1
|
Lu Z, Xiao P, Liu S, Huang C, Li W, Mao Y, Xu Y, Tian Y. Osteoimmunology: Crosstalk Between T Cells and Osteoclasts in Osteoporosis. Clin Rev Allergy Immunol 2025; 68:41. [PMID: 40208457 DOI: 10.1007/s12016-025-09046-1] [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] [Accepted: 03/22/2025] [Indexed: 04/11/2025]
Abstract
Osteoporosis, a common metabolic condition that affects the bones, increases the risk of fractures, thereby diminishing one's quality of life and, in severe cases, can even result in life-threatening conditions. Osteoporosis is becoming increasingly prevalent worldwide as the population ages. Previous research on osteoporosis has focused on skeletal cellular components such as osteoblasts and osteoclasts. The emerging field of "osteoimmunology" has recently been introduced through new research. The concept highlights the critical impact of bone-immune system interactions on osteoporosis progression. The pathogenesis of osteoporosis is significantly influenced by T cells, particularly cytotoxic and helper T cells, which modulate osteoclast differentiation and activity. A crucial aspect of understanding osteoporosis is how T lymphocytes interact with osteoclasts. However, the precise mechanisms underlying T cell-osteoclast crosstalk remain poorly understood. This review systematically examines T cell and osteoclast involvement in osteoimmunology, with a particular focus on their involvement in osteoporosis. It seeks to elucidate the immune mechanisms driving the progression of osteoporosis and identify key molecules involved in T cell-osteoclast interactions. This aims to discover novel molecular targets and intervention strategies to improve early diagnosis and management of osteoporosis. Furthermore, this article will explore the potential of intervening in T cell-osteoclast interactions using conventional therapies, traditional Chinese medicine, immunomodulatory agents, and nanomaterial-based treatments, providing new perspectives for future osteoporosis management.
Collapse
Affiliation(s)
- Zeyao Lu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Peilun Xiao
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shijia Liu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chongjun Huang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Weishang Li
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuanheng Mao
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ying Xu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Ye Tian
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
2
|
Hu K, Song M, Song T, Jia X, Song Y. Osteoimmunology in Osteoarthritis: Unraveling the Interplay of Immunity, Inflammation, and Joint Degeneration. J Inflamm Res 2025; 18:4121-4142. [PMID: 40125089 PMCID: PMC11930281 DOI: 10.2147/jir.s514002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/13/2025] [Indexed: 03/25/2025] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease influenced by multiple factors, with its etiology arising from intricate interactions among mechanical stress, inflammatory processes, and disruptions in bone metabolism. Recent research in bone immunology indicates that immune-mediated mechanisms significantly contribute to the progression of OA, highlighting the interactions among immune cells, cytokine networks, and bone components. Immune cells interact with osteoclasts, osteoblasts, and chondrocytes in a variety of ways. These interactions foster a pro-inflammatory microenvironment, contributing to cartilage breakdown, synovial inflammation, and the sclerosis of subchondral bone. In this article, we present a comprehensive review of bone immunology in OA, focusing on the critical role of immune cells and their cytokine-mediated feedback loops in the pathophysiology of OA. In addition, we are exploring novel therapeutic strategies targeting bone immune pathways, including macrophage polarization, T-cell differentiation, and stem cell therapy to restore the metabolic balance between immunity and bone. By integrating cutting-edge research in bone immunology, this review integrates the latest advancements in bone immunology to construct a comprehensive framework for unraveling the pathogenesis of OA, laying a theoretical foundation for the development of innovative precision therapies.
Collapse
Affiliation(s)
- Kangyi Hu
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Min Song
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Ting Song
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Xiao Jia
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Yongjia Song
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| |
Collapse
|
3
|
Chen Q, Wang X, Zhang P, Li B. Recent trends in human milk oligosaccharides: New synthesis technology, regulatory effects, and mechanisms of non-intestinal functions. Compr Rev Food Sci Food Saf 2025; 24:e70147. [PMID: 40091651 DOI: 10.1111/1541-4337.70147] [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: 10/18/2024] [Revised: 01/30/2025] [Accepted: 02/07/2025] [Indexed: 03/19/2025]
Abstract
Recently, the non-intestinal functions of human milk oligosaccharides (HMOs) have been widely documented, including their roles in promoting brain development and growth, as well as ameliorating anxiety, allergies, and obesity. Understanding their mechanisms of action is becoming increasingly critical. Furthermore, these effects are frequently associated with the type and structure of HMOs. As an innovative technology, "plant factory" is expected to complement traditional synthesis technology. This study reviews the novel "plant factory" synthesis techniques. Particular emphasis is placed on the processes, advantages, and limitations of "plant factory" synthesis of HMOs. This technology can express genes related to HMO synthesis instantaneously in plant leaves, thereby enabling the rapid and cost-effective generation of HMOs. However, "plant factory" technology remains underdeveloped, and challenges related to low yield and unsustainable production must be addressed. Furthermore, we present an overview of the most recent clinical and preclinical studies on the non-intestinal functions of HMOs. This review emphasizes the mechanisms of action underlying the non-intestinal functions of HMOs. HMOs primarily exert non-intestinal functions through the cleavage of beneficial monomer components, metabolism to produce advantageous metabolites, and regulation of immune responses.
Collapse
Affiliation(s)
- Qingxue Chen
- Food College, Northeast Agricultural University, Harbin, China
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Xiangxin Wang
- Food College, Northeast Agricultural University, Harbin, China
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Peng Zhang
- Food College, Northeast Agricultural University, Harbin, China
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Bailiang Li
- Food College, Northeast Agricultural University, Harbin, China
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| |
Collapse
|
4
|
Ye X, Li X, Zhu C, Cui L, Shen Z, Xu K, Shen G, Wu L, Zhang B. Associations between systemic inflammation response index and femur bone mineral density in adults: The NHANES 2005-2010, 2013-2014, and 2017-2018. Medicine (Baltimore) 2025; 104:e41565. [PMID: 39993115 PMCID: PMC11856988 DOI: 10.1097/md.0000000000041565] [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: 03/02/2024] [Revised: 12/13/2024] [Accepted: 01/30/2025] [Indexed: 02/26/2025] Open
Abstract
A unique measure of inflammatory evaluation, the systemic inflammation response index (SIRI) may offer useful data for the diagnosis and risk assessment of a number of diseases. The aim of this study was to investigate the relationship between SIRI and femur bone mineral density (BMD) in US adults. The association between SIRI and femur BMD was examined using multivariate logistic regression, sensitivity analysis, and smoothing curve fitting using data from the National Health and Nutrition Examination Survey (NHANES) 2005-2010, 2013-2014, and 2017-2018. Subgroup analysis and interaction tests were employed to examine the population-level stability of this connection. This present study included 18,022 participants older than 20 years from NHANES (2005-2010, 2013-2014, and 2017-2018). The present study showed a negative association between SIRI and femur BMD (including total femur BMD, femoral neck BMD, trochanter BMD, and intertrochanter BMD). In the fully adjusted model, we found a negative association between the SIRI and total femur BMD (Beta = -0.0032, 95% CI: -0.0053 to -0.0012), a negative association between the SIRI and femoral neck BMD (Beta = -0.0025, 95% CI: -0.0045 to -0.0005), a negative association between the SIRI and trochanter BMD (Beta = -0.0032, 95% CI: -0.0050 to -0.0013), a negative association between the SIRI and intertrochanter BMD (Beta = -0.0031, 95% CI: -0.0056 to -0.0007). This negative association was more pronounced in older adults > 65 years of age. In addition, we found a U-shaped association between SIRI and femur BMD by further smoothing curve-fitting methods. SIRI was negatively associated with femur BMD in US adults, and this association was more significant in older adults over 65 years. SIRI may be a useful, convenient, and practical indicator of inflammation. Moreover, older adults with high SIRI levels are likely to have low femur BMD.
Collapse
Affiliation(s)
- Xiaoang Ye
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Xinru Li
- Hangzhou Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Chaojin Zhu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Longkang Cui
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Zhe Shen
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Kuangying Xu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Gaobo Shen
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Lianguo Wu
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Bingbing Zhang
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| |
Collapse
|
5
|
Madel MB, Ibáñez L, Ciucci T, Halper J, Boutin A, Beldi G, Lavanant AC, Garchon HJ, Rouleau M, Mueller CG, Peyrin-Biroulet L, Moulin D, Blin-Wakkach C, Wakkach A. Dysregulated myeloid differentiation in colitis is induced by inflammatory osteoclasts in a TNFα-dependent manner. Mucosal Immunol 2025; 18:90-104. [PMID: 39332768 DOI: 10.1016/j.mucimm.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 09/14/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
Inflammatory bowel disease (IBD) is characterized by very severe intestinal inflammation associated with extra-intestinal manifestations. One of the most critical ones is bone destruction, which remains a major cause of morbidity and a risk factor for osteopenia and osteoporosis in IBD patients. In various mouse models of IBD, we and other have demonstrated concomitant bone loss due to a significant increase in osteoclast activity. Besides bone resorption, osteoclasts are known to control hematopoietic niches in vivo and modulate inflammatory responses in vitro, suggesting they may participate in chronic inflammation in vivo. Here, using different models of colitis, we showed that osteoclast inhibition significantly reduced disease severity and that induction of osteoclast differentiation by RANKL contributed to disease worsening. Our results demonstrate a direct link between osteoclast activity and myeloid cell accumulation in the intestine during colitis. RNAseq analysis of osteoclasts from colitic mice revealed overexpression of genes involved in the remodeling of hematopoietic stem cell niches. We also demonstrated that osteoclasts induced hematopoietic progenitor proliferation accompanied by a myeloid skewing in the early phases of colitis, which was confirmed in a model of RANKL-induced osteoclastogenesis. Mechanistically, inhibition of TNF-α reduced the induction of myeloid skewing by OCL both in vitro and in vivo. Lastly, we observed that osteoclastic activity and the proportion of myeloid cells in the blood are positively correlated in patients with Crohn's disease. Collectively, our results shed light on a new role of osteoclasts in colitis in vivo, demonstrating they exert their colitogenic activity through an early action on hematopoiesis, leading to an increase in myelopoiesis sustaining gut inflammation.
Collapse
Affiliation(s)
| | - Lidia Ibáñez
- Université Côte d'Azur, CNRS, LP2M, Nice, France
| | - Thomas Ciucci
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Julia Halper
- Université Côte d'Azur, CNRS, LP2M, Nice, France
| | | | - Ghada Beldi
- Université Côte d'Azur, CNRS, LP2M, Nice, France
| | - Alice C Lavanant
- CNRS UPR 3572, IBMC, University of Strasbourg, 67000 Strasbourg, France
| | - Henri-Jean Garchon
- Université Paris-Saclay, UVSQ, INSERM, Infection et inflammation, 78180 Montigny-Le-Bretonneux, France
| | | | | | - Laurent Peyrin-Biroulet
- Department of Gastroenterology, IHU INFINY, CHRU Nancy, F-54500 Vandœuvre-lès-Nancy, France; Université de Lorraine, INSERM, NGERE, F-54500 Vandoeuvre les Nancy, France
| | - David Moulin
- Université de Lorraine, CNRS, IMoPA, F-54500 Vandœuvre Les Nancy, France; IHU INFINY, Contrat d'interface, CHRU Nancy, France
| | | | | |
Collapse
|
6
|
Indrio F, Salatto A. Gut Microbiota-Bone Axis. ANNALS OF NUTRITION & METABOLISM 2025:1-10. [PMID: 39848230 DOI: 10.1159/000541999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/11/2024] [Indexed: 01/25/2025]
Abstract
BACKGROUND Knowledge of the complex interplay between gut microbiota and human health is gradually increasing as it has just recently been a field of such great interest. SUMMARY Recent studies have reported that communities of microorganisms inhabiting the gut influence the immune system through cellular responses and shape many physiological and pathophysiological aspects of the body, including muscle and bone metabolism (formation and resorption). Specifically, the gut microbiota affects skeletal homeostasis through changes in host metabolism, the immune system, hormone secretion, and the gut-brain axis. The major role on gut-bone axis is due to short-chain fatty acids (SCFAs). They have the ability to influence regulatory T-cell (Tregs) development and activate bone metabolism through the action of Wnt10. SCFA production may be a mechanism by which the microbial community, by increasing the serum level of insulin-like growth factor 1 (IGF-1), leads to the growth and regulation of bone homeostasis. A specific SCFA, butyrate, diffuses into the bone marrow where it expands Tregs. The Tregs induce production of the Wnt ligand Wnt10b by CD8+ T cells, leading to activation of Wnt signaling and stimulation of bone formation. At the hormonal level, the effect of the gut microbiota on bone homeostasis is expressed through the biphasic action of serotonin. Some microbiota, such as spore-forming microbes, regulate the level of serotonin in the gut, serum, and feces. Another group of bacterial species (Lactococcus, Mucispirillum, Lactobacillus, and Bifidobacterium) can increase the level of peripheral/vascular leptin, which in turn manages bone homeostasis through the action of brain serotonin.
Collapse
Affiliation(s)
- Flavia Indrio
- Department of Experimental Medicine, University of Salento, Lecce, Italy
| | - Alessia Salatto
- Department of Translational Medical Science, University of Naples Federico II, Napoli, Italy
| |
Collapse
|
7
|
Lyu Z, Yuan G, Zhang Y, Zhang F, Liu Y, Li Y, Li G, Wang Y, Zhang M, Hu Y, Guo Y, Liu D. Anaerostipes caccae CML199 enhances bone development and counteracts aging-induced bone loss through the butyrate-driven gut-bone axis: the chicken model. MICROBIOME 2024; 12:215. [PMID: 39438898 PMCID: PMC11495078 DOI: 10.1186/s40168-024-01920-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/28/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND The gut microbiota is a key regulator of bone metabolism. Investigating the relationship between the gut microbiota and bone remodeling has revealed new avenues for the treatment of bone-related disorders. Despite significant progress in understanding gut microbiota-bone interactions in mammals, research on avian species remains limited. Birds have unique bone anatomy and physiology to support egg-laying. However, whether and how the gut microbiota affects bone physiology in birds is still unknown. In this study, we utilized laying hens as a research model to analyze bone development patterns, elucidate the relationships between bone and the gut microbiota, and mine probiotics with osteomodulatory effects. RESULTS Aging led to a continuous increase in bone mineral density in the femur of laying hens. The continuous deposition of medullary bone in the bone marrow cavity of aged laying hens led to significant trabecular bone loss and weakened bone metabolism. The cecal microbial composition significantly shifted before and after sexual maturity, with some genera within the class Clostridia potentially linked to postnatal bone development in laying hens. Four bacterial strains associated with bone development, namely Blautia coccoides CML164, Fournierella sp002159185 CML151, Anaerostipes caccae CML199 (ANA), and Romboutsia lituseburensis CML137, were identified and assessed in chicks with low bacterial loads and chicken primary osteoblasts. Among these, ANA demonstrated the most significant promotion of bone formation both in vivo and in vitro, primarily attributed to butyrate in its fermentation products. A long-term feeding experiment of up to 72 weeks confirmed that ANA enhanced bone development during sexual maturity by improving the immune microenvironment of the bone marrow in laying hens. Dietary supplementation of ANA for 50 weeks prevented excessive medullary bone deposition and mitigated aging-induced trabecular bone loss. CONCLUSIONS These findings highlight the beneficial effects of ANA on bone physiology, offering new perspectives for microbial-based interventions for bone-related disorders in both poultry and possibly extending to human health. Video Abstract.
Collapse
Affiliation(s)
- Zhengtian Lyu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Gaoxiang Yuan
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yuying Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Fengwenhui Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yan Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Yifan Li
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Guang Li
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Ying Wang
- Sichuan Tieqilishi Industrial Co., Ltd, Mianyang, 621010, China
| | - Ming Zhang
- Sichuan Tieqilishi Industrial Co., Ltd, Mianyang, 621010, China
| | - Yongfei Hu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| | - Dan Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
8
|
Chen Z, Xu W, Luo J, Liu L, Peng X. Lonicera japonica Fermented by Lactobacillus plantarum Improve Multiple Patterns Driven Osteoporosis. Foods 2024; 13:2649. [PMID: 39272415 PMCID: PMC11393950 DOI: 10.3390/foods13172649] [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: 07/09/2024] [Revised: 08/08/2024] [Accepted: 08/19/2024] [Indexed: 09/15/2024] Open
Abstract
Osteoporosis (OP) represents a global health challenge. Certain functional food has the potential to mitigate OP. Honeysuckle (Lonicera japonica) solution has medicinal effects, such as anti-inflammatory and immune enhancement, and can be used in functional foods such as health drinks and functional snacks. The composition of honeysuckle changed significantly after fermentation, and 376 metabolites were enriched. In this study, we used dexamethasone to induce OP in the rat model. Research has confirmed the ability of FS (fermented Lonicera japonica solution) to enhance bone mineral density (BMD), repair bone microarchitectural damage, and increase blood calcium levels. Markers such as tartrate-resistant acid phosphatase-5b (TRACP-5b) and pro-inflammatory cytokines (TNF-α and IL-6) were notably decreased, whereas osteocalcin (OCN) levels increased after FS treatment. FS intervention in OP rats restored the abundance of 6 bacterial genera and the contents of 17 serum metabolites. The results of the Spearman correlation analysis showed that FS may alleviate OP by restoring the abundance of 6 bacterial genera and the contents of 17 serum metabolites, reducing osteoclast differentiation, promoting osteoblast differentiation, and reducing the inflammatory response. This study revealed that Lactobacillus plantarum-fermented honeysuckle alleviated OP through intestinal bacteria and serum metabolites and provided a theoretical basis for the development of related functional foods.
Collapse
Affiliation(s)
- Zimin Chen
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Weiye Xu
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Jianming Luo
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Liu Liu
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Xichun Peng
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China
| |
Collapse
|
9
|
Umur E, Bulut SB, Yiğit P, Bayrak E, Arkan Y, Arslan F, Baysoy E, Kaleli-Can G, Ayan B. Exploring the Role of Hormones and Cytokines in Osteoporosis Development. Biomedicines 2024; 12:1830. [PMID: 39200293 PMCID: PMC11351445 DOI: 10.3390/biomedicines12081830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/02/2024] Open
Abstract
The disease of osteoporosis is characterized by impaired bone structure and an increased risk of fractures. There is a significant impact of cytokines and hormones on bone homeostasis and the diagnosis of osteoporosis. As defined by the World Health Organization (WHO), osteoporosis is defined as having a bone mineral density (BMD) that is 2.5 standard deviations (SD) or more below the average for young and healthy women (T score < -2.5 SD). Cytokines and hormones, particularly in the remodeling of bone between osteoclasts and osteoblasts, control the differentiation and activation of bone cells through cytokine networks and signaling pathways like the nuclear factor kappa-B ligand (RANKL)/the receptor of RANKL (RANK)/osteoprotegerin (OPG) axis, while estrogen, parathyroid hormones, testosterone, and calcitonin influence bone density and play significant roles in the treatment of osteoporosis. This review aims to examine the roles of cytokines and hormones in the pathophysiology of osteoporosis, evaluating current diagnostic methods, and highlighting new technologies that could help for early detection and treatment of osteoporosis.
Collapse
Affiliation(s)
- Egemen Umur
- Department of Biomedical Engineering, İzmir Democracy University, İzmir 35140, Türkiye
| | - Safiye Betül Bulut
- Department of Biomedical Engineering, İzmir Democracy University, İzmir 35140, Türkiye
| | - Pelin Yiğit
- Department of Biomedical Engineering, İzmir Democracy University, İzmir 35140, Türkiye
| | - Emirhan Bayrak
- Department of Biomedical Engineering, İzmir Democracy University, İzmir 35140, Türkiye
| | - Yaren Arkan
- Department of Biomedical Engineering, İzmir Democracy University, İzmir 35140, Türkiye
| | - Fahriye Arslan
- Department of Biomedical Engineering, İzmir Democracy University, İzmir 35140, Türkiye
| | - Engin Baysoy
- Department of Biomedical Engineering, Bahçeşehir University, İstanbul 34353, Türkiye
| | - Gizem Kaleli-Can
- Department of Biomedical Engineering, İzmir Democracy University, İzmir 35140, Türkiye
| | - Bugra Ayan
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
10
|
Kim YT, Mills DA. Exploring the gut microbiome: probiotics, prebiotics, synbiotics, and postbiotics as key players in human health and disease improvement. Food Sci Biotechnol 2024; 33:2065-2080. [PMID: 39130661 PMCID: PMC11315840 DOI: 10.1007/s10068-024-01620-1] [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: 04/10/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 08/13/2024] Open
Abstract
The human gut microbiome accompanies us from birth, and it is developed and matured by diet, lifestyle, and environmental factors. During aging, the bacterial composition evolves in reciprocal communication with the host's physiological properties. Many diseases are closely related to the gut microbiome, which means the modulation of the gut microbiome can promote the disease targeting remote organs. This review explores the intricate interaction between the gut microbiome and other organs, and their improvement from disease by prebiotics, probiotics, synbiotics, and postbiotics. Each section of the review is supported by clinical trials that substantiate the benefits of modulation the gut microbiome through dietary intervention for improving primary health outcomes across various axes with the gut. In conclusion, the review underscores the significant potential of targeting the gut microbiome for therapeutic and preventative interventions in a wide range of diseases, calling for further research to fully unlock the microbiome's capabilities in enhancing human health.
Collapse
Affiliation(s)
- You-Tae Kim
- Department of Food Science and Technology, University of California-Davis, Davis, CA USA
| | - David A. Mills
- Department of Food Science and Technology, University of California-Davis, Davis, CA USA
| |
Collapse
|
11
|
Hong J, Luo F, Du X, Xian F, Li X. The immune cells in modulating osteoclast formation and bone metabolism. Int Immunopharmacol 2024; 133:112151. [PMID: 38685175 DOI: 10.1016/j.intimp.2024.112151] [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: 01/07/2024] [Revised: 04/10/2024] [Accepted: 04/22/2024] [Indexed: 05/02/2024]
Abstract
Osteoclasts are pivotal in regulating bone metabolism, with immune cells significantly influencing both physiological and pathological processes by modulating osteoclast functions. This is particularly evident in conditions of inflammatory bone resorption, such as rheumatoid arthritis and periodontitis. This review summarizes and comprehensively analyzes the research progress on the regulation of osteoclast formation by immune cells, aiming to unveil the underlying mechanisms and pathways through which diseases, such as rheumatoid arthritis and periodontitis, impact bone metabolism.
Collapse
Affiliation(s)
- Jiale Hong
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Fang Luo
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Xingyue Du
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Fa Xian
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Xinyi Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China.
| |
Collapse
|
12
|
Chen L, Zhao Y, Qiu J, Lin X. Analysis and validation of biomarkers of immune cell-related genes in postmenopausal osteoporosis: An observational study. Medicine (Baltimore) 2024; 103:e38042. [PMID: 38728482 PMCID: PMC11081595 DOI: 10.1097/md.0000000000038042] [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: 12/15/2023] [Accepted: 04/05/2024] [Indexed: 05/12/2024] Open
Abstract
Postmenopausal osteoporosis (PMOP) is a common metabolic inflammatory disease. In conditions of estrogen deficiency, chronic activation of the immune system leads to a hypo-inflammatory phenotype and alterations in its cytokine and immune cell profile, although immune cells play an important role in the pathology of osteoporosis, studies on this have been rare. Therefore, it is important to investigate the role of immune cell-related genes in PMOP. PMOP-related datasets were downloaded from the Gene Expression Omnibus database. Immune cells scores between high bone mineral density (BMD) and low BMD samples were assessed based on the single sample gene set enrichment analysis method. Subsequently, weighted gene co-expression network analysis was performed to identify modules highly associated with immune cells and obtain module genes. Differential analysis between high BMD and low BMD was also performed to obtain differentially expressed genes. Module genes are intersected with differentially expressed genes to obtain candidate genes, and functional enrichment analysis was performed. Machine learning methods were used to filter out the signature genes. The receiver operating characteristic (ROC) curves of the signature genes and the nomogram were plotted to determine whether the signature genes can be used as a molecular marker. Gene set enrichment analysis was also performed to explore the potential mechanism of the signature genes. Finally, RNA expression of signature genes was validated in blood samples from PMOP patients and normal control by real-time quantitative polymerase chain reaction. Our study of PMOP patients identified differences in immune cells (activated dendritic cell, CD56 bright natural killer cell, Central memory CD4 T cell, Effector memory CD4 T cell, Mast cell, Natural killer T cell, T follicular helper cell, Type 1 T-helper cell, and Type 17 T-helper cell) between high and low BMD patients. We obtained a total of 73 candidate genes based on modular genes and differential genes, and obtained 5 signature genes by least absolute shrinkage and selection operator and random forest model screening. ROC, principal component analysis, and t-distributed stochastic neighbor embedding down scaling analysis revealed that the 5 signature genes had good discriminatory ability between high and low BMD samples. A logistic regression model was constructed based on 5 signature genes, and both ROC and column line plots indicated that the model accuracy and applicability were good. Five signature genes were found to be associated with proteasome, mitochondria, and lysosome by gene set enrichment analysis. The real-time quantitative polymerase chain reaction results showed that the expression of the signature genes was significantly different between the 2 groups. HIST1H2AG, PYGM, NCKAP1, POMP, and LYPLA1 might play key roles in PMOP and be served as the biomarkers of PMOP.
Collapse
Affiliation(s)
- Lihua Chen
- Rehabilitation Department, Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, PR China
- Osteoporosis Department, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, PR China
- Postgraduate college, Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Yu Zhao
- Osteoporosis Department, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, PR China
- Postgraduate college, Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Jingjing Qiu
- Rehabilitation Department, Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, PR China
- Postgraduate college, Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Xiaosheng Lin
- Osteoporosis Department, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, PR China
| |
Collapse
|
13
|
Ren L, Liu G, Bai Y, Gu L, Wang Y, Sun L. NLRC3 attenuates osteoclastogenesis by limiting TNFα + Th17 cell response in osteoporosis. J Mol Med (Berl) 2024; 102:655-665. [PMID: 38436712 PMCID: PMC11055730 DOI: 10.1007/s00109-024-02422-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: 08/27/2022] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 03/05/2024]
Abstract
NOD-like receptor family CARD domain containing 3 (NLRC3) is the intracellular protein belonging to NLR (NOD-like receptor) family. NLRC3 can negatively regulate inflammatory signal transduction pathways within the adaptive and innate immunocytes. However, studies need to elucidate the biological role of NLRC3 in bone remodeling. Herein, our study proved that NLRC3 prevents bone loss by inhibiting TNFα+ Th17 cell responses. In osteoporosis, NLRC3 attenuated TNFα+ Th17 cell accumulation in the bone marrow. However, osteoporosis (OP) development was aggravated without affecting bone marrow macrophage (BMM) osteoclastogenesis in NLRC3-deficient ovariectomized (OVX) mice. In this study, we transferred the wild-type and NLRC3-/- CD4+ cells into Rag1-/- mice. Consequently, we evidenced the effects of NLRC3 in CD4+ T cells on inhibiting the accumulation of TNFα + Th17 cells, thus restricting bone loss in the OVX mice. Simultaneously, NLRC3-/- CD4+ T cells promoted the recruitment of osteoclast precursors and inflammatory monocytes into the OVX mouse bone marrow. Mechanism-wise, NLRC3 reduced the secretion of TNFα + Th17 cells of RANKL, MIP1α, and MCP1, depending on the T cells. In addition, NLRC3 negatively regulated the Th17 osteoclastogenesis promoting functions via limiting the NF-κB activation. Collectively, this study appreciated the effect of NLRC3 on modulating bone mass via adaptive immunity depending on CD4+ cells. According to findings of this study, NLRC3 may be the candidate anti-OP therapeutic target. KEY MESSAGES: NLRC3 negatively regulated the Th17 osteoclastogenesis promoting functions via limiting the NF-κB activation. NLRC3 may be the candidate anti-OP therapeutic target.
Collapse
Affiliation(s)
- Lingyan Ren
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province, 550003, China
- Antenatal Diagnosis Centre, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province, 550003, China
| | - Guangjun Liu
- Institute of Traumatic Orthopedics, The 80th, Army Hospital of the Chinese People's Liberation Army, Weifang Shandong Province, 500000, China
| | - Yun Bai
- Institute of Traumatic Orthopedics, The 80th, Army Hospital of the Chinese People's Liberation Army, Weifang Shandong Province, 500000, China
| | - Liling Gu
- Department of Rehabilitation, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province, 550003, China
| | - Yuan Wang
- Department of Orthopedics, TongRen Hospital, School of Medicine Shanghai, Jiao Tong University, Shanghai, 200336, China.
| | - Li Sun
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province, 550003, China.
| |
Collapse
|
14
|
Orsini F, Crotti C, Cincinelli G, Di Taranto R, Amati A, Ferrito M, Varenna M, Caporali R. Bone Involvement in Rheumatoid Arthritis and Spondyloartritis: An Updated Review. BIOLOGY 2023; 12:1320. [PMID: 37887030 PMCID: PMC10604370 DOI: 10.3390/biology12101320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/02/2023] [Accepted: 10/06/2023] [Indexed: 10/28/2023]
Abstract
Several rheumatologic diseases are primarily distinguished by their involvement of bone tissue, which not only serves as a mere target of the condition but often plays a pivotal role in its pathogenesis. This scenario is particularly prominent in chronic inflammatory arthritis such as rheumatoid arthritis (RA) and spondyloarthritis (SpA). Given the immunological and systemic nature of these diseases, in this review, we report an overview of the pathogenic mechanisms underlying specific bone involvement, focusing on the complex interactions that occur between bone tissue's own cells and the molecular and cellular actors of the immune system, a recent and fascinating field of interest defined as osteoimmunology. Specifically, we comprehensively elaborate on the distinct pathogenic mechanisms of bone erosion seen in both rheumatoid arthritis and spondyloarthritis, as well as the characteristic process of aberrant bone formation observed in spondyloarthritis. Lastly, chronic inflammatory arthritis leads to systemic bone involvement, resulting in systemic bone loss and consequent osteoporosis, along with increased skeletal fragility.
Collapse
Affiliation(s)
- Francesco Orsini
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy (A.A.)
- Department of Rheumatology and Medical Sciences, ASST G.Pini-CTO, 20122 Milan, Italy
| | - Chiara Crotti
- Bone Diseases Unit, Department of Rheumatology and Medical Sciences, ASST G.Pini-CTO, 20122 Milan, Italy
| | - Gilberto Cincinelli
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy (A.A.)
- Department of Rheumatology and Medical Sciences, ASST G.Pini-CTO, 20122 Milan, Italy
| | - Raffaele Di Taranto
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy (A.A.)
- Department of Rheumatology and Medical Sciences, ASST G.Pini-CTO, 20122 Milan, Italy
| | - Andrea Amati
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy (A.A.)
- Department of Rheumatology and Medical Sciences, ASST G.Pini-CTO, 20122 Milan, Italy
| | - Matteo Ferrito
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy (A.A.)
- Department of Rheumatology and Medical Sciences, ASST G.Pini-CTO, 20122 Milan, Italy
| | - Massimo Varenna
- Bone Diseases Unit, Department of Rheumatology and Medical Sciences, ASST G.Pini-CTO, 20122 Milan, Italy
| | - Roberto Caporali
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy (A.A.)
- Department of Rheumatology and Medical Sciences, ASST G.Pini-CTO, 20122 Milan, Italy
| |
Collapse
|
15
|
Yu E, Zhang M, Xu G, Liu X, Yan J. Consensus cluster analysis of apoptosis-related genes in patients with osteoarthritis and their correlation with immune cell infiltration. Front Immunol 2023; 14:1202758. [PMID: 37860011 PMCID: PMC10582959 DOI: 10.3389/fimmu.2023.1202758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 09/15/2023] [Indexed: 10/21/2023] Open
Abstract
Background Osteoarthritis (OA) progression involves multiple factors, including cartilage erosion as the basic pathological mechanism of degeneration, and is closely related to chondrocyte apoptosis. To analyze the correlation between apoptosis and OA development, we selected apoptosis genes from the differentially expressed genes (DEGs) between OA and normal samples from the Gene Expression Omnibus (GEO) database, used lasso regression analysis to identify characteristic genes, and performed consensus cluster analysis to further explore the pathogenesis of this disease. Methods The Gene expression profile datasets of OA samples, GSE12021 and GSE55235, were downloaded from GEO. The datasets were combined and analyzed for DEGs. Apoptosis-related genes (ARGs) were collected from the GeneCards database and intersected with DEGs for apoptosis-related DEGs (ARDEGs). Least absolute shrinkage and selection operator (LASSO) regression analysis was performed to obtain characteristic genes, and a nomogram was constructed based on these genes. A consensus cluster analysis was performed to divide the patients into clusters. The immune characteristics, functional enrichment, and immune infiltration statuses of the clusters were compared. In addition, a protein-protein interaction network of mRNA drugs, mRNA-transcription factors (TFs), and mRNA-miRNAs was constructed. Results A total of 95 DEGs were identified, of which 47 were upregulated and 48 were downregulated, and 31 hub genes were selected as ARDEGs. LASSO regression analysis revealed nine characteristic genes: growth differentiation factor 15 (GDF15), NAMPT, TLR7, CXCL2, KLF2, REV3L, KLF9, THBD, and MTHFD2. Clusters A and B were identified, and neutrophil activation and neutrophil activation involved in the immune response were highly enriched in Cluster B, whereas protein repair and purine salvage signal pathways were enriched in Cluster A. The number of activated natural killer cells in Cluster B was significantly higher than that in Cluster A. GDF15 and KLF9 interacted with 193 and 32 TFs, respectively, and CXCL2 and REV3L interacted with 48 and 82 miRNAs, respectively. Conclusion ARGs could predict the occurrence of OA and may be related to different degrees of OA progression.
Collapse
Affiliation(s)
| | | | | | | | - Jinglong Yan
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
16
|
Evenepoel P, Stenvinkel P, Shanahan C, Pacifici R. Inflammation and gut dysbiosis as drivers of CKD-MBD. Nat Rev Nephrol 2023; 19:646-657. [PMID: 37488276 DOI: 10.1038/s41581-023-00736-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2023] [Indexed: 07/26/2023]
Abstract
Two decades ago, Kidney Disease: Improving Global Outcomes coined the term chronic kidney disease-mineral and bone disorder (CKD-MBD) to describe the syndrome of biochemical, bone and extra-skeletal calcification abnormalities that occur in patients with CKD. CKD-MBD is a prevalent complication and contributes to the excessively high burden of fractures and cardiovascular disease, loss of quality of life and premature mortality in patients with CKD. Thus far, therapy has focused primarily on phosphate retention, abnormal vitamin D metabolism and parathyroid hormone disturbances, but these strategies have largely proved unsuccessful, thus calling for paradigm-shifting concepts and innovative therapeutic approaches. Interorgan crosstalk is increasingly acknowledged to have an important role in health and disease. Accordingly, mounting evidence suggests a role for both the immune system and the gut microbiome in bone and vascular biology. Gut dysbiosis, compromised gut epithelial barrier and immune cell dysfunction are prominent features of the uraemic milieu. These alterations might contribute to the inflammatory state observed in CKD and could have a central role in the pathogenesis of CKD-MBD. The emerging fields of osteoimmunology and osteomicrobiology add another level of complexity to the pathogenesis of CKD-MBD, but also create novel therapeutic opportunities.
Collapse
Affiliation(s)
- Pieter Evenepoel
- Laboratory of Nephrology, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Herestraat, Leuven, Belgium.
| | - Peter Stenvinkel
- Department of Renal Medicine M99, Karolinska University Hospital, Stockholm, Sweden
| | - Catherine Shanahan
- British Heart Foundation Centre of Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK
| | - Roberto Pacifici
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory Microbiome Research Center, and Immunology and Molecular Pathogenesis Program, Emory University, Atlanta, GA, USA
| |
Collapse
|
17
|
Yin P, Jiang Y, Fang X, Wang D, Li Y, Chen M, Deng H, Tang P, Zhang L. Cell-Based Therapies for Degenerative Musculoskeletal Diseases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207050. [PMID: 37199688 PMCID: PMC10375105 DOI: 10.1002/advs.202207050] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 04/29/2023] [Indexed: 05/19/2023]
Abstract
Degenerative musculoskeletal diseases (DMDs), including osteoporosis, osteoarthritis, degenerative disc disease, and sarcopenia, present major challenges in the aging population. Patients with DMDs present with pain, functional decline, and reduced exercise tolerance, which result in long-term or permanent deficits in their ability to perform daily activities. Current strategies for dealing with this cluster of diseases focus on relieving pain, but they have a limited capacity to repair function or regenerate tissue. Cell-based therapies have attracted considerable attention in recent years owing to their unique mechanisms of action and remarkable effects on regeneration. In this review, current experimental attempts to use cell-based therapies for DMDs are highlighted, and the modes of action of different cell types and their derivatives, such as exosomes, are generalized. In addition, the latest findings from state-of-the-art clinical trials are reviewed, approaches to improve the efficiency of cell-based therapies are summarized, and unresolved questions and potential future research directions for the translation of cell-based therapies are identified.
Collapse
Affiliation(s)
- Pengbin Yin
- Department of Orthopedicsthe Fourth Medical CenterChinese PLA General HospitalBeijing100853China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100853China
| | - Yuheng Jiang
- Department of Orthopedicsthe Fourth Medical CenterChinese PLA General HospitalBeijing100853China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100853China
- Department of OrthopedicsGeneral Hospital of Southern Theater Command of PLANo. 111, Liuhua AvenueGuangzhou510010China
| | - Xuan Fang
- Department of Anatomy, Histology and EmbryologySchool of Basic Medical SciencesPeking University Health Science CenterBeijing100191China
| | - Daofeng Wang
- Department of Orthopedicsthe Fourth Medical CenterChinese PLA General HospitalBeijing100853China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100853China
| | - Yi Li
- Department of Orthopedicsthe Fourth Medical CenterChinese PLA General HospitalBeijing100853China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100853China
| | - Ming Chen
- Department of Orthopedicsthe Fourth Medical CenterChinese PLA General HospitalBeijing100853China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100853China
| | - Hao Deng
- Department of OrthopedicsThird Affiliated Hospital of Jinzhou Medical UniversityJinzhouLiaoning Province121000China
| | - Peifu Tang
- Department of Orthopedicsthe Fourth Medical CenterChinese PLA General HospitalBeijing100853China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100853China
| | - Licheng Zhang
- Department of Orthopedicsthe Fourth Medical CenterChinese PLA General HospitalBeijing100853China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100853China
| |
Collapse
|
18
|
Lyu Z, Hu Y, Guo Y, Liu D. Modulation of bone remodeling by the gut microbiota: a new therapy for osteoporosis. Bone Res 2023; 11:31. [PMID: 37296111 PMCID: PMC10256815 DOI: 10.1038/s41413-023-00264-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 04/01/2023] [Accepted: 04/18/2023] [Indexed: 06/12/2023] Open
Abstract
The gut microbiota (GM) plays a crucial role in maintaining the overall health and well-being of the host. Recent studies have demonstrated that the GM may significantly influence bone metabolism and degenerative skeletal diseases, such as osteoporosis (OP). Interventions targeting GM modification, including probiotics or antibiotics, have been found to affect bone remodeling. This review provides a comprehensive summary of recent research on the role of GM in regulating bone remodeling and seeks to elucidate the regulatory mechanism from various perspectives, such as the interaction with the immune system, interplay with estrogen or parathyroid hormone (PTH), the impact of GM metabolites, and the effect of extracellular vesicles (EVs). Moreover, this review explores the potential of probiotics as a therapeutic approach for OP. The insights presented may contribute to the development of innovative GM-targeted therapies for OP.
Collapse
Affiliation(s)
- Zhengtian Lyu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yongfei Hu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Dan Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
19
|
Hascoët E, Blanchard F, Blin-Wakkach C, Guicheux J, Lesclous P, Cloitre A. New insights into inflammatory osteoclast precursors as therapeutic targets for rheumatoid arthritis and periodontitis. Bone Res 2023; 11:26. [PMID: 37217496 DOI: 10.1038/s41413-023-00257-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 03/02/2023] [Accepted: 03/09/2023] [Indexed: 05/24/2023] Open
Abstract
Rheumatoid arthritis (RA) and periodontitis are chronic inflammatory diseases leading to increased bone resorption. Preventing this inflammatory bone resorption is a major health challenge. Both diseases share immunopathogenic similarities and a common inflammatory environment. The autoimmune response or periodontal infection stimulates certain immune actors, leading in both cases to chronic inflammation that perpetuates bone resorption. Moreover, RA and periodontitis have a strong epidemiological association that could be explained by periodontal microbial dysbiosis. This dysbiosis is believed to be involved in the initiation of RA via three mechanisms. (i) The dissemination of periodontal pathogens triggers systemic inflammation. (ii) Periodontal pathogens can induce the generation of citrullinated neoepitopes, leading to the generation of anti-citrullinated peptide autoantibodies. (iii) Intracellular danger-associated molecular patterns accelerate local and systemic inflammation. Therefore, periodontal dysbiosis could promote or sustain bone resorption in distant inflamed joints. Interestingly, in inflammatory conditions, the existence of osteoclasts distinct from "classical osteoclasts" has recently been reported. They have proinflammatory origins and functions. Several populations of osteoclast precursors have been described in RA, such as classical monocytes, a dendritic cell subtype, and arthritis-associated osteoclastogenic macrophages. The aim of this review is to synthesize knowledge on osteoclasts and their precursors in inflammatory conditions, especially in RA and periodontitis. Special attention will be given to recent data related to RA that could be of potential value in periodontitis due to the immunopathogenic similarities between the two diseases. Improving our understanding of these pathogenic mechanisms should lead to the identification of new therapeutic targets involved in the pathological inflammatory bone resorption associated with these diseases.
Collapse
Affiliation(s)
- Emilie Hascoët
- Nantes Université, Oniris, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000, Nantes, France
| | - Frédéric Blanchard
- Nantes Université, Oniris, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000, Nantes, France
| | | | - Jérôme Guicheux
- Nantes Université, Oniris, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000, Nantes, France.
| | - Philippe Lesclous
- Nantes Université, Oniris, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000, Nantes, France
| | - Alexandra Cloitre
- Nantes Université, Oniris, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000, Nantes, France
| |
Collapse
|
20
|
Hu N, Wang J, Ju B, Li Y, Fan P, Jin X, Kang X, Wu S. Recent advances of osteoimmunology research in rheumatoid arthritis: From single-cell omics approach. Chin Med J (Engl) 2023:00029330-990000000-00608. [PMID: 37166215 DOI: 10.1097/cm9.0000000000002678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Indexed: 05/12/2023] Open
Abstract
ABSTRACT Cellular immune responses as well as generalized and periarticular bone loss are the key pathogenic features of rheumatoid arthritis (RA). Under the pathological conditions of RA, dysregulated inflammation and immune processes tightly interact with skeletal system, resulting in pathological bone damage via inhibition of bone formation or induction of bone resorption. Single-cell omics technologies are revolutionary tools in the field of modern biological research.They enable the display of the state and function of cells in various environments from a single-cell resolution, thus making it conducive to identify the dysregulated molecular mechanisms of bone destruction in RA as well as the discovery of potential therapeutic targets and biomarkers. Here, we summarize the latest findings of single-cell omics technologies in osteoimmunology research in RA. These results suggest that single-cell omics have made significant contributions to transcriptomics and dynamics of specific cells involved in bone remodeling, providing a new direction for our understanding of cellular heterogeneity in the study of osteoimmunology in RA.
Collapse
Affiliation(s)
- Nan Hu
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jing Wang
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Bomiao Ju
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yuanyuan Li
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Ping Fan
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xinxin Jin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Xiaomin Kang
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Shufang Wu
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| |
Collapse
|
21
|
Anginot A, Nguyen J, Abou Nader Z, Rondeau V, Bonaud A, Kalogeraki M, Boutin A, Lemos JP, Bisio V, Koenen J, Hanna Doumit Sakr L, Picart A, Coudert A, Provot S, Dulphy N, Aurrand-Lions M, Mancini SJC, Lazennec G, McDermott DH, Guidez F, Blin-Wakkach C, Murphy PM, Cohen-Solal M, Espéli M, Rouleau M, Balabanian K. WHIM Syndrome-linked CXCR4 mutations drive osteoporosis. Nat Commun 2023; 14:2058. [PMID: 37045841 PMCID: PMC10097661 DOI: 10.1038/s41467-023-37791-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 03/07/2023] [Indexed: 04/14/2023] Open
Abstract
WHIM Syndrome is a rare immunodeficiency caused by gain-of-function CXCR4 mutations. Here we report a decrease in bone mineral density in 25% of WHIM patients and bone defects leading to osteoporosis in a WHIM mouse model. Imbalanced bone tissue is observed in mutant mice combining reduced osteoprogenitor cells and increased osteoclast numbers. Mechanistically, impaired CXCR4 desensitization disrupts cell cycle progression and osteogenic commitment of skeletal stromal/stem cells, while increasing their pro-osteoclastogenic capacities. Impaired osteogenic differentiation is evidenced in primary bone marrow stromal cells from WHIM patients. In mice, chronic treatment with the CXCR4 antagonist AMD3100 normalizes in vitro osteogenic fate of mutant skeletal stromal/stem cells and reverses in vivo the loss of skeletal cells, demonstrating that proper CXCR4 desensitization is required for the osteogenic specification of skeletal stromal/stem cells. Our study provides mechanistic insights into how CXCR4 signaling regulates the osteogenic fate of skeletal cells and the balance between bone formation and resorption.
Collapse
Affiliation(s)
- Adrienne Anginot
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Julie Nguyen
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- Inflammation, Microbiome and Immunosurveillance, INSERM, Université Paris-Saclay, Orsay, France
| | - Zeina Abou Nader
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Vincent Rondeau
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Amélie Bonaud
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Maria Kalogeraki
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | | | - Julia P Lemos
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Valeria Bisio
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Joyce Koenen
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- Inflammation, Microbiome and Immunosurveillance, INSERM, Université Paris-Saclay, Orsay, France
| | - Lea Hanna Doumit Sakr
- Université Paris Cité, BIOSCAR Inserm U1132, Department of Rheumatology and Reference Center for Rare Bone Diseases, AP-HP Hospital Lariboisière, Paris, France
| | - Amandine Picart
- Université Paris Cité, BIOSCAR Inserm U1132, Department of Rheumatology and Reference Center for Rare Bone Diseases, AP-HP Hospital Lariboisière, Paris, France
| | - Amélie Coudert
- Université Paris Cité, BIOSCAR Inserm U1132, Department of Rheumatology and Reference Center for Rare Bone Diseases, AP-HP Hospital Lariboisière, Paris, France
| | - Sylvain Provot
- Université Paris Cité, BIOSCAR Inserm U1132, Department of Rheumatology and Reference Center for Rare Bone Diseases, AP-HP Hospital Lariboisière, Paris, France
| | - Nicolas Dulphy
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Michel Aurrand-Lions
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Stéphane J C Mancini
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Gwendal Lazennec
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- CNRS, SYS2DIAG-ALCEDIAG, Cap Delta, Montpellier, France
| | - David H McDermott
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Fabien Guidez
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1131, Paris, France
| | | | - Philip M Murphy
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Martine Cohen-Solal
- Université Paris Cité, BIOSCAR Inserm U1132, Department of Rheumatology and Reference Center for Rare Bone Diseases, AP-HP Hospital Lariboisière, Paris, France
| | - Marion Espéli
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | | | - Karl Balabanian
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France.
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France.
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France.
| |
Collapse
|
22
|
Ko FC, Jochum SB, Wilson BM, Adra A, Patel N, Lee H, Wilber S, Shaikh M, Forsyth C, Keshavarzian A, Swanson GR, Sumner DR. Colon epithelial cell-specific Bmal1 deletion impairs bone formation in mice. Bone 2023; 168:116650. [PMID: 36584784 PMCID: PMC9911378 DOI: 10.1016/j.bone.2022.116650] [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: 10/12/2022] [Revised: 12/16/2022] [Accepted: 12/17/2022] [Indexed: 12/29/2022]
Abstract
The circadian clock system regulates multiple metabolic processes, including bone metabolism. Previous studies have demonstrated that both central and peripheral circadian signaling regulate skeletal growth and homeostasis in mice. Disruption in central circadian rhythms has been associated with a decline in bone mineral density in humans and the global and osteoblast-specific disruption of clock genes in bone tissue leads to lower bone mass in mice. Gut physiology is highly sensitive to circadian disruption. Since the gut is also known to affect bone remodeling, we sought to test the hypothesis that circadian signaling disruption in colon epithelial cells affects bone. We therefore assessed structural, functional, and cellular properties of bone in 8 week old Ts4-Cre and Ts4-Cre;Bmal1fl/fl (cBmalKO) mice, where the clock gene Bmal1 is deleted in colon epithelial cells. Axial and appendicular trabecular bone volume was significantly lower in cBmalKO compared to Ts4-Cre 8-week old mice in a sex-dependent fashion, with male but not female mice showing the phenotype. Similarly, the whole bone mechanical properties were deteriorated in cBmalKO male mice. The tissue level mechanisms involved suppressed bone formation with normal resorption, as evidenced by serum markers and dynamic histomorphometry. Our studies demonstrate that colon epithelial cell-specific deletion of Bmal1 leads to failure to acquire trabecular and cortical bone in male mice.
Collapse
Affiliation(s)
- Frank C Ko
- Department of Anatomy& Cell Biology, Rush University Medical Center, Chicago, IL 60612, United States of America; Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612, United States of America; Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, United States of America.
| | - Sarah B Jochum
- Division of Digestive Diseases and Nutrition, Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612, United States of America
| | - Brittany M Wilson
- Department of Anatomy& Cell Biology, Rush University Medical Center, Chicago, IL 60612, United States of America
| | - Amal Adra
- Department of Anatomy& Cell Biology, Rush University Medical Center, Chicago, IL 60612, United States of America
| | - Nikhil Patel
- Department of Anatomy& Cell Biology, Rush University Medical Center, Chicago, IL 60612, United States of America
| | - Hoomin Lee
- Department of Anatomy& Cell Biology, Rush University Medical Center, Chicago, IL 60612, United States of America
| | - Sherry Wilber
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, United States of America
| | - Maliha Shaikh
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, United States of America
| | - Christopher Forsyth
- Department of Anatomy& Cell Biology, Rush University Medical Center, Chicago, IL 60612, United States of America; Division of Digestive Diseases and Nutrition, Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612, United States of America; Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, United States of America
| | - Ali Keshavarzian
- Department of Anatomy& Cell Biology, Rush University Medical Center, Chicago, IL 60612, United States of America; Division of Digestive Diseases and Nutrition, Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612, United States of America; Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, United States of America
| | - Garth R Swanson
- Department of Anatomy& Cell Biology, Rush University Medical Center, Chicago, IL 60612, United States of America; Division of Digestive Diseases and Nutrition, Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612, United States of America; Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, United States of America
| | - D Rick Sumner
- Department of Anatomy& Cell Biology, Rush University Medical Center, Chicago, IL 60612, United States of America; Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612, United States of America; Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, United States of America
| |
Collapse
|
23
|
Madel MB, Halper J, Ibáñez L, Claire L, Rouleau M, Boutin A, Mahler A, Pontier-Bres R, Ciucci T, Topi M, Hue C, Amiaud J, Iborra S, Sancho D, Heymann D, Garchon HJ, Czerucka D, Apparailly F, Duroux-Richard I, Wakkach A, Blin-Wakkach C. Specific targeting of inflammatory osteoclastogenesis by the probiotic yeast S. boulardii CNCM I-745 reduces bone loss in osteoporosis. eLife 2023; 12:e82037. [PMID: 36848406 PMCID: PMC9977286 DOI: 10.7554/elife.82037] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 02/12/2023] [Indexed: 03/01/2023] Open
Abstract
Bone destruction is a hallmark of chronic inflammation, and bone-resorbing osteoclasts arising under such a condition differ from steady-state ones. However, osteoclast diversity remains poorly explored. Here, we combined transcriptomic profiling, differentiation assays and in vivo analysis in mouse to decipher specific traits for inflammatory and steady-state osteoclasts. We identified and validated the pattern-recognition receptors (PRR) Tlr2, Dectin-1, and Mincle, all involved in yeast recognition as major regulators of inflammatory osteoclasts. We showed that administration of the yeast probiotic Saccharomyces boulardii CNCM I-745 (Sb) in vivo reduced bone loss in ovariectomized but not sham mice by reducing inflammatory osteoclastogenesis. This beneficial impact of Sb is mediated by the regulation of the inflammatory environment required for the generation of inflammatory osteoclasts. We also showed that Sb derivatives as well as agonists of Tlr2, Dectin-1, and Mincle specifically inhibited directly the differentiation of inflammatory but not steady-state osteoclasts in vitro. These findings demonstrate a preferential use of the PRR-associated costimulatory differentiation pathway by inflammatory osteoclasts, thus enabling their specific inhibition, which opens new therapeutic perspectives for inflammatory bone loss.
Collapse
Affiliation(s)
- Maria-Bernadette Madel
- Université Côte d’Azur, CNRS, LP2MNiceFrance
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur - Centre Scientifique de MonacoNice and MonacoFrance
| | - Julia Halper
- Université Côte d’Azur, CNRS, LP2MNiceFrance
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur - Centre Scientifique de MonacoNice and MonacoFrance
| | - Lidia Ibáñez
- Department of Pharmacy, Cardenal Herrera-CEU UniversityValenciaSpain
| | | | - Matthieu Rouleau
- Université Côte d’Azur, CNRS, LP2MNiceFrance
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur - Centre Scientifique de MonacoNice and MonacoFrance
| | - Antoine Boutin
- Université Côte d’Azur, CNRS, LP2MNiceFrance
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur - Centre Scientifique de MonacoNice and MonacoFrance
| | - Adrien Mahler
- Université Côte d’Azur, CNRS, LP2MNiceFrance
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur - Centre Scientifique de MonacoNice and MonacoFrance
| | - Rodolphe Pontier-Bres
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur - Centre Scientifique de MonacoNice and MonacoFrance
- Centre Scientifiquede MonacoMonaco
| | - Thomas Ciucci
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesdaUnited States
| | - Majlinda Topi
- Université Côte d’Azur, CNRS, LP2MNiceFrance
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur - Centre Scientifique de MonacoNice and MonacoFrance
| | - Christophe Hue
- Université Paris-Saclay, UVSQ, INSERM, Infection et inflammationMontigny-Le-BretonneuxFrance
| | | | - Salvador Iborra
- Department of Immunology, Ophthalmology and ENT. School of Medicine, Universidad Complutense de MadridMadridSpain
| | - David Sancho
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
| | - Dominique Heymann
- Université de Nantes, Institut de Cancérologie de l’OuestSaint HerblainFrance
| | - Henri-Jean Garchon
- Université Paris-Saclay, UVSQ, INSERM, Infection et inflammationMontigny-Le-BretonneuxFrance
- Genetics Division, Ambroise Paré Hospital, AP-HPBoulogne-BillancourtFrance
| | - Dorota Czerucka
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur - Centre Scientifique de MonacoNice and MonacoFrance
- Centre Scientifiquede MonacoMonaco
| | | | | | - Abdelilah Wakkach
- Université Côte d’Azur, CNRS, LP2MNiceFrance
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur - Centre Scientifique de MonacoNice and MonacoFrance
| | - Claudine Blin-Wakkach
- Université Côte d’Azur, CNRS, LP2MNiceFrance
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur - Centre Scientifique de MonacoNice and MonacoFrance
| |
Collapse
|
24
|
Guo J, Wang F, Hu Y, Luo Y, Wei Y, Xu K, Zhang H, Liu H, Bo L, Lv S, Sheng S, Zhuang X, Zhang T, Xu C, Chen X, Su J. Exosome-based bone-targeting drug delivery alleviates impaired osteoblastic bone formation and bone loss in inflammatory bowel diseases. Cell Rep Med 2023; 4:100881. [PMID: 36603578 PMCID: PMC9873828 DOI: 10.1016/j.xcrm.2022.100881] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 11/12/2022] [Accepted: 12/12/2022] [Indexed: 01/06/2023]
Abstract
Systematic bone loss is commonly complicated with inflammatory bowel diseases (IBDs) with unclear pathogenesis and uncertain treatment. In experimental colitis mouse models established by dextran sulfate sodium and IL-10 knockout induced with piroxicam, bone mass and quality are significantly decreased. Colitis mice demonstrate a lower bone formation rate and fewer osteoblasts in femur. Bone marrow mesenchymal stem/stromal cells (BMSCs) from colitis mice tend to differentiate into adipocytes rather than osteoblasts. Serum from patients with IBD promotes adipogenesis of human BMSCs. RNA sequencing reveals that colitis downregulates Wnt signaling in BMSCs. For treatment, exosomes with Golgi glycoprotein 1 inserted could carry Wnt agonist 1 and accumulate in bone via intravenous administration. They could alleviate bone loss, promote bone formation, and accelerate fracture healing in colitis mice. Collectively, BMSC commitment in inflammatory microenvironment contributes to lower bone quantity and quality and could be rescued by redirecting differentiation toward osteoblasts through bone-targeted drug delivery.
Collapse
Affiliation(s)
- Jiawei Guo
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Department of Orthopedics Trauma, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Fuxiao Wang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Yan Hu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Ying Luo
- Department of Orthopedics Trauma, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yan Wei
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Ke Xu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Hao Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Han Liu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Lumin Bo
- Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Shunli Lv
- Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Shihao Sheng
- Department of Orthopedics Trauma, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Xinchen Zhuang
- Department of Orthopedics Trauma, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Tao Zhang
- Department of Orthopedics Trauma, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Can Xu
- Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| | - Xiao Chen
- Department of Orthopedics Trauma, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Department of Orthopedics Trauma, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China; Organoid Research Center, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
25
|
Wu Y, Yang Y, Wang L, Chen Y, Han X, Sun L, Chen H, Chen Q. Effect of Bifidobacterium on osteoclasts: TNF-α/NF-κB inflammatory signal pathway-mediated mechanism. Front Endocrinol (Lausanne) 2023; 14:1109296. [PMID: 36967748 PMCID: PMC10034056 DOI: 10.3389/fendo.2023.1109296] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 02/14/2023] [Indexed: 03/11/2023] Open
Abstract
Osteoporosis is a systemic multifactorial bone disease characterized by low bone quality and density and bone microstructure damage, increasing bone fragility and fracture vulnerability. Increased osteoclast differentiation and activity are important factors contributing to bone loss, which is a common pathological manifestation of bone diseases such as osteoporosis. TNF-a/NF-κB is an inflammatory signaling pathway with a key regulatory role in regulating osteoclast formation, and the classical pathway RANKL/RANK/OPG assists osteoclast formation. Activation of this inflammatory pathway promotes the formation of osteoclasts and accelerates the process of osteoporosis. Recent studies and emerging evidence have consistently demonstrated the potential of probiotics to modulate bone health. Secretions of Bifidobacterium, a genus of probiotic bacteria in the phylum Actinobacteria, such as short-chain fatty acids, equol, and exopolysaccharides, have indicated beneficial effects on bone health. This review discusses the molecular mechanisms of the TNF-a/NF-κB inflammatory pathway in regulating osteoclast formation and describes the secretions produced by Bifidobacterium and their potential effects on bone health through this pathway, opening up new directions for future research.
Collapse
Affiliation(s)
- Yue Wu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yunjiao Yang
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lan Wang
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yiding Chen
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xuke Han
- College of Acupuncture & Tuina, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Lisha Sun
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huizhen Chen
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiu Chen
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Qiu Chen,
| |
Collapse
|
26
|
Chiu LS, Anderton RS. The role of the microbiota-gut-brain axis in long-term neurodegenerative processes following traumatic brain injury. Eur J Neurosci 2023; 57:400-418. [PMID: 36494087 PMCID: PMC10107147 DOI: 10.1111/ejn.15892] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022]
Abstract
Traumatic brain injury (TBI) can be a devastating and debilitating disease to endure. Due to improvements in clinical practice, declining mortality rates have led to research into the long-term consequences of TBI. For example, the incidence and severity of TBI have been associated with an increased susceptibility of developing neurodegenerative disorders, such as Parkinson's or Alzheimer's disease. However, the mechanisms linking this alarming association are yet to be fully understood. Recently, there has been a groundswell of evidence implicating the microbiota-gut-brain axis in the pathogenesis of these diseases. Interestingly, survivors of TBI often report gastrointestinal complaints and animal studies have demonstrated gastrointestinal dysfunction and dysbiosis following injury. Autonomic dysregulation and chronic inflammation appear to be the main driver of these pathologies. Consequently, this review will explore the potential role of the microbiota-gut-brain axis in the development of neurodegenerative diseases following TBI.
Collapse
Affiliation(s)
- Li Shan Chiu
- School of Medicine, The University Notre Dame Australia, Fremantle, Western Australia, Australia
- Ear Science Institute Australia, Nedlands, Western Australia, Australia
| | - Ryan S Anderton
- Institute for Health Research, The University Notre Dame Australia, Fremantle, Western Australia, Australia
| |
Collapse
|
27
|
Artoni de Carvalho JA, Magalhães LR, Polastri LM, Batista IET, de Castro Bremer S, Caetano HRDS, Rufino MN, Bremer-Neto H. Prebiotics improve osteoporosis indicators in a preclinical model: systematic review with meta-analysis. Nutr Rev 2022; 81:nuac097. [PMID: 36474436 DOI: 10.1093/nutrit/nuac097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024] Open
Abstract
CONTEXT Studies using experimental models have demonstrated that prebiotics are involved in antiosteoporotic mechanisms. OBJECTIVE This study was conducted to determine the impact of supplementation with prebiotics in the basal diet of ovariectomized rats with induced osteoporosis as a preclinical model. METHODS A comprehensive systematic search was carried out in the electronic databases PubMed, Science Direct, Web of Science, Scielo, and Google through March 2022 for studies that investigated the impact of prebiotics on bone mineral density (BMD), bone mineral content (BMC), and bone biomechanics. RESULTS The search returned 844 complete articles, abstracts, or book chapters. After detailed screening, 8 studies met the inclusion criteria. Rats (n = 206), were randomly divided between control and treatment groups. Weighted mean differences (WMDs) with the 95%CIs were used to estimate the combined effect size. Compared with the control group, dietary intake of prebiotics significantly increased bone density in the BMD subgroups, with WMDs as follows: 0.03 g/cm3, 95%CI, 0.01-0.05, P < 0.00001, n = 46; and 0.00 g/cm2, 95%CI, 0.00-0.02, P < 0.00001, n = 81; total BMD: WMD, 0.01, 95%CI, 0.01-0.02, P < 0.00001, n = 127; bone content in BMC: WMD, 0.02 g, 95%CI, 0.00-0.04, P = 0.05, n = 107; and the 3-point-bend test: WMD, 15.20 N, 95%CI, 5.92-24.47, P = 0.00001, n = 120. CONCLUSION Prebiotics improve indicators of osteoporosis, BMD, BMC, and bone biomechanics in ovariectomized rats. More studies are needed to increase the level of evidence. SYSTEMIC REVIEW REGISTRATION Systematic Review Protocol for Animal Intervention Studies.
Collapse
Affiliation(s)
- João Alberto Artoni de Carvalho
- Department of Orthopedics and Traumatology, Medicine School of Presidente Prudente, Western Sao Paulo University, Presidente Prudente, São Paulo, Brazil
| | - Leticia Rocha Magalhães
- Department of Orthopedics and Traumatology, Medicine School of Presidente Prudente, Western Sao Paulo University, Presidente Prudente, São Paulo, Brazil
| | - Laryssa Mayara Polastri
- Department of Orthopedics and Traumatology, Medicine School of Presidente Prudente, Western Sao Paulo University, Presidente Prudente, São Paulo, Brazil
| | - Ingrid Eloise Trombine Batista
- Department of Orthopedics and Traumatology, Medicine School of Presidente Prudente, Western Sao Paulo University, Presidente Prudente, São Paulo, Brazil
| | | | - Heliard Rodrigues Dos Santos Caetano
- Department of Functional Sciences, Health Technology Assessment Nucleus of the Medical School of Presidente Prudente, Western Sao Paulo University, Presidente Prudente, São Paulo, Brazil
| | - Marcos Natal Rufino
- Department of Functional Sciences, Health Technology Assessment Nucleus of the Medical School of Presidente Prudente, Western Sao Paulo University, Presidente Prudente, São Paulo, Brazil
| | - Hermann Bremer-Neto
- Department of Functional Sciences, Health Technology Assessment Nucleus of the Medical School of Presidente Prudente, Western Sao Paulo University, Presidente Prudente, São Paulo, Brazil
| |
Collapse
|
28
|
Shao C, Liu Y, Li J, Liu Z, Zhao Y, Jing Y, Lv Z, Fu T, Wang Z, Li G. Up-regulated IL-17 and Tnf signaling in bone marrow cells of young male osteogenesis imperfecta mice. PeerJ 2022; 10:e13963. [PMID: 36032950 PMCID: PMC9415356 DOI: 10.7717/peerj.13963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/08/2022] [Indexed: 01/19/2023] Open
Abstract
Osteogenesis imperfecta (OI) is a congenital bone dysplasia mainly caused by either defective production or assembly of type I collagen. The skeletal phenotypes especially fractures are often seen in OI adolescents. Studies have found that an increased number of osteoclasts and excessive bone resorption existed in collagen-related OI, which has not been well understood. Emerging evidence has suggested that inflammation may be associated with OI. We speculated that the bone marrow (BM) niche had similar inflammatory changes and performed RNA-sequencing (RNA-seq) in BM cells derived from young male mice to analyze the related differentially expressed genes (DEGs) and pathways. Data showed that there were 117 shared DEGs (Q ≤ 0.05, |log2FC| ≥ 1) in BM cells isolated from two types of OI murine models that respectively simulate different OI types. Gene Ontology (GO) (Q ≤ 0.05) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) (Q ≤ 0.05) analysis and real-time PCR validation indicated the dysregulated biology process of cellular response to interferon (Ifn) together with upregulated IL-17 signaling, tumor necrosis factor (Tnf) signaling and osteoclast differentiation in OI BM niche. Either defective collagen production or abnormal collagen assembly shared similar alterations in gene profiles and pathways involving inflammation and osteoclast activation. Data presented here not only contributed to understanding of the mechanism of the enhanced bone absorption in the bones of OI, but also provided more evidence to develop potential anti-inflammation therapies.
Collapse
Affiliation(s)
| | - Yi Liu
- Tianjin Medical University, Tianjin, China
| | - Jiaci Li
- Tianjin Pediatric Research Institute, Tianjin Children’s Hospital, Tianjin, Longyan Road, Beichen District, Tianjin, China
| | - Ziyun Liu
- Tianjin Medical University, Tianjin, China
| | - Yuxia Zhao
- Tianjin Medical University, Tianjin, China
| | | | - Zhe Lv
- Tianjin Medical University, Tianjin, China
| | - Ting Fu
- Tianjin Medical University, Tianjin, China
| | - Zihan Wang
- Tianjin Medical University, Tianjin, China
| | - Guang Li
- Tianjin Medical University, Tianjin, China
| |
Collapse
|
29
|
Peek CT, Ford CA, Eichelberger KR, Jacobse J, Torres TP, Maseda D, Latour YL, Piazuelo MB, Johnson JR, Byndloss MX, Wilson KT, Rathmell JC, Goettel JA, Cassat JE. Intestinal Inflammation Promotes MDL-1 + Osteoclast Precursor Expansion to Trigger Osteoclastogenesis and Bone Loss. Cell Mol Gastroenterol Hepatol 2022; 14:731-750. [PMID: 35835390 PMCID: PMC9420375 DOI: 10.1016/j.jcmgh.2022.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Inflammatory bowel disease (IBD) is characterized by severe gastrointestinal inflammation, but many patients experience extra-intestinal disease. Bone loss is one common extra-intestinal manifestation of IBD that occurs through dysregulated interactions between osteoclasts and osteoblasts. Systemic inflammation has been postulated to contribute to bone loss, but the specific pathologic mechanisms have not yet been fully elucidated. We hypothesized that intestinal inflammation leads to bone loss through increased abundance and altered function of osteoclast progenitors. METHODS We used chemical, T cell driven, and infectious models of intestinal inflammation to determine the impact of intestinal inflammation on bone volume, the skeletal cytokine environment, and the cellular changes to pre-osteoclast populations within bone marrow. Additionally, we evaluated the potential for monoclonal antibody treatment against an inflammation-induced osteoclast co-receptor, myeloid DNAX activation protein 12-associating lectin-1 (MDL-1) to reduce bone loss during colitis. RESULTS We observed significant bone loss across all models of intestinal inflammation. Bone loss was associated with an increase in pro-osteoclastogenic cytokines within the bone and an expansion of a specific Cd11b-/loLy6Chi osteoclast precursor (OCP) population. Intestinal inflammation led to altered OCP expression of surface receptors involved in osteoclast differentiation and function, including the pro-osteoclastogenic co-receptor MDL-1. OCPs isolated from mice with intestinal inflammation demonstrated enhanced osteoclast differentiation ex vivo compared to controls, which was abrogated by anti-MDL-1 antibody treatment. Importantly, in vivo anti-MDL-1 antibody treatment ameliorated bone loss during intestinal inflammation. CONCLUSIONS Collectively, these data implicate the pathologic expansion and altered function of OCPs expressing MDL-1 in bone loss during IBD.
Collapse
Affiliation(s)
- Christopher T Peek
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Caleb A Ford
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Kara R Eichelberger
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Justin Jacobse
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Teresa P Torres
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Damian Maseda
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Yvonne L Latour
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - M Blanca Piazuelo
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Joshua R Johnson
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Mariana X Byndloss
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Keith T Wilson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, Tennessee; Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Jeffrey C Rathmell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jeremy A Goettel
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, Tennessee
| | - James E Cassat
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
30
|
Ahmad SS, Ahmed F, Ali R, Ghoneim MM, Alshehri S, Najmi AK, Ahmad S, Ahmad MZ, Ahmad J, Khan MA. Immunology of osteoporosis: relevance of inflammatory targets for the development of novel interventions. Immunotherapy 2022; 14:815-831. [PMID: 35765988 DOI: 10.2217/imt-2021-0282] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Osteoporosis is recognized as low bone mass and deteriorated bone microarchitecture. It is the leading cause of fractures and consequent morbidity globally. The established pathophysiological evidence favors the endocrine factors for osteoporosis and the role of the immune system on the skeletal system has been recently identified. Due to the common developmental niche bone and immune system interactions have led to the emergence of osteoimmunology. Immune dysregulation can initiate inflammatory conditions that adversely affect bone integrity. The role of immune cells, such as T-lymphocytes subsets (Th17), cannot be neglected in the pathogenesis of osteoporosis. Local inflammation within the bone from any cause attracts immune cells that participate in the activation of osteoclasts. This work summarizes the present knowledge of osteoimmunology in reference to osteoporosis and identifies novel targets for immunotherapy of osteoporosis.
Collapse
Affiliation(s)
- Syed Sufian Ahmad
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Faraha Ahmed
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Ruhi Ali
- Delhi Institute of Pharmaceutical Education & Research (DIPSAR), DPSRU, New Delhi, 110017, India
| | - Mohammed M Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, AdDiriyah, 13713, Saudi Arabia
| | - Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Sayeed Ahmad
- Department of Pharmacognosy & Phytochemistry, Bioactive Natural Product Laboratory, School of Pharmaceutical Education & Research, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Mohammad Zaki Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, 11001, Saudi Arabia
| | - Javed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, 11001, Saudi Arabia
| | - Mohammad Ahmed Khan
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| |
Collapse
|
31
|
Tayyab S, Rual E, Nagari M. Improving compliance to DEXA in IBD population according to BSG guidelines in Morriston Hospital, Swansea. Clin Med (Lond) 2022; 22 Suppl 4:72-73. [PMID: 38614604 DOI: 10.7861/clinmed.22-4-s72] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
32
|
Marie JC, Bonnelye E. Effects of Estrogens on Osteoimmunology: A Role in Bone Metastasis. Front Immunol 2022; 13:899104. [PMID: 35677054 PMCID: PMC9168268 DOI: 10.3389/fimmu.2022.899104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/26/2022] [Indexed: 12/02/2022] Open
Abstract
Bone loss associated with estrogen deficiency indicates a fundamental role of these hormones in skeletal growth and bone remodeling. In the last decades, growing recent evidence demonstrated that estrogens can also affect the immune compartment of the bone. In this review, we summarize the impacts of estrogens on bone immune cells and their consequences on bone homeostasis, metastasis settlement into the bone and tumor progression. We also addressed the role of an orphan nuclear receptor ERRalpha (“Estrogen-receptor Related Receptor alpha”) on macrophages and T lymphocytes, and as an immunomodulator in bone metastases. Hence, this review links estrogens to bone immune cells in osteo-oncology.
Collapse
Affiliation(s)
- Julien C Marie
- Cancer Research Center of Lyon (CRCL), Tumor Escape Resistance Immunity Department, INSERM-1052, CNRS 5286, Centre Léon Bérard, Université Claude Bernard Lyon 1, Lyon, France
| | - Edith Bonnelye
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-UMR1277-Canther-Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| |
Collapse
|
33
|
Tian J, Chung HK, Moon JS, Nga HT, Lee HY, Kim JT, Chang JY, Kang SG, Ryu D, Che X, Choi J, Tsukasaki M, Sasako T, Lee S, Shong M, Yi H. Skeletal muscle mitoribosomal defects are linked to low bone mass caused by bone marrow inflammation in male mice. J Cachexia Sarcopenia Muscle 2022; 13:1785-1799. [PMID: 35306755 PMCID: PMC9178379 DOI: 10.1002/jcsm.12975] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 02/01/2022] [Accepted: 02/15/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Mitochondrial oxidative phosphorylation (OxPhos) is a critical regulator of skeletal muscle mass and function. Although muscle atrophy due to mitochondrial dysfunction is closely associated with bone loss, the biological characteristics of the relationship between muscle and bone remain obscure. We showed that muscle atrophy caused by skeletal muscle-specific CR6-interacting factor 1 knockout (MKO) modulates the bone marrow (BM) inflammatory response, leading to low bone mass. METHODS MKO mice with lower muscle OxPhos were fed a normal chow or high-fat diet and then evaluated for muscle mass and function, and bone mineral density. Immunophenotyping of BM immune cells was also performed. BM transcriptomic analysis was used to identify key factors regulating bone mass in MKO mice. To determine the effects of BM-derived CXCL12 (C-X-C motif chemokine ligand 12) on regulation of bone homeostasis, a variety of BM niche-resident cells were treated with recombinant CXCL12. Vastus lateralis muscle and BM immune cell samples from 14 patients with hip fracture were investigated to examine the association between muscle function and BM inflammation. RESULTS MKO mice exhibited significant reductions in both muscle mass and expression of OxPhos subunits but increased transcription of mitochondrial stress response-related genes in the extensor digitorum longus (P < 0.01). MKO mice showed a decline in grip strength and a higher drop rate in the wire hanging test (P < 0.01). Micro-computed tomography and von Kossa staining revealed that MKO mice developed a low mass phenotype in cortical and trabecular bone (P < 0.01). Transcriptomic analysis of the BM revealed that mitochondrial stress responses in skeletal muscles induce an inflammatory response and adipogenesis in the BM and that the CXCL12-CXCR4 (C-X-C chemokine receptor 4) axis is important for T-cell homing to the BM. Antagonism of CXCR4 attenuated BM inflammation and increased bone mass in MKO mice. In humans, patients with low body mass index (BMI = 17.2 ± 0.42 kg/m2 ) harboured a larger population of proinflammatory and cytotoxic senescent T-cells in the BMI (P < 0.05) and showed reduced expression of OxPhos subunits in the vastus lateralis, compared with controls with a normal BMI (23.7 ± 0.88 kg/m2 ) (P < 0.01). CONCLUSIONS Defects in muscle mitochondrial OxPhos promote BM inflammation in mice, leading to decreased bone mass. Muscle mitochondrial dysfunction is linked to BM inflammatory cytokine secretion via the CXCL12-CXCR4 signalling axis, which is critical for inducing low bone mass.
Collapse
Affiliation(s)
- Jingwen Tian
- Department of Medical ScienceChungnam National UniversityDaejeonKorea
- Laboratory of Endocrinology and Immune SystemChungnam National University School of MedicineDaejeonKorea
| | - Hyo Kyun Chung
- Research Center for Endocrine and Metabolic DiseasesChungnam National University School of MedicineDaejeonKorea
| | - Ji Sun Moon
- Laboratory of Endocrinology and Immune SystemChungnam National University School of MedicineDaejeonKorea
- Research Center for Endocrine and Metabolic DiseasesChungnam National University School of MedicineDaejeonKorea
| | - Ha Thi Nga
- Department of Medical ScienceChungnam National UniversityDaejeonKorea
- Laboratory of Endocrinology and Immune SystemChungnam National University School of MedicineDaejeonKorea
| | - Ho Yeop Lee
- Department of Medical ScienceChungnam National UniversityDaejeonKorea
- Laboratory of Endocrinology and Immune SystemChungnam National University School of MedicineDaejeonKorea
| | - Jung Tae Kim
- Department of Medical ScienceChungnam National UniversityDaejeonKorea
- Research Center for Endocrine and Metabolic DiseasesChungnam National University School of MedicineDaejeonKorea
| | - Joon Young Chang
- Department of Medical ScienceChungnam National UniversityDaejeonKorea
- Research Center for Endocrine and Metabolic DiseasesChungnam National University School of MedicineDaejeonKorea
| | - Seul Gi Kang
- Department of Medical ScienceChungnam National UniversityDaejeonKorea
- Research Center for Endocrine and Metabolic DiseasesChungnam National University School of MedicineDaejeonKorea
| | - Dongryeol Ryu
- Department of Molecular Cell BiologySungkyunkwan University School of MedicineSuwonKorea
- Samsung Biomedical Research InstituteSamsung Medical CenterSeoulKorea
| | - Xiangguo Che
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, School of MedicineKyungpook National UniversityDaeguKorea
- Department of Internal Medicine, Rheumatology and ImmunologyThe Affiliated Hospital of Yanbian UniversityYanjiChina
| | - Je‐Yong Choi
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, School of MedicineKyungpook National UniversityDaeguKorea
| | - Masayuki Tsukasaki
- Department of Immunology, Graduate School of Medicine and Faculty of MedicineThe University of TokyoTokyoJapan
| | - Takayoshi Sasako
- Department of Diabetes and Metabolic Diseases, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Sang‐Hee Lee
- Bio‐Electron Microscopy Research Center (104‐Dong)Korea Basic Science InstituteCheongjuKorea
| | - Minho Shong
- Department of Medical ScienceChungnam National UniversityDaejeonKorea
- Research Center for Endocrine and Metabolic DiseasesChungnam National University School of MedicineDaejeonKorea
| | - Hyon‐Seung Yi
- Department of Medical ScienceChungnam National UniversityDaejeonKorea
- Laboratory of Endocrinology and Immune SystemChungnam National University School of MedicineDaejeonKorea
- Research Center for Endocrine and Metabolic DiseasesChungnam National University School of MedicineDaejeonKorea
| |
Collapse
|
34
|
Fu H, Wang L, Bao Q, Ni D, Hu P, Shi J. Acid Neutralization and Immune Regulation by Calcium-Aluminum-Layered Double Hydroxide for Osteoporosis Reversion. J Am Chem Soc 2022; 144:8987-8999. [PMID: 35549335 DOI: 10.1021/jacs.2c00749] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Osteoporosis is a kind of global chronic bone disease characterized by progressive loss of bone mass and bone quality reduction, leading to a largely increased risk of bone fragility. In clinics, the current treatment of osteoporosis relies on the inhibition of bone damage by osteoclasts but ignores the function of immune cells in the progress of osteoporosis, leading to much compromised therapeutic efficacy. In this work, a highly effective osteoporosis-immunotherapeutic modality is established for the treatment of osteoporosis based on acid neutralization in synergy with immune microenvironment regulation by a specially designed nanocatalytic medicine, calcein functionalized calcium-aluminum-layered double hydroxide (CALC) nanosheets. Briefly, the mildly alkaline CALC nanosheets could neutralize the acidic microenvironment of osteoporosis accompanying the acidity-responsive LDH degradation. Subsequently, calcium phosphate nanoparticles (CAPs) are generated by the reaction between the released Ca2+ from LDH degradation and endogenous phosphates, resulting in M2 phenotype anti-inflammatory differentiation of bone macrophages through a c-Maf transcriptional factor pathway and the following activity enhancements of regulatory T cells (Treg) and the deactivation of T helper 17 cells (TH17). Both in vitro and in vivo results show an excellent therapeutic efficacy on osteoporosis featuring a significant BV/TV (%) enhancement of femurs from 6.2 to 10.7, demonstrating high feasibility of this therapeutic concept through the combined acid neutralization and immune regulation. Such an inorganic nanomaterial-based strategy provides a novel, efficient, and biosafe therapeutic modality for intractable osteoporosis treatment, which will benefit patients suffering from osteoporosis.
Collapse
Affiliation(s)
- Hao Fu
- State Key Laboratory of High-Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai 200050, P. R. China
| | - Lingtian Wang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, P. R. China
| | - Qunqun Bao
- State Key Laboratory of High-Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai 200050, P. R. China.,Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200331, P. R. China
| | - Dalong Ni
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, P. R. China
| | - Ping Hu
- State Key Laboratory of High-Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai 200050, P. R. China.,Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200331, P. R. China
| | - Jianlin Shi
- State Key Laboratory of High-Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai 200050, P. R. China.,Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200331, P. R. China
| |
Collapse
|
35
|
Connection between Mesenchymal Stem Cells Therapy and Osteoclasts in Osteoarthritis. Int J Mol Sci 2022; 23:ijms23094693. [PMID: 35563083 PMCID: PMC9102843 DOI: 10.3390/ijms23094693] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 12/12/2022] Open
Abstract
The use of mesenchymal stem cells constitutes a promising therapeutic approach, as it has shown beneficial effects in different pathologies. Numerous in vitro, pre-clinical, and, to a lesser extent, clinical trials have been published for osteoarthritis. Osteoarthritis is a type of arthritis that affects diarthritic joints in which the most common and studied effect is cartilage degradation. Nowadays, it is known that osteoarthritis is a disease with a very powerful inflammatory component that affects the subchondral bone and the rest of the tissues that make up the joint. This inflammatory component may induce the differentiation of osteoclasts, the bone-resorbing cells. Subchondral bone degradation has been suggested as a key process in the pathogenesis of osteoarthritis. However, very few published studies directly focus on the activity of mesenchymal stem cells on osteoclasts, contrary to what happens with other cell types of the joint, such as chondrocytes, synoviocytes, and osteoblasts. In this review, we try to gather the published bibliography in relation to the effects of mesenchymal stem cells on osteoclastogenesis. Although we find promising results, we point out the need for further studies that can support mesenchymal stem cells as a therapeutic tool for osteoclasts and their consequences on the osteoarthritic joint.
Collapse
|
36
|
Garza-Hernandez D, Sepulveda-Villegas M, Garcia-Pelaez J, Aguirre-Gamboa R, Lakatos PL, Estrada K, Martinez-Vazquez M, Trevino V. A systematic review and functional bioinformatics analysis of genes associated with Crohn's disease identify more than 120 related genes. BMC Genomics 2022; 23:302. [PMID: 35418025 PMCID: PMC9008988 DOI: 10.1186/s12864-022-08491-y] [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] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 02/28/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Crohn's disease is one of the two categories of inflammatory bowel diseases that affect the gastrointestinal tract. The heritability estimate has been reported to be 0.75. Several genes linked to Crohn's disease risk have been identified using a plethora of strategies such as linkage-based studies, candidate gene association studies, and lately through genome-wide association studies (GWAS). Nevertheless, to our knowledge, a compendium of all the genes that have been associated with CD is lacking. METHODS We conducted functional analyses of a gene set generated from a systematic review where genes potentially related to CD found in the literature were analyzed and classified depending on the genetic evidence reported and putative biological function. For this, we retrieved and analyzed 2496 abstracts comprising 1067 human genes plus 22 publications regarding 133 genes from GWAS Catalog. Then, each gene was curated and categorized according to the type of evidence associated with Crohn's disease. RESULTS We identified 126 genes associated with Crohn's disease risk by specific experiments. Additionally, 71 genes were recognized associated through GWAS alone, 18 to treatment response, 41 to disease complications, and 81 to related diseases. Bioinformatic analysis of the 126 genes supports their importance in Crohn's disease and highlights genes associated with specific aspects such as symptoms, drugs, and comorbidities. Importantly, most genes were not included in commercial genetic panels suggesting that Crohn's disease is genetically underdiagnosed. CONCLUSIONS We identified a total of 126 genes from PubMed and 71 from GWAS that showed evidence of association to diagnosis, 18 to treatment response, and 41 to disease complications in Crohn's disease. This prioritized gene catalog can be explored at http://victortrevino.bioinformatics.mx/CrohnDisease .
Collapse
Affiliation(s)
- Debora Garza-Hernandez
- Tecnologico de Monterrey, Escuela de Medicina, Cátedra de Bioinformática, Av. Morones Prieto No. 3000, Colonia Los Doctores, 64710, Monterrey, Nuevo León, Mexico
| | - Maricruz Sepulveda-Villegas
- Tecnologico de Monterrey, Escuela de Medicina, Cátedra de Bioinformática, Av. Morones Prieto No. 3000, Colonia Los Doctores, 64710, Monterrey, Nuevo León, Mexico
| | - Jose Garcia-Pelaez
- Instituto de Investigação e Inovação em Saude-i3S, Universidade do Porto, Porto, Portugal
- Ipatimup, Institute of Molecular Pathology and Immunology at the University of Porto, Porto, Portugal
| | | | - Peter L Lakatos
- McGill University Health Centre, Division of Gastroenterology, IBD Centre, Montreal General Hospital, 1650 Ave. Cedar, D16.173.1, Montreal, QC, H3G 1A4, Canada
| | - Karol Estrada
- Graduate Professional Studies, Brandeis University, Waltham, MA, 02453, USA
| | - Manuel Martinez-Vazquez
- Tecnologico de Monterrey, Instituto de Medicina Interna, Centro Médico Zambrano Hellion, Av. Batallón de San Patricio No. 112, Colonia Real San Agustín, 66278, San Pedro Garza García, Nuevo León, Mexico
| | - Victor Trevino
- Tecnologico de Monterrey, Escuela de Medicina, Cátedra de Bioinformática, Av. Morones Prieto No. 3000, Colonia Los Doctores, 64710, Monterrey, Nuevo León, Mexico.
- Tecnologico de Monterrey, The Institute for Obesity Research, Integrative Biology Unit, Eugenio Garza Sada 2501 Avenue, 64849, Monterrey, Nuevo Leon, Mexico.
| |
Collapse
|
37
|
Commensal gut bacterium critically regulates alveolar bone homeostasis. J Transl Med 2022; 102:363-375. [PMID: 34934182 PMCID: PMC8967765 DOI: 10.1038/s41374-021-00697-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 12/28/2022] Open
Abstract
The alveolar bone is a unique osseous tissue due to the presence of the teeth and the proximity of commensal oral microbes. Commensal microbe effects on alveolar bone homeostasis have been attributed to the oral microbiota, yet the impact of commensal gut microbes is unknown. Study purpose was to elucidate whether commensal gut microbes regulate osteoimmune mechanisms and skeletal homeostasis in alveolar bone. Male C57BL/6T germfree (GF) littermate mice were maintained as GF or monoassociated with segmented filamentous bacteria (SFB), a commensal gut bacterium. SFB has been shown to elicit broad immune response effects, including the induction of TH17/IL17A immunity, which impacts the development and homeostasis of host tissues. SFB colonized the gut, but not oral cavity, and increased IL17A levels in the ileum and serum. SFB had catabolic effects on alveolar bone and non-oral skeletal sites, which was attributed to enhanced osteoclastogenesis. The alveolar bone marrow of SFB vs. GF mice had increased dendritic cells, activated helper T-cells, TH1 cells, TH17 cells, and upregulated Tnf. Primary osteoblast cultures from SFB and GF mice were stimulated with vehicle-control, IL17A, or TNF to elucidate osteoblast-derived signaling factors contributing to the pro-osteoclastic phenotype in SFB mice. Treatment of RAW264.7 osteoclastic cells with supernatants from vehicle-stimulated SFB vs. GF osteoblasts recapitulated the osteoclast phenotype found in vivo. Supernatants from TNF-stimulated osteoblasts normalized RAW264.7 osteoclast endpoints across SFB and GF cultures, which was dependent on the induction of CXCL1 and CCL2. This report reveals that commensal gut microbes have the capacity to regulate osteoimmune processes in alveolar bone. Outcomes from this investigation challenge the current paradigm that alveolar bone health and homeostasis is strictly regulated by oral microbes.
Collapse
|
38
|
Interleukin-17 promotes osteoclastogenesis and periodontal damage via autophagy in vitro and in vivo. Int Immunopharmacol 2022; 107:108631. [PMID: 35219162 DOI: 10.1016/j.intimp.2022.108631] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 02/11/2022] [Accepted: 02/13/2022] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Because of its potent pro-inflammatory properties, interleukin-17 (IL-17) contributes to the pathogenesis of various inflammatory diseases. This study explored the effects of IL-17 on osteoclastogenesis in an osteoclast monoculture and osteoblast-osteoclast co-culture system, as tools to investigate the molecular mechanisms underlying the interactions between osteoclastogenesis and autophagy. METHODS Various ratios of calvarial osteoblasts (OB) and osteoclast precursor cells (mouse macrophage cell line RAW264.7, hereinafter referred to as OC) were tested. Tartrate-resistant acid phosphatase (TRAP) staining was used to detect the optimum osteoblasts:osteoclasts ratio. IL-17 was added to the co-culture system to test its effects on multinucleated osteoclast formation and osteoclast-related proteins. We assessed the effects of IL-17 on receptor activator of nuclear factor-kappa B ligand (RANKL) expression in osteoblasts, and determined if IL-17 alone could modulate osteoclast formation in an osteoclast monoculture. Administration of exogenous RANKL combined with IL-17 was employed to stimulate RAW264.7 cells osteoclastogenesis and to determine production of osteoclasts and autophagy-related proteins. We knocked down Beclin1 expression in RAW264.7 cells and examined the expression of autophagy-related and osteoclast-related proteins in RAW264.7 cells and the co-culture system, and the TAK1-binding protein 3 (TAB3)/ extracellular signal regulated kinase (ERK) pathway. RESULTS A ratio of 20 OB : 1 OC yielded the highest rate of osteoclast formation. Low IL-17 concentrations increased osteoclastogenesis in co-cultures significantly, but high levels of IL-17 had the opposite effect. IL-17 alone could not induce formation of TRAP+ multinucleated cells in RAW264.7 cells. Low IL-17 concentrations promoted osteoclast differentiation and autophagy in RAW264.7 cells induced by exogenous RANKL, but high IL-17 concentrations inhibited this process. Knockdown of Beclin1 reversed the enhanced effects of 0.1 ng/mL IL-17 on osteoclastogenesis and autophagy in RAW264.7 cells. The TAB3/ERK pathway was also blocked after autophagy inhibition. CONCLUSION In the co-culture model used in this study, a ratio of 20 OB:1 OC proved to be the optimal ratio to facilitate osteoclast formation. IL-17 regulated RANKL-induced osteoclastogenesis via autophagy. The Beclin1/TAB3/ERK pathway was involved in osteoclast autophagy.
Collapse
|
39
|
Ye F, Li J, Xu P, Xie Z, Zheng G, Liu W, Ye G, Yu W, Lin J, Su Z, Che Y, Zhang Z, Wang P, Wu Y, Shen H. Osteogenic differentiation of mesenchymal stem cells promotes c-Jun-dependent secretion of interleukin 8 and mediates the migration and differentiation of CD4+ T cells. Stem Cell Res Ther 2022; 13:58. [PMID: 35123547 PMCID: PMC8818240 DOI: 10.1186/s13287-022-02735-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/25/2021] [Indexed: 02/07/2023] Open
Abstract
Background The immune system and the skeletal system have complex interactions in the bone marrow and even in the joints, which has promoted the development of the concept of osteoimmunology. Some evidence has indicated that T cells and B cells contribute to the balance between the resorption and formation of bone. However, there has been little discussion on the regulation of CD4+ T lymphocytes by cells involved in bone metabolism. Mesenchymal stem cells (MSCs), which exert core functions related to immunoregulation and osteogenic differentiation, are crucial cells linked to both bone metabolism and the immune system. Previous studies have shown that the immunoregulatory capacity of MSCs changes following differentiation. However, it is still unclear whether the osteogenic differentiation of MSCs affects the migration and differentiation of CD4+ T cells. Methods MSCs were cultured in growth medium or osteogenic medium for 10 days and then cocultured with CD4+ T cells. CD4+ T cell migration and differentiation were detected by flow cytometry. Further, gene expression levels of specific cytokines were analyzed by quantitative real-time PCR and enzyme-linked immunosorbent assays. A Proteome Profiler Human XL Cytokine Array Kit was used to analyze supernatants collected from MSCs. Alizarin red S staining and Alkaline phosphatase assay were used to detect the osteogenic differentiation of MSCs. Results Here, we found that the migration of CD4+ T cells was elevated, and the capacity to induce the differentiation of regulatory T (Treg) cells was weakened during MSC osteogenic differentiation, while the differentiation of T helper 1 (Th1), T helper 2 (Th2) and T helper 17 (Th17) cells was not affected. Further studies revealed that interleukin (IL)-8 was significantly upregulated during MSC osteogenic differentiation. Both a neutralizing antibody and IL-8-specific siRNA significantly inhibited the migration of CD4+ T cells and promoted the differentiation of Treg cells. Finally, we found that the transcription factor c-Jun was involved in regulating the expression of IL-8 and affected the osteogenic differentiation of MSCs, thereby mediating the migration and differentiation of CD4+ T cells. Conclusion This study demonstrated that MSC osteogenic differentiation promoted c-Jun-dependent secretion of IL-8 and mediated the migration and differentiation of CD4+ T cells. These results provide a further understanding of the crosstalk between bone and the immune system and reveal information about the relationship between osteogenesis and inflammation in the field of osteoimmunology. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02735-0.
Collapse
|
40
|
Qiao D, Chen R, Li L, Zhu F, Zhang Y, Yan F. Accelerated Alveolar Bone Loss in a Mouse Model of Inflammatory Bowel Disease and its Relationship with Intestinal Inflammation. J Periodontol 2022; 93:1566-1577. [PMID: 35092308 DOI: 10.1002/jper.21-0374] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 01/11/2022] [Accepted: 01/16/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Dan Qiao
- Nanjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu People's Republic of China
| | - Rixin Chen
- Nanjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu People's Republic of China
| | - Lingjun Li
- Nanjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu People's Republic of China
| | - Feng Zhu
- Department of General Surgery Jinling Hospital, Medical School of Nanjing University Nanjing Jiangsu People's Republic of China
| | - Yangheng Zhang
- Nanjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu People's Republic of China
| | - Fuhua Yan
- Nanjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu People's Republic of China
| |
Collapse
|
41
|
Effector memory CD4 +T cells in mesenteric lymph nodes mediate bone loss in food-allergic enteropathy model mice, creating IL-4 dominance. Mucosal Immunol 2021; 14:1335-1346. [PMID: 34326478 DOI: 10.1038/s41385-021-00434-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 06/07/2021] [Accepted: 07/06/2021] [Indexed: 02/04/2023]
Abstract
Intestinal inflammation can be accompanied by osteoporosis, but their relationship, mediated by immune responses, remains unclear. Here, we investigated a non-IgE-mediated food-allergic enteropathy model of ovalbumin (OVA) 23-3 mice expressing OVA-specific T-cell-receptor transgenes. Mesenteric lymph nodes (MLNs) and their pathogenic CD4+T cells were important to enteropathy occurrence and exacerbation when the mice were fed an egg-white (EW) diet. EW-fed OVA23-3 mice also developed bone loss and increased CD44hiCD62LloCD4+T cells in the MLNs and bone marrow (BM); these changes were attenuated by MLN, but not spleen, resection. We fed an EW diet to F1 cross offspring from OVA23-3 mice and a mouse line expressing the photoconvertible protein KikGR to track MLN CD4+T cells. Photoconverted MLN CD44hiCD62LloCD4+T cells migrated predominantly to the BM; pit formation assay proved their ability to promote bone damage via osteoclasts. Significantly greater expression of IL-4 mRNA in MLN CD44hiCD62LloCD4+T cells and bone was observed in EW-fed OVA23-3 mice. Anti-IL-4 monoclonal antibody injection canceled bone loss in the primary inflammation phase in EW-fed mice, but less so in the chronic phase. This novel report shows the specific inflammatory relationship, via Th2-dominant-OVA-specific T cells and IL-4 production, between MLNs and bone, a distant organ, in food-allergic enteropathy.
Collapse
|
42
|
Zhou A, Wu B, Yu H, Tang Y, Liu J, Jia Y, Yang X, Xiang L. Current Understanding of Osteoimmunology in Certain Osteoimmune Diseases. Front Cell Dev Biol 2021; 9:698068. [PMID: 34485284 PMCID: PMC8416088 DOI: 10.3389/fcell.2021.698068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/15/2021] [Indexed: 02/05/2023] Open
Abstract
The skeletal system and immune system seem to be two independent systems. However, there in fact are extensive and multiple crosstalk between them. The concept of osteoimmunology was created to describe those interdisciplinary events, but it has been constantly updated over time. In this review, we summarize the interactions between the skeletal and immune systems in the co-development of the two systems and the progress of certain typical bone abnormalities and bone regeneration on the cellular and molecular levels according to the mainstream novel study. At the end of the review, we also highlighted the possibility of extending the research scope of osteoimmunology to other systemic diseases. In conclusion, we propose that osteoimmunology is a promising perspective to uncover the mechanism of related diseases; meanwhile, a study from the point of view of osteoimmunology may also provide innovative ideas and resolutions to achieve the balance of internal homeostasis.
Collapse
Affiliation(s)
- Anqi Zhou
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bingfeng Wu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hui Yu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yufei Tang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiayi Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yinan Jia
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaoyu Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lin Xiang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
43
|
Halper J, Madel MB, Blin-Wakkach C. Differentiation and Phenotyping of Murine Osteoclasts from Bone Marrow Progenitors, Monocytes, and Dendritic Cells. Methods Mol Biol 2021; 2308:21-34. [PMID: 34057711 DOI: 10.1007/978-1-0716-1425-9_2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
Bone physiology is dictated by various players, including osteoclasts (OCLs) as bone resorbing cells, osteoblasts (capable of bone formation), osteocytes, or mesenchymal stem cells, to mention the most important players. All these cells are in tight communication with each other and influence the constantly occurring process of bone remodeling to meet changing requirements on the skeletal system. In order to understand these interplays, one must investigate isolated functions of the various cell types. However, OCL research displays a special drawback: due to their giant size, low abundance, and tight attachment on the bone surface, ex vivo isolation of sufficient amounts of mature OCLs is limited or not conceivable in most species including mice. Moreover, OCLs can be obtained from different progenitors in vivo as well as in vitro. Thus, in vitro differentiation of OCLs from various progenitor cells remains essential in the analysis of OCL biology, underlining the importance of reliable gold standard protocols to be applied throughout OCL research. This chapter will deal with in vitro differentiation of OCLs from murine bone marrow cells, as well as isolated monocytes and dendritic cells that have already been validated in numerous studies.
Collapse
Affiliation(s)
- Julia Halper
- Université Côte d'Azur, CNRS, UMR7370, LP2M, Nice, France
| | - Maria-Bernadette Madel
- Université Côte d'Azur, CNRS, UMR7370, LP2M, Nice, France
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, USA
| | | |
Collapse
|
44
|
Frech M, Schuster G, Andes FT, Schett G, Zaiss MM, Sarter K. RANKL-Induced Btn2a2 - A T Cell Immunomodulatory Molecule - During Osteoclast Differentiation Fine-Tunes Bone Resorption. Front Endocrinol (Lausanne) 2021; 12:685060. [PMID: 34421818 PMCID: PMC8371446 DOI: 10.3389/fendo.2021.685060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/05/2021] [Indexed: 12/11/2022] Open
Abstract
Butyrophilins, which are members of the extended B7 family of immunoregulators structurally related to the B7 family, have diverse functions on immune cells as co-stimulatory and co-inhibitory molecules. Despite recent advances in the understanding on butyrophilins' role on adaptive immune cells during infectious or autoimmune diseases, nothing is known about their role in bone homeostasis. Here, we analyzed the role of one specific butyrophilin, namely Btn2a2, as we have recently shown that Btn2a2 is expressed on the monocyte/macrophage lineage that also gives rise to bone degrading osteoclasts. We found that expression of Btn2a2 on monocytes and pre-osteoclasts is upregulated by the receptor activator of nuclear factor κ-B ligand (RANKL), an essential protein required for osteoclast formation. Interestingly, in Btn2a2-deficient osteoclasts, typical osteoclast marker genes (Nfatc1, cathepsin K, TRAP, and RANK) were downregulated following RANKL stimulation. In vitro osteoclast assays resulted in decreased TRAP positive osteoclast numbers in Btn2a2-deficient cells. However, Btn2a2-deficient osteoclasts revealed abnormal fusion processes shown by their increased size. In vivo steady state µCT and histological analysis of bone architecture in complete Btn2a2-deficient mice showed differences in bone parameters further highlighting the fine-tuning effect of BTN2a2. Moreover, in rheumatoid arthritis patients and experimental arthritis, we detected significantly decreased serum levels of the secreted soluble Btn2a2 protein. Taken together, we identified the involvement of the immunomodulatory molecule Btn2a2 in osteoclast differentiation with potential future implications in basic and translational osteoimmunology.
Collapse
Affiliation(s)
- Michael Frech
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Gregor Schuster
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Fabian T. Andes
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Mario M. Zaiss
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Kerstin Sarter
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
45
|
Zhang YW, Li YJ, Lu PP, Dai GC, Chen XX, Rui YF. The modulatory effect and implication of gut microbiota on osteoporosis: from the perspective of "brain-gut-bone" axis. Food Funct 2021; 12:5703-5718. [PMID: 34048514 DOI: 10.1039/d0fo03468a] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Osteoporosis (OP) is a kind of systemic metabolic disease characterized by decreased bone mass and destruction of the bone microstructure. In recent years, it has become an expected research trend to explore the cross-linking relationship in the pathogenesis process of OP so as to develop reasonable and effective intervention strategies. With the further development of intestinal microbiology and the profound exploration of the gut microbiota (GM), it has been further revealed that the "brain-gut" axis may be a potential target for the bone, thereby affecting the occurrence and progression of OP. Hence, based on the concept of "brain-gut-bone" axis, we look forward to deeply discussing and summarizing the cross-linking relationship of OP in the next three parts, including the "brain-bone" connection, "gut-bone" connection, and "brain-gut" connection, so as to provide an emerging thought for the prevention strategies and mechanism researches of OP.
Collapse
Affiliation(s)
- Yuan-Wei Zhang
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China. and Multidisciplinary Team (MDT) for Geriatric Hip Fracture Management, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China and School of Medicine, Southeast University, Nanjing, Jiangsu, China and Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, Jiangsu, China and Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Ying-Juan Li
- Multidisciplinary Team (MDT) for Geriatric Hip Fracture Management, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China and Department of Geriatrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Pan-Pan Lu
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China. and Multidisciplinary Team (MDT) for Geriatric Hip Fracture Management, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China and School of Medicine, Southeast University, Nanjing, Jiangsu, China and Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, Jiangsu, China and Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Guang-Chun Dai
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China. and Multidisciplinary Team (MDT) for Geriatric Hip Fracture Management, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China and School of Medicine, Southeast University, Nanjing, Jiangsu, China and Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, Jiangsu, China and Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Xiang-Xu Chen
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China. and Multidisciplinary Team (MDT) for Geriatric Hip Fracture Management, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China and School of Medicine, Southeast University, Nanjing, Jiangsu, China and Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, Jiangsu, China and Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Yun-Feng Rui
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China. and Multidisciplinary Team (MDT) for Geriatric Hip Fracture Management, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China and School of Medicine, Southeast University, Nanjing, Jiangsu, China and Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, Jiangsu, China and Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| |
Collapse
|
46
|
Wang B, Dong Y, Tian Z, Chen Y, Dong S. The role of dendritic cells derived osteoclasts in bone destruction diseases. Genes Dis 2021; 8:401-411. [PMID: 34179305 PMCID: PMC8209356 DOI: 10.1016/j.gendis.2020.03.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/14/2020] [Accepted: 03/20/2020] [Indexed: 02/06/2023] Open
Abstract
The bone is previously considered as a dominant organ involved in the processes of locomotion. However, in the past two decades, a large number of studies have suggested that the skeletal system closely coordinated with the immune system so as to result in the emerging area of 'osteoimmunology'. In the evolution of many kinds of bone destruction-related diseases, osteoclasts could differentiate from dendritic cells, which contributed to increased expression of osteoclast-related membrane receptors and relatively higher activity of bone destruction, inducing severe bone destruction under inflammatory conditions. Numerous factors could influence the interaction between osteoclasts and dendritic cells, contributing to the pathogenesis of several bone diseases in the context of inflammation, including both immunocytes and a large number of cytokines. In addition, the products of osteoclasts released from bone destruction area serve as important signals for the differentiation and activation of immature dendritic cells. Therefore, the border between the dendritic cell-related immune response and osteoclast-related bone destruction has gradually unravelled. Dendritic cells and osteoclasts cooperate with each other to mediate bone destruction and bone remodelling under inflammatory conditions. In this review, we will pay attention to the interactions between dendritic cells and osteoclasts in physiological and pathological conditions to further understand the skeletal system and identify potential new therapeutic targets for the future by summarizing their significant roles and molecular mechanisms in bone destruction.
Collapse
Affiliation(s)
- Bo Wang
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
- Battalion of Basic Medical Sciences, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Yutong Dong
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
- Battalion of Basic Medical Sciences, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Zhansong Tian
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Yueqi Chen
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Shiwu Dong
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
- State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| |
Collapse
|
47
|
Rajput S, Mehta P, Mittal M, Rajender S, Chattopadhyay N. Human Relevance of Preclinical Studies on the Skeletal Impact of Inflammatory Bowel Disease: A Systematic Review and Meta-Analysis. Calcif Tissue Int 2021; 108:708-724. [PMID: 33558960 DOI: 10.1007/s00223-021-00808-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 01/12/2021] [Indexed: 01/02/2023]
Abstract
Inflammatory bowel disease (IBD) is a relapsing chronic idiopathic inflammatory condition. The increased risks of fractures in the spine and decreased BMD at all weight-bearing skeletal sites have been reported in IBD patients. The understanding of the mechanisms of IBD-induced bone loss is far from complete. Appropriate animal models are a prerequisite for studying IBD-induced bone loss, which prompted us to undertake quantitative meta-analyses by pooling data from the available IBD models that assessed various bone parameters. Sufficient data for meta-analysis are obtained from chemically- but not genetically induced models. Among the chemically induced models, only the effects of dextran sulfate sodium (DSS) and 2,4,6-trinitrobenzene sulfonic acid (TNBS) on bone parameters have been reported. Meta-analysis showed that both DSS (Hedge's g = 2.124, p = 0.001) and TNBS (Hedge's g = 6.292, p = 0.000) increased inflammatory disease severity. In pooled analysis, bone volumes in femur (Hedge's g = - 3.42, p = 0.000) and tibia (Hedge's g = - 2.49, p = 0.000) showed significant losses upon DSS administration. Similarly, bone formation rate was significantly reduced upon IBD induction (Hedge's g = - 3.495, p = 0.006). Besides, cortical thickness was reduced and trabecular microstructure deteriorated by IBD induction. Insufficient data precluded us from determining the effect of IBD on bone strength and calciotropic hormones, as well as the impact of proinflammatory cytokines on bone turnover. This meta-analysis showed that IBD induction in rodents causes significant bone loss. Impaired osteoblast function appears to be the cause of this impact.
Collapse
Affiliation(s)
- Swati Rajput
- Division of Endocrinology and Centre for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Poonam Mehta
- Division of Endocrinology and Centre for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Monika Mittal
- Division of Endocrinology and Centre for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Singh Rajender
- Division of Endocrinology and Centre for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| | - Naibedya Chattopadhyay
- Division of Endocrinology and Centre for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
48
|
Fischer V, Haffner-Luntzer M. Interaction between bone and immune cells: Implications for postmenopausal osteoporosis. Semin Cell Dev Biol 2021; 123:14-21. [PMID: 34024716 DOI: 10.1016/j.semcdb.2021.05.014] [Citation(s) in RCA: 329] [Impact Index Per Article: 82.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/11/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022]
Abstract
Postmenopausal osteoporosis is a systemic disease characterized by the loss of bone mass and increased bone fracture risk largely resulting from significantly reduced levels of the hormone estrogen after menopause. Besides the direct negative effects of estrogen-deficiency on bone, indirect effects of altered immune status in postmenopausal women might contribute to ongoing bone destruction, as postmenopausal women often display a chronic low-grade inflammatory phenotype with altered cytokine expression and immune cell profile. In this context, it was previously shown that various immune cells interact with osteoblasts and osteoclasts either via direct cell-cell contact, or more likely via paracrine mechanisms. For example, specific subtypes of T lymphocytes express TNFα, which was shown to increase osteoblast apoptosis and to indirectly stimulate osteoclastogenesis via B cell-produced receptor-activator of NF-κB ligand (RANKL), thereby triggering bone loss during postmenopausal osteoporosis. Th17 cells release interleukin-17 (IL-17), which directs mesenchymal stem cell differentiation towards the osteogenic lineage, but also indirectly increases osteoclast differentiation. B lymphocytes are a major regulator of osteoclast formation via granulocyte colony-stimulating factor secretion and the RANKL/osteoprotegerin system under estrogen-deficient conditions. Macrophages might act differently on bone cells dependent on their polarization profile and their secreted paracrine factors, which might have implications for the development of postmenopausal osteoporosis, because macrophage polarization is altered during disease progression. Likewise, neutrophils play an important role during bone homeostasis, but their over-activation under estrogen-deficient conditions contributes to osteoblast apoptosis via the release of reactive oxygen species and increased osteoclastogenesis via RANKL signaling. Furthermore, mast cells might be involved in the development of postmenopausal osteoporosis, because they store high levels of osteoclastic mediators, including IL-6 and RANKL, in their granules and their numbers are greatly increased in osteoporotic bone. Additionally, bone fracture healing is altered under estrogen-deficient conditions with the increased presence of pro-inflammatory cytokines, including IL-6 and Midkine, which might contribute to healing disturbances. Consequently, in addition to the direct negative influence of estrogen-deficiency on bone, immune cell alterations contribute to the pathogenesis of postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Verena Fischer
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Helmholtzstraße 14, 89081 Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Helmholtzstraße 14, 89081 Ulm, Germany.
| |
Collapse
|
49
|
Yu J, Cao G, Yuan S, Luo C, Yu J, Cai M. Probiotic supplements and bone health in postmenopausal women: a meta-analysis of randomised controlled trials. BMJ Open 2021; 11:e041393. [PMID: 33653743 PMCID: PMC7929795 DOI: 10.1136/bmjopen-2020-041393] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 02/10/2021] [Accepted: 02/17/2021] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Osteoporosis is a common disease in postmenopausal women. Several studies have analysed the associations between dietary supplementation with probiotics and bone health in postmenopausal women, but the results are still controversial. We conducted this meta-analysis to assess the effects of probiotics supplement on bone mineral density (BMD) and bone turnover markers for postmenopausal women. DESIGN Systematic review and meta-analysis. METHODS We systematically searched PubMed, EMBASE and the Cochrane Library from their inception to November 2020 for randomised controlled trials (RCTs) assessing probiotic supplements and osteoporosis in postmenopausal women. Study-specific risk estimates were combined using random-effect models. RESULTS Five RCTs (n=497) were included. Probiotic supplements were associated with a significantly higher BMD in the lumbar spine (standardised mean difference, SMD=0.27, 95% CI 0.09 to 0.44) than in control. There was no difference between probiotic supplements and BMD in hips (SMD=0.22, 95% CI -0.07 to 0.52). Collagen type 1 cross-linked C-telopeptide levels in the treatment groups were significantly lower than those of the placebo group (SMD=-0.34, 95% CI -0.60 to -0.09). In subgroup meta-analysis, levels of bone-specific alkaline phosphatase, osteoprotegerin, osteocalcin and tumour necrosis factor did not differ between the probiotic and placebo groups. CONCLUSIONS We conclude cautiously that supplementation with probiotics could increase lumbar BMD. More RCTs are recommended to validate or update these results.
Collapse
Affiliation(s)
- Jiawei Yu
- Department of Orthopedic Surgery, Zhuji Affiliated Hospital of Wenzhou Medical University, Shaoxing, China
| | - Gaoyang Cao
- Department of Colorectal Surgery, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Hangzhou, China
| | - Shuohui Yuan
- Department of Orthopedic Surgery, Zhuji Affiliated Hospital of Wenzhou Medical University, Shaoxing, China
| | - Cong Luo
- Department of Orthopedic Surgery, Zhuji Affiliated Hospital of Wenzhou Medical University, Shaoxing, China
| | - Jiafeng Yu
- Department of Orthopedic Surgery, Zhuji Affiliated Hospital of Wenzhou Medical University, Shaoxing, China
| | - Ming Cai
- Department of Orthopedic Surgery, Zhuji Affiliated Hospital of Wenzhou Medical University, Shaoxing, China
| |
Collapse
|
50
|
Sahbani K, Shultz LC, Cardozo CP, Bauman WA, Tawfeek HA. Absence of αβ T cells accelerates disuse bone loss in male mice after spinal cord injury. Ann N Y Acad Sci 2021; 1487:43-55. [PMID: 33107070 DOI: 10.1111/nyas.14518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/07/2020] [Accepted: 10/07/2020] [Indexed: 11/30/2022]
Abstract
Whether T cells promote bone loss following immobilization after spinal cord injury (SCI) remains undetermined. Therefore, wild-type (WT) and T cell-deficient (Tcrb-/- ) male mice underwent sham or contusion SCI to cause hindlimb paralysis. Femurs were isolated and distal and midshaft regions were evaluated by microcomputed tomography scanning. Bone marrow (BM) levels of bone turnover markers, as well as receptor activator of nuclear factor-kappa B ligand (RANKL) and osteoprotegerin (OPG), were measured by ELISA. At 2 weeks post-SCI, immobilization resulted in marked reduction in trabecular fractional bone volume (55%), thickness (40%), connectivity, and cortical thickness only in the Tcrb-/- animals (interaction with P < 0.05). BM analysis revealed lower bone formation (procollagen type 1 intact N-terminal propeptide), higher bone resorption (tartrate-resistant acid phosphatase-5b), and a higher RANKL/OPG ratio in the Tcrb-/- SCI animals. At 5 weeks post-SCI, while both WT and Tcrb-/- paralyzed animals showed deterioration of all indices of bone structure, they were more severe in Tcrb-/- animals. In summary, unlike other skeletal disorders, loss of αβ T cells compromises, rather than preserves, skeletal integrity under conditions of immobilization.
Collapse
MESH Headings
- Animals
- Bone Density/genetics
- Bone Density/immunology
- Bone Diseases, Metabolic/genetics
- Bone Diseases, Metabolic/immunology
- Bone Diseases, Metabolic/metabolism
- Bone Diseases, Metabolic/pathology
- Bone Resorption/genetics
- Bone Resorption/immunology
- Bone Resorption/metabolism
- Cell Count
- Genes, T-Cell Receptor beta/genetics
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, Antigen, T-Cell, alpha-beta/deficiency
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Spinal Cord Injuries/complications
- Spinal Cord Injuries/genetics
- Spinal Cord Injuries/immunology
- Spinal Cord Injuries/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/pathology
- X-Ray Microtomography
Collapse
Affiliation(s)
- Karim Sahbani
- National Center for the Medical Consequences of Spinal Cord Injury, James J Peters Veterans Affairs Medical Center, Bronx, New York
- Bronx Veterans Medical Research Foundation Inc., Bronx, New York
| | - Laura C Shultz
- Veterinary Medical Unit, James J Peters Veterans Affairs Medical Center, Bronx, New York
| | - Christopher P Cardozo
- National Center for the Medical Consequences of Spinal Cord Injury, James J Peters Veterans Affairs Medical Center, Bronx, New York
- Bronx Veterans Medical Research Foundation Inc., Bronx, New York
- Department of Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Rehabilitation Medicine and Human Performance, The Icahn School of Medicine at Mount Sinai, New York, New York
| | - William A Bauman
- National Center for the Medical Consequences of Spinal Cord Injury, James J Peters Veterans Affairs Medical Center, Bronx, New York
- Bronx Veterans Medical Research Foundation Inc., Bronx, New York
- Department of Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York
| | - Hesham A Tawfeek
- National Center for the Medical Consequences of Spinal Cord Injury, James J Peters Veterans Affairs Medical Center, Bronx, New York
- Bronx Veterans Medical Research Foundation Inc., Bronx, New York
- Department of Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|