1
|
Ranjan G, Ranjan S, Sunita P, Pattanayak SP. Thiazolidinedione derivatives in cancer therapy: exploring novel mechanisms, therapeutic potentials, and future horizons in oncology. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4705-4725. [PMID: 39621087 DOI: 10.1007/s00210-024-03661-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/20/2024] [Indexed: 04/11/2025]
Abstract
Thiazolidinedione derivatives have shown significant potential as targeted cancer therapies by leveraging their various mechanisms of action. These include suppressing cell proliferation, triggering apoptosis, and influencing signaling pathways associated with tumor development. Their multifaceted effects make them promising candidates for advancing cancer treatment strategies. They have shown significant promise as anti-cancer agents, particularly through their ability to inhibit lipogenesis pathways and apoptosis essential for cancer cell survival and proliferation. This review comprehensively examines the anti-cancer potential of thiazolidinedione derivatives by targeting key aspects of lipid metabolism, apoptosis, and various mechanistic pathways. This review provides an in-depth examination of the anti-cancer potential of TZD derivatives, focusing on their mechanisms of action, therapeutic applications, and future directions in oncology. The anti-tumor effects of TZDs primarily involve the stimulation of peroxisome proliferator-activated receptor gamma (PPAR-γ), suppressing cell proliferation, induction of apoptosis, and inhibition of angiogenesis. Moreover, recent evidence highlights their ability to modulate non-PPAR-γ pathways, such as PI3K/Akt, NF-κB, and MAPK, further expanding their role in overcoming drug resistance and enhancing therapeutic outcomes. This review explores the preclinical (in vitro and in vivo) and clinical research investigating TZD derivatives efficacy in various cancer types. The insights underscore the significance of targeting lipogenesis as a novel anti-cancer strategy, positioning thiazolidinedione derivatives as potent candidates for future cancer therapeutics. As the oncology landscape evolves, TZD derivatives (rosiglitazone, pioglitazone, inolitazone, troglitazone, and 2,4-thiazolidinedione derivatives) represent a promising class of agents with the potential to contribute meaningfully to cancer treatment. By integrating existing knowledge with recent advancements, this study provides valuable insights into the role of thiazolidinedione derivatives in cancer treatment, paving the way for further research and clinical applications.
Collapse
Affiliation(s)
- Gaurav Ranjan
- Department of Pharmacy, School of Health Sciences, Central University of South Bihar, Gaya, 824236, India
| | - Shashi Ranjan
- Department of Pharmacy, School of Health Sciences, Central University of South Bihar, Gaya, 824236, India
| | - Priyashree Sunita
- Department of Surgery, Case Comprehensive Cancer Centre, Case Western Reserve University, Wolstein Research Building 2103 Cornell Rd, Cleveland, OH, 44106, USA
| | - Shakti Prasad Pattanayak
- Department of Pharmacy, School of Health Sciences, Central University of South Bihar, Gaya, 824236, India.
- Department of Biochemistry, School of Medicine, Case Western Reserve University, Woods Building, W437, 2109 Adelbert Road, Cleaveland, OH, 44106, USA.
| |
Collapse
|
2
|
Xue C, Chu Q, Shi Q, Zeng Y, Lu J, Li L. Wnt signaling pathways in biology and disease: mechanisms and therapeutic advances. Signal Transduct Target Ther 2025; 10:106. [PMID: 40180907 PMCID: PMC11968978 DOI: 10.1038/s41392-025-02142-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/13/2024] [Accepted: 12/29/2024] [Indexed: 04/05/2025] Open
Abstract
The Wnt signaling pathway is critically involved in orchestrating cellular functions such as proliferation, migration, survival, and cell fate determination during development. Given its pivotal role in cellular communication, aberrant Wnt signaling has been extensively linked to the pathogenesis of various diseases. This review offers an in-depth analysis of the Wnt pathway, detailing its signal transduction mechanisms and principal components. Furthermore, the complex network of interactions between Wnt cascades and other key signaling pathways, such as Notch, Hedgehog, TGF-β, FGF, and NF-κB, is explored. Genetic mutations affecting the Wnt pathway play a pivotal role in disease progression, with particular emphasis on Wnt signaling's involvement in cancer stem cell biology and the tumor microenvironment. Additionally, this review underscores the diverse mechanisms through which Wnt signaling contributes to diseases such as cardiovascular conditions, neurodegenerative disorders, metabolic syndromes, autoimmune diseases, and cancer. Finally, a comprehensive overview of the therapeutic progress targeting Wnt signaling was given, and the latest progress in disease treatment targeting key components of the Wnt signaling pathway was summarized in detail, including Wnt ligands/receptors, β-catenin destruction complexes, and β-catenin/TCF transcription complexes. The development of small molecule inhibitors, monoclonal antibodies, and combination therapy strategies was emphasized, while the current potential therapeutic challenges were summarized. This aims to enhance the current understanding of this key pathway.
Collapse
Affiliation(s)
- Chen Xue
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingmiao Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yifan Zeng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
3
|
Jiang H, Xi Y, Jiang Q, Dai W, Qin X, Zhang J, Jiang Z, Yang G, Chen Q. LRP5 Down-Regulation Exacerbates Inflammation and Alveolar Bone Loss in Periodontitis by Inhibiting PI3K/c-FOS Signalling. J Clin Periodontol 2025; 52:637-650. [PMID: 39837316 DOI: 10.1111/jcpe.14112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/11/2024] [Accepted: 12/15/2024] [Indexed: 01/23/2025]
Abstract
AIM To investigate the involvement of low-density lipoprotein receptor-related protein 5 (LRP5) in inflammation and alveolar bone loss in periodontitis. MATERIALS AND METHODS Gingival tissues were obtained from 10 periodontitis patients and 10 healthy individuals. Wild-type (WT) and osteoblast-specific Lrp5 conditional knock-out C57BL/6 (LRP5fl/fl;Oc-Cre) mice were used to establish a ligature-induced mouse model of periodontitis. Human periodontal ligament stem cells (hPDLSCs) were isolated and used to further verify the mechanism through which LRP5 mediates periodontitis in vitro. Micro-computed tomography, haematoxylin and eosin staining, immunohistochemistry, quantitative reverse transcription PCR, western blotting, enzyme-linked immunosorbent assay and RNA sequencing were performed to explore the role of LRP5 in periodontitis and the underlying mechanism. RESULTS LRP5 expression was down-regulated in human/mouse periodontal tissues compared to that in healthy controls. Compared to those in wild-type mice, the periodontal tissues of LRP5fl/fl;Oc-Cre mice had increased alveolar bone loss, higher proinflammatory cytokine levels, and lower osteogenesis-related factor expression. LRP5 expression was down-regulated in hPDLSCs after lipopolysaccharide treatment in vitro. LRP5 knockdown increased proinflammatory cytokine production and inhibited osteoblastogenesis by inhibiting PI3K/c-FOS signalling. CONCLUSION LRP5 down-regulation exacerbates inflammation and alveolar bone loss in periodontitis by inhibiting PI3K/c-FOS signalling, suggesting LRP5 as a potential therapeutic target for periodontitis.
Collapse
Affiliation(s)
- Hui Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Yue Xi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Qifeng Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Wei Dai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Xiaoru Qin
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Jing Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Zhiwei Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Qianming Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| |
Collapse
|
4
|
Poudel SB, Kim MH, Bhattarai G, So HS, Kook SH, Lee JC. n-acetyl-l-cysteine stimulates bone healing by recovering the age-associated degenerative complications relative to osteoblastic Wntless ablation. Biomed Pharmacother 2025; 182:117761. [PMID: 39700869 DOI: 10.1016/j.biopha.2024.117761] [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: 09/16/2024] [Revised: 12/10/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024] Open
Abstract
Dysregulated Wnt signaling causes age-related characteristics such as oxidative stress, stem cell senescence, and abnormal bone homeostasis. Here we explored whether supplemental n-acetyl-l-cysteine (NAC) recovers the age-associated complications relative to osteoblastic Wntless (Wls) ablation and examined the possible mechanisms therein. For this work, we administered Col2.3-Cre;Wlsfl/fl mutant and littermate control (Wlsfl/fl) mice (14 weeks of age) with NAC (40 mM)-supplemented or NAC-free water for four weeks. A proportion of these mice received non-critical-sized femoral defects at 16 weeks of age. Blood, bone, and bone marrow (BM) samples were collected and adjusted for in vivo, ex vivo, and in vitro analyses. Osteoblastic Wls deletion delayed bone mass accrual and the healing of bone defects, stimulated osteoclastic activation and inflammatory factor expression, and decreased antioxidant enzyme activity in the BM. Osteoblastic Wls deletion also promoted oxidative stress, apoptosis, and senescence in BM stromal cells (BMSCs) and decreased BMSC' multipotencies. Supplementation of Wlsfl/fl mice with NAC enhanced bone mass accrual and regenerative bone healing via a Wnt signal-associated osteogenic activation. However, supplemental NAC induced new bone formation in the mutant mice by inhibiting the age-related complications of BM/BMSCs, as well as by restoring endogenous antioxidant system without any alterations in Wnt ligand secretion, hematopoiesis, and expression of osteogenic and growth factors. This study indicates that supplemental NAC protects mice against Wnt deficiency-mediated and age-associated degenerative complications. Overall, this study highlights the therapeutic potency of NAC for restoring the antioxidant system, stem cell function, and regenerative bone homeostasis in osteoblastic Wls-dispensable manner.
Collapse
Affiliation(s)
- Sher Bahadur Poudel
- Department of Basic Science & Craniofacial Biology, College of Dentistry, New York University, New York, NY 10010, USA
| | - Min-Hye Kim
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju 54896, South Korea
| | - Govinda Bhattarai
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences, School of Dentistry, Jeonbuk National University, Jeonju 54896, South Korea
| | - Han-Sol So
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju 54896, South Korea
| | - Sung-Ho Kook
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju 54896, South Korea; Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences, School of Dentistry, Jeonbuk National University, Jeonju 54896, South Korea.
| | - Jeong-Chae Lee
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju 54896, South Korea; Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences, School of Dentistry, Jeonbuk National University, Jeonju 54896, South Korea.
| |
Collapse
|
5
|
Lu X, Zhao Y, Peng X, Lu C, Wu Z, Xu H, Qin Y, Xu Y, Wang Q, Hao Y, Geng D. Comprehensive Overview of Interface Strategies in Implant Osseointegration. ADVANCED FUNCTIONAL MATERIALS 2024. [DOI: 10.1002/adfm.202418849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Indexed: 01/05/2025]
Abstract
AbstractWith the improvement of implant design and the expansion of application scenarios, orthopedic implants have become a common surgical option for treating fractures and end‐stage osteoarthritis. Their common goal is rapidly forming and long‐term stable osseointegration. However, this fixation effect is limited by implant surface characteristics and peri‐implant bone tissue activity. Therefore, this review summarizes the strategies of interface engineering (osteogenic peptides, growth factors, and metal ions) and treatment methods (porous nanotubes, hydrogel embedding, and other load‐release systems) through research on its biological mechanism, paving the way to achieve the adaptation of both and coordination between different strategies. With the transition of the osseointegration stage, interface engineering strategies have demonstrated varying therapeutic effects. Especially, the activity of osteoblasts runs almost through the entire process of osseointegration, and their physiological activities play a dominant role in bone formation. Furthermore, diseases impacting bone metabolism exacerbate the difficulty of achieving osseointegration. This review aims to assist future research on osseointegration engineering strategies to improve implant‐bone fixation, promote fracture healing, and enhance post‐implantation recovery.
Collapse
Affiliation(s)
- Xiaoheng Lu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yuhu Zhao
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Xiaole Peng
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University 1 Youyi Street Chongqing 400016 China
| | - Chengyao Lu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Zebin Wu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Hao Xu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yi Qin
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yaozeng Xu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Qing Wang
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yuefeng Hao
- Orthopedics and Sports Medicine Center The Affiliated Suzhou Hospital of Nanjing Medical University 242 Guangji Street Suzhou Jiangsu 215006 China
| | - Dechun Geng
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| |
Collapse
|
6
|
Shi V, Morgan EF. Estrogen and estrogen receptors mediate the mechanobiology of bone disease and repair. Bone 2024; 188:117220. [PMID: 39106937 PMCID: PMC11392539 DOI: 10.1016/j.bone.2024.117220] [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: 06/08/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/09/2024]
Abstract
It is well understood that the balance of bone formation and resorption is dependent on both mechanical and biochemical factors. In addition to cell-secreted cytokines and growth factors, sex hormones like estrogen are critical to maintaining bone health. Although the direct osteoprotective function of estrogen and estrogen receptors (ERs) has been reported extensively, evidence that estrogen signaling also has a role in mediating the effects of mechanical loading on maintenance of bone mass and healing of bone injuries has more recently emerged. Recent studies have underscored the role of estrogen and ERs in many pathways of bone mechanosensation and mechanotransduction. Estrogen and ERs have been shown to augment integrin-based mechanotransduction as well as canonical Wnt/b-catenin, RhoA/ROCK, and YAP/TAZ pathways. Estrogen and ERs also influence the mechanosensitivity of not only osteocytes but also osteoblasts, osteoclasts, and marrow stromal cells. The current review will highlight these roles of estrogen and ERs in cellular mechanisms underlying bone mechanobiology and discuss their implications for management of osteoporosis and bone fractures. A greater understanding of the mechanisms behind interactions between estrogen and mechanical loading may be crucial to addressing the shortcomings of current hormonal and pharmaceutical therapies. A combined therapy approach including high-impact exercise therapy may mitigate adverse side effects and allow an effective long-term solution for the prevention, treatment, and management of bone fragility in at-risk populations. Furthermore, future implications to novel local delivery mechanisms of hormonal therapy for osteoporosis treatment, as well as the effects on bone health of applications of sex hormone therapy outside of bone disease, will be discussed.
Collapse
Affiliation(s)
- Vivian Shi
- Boston University, Department of Biomedical Engineering, 44 Cummington St, Boston 02215, MA, USA; Center for Multiscale and Translational Mechanobiology, Boston University, 44 Cummington St, Boston 02215, MA, USA
| | - Elise F Morgan
- Boston University, Department of Biomedical Engineering, 44 Cummington St, Boston 02215, MA, USA; Center for Multiscale and Translational Mechanobiology, Boston University, 44 Cummington St, Boston 02215, MA, USA.
| |
Collapse
|
7
|
Rajput S, Kulkarni C, Sharma S, Tomar MS, Khatoon S, Gupta A, Sanyal S, Shrivastava A, Ghosh JK, Chattopadhyay N. Osteogenic effect of an adiponectin-derived short peptide that rebalances bone remodeling: a potential disease-modifying approach for postmenopausal osteoporosis therapy. Arch Pharm Res 2024; 47:736-755. [PMID: 39073743 DOI: 10.1007/s12272-024-01509-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 07/15/2024] [Indexed: 07/30/2024]
Abstract
Adiponectin, an adipokine, regulates metabolic processes, including glucose flux, lipid breakdown, and insulin response, by activating adiponectin receptors 1 and 2 (AdipoR1 and AdipoR2). We have previously shown that globular adiponectin (gAd), an endogenous form of adiponectin, has osteoanabolic and anti-catabolic effects in rodent models of postmenopausal osteopenia. Moreover, we reported the identification of a 13-mer peptide (ADP-1) from the collagen domain of adiponectin, which exhibited significant adiponectin-mimetic properties. Since the clinical development of gAd is constrained by its large size, here, we investigated the osteogenic property of ADP-1. ADP-1 induced osteoblast differentiation more potently than gAd. ADP-1 elicited osteoblast differentiation through two downstream pathways that involved the participation of adiponectin receptors. Firstly, it enhanced mitochondrial biogenesis and OxPhos, leading to osteoblast differentiation. Secondly, it activated the Akt-glycogen synthase kinase 3β-Wnt pathway, thereby increasing osteoblast differentiation. Additionally, ADP-1 suppressed the production of receptor-activator of nuclear kappa B ligand from osteoblasts, enabling it to act as a dual-action molecule (suppressing osteoclast function besides promoting osteoblast function). In osteopenic ovariectomized rats, ADP-1 increased bone mass and strength and improved trabecular integrity by stimulating bone formation and inhibiting bone resorption. Furthermore, by increasing ATP-producing intermediates within the tricarboxylic acid cycle in bones, ADP-1 likely fueled osteoblast function. Given its dual-action mechanism and high potency, ADP-1 offers a unique opportunity to address the unmet clinical need to reset the aberrant bone remodeling in osteoporosis to normalcy, potentially offering a disease-modifying 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, 201002, India
| | - Chirag Kulkarni
- 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, 201002, India
| | - Shivani Sharma
- 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, 201002, India
| | - Manendra Singh Tomar
- Centre for Advance Research, Faculty of Medicine, King George's Medical University, Lucknow, India
| | - Shamima Khatoon
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Arvind Gupta
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Sabyasachi Sanyal
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Ashutosh Shrivastava
- Centre for Advance Research, Faculty of Medicine, King George's Medical University, Lucknow, India
| | - Jimut Kanti Ghosh
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, 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, 201002, India.
| |
Collapse
|
8
|
Vrščaj LA, Marc J, Ostanek B. Towards an enhanced understanding of osteoanabolic effects of PTH-induced microRNAs on osteoblasts using a bioinformatic approach. Front Endocrinol (Lausanne) 2024; 15:1380013. [PMID: 39086902 PMCID: PMC11289717 DOI: 10.3389/fendo.2024.1380013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/03/2024] [Indexed: 08/02/2024] Open
Abstract
In this study, we used a bioinformatic approach to construct a miRNA-target gene interaction network potentially involved in the anabolic effect of parathyroid hormone analogue teriparatide [PTH (1-34)] on osteoblasts. We extracted a dataset of 26 microRNAs (miRNAs) from previously published studies and predicted miRNA target interactions (MTIs) using four software tools: DIANA, miRWalk, miRDB, and TargetScan. By constructing an interactome of PTH-regulated miRNAs and their predicted target genes, we elucidated signaling pathways regulating pluripotency of stem cells, the Hippo signaling pathway, and the TGF-beta signaling pathway as the most significant pathways in the effects of PTH on osteoblasts. Furthermore, we constructed intersection of MTI networks for these three pathways and added validated interactions. There are 8 genes present in all three selected pathways and a set of 18 miRNAs are predicted to target these genes, according to literature data. The most important genes in all three pathways were BMPR1A, BMPR2 and SMAD2 having the most interactions with miRNAs. Among these miRNAs, only miR-146a-5p and miR-346 have validated interactions in these pathways and were shown to be important regulators of these pathways. In addition, we also propose miR-551b-5p and miR-338-5p for further experimental validation, as they have been predicted to target important genes in these pathways but none of their target interactions have yet been verified. Our wet-lab experiment on miRNAs differentially expressed between PTH (1-34) treated and untreated mesenchymal stem cells supports miR-186-5p from the literature obtained data as another prominent miRNA. The meticulous selection of miRNAs outlined will significantly support and guide future research aimed at discovering and understanding the crucial pathways of osteoanabolic PTH-epigenetic effects on osteoblasts. Additionally, they hold potential for the discovery of new PTH target genes, innovative biomarkers for the effectiveness and safety of osteoporosis-affected treatment, as well as novel therapeutic targets.
Collapse
Affiliation(s)
- Lucija Ana Vrščaj
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Janja Marc
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
- Clinical Institute of Clinical Chemistry and Biochemistry, University Clinical Centre Ljubljana, Ljubljana, Slovenia
| | - Barbara Ostanek
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
9
|
Nogueira-Júnior V, Sousa FRN, da S M Rebouças C, Braz HLB, Dos S Morais MLG, Goes P, de C Brito GA, Jorge RJB, Barbosa FG, Mafezoli J, Silva-Filho CJA, de O Capistrano AL, Bezerra MM, de C Leitão RF. Exploring the osteogenic potential of semisynthetic triterpenes from Combretum leprosum: An in vitro and in silico study. In Vitro Cell Dev Biol Anim 2024:10.1007/s11626-024-00928-w. [PMID: 38992216 DOI: 10.1007/s11626-024-00928-w] [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: 11/14/2023] [Accepted: 05/13/2024] [Indexed: 07/13/2024]
Abstract
Combretum leprosum Mart. is a plant of the Combretaceae family, widely distributed in the Northeast region of Brazil, popularly used as an anti-inflammatory agent, and rich in triterpenes. This study evaluated in vitro and in silico potential osteogenic of two semisynthetic triterpenes (CL-P2 and CL-P2A) obtained from the pentacyclic triterpene 3β,6β,16β-trihydroxylup-20(29)-ene (CL-1) isolated from C. leprosum. Assays were carried out in cultured murine osteoblasts (OFCOL II), first investigating the possible toxicity of the compounds on these cells through viability assays (MTT). Cell proliferation and activation were investigated by immunohistochemical evaluation of Ki-67, bone alkaline phosphatase (ALP) activity, and mineralization test by Von Kossa. Molecular docking analysis was performed to predict the binding affinity of CL-P2 and CL-P2A to target proteins involved in the regulation of osteogenesis, including: bone morphogenetic protein 2 (BMP-2), proteins related to Wingless-related integration (WNT) pathway (Low-density lipoprotein receptor-related protein 6-LRP6 and sclerostin-SOST), and receptor activator of nuclear factor (NF)-kB-ligand (RANK-L). Next, Western Blot and immunofluorescence investigated BMP-2, WNT, RANK-L, and OPG protein expressions in cultured murine osteoblasts (OFCOL II). None of the CL-P2 and CL-P2A concentrations were toxic to osteoblasts. Increased cell proliferation, ALP activity, and bone mineralization were observed. Molecular docking assays demonstrated interactions with BMP-2, LRP6, SOST, and RANK-L/OPG. There was observed increased expression of BMP-2, WNT, and RANK-L/OPG proteins. These results suggest, for the first time, the osteogenic potential of CL-P2 and CL-P2A.
Collapse
Affiliation(s)
- Valdo Nogueira-Júnior
- Postgraduate Program in Morphofunctional Sciences, Department of Morphology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Fátima Regina N Sousa
- Department of Morphology, Medical School, Federal University of Piaui, Picos, Piauí, Brazil
| | - Conceição da S M Rebouças
- Postgraduate Program in Morphofunctional Sciences, Department of Morphology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Helyson L B Braz
- Postgraduate Program in Morphofunctional Sciences, Department of Morphology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Maria Luana G Dos S Morais
- Postgraduate Program in Morphofunctional Sciences, Department of Morphology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Paula Goes
- Postgraduate Program in Morphofunctional Sciences, Department of Morphology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Gerly Anne de C Brito
- Postgraduate Program in Morphofunctional Sciences, Department of Morphology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Roberta Jeane B Jorge
- Postgraduate Program in Morphofunctional Sciences, Department of Morphology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
- Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Francisco Geraldo Barbosa
- Department of Organic and Inorganic Chemistry, Science Centre, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Jair Mafezoli
- Department of Organic and Inorganic Chemistry, Science Centre, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Carlos José A Silva-Filho
- Department of Organic and Inorganic Chemistry, Science Centre, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - André Luiz de O Capistrano
- Department of Organic and Inorganic Chemistry, Science Centre, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Mirna M Bezerra
- Postgraduate Program in Health Sciences, School of Medicine, Federal University of Ceará, Sobral, Ceará, Brazil.
| | - Renata F de C Leitão
- Postgraduate Program in Morphofunctional Sciences, Department of Morphology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| |
Collapse
|
10
|
Gou Y, Li H, Sun X, Chen D, Tian F. Parathyroid hormone (1-34) retards the lumbar facet joint degeneration and activates Wnt/β-catenin signaling pathway in ovariectomized rats. J Orthop Surg Res 2024; 19:352. [PMID: 38877549 PMCID: PMC11177467 DOI: 10.1186/s13018-024-04817-6] [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: 12/06/2023] [Accepted: 05/28/2024] [Indexed: 06/16/2024] Open
Abstract
PURPOSE Facet joint degeneration (FJD) is a major cause of low back pain. Parathyroid hormone (PTH) (1-34) is commonly used to treat osteoporosis. However, little is known about its effects on FJD induced by estrogen deficiency. This study aims to investigate the effects of PTH (1-34) on FJD induced by estrogen deficiency and the underlying pathogenesis of the disease. METHODS Forty 3-month-old female Sprague-Dawley rats were randomly divided into four groups: 30 received bilateral ovariectomy (OVX) followed by 12 weeks of treatment with normal saline, PTH (1-34) or 17β-estradiol (E2), and 10 received sham surgery followed by administration of normal saline. Status and Wnt/β-catenin signaling activity in the cartilage and subchondral bone of the L4-L5 FJs and serum biomarkers were analyzed. RESULTS Administration of PTH (1-34) and E2 ameliorated cartilage lesions, and significantly decreased MMP-13 and caspase-3 levels and chondrocyte apoptosis. PTH (1-34) but not E2 significantly increased cartilage thickness, number of chondrocytes, and the expression of aggrecan. PTH (1-34) significantly improved microarchitecture parameters of subchondral bone, increased the expression of collagen I and osteocalcin, and decreased RANKL/OPG ratio. E2 treatment significantly increased the OPG level and decreased the RANKL/OPG ratio in the subchondral bone of ovariectomized rats, but it did not significantly improve the microarchitecture parameters of subchondral bone. Wnt3a and β-catenin expression was significantly reduced in the articular cartilage and subchondral bone in OVX rats, but PTH (1-34) could increase the expression of these proteins. E2 significantly increased the activity of Wnt/β-catenin pathway only in cartilage, but not in subchondral bone. The restoration of Wnt/β-catenin signaling had an obvious correlation with the improvement of some parameters associated with the FJs status. CONCLUSION Wnt/β-catenin signaling may be a potential therapeutic target for FJD induced by estrogen deficiency. PTH (1-34) is effective in treating this disease with better efficacy than 17β-estradiol, and the efficacy may be attributed to its restoration of Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Yu Gou
- Department of Orthopaedic Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Hetong Li
- Department of Orthopaedics, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Xun Sun
- Department of Orthopaedic Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Desheng Chen
- Department of Orthopaedic Surgery, Tianjin Hospital, Tianjin University, Tianjin, China.
| | - Faming Tian
- School of Public Health, North China University of Science and Technology, Tangshan, China.
| |
Collapse
|
11
|
Neri S, Assirelli E, Manzetti M, Viroli G, Ialuna M, Traversari M, Ciaffi J, Ursini F, Faldini C, Ruffilli A. Identification of Epigenetic Biomarkers of Adolescent Idiopathic Scoliosis Progression: A Workflow to Assess Local Gene Expression. Int J Mol Sci 2024; 25:5329. [PMID: 38791368 PMCID: PMC11120692 DOI: 10.3390/ijms25105329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/30/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Adolescent idiopathic scoliosis (AIS) is a three-dimensional structural deformity of the spine that affects 2-3% of adolescents under the age of 16. AIS etiopathogenesis is not completely understood; however, the disease phenotype is correlated to multiple genetic loci and results from genetic-environmental interactions. One of the primary, still unresolved issues is the implementation of reliable diagnostic and prognostic markers. For clinical management improvement, predictors of curve progression are particularly needed. Recently, an epigenetic contribution to AIS development and progression was proposed; nevertheless, validation of data obtained in peripheral tissues and identification of the specific mechanisms and genes under epigenetic control remain limited. In this study, we propose a methodological approach for the identification of epigenetic markers of AIS progression through an original workflow based on the preliminary characterization of local expression of candidate genes in tissues directly involved in the pathology. The feasibility of the proposed methodological protocol has been originally tested here in terms of identification of the putative epigenetic markers of AIS progression, collection of the different tissues, retrieval of an appropriate amount and quality of RNA and DNA, and identification of suitable reference genes.
Collapse
Affiliation(s)
- Simona Neri
- Medicine and Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (E.A.); (J.C.); (F.U.)
| | - Elisa Assirelli
- Medicine and Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (E.A.); (J.C.); (F.U.)
| | - Marco Manzetti
- 1st Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.M.); (G.V.); (M.I.); (M.T.); (C.F.); (A.R.)
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Giovanni Viroli
- 1st Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.M.); (G.V.); (M.I.); (M.T.); (C.F.); (A.R.)
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Marco Ialuna
- 1st Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.M.); (G.V.); (M.I.); (M.T.); (C.F.); (A.R.)
| | - Matteo Traversari
- 1st Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.M.); (G.V.); (M.I.); (M.T.); (C.F.); (A.R.)
| | - Jacopo Ciaffi
- Medicine and Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (E.A.); (J.C.); (F.U.)
| | - Francesco Ursini
- Medicine and Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (E.A.); (J.C.); (F.U.)
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Cesare Faldini
- 1st Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.M.); (G.V.); (M.I.); (M.T.); (C.F.); (A.R.)
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Alberto Ruffilli
- 1st Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.M.); (G.V.); (M.I.); (M.T.); (C.F.); (A.R.)
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
12
|
Xu J, Ze X, Zhao L, Sheng L, Ze Y. Titanium dioxide nanoparticles oral exposure induce osteoblast apoptosis, inhibit osteogenic ability and increase lipogenesis in mouse. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 277:116367. [PMID: 38669870 DOI: 10.1016/j.ecoenv.2024.116367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/26/2024] [Accepted: 04/20/2024] [Indexed: 04/28/2024]
Abstract
Titanium dioxide nanoparticles (TiO2-NPs) are widely used in food, paint, coating, cosmetic, and composite orthodontic material. As a common food additive, TiO2-NPs can accumulate in various organs of human body, but the effect and underlying mechanism of bone remain unclear. Here mice were exposed to TiO2-NPs by oral gavage, and histological staining of femoral sections showed that TiO2-NPs reduced bone formation and enhanced osteoclast activity and lipogenesis, contributing to decreased trabecula bone. Transmission electron microscope (TEM) as well as biochemical and flow cytometry analysis of osteoblast exhibited that TiO2-NPs accumulated in osteoblast cytoplasm and impaired mitochondria ultrastructure with increased reactive oxygen species (ROS) and lipid hyperoxide, resulting in osteoblast apoptosis. In terms of mechanism, TiO2-NPs treatment inhibited expression of AKT and then increased pro-apoptotic protein Bax expression which was failure to form heterodimers with decreased anti-apoptotic Bcl-2, activating downstream Caspase-9 and Caspase-3 and inducing apoptosis. Additionally, TiO2-NPs suppressed Wnt3a level and then activated anti-Glycogen synthesis kinase (GSK-3β) phosphorylation, and ultimately resulted in degradation of β-catenin which down-regulated Runt-related transcription factor 2 (Runx2) and Osterix, inhibiting expression of osteogenic related proteins. Together, these results revealed that exposure of TiO2-NPs induced apoptosis and inhibited osteoblast differentiation through suppressing PI3K/AKT and Wnt/β-catenin signaling pathways, resulting in reduction of trabecula bone.
Collapse
Affiliation(s)
- Jingxi Xu
- Orthopedic Institute, Medical College, Soochow University, 178 Ganjiang Road, Suzhou, Jiangsu 215007, China
| | - Xiao Ze
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu 215004, China
| | - Linchuan Zhao
- Department of Biological Sciences, School of Basic Medical and Biological Sciences, Soochow University, 199 Ren-ai Road, Soochow, Jiangsu 215123, China
| | - Lei Sheng
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu 215004, China.
| | - Yuguan Ze
- Department of Biochemistry and Molecular Biology, School of Basic Medical and Biological Sciences, Soochow University, 199 Ren-ai Road, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
13
|
Cao S, Wei Y, Yue Y, Xiong A, Zeng H. Zooming in and Out of Programmed Cell Death in Osteoarthritis: A Scientometric and Visualized Analysis. J Inflamm Res 2024; 17:2479-2498. [PMID: 38681072 PMCID: PMC11055561 DOI: 10.2147/jir.s462722] [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: 02/03/2024] [Accepted: 04/09/2024] [Indexed: 05/01/2024] Open
Abstract
During the past decade, mounting evidence has increasingly linked programmed cell death (PCD) to the progression and development of osteoarthritis (OA). There is a significant need for a thorough scientometric analysis that recapitulates the relationship between PCD and OA. This study aimed to collect articles and reviews focusing on PCD in OA, extracting data from January 1st, 2013, to October 31st, 2023, using the Web of Science. Various tools, including VOSviewer, CiteSpace, Pajek, Scimago Graphica, and the R package, were employed for scientometric and visualization analyses. Notably, China, the USA, and South Korea emerged as major contributors, collectively responsible for more than 85% of published papers and significantly influencing research in this field. Among different institutions, Shanghai Jiao Tong University, Xi'an Jiao Tong University, and Zhejiang University exhibited the highest productivity. Prolific authors included Wang Wei, Wang Jing, and Zhang Li. Osteoarthritis and Cartilage had the most publications in this area. Keywords related to PCD in OA prominently highlighted 'chondrocytes', 'inflammation', and 'oxidative stress', recognized as pivotal mechanisms contributing to PCD within OA. This study presents the first comprehensive scientometric analysis, offering a broad perspective on the knowledge framework and evolving patterns concerning PCD in relation to OA over the last decade. Such insights can aid researchers in comprehensively understanding this field and provide valuable directions for future explorations.
Collapse
Affiliation(s)
- Siyang Cao
- National & Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People’s Republic of China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People’s Republic of China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People’s Republic of China
| | - Yihao Wei
- National & Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People’s Republic of China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People’s Republic of China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People’s Republic of China
| | - Yaohang Yue
- National & Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People’s Republic of China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People’s Republic of China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People’s Republic of China
| | - Ao Xiong
- National & Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People’s Republic of China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People’s Republic of China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People’s Republic of China
| | - Hui Zeng
- National & Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People’s Republic of China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People’s Republic of China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People’s Republic of China
| |
Collapse
|
14
|
Han D, Wang W, Gong J, Ma Y, Li Y. Microbiota metabolites in bone: Shaping health and Confronting disease. Heliyon 2024; 10:e28435. [PMID: 38560225 PMCID: PMC10979239 DOI: 10.1016/j.heliyon.2024.e28435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 02/16/2024] [Accepted: 03/19/2024] [Indexed: 04/04/2024] Open
Abstract
The intricate interplay between the gut microbiota and bone health has become increasingly recognized as a fundamental determinant of skeletal well-being. Microbiota-derived metabolites play a crucial role in dynamic interaction, specifically in bone homeostasis. In this sense, short-chain fatty acids (SCFAs), including acetate, propionate, and butyrate, indirectly promote bone formation by regulating insulin-like growth factor-1 (IGF-1). Trimethylamine N-oxide (TMAO) has been found to increase the expression of osteoblast genes, such as Runt-related transcription factor 2 (RUNX2) and bone morphogenetic protein-2 (BMP2), thus enhancing osteogenic differentiation and bone quality through BMP/SMADs and Wnt signaling pathways. Remarkably, in the context of bone infections, the role of microbiota metabolites in immune modulation and host defense mechanisms potentially affects susceptibility to infections such as osteomyelitis. Furthermore, ongoing research elucidates the precise mechanisms through which microbiota-derived metabolites influence bone cells, such as osteoblasts and osteoclasts. Understanding the multifaceted influence of microbiota metabolites on bone, from regulating homeostasis to modulating susceptibility to infections, has the potential to revolutionize our approach to bone health and disease management. This review offers a comprehensive exploration of this evolving field, providing a holistic perspective on the impact of microbiota metabolites on bone health and diseases.
Collapse
Affiliation(s)
- Dong Han
- Department of Trauma Orthopedics, Yantaishan Hospital, Yantai 264000, China
| | - Weijiao Wang
- Department of Otolaryngology, Yantaishan Hospital, Yantai 264000, China
| | - Jinpeng Gong
- Department of Trauma Orthopedics, Yantaishan Hospital, Yantai 264000, China
| | - Yupeng Ma
- Department of Trauma Orthopedics, Yantaishan Hospital, Yantai 264000, China
| | - Yu Li
- Department of Trauma Orthopedics, Yantaishan Hospital, Yantai 264000, China
| |
Collapse
|
15
|
Wadke A, Kommuri K, Talluri S, Kalladka M, Kalladka G, Khan J. Effect of Lithium on Orthodontic Tooth Movement: a Systematic Review of Animal Studies. Turk J Orthod 2024; 37:63-71. [PMID: 38556955 PMCID: PMC10986460 DOI: 10.4274/turkjorthod.2023.2022.149] [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: 09/30/2022] [Accepted: 03/15/2023] [Indexed: 04/02/2024]
Abstract
Objective This study aimed to systematically review the effect of lithium on orthodontic tooth movement (OTM). Methods The focus question was "does lithium have an effect on OTM?" A systematic search was conducted using indexed databases and the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines were followed. The quality assessment of the selected studies was performed according to the systematic review center for laboratory animal experimentation. Results Five of the initially identified 656 articles fulfilled the eligibility criteria and were selected for this review. The studies reported that lithium administration lowered the rate of OTM by inducing a reduction in the number of osteoclasts and possibly inhibiting osteoclastogenesis. These studies further showed an increase in bone density and bone volume by promoting the Wnt/ß-catenin signaling pathway and osteoblastogenesis. It was also noted that lithium reduced orthodontically induced root resorption during experimental OTM. Further, standardized studies are warranted to understand the impact of lithium in OTM. Overall, the risk of bias for 3 studies was very high, high in 1 study, and moderate in 1 study. Conclusion On an experimental level in animals, lithium decreased the rate of OTM during the active treatment phase by increasing bone density and bone volume and reducing root resorption. In addition, lithium may enhance alveolar bone formation during orthodontic retention. Clinically, this may impact the orthodontic treatment duration in patients receiving lithium, and further studies are needed to understand the true impact of lithium on OTM.
Collapse
Affiliation(s)
- Amit Wadke
- Orofacial Pain and TMJ Disorders, Eastman Institute for Oral Health, University of Rochester, NY, USA
| | - Karthik Kommuri
- Orthodontics and Craniofacial Orthopedics, Eastman Institute for Oral Health, University of Rochester, NY, USA
| | - Sandeep Talluri
- Orofacial Pain and TMJ Disorders, Eastman Institute for Oral Health, University of Rochester, NY, USA
| | - Mythili Kalladka
- Orofacial Pain and TMJ Disorders, Eastman Institute for Oral Health, University of Rochester, NY, USA
| | - Goutham Kalladka
- Dayananda Sagar College of Dental Sciences, Department of Orthodontics, Karnataka, India
| | - Junad Khan
- Orofacial Pain and TMJ Disorders, Eastman Institute for Oral Health, University of Rochester, NY, USA
| |
Collapse
|
16
|
ElGhazaly M, Collins MO, Ibler AEM, Humphreys D. Typhoid toxin hijacks Wnt5a to establish host senescence and Salmonella infection. Cell Rep 2023; 42:113181. [PMID: 37792529 DOI: 10.1016/j.celrep.2023.113181] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 06/15/2023] [Accepted: 09/13/2023] [Indexed: 10/06/2023] Open
Abstract
Damage to our genome causes acute senescence in mammalian cells, which undergo growth arrest and release a senescence-associated secretory phenotype (SASP) that propagates the stress response to bystander cells. Thus, acute senescence is a powerful tumor suppressor. Salmonella enterica hijacks senescence through its typhoid toxin, which usurps unidentified factors in the stress secretome of senescent cells to mediate intracellular infections. Here, transcriptomics of toxin-induced senescent cells (TxSCs) and proteomics of their secretome identify the factors as Wnt5a, INHBA, and GDF15. Wnt5a establishes a positive feedback loop, driving INHBA and GDF15 expression. In fibroblasts, Wnt5a and INHBA mediate autocrine senescence in TxSCs and paracrine senescence in naive cells. Wnt5a synergizes with GDF15 to increase Salmonella invasion. Intestinal TxSCs undergo apoptosis without Wnt5a, which is required for establishing intestinal TxSCs. The study reveals how an innate defense against cancer is co-opted by a bacterial pathogen to cause widespread damage and mediate infections.
Collapse
Affiliation(s)
- Mohamed ElGhazaly
- School of Biosciences, University of Sheffield, Sheffield, South Yorkshire S10 2TN, UK
| | - Mark O Collins
- School of Biosciences, University of Sheffield, Sheffield, South Yorkshire S10 2TN, UK
| | - Angela E M Ibler
- School of Biosciences, University of Sheffield, Sheffield, South Yorkshire S10 2TN, UK
| | - Daniel Humphreys
- School of Biosciences, University of Sheffield, Sheffield, South Yorkshire S10 2TN, UK.
| |
Collapse
|
17
|
Vimalraj S, Sekaran S. RUNX Family as a Promising Biomarker and a Therapeutic Target in Bone Cancers: A Review on Its Molecular Mechanism(s) behind Tumorigenesis. Cancers (Basel) 2023; 15:3247. [PMID: 37370857 DOI: 10.3390/cancers15123247] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/10/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
The transcription factor runt-related protein (RUNX) family is the major transcription factor responsible for the formation of osteoblasts from bone marrow mesenchymal stem cells, which are involved in bone formation. Accumulating evidence implicates the RUNX family for its role in tumor biology and cancer progression. The RUNX family has been linked to osteosarcoma via its regulation of many tumorigenicity-related factors. In the regulatory network of cancers, with numerous upstream signaling pathways and its potential target molecules downstream, RUNX is a vital molecule. Hence, a pressing need exists to understand the precise process underpinning the occurrence and prognosis of several malignant tumors. Until recently, RUNX has been regarded as one of the therapeutic targets for bone cancer. Therefore, in this review, we have provided insights into various molecular mechanisms behind the tumorigenic role of RUNX in various important cancers. RUNX is anticipated to grow into a novel therapeutic target with the in-depth study of RUNX family-related regulatory processes, aid in the creation of new medications, and enhance clinical efficacy.
Collapse
Affiliation(s)
- Selvaraj Vimalraj
- Department of Prosthodontics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Saravanan Sekaran
- Department of Prosthodontics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai 600077, Tamil Nadu, India
| |
Collapse
|
18
|
Castillo-Azofeifa D, Wald T, Reyes EA, Gallagher A, Schanin J, Vlachos S, Lamarche-Vane N, Bomidi C, Blutt S, Estes MK, Nystul T, Klein OD. A DLG1-ARHGAP31-CDC42 axis is essential for the intestinal stem cell response to fluctuating niche Wnt signaling. Cell Stem Cell 2023; 30:188-206.e6. [PMID: 36640764 PMCID: PMC9922544 DOI: 10.1016/j.stem.2022.12.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 10/13/2022] [Accepted: 12/12/2022] [Indexed: 01/15/2023]
Abstract
A central factor in the maintenance of tissue integrity is the response of stem cells to variations in the levels of niche signals. In the gut, intestinal stem cells (ISCs) depend on Wnt ligands for self-renewal and proliferation. Transient increases in Wnt signaling promote regeneration after injury or in inflammatory bowel diseases, whereas constitutive activation of this pathway leads to colorectal cancer. Here, we report that Discs large 1 (Dlg1), although dispensable for polarity and cellular turnover during intestinal homeostasis, is required for ISC survival in the context of increased Wnt signaling. RNA sequencing (RNA-seq) and genetic mouse models demonstrated that DLG1 regulates the cellular response to increased canonical Wnt ligands. This occurs via the transcriptional regulation of Arhgap31, a GTPase-activating protein that deactivates CDC42, an effector of the non-canonical Wnt pathway. These findings reveal a DLG1-ARHGAP31-CDC42 axis that is essential for the ISC response to increased niche Wnt signaling.
Collapse
Affiliation(s)
- David Castillo-Azofeifa
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA; Department of Regenerative Medicine, Genentech, Inc., South San Francisco, CA, USA
| | - Tomas Wald
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Efren A Reyes
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA; Department of Pharmaceutical Chemistry and TETRAD Program, University of California, San Francisco, San Francisco, CA, USA
| | - Aaron Gallagher
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Julia Schanin
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Stephanie Vlachos
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Nathalie Lamarche-Vane
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada; Department of Anatomy and Cell Biology, McGill University, Montréal, QC, Canada
| | - Carolyn Bomidi
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Sarah Blutt
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Mary K Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Todd Nystul
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Ophir D Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA; Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA; Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
19
|
Vlashi R, Zhang X, Wu M, Chen G. Wnt signaling: essential roles in osteoblast differentiation, bone metabolism and therapeutic implications for bone and skeletal disorders. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
|
20
|
Gassel LC, Schneider S, Banke IJ, Braun KF, Volkering C, Zeeb L, Burgkart RHH, von Eisenhart-Rothe R, Biberthaler P, van Griensven M, Haug AT. Dysregulation of Wnt signaling in bone of type 2 diabetes mellitus and diabetic Charcot arthropathy. BMC Musculoskelet Disord 2022; 23:365. [PMID: 35436882 PMCID: PMC9017014 DOI: 10.1186/s12891-022-05314-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 04/04/2022] [Indexed: 12/03/2022] Open
Abstract
Background Type 2 diabetes mellitus (T2DM) patients show a markedly higher fracture risk and impaired fracture healing when compared to non-diabetic patients. However in contrast to type 1 diabetes mellitus, bone mineral density in T2DM is known to be normal or even regionally elevated, also known as diabetic bone disease. Charcot arthropathy is a severe and challenging complication leading to bone destruction and mutilating bone deformities. Wnt signaling is involved in increasing bone mineral density, bone homeostasis and apoptotic processes. It has been shown that type 2 diabetes mellitus is strongly associated with gene variants of the Wnt signaling pathway, specifically polymorphisms of TCF7L2 (transcription factor 7 like 2), which is an effector transcription factor of this pathway. Methods Bone samples of 19 T2DM patients and 7 T2DM patients with additional Charcot arthropathy were compared to 19 non-diabetic controls. qPCR analysis for selected members of the Wnt-signaling pathway (WNT3A, WNT5A, catenin beta, TCF7L2) and bone gamma-carboxyglutamate (BGLAP, Osteocalcin) was performed and analyzed using the 2-ΔΔCt- Method. Statistical analysis comprised one-way analysis of variance (ANOVA). Results In T2DM patients who had developed Charcot arthropathy WNT3A and WNT5A gene expression was down-regulated by 89 and 58% compared to healthy controls (p < 0.0001). TCF7L2 gene expression showed a significant reduction by 63% (p < 0.0001) and 18% (p = 0.0136) in diabetic Charcot arthropathy. In all diabetic patients BGLAP (Osteocalcin) was significantly decreased by at least 59% (p = 0.0019). Conclusions For the first time with this study downregulation of members of the Wnt-signaling pathway has been shown in the bone of diabetic patients with and without Charcot arthropathy. This may serve as future therapeutic target for this severe disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12891-022-05314-9.
Collapse
Affiliation(s)
- Laurens Christian Gassel
- Department of Experimental Trauma Surgery, and Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Strasse 22, 81675, Munich, Germany
| | - Sandra Schneider
- Department of Experimental Trauma Surgery, and Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Strasse 22, 81675, Munich, Germany
| | - Ingo Jörg Banke
- Department of Orthopedics and Sports Orthopedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Strasse 22, 81675, Munich, Germany
| | - Karl Friedrich Braun
- Charité - Berlin University of Medicine, Center for Musculoskeletal Surgery, Campus Virchow-Klinikum (CVK), Augustenburger Platz 1, 13353, Berlin, Germany.,Department of Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Strasse 22, 81675, Munich, Germany
| | | | - Leonie Zeeb
- Department of Experimental Trauma Surgery, and Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Strasse 22, 81675, Munich, Germany
| | - Rainer Hans Hermann Burgkart
- Department of Orthopedics and Sports Orthopedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Strasse 22, 81675, Munich, Germany
| | - Rüdiger von Eisenhart-Rothe
- Department of Orthopedics and Sports Orthopedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Strasse 22, 81675, Munich, Germany
| | - Peter Biberthaler
- Department of Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Strasse 22, 81675, Munich, Germany
| | - Martijn van Griensven
- Department of Experimental Trauma Surgery, and Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Strasse 22, 81675, Munich, Germany.,Department cBITE, MERLN Institute, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Alexander Tobias Haug
- Department of Experimental Trauma Surgery and, Department of Orthopedics and Sports Orthopedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Strasse 22, 81675, Munich, Germany.
| |
Collapse
|
21
|
Martínez-Gil N, Ugartondo N, Grinberg D, Balcells S. Wnt Pathway Extracellular Components and Their Essential Roles in Bone Homeostasis. Genes (Basel) 2022; 13:genes13010138. [PMID: 35052478 PMCID: PMC8775112 DOI: 10.3390/genes13010138] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 12/11/2022] Open
Abstract
The Wnt pathway is involved in several processes essential for bone development and homeostasis. For proper functioning, the Wnt pathway is tightly regulated by numerous extracellular elements that act by both activating and inhibiting the pathway at different moments. This review aims to describe, summarize and update the findings regarding the extracellular modulators of the Wnt pathway, including co-receptors, ligands and inhibitors, in relation to bone homeostasis, with an emphasis on the animal models generated, the diseases associated with each gene and the bone processes in which each member is involved. The precise knowledge of all these elements will help us to identify possible targets that can be used as a therapeutic target for the treatment of bone diseases such as osteoporosis.
Collapse
|
22
|
Abstract
Osteocytes, former osteoblasts encapsulated by mineralized bone matrix, are far from being passive and metabolically inactive bone cells. Instead, osteocytes are multifunctional and dynamic cells capable of integrating hormonal and mechanical signals and transmitting them to effector cells in bone and in distant tissues. Osteocytes are a major source of molecules that regulate bone homeostasis by integrating both mechanical cues and hormonal signals that coordinate the differentiation and function of osteoclasts and osteoblasts. Osteocyte function is altered in both rare and common bone diseases, suggesting that osteocyte dysfunction is directly involved in the pathophysiology of several disorders affecting the skeleton. Advances in osteocyte biology initiated the development of novel therapeutics interfering with osteocyte-secreted molecules. Moreover, osteocytes are targets and key distributors of biological signals mediating the beneficial effects of several bone therapeutics used in the clinic. Here we review the most recent discoveries in osteocyte biology demonstrating that osteocytes regulate bone homeostasis and bone marrow fat via paracrine signaling, influence body composition and energy metabolism via endocrine signaling, and contribute to the damaging effects of diabetes mellitus and hematologic and metastatic cancers in the skeleton.
Collapse
Affiliation(s)
- Jesus Delgado-Calle
- 1Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas,2Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Teresita Bellido
- 1Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas,2Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas,3Central Arkansas Veterans Healthcare System, Little Rock, Arkansas
| |
Collapse
|
23
|
The concurrent stimulation of Wnt and FGF8 signaling induce differentiation of dental mesenchymal cells into odontoblast-like cells. Med Mol Morphol 2021; 55:8-19. [PMID: 34739612 PMCID: PMC8885561 DOI: 10.1007/s00795-021-00297-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/13/2021] [Indexed: 11/05/2022]
Abstract
Fibroblast growth factor 8 (FGF8) is known to be a potent stimulator of canonical Wnt/β-catenin activity, an essential factor for tooth development. In this study, we analyzed the effects of co-administration of FGF8 and a CHIR99021 (GSK3β inhibitor) on differentiation of dental mesenchymal cells into odontoblasts. Utilizing Cre-mediated EGFP reporter mice, dentin matrix protein 1 (Dmp1) expression was examined in mouse neonatal molar tooth germs. At birth, expression of Dmp1-EGFP was not found in mesenchymal cells but rather epithelial cells, after which Dmp1-positive cells gradually emerged in the mesenchymal area along with disappearance in the epithelial area. Primary cultured mesenchymal cells from neonatal tooth germ specimens showed loss of Dmp1-EGFP positive signals, whereas addition of Wnt3a or the CHIR99021 significantly regained Dmp1 positivity within approximately 2 weeks. Other odontoblast markers such as dentin sialophosphoprotein (Dspp) could not be clearly detected. Concurrent stimulation of primary cultured mesenchymal cells with the CHIR99021 and FGF8 resulted in significant upregulation of odonto/osteoblast proteins. Furthermore, increased expression levels of runt-related transcription factor 2 (Runx2), osterix, and osteocalcin were also observed. The present findings indicate that coordinated action of canonical Wnt/β-catenin and FGF8 signals is essential for odontoblast differentiation of tooth germs in mice.
Collapse
|
24
|
Cerqueni G, Scalzone A, Licini C, Gentile P, Mattioli-Belmonte M. Insights into oxidative stress in bone tissue and novel challenges for biomaterials. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 130:112433. [PMID: 34702518 DOI: 10.1016/j.msec.2021.112433] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/08/2021] [Accepted: 09/11/2021] [Indexed: 12/28/2022]
Abstract
The presence of Reactive Oxygen Species (ROS) in bone can influence resident cells behaviour as well as the extra-cellular matrix composition and the tissue architecture. Aging, in addition to excessive overloads, unbalanced diet, smoking, predisposing genetic factors, lead to an increase of ROS and, if it is accompanied with an inappropriate production of scavengers, promotes the generation of oxidative stress that encourages bone catabolism. Furthermore, bone injuries can be triggered by numerous events such as road and sports accidents or tumour resection. Although bone tissue possesses a well-known repair and regeneration capacity, these mechanisms are inefficient in repairing large size defects and bone grafts are often necessary. ROS play a fundamental role in response after the implant introduction and can influence its success. This review provides insights on the mechanisms of oxidative stress generated by an implant in vivo and suitable ways for its modulation. The local delivery of active molecules, such as polyphenols, enhanced bone biomaterial integration evidencing that the management of the oxidative stress is a target for the effectiveness of an implant. Polyphenols have been widely used in medicine for cardiovascular, neurodegenerative, bone disorders and cancer, thanks to their antioxidant and anti-inflammatory properties. In addition, the perspective of new smart biomaterials and molecular medicine for the oxidative stress modulation in a programmable way, by the use of ROS responsive materials or by the targeting of selective molecular pathways involved in ROS generation, will be analysed and discussed critically.
Collapse
Affiliation(s)
- Giorgia Cerqueni
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Via Tronto 10/a, Ancona 60126, Italy
| | - Annachiara Scalzone
- School of Engineering, Newcastle University, Stephenson Building, Claremont Road, Newcastle upon Tyne NE1 7RU, UK
| | - Caterina Licini
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Via Tronto 10/a, Ancona 60126, Italy; Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 204, 10129 Torino, Italy
| | - Piergiorgio Gentile
- School of Engineering, Newcastle University, Stephenson Building, Claremont Road, Newcastle upon Tyne NE1 7RU, UK
| | - Monica Mattioli-Belmonte
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Via Tronto 10/a, Ancona 60126, Italy.
| |
Collapse
|
25
|
Debnath N, Kumar R, Kumar A, Mehta PK, Yadav AK. Gut-microbiota derived bioactive metabolites and their functions in host physiology. Biotechnol Genet Eng Rev 2021; 37:105-153. [PMID: 34678130 DOI: 10.1080/02648725.2021.1989847] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Every individual harbours a complex, diverse and mutualistic microbial flora in their intestine and over the time it became an integral part of the body, affecting a plethora of activities of the host. Interaction between host and gut-microbiota affects several aspects of host physiology. Gut-microbiota affects host metabolism by fermenting unabsorbed/undigested carbohydrates in the large intestine. Not only the metabolic functions, any disturbances in the composition of the gut-microbiota during first 2-3 years of life may impact on the brain development and later affects cognition and behaviour. Thus, gut-dysbiosis causes certain serious pathological conditions in the host including metabolic disorders, inflammatory bowel disease and mood alterations, etc. Microbial-metabolites in recent times have emerged as key mediators and are responsible for microbiota induced beneficial effects on host. This review provides an overview of the mechanism of microbial-metabolite production, their respective physiological functions and the impact of gut-microbiome in health and diseases. Metabolites from dietary fibres, aromatic amino acids such as tryptophan, primary bile acids and others are the potential substances and link microbiota to host physiology. Many of these metabolites act as signalling molecules to a number of cells types and also help in the secretion of hormones. Moreover, interaction of microbiota derived metabolites with their host, immunity boosting mechanisms, protection against pathogens and modulation of metabolism is also highlighted here. Understanding all these functional attributes of metabolites produced from gut-microbiota may lead to the opening of a new avenue for preventing and developing potent therapies against several diseases.
Collapse
Affiliation(s)
- Nabendu Debnath
- Centre for Molecular Biology, Central University of Jammu, Samba, Jammu & Kashmir, India
| | | | - Ashwani Kumar
- Department of Nutrition Biology, Central University of Haryana, Mahendergarh, Jant-Pali, India
| | - Praveen Kumar Mehta
- Centre for Molecular Biology, Central University of Jammu, Samba, Jammu & Kashmir, India
| | - Ashok Kumar Yadav
- Centre for Molecular Biology, Central University of Jammu, Samba, Jammu & Kashmir, India
| |
Collapse
|
26
|
Lojk J, Marc J. Roles of Non-Canonical Wnt Signalling Pathways in Bone Biology. Int J Mol Sci 2021; 22:10840. [PMID: 34639180 PMCID: PMC8509327 DOI: 10.3390/ijms221910840] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/01/2021] [Accepted: 10/05/2021] [Indexed: 01/15/2023] Open
Abstract
The Wnt signalling pathway is one of the central signalling pathways in bone development, homeostasis and regulation of bone mineral density. It consists of numerous Wnt ligands, receptors and co-receptors, which ensure tight spatiotemporal regulation of Wnt signalling pathway activity and thus tight regulation of bone tissue homeostasis. This enables maintenance of optimal mineral density, tissue healing and adaptation to changes in bone loading. While the role of the canonical/β-catenin Wnt signalling pathway in bone homeostasis is relatively well researched, Wnt ligands can also activate several non-canonical, β-catenin independent signalling pathways with important effects on bone tissue. In this review, we will provide a thorough overview of the current knowledge on different non-canonical Wnt signalling pathways involved in bone biology, focusing especially on the pathways that affect bone cell differentiation, maturation and function, processes involved in bone tissue structure regulation. We will describe the role of the two most known non-canonical pathways (Wnt/planar cell polarity pathways and Wnt/Ca2+ pathway), as well as other signalling pathways with a strong role in bone biology that communicate with the Wnt signalling pathway through non-canonical Wnt signalling. Our goal is to bring additional attention to these still not well researched but important pathways in the regulation of bone biology in the hope of prompting additional research in the area of non-canonical Wnt signalling pathways.
Collapse
Affiliation(s)
- Jasna Lojk
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia;
| | - Janja Marc
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia;
- University Clinical Center Ljubljana, Clinical Department of Clinical Chemistry and Biochemistry, 1000 Ljubljana, Slovenia
| |
Collapse
|
27
|
Study on the Mechanism of Salvia miltiorrhiza in the Treatment of Traumatic Bone Defects. J CHEM-NY 2021. [DOI: 10.1155/2021/8646394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Traumatic bone defect is one of the major orthopedic diseases in clinics, and its incidence is increasing year by year. And repairing traumatic bone defects is a very difficult problem in clinics at present. The surface of medical titanium-based alloy has good biological properties, and its implant has a certain role in promoting bone in bone tissue. However, titanium-based materials are biologically inert and have no biological activity. As a traditional Chinese medicine, Salvia miltiorrhiza has the efficacy of treating bone diseases and promoting bone healing. The curative effect can be better exerted by loading the traditional Chinese medicine active compound Salvia miltiorrhiza on the surface of the titanium implant in a certain way. At present, due to the complex chemical composition of Salvia miltiorrhiza, the mechanism of its use for the treatment of traumatic bone defects is still unclear. Therefore, in this study, we mainly discussed the potential target and mechanism of Salvia miltiorrhiza in the treatment of traumatic bone defects through network pharmacology, which may provide a scientific basis for the treatment of traumatic bone defects with Salvia miltiorrhiza loaded on the surface of medical titanium-based alloy. We screened out effective compounds and targets of Salvia miltiorrhiza and targets related to traumatic bone defects with the help of relevant databases. The targets of Salvia miltiorrhiza for traumatic bone defects were analyzed by STRING and GeneCards databases, and the results were visualized by constructing a compound-target network, protein-protein interaction network, and compound-target-disease network with Cytoscape 3.7.1 analysis software. Finally, the selected core targets carried out GO and KEGG enrichment. The results showed that 60 main active components were screened from Salvia miltiorrhiza Bunge, which could act on 149 targets. There were 33 active components and 70 targets related to traumatic bone defects, respectively. The core targets of Salvia miltiorrhiza in the treatment of traumatic bone defects were MAPK1, MAPK10, MAPK14, TGFB1, and TNF. The results of enrichment analysis showed that Salvia miltiorrhiza might treat traumatic bone defects through an osteogenic differentiation pathway.
Collapse
|
28
|
Peng CH, Lin WY, Yeh KT, Chen IH, Wu WT, Lin MD. The molecular etiology and treatment of glucocorticoid-induced osteoporosis. Tzu Chi Med J 2021; 33:212-223. [PMID: 34386357 PMCID: PMC8323641 DOI: 10.4103/tcmj.tcmj_233_20] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/19/2020] [Accepted: 12/30/2020] [Indexed: 12/30/2022] Open
Abstract
Glucocorticoid-induced osteoporosis (GIOP) is the most common form of secondary osteoporosis, accounting for 20% of osteoporosis diagnoses. Using glucocorticoids for >6 months leads to osteoporosis in 50% of patients, resulting in an increased risk of fracture and death. Osteoblasts, osteocytes, and osteoclasts work together to maintain bone homeostasis. When bone formation and resorption are out of balance, abnormalities in bone structure or function may occur. Excess glucocorticoids disrupt the bone homeostasis by promoting osteoclast formation and prolonging osteoclasts' lifespan, leading to an increase in bone resorption. On the other hand, glucocorticoids inhibit osteoblasts' formation and facilitate apoptosis of osteoblasts and osteocytes, resulting in a reduction of bone formation. Several signaling pathways, signaling modulators, endocrines, and cytokines are involved in the molecular etiology of GIOP. Clinically, adults ≥40 years of age using glucocorticoids chronically with a high fracture risk are considered to have medical intervention. In addition to vitamin D and calcium tablet supplementations, the major therapeutic options approved for GIOP treatment include antiresorption drug bisphosphonates, parathyroid hormone N-terminal fragment teriparatide, and the monoclonal antibody denosumab. The selective estrogen receptor modulator can only be used under specific condition for postmenopausal women who have GIOP but fail to the regular GIOP treatment or have specific therapeutic contraindications. In this review, we focus on the molecular etiology of GIOP and the molecular pharmacology of the therapeutic drugs used for GIOP treatment.
Collapse
Affiliation(s)
- Cheng-Huan Peng
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Wen-Ying Lin
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Kuang-Ting Yeh
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Ing-Ho Chen
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Wen-Tien Wu
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Ming-Der Lin
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan.,Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
29
|
Yan T, Xie Y, He H, Fan W, Huang F. Role of nitric oxide in orthodontic tooth movement (Review). Int J Mol Med 2021; 48:168. [PMID: 34278439 PMCID: PMC8285047 DOI: 10.3892/ijmm.2021.5001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/08/2021] [Indexed: 12/14/2022] Open
Abstract
Nitric oxide (NO) is an ubiquitous signaling molecule that mediates numerous cellular processes associated with cardiovascular, nervous and immune systems. NO also plays an essential role in bone homeostasis regulation. The present review article summarized the effects of NO on bone metabolism during orthodontic tooth movement in order to provide insight into the regulatory role of NO in orthodontic tooth movement. Orthodontic tooth movement is a process in which the periodontal tissue and alveolar bone are reconstructed due to the effect of orthodontic forces. Accumulating evidence has indicated that NO and its downstream signaling molecule, cyclic guanosine monophosphate (cGMP), mediate the mechanical signals during orthodontic-related bone remodeling, and exert complex effects on osteogenesis and osteoclastogenesis. NO has a regulatory effect on the cellular activities and functional states of osteoclasts, osteocytes and periodontal ligament fibroblasts involved in orthodontic tooth movement. Variations of NO synthase (NOS) expression levels and NO production in periodontal tissues or gingival crevicular fluid (GCF) have been found on the tension and compression sides during tooth movement in both orthodontic animal models and patients. Furthermore, NO precursor and NOS inhibitor administration increased and reduced the tooth movement in animal models, respectively. Further research is required in order to further elucidate the underlying mechanisms and the clinical application prospect of NO in orthodontic tooth movement.
Collapse
Affiliation(s)
- Tong Yan
- Department of Pediatric Dentistry, Hospital of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Yongjian Xie
- Department of Orthodontic Dentistry, Hospital of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Hongwen He
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Wenguo Fan
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Fang Huang
- Department of Pediatric Dentistry, Hospital of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| |
Collapse
|
30
|
Wang F, Tu P, Zeng K, Jiang Y. Total glycosides and polysaccharides of Cistanche deserticola prevent osteoporosis by activating Wnt/β-catenin signaling pathway in SAMP6 mice. JOURNAL OF ETHNOPHARMACOLOGY 2021; 271:113899. [PMID: 33549763 DOI: 10.1016/j.jep.2021.113899] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 12/31/2020] [Accepted: 01/27/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese medicine Cistanche deserticola Y. C. Ma has effect of "tonifying kidney and strengthening bone". However, the specific active extracts of C. deserticola and mechanisms for treatment of osteoporotic are not clear. AIM OF THE STUDY We wanted to identify the effective component extracts of C. deserticola for the treatment of osteoporosis and the potential mechanisms. MATERIALS AND METHODS Our group researched the extracts of C. deserticola with anti-osteoporotic activity, including total glycosides (TGs), polysaccharides (PSs), and oligosaccharides (OSs) in senescence accelerated mouse prone 6 (SAMP6) mice. The Goldner's Trichrome, Van Gieson's (VG), Safranin O-Fast Green staining and Von Kossa staining were performed to investigate the bone structure formation and calcium deposits. Serum was collected for detecting biochemical markers. Bone micro-architecture was detected by micro-CT. Expressions of bone morphogenetic protein-2 (BMP-2), osteocalcin (OCN), osteoprotegerin (OPG), receptor activator of nuclear factor-κ B ligand (RANKL), p-glycogen synthetase kinase-3β (p-GSK-3β), and p-β-catenin were analyzed by western blotting and immunohistochemistry. RESULTS TGs and PSs ameliorated bone histopathological damages, promoted the formation of new bone, collagenous fiber, and chondrocytes, and accelerated the calcium deposits. Moreover, they remarkable altered the biomarkers of bone turnover and effectively ameliorated bone microarchitecture. The further mechanisms study showed that TGs and PSs significantly decreased the expressions of RANKL, p-β-catenin, as well as up-regulated the expression of BMP-2, OCN, OPG, and p-GSK-3β (Ser9). CONCLUSION The findings of this study suggest that TGs and PSs can promote osteoblastogenic bone formation and improve bone microstructure damage in SAMP6 mice, and their therapeutic effect on osteoporosis is via activating Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Fujiang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Pengfei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Kewu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| | - Yong Jiang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| |
Collapse
|
31
|
Camal Ruggieri IN, Cícero AM, Issa JPM, Feldman S. Bone fracture healing: perspectives according to molecular basis. J Bone Miner Metab 2021; 39:311-331. [PMID: 33151416 DOI: 10.1007/s00774-020-01168-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 10/13/2020] [Indexed: 12/11/2022]
Abstract
Fractures have a great impact on health all around the world and with fracture healing optimization; this problem could be resolved partially. To make a practical contribution to this issue, the knowledge of bone tissue, cellularity, and metabolism is essential, especially cytoskeletal architecture and its transformations according to external pressures. Special physical and chemical characteristics of the extracellular matrix (ECM) allow the transmission of mechanical stimuli from outside the cell to the plasmatic membrane. The osteocyte cytoskeleton is conformed by a complex network of actin and microtubules combined with crosslinker proteins like vinculin and fimbrin, connecting and transmitting outside stimuli through EMC to cytoplasm. Herein, critical signaling pathways like Cx43-depending ones, MAPK/ERK, Wnt, YAP/TAZ, Rho-ROCK, and others are activated due to mechanical stimuli, resulting in osteocyte cytoskeletal changes and ECM remodeling, altering the tissue and, therefore, the bone. In recent years, the osteocyte has gained more interest and value in relation to bone homeostasis as a great coordinator of other cell populations, thanks to its unique functions. By integrating the latest advances in relation to intracellular signaling pathways, mechanotransmission system of the osteocyte and bone tissue engineering, there are promising experimental strategies, while some are ready for clinical trials. This work aims to show clearly and precisely the integration between cytoskeleton and main molecular pathways in relation to mechanotransmission mechanism in osteocytes, and the use of this theoretical knowledge in therapeutic tools for bone fracture healing.
Collapse
Affiliation(s)
- Iván Nadir Camal Ruggieri
- School of Medicine, LABOATEM (Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory), Biological Chemistry Cat, School of Medicine, Rosario National University, Rosario, Argentina.
| | - Andrés Mauricio Cícero
- School of Medicine, LABOATEM (Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory), Biological Chemistry Cat, School of Medicine, Rosario National University, Rosario, Argentina
| | | | - Sara Feldman
- School of Medicine, LABOATEM (Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory), Biological Chemistry Cat, School of Medicine, Rosario National University, Rosario, Argentina
- Research Council of the Rosario National University (CIUNR) and CONICET, Rosario, Argentina
| |
Collapse
|
32
|
Abstract
Exposed surfaces of mammals are colonized with 100 trillion indigenous bacteria, fungi, and viruses, creating a diverse ecosystem known as the human microbiome. The gut microbiome is the richest microbiome and is now known to regulate postnatal skeletal development and the activity of the major endocrine regulators of bone. Parathyroid hormone (PTH) is one of the bone-regulating hormone that requires elements of the gut microbiome to exert both its bone catabolic and its bone anabolic effects. How the gut microbiome regulates the skeletal response to PTH is object of intense research. Involved mechanisms include absorption and diffusion of bacterial metabolites, such as short-chain fatty acids, and trafficking of immune cells from the gut to the bone marrow. This review will focus on how the gut microbiome communicates and regulates bone marrow cells in order to modulate the skeletal effects of PTH.
Collapse
Affiliation(s)
- Roberto Pacifici
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University, Atlanta, GA, USA
- Emory Microbiome Research Center, Emory University, Atlanta, GA, USA
- Immunology and Molecular Pathogenesis Program, Emory University, Atlanta, GA, USA
| |
Collapse
|
33
|
Multiple Myeloma Bone Disease: Implication of MicroRNAs in Its Molecular Background. Int J Mol Sci 2021; 22:ijms22052375. [PMID: 33673480 PMCID: PMC7956742 DOI: 10.3390/ijms22052375] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 02/23/2021] [Accepted: 02/26/2021] [Indexed: 12/16/2022] Open
Abstract
Multiple myeloma (MM) is a common hematological malignancy arising from terminally differentiated plasma cells. In the majority of cases, symptomatic disease is characterized by the presence of bone disease. Multiple myeloma bone disease (MMBD) is a result of an imbalance in the bone-remodeling process that leads to increased osteoclast activity and decreased osteoblast activity. The molecular background of MMBD appears intriguingly complex, as several signaling pathways and cell-to-cell interactions are implicated in the pathophysiology of MMBD. MicroRNAs (miRNAs) are small non-coding RNA molecules that regulate the expression of their target mRNAs. Numerous miRNAs have been witnessed to be involved in cancer and hematological malignancies and their role has been characterized either as oncogenic or oncosuppressive. Recently, scientific research turned towards miRNAs as regulators of MMBD. Scientific data support that miRNAs finely regulate the majority of the signaling pathways implicated in MMBD. In this review, we provide concise information regarding the molecular pathways with a significant role in MMBD and the miRNAs implicated in their regulation. Moreover, we discuss their utility as molecular biomarkers and highlight the putative usage of miRNAs as novel molecular targets for targeted therapy in MMBD.
Collapse
|
34
|
Shen S, Zhang Y, Zhang S, Wang B, Shang L, Shao J, Lin M, Cui Y, Sun S, Ge S. 6-Bromoindirubin-3'-oxime Promotes Osteogenic Differentiation of Periodontal Ligament Stem Cells and Facilitates Bone Regeneration in a Mouse Periodontitis Model. ACS Biomater Sci Eng 2020; 7:232-241. [PMID: 33320531 DOI: 10.1021/acsbiomaterials.0c01078] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Effective bone tissue engineering is important to overcome the unmet clinical challenges of periodontal tissue regeneration. Successful bone tissue engineering comprises three key factors: stem cells, growth factors, and scaffolds. 6-Bromoindirubin-3'-oxime (BIO) is an inhibitor of glycogen synthase kinase-3 (GSK-3) that can activate the Wnt signaling pathway by enhancing β-catenin activity. In this study, the effects of BIO on the proliferation, migration, and osteogenic differentiation of periodontal ligament stem cells (PDLSCs) were investigated. Poly(lactic-co-glycolic acid) (PLGA) and hyaluronic acid (HA) emerged as promising biomaterials; thus, we developed a novel HA hydrogel embedded with BIO-encapsulated PLGA microspheres and injected the formulation into the gingival sulcus of mice with experimental periodontitis. The release speed of this system was fast in the first week and followed a sustained release phase until week 4. In vivo experiments showed that this PLGA-BIO-HA hydrogel system can inhibit periodontal inflammation, promote bone regeneration, and induce the expression of bone-forming markers alkaline phosphatase (ALP), runt-related transcription factor 2 (Runx2), and osteocalcin (OCN) in a mouse periodontitis model. Therefore, this PLGA-BIO-HA hydrogel system provides a promising therapeutic strategy for periodontal bone regeneration.
Collapse
Affiliation(s)
- Song Shen
- Department of Periodontology & Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong University, 250012 Jinan, Shandong, China
| | - Yilin Zhang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021 Jinan, Shandong, China
| | - Songmei Zhang
- Eastman Institute for Oral Health, School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, 14642 New York, United States
| | - Bing Wang
- Department of Periodontology & Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong University, 250012 Jinan, Shandong, China
| | - Lingling Shang
- Department of Periodontology & Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong University, 250012 Jinan, Shandong, China
| | - Jinlong Shao
- Department of Periodontology & Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong University, 250012 Jinan, Shandong, China
| | - Meng Lin
- School of Chemistry and Chemical Engineering, Shandong University, 250012 Jinan, Shandong, China
| | - Yating Cui
- Department of Periodontology & Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong University, 250012 Jinan, Shandong, China
| | - Shengjun Sun
- Department of Periodontology & Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong University, 250012 Jinan, Shandong, China
| | - Shaohua Ge
- Department of Periodontology & Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong University, 250012 Jinan, Shandong, China
| |
Collapse
|
35
|
Alcorta-Sevillano N, Macías I, Infante A, Rodríguez CI. Deciphering the Relevance of Bone ECM Signaling. Cells 2020; 9:E2630. [PMID: 33297501 PMCID: PMC7762413 DOI: 10.3390/cells9122630] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 12/11/2022] Open
Abstract
Bone mineral density, a bone matrix parameter frequently used to predict fracture risk, is not the only one to affect bone fragility. Other factors, including the extracellular matrix (ECM) composition and microarchitecture, are of paramount relevance in this process. The bone ECM is a noncellular three-dimensional structure secreted by cells into the extracellular space, which comprises inorganic and organic compounds. The main inorganic components of the ECM are calcium-deficient apatite and trace elements, while the organic ECM consists of collagen type I and noncollagenous proteins. Bone ECM dynamically interacts with osteoblasts and osteoclasts to regulate the formation of new bone during regeneration. Thus, the composition and structure of inorganic and organic bone matrix may directly affect bone quality. Moreover, proteins that compose ECM, beyond their structural role have other crucial biological functions, thanks to their ability to bind multiple interacting partners like other ECM proteins, growth factors, signal receptors and adhesion molecules. Thus, ECM proteins provide a complex network of biochemical and physiological signals. Herein, we summarize different ECM factors that are essential to bone strength besides, discussing how these parameters are altered in pathological conditions related with bone fragility.
Collapse
Affiliation(s)
| | | | - Arantza Infante
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, Barakaldo, 48903 Bizkaia, Spain; (N.A.-S.); (I.M.)
| | - Clara I. Rodríguez
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, Barakaldo, 48903 Bizkaia, Spain; (N.A.-S.); (I.M.)
| |
Collapse
|
36
|
Fibronectin 1 activates WNT/β-catenin signaling to induce osteogenic differentiation via integrin β1 interaction. J Transl Med 2020; 100:1494-1502. [PMID: 32561820 DOI: 10.1038/s41374-020-0451-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 02/06/2023] Open
Abstract
Osteoporosis (OP) is a systemic skeletal disease leading to fragility fractures and is a major health issue globally. WNT/β-catenin signaling regulates bone-remodeling processes and plays vital roles in OP development. However, the underlying regulatory mechanisms behind WNT/β-catenin signaling in OP requires clarification, as further studies are required to identify novel alternate therapeutic agents to improve OP. Here we report that fibronectin 1 (FN-1) promoted differentiation and mineralization of osteoblasts by activating WNT/β-catenin pathway, in cultured pre-osteoblasts. With isobaric tags for relative and absolute quantitation labeling proteomics analysis, we investigated protein changes in bone samples from OP patients and normal controls. FN-1 accumulated in osteoblasts in bone samples from OP patients and age-related OP mice compared to control group. In addition, we observed that integrin β1 (ITGB1) acts as an indispensable signaling molecule for the interplay between FN-1 and β-catenin, and that FN-1 expression increased, but ITGB1 expression decreased in osteoblasts during OP progression. Therefore, our study reveals a novel explanation for WNT/β-catenin pathway inactivation in OP pathology. Supplying of FN-1 and ITGB1 may provide a potential therapeutic strategy in improving bone formation during OP.
Collapse
|
37
|
Changes in Bone Metabolism and Structure in Primary Hyperparathyroidism. ACTA MEDICA BULGARICA 2020. [DOI: 10.2478/amb-2020-0050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Parathyroid hormone (PTH) is a key regulator of bone turnover. Depending on the duration of action, the hormone causes catabolic and anabolic effects by binding with specific receptors (PTHR1) in the bone. Various cells expressing PTHR1 on their surface are involved in the process – osteoblasts, osteocytes, bone marrow stromal cells, T-lymphocytes and macrophages. In physiological conditions PTH balances the bone metabolism. Intermittent pharmacological doses of PTH lead to the prevalence of bone formation and are used in the treatment of osteoporosis. Persistently elevated levels of PTH stimulate bone resorption by impacting mainly the cortical bone. New imaging and analysis techniques show that high PTH levels can also have an adverse effect on trabecular microarchitecture. Primary hyperparathyroidism (PHPT) is a disease characterized by increased bone metabolism, decreased bone mineral density (BMD), inadequate osteoid mineralization and an increased risk of fractures. Prolonged overproduction of PTH leads to stimulation of bone resorption and defects in bone formation, mainly causing loss of cortical bone mass, while in the trabecular bone predominate demineralization processes. One explanation of these findings is the enhanced stimulation of RANKL expression by osteoblasts with decreased OPG expression and bone formation at the same time.
Collapse
|
38
|
Poudel SB, So HS, Sim HJ, Cho JS, Cho ES, Jeon YM, Kook SH, Lee JC. Osteoblastic Wntless deletion differentially regulates the fate and functions of bone marrow-derived stem cells in relation to age. Stem Cells 2020; 39:103-114. [PMID: 33038284 DOI: 10.1002/stem.3289] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/22/2020] [Indexed: 11/10/2022]
Abstract
Although functional association between Wnt signaling and bone homeostasis has been well described through genetic ablation of Wntless (Wls), the mechanisms of how osteoblastic Wls regulates the fate of bone marrow stromal cells (BMSCs) and hematopoietic stem cells (HSCs) in relation to age are not yet understood. Here, we generated Col2.3-Cre;Wlsfl/fl mice that were free from premature lethality and investigated age-related impacts of osteoblastic Wls deficiency on hematopoiesis, BM microenvironment, and maintenance of BMSCs (also known as BM-derived mesenchymal stem/stromal cells) and HSCs. Ablation of osteoblastic Wls deteriorated BM microenvironment and bone mass accrual along with age-independent effects on functions of BMSCs. Osteoblastic Wls deletion impaired HSC repopulation and progeny with skewing toward myeloid lineage cells only at old stage. As proven by hallmarks of stem cell senescence, osteoblastic Wls ablation differentially induced senescence of BMSCs and HSCs in relation to age without alteration in their BM frequency. Our findings support that deletion of Wls in Col2.3-expressing cells induces senescence of BMSCs and impairs BM microenvironment in age-independent manner. Overall, long-term deterioration in BM microenvironment contributes to age-related HSC senescence with impaired progeny and hematopoiesis, which also suggests possible roles of osteoblastic Wls on the maintenance of BM HSCs.
Collapse
Affiliation(s)
- Sher Bahadur Poudel
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, New York, USA
| | - Han-Sol So
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, South Korea
| | - Hyun-Jaung Sim
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, South Korea
| | - Joon-Seok Cho
- Department of Medicine-Endocrinology, Gerontology and Metabolism, Stanford University School of Medicine, Stanford, California, USA
| | - Eui-Sic Cho
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences (BK21 Program) and School of Dentistry, Jeonbuk National University, Jeonju, South Korea
| | - Young-Mi Jeon
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences (BK21 Program) and School of Dentistry, Jeonbuk National University, Jeonju, South Korea
| | - Sung-Ho Kook
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, South Korea
| | - Jeong-Chae Lee
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, South Korea.,Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences (BK21 Program) and School of Dentistry, Jeonbuk National University, Jeonju, South Korea
| |
Collapse
|
39
|
Raimondi L, De Luca A, Giavaresi G, Raimondo S, Gallo A, Taiana E, Alessandro R, Rossi M, Neri A, Viglietto G, Amodio N. Non-Coding RNAs in Multiple Myeloma Bone Disease Pathophysiology. Noncoding RNA 2020; 6:ncrna6030037. [PMID: 32916806 PMCID: PMC7549375 DOI: 10.3390/ncrna6030037] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 08/27/2020] [Accepted: 09/08/2020] [Indexed: 12/11/2022] Open
Abstract
Bone remodeling is uncoupled in the multiple myeloma (MM) bone marrow niche, resulting in enhanced osteoclastogenesis responsible of MM-related bone disease (MMBD). Several studies have disclosed the mechanisms underlying increased osteoclast formation and activity triggered by the various cellular components of the MM bone marrow microenvironment, leading to the identification of novel targets for therapeutic intervention. In this regard, recent attention has been given to non-coding RNA (ncRNA) molecules, that finely tune gene expression programs involved in bone homeostasis both in physiological and pathological settings. In this review, we will analyze major signaling pathways involved in MMBD pathophysiology, and report emerging evidence of their regulation by different classes of ncRNAs.
Collapse
Affiliation(s)
- Lavinia Raimondi
- IRCSS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche–SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, 40136 Bologna, Italy; (A.D.L.); (G.G.)
- Correspondence: (L.R.); (N.A.); Tel.: +39-091-6236011 (L.R.); +39-0961-3694159 (N.A.)
| | - Angela De Luca
- IRCSS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche–SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, 40136 Bologna, Italy; (A.D.L.); (G.G.)
| | - Gianluca Giavaresi
- IRCSS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche–SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, 40136 Bologna, Italy; (A.D.L.); (G.G.)
| | - Stefania Raimondo
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (Bi.N.D), Section of Biology and Genetics, University of Palermo, 90133 Palermo, Italy; (S.R.); (R.A.)
| | - Alessia Gallo
- IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Research Department, 90127 Palermo, Italy;
| | - Elisa Taiana
- Department of Oncology and Hemato-oncology, University of Milan, 20122 Milan, Italy; (E.T.); (A.N.)
- Hematology, Fondazione Cà Granda IRCCS Policlinico, 20122 Milan, Italy
| | - Riccardo Alessandro
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (Bi.N.D), Section of Biology and Genetics, University of Palermo, 90133 Palermo, Italy; (S.R.); (R.A.)
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), 90146 Palermo, Italy
| | - Marco Rossi
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (M.R.); (G.V.)
| | - Antonino Neri
- Department of Oncology and Hemato-oncology, University of Milan, 20122 Milan, Italy; (E.T.); (A.N.)
- Hematology, Fondazione Cà Granda IRCCS Policlinico, 20122 Milan, Italy
| | - Giuseppe Viglietto
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (M.R.); (G.V.)
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (M.R.); (G.V.)
- Correspondence: (L.R.); (N.A.); Tel.: +39-091-6236011 (L.R.); +39-0961-3694159 (N.A.)
| |
Collapse
|
40
|
Shu B, Zhao Y, Zhao S, Pan H, Xie R, Yi D, Lu K, Yang J, Xue C, Huang J, Wang J, Zhao D, Xiao G, Wang Y, Chen D. Inhibition of Axin1 in osteoblast precursor cells leads to defects in postnatal bone growth through suppressing osteoclast formation. Bone Res 2020; 8:31. [PMID: 32821442 PMCID: PMC7424530 DOI: 10.1038/s41413-020-0104-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 03/05/2020] [Indexed: 12/11/2022] Open
Abstract
Axin1 is a negative regulator of β-catenin signaling and its role in osteoblast precursor cells remains undefined. In the present studies, we determined changes in postnatal bone growth by deletion of Axin1 in osteoblast precursor cells and analyzed bone growth in newborn and postnatal Axin1Osx mice and found that hypertrophic cartilage area was largely expanded in Axin1Osx KO mice. A larger number of chondrocytes and unabsorbed cartilage matrix were found in the bone marrow cavity of Axin1Osx KO mice. Osteoclast formation in metaphyseal and subchondral bone areas was significantly decreased, demonstrated by decreased TRAP-positive cell numbers, associated with reduction of MMP9- and cathepsin K-positive cell numbers in Axin1Osx KO mice. OPG expression and the ratio of Opg to Rankl were significantly increased in osteoblasts of Axin1Osx KO mice. Osteoclast formation in primary bone marrow derived microphage (BMM) cells was significantly decreased when BMM cells were cultured with conditioned media (CM) collected from osteoblasts derived from Axin1Osx mice compared with BMM cells cultured with CM derived from WT mice. Thus, the loss of Axin1 in osteoblast precursor cells caused increased OPG and the decrease in osteoclast formation, leading to delayed bone growth in postnatal Axin1Osx KO mice.
Collapse
Affiliation(s)
- Bing Shu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Key Laboratory, Ministry of Education of China, 725 WanPing South Road, Shanghai, 200032 China
| | - Yongjian Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Key Laboratory, Ministry of Education of China, 725 WanPing South Road, Shanghai, 200032 China
| | - Shitian Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Key Laboratory, Ministry of Education of China, 725 WanPing South Road, Shanghai, 200032 China
| | - Haobo Pan
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055 China
| | - Rong Xie
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612 USA
| | - Dan Yi
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055 China
| | - Ke Lu
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612 USA
| | - Junjie Yang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Key Laboratory, Ministry of Education of China, 725 WanPing South Road, Shanghai, 200032 China
| | - Chunchun Xue
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Key Laboratory, Ministry of Education of China, 725 WanPing South Road, Shanghai, 200032 China
| | - Jian Huang
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612 USA
| | - Jing Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Key Laboratory, Ministry of Education of China, 725 WanPing South Road, Shanghai, 200032 China
| | - Dongfeng Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Key Laboratory, Ministry of Education of China, 725 WanPing South Road, Shanghai, 200032 China
| | - Guozhi Xiao
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055 China
| | - Yongjun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Key Laboratory, Ministry of Education of China, 725 WanPing South Road, Shanghai, 200032 China
| | - Di Chen
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055 China
| |
Collapse
|
41
|
Kim HN, Ponte F, Nookaew I, Ucer Ozgurel S, Marques-Carvalho A, Iyer S, Warren A, Aykin-Burns N, Krager K, Sardao VA, Han L, de Cabo R, Zhao H, Jilka RL, Manolagas SC, Almeida M. Estrogens decrease osteoclast number by attenuating mitochondria oxidative phosphorylation and ATP production in early osteoclast precursors. Sci Rep 2020; 10:11933. [PMID: 32686739 PMCID: PMC7371870 DOI: 10.1038/s41598-020-68890-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 06/02/2020] [Indexed: 02/08/2023] Open
Abstract
Loss of estrogens at menopause is a major cause of osteoporosis and increased fracture risk. Estrogens protect against bone loss by decreasing osteoclast number through direct actions on cells of the myeloid lineage. Here, we investigated the molecular mechanism of this effect. We report that 17β-estradiol (E2) decreased osteoclast number by promoting the apoptosis of early osteoclast progenitors, but not mature osteoclasts. This effect was abrogated in cells lacking Bak/Bax-two pro-apoptotic members of the Bcl-2 family of proteins required for mitochondrial apoptotic death. FasL has been previously implicated in the pro-apoptotic actions of E2. However, we show herein that FasL-deficient mice lose bone mass following ovariectomy indistinguishably from FasL-intact controls, indicating that FasL is not a major contributor to the anti-osteoclastogenic actions of estrogens. Instead, using microarray analysis we have elucidated that ERα-mediated estrogen signaling in osteoclast progenitors decreases "oxidative phosphorylation" and the expression of mitochondria complex I genes. Additionally, E2 decreased the activity of complex I and oxygen consumption rate. Similar to E2, the complex I inhibitor Rotenone decreased osteoclastogenesis by promoting osteoclast progenitor apoptosis via Bak/Bax. These findings demonstrate that estrogens decrease osteoclast number by attenuating respiration, and thereby, promoting mitochondrial apoptotic death of early osteoclast progenitors.
Collapse
Affiliation(s)
- Ha-Neui Kim
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, 4301 W. Markham St. #587, Little Rock, 72205-7199, USA
| | - Filipa Ponte
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, 4301 W. Markham St. #587, Little Rock, 72205-7199, USA
| | - Intawat Nookaew
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Serra Ucer Ozgurel
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, 4301 W. Markham St. #587, Little Rock, 72205-7199, USA
| | - Adriana Marques-Carvalho
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, UC-Biotech, Biocant Park, Cantanhede, Portugal
| | - Srividhya Iyer
- Department of Orthopedic Surgery, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Aaron Warren
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, 4301 W. Markham St. #587, Little Rock, 72205-7199, USA
| | - Nukhet Aykin-Burns
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Kimberly Krager
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Vilma A Sardao
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, UC-Biotech, Biocant Park, Cantanhede, Portugal
| | - Li Han
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, 4301 W. Markham St. #587, Little Rock, 72205-7199, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, NIA, NIH, Baltimore, MD, USA
| | - Haibo Zhao
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, 4301 W. Markham St. #587, Little Rock, 72205-7199, USA
| | - Robert L Jilka
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, 4301 W. Markham St. #587, Little Rock, 72205-7199, USA
| | - Stavros C Manolagas
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, 4301 W. Markham St. #587, Little Rock, 72205-7199, USA.,Department of Orthopedic Surgery, University of Arkansas for Medical Sciences, Little Rock, USA.,Central Arkansas Veterans Healthcare System, Little Rock, AR, 72205, USA
| | - Maria Almeida
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, 4301 W. Markham St. #587, Little Rock, 72205-7199, USA. .,Department of Orthopedic Surgery, University of Arkansas for Medical Sciences, Little Rock, USA. .,Central Arkansas Veterans Healthcare System, Little Rock, AR, 72205, USA.
| |
Collapse
|
42
|
Villarroel A, Del Valle-Pérez B, Fuertes G, Curto J, Ontiveros N, Garcia de Herreros A, Duñach M. Src and Fyn define a new signaling cascade activated by canonical and non-canonical Wnt ligands and required for gene transcription and cell invasion. Cell Mol Life Sci 2020; 77:919-935. [PMID: 31312879 PMCID: PMC11104847 DOI: 10.1007/s00018-019-03221-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 07/01/2019] [Accepted: 07/05/2019] [Indexed: 01/12/2023]
Abstract
Wnt ligands signal through canonical or non-canonical signaling pathways. Although both routes share common elements, such as the Fz2 receptor, they differ in the co-receptor and in many of the final responses; for instance, whereas canonical Wnts increase β-catenin stability, non-canonical ligands downregulate it. However, both types of ligands stimulate tumor cell invasion. We show here that both the canonical Wnt3a and the non-canonical Wnt5a stimulate Fz2 tyrosine phosphorylation, Fyn binding to Fz2, Fyn activation and Fyn-dependent Stat3 phosphorylation. Wnt3a and Wnt5a require Src for Fz2 tyrosine phosphorylation; Src binds to canonical and non-canonical co-receptors (LRP5/6 and Ror2, respectively) and is activated by Wnt3a and Wnt5a. This Fz2/Fyn/Stat3 branch is incompatible with the classical Fz2/Dvl2 pathway as shown by experiments of over-expression or depletion. Fyn is necessary for transcription of genes associated with invasiveness, such as Snail1, and for activation of cell invasion by both Wnt ligands. Our results extend the knowledge about canonical Wnt pathways, demonstrating additional roles for Fyn in this pathway and describing how this protein kinase is activated by both canonical and non-canonical Wnts.
Collapse
Affiliation(s)
- Aida Villarroel
- Departament de Bioquímica i Biologia Molecular, CEB, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Beatriz Del Valle-Pérez
- Departament de Bioquímica i Biologia Molecular, CEB, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Guillem Fuertes
- Departament de Bioquímica i Biologia Molecular, CEB, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Josué Curto
- Departament de Bioquímica i Biologia Molecular, CEB, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Neus Ontiveros
- Departament de Bioquímica i Biologia Molecular, CEB, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Antonio Garcia de Herreros
- Programa de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Unidad Asociada CSIC, Parc de Recerca Biomèdica de Barcelona, c/Doctor Aiguader 88, 08003, Barcelona, Spain.
- Departament de Ciències, Experimentals i de la Salut, Universitat Pompeu Fabra, 08003, Barcelona, Spain.
| | - Mireia Duñach
- Departament de Bioquímica i Biologia Molecular, CEB, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.
| |
Collapse
|
43
|
Reddy D, Ghosh P, Kumavath R. Strophanthidin Attenuates MAPK, PI3K/AKT/mTOR, and Wnt/β-Catenin Signaling Pathways in Human Cancers. Front Oncol 2020; 9:1469. [PMID: 32010609 PMCID: PMC6978703 DOI: 10.3389/fonc.2019.01469] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 12/09/2019] [Indexed: 12/16/2022] Open
Abstract
Lung cancer is the most prevalent in cancer-related deaths, while breast carcinoma is the second most dominant cancer in women, accounting for the most number of deaths worldwide. Cancers are heterogeneous diseases that consist of several subtypes based on the presence or absence of hormone receptors and human epidermal growth factor receptor 2. Several drugs have been developed targeting cancer biomarkers; nonetheless, their efficiency are not adequate due to the high reemergence rate of cancers and fundamental or acquired resistance toward such drugs, which leads to partial therapeutic possibilities. Recent studies on cardiac glycosides (CGs) positioned them as potent cytotoxic agents that target multiple pathways to initiate apoptosis and autophagic cell death in many cancers. In the present study, our aim is to identify the anticancer activity of a naturally available CG (strophanthidin) in human breast (MCF-7), lung (A549), and liver cancer (HepG2) cells. Our results demonstrate a dose-dependent cytotoxic effect of strophanthidin in MCF-7, A549, and HepG2 cells, which was further supported by DNA damage on drug treatment. Strophanthidin arrested the cell cycle at the G2/M phase; this effect was further validated by checking the inhibited expressions of checkpoint and cyclin-dependent kinases in strophanthidin-induced cells. Moreover, strophanthidin inhibited the expression of several key proteins such as MEK1, PI3K, AKT, mTOR, Gsk3α, and β-catenin from MAPK, PI3K/AKT/mTOR, and Wnt/β-catenin signaling. The current study adequately exhibits the role of strophanthidin in modulating the expression of various key proteins involved in cell cycle arrest, apoptosis, and autophagic cell death. Our in silico studies revealed that strophanthidin can interact with several key proteins from various pathways. Taken together, this study demonstrates the viability of strophanthidin as a promising anticancer agent, which may serve as a new anticancer drug.
Collapse
Affiliation(s)
- Dhanasekhar Reddy
- Department of Genomic Science, School of Biological Sciences, Central University of Kerala, Kasaragod, India
| | - Preetam Ghosh
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA, United States
| | - Ranjith Kumavath
- Department of Genomic Science, School of Biological Sciences, Central University of Kerala, Kasaragod, India
| |
Collapse
|
44
|
Roberts JL, Liu G, Paglia DN, Kinter CW, Fernandes LM, Lorenzo J, Hansen MF, Arif A, Drissi H. Deletion of
Wnt5a
in osteoclasts results in bone loss through decreased bone formation. Ann N Y Acad Sci 2020; 1463:45-59. [DOI: 10.1111/nyas.14293] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/25/2019] [Accepted: 12/04/2019] [Indexed: 12/29/2022]
Affiliation(s)
- Joseph L. Roberts
- Department of Orthopaedics Emory University School of Medicine Atlanta Georgia
| | - Guanglu Liu
- Department of Orthopaedics Emory University School of Medicine Atlanta Georgia
| | - David N. Paglia
- Department of Orthopaedics, New Jersey Medical School Rutgers University Newark New Jersey
| | | | | | - Joseph Lorenzo
- Department of Medicine and Department of Orthopaedic Surgery University of Connecticut Health Farmington Connecticut
| | - Marc F. Hansen
- Center for Molecular Medicine University of Connecticut Health Farmington Connecticut
| | - Abul Arif
- Department of Orthopaedics Emory University School of Medicine Atlanta Georgia
| | - Hicham Drissi
- Department of Orthopaedics Emory University School of Medicine Atlanta Georgia
| |
Collapse
|
45
|
Yang J, Li J, Cui X, Li W, Xue Y, Shang P, Zhang H. Blocking glucocorticoid signaling in osteoblasts and osteocytes prevents mechanical unloading-induced cortical bone loss. Bone 2020; 130:115108. [PMID: 31704341 DOI: 10.1016/j.bone.2019.115108] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 09/25/2019] [Accepted: 10/10/2019] [Indexed: 12/13/2022]
Abstract
Bone loss has been supposed to be the greatest damage to the health of astronauts. It is generally believed that the mechanical unloading induced by microgravity is the main cause of bone loss. However, besides mechanical unloading, many evidences from animal models and spaceflight missions indicate that microgravity conditions can cause some stress reactions and elevated endogenous glucocorticoid (GC) levels. High levels of GCs can lead to bone loss. This study aimed to investigate whether elevated GC levels are involved in hindlimb unloading (HLU)-induced bone loss in mice. Col2.3-11β-hydroxysteroid dehydrogenase type 2 (Col2.3-11β-HSD2) transgenic mice which are characterized by specific blocking GC signaling in mature osteoblasts and osteocytes were used. Male 14-week-old Col2.3-11β-HSD2 transgenic mice and wild type littermates were tail-suspended or kept under ambulatory conditions. At the endpoint, the tibias were examined by micro-computed tomography and histomorphometry, and bone turnover was analyzed by serum biochemistry, histochemistry staining, immunohistochemistry, and real-time PCR. Mice exposed to unloading occurred a significant increase in serum GC concentrations. Compared with non-unloaded controls, HLU led to a severe damage in cortical bone microstructure and bone strength of the tibia in wild type mice but not transgenic littermates. Osteoblast activity and bone formation were inhibited, whereas osteoclast activity and bone resorption were promoted in the tibial cortical bone of wild type mice following HLU, features absented in transgenic mice. Furthermore, HLU resulted in a significant increase in the number of sclerostin-producing and receptor activator of nuclear factor-κ B ligand (RANKL)-positive osteocytes, and apoptotic osteoblasts and osteocytes in wild type mice of unloading but not in unloaded transgenic mice. In conclusion, cortical bone loss during HLU is mediated through enhancing GC signaling in osteoblasts and osteocytes and subsequently restraining bone formation and activating bone resorption. It suggests that elevated GC levels play an important role in cortical bone loss in response to mechanical unloading.
Collapse
Affiliation(s)
- Jiancheng Yang
- Department of Spinal Surgery, People's Hospital of Longhua Shenzhen, Shenzhen, China; School of Life Sciences, Northwestern Polytechnical University, Xi'an, China; Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, China
| | - Jingbao Li
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China; Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, China
| | - Xiaobin Cui
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China; Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, China
| | - Wenbin Li
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China; Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, China
| | - Yanru Xue
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China; Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, China
| | - Peng Shang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China; Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, China; Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, China.
| | - Hao Zhang
- Department of Spinal Surgery, People's Hospital of Longhua Shenzhen, Shenzhen, China.
| |
Collapse
|
46
|
Zhou Y, Shi WY, He W, Yan ZW, Liu MH, Chen J, Yang YS, Wang YQ, Chen GQ, Huang Y. FAM122A supports the growth of hepatocellular carcinoma cells and its deletion enhances Doxorubicin-induced cytotoxicity. Exp Cell Res 2019; 387:111714. [PMID: 31711919 DOI: 10.1016/j.yexcr.2019.111714] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 11/02/2019] [Accepted: 11/05/2019] [Indexed: 12/29/2022]
Abstract
FAM122A is a highly conserved protein in mammals, however its function is still largely unknown so far. In this study, we investigated the potential role of FAM122A in hepatocellular carcinoma (HCC). By analyzing HCC patient cohorts from RNA sequencing datasets, we found the expression level of FAM122A mRNA is significantly upregulated in HCC patients. Moreover, this abnormally higher expression pattern of FAM122A protein was also found in partial HCC tumor tissues, compared with the normal parts. Further, we demonstrated that CRISPR/Cas9-mediated FAM122A knockout significantly inhibits the growth, clonogenic potential and xenografts of HCC cells, induces cell cycle arrest and reduces the expression of proliferation-related genes. Interestingly, FAM122A deletion significantly enhances the cytotoxicity effect of Doxorubicin (Dox), a drug used in standard chemotherapy in HCC patients. In contrary, overexpression of FAM122A not only promotes HCC cell growth, but also inhibits Dox-induced DNA damage and cell death. Considering that FAM122A is previously identified as an endogenous inhibitor of PP2A, we asked whether FAM122A regulating HCC cell growth is associated with PP2A. The results showed FAM122A can also modulate PP2A activity in HCC cells although the modulated effect is relatively slight, however, treatment with a PP2A inhibitor okadaic acid did not rescue the inhibitory effects of cell growth and proliferation in FAM122A deletion cells, indicating that FAM122A may support HCC cell growth independent of its ability to modulate PP2A. Collectively, these results suggest that FAM122A is required for maintaining HCC cell growth, and its elimination combined with chemotherapy may represent a potential novel therapeutic strategy for HCC patients.
Collapse
Affiliation(s)
- Yong Zhou
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wen-Yang Shi
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wei He
- Department of Pathology, Ren-Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200027, China
| | - Zhao-Wen Yan
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Man-Hua Liu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jing Chen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yun-Sheng Yang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yin-Qi Wang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Guo-Qiang Chen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ying Huang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
47
|
Sun Q, Li Z, Liu B, Yuan X, Guo S, Helms JA. Improving intraoperative storage conditions for autologous bone grafts: An experimental investigation in mice. J Tissue Eng Regen Med 2019; 13:2169-2180. [PMID: 31617958 DOI: 10.1002/term.2970] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 08/27/2019] [Accepted: 09/26/2019] [Indexed: 11/07/2022]
Affiliation(s)
- Qiang Sun
- Department of Plastic SurgeryThe First Hospital of China Medical University Shenyang China
- Division of Plastic and Reconstructive Surgery, Department of SurgeryStanford School of Medicine Stanford CA
| | - Zhijun Li
- Division of Plastic and Reconstructive Surgery, Department of SurgeryStanford School of Medicine Stanford CA
- Department of OrthopedicsTianjin Medical University General Hospital Tianjin China
| | - Bo Liu
- Ankasa Regenerative Therapeutics, Inc. South San Francisco CA
| | - Xue Yuan
- Division of Plastic and Reconstructive Surgery, Department of SurgeryStanford School of Medicine Stanford CA
| | - Shu Guo
- Department of Plastic SurgeryThe First Hospital of China Medical University Shenyang China
| | - Jill A. Helms
- Division of Plastic and Reconstructive Surgery, Department of SurgeryStanford School of Medicine Stanford CA
- Ankasa Regenerative Therapeutics, Inc. South San Francisco CA
| |
Collapse
|
48
|
Soysa NS, Alles N. Positive and negative regulators of osteoclast apoptosis. Bone Rep 2019; 11:100225. [PMID: 31720316 PMCID: PMC6838739 DOI: 10.1016/j.bonr.2019.100225] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 08/22/2019] [Accepted: 10/02/2019] [Indexed: 12/26/2022] Open
Abstract
Survival and apoptosis are of major importance in the osteoclast life cycle. As osteoclasts have short lifespan, any alteration that prolongs their viability may cause enhanced osteoclast activity. Hence, the regulation of OC apoptosis has been recognized as a critical factor in bone remodeling. An imbalance in bone remodeling due to increased osteoclast activity leads to most adult bone diseases such as osteoporosis, rheumatoid arthritis and multiple myeloma. Therefore, manipulating osteoclast death would be a viable therapeutic approach in ameliorating bone diseases, with accelerated resorption. Over the last few decades we have witnessed the unraveling of many of the intracellular mechanisms responsible for osteoclast apoptosis. Thus, an understanding of the underlying mechanisms by which osteoclasts undergo programmed cell death and the regulators that modulate that activity will undoubtedly provide an insight into the development of pharmacological agents to treat such pathological bone diseases.
Collapse
Affiliation(s)
- Niroshani Surangika Soysa
- Division of Pharmacology, Department of Oral Medicine and Periodontology, Faculty of Dental Sciences, University of Peradeniya, Sri Lanka
| | - Neil Alles
- Department of Biochemistry, Faculty of Medicine, University of Peradeniya, Sri Lanka
| |
Collapse
|
49
|
García de Herreros A, Duñach M. Intracellular Signals Activated by Canonical Wnt Ligands Independent of GSK3 Inhibition and β-Catenin Stabilization. Cells 2019; 8:cells8101148. [PMID: 31557964 PMCID: PMC6829497 DOI: 10.3390/cells8101148] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/19/2019] [Accepted: 09/21/2019] [Indexed: 12/31/2022] Open
Abstract
In contrast to non-canonical ligands, canonical Wnts promote the stabilization of β-catenin, which is a prerequisite for formation of the TCF4/β-catenin transcriptional complex and activation of its target genes. This pathway is initiated by binding of Wnt ligands to the Frizzled/LRP5/6 receptor complex, and it increases the half-life of β-catenin by precluding the phosphorylation of β-catenin by GSK3 and its binding to the βTrCP1 ubiquitin ligase. Other intercellular signals are also activated by Wnt ligands that do not inhibit GSK3 and increase β-catenin protein but that either facilitate β-catenin transcriptional activity or stimulate other transcriptional factors that cooperate with it. In this review, we describe the layers of complexity of these signals and discuss their crosstalk with β-catenin in activation of transcriptional targets.
Collapse
Affiliation(s)
- Antonio García de Herreros
- Programa de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Unidad Asociada CSIC, and Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, E-08003 Barcelona, Spain.
| | - Mireia Duñach
- Departament de Bioquímica i Biologia Molecular, CEB, Facultat de Medicina, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Spain.
| |
Collapse
|
50
|
Kim SM, Kim EM, Ji KY, Lee HY, Yee SM, Woo SM, Yi JW, Yun CH, Choi H, Kang HS. TREM2 Acts as a Tumor Suppressor in Colorectal Carcinoma through Wnt1/ β-catenin and Erk Signaling. Cancers (Basel) 2019; 11:cancers11091315. [PMID: 31489935 PMCID: PMC6770495 DOI: 10.3390/cancers11091315] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 08/30/2019] [Accepted: 09/05/2019] [Indexed: 01/22/2023] Open
Abstract
TREM2 (triggering receptor expressed on myeloid cells) is involved in the development of malignancies. However, the function of TREM2 in colorectal cancer has not been clearly elucidated. Here, we investigated TREM2 function for the first time in colorectal epithelial cancer cells and demonstrated that TREM2 is a novel tumor suppressor in colorectal carcinoma. Blockade of TREM2 significantly promoted the proliferation of HT29 colorectal carcinoma cells by regulating cell cycle-related factors, such as p53 phosphorylation and p21 and cyclin D1 protein levels. HT29 cell migration was also increased by TREM2 inhibition via MMP9 (matrix metalloproteinase 9) expression upregulation. Furthermore, we found that the tumor suppressor effects of TREM2 were associated with Wnt/β-catenin and extracellular signal-regulated kinase (ERK) signaling. Importantly, the effect of TREM2 in the suppression of tumor development was demonstrated by in vivo and in vitro assays, as well as in human colon cancer patient tissue arrays. Overall, our results identify TREM2 as a potential prognostic biomarker and therapeutic target for colorectal cancer.
Collapse
Affiliation(s)
- Su-Man Kim
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Korea.
| | - Eun-Mi Kim
- Korea Institute of Toxicology, Daejeon, 34114, Korea.
| | - Kon-Young Ji
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea.
| | - Hwa-Youn Lee
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 701-310, Korea.
| | - Su-Min Yee
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Korea.
| | - Su-Min Woo
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Korea.
| | - Ja-Woon Yi
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Korea.
| | - Chul-Ho Yun
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Korea.
| | - Harim Choi
- Department of Nursing, Nambu University, Gwangju 506-706, Korea.
| | - Hyung-Sik Kang
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Korea.
| |
Collapse
|