1
|
Mi Y, Wei D, Du B, Zhang R, Li J, Huang S, Zhang B, Ren J, Wu X. Effect of type 2 diabetes mellitus microenvironment on osteogenic capacity of bone marrow mesenchymal stem cells. Int Immunopharmacol 2025; 157:114724. [PMID: 40300360 DOI: 10.1016/j.intimp.2025.114724] [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: 02/27/2025] [Revised: 04/07/2025] [Accepted: 04/21/2025] [Indexed: 05/01/2025]
Abstract
Type 2 diabetes mellitus (T2DM) often leads to delayed bone regeneration such as slow healing of fractures and bone defects. The number, status and osteogenic differentiation capacity of bone marrow mesenchymal stem cells (BMSCs) are extremely important in bone healing and bone regeneration. The T2DM microenvironment can have irreversible negative effects on BMSCs. In this paper, we review the molecular expression and altered proliferation, migration, and osteogenic differentiation capacity of BMSCs in the microenvironment of T2DM, it provides a new perspective to restore the normal function of T2DM-BMSCs, so as to save the damaged bone regeneration capacity.
Collapse
Affiliation(s)
- Yanling Mi
- Shanxi Medical University, School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| | - Danni Wei
- Shanxi Medical University, School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| | - Bingli Du
- Shanxi Medical University, School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| | - Ran Zhang
- Shanxi Medical University, School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| | - Jiadi Li
- Shanxi Medical University, School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| | - Shuo Huang
- Shanxi Medical University, School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| | - Binbin Zhang
- Shanxi Medical University, School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| | - Juan Ren
- Shanxi Medical University, School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China.
| | - Xiuping Wu
- Shanxi Medical University, School and Hospital of Stomatology, Taiyuan 030001, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China.
| |
Collapse
|
2
|
Su S, Bai J, Wang R, Gao S, Zhou R, Zhou F. A novel strategy for bone defect repair: Stromal cell-derived factor 1α sustained-release acellular fish scale scaffolds combined with injection of bone marrow mesenchymal stem cells promote bone regeneration. Mater Today Bio 2025; 32:101759. [PMID: 40270891 PMCID: PMC12017916 DOI: 10.1016/j.mtbio.2025.101759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
Patients with bone defects often have weak cell vitality and differentiation ability of endogenous bone marrow mesenchymal stem cells (BMSCs), which makes bone regeneration face challenges. At present, the bone tissue engineering strategies are mainly to build grafts by loading cells on scaffolds in vitro. These strategies face many difficulties that limit their clinical application. To this end, we developed a new strategy for bone defect repair, namely chemotactic cell-free scaffolds combined with BMSCs injection. We first prepared a polydopamine-functionalized acellular fish scale scaffold that can continuously release stromal cell-derived factor 1α (SDF-1α) (termed as SDF-1α/PAFS) in vivo for at least 10 days. The study results showed that the scaffold not only has excellent mechanical properties and good biocompatibility but also has reactive oxygen scavenging activity, immunomodulation, angiogenesis, and osteogenesis. More importantly, SDF-1α/PAFS can recruit postoperatively injected BMSCs into bone defects for bone repair. We constructed the mouse cranial bone defect model, and in vivo experimental results confirmed that the strategy of combining SDF-1α/PAFS with BMSCs injection can effectively promote bone defect repair. Overall, this study provides a promising strategy for bone defect repair, with better clinical convenience and operability.
Collapse
Affiliation(s)
- Shilong Su
- Department of Orthopedics, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
| | - Jinwu Bai
- Department of Orthopedics, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
| | - Ruideng Wang
- Department of Orthopedics, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
| | - Shan Gao
- Department of Orthopedics, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
| | - Rubing Zhou
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, No.95 Yong'an Road, Xicheng, 100050, Beijing, China
| | - Fang Zhou
- Department of Orthopedics, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
| |
Collapse
|
3
|
Seto T, Yukata K, Tsuji S, Takeshima Y, Honda T, Sakamoto A, Takemoto K, Sakai H, Matsuo M, Sasaki Y, Kaneda M, Yoshimura M, Mihara A, Uehara K, Matsugaki A, Nakano T, Harada K, Tahara Y, Iwaisako K, Yanai R, Takeda N, Sakai T, Asagiri M. Methylglyoxal compromises callus mineralization and impairs fracture healing through suppression of osteoblast terminal differentiation. Biochem Biophys Res Commun 2025; 747:151312. [PMID: 39799866 DOI: 10.1016/j.bbrc.2025.151312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 12/27/2024] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
Impaired fracture healing in diabetic patients leads to prolonged morbidity and increased healthcare costs. Methylglyoxal (MG), a reactive metabolite elevated in diabetes, is implicated in various complications, but its direct impact on bone healing remains unclear. Here, using a non-diabetic murine tibial fracture model, we demonstrate that MG directly impairs fracture healing. Micro-computed tomography revealed decreased volumetric bone mineral density in the callus, while callus volume remained unchanged, resulting in a brittle bone structure. This was accompanied by reduced expression of osteocalcin and bone sialoprotein, both critical for mineralization. Biomechanical analysis indicated that MG reduced the mechanical resilience of the fracture site without altering its elastic strength, suggesting that the impairment was not primarily due to the accumulation of advanced glycation end-products in the bone extracellular matrix. In vitro studies confirmed that non-cytotoxic concentrations of MG inhibited osteoblast maturation and mineralization. Transcriptomic analysis identified downregulation of Osterix, a key transcription factor for osteoblast maturation, without altering Runx2 levels, leading to decreased expression of key mineralization-related factors like osteocalcin. These findings align with clinical observations of reduced circulating osteocalcin levels in diabetic patients, suggesting that the detrimental effects of MG on osteoblasts may extend beyond bone metabolism. Our study highlights MG and MG-sensitive pathways as potential therapeutic targets for improving bone repair in individuals with diabetes and other conditions characterized by elevated MG levels.
Collapse
Affiliation(s)
- Tetsuya Seto
- Department of Pharmacology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan; Department of Orthopedic Surgery, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Kiminori Yukata
- Department of Orthopedic Surgery, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Shunya Tsuji
- Department of Pharmacology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan; Research Institute for Cell Design Medical Science, Yamaguchi University, Yamaguchi, Japan.
| | - Yusuke Takeshima
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Takeshi Honda
- Department of Pharmacology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Akihiko Sakamoto
- Department of Pharmacology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Kenji Takemoto
- Department of Pharmacology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Hiroki Sakai
- Department of Pharmacology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Mayu Matsuo
- Department of Pharmacology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Yurika Sasaki
- Department of Pharmacology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Mizuki Kaneda
- Department of Pharmacology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Mikako Yoshimura
- Department of Pharmacology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Atsushi Mihara
- Department of Orthopedic Surgery, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Kazuya Uehara
- Department of Pharmacology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan; Department of Orthopedic Surgery, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Aira Matsugaki
- Division of Materials and Manufacturing Science, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Takayoshi Nakano
- Division of Materials and Manufacturing Science, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Koji Harada
- Department of Nursing, Faculty of Health Sciences, Hiroshima Cosmopolitan University, Hiroshima, Japan
| | - Yoshiro Tahara
- Department of Chemical Engineering and Materials Science, Doshisha University, Kyoto, Japan
| | - Keiko Iwaisako
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan; Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyoto, Japan
| | - Ryoji Yanai
- Department of Ophthalmology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Norihiko Takeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takashi Sakai
- Department of Orthopedic Surgery, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Masataka Asagiri
- Department of Pharmacology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan; Research Institute for Cell Design Medical Science, Yamaguchi University, Yamaguchi, Japan.
| |
Collapse
|
4
|
Nasadiuk K, Kolanowski T, Kowalewski C, Wozniak K, Oldak T, Rozwadowska N. Harnessing Mesenchymal Stromal Cells for Advanced Wound Healing: A Comprehensive Review of Mechanisms and Applications. Int J Mol Sci 2024; 26:199. [PMID: 39796055 PMCID: PMC11719717 DOI: 10.3390/ijms26010199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/20/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Chronic wounds and injuries remain a substantial healthcare challenge, with significant burdens on patient quality of life and healthcare resources. Mesenchymal stromal cells (MSCs) present an innovative approach to enhance tissue repair and regeneration in the context of wound healing. The intrinsic presence of MSCs in skin tissue, combined with their roles in wound repair, ease of isolation, broad secretory profile, and low immunogenicity, makes them especially promising for treating chronic wounds. This review explores the current landscape of MSC application, focusing on preclinical and clinical data across chronic wounds, diabetic ulcers, burns, non-union bone fractures, lower extremity venous ulcers, pressure ulcers, and genetic skin conditions like epidermolysis bullosa. Special emphasis is given to the mechanisms through which MSCs exert their regenerative effects, underscoring their potential in advancing wound healing therapies and supporting the broader field of regenerative medicine.
Collapse
Affiliation(s)
- Khrystyna Nasadiuk
- Research and Development Department, Polski Bank Komórek Macierzystych S.A. (FamiCord Group), 00-867 Warsaw, Poland; (K.N.); (T.K.)
| | - Tomasz Kolanowski
- Research and Development Department, Polski Bank Komórek Macierzystych S.A. (FamiCord Group), 00-867 Warsaw, Poland; (K.N.); (T.K.)
- Institute of Human Genetics, Polish Academy of Sciences, 60-479 Poznan, Poland
| | - Cezary Kowalewski
- Department of Dermatology, National Medical Institute of the Ministry of the Interior and Administration, 02-507 Warsaw, Poland; (C.K.); (K.W.)
| | - Katarzyna Wozniak
- Department of Dermatology, National Medical Institute of the Ministry of the Interior and Administration, 02-507 Warsaw, Poland; (C.K.); (K.W.)
| | - Tomasz Oldak
- Research and Development Department, Polski Bank Komórek Macierzystych S.A. (FamiCord Group), 00-867 Warsaw, Poland; (K.N.); (T.K.)
| | - Natalia Rozwadowska
- Institute of Human Genetics, Polish Academy of Sciences, 60-479 Poznan, Poland
| |
Collapse
|
5
|
Joladarashi D, Thej C, Mallaredy V, Magadum A, Cimini M, Gonzalez C, Truongcao M, Nigro JT, Sethi MK, Gibb AA, Benedict C, Koch WJ, Kishore R. GPC3-mediated metabolic rewiring of diabetic mesenchymal stromal cells enhances their cardioprotective functions via PKM2 activation. iScience 2024; 27:111021. [PMID: 39429777 PMCID: PMC11490746 DOI: 10.1016/j.isci.2024.111021] [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: 05/03/2024] [Revised: 08/02/2024] [Accepted: 09/20/2024] [Indexed: 10/22/2024] Open
Abstract
Mesenchymal stromal cells (MSC) are promising stem cell therapy for treating cardiovascular and other degenerative diseases. Diabetes affects the functional capability of MSC and impedes cell-based therapy. Despite numerous studies, the impact of diabetes on MSC myocardial reparative activity, metabolic fingerprint, and the mechanism of dysfunction remains inadequately perceived. We demonstrated that the transplantation of diabetic-MSC (db/db-MSC) into the ischemic myocardium of mice does not confer cardiac benefit post-MI. Metabolomic studies identified defective energy metabolism in db/db-MSC. Furthermore, we found that glypican-3 (GPC3), a heparan sulfate proteoglycan, is highly upregulated in db/db-MSC and is involved in metabolic alterations in db/db-MSC via pyruvate kinase M2 (PKM2) activation. GPC3-knockdown reprogrammed-db/db-MSC restored their energy metabolic rates, immunomodulation, angiogenesis, and cardiac reparative activities. Together, these data indicate that GPC3-metabolic reprogramming in diabetic MSC may represent a strategy to enhance MSC-based therapeutics for myocardial repair in diabetic patients.
Collapse
Affiliation(s)
- Darukeshwara Joladarashi
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Charan Thej
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Vandana Mallaredy
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Ajit Magadum
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Maria Cimini
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Carolina Gonzalez
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - May Truongcao
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Joseph T. Nigro
- Center for Biomedical Mass Spectrometry, Department of Biochemistry & Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Manveen K. Sethi
- Center for Biomedical Mass Spectrometry, Department of Biochemistry & Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Andrew A. Gibb
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville, 580 South Preston Street, Louisville, KY, USA
| | - Cindy Benedict
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Walter J. Koch
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Raj Kishore
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
- Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
6
|
Wang X, He W, Huang H, Han J, Wang R, Li H, Long Y, Wang G, Han X. Recent Advances in Hydrogel Technology in Delivering Mesenchymal Stem Cell for Osteoarthritis Therapy. Biomolecules 2024; 14:858. [PMID: 39062572 PMCID: PMC11274544 DOI: 10.3390/biom14070858] [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: 05/28/2024] [Revised: 07/06/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Osteoarthritis (OA), a chronic joint disease affecting over 500 million individuals globally, is characterized by the destruction of articular cartilage and joint inflammation. Conventional treatments are insufficient for repairing damaged joint tissue, necessitating novel therapeutic approaches. Mesenchymal stem cells (MSCs), with their potential for differentiation and self-renewal, hold great promise as a treatment for OA. However, challenges such as MSC viability and apoptosis in the ischemic joint environment hinder their therapeutic effectiveness. Hydrogels with biocompatibility and degradability offer a three-dimensional scaffold that support cell viability and differentiation, making them ideal for MSC delivery in OA treatment. This review discusses the pathological features of OA, the properties of MSCs, the challenges associated with MSC therapy, and methods for hydrogel preparation and functionalization. Furthermore, it highlights the advantages of hydrogel-based MSC delivery systems while providing insights into future research directions and the clinical potential of this approach.
Collapse
Affiliation(s)
- Xiangjiang Wang
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China; (X.W.); (W.H.); (J.H.); (R.W.); (H.L.); (Y.L.)
| | - Wentao He
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China; (X.W.); (W.H.); (J.H.); (R.W.); (H.L.); (Y.L.)
| | - Hao Huang
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Collage of Physics and Optoelectronics Engineering, Shenzhen University, Shenzhen 518060, China;
| | - Jiali Han
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China; (X.W.); (W.H.); (J.H.); (R.W.); (H.L.); (Y.L.)
| | - Ruren Wang
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China; (X.W.); (W.H.); (J.H.); (R.W.); (H.L.); (Y.L.)
| | - Hongyi Li
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China; (X.W.); (W.H.); (J.H.); (R.W.); (H.L.); (Y.L.)
| | - Ying Long
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China; (X.W.); (W.H.); (J.H.); (R.W.); (H.L.); (Y.L.)
| | - Guiqing Wang
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China; (X.W.); (W.H.); (J.H.); (R.W.); (H.L.); (Y.L.)
| | - Xianjing Han
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China; (X.W.); (W.H.); (J.H.); (R.W.); (H.L.); (Y.L.)
| |
Collapse
|
7
|
Wang X, Xiang C, Huang C, Cheng H, Zhou Z, Zhang J, Xie H. The treatment efficacy of bone tissue engineering strategy for repairing segmental bone defects under diabetic condition. Front Bioeng Biotechnol 2024; 12:1379679. [PMID: 38737542 PMCID: PMC11082311 DOI: 10.3389/fbioe.2024.1379679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/01/2024] [Indexed: 05/14/2024] Open
Abstract
Background Diabetes mellitus is a systematic disease which exert detrimental effect on bone tissue. The repair and reconstruction of bone defects in diabetic patients still remain a major clinical challenge. This study aims to investigate the potential of bone tissue engineering approach to improve bone regeneration under diabetic condition. Methods In the present study, decalcified bone matrix (DBM) scaffolds were seeded with allogenic fetal bone marrow-derived mesenchymal stem cells (BMSCs) and cultured in osteogenic induction medium to fabricate BMSC/DBM constructs. Then the BMSC/DBM constructs were implanted in both subcutaneous pouches and large femoral bone defects in diabetic (BMSC/DBM in DM group) and non-diabetic rats (BMSC/DBM in non-DM group), cell-free DBM scaffolds were implanted in diabetic rats to serve as the control group (DBM in DM group). X-ray, micro-CT and histological analyses were carried out to evaluate the bone regenerative potential of BMSC/DBM constructs under diabetic condition. Results In the rat subcutaneous implantation model, quantitative micro-CT analysis demonstrated that BMSC/DBM in DM group showed impaired bone regeneration activity compared with the BMSC/DBM in non-DM group (bone volume: 46 ± 4.4 mm3 vs 58.9 ± 7.15 mm3, *p < 0.05). In the rat femoral defect model, X-ray examination demonstrated that bone union was delayed in BMSC/DBM in DM group compared with BMSC/DBM in non-DM group. However, quantitative micro-CT analysis showed that after 6 months of implantation, there was no significant difference in bone volume and bone density between the BMSC/DBM in DM group (199 ± 63 mm3 and 593 ± 65 mg HA/ccm) and the BMSC/DBM in non-DM group (211 ± 39 mm3 and 608 ± 53 mg HA/ccm). Our data suggested that BMSC/DBM constructs could repair large bone defects in diabetic rats, but with delayed healing process compared with non-diabetic rats. Conclusion Our study suggest that biomaterial sacffolds seeded with allogenic fetal BMSCs represent a promising strategy to induce and improve bone regeneration under diabetic condition.
Collapse
Affiliation(s)
- Xiangsheng Wang
- Department of Plastic Surgery, Jingshan Union Hospital, Union Hospital, Huazhong University of Science and Technology, Hubei, China
- Department of Plastic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Can Xiang
- Department of Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chunhua Huang
- Department of Plastic Surgery, Jingshan Union Hospital, Union Hospital, Huazhong University of Science and Technology, Hubei, China
| | - Hanxiao Cheng
- Department of Plastic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhentao Zhou
- Department of Plastic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jufang Zhang
- Department of Plastic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hui Xie
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
8
|
Mateen MA, Alaagib N, Haider KH. High glucose microenvironment and human mesenchymal stem cell behavior. World J Stem Cells 2024; 16:237-244. [PMID: 38577235 PMCID: PMC10989287 DOI: 10.4252/wjsc.v16.i3.237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 01/11/2024] [Accepted: 01/29/2024] [Indexed: 03/25/2024] Open
Abstract
High glucose (HG) culture conditions in vitro and persistent exposure to hyperglycemia in diabetes patients are detrimental to stem cells, analogous to any other cell type in our body. It interferes with diverse signaling pathways, i.e. mammalian target of rapamycin (mTOR)-phosphoinositide 3-kinase (PI3K)-Akt signaling, to impact physiological cellular functions, leading to low cell survival and higher cell apoptosis rates. While elucidating the underlying mechanism responsible for the apoptosis of adipose tissue-derived mesenchymal stem cells (MSCs), a recent study has shown that HG culture conditions dysregulate mTOR-PI3K-Akt signaling in addition to mitochondrial malfunctioning due to defective mitochondrial membrane potential (MtMP) that lowers ATP production. This organelle-level dysfunction energy-starves the cells and increases oxidative stress and ultrastructural abnormalities. Disruption of the mitochondrial electron transport chain produces an altered mitochondrial NAD+/NADH redox state as evidenced by a low NAD+/NADH ratio that primarily contributes to the reduced cell survival in HG. Some previous studies have also reported altered mitochondrial membrane polarity (causing hyperpolarization) and reduced mitochondrial cell mass, leading to perturbed mitochondrial homeostasis. The hostile microenvironment created by HG exposure creates structural and functional changes in the mitochondria, altering their bioenergetics and reducing their capacity to produce ATP. These are significant data, as MSCs are extensively studied for tissue regeneration and restoring their normal functioning in cell-based therapy. Therefore, MSCs from hyperglycemic donors should be cautiously used in clinical settings for cell-based therapy due to concerns of their poor survival rates and increased rates of post engraftment proliferation. As hyperglycemia alters the bioenergetics of donor MSCs, rectifying the loss of MtMP may be an excellent target for future research to restore the normal functioning of MSCs in hyperglycemic patients.
Collapse
Affiliation(s)
| | | | - Khawaja Husnain Haider
- Cellular and Molecular Pharmacology, Sulaiman AlRajhi Medical School, Al Bukairiyah 51941, Saudi Arabia.
| |
Collapse
|
9
|
Zhang G, Song S, Chen Z, Liu X, Zheng J, Wang Y, Chen X, Song Y. Inhibition of PTEN promotes osteointegration of titanium implants in type 2 diabetes by enhancing anti-inflammation and osteogenic capacity of adipose-derived stem cells. Front Bioeng Biotechnol 2024; 12:1358802. [PMID: 38425992 PMCID: PMC10902433 DOI: 10.3389/fbioe.2024.1358802] [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: 12/20/2023] [Accepted: 02/02/2024] [Indexed: 03/02/2024] Open
Abstract
Background: The low osteogenic differentiation potential and attenuated anti-inflammatory effect of adipose-derived stem cells (ADSCs) from animals with type 2 diabetes mellitus (T2DM) limits osseointegration of the implant. However, the underlying mechanisms are not fully understood. Methods: Western blotting and qRT-PCR analyses were performed to investigate the effects of PTEN on the osteogenic capacity of ADSCs of T2DM rats (TADSCs). We conducted animal experiments in T2DM-Sprague Dawley (SD) rats to evaluate the osteogenic capacity of modified TADSC sheets in vivo. New bone formation was assessed by micro-CT and histological analyses. Results: In this study, adipose-derived stem cells of T2DM rats exhibited an impaired osteogenic capacity. RNA-seq analysis showed that PTEN mRNA expression was upregulated in TADSCs, which attenuated the osteogenic capacity of TADSCs by inhibiting the AKT/mTOR/HIF-1α signaling pathway. miR-140-3p, which inhibits PTEN, was suppressed in TADSCs. Overexpression or inhibition of PTEN could correspondingly reduce or enhance the osteogenic ability of TADSCs by regulating the AKT/mTOR/HIF-1α signaling pathway. TADSCs transfected with PTEN siRNA resulted in higher and lower expressions of genes encoded in M2 macrophages (Arg1) and M1 macrophages (iNOS), respectively. In the T2DM rat model, PTEN inhibition in TADSC sheets promoted macrophage polarization toward the M2 phenotype, attenuated inflammation, and enhanced osseointegration around implants. Conclusion: Upregulation of PTEN, which was partially due to the inhibition of miR-140-3p, is important for the attenuated osteogenesis by TADSCs owing to the inhibition of the AKT/mTOR/HIF-1α signaling pathway. Inhibition of PTEN significantly improves the anti-inflammatory effect and osteogenic capacity of TADSCs, thus promoting peri-implant bone formation in T2DM rats. Our findings offer a potential therapeutic approach for modifying stem cells derived from patients with T2DM to enhance osseointegration.
Collapse
Affiliation(s)
- Guanhua Zhang
- Department of Implant Dentistry, School of Stomatology, State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Shuang Song
- College of Stomatology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, China
| | - Zijun Chen
- Department of Implant Dentistry, School of Stomatology, State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Xiangdong Liu
- Department of Implant Dentistry, School of Stomatology, State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Jian Zheng
- Department of Implant Dentistry, School of Stomatology, State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yuxi Wang
- Department of Implant Dentistry, School of Stomatology, State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Xutao Chen
- Department of Implant Dentistry, School of Stomatology, State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi’an, Shaanxi, China
- Department of Immunology, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yingliang Song
- Department of Implant Dentistry, School of Stomatology, State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi’an, Shaanxi, China
| |
Collapse
|
10
|
Ren Y, Zhang C, Liu Y, Kong W, Yang X, Niu H, Qiang L, Yang H, Yang F, Wang C, Wang J. Advances in 3D Printing of Highly Bioadaptive Bone Tissue Engineering Scaffolds. ACS Biomater Sci Eng 2024; 10:255-270. [PMID: 38118130 DOI: 10.1021/acsbiomaterials.3c01129] [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] [Indexed: 12/22/2023]
Abstract
The number of patients with bone defects caused by trauma, bone tumors, and osteoporosis has increased considerably. The repair of irregular, recurring, and large bone defects poses a great challenge to clinicians. Bone tissue engineering is emerging as an appropriate strategy to replace autologous bone grafting in the repair of critically sized bone defects. However, the suitability of bone tissue engineering scaffolds in terms of structure, mechanics, degradation, and the microenvironment is inadequate. Three-dimensional (3D) printing is an advanced additive-manufacturing technology widely used for bone repair. 3D printing constructs personalized structurally adapted scaffolds based on 3D models reconstructed from CT images. The contradiction between the mechanics and degradation is resolved by altering the stacking structure. The local microenvironment of the implant is improved by designing an internal pore structure and a spatiotemporal factor release system. Therefore, there has been a boom in the 3D printing of personalized bone repair scaffolds. In this review, successful research on the preparation of highly bioadaptive bone tissue engineering scaffolds using 3D printing is presented. The mechanisms of structural, mechanical, degradation, and microenvironmental adaptations of bone prostheses and their interactions were elucidated to provide a feasible strategy for constructing highly bioadaptive bone tissue engineering scaffolds.
Collapse
Affiliation(s)
- Ya Ren
- School of Rehabilitation Medicine, Weifang Medical University, Shandong 261041, China
- Southwest JiaoTong University College of Medicine, No. 111 North first Section of Second Ring Road, Chengdu 610036, China
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
| | - Changru Zhang
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
| | - Yihao Liu
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
| | - Weiqing Kong
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao 266000, Shandong Province, China
| | - Xue Yang
- Southwest JiaoTong University College of Medicine, No. 111 North first Section of Second Ring Road, Chengdu 610036, China
| | - Haoyi Niu
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
| | - Lei Qiang
- Key Laboratory of Advanced Technologies of Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Han Yang
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Fei Yang
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
| | - Chengwei Wang
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
| | - Jinwu Wang
- School of Rehabilitation Medicine, Weifang Medical University, Shandong 261041, China
- Southwest JiaoTong University College of Medicine, No. 111 North first Section of Second Ring Road, Chengdu 610036, China
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China
| |
Collapse
|
11
|
Maisenbacher TC, Ehnert S, Histing T, Nüssler AK, Menger MM. Advantages and Limitations of Diabetic Bone Healing in Mouse Models: A Narrative Review. Biomedicines 2023; 11:3302. [PMID: 38137522 PMCID: PMC10741210 DOI: 10.3390/biomedicines11123302] [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: 10/30/2023] [Revised: 11/29/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Diabetes represents a major risk factor for impaired fracture healing. Type 2 diabetes mellitus is a growing epidemic worldwide, hence an increase in diabetes-related complications in fracture healing can be expected. However, the underlying mechanisms are not yet completely understood. Different mouse models are used in preclinical trauma research for fracture healing under diabetic conditions. The present review elucidates and evaluates the characteristics of state-of-the-art murine diabetic fracture healing models. Three major categories of murine models were identified: Streptozotocin-induced diabetes models, diet-induced diabetes models, and transgenic diabetes models. They all have specific advantages and limitations and affect bone physiology and fracture healing differently. The studies differed widely in their diabetic and fracture healing models and the chosen models were evaluated and discussed, raising concerns in the comparability of the current literature. Researchers should be aware of the presented advantages and limitations when choosing a murine diabetes model. Given the rapid increase in type II diabetics worldwide, our review found that there are a lack of models that sufficiently mimic the development of type II diabetes in adult patients over the years. We suggest that a model with a high-fat diet that accounts for 60% of the daily calorie intake over a period of at least 12 weeks provides the most accurate representation.
Collapse
Affiliation(s)
- Tanja C. Maisenbacher
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, BG Clinic Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany; (T.H.); (M.M.M.)
- Siegfried Weller Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany; (S.E.); (A.K.N.)
| | - Sabrina Ehnert
- Siegfried Weller Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany; (S.E.); (A.K.N.)
| | - Tina Histing
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, BG Clinic Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany; (T.H.); (M.M.M.)
| | - Andreas K. Nüssler
- Siegfried Weller Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany; (S.E.); (A.K.N.)
| | - Maximilian M. Menger
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, BG Clinic Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany; (T.H.); (M.M.M.)
| |
Collapse
|
12
|
Sheng N, Xing F, Wang J, Zhang QY, Nie R, Li-Ling J, Duan X, Xie HQ. Recent progress in bone-repair strategies in diabetic conditions. Mater Today Bio 2023; 23:100835. [PMID: 37928253 PMCID: PMC10623372 DOI: 10.1016/j.mtbio.2023.100835] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 10/02/2023] [Accepted: 10/14/2023] [Indexed: 11/07/2023] Open
Abstract
Bone regeneration following trauma, tumor resection, infection, or congenital disease is challenging. Diabetes mellitus (DM) is a metabolic disease characterized by hyperglycemia. It can result in complications affecting multiple systems including the musculoskeletal system. The increased number of diabetes-related fractures poses a great challenge to clinical specialties, particularly orthopedics and dentistry. Various pathological factors underlying DM may directly impair the process of bone regeneration, leading to delayed or even non-union of fractures. This review summarizes the mechanisms by which DM hampers bone regeneration, including immune abnormalities, inflammation, reactive oxygen species (ROS) accumulation, vascular system damage, insulin/insulin-like growth factor (IGF) deficiency, hyperglycemia, and the production of advanced glycation end products (AGEs). Based on published data, it also summarizes bone repair strategies in diabetic conditions, which include immune regulation, inhibition of inflammation, reduction of oxidative stress, promotion of angiogenesis, restoration of stem cell mobilization, and promotion of osteogenic differentiation, in addition to the challenges and future prospects of such approaches.
Collapse
Affiliation(s)
- Ning Sheng
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Fei Xing
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Jie Wang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Qing-Yi Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Rong Nie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Jesse Li-Ling
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
- Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Xin Duan
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Hui-Qi Xie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
- Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China
| |
Collapse
|
13
|
Wang H, Yu H, Huang T, Wang B, Xiang L. Hippo-YAP/TAZ signaling in osteogenesis and macrophage polarization: Therapeutic implications in bone defect repair. Genes Dis 2023; 10:2528-2539. [PMID: 37554194 PMCID: PMC10404961 DOI: 10.1016/j.gendis.2022.12.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/16/2022] [Accepted: 12/08/2022] [Indexed: 01/18/2023] Open
Abstract
Bone defects caused by diseases or surgery are a common clinical problem. Researchers are devoted to finding biological mechanisms that accelerate bone defect repair, which is a complex and continuous process controlled by many factors. As members of transcriptional costimulatory molecules, Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) play an important regulatory role in osteogenesis, and they affect cell function by regulating the expression of osteogenic genes in osteogenesis-related cells. Macrophages are an important group of cells whose function is regulated by YAP/TAZ. Currently, the relationship between YAP/TAZ and macrophage polarization has attracted increasing attention. In bone tissue, YAP/TAZ can realize diverse osteogenic regulation by mediating macrophage polarization. Macrophages polarize into M1 and M2 phenotypes under different stimuli. M1 macrophages dominate the inflammatory response by releasing a number of inflammatory mediators in the early phase of bone defect repair, while massive aggregation of M2 macrophages is beneficial for inflammation resolution and tissue repair, as they secrete many anti-inflammatory and osteogenesis-related cytokines. The mechanism of YAP/TAZ-mediated macrophage polarization during osteogenesis warrants further study and it is likely to be a promising strategy for bone defect repair. In this article, we review the effect of Hippo-YAP/TAZ signaling and macrophage polarization on bone defect repair, and highlight the regulation of macrophage polarization by YAP/TAZ.
Collapse
Affiliation(s)
- Haochen Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hui Yu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Tianyu Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Bin Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lin Xiang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
14
|
Lao A, Wu J, Li D, Shen A, Li Y, Zhuang Y, Lin K, Wu J, Liu J. Functionalized Metal-Organic Framework-Modified Hydrogel That Breaks the Vicious Cycle of Inflammation and ROS for Repairing of Diabetic Bone Defects. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206919. [PMID: 37183293 DOI: 10.1002/smll.202206919] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/24/2023] [Indexed: 05/16/2023]
Abstract
The regeneration of diabetic bone defects remains challenging. Hyperglycemia causes inflammation state and excessive reactive oxygen species (ROS) during bone regeneration period. These two effects reinforce one another and create an endless loop that is also accompanied by mitochondrial dysfunction. However, there is still no effective and inclusive method targeting at the two aspects and breaking the vicious cycle. Herein, nanoparticles-Met@ZIF-8(metformin loaded zeolitic imidazolate frameworks) modified hydrogel that is capable of releasing metformin and Zn elements are constructed. This hydrogel treats hyperglycemia while also controlling mitochondrial function, reducing inflammation, and restoring homeostasis. In addition, the synergetic effect from metformin and Zn ions inhibits ROS-inflammation cascade generation and destroys the continuous progress by taking effects in both ROS and inflammation and further keeping organelles' homeostasis. Furthermore, with the recovery of mitochondria and breakdown of the ROS-inflammation cascade cycle, osteogenesis under a diabetic microenvironment is enhanced in vivo and in vitro. In conclusion, the study provides critical insight into the biological mechanism and potential therapy for diabetic bone regeneration.
Collapse
Affiliation(s)
- An Lao
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200120, China
- Department of Stomatology, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jiaqing Wu
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200120, China
| | - Dejian Li
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200120, China
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201301, China
| | - Aili Shen
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200120, China
| | - Yaxin Li
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200120, China
| | - Yu Zhuang
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200120, China
| | - Kaili Lin
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200120, China
| | - Jianyong Wu
- Department of Stomatology, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jiaqiang Liu
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200120, China
| |
Collapse
|
15
|
Chinipardaz Z, Yuan G, Liu M, Graves DT, Yang S. Diabetes impairs fracture healing through Foxo1 mediated disruption of ciliogenesis. Cell Death Discov 2023; 9:299. [PMID: 37591875 PMCID: PMC10435563 DOI: 10.1038/s41420-023-01562-3] [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: 02/08/2023] [Revised: 05/12/2023] [Accepted: 07/17/2023] [Indexed: 08/19/2023] Open
Abstract
Foxo1 upregulation is linked to defective fracture healing under diabetic conditions. Previous studies demonstrated that diabetes upregulates Foxo1 expression and activation and diabetes impairs ciliogenesis resulting in defective fracture repair. However, the mechanism by which diabetes causes cilia loss during fracture healing remains elusive. We report here that streptozotocin (STZ)-induced type 1 diabetes mellitus (T1DM) dramatically increased Foxo1 expression in femoral fracture calluses, which thereby caused a significant decrease in the expression of IFT80 and primary cilia number. Ablation of Foxo1 in osteoblasts in OSXcretTAFoxo1f/f mice rescued IFT80 expression and ciliogenesis and restored bone formation and mechanical strength in diabetic fracture calluses. In vitro, advanced glycation end products (AGEs) impaired cilia formation in osteoblasts and reduced the production of a mineralizing matrix, which were rescued by Foxo1 deletion. Mechanistically, AGEs increased Foxo1 expression and transcriptional activity to inhibit IFT80 expression causing impaired cilia formation. Thus, our findings demonstrate that diabetes impairs fracture healing through Foxo1 mediated inhibition of ciliary IFT80 expression and primary cilia formation, resulting in impaired osteogenesis. Inhibition of Foxo1 and/or restoration of cilia formation has the potential to promote diabetes-impaired fracture healing.
Collapse
Affiliation(s)
- Zahra Chinipardaz
- Department of Basic and Translation Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Periodontology, Tufts University School of Dental Medicine, Boston, MA, 02111, USA
| | - Gongsheng Yuan
- Department of Basic and Translation Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Min Liu
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dana T Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Shuying Yang
- Department of Basic and Translation Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Center for Innovation & Precision Dentistry, School of Dental Medicine, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- The Penn Center for Musculoskeletal Disorders, School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
16
|
Alharbi MA, Graves DT. FOXO 1 deletion in chondrocytes rescues diabetes-impaired fracture healing by restoring angiogenesis and reducing apoptosis. Front Endocrinol (Lausanne) 2023; 14:1136117. [PMID: 37576976 PMCID: PMC10421747 DOI: 10.3389/fendo.2023.1136117] [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: 01/02/2023] [Accepted: 06/12/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction Diabetes mellitus is associated with higher risks of long bone and jaw fractures. It is also associated with a higher incidence of delayed union or non-union. Our previous investigations concluded that a dominant mechanism was the premature loss of cartilage during endochondral bone formation associated with increased osteoclastic activities. We tested the hypothesis that FOXO1 plays a key role in diabetes-impaired angiogenesis and chondrocyte apoptosis. Methods Closed fractures of the femur were induced in mice with lineage-specific FOXO1 deletion in chondrocytes. The control group consisted of mice with the FOXO1 gene present. Mice in the diabetic group were rendered diabetic by multiple streptozotocin injections, while mice in the normoglycemic group received vehicle. Specimens were collected 16 days post fracture. The samples were fixed, decalcified, and embedded in paraffin blocks for immunostaining utilizing anti cleaved caspase-3 or CD31 specific antibodies compared with matched control IgG antibody, and apoptosis by the TUNEL assay. Additionally, ATDC5 chondrocytes were examined in vitro by RT-PCR, luciferase reporter and chromatin immunoprecipitation assays. Results Diabetic mice had ~ 50% fewer blood vessels compared to normoglycemic mice FOXO1 deletion in diabetic mice partially rescued the low number of blood vessels (p < 0.05). Additionally, diabetes increased caspase-3 positive and apoptotic chondrocytes by 50%. FOXO1 deletion in diabetic animals blocked the increase in both to levels comparable to normoglycemic animals (p < 0.05). High glucose (HG) and high advanced glycation end products (AGE) levels stimulated FOXO1 association with the caspase-3 promoter in vitro, and overexpression of FOXO1 increased caspase-3 promoter activity in luciferase reporter assays. Furthermore, we review previous mechanistic studies demonstrating that tumor necrosis factor (TNF) inhibition reverses impaired angiogenesis and reverses high levels of chondrocyte apoptosis that occur in fracture healing. Discussion New results presented here, in combination with recent studies, provide a comprehensive overview of how diabetes, through high glucose levels, AGEs, and increased inflammation, impair the healing process by interfering with angiogenesis and stimulating chondrocyte apoptosis. FOXO1 in diabetic fractures plays a negative role by reducing new blood vessel formation and increasing chondrocyte cell death which is distinct from its role in normal fracture healing.
Collapse
Affiliation(s)
- Mohammed A. Alharbi
- Department of Endodontics, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Dana T. Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
17
|
Fujikawa H, Kojima H, Terashima T, Katagi M, Yayama T, Kumagai K, Mori K, Saito H, Imai S. Expression of proinflammatory cytokines and proinsulin by bone marrow-derived cells for fracture healing in long-term diabetic mice. BMC Musculoskelet Disord 2023; 24:585. [PMID: 37464323 DOI: 10.1186/s12891-023-06710-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 07/09/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Diabetes mellitus (DM) causes bone dysfunction due to poor bone quality, leading to severe deterioration in patient of quality of life. The mechanisms of bone metabolism in DM remain unclear, although chemical and/or mechanical factors are known to disrupt the homeostasis of osteoblasts and osteoclasts. The purpose of this study was to identify the changes of osteoblasts and osteoclasts under long-term hyperglycaemic conditions, using a mouse fracture model of long-term hyperglycemia (LT-HG). METHODS C57BL/6J mice and green fluorescent protein (GFP) -positive bone marrow transplanted C57BL/6J mice with LT-HG, maintained under a state of hyperglycaemia for 2 months, were used in this study. After the experimental fracture, we examined the immunohistochemical expression of proinsulin and tumor necrosis factor (TNF) -α at the fracture site. C57BL/6J fracture model mice without hyperglycaemia were used as controls. RESULTS In the LT-HG mice, chondrocyte resorption was delayed, and osteoblasts showed an irregular arrangement at the callus site. The osteoclasts were scattered with a decrement in the number of nuclei. The expression of proinsulin was confirmed in bone marrow derived cells (BMDCs) with neovascularization 2 and 3 weeks after fracture. Immunopositivity for TNF-α was also confirmed in immature chondrocytes and BMDCs with neovascularization at 2 weeks, and the number of positive cells was not decreased at 3 weeks. Examination of GFP-grafted hyperglycaemic mice showed that the majority of cells at the fracture site were GFP-positive. Immunohistochemistry showed that the rate of double positives was 15% for GFP and proinsulin and 47% for GFP and TNF-α. CONCLUSION LT-HG induces an increase in the number of proinsulin and TNF-α positive cells derived from BMDCs. We suggest that proinsulin and TNF-α positive cells are involved in both bone formation and bone resorption after fracture under hyperglycaemic conditions, resulting in the delay of bone healing.
Collapse
Affiliation(s)
- Hitomi Fujikawa
- Department of Orthopaedic Surgery, Shiga University of Medical Science, Setatsukinowa-cho, Otsu, 520-2192, Shiga, Japan.
| | - Hideto Kojima
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Otsu, 520-2192, Shiga, Japan
| | - Tomoya Terashima
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Otsu, 520-2192, Shiga, Japan
| | - Miwako Katagi
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Otsu, 520-2192, Shiga, Japan
| | - Takafumi Yayama
- Department of Orthopaedic Surgery, Shiga University of Medical Science, Setatsukinowa-cho, Otsu, 520-2192, Shiga, Japan
| | - Kosuke Kumagai
- Department of Orthopaedic Surgery, Shiga University of Medical Science, Setatsukinowa-cho, Otsu, 520-2192, Shiga, Japan
| | - Kanji Mori
- Department of Orthopaedic Surgery, Shiga University of Medical Science, Setatsukinowa-cho, Otsu, 520-2192, Shiga, Japan.
| | - Hideki Saito
- Department of Orthopaedic Surgery, Shiga University of Medical Science, Setatsukinowa-cho, Otsu, 520-2192, Shiga, Japan
| | - Shinji Imai
- Department of Orthopaedic Surgery, Shiga University of Medical Science, Setatsukinowa-cho, Otsu, 520-2192, Shiga, Japan
| |
Collapse
|
18
|
Luo M, Zhao Z, Yi J. Osteogenesis of bone marrow mesenchymal stem cell in hyperglycemia. Front Endocrinol (Lausanne) 2023; 14:1150068. [PMID: 37415664 PMCID: PMC10321525 DOI: 10.3389/fendo.2023.1150068] [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: 01/23/2023] [Accepted: 06/05/2023] [Indexed: 07/08/2023] Open
Abstract
Diabetes mellitus (DM) has been shown to be a clinical risk factor for bone diseases including osteoporosis and fragility. Bone metabolism is a complicated process that requires coordinated differentiation and proliferation of bone marrow mesenchymal stem cells (BMSCs). Owing to the regenerative properties, BMSCs have laid a robust foundation for their clinical application in various diseases. However, mounting evidence indicates that the osteogenic capability of BMSCs is impaired under high glucose conditions, which is responsible for diabetic bone diseases and greatly reduces the therapeutic efficiency of BMSCs. With the rapidly increasing incidence of DM, a better understanding of the impacts of hyperglycemia on BMSCs osteogenesis and the underlying mechanisms is needed. In this review, we aim to summarize the current knowledge of the osteogenesis of BMSCs in hyperglycemia, the underlying mechanisms, and the strategies to rescue the impaired BMSCs osteogenesis.
Collapse
Affiliation(s)
- Meng Luo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jianru Yi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Johnson MJ, Kandasamy S, Raspovic KM, Manchanda K, Liu GT, VanPelt MD, Lavery LA, Wukich DK. Fractures and dislocations of the foot and ankle in people with diabetes: a literature review. Ther Adv Endocrinol Metab 2023; 14:20420188231163794. [PMID: 37323164 PMCID: PMC10265356 DOI: 10.1177/20420188231163794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 02/27/2023] [Indexed: 06/17/2023] Open
Abstract
Diabetes (DM) increases fracture risk, and bone quality depends on type diabetes type, duration, and other comorbidities. Diabetes is associated with a 32% increased relative risk (RR) of total fractures and 24% increased RR of ankle fractures compared with patients without DM. Type 2 DM is associated with a 37% increased RR of foot fractures compared with patients without DM. The incidence of ankle fractures in the general population is 169/100,000 per year, while foot fractures occur less frequently, with an incidence of 142/100,000 per year. Biomechanical properties of bone are negatively impacted by stiff collagen, contributing to the increased risk of fragility fractures in patients with DM. Systemic elevation of proinflammatory cytokines, such as tumor necrosis factor-alpha (TNFα), interleukin-1β (IL-1β), and interleukin 6 (IL-6), impact bone healing in patients with DM. Fractures in patients with DM, can be associated with poorly regulated levels of RANKL (receptor activator of nuclear transcription factor kappa-b ligand) leading to prolonged osteoclastogenesis, and net bone resorption. One of the most salient factors in treating fractures and dislocations of the foot and ankle is to recognize the difference between patients with uncomplicated and complicated DM. Complicated diabetes is defined as 'end organ damage', and for the purposes of this review, includes patients with neuropathy, peripheral artery disease (PAD) and/or chronic renal disease. Uncomplicated diabetes is not associated with 'end organ damage'. Foot and ankle fractures in patients with complicated DM pose challenges, and surgery is associated with increased risks of impaired wound healing, delayed fracture healing, malunion, infection, surgical site infection, and revision surgery. While patients with uncomplicated DM can be treated like patients without DM, patients with complicated DM require close follow-up and robust fixation methods should be considered to withstand the anticipated prolonged healing period. The aims of this review are as follows: (1) to review pertinent aspects of DM bone physiology and fracture healing, (2) to review the recent literature on treatment of foot and ankle fractures in patients with complicated DM, and (3) to provide treatment protocols based on the recent published evidence.
Collapse
Affiliation(s)
- Matthew J. Johnson
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Suganthi Kandasamy
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Katherine M. Raspovic
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Plastic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kshitij Manchanda
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - George Tye Liu
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Michael D. VanPelt
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lawrence A. Lavery
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Plastic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | |
Collapse
|
20
|
Wee NKY, Novak S, Ghosh D, Root SH, Dickerson IM, Kalajzic I. Inhibition of CGRP signaling impairs fracture healing in mice. J Orthop Res 2023; 41:1228-1239. [PMID: 36281531 PMCID: PMC10123175 DOI: 10.1002/jor.25474] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/06/2022] [Accepted: 10/18/2022] [Indexed: 02/04/2023]
Abstract
Calcitonin gene-related peptide (CGRP) is a neuropeptide produced by sensory nerves and functions as a pain sensor. It acts by binding to the calcitonin-like receptor (CLR, protein; Calcrl, gene). CGRP inhibition has been recently introduced as therapeutic treatment of migraine-associated pain. Previous studies have shown that CGRP stimulates bone formation. The aim of our study is to determine whether the inhibition of CGRP signaling negatively impacted fracture healing. Using α-smooth muscle actin (αSMA) Cre animals crossed with Ai9 reporter mice, we showed that CGRP-expressing nerves are near αSMA + cells in the periosteum. In vitro experiments revealed that periosteal cells express Calcrl and receptor activity modifying protein 1; and CGRP stimulation increased periosteal cell proliferation. Using a tamoxifen-inducible model αSMACre/CLRfl/fl , we targeted the deletion of CLR to periosteal progenitor cells and examined fracture healing. Microcomputed tomography of fractured femurs showed a reduction in bone mass in αSMACre+/CLRfl/fl female mice relative to controls and callus volume in males. Pharmacological CGRP-CLR inhibition was achieved by subcutaneous delivery of customized pellets with small molecule inhibitor olcegepant (BIBN-4096) at a dose of 10 μg/day. BIBN-4096-treated C57BL/6J mice had a higher latency toward thermal nociception than placebo-treated mice, indicating impaired sensory function through CGRP inhibition. CGRP inhibition also resulted in reduced callus volume, bone mass, and bone strength compared to placebo controls. These results indicate that inhibiting CGRP by deleting CLR or by using BIBN-4096, contributes to delayed bone healing.
Collapse
Affiliation(s)
- Natalie KY Wee
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
- Bone Cell Biology and Disease Unit, St Vincent’s Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Sanja Novak
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Debolina Ghosh
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Sierra H Root
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Ian M Dickerson
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
| |
Collapse
|
21
|
Luo B, Luo Y, He L, Cao Y, Jiang Q. Residual periodontal ligament in the extraction socket promotes the dentin regeneration potential of DPSCs in the rabbit jaw. Stem Cell Res Ther 2023; 14:47. [PMID: 36941706 PMCID: PMC10029302 DOI: 10.1186/s13287-023-03283-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 05/01/2022] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Because of the low regeneration efficiency and unclear underlying molecular mechanism, tooth regeneration applications are limited. In this study, we explored the influence of residual periodontal ligament on the dentin regeneration potential of dental pulp stem cells (DPSCs) in the jaw. METHODS To establish a tooth regeneration model, the incisors of New Zealand white rabbits were extracted while preserving residual periodontal ligament, followed by the implantation of DPSCs. After 3 months, micro-computed tomography (micro-CT), stereomicroscopy and scanning electron microscopy (SEM) were used to observe the volume, morphology and microstructure of regenerated tissue. Histological staining and immunostaining analyses were used to observe the morphological characteristics and expression of the dentin-specific proteins DMP1 and DSPP. To explore the mechanism, DPSCs and periodontal ligament stem cells (PDLSCs) were cocultured in vitro, and RNA was collected from the DPSCs for RNA-seq and bioinformatic analysis. RESULTS The results of micro-CT and stereomicroscopy showed that the number of sites with regeneration and the volume of regenerated tissue in the DPSCs/PDL group (6/8, 1.07 ± 0.93 cm3) were larger than those in the DPSCs group (3/8, 0.23 ± 0.41 cm3). The results of SEM showed that the regenerated dentin-like tissue in the DPSCs and DPSCs/PDL groups contained dentin tubules. Haematoxylin and eosin staining and immunohistochemical staining indicated that compared with the DPSCs group, the DPSCs/PDL group showed more regular regenerated tissue and higher expression levels of the dentin-specific proteins DMP1 and DSPP (DMP1: P = 0.02, DSPP: P = 0.01). RNA-seq showed that the coculture of DPSCs with PDLSCs resulted in the DPSCs differentially expressing 427 mRNAs (285 upregulated and 142 downregulated), 41 lncRNAs (26 upregulated and 15 downregulated), 411 circRNAs (224 upregulated and 187 downregulated), and 19 miRNAs (13 upregulated and 5 downregulated). Bioinformatic analysis revealed related Gene Ontology function and signalling pathways, including extracellular matrix (ECM), tumour necrosis factor (TNF) signalling and chemokine signalling pathways. CONCLUSIONS Residual periodontal ligament in the extraction socket promotes the dentin regeneration potential of DPSCs in the jaw. RNA-seq and bioinformatic analysis revealed that ECM, TNF signalling and chemokine signalling pathways may represent the key factors and signalling pathways.
Collapse
Affiliation(s)
- Bin Luo
- Department of Prosthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, 100050, China
| | - Yu Luo
- Department of Prosthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, 100050, China
| | - Lin He
- Department of Prosthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, 100050, China
| | - Yangyang Cao
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, 100050, China
| | - Qingsong Jiang
- Department of Prosthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
22
|
Niu Y, Du SZ, He R. TNF-α interference ameliorates brain damage in neonatal hypoxic-ischemic encephalopathy rats by regulating the expression of NT-3 and TRKC. IBRAIN 2023; 9:381-389. [PMID: 38680513 PMCID: PMC11045181 DOI: 10.1002/ibra.12089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 01/06/2023] [Accepted: 01/13/2023] [Indexed: 05/01/2024]
Abstract
The aim of this study is to explore the effect of tumor necrosis factor-α (TNF-α) inhibition in rats with neonatal hypoxic-ischemic encephalopathy (HIE) and ascertain the relevant signaling pathways. The Zea-Longa score was used to evaluate the neurological function of the rats. ImageJ was used for quantification of the brain edema volume. Triphenyl tetrazolium chloride (TTC) staining of brain tissue was performed 24 h after hypoxic-ischemic (HI) to detect right brain infarction. The expression of TNF-α was detected by real-time quantitative polymerase chain reaction (RT-qPCR). Immunofluorescence staining was used to identify the localization of TNF-α; Then, the effective shRNA fragment of TNF-α was used to validate the role of TNF-α in HIE rats, and the change of neurotrofin-3 (NT-3) and tyrosine kinase receptor-C (TRKC) was examined after TNF-α-shRNA lentivirus transfection to determine downstream signaling associated with TNF-α. Protein interaction analysis was carried out to predict the links among TNF-α, NT-3, and TRKC. Cerebral edema volume and infarction increased in the right brain after the HI operation. The Zea-Longa score significantly increased within 24 h after the HI operation. The relative expression of TNF-α was upregulated after the HI operation. TNF-α was highly expressed in the right hippocampus post HI through immunofluorescence staining. Bioinformatics analysis found a direct or an indirect link among TNF-α, NT-3, and TRKC. Moreover, the interference of TNF-α increased the expression of NT-3 and TRKC. TNF-α interference might alleviate brain injury in HIE by upregulating NT-3 and TRKC.
Collapse
Affiliation(s)
- Yong‐Min Niu
- Institute of NeuroscienceKunming Medical UniversityKunmingChina
| | - Steven Z. Du
- Department of Integrative BiologyUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Rong He
- Animal Zoology DepartmentKunming Medical UniversityKunmingChina
| |
Collapse
|
23
|
Zhang E, Miramini S, Patel M, Richardson M, Ebeling P, Zhang L. The effects of mechanical instability on PDGF mediated inflammatory response at early stage of fracture healing under diabetic condition. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 229:107319. [PMID: 36586180 DOI: 10.1016/j.cmpb.2022.107319] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/09/2022] [Accepted: 12/17/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND AND OBJECTIVE Mechanical stability plays an important role in fracture healing process. Excessive interfragmentary movement will continuously damage the tissue and newly formed capillaries at the fracture site, which leads to overproduction of platelet-derived growth factor (PDGF) that attracts more macrophages into fracture callus, ultimately persistent and enhanced inflammatory response happens. For diabetic condition, the impact of mechanical instability of fracture site on inflammatory response could be further compliciated and the relevant research in this field is relatively limited. METHODS Building on previous experimental studies, this study presents a numerical model consisting of a system of reactive-transport equations representing the transport as well as interactions of different cells and cytokines within the fracture callus. The model is initially validated by available experimental data, and then implemented to investigate the role of mechanical stability of fracture site in inflammatory response during early stage of healing. It is assumed that there is an increased release of PDGF due to the rupture of blood vessels resulting from mechanical instability, which leads to increased production of inflammatory cytokines (i.e., TNF-α). The bone healing process under three different conditions were investigated, i.e., mechanically stable condition with normal inflammatory response (Control, Case 1), mechanically unstable condition with normal inflammatory response (Case 2) and mechanically unstable condition with diabetes (Case 3). RESULTS Mechanical instability can promote the macrophage infiltration and thus induce an enhanced and prolonged inflammatory response, which could impede the MSCs proliferation during the early fracture healing stage (e.g., compared with the control condition, the MSCs concentration in unstable fracture with normal inflammatory response can be reduced by 3.2% and 5.2% on day 2 and day 10 post-fracture, respectively). Under diabetic condition, the mechanical instability of fracture site could lead to a significant increase of TNF-α concentration in fracture callus (Case 3) in comparison to control (Case 1) (e.g., three-fold increase in TNF-α concentration compared to control). In addition, the results show that the mechanical instability affects the cell differentiation and proliferation in fracture callus in a spatially dependent manner, e.g., for diabetic fracture patients, the mechanical instability could potentially decrease the concentration of MSCs, osteoblasts and chondrocytes by around 39%, 30% and 29% in cortical callus, respectively, in comparison to control. CONCLUSION The mechanical instability together with diabetic condition can significantly affect the natural resolution of inflammation during early stage of healing by turning acute inflammation into chronic inflammation which is characterized by a continuously upregulated TNF-α pathway.
Collapse
Affiliation(s)
- Enhao Zhang
- Department of Infrastructure Engineering, The University of Melbourne, Parkville, Victoria, Australia
| | - Saeed Miramini
- Department of Infrastructure Engineering, The University of Melbourne, Parkville, Victoria, Australia
| | - Minoo Patel
- Epworth Hospital Richmond, Richmond, Victoria, Australia
| | | | - Peter Ebeling
- Department of Medicine, School of Clinical Sciences, Monash University, Monash Medical Centre, Victoria, Australia
| | - Lihai Zhang
- Department of Infrastructure Engineering, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
24
|
Marino F, Scalise M, Salerno N, Salerno L, Molinaro C, Cappetta D, Torella M, Greco M, Foti D, Sasso FC, Mastroroberto P, De Angelis A, Ellison-Hughes GM, Sampaolesi M, Rota M, Rossi F, Urbanek K, Nadal-Ginard B, Torella D, Cianflone E. Diabetes-Induced Cellular Senescence and Senescence-Associated Secretory Phenotype Impair Cardiac Regeneration and Function Independently of Age. Diabetes 2022; 71:1081-1098. [PMID: 35108360 PMCID: PMC9490451 DOI: 10.2337/db21-0536] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 01/30/2022] [Indexed: 11/13/2022]
Abstract
Diabetes mellitus (DM) affects the biology of multipotent cardiac stem/progenitor cells (CSCs) and adult myocardial regeneration. We assessed the hypothesis that senescence and senescence-associated secretory phenotype (SASP) are main mechanisms of cardiac degenerative defect in DM. Accordingly, we tested whether ablation of senescent CSCs would rescue the cardiac regenerative/reparative defect imposed by DM. We obtained cardiac tissue from nonaged (50- to 64-year-old) patients with type 2 diabetes mellitus (T2DM) and without DM (NDM) and postinfarct cardiomyopathy undergoing cardiac surgery. A higher reactive oxygen species production in T2DM was associated with an increased number of senescent/dysfunctional T2DM-human CSCs (hCSCs) with reduced proliferation, clonogenesis/spherogenesis, and myogenic differentiation versus NDM-hCSCs in vitro. T2DM-hCSCs showed a defined pathologic SASP. A combination of two senolytics, dasatinib (D) and quercetin (Q), cleared senescent T2DM-hCSCs in vitro, restoring their expansion and myogenic differentiation capacities. In a T2DM model in young mice, diabetic status per se (independently of ischemia and age) caused CSC senescence coupled with myocardial pathologic remodeling and cardiac dysfunction. D + Q treatment efficiently eliminated senescent cells, rescuing CSC function, which resulted in functional myocardial repair/regeneration, improving cardiac function in murine DM. In conclusion, DM hampers CSC biology, inhibiting CSCs' regenerative potential through the induction of cellular senescence and SASP independently from aging. Senolytics clear senescence, abrogating the SASP and restoring a fully proliferative/differentiation-competent hCSC pool in T2DM with normalization of cardiac function.
Collapse
Affiliation(s)
- Fabiola Marino
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
- Translational Cardiomyology Laboratory, Stem Cell Biology and Embryology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Mariangela Scalise
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Nadia Salerno
- Department of Medical and Surgical Sciences, Magna Græcia University, Catanzaro, Italy
| | - Luca Salerno
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Claudia Molinaro
- Department of Medical and Surgical Sciences, Magna Græcia University, Catanzaro, Italy
| | - Donato Cappetta
- Department of Experimental Medicine, Section of Pharmacology, University of Campania “L. Vanvitelli,” Naples, Italy
| | - Michele Torella
- Department of Translational Medicine, University of Campania “L. Vanvitelli,” Naples, Italy
| | - Marta Greco
- Department of Health Sciences, Magna Græcia University, Catanzaro, Italy
| | - Daniela Foti
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Ferdinando C. Sasso
- Department of Translational Medicine, University of Campania “L. Vanvitelli,” Naples, Italy
| | - Pasquale Mastroroberto
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, Section of Pharmacology, University of Campania “L. Vanvitelli,” Naples, Italy
| | - Georgina M. Ellison-Hughes
- Centre for Human and Applied Physiological Sciences and Centre for Stem Cells and Regenerative Medicine, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, King’s College London, London, U.K
| | - Maurilio Sampaolesi
- Translational Cardiomyology Laboratory, Stem Cell Biology and Embryology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Marcello Rota
- Department of Physiology, New York Medical College, Valhalla, NY
| | - Francesco Rossi
- Department of Experimental Medicine, Section of Pharmacology, University of Campania “L. Vanvitelli,” Naples, Italy
| | - Konrad Urbanek
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Bernardo Nadal-Ginard
- Department of Medical and Surgical Sciences, Magna Græcia University, Catanzaro, Italy
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
- Corresponding authors: Daniele Torella, , and Eleonora Cianflone,
| | - Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Græcia University, Catanzaro, Italy
- Department of Physiology, New York Medical College, Valhalla, NY
- Corresponding authors: Daniele Torella, , and Eleonora Cianflone,
| |
Collapse
|
25
|
Paco-Meza LM, Carmona MD, Cañadillas S, Lopez-Diaz A, Muñoz-López F, Jimenez-Arranz A, Guler I, Herrera C. Identification of molecular pathways and protein-protein interactions in adipose tissue-derived mesenchymal stromal cells (ASCs) under physiological oxygen concentration in a diabetic rat model. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:155-163. [PMID: 35655589 PMCID: PMC9124531 DOI: 10.22038/ijbms.2022.59004.13107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 01/24/2022] [Indexed: 11/06/2022]
Abstract
Objectives Adipose tissue-derived mesenchymal stromal cells (ASCs) are useful in cell-based therapy. However, it is well known that diabetes mellitus (DM) alters ASCs' functionality. The majority of in vitro studies related to ASCs are developed under non-physiological oxygen conditions. Therefore, they may not reflect the full effects of DM on ASCs, in vivo. The main aim of the current study is to identify molecular pathways and underlying biological mechanisms affected by diabetes on ASCs in physiological oxygen conditions. Materials and Methods ASCs derived from healthy (ASCs-C) and diabetic (ASCs-D) rats were expanded under standard culture conditions (21% O2) or cultured in physiological oxygen conditions (3% O2) and characterized. Differential gene expressions (DEGs) of ASCs-D with respect to ASCs-C were identified and analyzed with bioinformatic tools. Protein-protein interaction (PPI) networks, from up- and down-regulated DEGs, were also constructed. Results The bioinformatic analysis revealed 1354 up-regulated and 859 down-regulated DEGs in ASCs-D, with 21 and 78 terms over and under-represented, respectively. Terms linked with glycosylation and ribosomes were over-represented and terms related to the activity of RNA-polymerase II and transcription regulation were under-represented. PPI network disclosed RPL11-RPS5 and KDR-VEGFA as the main interactions from up- and down-regulated DEGs, respectively. Conclusion These results provide valuable information about gene pathways and underlying molecular mechanisms by which diabetes disturbs ASCs biology in physiological oxygen conditions. Furthermore, they reveal, molecular targets to improve the use of ASCs in autologous transplantation.
Collapse
Affiliation(s)
- Luis-Miguel Paco-Meza
- Maimonides Institute of Biomedical Research in Cordoba (IMIBIC), Spain. Avenida Menéndez Pidal s/n, CP 14004 Córdoba, Spain
- These authors contributed equally to this work
| | - MDolores Carmona
- Maimonides Institute of Biomedical Research in Cordoba (IMIBIC), Spain. Avenida Menéndez Pidal s/n, CP 14004 Córdoba, Spain
- Cellular Therapy Unit, Reina Sofia University Hospital, Cordoba, Spain. Avenida Menéndez Pidal s/n, CP 14004 Córdoba, Spain
- University of Cordoba, Spain. Avenida Menéndez Pidal s/n, CP 14004 Córdoba, Spain
- These authors contributed equally to this work
| | - Sagrario Cañadillas
- Maimonides Institute of Biomedical Research in Cordoba (IMIBIC), Spain. Avenida Menéndez Pidal s/n, CP 14004 Córdoba, Spain
| | - Ana Lopez-Diaz
- Maimonides Institute of Biomedical Research in Cordoba (IMIBIC), Spain. Avenida Menéndez Pidal s/n, CP 14004 Córdoba, Spain
| | - Francisco Muñoz-López
- Bio-Knowledge Lab, Glorieta de los Países Bálticos, s/n. Edificio Baobab 1, Oficina 15, Polígono Tecnocórdoba, 14014 Córdoba, Spain
| | - Alvaro Jimenez-Arranz
- Maimonides Institute of Biomedical Research in Cordoba (IMIBIC), Spain. Avenida Menéndez Pidal s/n, CP 14004 Córdoba, Spain
| | - Ipek Guler
- Maimonides Institute of Biomedical Research in Cordoba (IMIBIC), Spain. Avenida Menéndez Pidal s/n, CP 14004 Córdoba, Spain
| | - Concha Herrera
- Maimonides Institute of Biomedical Research in Cordoba (IMIBIC), Spain. Avenida Menéndez Pidal s/n, CP 14004 Córdoba, Spain
- Cellular Therapy Unit, Reina Sofia University Hospital, Cordoba, Spain. Avenida Menéndez Pidal s/n, CP 14004 Córdoba, Spain
- University of Cordoba, Spain. Avenida Menéndez Pidal s/n, CP 14004 Córdoba, Spain
| |
Collapse
|
26
|
Zhang E, Miramini S, Patel M, Richardson M, Ebeling P, Zhang L. Role of TNF-α in early-stage fracture healing under normal and diabetic conditions. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 213:106536. [PMID: 34823199 DOI: 10.1016/j.cmpb.2021.106536] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/14/2021] [Accepted: 11/12/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND AND OBJECTIVE Inflammatory response plays a crucial role in the early stage of fracture healing. Immediately after fracture, the debris and immune cells (e.g., macrophages), recruited into the fracture callus, lead to the secretion of inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α), which governs the mesenchymal stem cells (MSCs) mediated healing processes. However, it is still unclear how chronic inflammatory diseases (e.g., diabetes) affect the level of TNF-α in fracture callus, ultimately the healing outcomes at the early stage of healing. Therefore, the purpose of this study is to develop a numerical model for investigating TNF-α mediated bone fracture healing. METHODS A mathematical model consisting of a system of partial differential equations that represent the reactive transport of cells and cytokines in the fracture callus is developed in this study. The model is first calibrated by using available experimental data and then implemented to study the effect of TNF-α on the early stage of fracture healing under normal and diabetic conditions. RESULTS There is a significant elevation of TNF-α level in facture callus during the first 24 h post-fracture in normal condition, and its influence in the concentration of MSCs and cell differentiation becomes significant three days post-fracture (e.g., the absence of TNF-α signaling could reduce the concentration of MSCs more than 20% in cortical callus). In addition, the excessive secretion of TNF-α induced by diabetes could decrease the concentration of MSCs at the initial stage of healing, particularly reduce the concentration of MSCs in cortical callus by around 25%. CONCLUSION The model predictions suggested that there should be an optimal concentration of TNF-α in fracture callus, which enhances the early stage of healing, and excessive or insufficient secretion of TNF-α might significantly hinder the healing process.
Collapse
Affiliation(s)
- Enhao Zhang
- Department of Infrastructure Engineering, The University of Melbourne, 700 Swanston St, Parkville, Victoria 3010, Australia
| | - Saeed Miramini
- Department of Infrastructure Engineering, The University of Melbourne, 700 Swanston St, Parkville, Victoria 3010, Australia
| | - Minoo Patel
- Centre for Limb Lengthening and Reconstruction, Epworth Hospital Richmond, Richmond, Victoria, Australia
| | | | - Peter Ebeling
- Department of Medicine, School of Clinical Sciences, Monash University, Monash Medical Centre, Victoria, Australia
| | - Lihai Zhang
- Department of Infrastructure Engineering, The University of Melbourne, 700 Swanston St, Parkville, Victoria 3010, Australia.
| |
Collapse
|
27
|
Okada K, Kawao N, Nakai D, Wakabayashi R, Horiuchi Y, Okumoto K, Kurashimo S, Takafuji Y, Matsuo O, Kaji H. Role of Macrophages and Plasminogen Activator Inhibitor-1 in Delayed Bone Repair Induced by Glucocorticoids in Mice. Int J Mol Sci 2022; 23:478. [PMID: 35008904 PMCID: PMC8745285 DOI: 10.3390/ijms23010478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/24/2021] [Accepted: 12/29/2021] [Indexed: 11/16/2022] Open
Abstract
Glucocorticoids delay fracture healing and induce osteoporosis. However, the mechanisms by which glucocorticoids delay bone repair have yet to be clarified. Plasminogen activator inhibitor-1 (PAI-1) is the principal inhibitor of plasminogen activators and an adipocytokine that regulates metabolism. We herein investigated the roles of macrophages in glucocorticoid-induced delays in bone repair after femoral bone injury using PAI-1-deficient female mice intraperitoneally administered with dexamethasone (Dex). Dex significantly decreased the number of F4/80-positive macrophages at the damaged site two days after femoral bone injury. It also attenuated bone injury-induced decreases in the number of hematopoietic stem cells in bone marrow in wild-type and PAI-1-deficient mice. PAI-1 deficiency significantly weakened Dex-induced decreases in macrophage number and macrophage colony-stimulating factor (M-CSF) mRNA levels at the damaged site two days after bone injury. It also significantly ameliorated the Dex-induced inhibition of macrophage phagocytosis at the damaged site. In conclusion, we herein demonstrated that Dex decreased the number of macrophages at the damaged site during early bone repair after femoral bone injury partly through PAI-1 and M-CSF in mice.
Collapse
Affiliation(s)
- Kiyotaka Okada
- Department of Arts and Sciences, Faculty of Medicine, Kindai University, Osaka 589-8511, Japan;
- Department of Physiology and Regenerative Medicine, Faculty of Medicine, Kindai University, Osaka 589-8511, Japan; (N.K.); (D.N.); (R.W.); (Y.T.); (O.M.)
| | - Naoyuki Kawao
- Department of Physiology and Regenerative Medicine, Faculty of Medicine, Kindai University, Osaka 589-8511, Japan; (N.K.); (D.N.); (R.W.); (Y.T.); (O.M.)
| | - Daisho Nakai
- Department of Physiology and Regenerative Medicine, Faculty of Medicine, Kindai University, Osaka 589-8511, Japan; (N.K.); (D.N.); (R.W.); (Y.T.); (O.M.)
| | - Rei Wakabayashi
- Department of Physiology and Regenerative Medicine, Faculty of Medicine, Kindai University, Osaka 589-8511, Japan; (N.K.); (D.N.); (R.W.); (Y.T.); (O.M.)
| | - Yoshitaka Horiuchi
- Life Science Research Institute, Kindai University, Osaka 589-8511, Japan; (Y.H.); (K.O.); (S.K.)
| | - Katsumi Okumoto
- Life Science Research Institute, Kindai University, Osaka 589-8511, Japan; (Y.H.); (K.O.); (S.K.)
| | - Shinji Kurashimo
- Life Science Research Institute, Kindai University, Osaka 589-8511, Japan; (Y.H.); (K.O.); (S.K.)
| | - Yoshimasa Takafuji
- Department of Physiology and Regenerative Medicine, Faculty of Medicine, Kindai University, Osaka 589-8511, Japan; (N.K.); (D.N.); (R.W.); (Y.T.); (O.M.)
| | - Osamu Matsuo
- Department of Physiology and Regenerative Medicine, Faculty of Medicine, Kindai University, Osaka 589-8511, Japan; (N.K.); (D.N.); (R.W.); (Y.T.); (O.M.)
| | - Hiroshi Kaji
- Department of Physiology and Regenerative Medicine, Faculty of Medicine, Kindai University, Osaka 589-8511, Japan; (N.K.); (D.N.); (R.W.); (Y.T.); (O.M.)
| |
Collapse
|
28
|
Ehara H, Tatsumi K, Takafuji Y, Kawao N, Ishida M, Okada K, Mackman N, Kaji H. Role of tissue factor in delayed bone repair induced by diabetic state in mice. PLoS One 2021; 16:e0260754. [PMID: 34855855 PMCID: PMC8638858 DOI: 10.1371/journal.pone.0260754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/17/2021] [Indexed: 11/19/2022] Open
Abstract
Background Tissue factor (TF) is the primary activator of the extrinsic coagulation protease cascade. Although TF plays roles in various pathological states, such as thrombosis, inflammatory diseases, cancer, and atherosclerosis, its involvement in bone metabolism remains unknown. Materials and methods The present study examined the roles of TF in delayed bone repair induced by a diabetic state in mice using wild-type (WT) and low TF-expressing (LTF) male mice. A diabetic state was induced by intraperitoneal injections of streptozotocin (STZ). Results A prolonged diabetic state significantly reduced total and trabecular bone mineral densities (BMD) as well as cortical bone thickness in WT and LTF mice; these BMD parameters were similar between WT and LTF mice treated with or without STZ. The diabetic state induced in WT mice delayed the repair of the femur following injury. The diabetic state induced in LTF mice was associated with further delays in bone repair. In in vitro experiments, TF significantly decreased receptor activator of nuclear factor-κB ligand-induced osteoclast formation and osteoclastogenic gene expression in RAW264.7 cells. However, it did not affect the gene expression levels of runt-related transcription factor 2 and osterix as well as alkaline phosphatase activity in mouse primary osteoblasts. Conclusion Low TF state was associated with enhanced bone repair delay induced by diabetic state in mice. The TF-induced suppression of bone remodeling may be a contributing factor to the protective effects of TF against delayed bone repair in a diabetic state.
Collapse
Affiliation(s)
- Hiroki Ehara
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Kohei Tatsumi
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
- Advanced Medical Science of Thrombosis and Hemostasis, Nara Medical University, Kashihara, Nara, Japan
| | - Yoshimasa Takafuji
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Naoyuki Kawao
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Masayoshi Ishida
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Kiyotaka Okada
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Nigel Mackman
- Department of Medicine, Division of Hematology, UNC Blood Research Institute, University of North Carolina, Chapel Hill, NC, United States of America
| | - Hiroshi Kaji
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
- * E-mail:
| |
Collapse
|
29
|
Chinipardaz Z, Liu M, Graves D, Yang S. Diabetes impairs fracture healing through disruption of cilia formation in osteoblasts. Bone 2021; 153:116176. [PMID: 34508881 PMCID: PMC9160738 DOI: 10.1016/j.bone.2021.116176] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/22/2021] [Accepted: 09/05/2021] [Indexed: 02/06/2023]
Abstract
Diabetes-associated fracture risk and impaired fracture healing represents a serious health threat. It is well known that type 1 diabetes mellitus (T1DM) impairs fracture healing due to its effect on osteoblasts and their progenitor cells. Previous studies have showed that primary cilia and intraflagellar transport protein 80 (IFT80) are critical for bone formation. However, whether TIDM impairs fracture healing due to influencing ciliary gene expression and cilia formation is unknown. Here, we investigated the effect of T1DM on primary cilia in a streptozotocin induced diabetes mouse model and examined the impact of cilia on fracture healing in osteoblasts by deletion of IFT80 in osteoblast linage using osterix (OSX)-cre (OSXcretTAIFT80f/f). The results showed that diabetes inhibited ciliary gene expression and primary cilia formation to an extent that was similar to normoglycemic mice with IFT80 deletion. Moreover, diabetic mice and normoglycemic mice with cilia loss in osteoblasts (OSXcretTAIFT80f/f) both exhibited delayed fracture healing with significantly reduced bone density and mechanical strength as well as with reduced expression of osteoblast markers, decreased angiogenesis and proliferation of bone lining cells at the fracture sites. In vitro studies showed that advanced glycation end products (AGEs) downregulated IFT80 expression in osteoblast progenitors. Moreover, AGEs and IFT80 deletion significantly reduced cilia number and length which inhibited differentiation of primary osteoblast precursors. Thus, this study for the first time report that primary cilia are essential for bone regeneration during fracture healing and loss of cilia caused by diabetes in osteoblasts resulted in defective diabetic fracture healing.
Collapse
Affiliation(s)
- Zahra Chinipardaz
- Department of Basic and Translation Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Min Liu
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dana Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Shuying Yang
- Department of Basic and Translation Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Innovation & Precision Dentistry, School of Dental Medicine, School of Engineering and Applied Sciences, University of Pennsylvania, PA 19104, USA; The Penn Center for Musculoskeletal Disorders, School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
30
|
Xu J, Zuo C. The Fate Status of Stem Cells in Diabetes and its Role in the Occurrence of Diabetic Complications. Front Mol Biosci 2021; 8:745035. [PMID: 34796200 PMCID: PMC8592901 DOI: 10.3389/fmolb.2021.745035] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/20/2021] [Indexed: 12/19/2022] Open
Abstract
Diabetes mellitus (DM) is becoming a growing risk factor for public health worldwide. It is a very common disease and is widely known for its susceptibility to multiple complications which do great harm to the life and health of patients, some even lead to death. To date, there are many mechanisms for the complications of diabetes, including the generation of reactive oxygen species (ROS) and the abnormal changes of gas transmitters, which ultimately lead to injuries of cells, tissues and organs. Normally, even if injured, the body can quickly repair and maintain its homeostasis. This is closely associated with the repair and regeneration ability of stem cells. However, many studies have demonstrated that stem cells happen to be damaged under DM, which may be a nonnegligible factor in the occurrence and progression of diabetic complications. Therefore, this review summarizes how diabetes causes the corresponding complications by affecting stem cells from two aspects: stem cells dysfunctions and stem cells quantity alteration. In addition, since mesenchymal stem cells (MSCs), especially bone marrow mesenchymal stem cells (BMMSCs), have the advantages of strong differentiation ability, large quantity and wide application, we mainly focus on the impact of diabetes on them. The review also puts forward the basis of using exogenous stem cells to treat diabetic complications. It is hoped that through this review, researchers can have a clearer understanding of the roles of stem cells in diabetic complications, thus promoting the process of using stem cells to treat diabetic complications.
Collapse
Affiliation(s)
- Jinyi Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Chengguo Zuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
31
|
Ko KI, Sculean A, Graves DT. Diabetic wound healing in soft and hard oral tissues. Transl Res 2021; 236:72-86. [PMID: 33992825 PMCID: PMC8554709 DOI: 10.1016/j.trsl.2021.05.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/06/2021] [Accepted: 05/06/2021] [Indexed: 12/17/2022]
Abstract
There is significant interest in understanding the cellular mechanisms responsible for expedited healing response in various oral tissues and how they are impacted by systemic diseases. Depending upon the types of oral tissue, wound healing may occur by predominantly re-eptihelialization, by re-epithelialization with substantial new connective tissue formation, or by a a combination of both plus new bone formation. As a result, the cells involved differ and are impacted by systemic diaseses in various ways. Diabetes mellitus is a prevalent metabolic disorder that impairs barrier function and healing responses throughout the human body. In the oral cavity, diabetes is a known risk factor for exacerbated periodontal disease and delayed wound healing, which includes both soft and hard tissue components. Here, we review the mechanisms of diabetic oral wound healing, particularly on impaired keratinocyte proliferation and migration, altered level of inflammation, and reduced formation of new connective tissue and bone. In particular, diabetes inhibits the expression of mitogenic growth factors whereas that of pro-inflammatory cytokines is elevated through epigenetic mechanisms. Moreover, hyperglycemia and oxidative stress induced by diabetes prevents the expansion of mesengenic cells that are involved in both soft and hard tissue oral wounds. A better understanding of how diabetes influences the healing processes is crucial for the prevention and treatment of diabetes-associated oral complications.
Collapse
Affiliation(s)
- Kang I Ko
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, 19104
| | - Anton Sculean
- Department of Periodontology, School of Dental Medicine, University of Bern, Freiburgstrasse 7, CH-3010, Bern, Switzerland
| | - Dana T Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, 19104.
| |
Collapse
|
32
|
Shang F, Yu Y, Liu S, Ming L, Zhang Y, Zhou Z, Zhao J, Jin Y. Advancing application of mesenchymal stem cell-based bone tissue regeneration. Bioact Mater 2021; 6:666-683. [PMID: 33005830 PMCID: PMC7509590 DOI: 10.1016/j.bioactmat.2020.08.014] [Citation(s) in RCA: 155] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 08/07/2020] [Accepted: 08/15/2020] [Indexed: 12/11/2022] Open
Abstract
Reconstruction of bone defects, especially the critical-sized defects, with mechanical integrity to the skeleton is important for a patient's rehabilitation, however, it still remains challenge. Utilizing biomaterials of human origin bone tissue for therapeutic purposes has provided a facilitated approach that closely mimics the critical aspects of natural bone tissue with regard to its properties. However, not only efficacious and safe but also cost-effective and convenient are important for regenerative biomaterials to achieve clinical translation and commercial success. Advances in our understanding of regenerative biomaterials and their roles in new bone formation potentially opened a new frontier in the fast-growing field of regenerative medicine. Taking inspiration from the role and multicomponent construction of native extracellular matrix (ECM) for cell accommodation, the ECM-mimicking biomaterials and the naturally decellularized ECM scaffolds were used to create new tissues for bone restoration. On the other hand, with the going deep in understanding of mesenchymal stem cells (MSCs), they have shown great promise to jumpstart and facilitate bone healing even in diseased microenvironments with pharmacology-based endogenous MSCs rescue/mobilization, systemic/local infusion of MSCs for cytotherapy, biomaterials-based approaches, cell-sheets/-aggregates technology and usage of subcellular vesicles of MSCs to achieve scaffolds-free or cell-free delivery system, all of them have been shown can improve MSCs-mediated regeneration in preclinical studies and several clinical trials. Here, following an overview discussed autogenous/allogenic and ECM-based bone biomaterials for reconstructive surgery and applications of MSCs-mediated bone healing and tissue engineering to further offer principles and effective strategies to optimize MSCs-based bone regeneration.
Collapse
Affiliation(s)
- Fengqing Shang
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Department of Stomatology, The 306th Hospital of PLA, Beijing, 100101, China
| | - Yang Yu
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, 250012, China
| | - Shiyu Liu
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Leiguo Ming
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yongjie Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Zhifei Zhou
- Department of Stomatology, General Hospital of Tibetan Military Command, Lhasa, 850000, China
| | - Jiayu Zhao
- Bureau of Service for Veteran Cadres of PLA in Beijing, Beijing, 100001, China
| | - Yan Jin
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| |
Collapse
|
33
|
Ding Y, Wei J, Hettinghouse A, Li G, Li X, Einhorn TA, Liu CJ. Progranulin promotes bone fracture healing via TNFR pathways in mice with type 2 diabetes mellitus. Ann N Y Acad Sci 2021; 1490:77-89. [PMID: 33543485 DOI: 10.1111/nyas.14568] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 01/08/2021] [Accepted: 01/15/2021] [Indexed: 12/12/2022]
Abstract
Type 2 diabetes mellitus (T2DM) significantly increases bone fragility and fracture risk. Progranulin (PGRN) promotes bone fracture healing in both physiological and type 1 diabetic conditions. The present study aimed to investigate the role of PGRN in T2DM bone fracture healing. MKR mice (with an FVB/N genetic background) were used as the T2DM model. Drill-hole and Bonnarens and Einhorn models were used to investigate the role of PGRN in T2DM fracture healing in vivo. Primary bone marrow cells were isolated for molecular and signaling studies, and reverse transcription-polymerase chain reaction, immunohistochemical staining, and western blotting were performed to assess PGRN effects in vitro. PGRN mRNA and protein expression were upregulated in the T2DM model. Local administration of recombinant PGRN effectively promoted T2DM bone fracture healing in vivo. Additionally, PGRN could induce anabolic metabolism during endochondral ossification through the TNFR2-Akt and Erk1/2 pathways. Furthermore, PGRN showed anti-inflammatory activity in the T2DM bone regeneration process. These findings suggest that local administration of exogenous PGRN may be an alternative strategy to support bone regeneration in patients with T2DM. Additionally, PGRN might hold therapeutic potential for other TNFR-related metabolic disorders.
Collapse
Affiliation(s)
- Yuanjing Ding
- Department of Orthopaedic Surgery, New York University Medical Center, New York, New York.,Department of Orthopaedic Surgery, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jianlu Wei
- Department of Orthopaedic Surgery, New York University Medical Center, New York, New York.,Department of Orthopaedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Aubryanna Hettinghouse
- Department of Orthopaedic Surgery, New York University Medical Center, New York, New York
| | - Guangfei Li
- Department of Orthopaedic Surgery, New York University Medical Center, New York, New York
| | - Xin Li
- College of Dentistry, New York University, New York, New York
| | - Thomas A Einhorn
- Department of Orthopaedic Surgery, New York University Medical Center, New York, New York
| | - Chuan-Ju Liu
- Department of Orthopaedic Surgery, New York University Medical Center, New York, New York.,Department of Cell Biology, New York University School of Medicine, New York, New York
| |
Collapse
|
34
|
Doherty L, Wan M, Kalajzic I, Sanjay A. Diabetes impairs periosteal progenitor regenerative potential. Bone 2021; 143:115764. [PMID: 33221502 PMCID: PMC7770068 DOI: 10.1016/j.bone.2020.115764] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 01/01/2023]
Abstract
Diabetics are at increased risk for fracture, and experience severely impaired skeletal healing characterized by delayed union or nonunion of the bone. The periosteum harbors osteochondral progenitors that can differentiate into chondrocytes and osteoblasts, and this connective tissue layer is required for efficient fracture healing. While bone marrow-derived stromal cells have been studied extensively in the context of diabetic skeletal repair and osteogenesis, the effect of diabetes on the periosteum and its ability to contribute to bone regeneration has not yet been explicitly evaluated. Within this study, we utilized an established murine model of type I diabetes to evaluate periosteal cell differentiation capacity, proliferation, and availability under the effect of a diabetic environment. Periosteal cells from diabetic mice were deficient in osteogenic differentiation ability in vitro, and diabetic mice had reduced periosteal populations of mesenchymal progenitors with a corresponding reduction in proliferation capacity following injury. Additionally, fracture callus mineralization and mature osteoblast activity during periosteum-mediated healing was impaired in diabetic mice compared to controls. We propose that the effect of diabetes on periosteal progenitors and their ability to aid in skeletal repair directly impairs fracture healing.
Collapse
Affiliation(s)
- Laura Doherty
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Health, Farmington, CT, USA
| | - Matthew Wan
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Health, Farmington, CT, USA
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, UConn School of Dental Medicine, Farmington, CT, USA
| | - Archana Sanjay
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Health, Farmington, CT, USA.
| |
Collapse
|
35
|
Ribot J, Denoeud C, Frescaline G, Landon R, Petite H, Pavon-Djavid G, Bensidhoum M, Anagnostou F. Experimental Type 2 Diabetes Differently Impacts on the Select Functions of Bone Marrow-Derived Multipotent Stromal Cells. Cells 2021; 10:268. [PMID: 33572905 PMCID: PMC7912056 DOI: 10.3390/cells10020268] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 12/16/2022] Open
Abstract
Bone marrow-derived multipotent stromal cells (BMMSCs) represent an attractive therapeutic modality for cell therapy in type 2 diabetes mellitus (T2DM)-associated complications. T2DM changes the bone marrow environment; however, its effects on BMMSC properties remain unclear. The present study aimed at investigating select functions and differentiation of BMMSCs harvested from the T2DM microenvironment as potential candidates for regenerative medicine. BMMSCs were obtained from Zucker diabetic fatty (ZDF; an obese-T2DM model) rats and their lean littermates (ZL; controls), and cultured under normoglycemic conditions. The BMMSCs derived from ZDF animals were fewer in number, with limited clonogenicity (by 2-fold), adhesion (by 2.9-fold), proliferation (by 50%), migration capability (by 25%), and increased apoptosis rate (by 2.5-fold) compared to their ZL counterparts. Compared to the cultured ZL-BMMSCs, the ZDF-BMMSCs exhibited (i) enhanced adipogenic differentiation (increased number of lipid droplets by 2-fold; upregulation of the Pparg, AdipoQ, and Fabp genes), possibly due to having been primed to undergo such differentiation in vivo prior to cell isolation, and (ii) different angiogenesis-related gene expression in vitro and decreased proangiogenic potential after transplantation in nude mice. These results provided evidence that the T2DM environment impairs BMMSC expansion and select functions pertinent to their efficacy when used in autologous cell therapies.
Collapse
Affiliation(s)
- Jonathan Ribot
- Université de Paris, CNRS, INSERM, B3OA, 75010 Paris, France; (J.R.); (C.D.); (G.F.); (R.L.); (H.P.); (M.B.)
| | - Cyprien Denoeud
- Université de Paris, CNRS, INSERM, B3OA, 75010 Paris, France; (J.R.); (C.D.); (G.F.); (R.L.); (H.P.); (M.B.)
| | - Guilhem Frescaline
- Université de Paris, CNRS, INSERM, B3OA, 75010 Paris, France; (J.R.); (C.D.); (G.F.); (R.L.); (H.P.); (M.B.)
| | - Rebecca Landon
- Université de Paris, CNRS, INSERM, B3OA, 75010 Paris, France; (J.R.); (C.D.); (G.F.); (R.L.); (H.P.); (M.B.)
| | - Hervé Petite
- Université de Paris, CNRS, INSERM, B3OA, 75010 Paris, France; (J.R.); (C.D.); (G.F.); (R.L.); (H.P.); (M.B.)
| | - Graciela Pavon-Djavid
- INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Université Sorbonne Paris Nord, 93430 Villetaneuse, France;
| | - Morad Bensidhoum
- Université de Paris, CNRS, INSERM, B3OA, 75010 Paris, France; (J.R.); (C.D.); (G.F.); (R.L.); (H.P.); (M.B.)
| | - Fani Anagnostou
- Université de Paris, CNRS, INSERM, B3OA, 75010 Paris, France; (J.R.); (C.D.); (G.F.); (R.L.); (H.P.); (M.B.)
- Department of Periodontology, Service of Odontology–Pitié Salpêtrière Hospital, AP-HP et U.F.R. of Odontology, 75013 Paris, France
| |
Collapse
|
36
|
Sun X, Ma Z, Zhao X, Jin W, Zhang C, Ma J, Qiang L, Wang W, Deng Q, Yang H, Zhao J, Liang Q, Zhou X, Li T, Wang J. Three-dimensional bioprinting of multicell-laden scaffolds containing bone morphogenic protein-4 for promoting M2 macrophage polarization and accelerating bone defect repair in diabetes mellitus. Bioact Mater 2020; 6:757-769. [PMID: 33024897 PMCID: PMC7522044 DOI: 10.1016/j.bioactmat.2020.08.030] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/09/2020] [Accepted: 08/23/2020] [Indexed: 12/18/2022] Open
Abstract
Critical-sized bone defect repair in patients with diabetes mellitus remains a challenge in clinical treatment because of dysfunction of macrophage polarization and the inflammatory microenvironment in the bone defect region. Three-dimensional (3D) bioprinted scaffolds loaded with live cells and bioactive factors can improve cell viability and the inflammatory microenvironment and further accelerating bone repair. Here, we used modified bioinks comprising gelatin, gelatin methacryloyl (GelMA), and 4-arm poly (ethylene glycol) acrylate (PEG) to fabricate 3D bioprinted scaffolds containing BMSCs, RAW264.7 macrophages, and BMP-4-loaded mesoporous silica nanoparticles (MSNs). Addition of MSNs effectively improved the mechanical strength of GelMA/gelatin/PEG scaffolds. Moreover, MSNs sustainably released BMP-4 for long-term effectiveness. In 3D bioprinted scaffolds, BMP-4 promoted the polarization of RAW264.7 to M2 macrophages, which secrete anti-inflammatory factors and thereby reduce the levels of pro-inflammatory factors. BMP-4 released from MSNs and BMP-2 secreted from M2 macrophages collectively stimulated the osteogenic differentiation of BMSCs in the 3D bioprinted scaffolds. Furthermore, in calvarial critical-size defect models of diabetic rats, 3D bioprinted scaffolds loaded with MSNs/BMP-4 induced M2 macrophage polarization and improved the inflammatory microenvironment. And 3D bioprinted scaffolds with MSNs/BMP-4, BMSCs, and RAW264.7 cells significantly accelerated bone repair. In conclusion, our results indicated that implanting 3D bioprinted scaffolds containing MSNs/BMP-4, BMSCs, and RAW264.7 cells in bone defects may be an effective method for improving diabetic bone repair, owing to the direct effects of BMP-4 on promoting osteogenesis of BMSCs and regulating M2 type macrophage polarization to improve the inflammatory microenvironment and secrete BMP-2. The GelMA/gelatin/PEG/MSN composite bioinks showed satisfactory printability, mechanical stability, and biocompatibility. The sustained release of BMP-4 from MSNs induced M2 macrophage polarization and thereby inhibited inflammatory reactions. Loading of BMP-4 and secretion of BMP-2 by M2 type macrophages accelerated bone repair in DM bone defects.
Collapse
Affiliation(s)
- Xin Sun
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Zhenjiang Ma
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Xue Zhao
- Department of Radiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China.,Department of Radiology, Minhang Hospital of Fudan University, Minhang Central Hospital, No. 170 Xinsong Road, Shanghai 201100, China
| | - Wenjie Jin
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Chenyu Zhang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Jie Ma
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Lei Qiang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China.,Southwest JiaoTong University College of Medicine, No. 111 North 1st Section of Second Ring Road, Chengdu, 610031, China
| | - Wenhao Wang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China.,Southwest JiaoTong University College of Medicine, No. 111 North 1st Section of Second Ring Road, Chengdu, 610031, China
| | - Qian Deng
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China.,Southwest JiaoTong University College of Medicine, No. 111 North 1st Section of Second Ring Road, Chengdu, 610031, China
| | - Han Yang
- School of Biomedical Engineering, Shanghai JiaoTong University, No. 1956 Huashan Road, Shanghai, 200030, China
| | - Jinzhong Zhao
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, Shanghai 200233, China
| | - Qianqian Liang
- Spine Institute, Shanghai University of Traditional Chinese Medicine, No.1200 Cailun Road, Shanghai 200032, China
| | - Xiaojun Zhou
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, No. 2999, North Renmin Road, Shanghai 201620, China
| | - Tao Li
- Department of Orthopaedics, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Jinwu Wang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
| |
Collapse
|
37
|
王 宁, 陈 俊, 陈 西, 朱 伦, 段 江, 王 烨, 贝 朝. [Effect of lentivirus-mediated silencing of P75 neurotrophin receptor gene on osteogenic differentiation of bone marrow mesenchymal stem cells in rats]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2020; 34:1052-1058. [PMID: 32794678 PMCID: PMC8171920 DOI: 10.7507/1002-1892.201912086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/23/2020] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To investigate the effect of small interfering RNA (siRNA) lentivirus-mediated silencing of P75 neurotrophin receptor (P75NTR) gene on osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) in rats. METHODS Three lentivirus-mediated P75NTR gene siRNA sequences (P75NTR-siRNA-1, 2, 3) and negative control (NC)-siRNA were designed and transfected into the 3rd generation Sprague Dawley (SD) rat BMSCs. The cells morphological changes were observed under an inverted microscope, and the expressions of P75NTR gene and protein in cells were detected by real-time fluorescence quantitative PCR and Western blot. Then the best silencing P75NTR-siRNA for subsequent osteogenic differentiation experiments was screened out. The 3rd generation SD rat BMSCs were randomly divided into experimental group, negative control group, and blank control group (normal BMSCs). The BMSCs of negative control group and experimental group were transfected with NC-siRNA and the selected P75NTR-siRNA lentiviral vector, respectively. The cells of each group were cultured by osteogenic induction. The expressions of osteogenic related proteins [osteocalcin (OCN) and Runx related transcription factor 2 (Runx2)] were detected by Western blot; the collagen type Ⅰ expression was observed by immunohistochemical staining; the osteogenesis of BMSCs was observed by alkaline phosphatase (ALP) detection and alizarin red staining. RESULTS After lentivirus-mediated P75NTR transfected into BMSCs, the expressions of P75NTR mRNA and protein significantly reduced ( P<0.05), and the best silencing P75NTR-siRNA was P75NTR-siRNA-3. After P75NTR gene was silenced, MTT test showed that the cell proliferation in the experimental group was significantly faster than those in the two control groups ( P<0.05). After osteogenic induction, the relative expressions of OCN and Runx2 proteins, collagen type Ⅰ expression, and ALP activity were significantly higher in the experimental group than in the two control groups, the differences were significant ( P<0.05). With the prolongation of osteogenic induction, the mineralized nodules in the experimental group gradually increased. CONCLUSION Silencing the P75NTR gene with siRNA lentivirus can promote the osteogenic differentiation of rat BMSCs and provide a new idea for the treatment of bone defects.
Collapse
Affiliation(s)
- 宁 王
- 桂林医学院附属医院四肢创伤手外科(广西桂林 541000)Department of Trauma and Limb Surgery, Affiliated Hospital of Guilin Medical College, Guilin Guangxi, 541000, P.R.China
| | - 俊毅 陈
- 桂林医学院附属医院四肢创伤手外科(广西桂林 541000)Department of Trauma and Limb Surgery, Affiliated Hospital of Guilin Medical College, Guilin Guangxi, 541000, P.R.China
| | - 西淼 陈
- 桂林医学院附属医院四肢创伤手外科(广西桂林 541000)Department of Trauma and Limb Surgery, Affiliated Hospital of Guilin Medical College, Guilin Guangxi, 541000, P.R.China
| | - 伦井 朱
- 桂林医学院附属医院四肢创伤手外科(广西桂林 541000)Department of Trauma and Limb Surgery, Affiliated Hospital of Guilin Medical College, Guilin Guangxi, 541000, P.R.China
| | - 江涛 段
- 桂林医学院附属医院四肢创伤手外科(广西桂林 541000)Department of Trauma and Limb Surgery, Affiliated Hospital of Guilin Medical College, Guilin Guangxi, 541000, P.R.China
| | - 烨 王
- 桂林医学院附属医院四肢创伤手外科(广西桂林 541000)Department of Trauma and Limb Surgery, Affiliated Hospital of Guilin Medical College, Guilin Guangxi, 541000, P.R.China
| | - 朝涌 贝
- 桂林医学院附属医院四肢创伤手外科(广西桂林 541000)Department of Trauma and Limb Surgery, Affiliated Hospital of Guilin Medical College, Guilin Guangxi, 541000, P.R.China
| |
Collapse
|
38
|
Wang X, Wang H, Zhang T, Cai L, Kong C, He J. Current Knowledge Regarding the Interaction Between Oral Bone Metabolic Disorders and Diabetes Mellitus. Front Endocrinol (Lausanne) 2020; 11:536. [PMID: 32903738 PMCID: PMC7438828 DOI: 10.3389/fendo.2020.00536] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 07/01/2020] [Indexed: 12/16/2022] Open
Abstract
Diabetes mellitus, a major chronic disease affecting human health, has been increasing in prevalence in recent years. Diabetes mellitus can cause bone metabolic disorders in patients, leading to osteoporosis, a higher risk of traumatic fracture, and other bone diseases. Bone metabolic disorders in the oral cavity principally manifest as periodontitis, loss of alveolar bone, and failure of implant osseointegration. In recent years, numerous studies have shown that there is a complex interaction between bone metabolic disorders and diabetes mellitus. This paper reviews the adverse effects of diabetes on oral bone metabolism disorders such as alveolar osteoporosis and bone loss in patients with periodontitis, discusses the potential mechanisms of diabetic bone loss, and suggests potential ways to prevent and treat oral bone loss in patients with diabetes mellitus.
Collapse
Affiliation(s)
- Xiaofeng Wang
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun, China
- Department of Pediatrics, Pediatric Research Institute, The University of Louisville School of Medicine, Louisville, KY, United States
| | - Huiyu Wang
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Tianfu Zhang
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Lu Cai
- Department of Pediatrics, Pediatric Research Institute, The University of Louisville School of Medicine, Louisville, KY, United States
- Departments of Radiation Oncology, Pharmacology, and Toxicology, University of Louisville, Louisville, KY, United States
| | - Chenfei Kong
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jinting He
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
39
|
Khajuria DK, Soliman M, Elfar JC, Lewis GS, Abraham T, Kamal F, Elbarbary RA. Aberrant structure of fibrillar collagen and elevated levels of advanced glycation end products typify delayed fracture healing in the diet-induced obesity mouse model. Bone 2020; 137:115436. [PMID: 32439570 PMCID: PMC7938873 DOI: 10.1016/j.bone.2020.115436] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/17/2020] [Accepted: 05/14/2020] [Indexed: 12/16/2022]
Abstract
Impaired fracture healing in patients with obesity-associated type 2 diabetes (T2D) is a significant unmet clinical problem that affects millions of people worldwide. However, the underlying causes are poorly understood. Additionally, limited clinical information is available on how pre-diabetic hyperglycemia in obese individuals impacts bone healing. Here, we use the diet-induced obesity (DIO) mouse (C57BL/6J) model to study the impact of obesity-associated pre-diabetic hyperglycemia on bone healing and fibrillar collagen organization as healing proceeds from one phase to another. We show that DIO mice exhibit defective healing characterized by reduced bone mineral density, bone volume, and bone volume density. Differences in the healing pattern between lean and DIO mice occur early in the healing process as evidenced by faster resorption of the fibrocartilaginous callus in DIO mice. However, the major differences between lean and DIO mice occur during the later phases of endochondral ossification and bone remodeling. Comprehensive analyses of fibrillar collagen microstructure and expression pattern during these phases, using a set of complementary techniques that include histomorphometry, immunofluorescence staining, and second harmonic generation microscopy, demonstrate significant defects in DIO mice. Defects include strikingly sparse and disorganized collagen fibers, as well as pathological accumulation of unfolded collagen triple helices. We also demonstrate that DIO-associated changes in fibrillar collagen structure are attributable, at least in part, to the accumulation of advanced glycation end products, which increase the collagen-fiber crosslink density. These major changes impair fibrillar collagens functions, culminating in defective callus mineralization, remodeling, and strength. Our data extend the understanding of mechanisms by which obesity and its associated hyperglycemia impair fracture healing and underline defective fibrillar collagen microstructure as a novel and important contributor.
Collapse
Affiliation(s)
- Deepak Kumar Khajuria
- Department of Orthopaedics and Rehabilitation, The Pennsylvania State University College of Medicine, Hershey, PA, USA; Center for Orthopaedic Research and Translational Science (CORTS), The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Marwa Soliman
- Department of Orthopaedics and Rehabilitation, The Pennsylvania State University College of Medicine, Hershey, PA, USA; Center for Orthopaedic Research and Translational Science (CORTS), The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - John C Elfar
- Department of Orthopaedics and Rehabilitation, The Pennsylvania State University College of Medicine, Hershey, PA, USA; Center for Orthopaedic Research and Translational Science (CORTS), The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Gregory S Lewis
- Department of Orthopaedics and Rehabilitation, The Pennsylvania State University College of Medicine, Hershey, PA, USA; Center for Orthopaedic Research and Translational Science (CORTS), The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Thomas Abraham
- Microscopy Imaging Facility, The Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Neural and Behavioural Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Fadia Kamal
- Department of Orthopaedics and Rehabilitation, The Pennsylvania State University College of Medicine, Hershey, PA, USA; Center for Orthopaedic Research and Translational Science (CORTS), The Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Reyad A Elbarbary
- Department of Orthopaedics and Rehabilitation, The Pennsylvania State University College of Medicine, Hershey, PA, USA; Center for Orthopaedic Research and Translational Science (CORTS), The Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA; Center for RNA Molecular Biology, Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
40
|
Chen L, Wang CT, Forsyth NR, Wu P. Transcriptional profiling reveals altered biological characteristics of chorionic stem cells from women with gestational diabetes. Stem Cell Res Ther 2020; 11:319. [PMID: 32711583 PMCID: PMC7382800 DOI: 10.1186/s13287-020-01828-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 06/30/2020] [Accepted: 07/13/2020] [Indexed: 12/17/2022] Open
Abstract
Background Gestational diabetes (GDM) is a common complication of pregnancy. The impact of pregnancy complications on placental function suggests that extraembryonic stem cells in the placenta may also be affected during pregnancy. Neonatal tissue-derived stem cells, with the advantages of their differentiation capacity and non-invasive isolation processes, have been proposed as a promising therapeutic avenue for GDM management through potential cell therapy approaches. However, the influence of GDM on autologous stem cells remains unclear. Thus, studies that provide comprehensive understanding of stem cells isolated from women with GDM are essential to guide future clinical applications. Methods Human chorionic membrane-derived stem cells (CMSCs) were isolated from placentas of healthy and GDM pregnancies. Transcriptional profiling was performed by DNA microarray, and differentially regulated genes between GDM- and Healthy-CMSCs were used to analyse molecular functions, differentiation, and pathway enrichment. Altered genes and biological functions were validated via real-time PCR and in vitro assays. Results GDM-CMSCs displayed, vs. Healthy-CMSCs, 162 upregulated genes associated with increased migration ability, epithelial development, and growth factor-associated signal transduction while the 269 downregulated genes were strongly linked to angiogenesis and cellular metabolic processes. Notably, significantly reduced expression of detoxification enzymes belonging to the aldehyde dehydrogenase gene families (ALDH1A1/1A2, ALDH2, ALDH3) accounted for downregulation across several metabolic pathways. ALDH activity and inhibitor assays indicated that reduced gene expression of ALDHs affected ALDH enzymatic functions and resulted in oxidative stress dysregulation in GDM-CMSCs. Conclusion Our combined transcriptional analysis and in vitro functional characterisation have provided novel insights into fundamental biological differences in GDM- and Healthy-CMSCs. Enhanced mobility of GDM-CMSCs may promote MSC migration toward injured sites; however, impaired cellular metabolic activity may negatively affect any perceived benefit.
Collapse
Affiliation(s)
- Liyun Chen
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Thornburrow Drive, Stoke-on-Trent, UK.,Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - Chung-Teng Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Nicholas R Forsyth
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Thornburrow Drive, Stoke-on-Trent, UK. .,School of Life Science, Guangzhou University, Guangzhou, 510006, China.
| | - Pensee Wu
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Thornburrow Drive, Stoke-on-Trent, UK.,Academic Unit of Obstetrics and Gynaecology, University Hospital of North Midlands, Stoke-on-Trent, UK.,Keele Cardiovascular Research Group, School of Primary, Community, and Social Care, Keele University, Stoke-on-Trent, UK
| |
Collapse
|
41
|
Skubis-Sikora A, Sikora B, Witkowska A, Mazurek U, Gola J. Osteogenesis of adipose-derived stem cells from patients with glucose metabolism disorders. Mol Med 2020; 26:67. [PMID: 32615920 PMCID: PMC7331176 DOI: 10.1186/s10020-020-00192-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 06/19/2020] [Indexed: 12/13/2022] Open
Abstract
Background Adipose derived stem cells (ADSCs) are clinically widely used somatic stem cells obtained from white adipose tissue. They are characterized by ability to differentiate e.g. into osteoblasts and might successfully regenerate bone tissue in fracture repair. However, the main problem of somatic stem cells is a documented influence of various diseases, drugs or age which can inhibit cells activity. Therefore, in the present study, we investigated the influence of insulin resistance (IR) and type 2 diabetes (T2D) on the proliferation and differentiation potential of ADSCs. Methods The fat from subcutaneous abdominal adipose tissue was acquired by lipoaspiration from 23 voluntary participants, divided into three groups: with diabetes type 2, with insulin resistance and control healthy donors. The proliferative potential was analyzed by cell cytotoxicity assays and by mRNA expression of genes connected with proliferation. Flow cytometry was done for identifying proteins characteristic for mesenchymal stem cells and an analysis of osteogenic differentiation potential based on the assessment of osteogenic markers by real time RT-qPCR, and the evaluation of calcium deposition were also performed. Results The results showed that diabetes type 2 lowered the activity of ADSCs in proliferation assays and changed their phenotypical characteristics. Interestingly, we observed differences in the proliferation potential of ADSCs in patients with insulin resistance, which is often the first phase of diabetes, compared to the control. It might suggest that insulin resistance, early-stage T2D, alters the activity of cells. Moreover, expression of osteogenesis markers was higher in cells from T2D patients than in cells from patients with IR and control. Conclusion We conclude that type 2 diabetes changes the activity of stem cells, and insulin resistance influences on the proliferation of ADSCs.
Collapse
Affiliation(s)
- Aleksandra Skubis-Sikora
- Department of Cytophysiology, Chair of Histology and Embryology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, ul. Medyków 18, C2/108, 40-752, Katowice, Poland
| | - Bartosz Sikora
- Department of Cytophysiology, Chair of Histology and Embryology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, ul. Medyków 18, C2/108, 40-752, Katowice, Poland.
| | | | - Urszula Mazurek
- Józef Tyszkiewicz Higher School in Bielsko-Biała, ul, Nadbrzeżna 12, 43-300, Bielsko-Biała, Poland
| | - Joanna Gola
- Department of Molecular Biology, Chair of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Katowice, Poland
| |
Collapse
|
42
|
Muire PJ, Mangum LH, Wenke JC. Time Course of Immune Response and Immunomodulation During Normal and Delayed Healing of Musculoskeletal Wounds. Front Immunol 2020; 11:1056. [PMID: 32582170 PMCID: PMC7287024 DOI: 10.3389/fimmu.2020.01056] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 05/01/2020] [Indexed: 12/20/2022] Open
Abstract
Single trauma injuries or isolated fractures are often manageable and generally heal without complications. In contrast, high-energy trauma results in multi/poly-trauma injury patterns presenting imbalanced pro- and anti- inflammatory responses often leading to immune dysfunction. These injuries often exhibit delayed healing, leading to fibrosis of injury sites and delayed healing of fractures depending on the intensity of the compounding traumas. Immune dysfunction is accompanied by a temporal shift in the innate and adaptive immune cells distribution, triggered by the overwhelming release of an arsenal of inflammatory mediators such as complements, cytokines and damage associated molecular patterns (DAMPs) from necrotic cells. Recent studies have implicated this dysregulated inflammation in the poor prognosis of polytraumatic injuries, however, interventions focusing on immunomodulating inflammatory cellular composition and activation, if administered incorrectly, can result in immune suppression and unintended outcomes. Immunomodulation therapy is promising but should be conducted with consideration for the spatial and temporal distribution of the immune cells during impaired healing. This review describes the current state of knowledge in the spatiotemporal distribution patterns of immune cells at various stages during musculoskeletal wound healing, with a focus on recent advances in the field of Osteoimmunology, a study of the interface between the immune and skeletal systems, in long bone fractures. The goals of this review are to (1) discuss wound and fracture healing processes of normal and delayed healing in skeletal muscles and long bones; (2) provide a balanced perspective on temporal distributions of immune cells and skeletal cells during healing; and (3) highlight recent therapeutic interventions used to improve fracture healing. This review is intended to promote an understanding of the importance of inflammation during normal and delayed wound and fracture healing. Knowledge gained will be instrumental in developing novel immunomodulatory approaches for impaired healing.
Collapse
Affiliation(s)
- Preeti J. Muire
- Orthopaedic Trauma Research Department, US Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | | | | |
Collapse
|
43
|
Administration of Human Non-Diabetic Mesenchymal Stromal Cells to a Murine Model of Diabetic Fracture Repair: A Pilot Study. Cells 2020; 9:cells9061394. [PMID: 32503335 PMCID: PMC7348854 DOI: 10.3390/cells9061394] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 01/05/2023] Open
Abstract
Individuals living with type 1 diabetes mellitus may experience an increased risk of long bone fracture. These fractures are often slow to heal, resulting in delayed reunion or non-union. It is reasonable to theorize that the underlying cause of these diabetes-associated osteopathies is faulty repair dynamics as a result of compromised bone marrow progenitor cell function. Here it was hypothesized that the administration of non-diabetic, human adult bone marrow-derived mesenchymal stromal cells (MSCs) would enhance diabetic fracture healing. Human MSCs were locally introduced to femur fractures in streptozotocin-induced diabetic mice, and the quality of de novo bone was assessed eight weeks later. Biodistribution analysis demonstrated that the cells remained in situ for three days following administration. Bone bridging was evident in all animals. However, a large reparative callus was retained, indicating non-union. µCT analysis elucidated comparable callus dimensions, bone mineral density, bone volume/total volume, and volume of mature bone in all groups that received cells as compared to the saline-treated controls. Four-point bending evaluation of flexural strength, flexural modulus, and total energy to re-fracture did not indicate a statistically significant change as a result of cellular administration. An ex vivo lymphocytic proliferation recall assay indicated that the xenogeneic administration of human cells did not result in an immune response by the murine recipient. Due to this dataset, the administration of non-diabetic bone marrow-derived MSCs did not support fracture healing in this pilot study.
Collapse
|
44
|
Zamarioli A, de Andrade Staut C, Volpon JB. Review of Secondary Causes of Osteoporotic Fractures Due to Diabetes and Spinal Cord Injury. Curr Osteoporos Rep 2020; 18:148-156. [PMID: 32147752 DOI: 10.1007/s11914-020-00571-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW The aim of this review is to gain a better understanding of osteoporotic fractures and the different mechanisms that are driven in the scenarios of bone disuse due to spinal cord injury and osteometabolic disorders due to diabetes. RECENT FINDINGS Despite major advances in understanding the pathogenesis, prevention, and treatment of osteoporosis, the high incidence of impaired fracture healing remains an important complication of bone loss, leading to marked impairment of the health of an individual and economic burden to the medical system. This review underlines several pathways leading to bone loss and increased risk for fractures. Specifically, we addressed the different mechanisms leading to bone loss after a spinal cord injury and diabetes. Finally, it also encompasses the changes responsible for impaired bone repair in these scenarios, which may be of great interest for future studies on therapeutic approaches to treat osteoporosis and osteoporotic fractures.
Collapse
Affiliation(s)
- Ariane Zamarioli
- Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil.
| | - Caio de Andrade Staut
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - José B Volpon
- Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
45
|
Alder KD, White AHA, Chung Y, Lee I, Back J, Kwon H, Cahill SV, Hao Z, Li L, Chen F, Lee S, Riedel MD, Lee FY. Systemic Parathyroid Hormone Enhances Fracture Healing in Multiple Murine Models of Type 2 Diabetes Mellitus. JBMR Plus 2020; 4:e10359. [PMID: 32382692 PMCID: PMC7202418 DOI: 10.1002/jbm4.10359] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/28/2020] [Accepted: 03/07/2020] [Indexed: 12/30/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a multisystemic disease that afflicts more than 415 million people globally-the incidence and prevalence of T2DM continues to rise. It is well-known that T2DM has detrimental effects on bone quality that increase skeletal fragility, which predisposes subjects to an increased risk of fracture and fracture healing that results in non- or malunion. Diabetics have been found to have perturbations in metabolism, hormone production, and calcium homeostasis-particularly PTH expression-that contribute to the increased risk of fracture and decreased fracture healing. Given the perturbations in PTH expression and the establishment of hPTH (1-34) for use in age-related osteoporosis, it was determined logical to attempt to ameliorate the bone phenotype found in T2DM using hPTH (1-34). Therefore, the present study had two aims: (i) to establish a suitable murine model of the skeletal fragility present in T2DM because no current consensus model exists; and (ii) to determine the effects of hPTH (1-34) on bone fractures in T2DM. The results of the present study suggest that the polygenic mouse of T2DM, TALLYHO/JngJ, most accurately recapitulates the diabetic osteoporotic phenotype seen in humans and that the intermittent systemic administration of hPTH (1-34) increases fracture healing in T2DM murine models by increasing the proliferation of mesenchymal stem cells. © 2020 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Kareme D Alder
- Department of Orthopædics & RehabilitationYale University, School of MedicineNew HavenCTUSA
| | - Andrew HA White
- Department of Orthopædics & RehabilitationYale University, School of MedicineNew HavenCTUSA
| | - Yeon‐Ho Chung
- Department of Orthopædics & RehabilitationYale University, School of MedicineNew HavenCTUSA
| | - Inkyu Lee
- Department of Orthopædics & RehabilitationYale University, School of MedicineNew HavenCTUSA
- Department of Life ScienceChung‐Ang UniversitySeoulRepublic of Korea
| | - JungHo Back
- Department of Orthopædics & RehabilitationYale University, School of MedicineNew HavenCTUSA
| | - Hyuk‐Kwon Kwon
- Department of Orthopædics & RehabilitationYale University, School of MedicineNew HavenCTUSA
| | - Sean V Cahill
- Department of Orthopædics & RehabilitationYale University, School of MedicineNew HavenCTUSA
| | - Zichen Hao
- Department of Orthopædics & RehabilitationYale University, School of MedicineNew HavenCTUSA
| | - Lu Li
- Department of Orthopædics & RehabilitationYale University, School of MedicineNew HavenCTUSA
| | - Fancheng Chen
- Department of Orthopædics & RehabilitationYale University, School of MedicineNew HavenCTUSA
| | - Saelim Lee
- Department of Orthopædics & RehabilitationYale University, School of MedicineNew HavenCTUSA
| | - Matthew D Riedel
- Department of Orthopædics & RehabilitationYale University, School of MedicineNew HavenCTUSA
| | - Francis Y Lee
- Department of Orthopædics & RehabilitationYale University, School of MedicineNew HavenCTUSA
| |
Collapse
|
46
|
Wang S, Wang L, Shi S, Wang X, He C, Yuan L, Ding F, Song Y, Zhang S. Inhibition of GDF11 could promote bone healing in the tooth extraction socket and facilitate mesenchymal stem cell osteogenic differentiation in T2DM pigs. J Periodontol 2020; 91:1645-1652. [PMID: 32281654 DOI: 10.1002/jper.20-0011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/29/2020] [Accepted: 03/18/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND Growth differentiation factor 11 (GDF11) might be a key factor responsible for the weakening of mesenchymal stem cell (MSC) osteogenic differentiation in tooth extraction sockets in patients with type 2 diabetes mellitus (T2DM). This study aimed to confirm that inhibition of GDF11 could promote bone healing in tooth extraction sockets and facilitate MSC osteogenic differentiation under T2DM conditions. METHODS Three streptozotocin-induced T2DM pig models and two control pig models were established. The T2DM pigs were treated with an intrasocket injection of GDF11 inhibitor in the left mandible, whereas the right side was maintained for natural healing. The postextraction socket healing of the T2DM pigs was compared with that of nondiabetic controls. Healing was quantitatively verified by microcomputed tomography, and the GDF11 expression level was detected. MSCs from T2DM pig sockets were cultured and treated with a GDF11 inhibitor. The osteogenic differentiation ability of MSCs was also compared among groups. RESULTS The expression of GDF11 in the tooth extraction sockets from T2DM pigs increased significantly post extraction. Bone healing was promoted by periodic injection of the GDF11 inhibitor into the extraction sockets of T2DM pigs. Furthermore, the osteogenic differentiation ability of T2DM-MSCs was improved in pigs treated with the GDF11 inhibitor. CONCLUSIONS GDF11 inhibition could promote bone healing in the tooth extraction socket and facilitate MSC osteogenic differentiation under T2DM conditions. GDF11 could be a potential therapeutic target for undesirable alveolar bone healing in T2DM patients.
Collapse
Affiliation(s)
- Shuyan Wang
- Department of Oral Disease, School of Stomatology, The Air Force Medical University, Xi'an, China
| | - Lei Wang
- Department of Implant Dentistry, School of Stomatology, The Air Force Medical University, Xi'an, China
| | - Shaojie Shi
- Department of Implant Dentistry, School of Stomatology, The Air Force Medical University, Xi'an, China
| | - Xingxing Wang
- Department of Implant Dentistry, School of Stomatology, The Air Force Medical University, Xi'an, China
| | - Chunxia He
- Institute of Basic Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Lijuan Yuan
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Feng Ding
- Department of Implant Dentistry, School of Stomatology, The Air Force Medical University, Xi'an, China
| | - Yingliang Song
- Department of Implant Dentistry, School of Stomatology, The Air Force Medical University, Xi'an, China
| | - Sijia Zhang
- Department of Implant Dentistry, School of Stomatology, The Air Force Medical University, Xi'an, China.,Department of Biochemistry and Molecular Biology, Center for DNA Typing, Air Force Medical University, Xi'an, China
| |
Collapse
|
47
|
Abstract
The susceptibility and severity of periodontal diseases is made more severe by diabetes, with the impact on the disease process inversely proportional to the level of glycemic control. Although type 1 diabetes mellitus and type 2 diabetes mellitus have different etiologies, and their impact on bone is not identical, they share many of the same complications. Studies in animals and humans agree that both forms of diabetes increase inflammatory events in periodontal tissue, impair new bone formation, and increase expression of RANKL in response to bacterial challenge. High levels of glucose, reactive oxygen species, and advanced glycation end-products are found in the periodontium of diabetic individuals and lead to increased activation of nuclear factor-kappa B and expression of inflammatory cytokines such as tumor necrosis factor and interleukin-1. Studies in animals, moreover, suggest that there are multiple cell types in periodontal tissues that are affected by diabetes, including leukocytes, vascular cells, mesenchymal stem cells, periodontal ligament fibroblasts, osteoblasts, and osteocytes. The etiology of periodontal disease involves the host response to bacterial challenge that is affected by diabetes, which increases the expression of RANKL and reduces coupled bone formation. In addition, the inflammatory response also modifies the oral microbiota to render it more pathogenic, as demonstrated by increased inflammation and bone loss in animals where bacteria are transferred from diabetic donors to germ-free hosts compared with transfer from normoglycemic donors. This approach has the advantage of not relying upon limited knowledge of the specific bacterial taxa to determine pathogenicity, and examines the overall impact of the microbiota rather than the presumed pathogenicity of a few bacterial groups. Thus, animal studies have provided new insights into pathogenic mechanisms that identify cause-and-effect relationships that are difficult to perform in human studies.
Collapse
Affiliation(s)
- Dana T Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Zhenjiang Ding
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Pediatric Dentistry, School of Stomatology, China Medical University, Shenyang, China
| | - Yingming Yang
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Preventive Dentistry, West China School of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
48
|
Calciolari E, Donos N. Proteomic and Transcriptomic Approaches for Studying Bone Regeneration in Health and Systemically Compromised Conditions. Proteomics Clin Appl 2020; 14:e1900084. [PMID: 32131137 DOI: 10.1002/prca.201900084] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 02/05/2020] [Indexed: 01/04/2023]
Abstract
Bone regeneration is a complex biological process, where the molecular mechanisms are only partially understood. In an ageing population, where the prevalence of chronic diseases with an impact on bone metabolism is increasing, it becomes crucial to identify new strategies that would improve regenerative outcomes also in medically compromised patients. In this context, omics are demonstrating a great potential, as they offer new insights on the molecular mechanisms regulating physiologic/pathologic bone healing and, at the same time, allow the identification of new diagnostic and therapeutic targets. This review provides an overview on the current evidence on the use of transcriptomic and proteomic approaches in bone regeneration research, particularly in relation to type 1 diabetes and osteoporosis, and discusses future scenarios and potential benefits and limitations on the integration of multi-omics. It is suggested that future research will leverage the synergy of omics with statistical modeling and bioinformatics to prompt the understanding of the biology underpinning bone formation in health and medically compromised conditions. With an eye toward personalized medicine, new strategies combining the mining of large datasets and bioinformatic data with a detailed characterization of relevant phenotypes will need to be pursued to further the understanding of disease mechanisms.
Collapse
Affiliation(s)
- Elena Calciolari
- Centre for Oral Immunobiology and Regenerative Medicine & Centre for Oral Clinical Research, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Turner Street, London, E1 2AD, UK.,Department of Medicine and Surgery, School of Dental Medicine, University of Parma, via Gramsci 14, Parma, 43126, Italy
| | - Nikolaos Donos
- Centre for Oral Immunobiology and Regenerative Medicine & Centre for Oral Clinical Research, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Turner Street, London, E1 2AD, UK
| |
Collapse
|
49
|
Rios-Arce ND, Dagenais A, Feenstra D, Coughlin B, Kang HJ, Mohr S, McCabe LR, Parameswaran N. Loss of interleukin-10 exacerbates early Type-1 diabetes-induced bone loss. J Cell Physiol 2020; 235:2350-2365. [PMID: 31538345 PMCID: PMC6899206 DOI: 10.1002/jcp.29141] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 08/23/2019] [Indexed: 01/08/2023]
Abstract
Type-1 diabetes (T1D) increases systemic inflammation, bone loss, and risk for bone fractures. Levels of the anti-inflammatory cytokine interleukin-10 (IL-10) are decreased in T1D, however their role in T1D-induced osteoporosis is unknown. To address this, diabetes was induced in male IL-10 knockout (KO) and wild-type (WT) mice. Analyses of femur and vertebral trabecular bone volume fraction identified bone loss in T1D-WT mice at 4 and 12 weeks, which in T1D-IL-10-KO mice was further reduced at 4 weeks but not 12 weeks. IL-10 deficiency also increased the negative effects of T1D on cortical bone. Osteoblast marker osterix was decreased, while osteoclast markers were unchanged, suggesting that IL-10 promotes anabolic processes. MC3T3-E1 osteoblasts cultured under high glucose conditions displayed a decrease in osterix which was prevented by addition of IL-10. Taken together, our results suggest that IL-10 is important for promoting osteoblast maturation and reducing bone loss during early stages of T1D.
Collapse
Affiliation(s)
- Naiomy Deliz Rios-Arce
- Department of Physiology, Michigan State University, East Lansing, Michigan
- Comparative Medicine and Integrative Biology Program, Michigan State University, East Lansing, Michigan
| | - Andrew Dagenais
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Derrick Feenstra
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Brandon Coughlin
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Ho Jun Kang
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Susanne Mohr
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Laura R. McCabe
- Department of Physiology, Michigan State University, East Lansing, Michigan
- Department of Radiology, Michigan State University, East Lansing, Michigan
- Biomedical Imaging Research Center, Michigan State University, East Lansing, Michigan
- These authors contributed equally to this work are co-senior and co-corresponding authors
| | - Narayanan Parameswaran
- Department of Physiology, Michigan State University, East Lansing, Michigan
- Comparative Medicine and Integrative Biology Program, Michigan State University, East Lansing, Michigan
- These authors contributed equally to this work are co-senior and co-corresponding authors
| |
Collapse
|
50
|
LIU MIN, ALHARBI MOHAMMED, GRAVES DANA, YANG SHUYING. IFT80 Is Required for Fracture Healing Through Controlling the Regulation of TGF-β Signaling in Chondrocyte Differentiation and Function. J Bone Miner Res 2020; 35:571-582. [PMID: 31643106 PMCID: PMC7525768 DOI: 10.1002/jbmr.3902] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 10/10/2019] [Accepted: 10/17/2019] [Indexed: 12/14/2022]
Abstract
Primary cilia are essential cellular organelles that are anchored at the cell surface membrane to sense and transduce signaling. Intraflagellar transport (IFT) proteins are indispensable for cilia formation and function. Although major advances in understanding the roles of these proteins in bone development have been made, the mechanisms by which IFT proteins regulate bone repair have not been identified. We investigated the role of the IFT80 protein in chondrocytes during fracture healing by creating femoral fractures in mice with conditional deletion of IFT80 in chondrocytes utilizing tamoxifen inducible Col2α1-CreER mice. Col2α1cre IFT80f/f mice had smaller fracture calluses than IFT80f/f (control) mice. The max-width and max-callus area were 31% and 48% smaller than those of the control mice, respectively. Col2α1cre IFT80f/f mice formed low-density/porous woven bony tissue with significantly lower ratio of bone volume, Trabecular (Tb) number and Tb thickness, and greater Tb spacing compared to control mice. IFT80 deletion significantly downregulated the expression of angiogenesis markers-VEGF, PDGF and angiopoietin and inhibited fracture callus vascularization. Mechanistically, loss of IFT80 in chondrocytes resulted in a decrease in cilia formation and chondrocyte proliferation rate in fracture callus compared to the control mice. Meanwhile, IFT80 deletion downregulated the TGF-β signaling pathway by inhibiting the expression of TGF-βI, TGF-βR, and phosphorylation of Smad2/3 in the fracture callus. In primary chondrocyte cultures in vitro, IFT80 deletion dramatically reduced chondrocyte proliferation, cilia assembly, and chondrogenic gene expression and differentiation. Collectively, our findings demonstrate that IFT80 and primary cilia play an essential role in fracture healing, likely through controlling chondrocyte proliferation and differentiation, and the TGF-β signaling pathway. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- MIN LIU
- Dept. of Anatomy and Cell Biology, University of
Pennsylvania, Philadelphia, PA
| | - MOHAMMED ALHARBI
- Dept. of Endodontics, Faculty of Dentistry, King Abdulaziz
University, Saudi Arabia
| | - DANA GRAVES
- Dept. of Periodontics, School of Dental Medicine,
University of Pennsylvania, Philadelphia, PA
| | - SHUYING YANG
- Dept. of Anatomy and Cell Biology, University of
Pennsylvania, Philadelphia, PA
| |
Collapse
|