muscle and physical exercise is currently unknown. We observed that following acute high‐intensity exercise (“Gran Sasso d'Italia” vertical run), LCN2 serum levels were increased. The Wnt pathway antagonist, DKK1, was also increased after the run, positively correlating with LCN2, and the same was found for the cytokine Interleukin 6. We, therefore, investigated the involvement of LCN2 in muscle physiology employing an Lcn2 global knockout (Lcn2−/−) mouse model. Lcn2−/− mice presented with smaller muscle fibres but normal muscle performance (grip strength metre) and muscle weight. At variance with wild type (WT) mice, the inflammatory cytokine Interleukin 6 was undetectable in Lcn2−/− mice at all ages. Intriguingly, Lcn2−/− mice did not lose gastrocnemius and quadriceps muscle mass and muscle performance following hindlimb suspension, while at variance with WT, they lose soleus muscle mass. In vitro, LCN2 treatment reduced the myogenic differentiation of C2C12 and primary mouse myoblasts and influenced their gene expression. Treating myoblasts with LCN2 reduced myogenesis, suggesting that LCN2 may negatively affect muscle physiology when upregulated following high‐intensity exercise.
Collapse
Affiliation(s)
- Marco Ponzetti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Federica Aielli
- Medical Oncology Department, Giuseppe Mazzini Hospital, Teramo, Italy
| | - Argia Ucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Alfredo Cappariello
- Research Laboratories, Department of Onco-haematology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry & Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Poland
| | - Anna Teti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Nadia Rucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
43
|
Gerosa L, Lombardi G. Bone-to-Brain: A Round Trip in the Adaptation to Mechanical Stimuli. Front Physiol 2021; 12:623893. [PMID: 33995117 PMCID: PMC8120436 DOI: 10.3389/fphys.2021.623893] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
Besides the classical ones (support/protection, hematopoiesis, storage for calcium, and phosphate) multiple roles emerged for bone tissue, definitively making it an organ. Particularly, the endocrine function, and in more general terms, the capability to sense and integrate different stimuli and to send signals to other tissues, has highlighted the importance of bone in homeostasis. Bone is highly innervated and hosts all nervous system branches; bone cells are sensitive to most of neurotransmitters, neuropeptides, and neurohormones that directly affect their metabolic activity and sensitivity to mechanical stimuli. Indeed, bone is the principal mechanosensitive organ. Thanks to the mechanosensing resident cells, and particularly osteocytes, mechanical stimulation induces metabolic responses in bone forming (osteoblasts) and bone resorbing (osteoclasts) cells that allow the adaptation of the affected bony segment to the changing environment. Once stimulated, bone cells express and secrete, or liberate from the entrapping matrix, several mediators (osteokines) that induce responses on distant targets. Brain is a target of some of these mediator [e.g., osteocalcin, lipocalin2, sclerostin, Dickkopf-related protein 1 (Dkk1), and fibroblast growth factor 23], as most of them can cross the blood-brain barrier. For others, a role in brain has been hypothesized, but not yet demonstrated. As exercise effectively modifies the release and the circulating levels of these osteokines, it has been hypothesized that some of the beneficial effects of exercise on brain functions may be associated to such a bone-to-brain communication. This hypothesis hides an interesting clinical clue: may well-addressed physical activities support the treatment of neurodegenerative diseases, such as Alzheimer’s and Parkinson’s diseases?
Collapse
Affiliation(s)
| | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry & Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy.,Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Poland
| |
Collapse
|
44
|
Lipocalin 2 serum levels correlate with age and bone turnover biomarkers in healthy subjects but not in postmenopausal osteoporotic women. Bone Rep 2021; 14:101059. [PMID: 34026950 PMCID: PMC8121999 DOI: 10.1016/j.bonr.2021.101059] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 03/16/2021] [Accepted: 03/24/2021] [Indexed: 12/25/2022] Open
Abstract
Purpose Lipocalin 2 (LCN2) is an adipokine involved in many physiological functions, including bone metabolism. We previously demonstrated its implication in mouse models of mechanical unloading-induced osteoporosis and in a cohort of bed rest volunteers. We therefore aimed at studying its involvement in postmenopausal osteoporosis. Methods We measured serum LCN2 and correlated its levels to Dickkopf WNT Signaling Pathway Inhibitor 1 (DKK1), Tartrate Resistant Acid Phosphatase 5B (TRAcP5B), sclerostin, urinary N-terminal telopeptide of type I collagen (NTX), serum C-terminal telopeptide of type I collagen (CTX), parathyroid hormone and vitamin K by ELISA performed in a cohort of younger (50–65 years) and older (66–90 years) osteoporotic women in comparison to healthy subjects. A cohort of male healthy and osteoarthritic patients was also included. Sobel mediation analysis was used to test indirect associations among age, LCN2 and DKK1 or NTX. Results LCN2 levels were unchanged in osteoporotic and in osteoarthritis patients when compared to healthy subjects and did not correlate with BMD. However, serum LCN2 correlated with age in healthy women (R = 0.44; P = 0.003) and men (R = 0.5; P = 0.001) and serum concentrations of DKK1 (R = 0.47; P = 0.003) and urinary NTX (R = 0.34; P = 0.04). Sobel mediation analysis showed that LCN2 mediates an indirect relationship between age and DKK1 (P = 0.02), but not with NTX, in healthy subjects. Conclusions Taken together, the results suggest a hitherto unknown association between LCN2, DKK1 and age in healthy individuals, but not in postmenopausal osteoporotic women.
Collapse
Key Words
- BALP, bone-specific alkaline phosphatase
- BMD, bone mineral density
- BMI, body mass index
- CTX, C-terminal telopeptide of type I collagen
- DKK1
- DKK1, Dickkopf WNT Signaling Pathway Inhibitor 1
- IL, interleukin
- LCN2, lipocalin 2
- Lipocalin-2
- NGAL
- NTX, N-terminal telopeptide of type I collagen
- NfκB, nuclear factor kappa-B
- Osteoarthritis
- Osteoporosis
- PTH, parathyroid hormone
- RANKL, receptor activator of nuclear factor kappa-B
- TNF, tumor necrosis factor
- TRAcP5B, tartrate-resistant acid phosphatase 5B
- Wnt
Collapse
|
45
|
Tang MJ, Graham HK, Davidson KE. Botulinum Toxin A and Osteosarcopenia in Experimental Animals: A Scoping Review. Toxins (Basel) 2021; 13:213. [PMID: 33799488 PMCID: PMC8001269 DOI: 10.3390/toxins13030213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/11/2021] [Accepted: 03/11/2021] [Indexed: 01/24/2023] Open
Abstract
We conducted a scoping review to investigate the effects of intramuscular injection of Botulinum Toxin A (BoNT-A) on bone morphology. We investigated if the muscle atrophy associated with Injection of BoNT-A had effects on the neighboring bone. We used the search terms: osteopenia, bone atrophy, Botulinum Toxin A, Micro-CT, mice or rat. The following databases were searched: Medline, Embase, PubMed and the Cochrane Library, between 1990 and 2020. After removal of duplicates, 228 abstracts were identified of which 49 studies satisfied our inclusion and exclusion criteria. The majority of studies (41/49) reported a quantitative reduction in at least one measure of bone architecture based on Micro-CT. The reduction in the ratio of bone volume to tissue volume varied from 11% to 81% (mean 43%) according to the experimental set up and study time points. While longer term studies showed muscle recovery, no study showed complete recovery of all bone properties at the termination of the study. In experimental animals, intramuscular injection of BoNT-A resulted in acute muscle atrophy and acute degradation of the neighboring bone segment. These findings may have implications for clinical protocols in the use of Botulinum Toxin in children with cerebral palsy, with restraint recommended in injection protocols and consideration for monitoring bone density. Clinical studies in children with cerebral palsy receiving injections of Botulinum are indicated.
Collapse
Affiliation(s)
- Min Jia Tang
- Department of Orthopaedics, The Royal Children’s Hospital, Parkville, VIC 3052, Australia;
| | - H. Kerr Graham
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3010, Australia;
| | - Kelsey E. Davidson
- Department of Orthopedics, Shriners Hospitals for Children, Chicago, IL 60707, USA
| |
Collapse
|
46
|
Al-Absi B, AL-Habori M, Saif-Ali R. Plasma Lipocalin-2 and Adiponectin are Affected by Obesity Rather Than Type 2 Diabetes Mellitus per se. Diabetes Metab Syndr Obes 2021; 14:4547-4556. [PMID: 34815681 PMCID: PMC8605802 DOI: 10.2147/dmso.s338254] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/03/2021] [Indexed: 04/20/2023] Open
Abstract
PURPOSE Changes in plasma adipocytokines and inflammatory markers in type 2 DM remain controversial as to whether they are due to obesity or directly associated with the diabetic state. Our objective was to study the effect of obesity and diabetes on plasma lipocalin-2 (LCN2), adiponectin, and interleukin-1β (IL-1β) by comparing their levels in non-diabetic obese subjects and non-obese type 2 DM patients, as well as determining the association of these adipocytokines with metabolic syndrome factors and diabetic parameters. PATIENTS AND METHODS In this study, 85 Yemeni male volunteers aged 30-60 years old were enrolled, 25 of whom were healthy subjects with BMI < 25 kg/m2 served as control; 30 non-diabetic obese subjects (BMI ≥ 30 kg/m2 and FBG < 6.1 mmol/l); and 30 non-obese type 2 DM patients (BMI < 25 kg/m2 and FBG > 7 mmol/l). RESULTS Lipocalin-2 and adiponectin were significantly (p = 0.043 and p = 0.034) lower in non-diabetic obese subjects by 16.2% and 29.7% with respect to control group, with no effect in the non-obese type 2 DM patients. Moreover, LCN2 was significantly (p = 0.04) lower in the non-diabetic obese subjects by 15.8% as compared with the non-obese type 2 DM patients, with no significant difference in adiponectin levels. In contrast, serum IL-1β was significantly (p = 0.001 and p = 0.003) higher in both non-diabetic obese subjects and the non-obese type 2 DM patients by 76.5% and 67.7% as compared to control group. The significant decrease in both LCN2 and adiponectin and the significant increase in IL-1β in the non-diabetic obese subjects disappeared upon adjustment for waist circumference (WC). In contrast, the significant increase in IL-1β in the non-obese Type 2 DM patients was not affected upon adjustment for WC. CONCLUSION Plasma LCN2 and adiponectin were not affected by diabetes per se, suggesting that the observed changes in LCN2 and adiponectin in type 2 DM may be due to obesity rather than the diabetic state, whereas IL-1β levels were affected by both obesity and diabetes.
Collapse
Affiliation(s)
- Boshra Al-Absi
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Health Sciences, University of Sana`a, Sana`a, Yemen
| | - Molham AL-Habori
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Health Sciences, University of Sana`a, Sana`a, Yemen
- Correspondence: Molham AL-Habori Email
| | - Riyadh Saif-Ali
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Health Sciences, University of Sana`a, Sana`a, Yemen
| |
Collapse
|
47
|
Petropoulou PI, Mosialou I, Shikhel S, Hao L, Panitsas K, Bisikirska B, Luo N, Bahna F, Kim J, Carberry P, Zanderigo F, Simpson N, Bakalian M, Kassir S, Shapiro L, Underwood MD, May CM, Soligapuram Sai KK, Jorgensen MJ, Confavreux CB, Shapses S, Laferrère B, Mintz A, Mann JJ, Rubin M, Kousteni S. Lipocalin-2 is an anorexigenic signal in primates. eLife 2020; 9:58949. [PMID: 33231171 PMCID: PMC7685704 DOI: 10.7554/elife.58949] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022] Open
Abstract
In the mouse, the osteoblast-derived hormone Lipocalin-2 (LCN2) suppresses food intake and acts as a satiety signal. We show here that meal challenges increase serum LCN2 levels in persons with normal or overweight, but not in individuals with obesity. Postprandial LCN2 serum levels correlate inversely with hunger sensation in challenged subjects. We further show through brain PET scans of monkeys injected with radiolabeled recombinant human LCN2 (rh-LCN2) and autoradiography in baboon, macaque, and human brain sections, that LCN2 crosses the blood-brain barrier and localizes to the hypothalamus in primates. In addition, daily treatment of lean monkeys with rh-LCN2 decreases food intake by 21%, without overt side effects. These studies demonstrate the biology of LCN2 as a satiety factor and indicator and anorexigenic signal in primates. Failure to stimulate postprandial LCN2 in individuals with obesity may contribute to metabolic dysregulation, suggesting that LCN2 may be a novel target for obesity treatment.
Collapse
Affiliation(s)
| | - Ioanna Mosialou
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, United States
| | - Steven Shikhel
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, United States
| | - Lihong Hao
- Department of Nutritional Sciences, Rutgers University, New Brunswick, United States
| | - Konstantinos Panitsas
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, United States
| | - Brygida Bisikirska
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, United States
| | - Na Luo
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, United States
| | - Fabiana Bahna
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, United States
| | - Jongho Kim
- Department of Radiology, Columbia University Medical Center, New York, United States
| | - Patrick Carberry
- Department of Radiology, Columbia University Medical Center, New York, United States
| | - Francesca Zanderigo
- Department of Psychiatry, Columbia University Medical Center, New York, United States.,Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, United States
| | - Norman Simpson
- Department of Psychiatry, Columbia University Medical Center, New York, United States
| | - Mihran Bakalian
- Department of Psychiatry, Columbia University Medical Center, New York, United States
| | - Suham Kassir
- Department of Psychiatry, Columbia University Medical Center, New York, United States.,Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, United States
| | - Lawrence Shapiro
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, United States
| | - Mark D Underwood
- Department of Psychiatry, Columbia University Medical Center, New York, United States.,Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, United States
| | - Christina M May
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, United States
| | | | - Matthew J Jorgensen
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, United States
| | | | - Sue Shapses
- Department of Nutritional Sciences, Rutgers University, New Brunswick, United States.,Department of Medicine, Rutgers - RWJ Medical School, Rutgers University, New Brunswick, United States
| | - Blandine Laferrère
- New York Obesity Nutrition Research Center, Columbia University, New York, United States.,Department of Medicine, Division of Endocrinology, Columbia University Irving Medical Center, New York, United States
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, United States
| | - J John Mann
- Department of Radiology, Columbia University Medical Center, New York, United States.,Department of Psychiatry, Columbia University Medical Center, New York, United States.,Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, United States
| | - Mishaela Rubin
- New York Obesity Nutrition Research Center, Columbia University, New York, United States
| | - Stavroula Kousteni
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, United States
| |
Collapse
|
48
|
Carina V, Della Bella E, Costa V, Bellavia D, Veronesi F, Cepollaro S, Fini M, Giavaresi G. Bone's Response to Mechanical Loading in Aging and Osteoporosis: Molecular Mechanisms. Calcif Tissue Int 2020; 107:301-318. [PMID: 32710266 DOI: 10.1007/s00223-020-00724-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/08/2020] [Indexed: 02/07/2023]
Abstract
Mechanotransduction is pivotal in the maintenance of homeostasis in different tissues and involves multiple cell signaling pathways. In bone, mechanical stimuli regulate the balance between bone formation and resorption; osteocytes play a central role in this regulation. Dysfunctions in mechanotransduction signaling or in osteocytes response lead to an imbalance in bone homeostasis. This alteration is very relevant in some conditions such as osteoporosis and aging. Both are characterized by increased bone weakness due to different causes, for example, the increase of osteocyte apoptosis that cause an alteration of fluid space, or the alteration of molecular pathways. There are intertwined yet very different mechanisms involved among the cell-intrinsic effects of aging on bone, the cell-intrinsic and tissue-level effects of estrogen/androgen withdrawal on bone, and the effects of reduced mechanical loading on bone, which are all involved to some degree in how aged bone fails to respond properly to stress/strain compared to younger bone. This review aims at clarifying how the cellular and molecular pathways regulated and induced in bone by mechanical stimulation are altered with aging and in osteoporosis, to highlight new possible targets for antiresorptive or anabolic bone therapeutic approaches.
Collapse
Affiliation(s)
- Valeria Carina
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Via Di Barbiano, 1/10, 40136, Bologna, Italy.
| | | | - Viviana Costa
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Via Di Barbiano, 1/10, 40136, Bologna, Italy
| | - Daniele Bellavia
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Via Di Barbiano, 1/10, 40136, Bologna, Italy
| | - Francesca Veronesi
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Via Di Barbiano, 1/10, 40136, Bologna, Italy
| | - Simona Cepollaro
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Via Di Barbiano, 1/10, 40136, Bologna, Italy
| | - Milena Fini
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Via Di Barbiano, 1/10, 40136, Bologna, Italy
| | - Gianluca Giavaresi
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Via Di Barbiano, 1/10, 40136, Bologna, Italy
| |
Collapse
|
49
|
Lin X, Onda DA, Yang CH, Lewis JR, Levinger I, Loh K. Roles of bone-derived hormones in type 2 diabetes and cardiovascular pathophysiology. Mol Metab 2020; 40:101040. [PMID: 32544571 PMCID: PMC7348059 DOI: 10.1016/j.molmet.2020.101040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/28/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023] Open
Abstract
Background Emerging evidence demonstrates that bone is an endocrine organ capable of influencing multiple physiological and pathological processes through the secretion of hormones. Recent research suggests complex crosstalk between the bone and other metabolic and cardiovascular tissues. It was uncovered that three of these bone-derived hormones—osteocalcin, lipocalin 2, and sclerostin—are involved in the endocrine regulations of cardiometabolic health and play vital roles in the pathophysiological process of developing cardiometabolic syndromes such as type 2 diabetes and cardiovascular disease. Chronic low-grade inflammation is one of the hallmarks of cardiometabolic diseases and a major contributor to disease progression. Novel evidence also implicates important roles of bone-derived hormones in the regulation of chronic inflammation. Scope of review In this review, we provide a detailed overview of the physiological and pathological roles of osteocalcin, lipocalin 2, and sclerostin in cardiometabolic health regulation and disease development, with a focus on the modulation of chronic inflammation. Major conclusions Evidence supports that osteocalcin has a protective role in cardiometabolic health, and an increase of lipocalin 2 contributes to the development of cardiometabolic diseases partly via pro-inflammatory effects. The roles of sclerostin appear to be complicated: It exerts pro-adiposity and pro-insulin resistance effects in type 2 diabetes and has an anti-calcification effect during cardiovascular disease. A better understanding of the actions of these bone-derived hormones in the pathophysiology of cardiometabolic diseases will provide crucial insights to help further research develop new therapeutic strategies to treat these diseases.
Collapse
Affiliation(s)
- Xuzhu Lin
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia.
| | - Danise-Ann Onda
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
| | - Chieh-Hsin Yang
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
| | - Joshua R Lewis
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia; Medical School, University of Western Australia, Perth, Australia
| | - Itamar Levinger
- Institute for Health and Sport (IHES), Victoria University, Footscray, VIC, Australia; Australian Institute for Musculoskeletal Science (AIMSS), University of Melbourne and Western Health, St Albans, VIC, Australia
| | - Kim Loh
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia; Department of Medicine, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
50
|
Qin L, Liu W, Cao H, Xiao G. Molecular mechanosensors in osteocytes. Bone Res 2020; 8:23. [PMID: 32550039 PMCID: PMC7280204 DOI: 10.1038/s41413-020-0099-y] [Citation(s) in RCA: 222] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/07/2020] [Accepted: 04/17/2020] [Indexed: 12/11/2022] Open
Abstract
Osteocytes, the most abundant and long-lived cells in bone, are the master regulators of bone remodeling. In addition to their functions in endocrine regulation and calcium and phosphate metabolism, osteocytes are the major responsive cells in force adaptation due to mechanical stimulation. Mechanically induced bone formation and adaptation, disuse-induced bone loss and skeletal fragility are mediated by osteocytes, which sense local mechanical cues and respond to these cues in both direct and indirect ways. The mechanotransduction process in osteocytes is a complex but exquisite regulatory process between cells and their environment, between neighboring cells, and between different functional mechanosensors in individual cells. Over the past two decades, great efforts have focused on finding various mechanosensors in osteocytes that transmit extracellular mechanical signals into osteocytes and regulate responsive gene expression. The osteocyte cytoskeleton, dendritic processes, Integrin-based focal adhesions, connexin-based intercellular junctions, primary cilium, ion channels, and extracellular matrix are the major mechanosensors in osteocytes reported so far with evidence from both in vitro and in vitro studies. This review aims to give a systematic introduction to osteocyte mechanobiology, provide details of osteocyte mechanosensors, and discuss the roles of osteocyte mechanosensitive signaling pathways in the regulation of bone homeostasis.
Collapse
Affiliation(s)
- Lei Qin
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, and School of Medicine, Southern University of Science and Technology, Shenzhen, 518055 China
| | - Wen Liu
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, and School of Medicine, Southern University of Science and Technology, Shenzhen, 518055 China
| | - Huiling Cao
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, and School of Medicine, Southern University of Science and Technology, Shenzhen, 518055 China
| | - Guozhi Xiao
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, and School of Medicine, Southern University of Science and Technology, Shenzhen, 518055 China
| |
Collapse
|