1
|
Liu ZX, Liu GQ, Lin ZX, Chen YQ, Chen P, Hu YJ, Yu B, Jiang N. Effects of Staphylococcus aureus on stem cells and potential targeted treatment of inflammatory disorders. Stem Cell Res Ther 2024; 15:187. [PMID: 38937829 PMCID: PMC11210046 DOI: 10.1186/s13287-024-03781-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/02/2024] [Indexed: 06/29/2024] Open
Abstract
Due to the advanced studies on stem cells in developmental biology, the roles of stem cells in the body and their phenotypes in related diseases have not been covered clearly. Meanwhile, with the intensive research on the mechanisms of stem cells in regulating various diseases, stem cell therapy is increasingly being attention because of its effectiveness and safety. As one of the most widely used stem cell in stem cell therapies, hematopoietic stem cell transplantation shows huge advantage in treatment of leukemia and other blood-malignant diseases. Besides, due to the effect of anti-inflammatory and immunomodulatory, mesenchymal stem cells could be a potential therapeutic strategy for variety infectious diseases. In this review, we summarized the effects of Staphylococcus aureus (S. aureus) and its components on different types of adult stem cells and their downstream signaling pathways. Also, we reviewed the roles of different kinds of stem cells in various disease models caused by S. aureus, providing new insights for applying stem cell therapy to treat infectious diseases.
Collapse
Affiliation(s)
- Zi-Xian Liu
- Division of Orthopaedics & Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Guan-Qiao Liu
- Division of Orthopaedics & Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
| | - Ze-Xin Lin
- Division of Orthopaedics & Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
| | - Ying-Qi Chen
- Division of Orthopaedics & Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
| | - Peng Chen
- Division of Orthopaedics & Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
| | - Yan-Jun Hu
- Division of Orthopaedics & Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
| | - Bin Yu
- Division of Orthopaedics & Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China.
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China.
| | - Nan Jiang
- Division of Orthopaedics & Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China.
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China.
| |
Collapse
|
2
|
Wang H, Lin S, Feng L, Huang B, Lu X, Yang Z, Jiang Z, Li Y, Zhang X, Wang M, Wang B, Kong L, Pan Q, Bai S, Li Y, Yang Y, Lee WYW, Currie PD, Lin C, Jiang Y, Chen J, Tortorella MD, Li H, Li G. Low-Dose Staphylococcal Enterotoxin C2 Mutant Maintains Bone Homeostasis via Regulating Crosstalk between Bone Formation and Host T-Cell Effector Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300989. [PMID: 37552005 PMCID: PMC10558680 DOI: 10.1002/advs.202300989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/22/2023] [Indexed: 08/09/2023]
Abstract
Studies in recent years have highlighted an elaborate crosstalk between T cells and bone cells, suggesting that T cells may be alternative therapeutic targets for the maintenance of bone homeostasis. Here, it is reported that systemic administration of low-dose staphylococcal enterotoxin C2 (SEC2) 2M-118, a form of mutant superantigen, dramatically alleviates ovariectomy (OVX)-induced bone loss via modulating T cells. Specially, SEC2 2M-118 treatment increases trabecular bone mass significantly via promoting bone formation in OVX mice. These beneficial effects are largely diminished in T-cell-deficient nude mice and can be rescued by T-cell reconstruction. Neutralizing assays determine interferon gamma (IFN-γ) as the key factor that mediates the beneficial effects of SEC2 2M-118 on bone. Mechanistic studies demonstrate that IFN-γ stimulates Janus kinase/signal transducer and activator of transcription (JAK-STAT) signaling, leading to enhanced production of nitric oxide, which further activates p38 mitogen-activated protein kinase (MAPK) and Runt-related transcription factor 2 (Runx2) signaling and promotes osteogenic differentiation. IFN-γ also directly inhibits osteoclast differentiation, but this effect is counteracted by proabsorptive factors tumor necrosis factor alpha (TNF-α) and interleukin 1 beta (IL-1β) secreted from IFN-γ-stimulated macrophages. Taken together, this work provides clues for developing innovative approaches which target T cells for the prevention and treatment of osteoporosis.
Collapse
Affiliation(s)
- Haixing Wang
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong999077China
- Centre for Regenerative Medicine and HealthHong Kong Institute of Science & InnovationChinese Academy of SciencesHong Kong999077China
| | - Sien Lin
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong999077China
| | - Lu Feng
- Centre for Regenerative Medicine and HealthHong Kong Institute of Science & InnovationChinese Academy of SciencesHong Kong999077China
| | - Baozhen Huang
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong999077China
| | - Xuan Lu
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong999077China
| | - Zhengmeng Yang
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong999077China
| | - Zhaowei Jiang
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong999077China
| | - Yu‐Cong Li
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong999077China
| | - Xiaoting Zhang
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong999077China
| | - Ming Wang
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong999077China
| | - Bin Wang
- Greater Bay Area Institute of Precision Medicine (Guangzhou)Fudan University2nd Nanjiang Rd, Nansha DistrictGuangzhou511458China
| | - Lingchi Kong
- Department of Orthopaedic SurgeryShanghai Jiao Tong University Affiliated Sixth People's HospitalYishan Rd. 600Shanghai200233China
| | - Qi Pan
- Department of OrthopaedicsSouth China HospitalShenzhen UniversityShenzhen518116China
| | - Shanshan Bai
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong999077China
| | - Yuan Li
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong999077China
| | - Yongkang Yang
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong999077China
| | - Wayne Yuk Wai Lee
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong999077China
| | - Peter D. Currie
- Australian Regenerative Medicine InstituteMonash UniversityWellington RoadClaytonVictoria3800Australia
| | - Changshuang Lin
- Shenyang Xiehe Biopharmaceutical Co. Ltd.ShenyangLiaoning Province110179China
| | - Yanfu Jiang
- Shenyang Xiehe Biopharmaceutical Co. Ltd.ShenyangLiaoning Province110179China
| | - Juyu Chen
- Shenyang Xiehe Biopharmaceutical Co. Ltd.ShenyangLiaoning Province110179China
| | - Micky D. Tortorella
- Centre for Regenerative Medicine and HealthHong Kong Institute of Science & InnovationChinese Academy of SciencesHong Kong999077China
| | - Hongyi Li
- Shenyang Xiehe Biopharmaceutical Co. Ltd.ShenyangLiaoning Province110179China
| | - Gang Li
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong999077China
| |
Collapse
|
3
|
Feng Z, Jin M, Liang J, Kang J, Yang H, Guo S, Sun X. Insight into the effect of biomaterials on osteogenic differentiation of mesenchymal stem cells: A review from a mitochondrial perspective. Acta Biomater 2023; 164:1-14. [PMID: 36972808 DOI: 10.1016/j.actbio.2023.03.032] [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: 11/16/2022] [Revised: 03/02/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023]
Abstract
Bone damage may be triggered by a variety of factors, and the damaged area often requires a bone graft. Bone tissue engineering can serve as an alternative strategy for repairing large bone defects. Mesenchymal stem cells (MSCs), the progenitor cells of connective tissue, have become an important tool for tissue engineering due to their ability to differentiate into a variety of cell types. The precise regulation of the growth and differentiation of the stem cells used for bone regeneration significantly affects the efficiency of this type of tissue engineering. During the process of osteogenic induction, the dynamics and function of localized mitochondria are altered. These changes may also alter the microenvironment of the therapeutic stem cells and result in mitochondria transfer. Mitochondrial regulation not only affects the induction/rate of differentiation, but also influences its direction, determining the final identity of the differentiated cell. To date, bone tissue engineering research has mainly focused on the influence of biomaterials on phenotype and nuclear genotype, with few studies investigating the role of mitochondria. In this review, we provide a comprehensive summary of researches into the role of mitochondria in MSCs differentiation and critical analysis regarding smart biomaterials that are able to "programme" mitochondria modulation was proposed. STATEMENT OF SIGNIFICANCE: : • This review proposed the precise regulation of the growth and differentiation of the stem cells used to seed bone regeneration. • This review addressed the dynamics and function of localized mitochondria during the process of osteogenic induction and the effect of mitochondria on the microenvironment of stem cells. • This review summarized biomaterials which affect the induction/rate of differentiation, but also influences its direction, determining the final identity of the differentiated cell through the regulation of mitochondria.
Collapse
Affiliation(s)
- Ziyi Feng
- Department of Plastic Surgery, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110002 Liaoning Province, China
| | - Meiqi Jin
- School of Intelligent Medicine, China Medical University, No.77, Puhe Road, Shenyang, 110122, Liaoning Province, China
| | - Junzhi Liang
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping, Shenyang, 110004 Liaoning Province, China
| | - Junning Kang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping, Shenyang, 110004 Liaoning Province, China
| | - Huazhe Yang
- School of Intelligent Medicine, China Medical University, No.77, Puhe Road, Shenyang, 110122, Liaoning Province, China.
| | - Shu Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110002 Liaoning Province, China.
| | - Xiaoting Sun
- School of Forensic Medicine, China Medical University, No.77, Puhe Road, Shenyang, 110122, Liaoning Province, China.
| |
Collapse
|
4
|
Affiliation(s)
- Shao-Ting J. Tsang
- Department of Orthopaedic Surgery, University of Edinburgh, Edinburgh, UK
- Department of Trauma and Orthopaedics, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Nando Ferreira
- Division Orthopaedic Surgery Department of Surgical Sciences, Faculty of Medicine and Health Sciences Stellenbosch University, Cape Town, South Africa
| | | |
Collapse
|
5
|
Li W, Li T, Tang Z, Qi X, Zhou Y, Tang X, Xu W, Xiong H. Taohong Siwu Decoction promotes the process of fracture healing by activating the VEGF-FAK signal pathway and systemically regulating the gut microbiota. J Appl Microbiol 2022; 133:1363-1377. [PMID: 35475538 DOI: 10.1111/jam.15598] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 04/07/2022] [Accepted: 04/25/2022] [Indexed: 11/27/2022]
Abstract
AIMS This study aimed to explore the effect of Taohong Siwu Decoction (THSWD) on Bone marrow mesenchymal stem cells (BMSCs) at the cellular level and the possible mechanism of systemic regulation of gut microbiota on fracture recovery. METHODS AND RESULTS Cell Counting Kit-8 (CCK-8) experiments show that THSWD effectively promotes the proliferation of BMSCs. Transwell and wound healing assays show that THSWD effectively promotes the invasion and migration of BMSCs. Alizarin red staining showed that the THSWD model enhanced the osteogenic differentiation of BMSCs. Moreover, the effect of THSWD on BMSCs is time- and concentration-dependent. RT-qPCR and Western blot results showed that THSWD treatment up-regulated the expression of vascular endothelial growth factor (VEGF) and focal adhesion kinase (FAK) at mRNA and protein levels, respectively. Hematoxylin-eosin and crocin O-quick green staining showed that rats with right femoral shaft fractures, after 14 days of THSWD treatment, the area of callus and cartilage regeneration at the fracture site increased significantly. Gut microbiota was changed in fractured rats, such as the abundance of Bacteroidetes and Firmicutes was increased. THSWD showed positive regulation of both to a certain extent. CONCLUSION THSWD up-regulates VEGF and activates the FAK signaling pathway to enhance the development and differentiation of BMSCs, and systematically regulates the gut microbiota to promote fracture healing. SIGNIFICANCE AND IMPACT OF STUDY This study provides new insights on the cellular and systemic level to understand the mechanism of THSWD in the treatment of fractures.
Collapse
Affiliation(s)
- Wangyang Li
- Hunan University of Chinese Medicine, Changsha, Hunan
| | - Tiao Li
- Department of Respiratory Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan
| | - Zhi Tang
- Xiangtan Chinese Medicine hospital, Xiangtan, Hunan
| | - Xinyu Qi
- Hunan University of Chinese Medicine, Changsha, Hunan
| | - Youliang Zhou
- Department of Emergency First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, Hunan
| | - Xiaolu Tang
- Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan
| | - Weijie Xu
- Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan
| | - Hui Xiong
- Hunan University of Chinese Medicine, Changsha, Hunan
| |
Collapse
|
6
|
Ramirez-GarciaLuna JL, Rangel-Berridi K, Olasubulumi OO, Rosenzweig DH, Henderson JE, Gawri R, Martineau PA. Enhanced Bone Remodeling After Fracture Priming. Calcif Tissue Int 2022; 110:349-366. [PMID: 34668029 DOI: 10.1007/s00223-021-00921-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 10/01/2021] [Indexed: 11/30/2022]
Abstract
The immune system is an active component of bone repair. Mast cells influence the recruitment of macrophages, osteoclasts and blood vessels into the repair tissue. We hypothesized that if mast cells and other immune cells are sensitized to recognize broken bone, they will mount an increased response to subsequent fractures that may be translated into enhanced healing. To test this, we created a bone defect on the left leg of anesthetized mice and 2 weeks later, a second one on the right leg. Bone repair in the right legs was then compared to control mice that underwent the creation of bilateral window bone defects at the same time. Mice were euthanized at 14 and 56 days. Mineralized tissue quantity and morphometric parameters were assessed using micro-CT and histology. The activity of osteoblasts, osteoclasts, vascular endothelial cells, mast cells, and macrophages was evaluated using histochemistry. Our main findings were (1) no significant differences in the amount of bone produced at 14- or 56 days post-operative between groups; (2) mice exposed to subsequent fractures showed significantly better bone morphometric parameters after 56 days post-operative; and (3) significant increases in the content of blood vessels, osteoclasts, and the number of macrophages in the subsequent fracture group. Our results provide strong evidence that a transient increase in the inflammatory state of a healing injury promotes faster bone remodelling and increased neo-angiogenesis. This phenomenon is also characterized by changes in mast cell and macrophage content that translate into more active recruitment of mesenchymal stromal cells.
Collapse
Affiliation(s)
- Jose L Ramirez-GarciaLuna
- Bone Engineering Labs, Injury, Repair & Recovery Program, Research Institute, McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada
- Experimental Surgery, Faculty of Medicine, McGill University, 3605 Rue de la Montagne, Montreal, QC, H3G 2M1, Canada
| | - Karla Rangel-Berridi
- Bone Engineering Labs, Injury, Repair & Recovery Program, Research Institute, McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada
- Biofabrication and Bioengineering Labs, Injury, Repair & Recovery Program, Research Institute, McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada
- Experimental Surgery, Faculty of Medicine, McGill University, 3605 Rue de la Montagne, Montreal, QC, H3G 2M1, Canada
| | - Ore-Oluwa Olasubulumi
- Bone Engineering Labs, Injury, Repair & Recovery Program, Research Institute, McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada
| | - Derek H Rosenzweig
- Biofabrication and Bioengineering Labs, Injury, Repair & Recovery Program, Research Institute, McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada
- Experimental Surgery, Faculty of Medicine, McGill University, 3605 Rue de la Montagne, Montreal, QC, H3G 2M1, Canada
| | - Janet E Henderson
- Bone Engineering Labs, Injury, Repair & Recovery Program, Research Institute, McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada
- Experimental Surgery, Faculty of Medicine, McGill University, 3605 Rue de la Montagne, Montreal, QC, H3G 2M1, Canada
- Experimental Medicine, Faculty of Medicine, McGill University, 3605 Rue de la Montagne, Montreal, QC, H3G 2M1, Canada
| | - Rahul Gawri
- Regenerative Orthopaedics and Innovation Laboratory, Injury, Repair & Recovery Program, Research Institute-McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada.
- Experimental Surgery, Faculty of Medicine, McGill University, 3605 Rue de la Montagne, Montreal, QC, H3G 2M1, Canada.
| | - Paul A Martineau
- Bone Engineering Labs, Injury, Repair & Recovery Program, Research Institute, McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada
- Regenerative Orthopaedics and Innovation Laboratory, Injury, Repair & Recovery Program, Research Institute-McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada
- Experimental Surgery, Faculty of Medicine, McGill University, 3605 Rue de la Montagne, Montreal, QC, H3G 2M1, Canada
| |
Collapse
|
7
|
Gu F, Zhang K, Li J, Xie X, Wen Q, Sui Z, Su Z, Yu T. Changes of Migration, Immunoregulation and Osteogenic Differentiation of Mesenchymal Stem Cells in Different Stages of Inflammation. Int J Med Sci 2022; 19:25-33. [PMID: 34975296 PMCID: PMC8692114 DOI: 10.7150/ijms.58428] [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: 01/20/2021] [Accepted: 10/25/2021] [Indexed: 12/17/2022] Open
Abstract
Bone infection has always been the focus of orthopedic research. Mesenchymal stem cells (MSCs) are the natural progenitors of osteoblasts, and the process of osteogenesis is triggered in response to different signals from the extracellular matrix. MSCs exert important functions including secretion and immune regulation and also play a key role in bone regeneration. The biological behavior of MSCs in acute and chronic inflammation, especially the transformation between acute inflammation and chronic inflammation, has aroused great interest among researchers. This paper reviews the recent literature and summarizes the behavior and biological characteristics of MSCs in acute and chronic inflammation to stimulate further research on MSCs and treatment of bone diseases.
Collapse
Affiliation(s)
- Feng Gu
- Department of Orthopedics, First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Ke Zhang
- Department of Orthopedics, First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Jiangbi Li
- Department of Orthopedics, First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Xiaoping Xie
- Department of Orthopedics, First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Qiangqiang Wen
- Department of Orthopedics, First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Zhenjiang Sui
- Department of Orthopedics, First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Zilong Su
- Department of Orthopedics, First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Tiecheng Yu
- Department of Orthopedics, First Hospital of Jilin University, Changchun 130021, Jilin, China
| |
Collapse
|
8
|
Patient-specific effects of soluble factors from Staphylococcus aureus and Staphylococcus epidermidis biofilms on osteogenic differentiation of primary human osteoblasts. Sci Rep 2021; 11:17282. [PMID: 34446785 PMCID: PMC8390505 DOI: 10.1038/s41598-021-96719-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 08/13/2021] [Indexed: 11/08/2022] Open
Abstract
Due to the frequency of biofilm-forming Staphylococcus aureus and Staphylococcus epidermidis in orthopedics, it is crucial to understand the interaction between the soluble factors produced by prokaryotes and their effects on eukaryotes. Our knowledge concerning the effect of soluble biofilm factors (SBF) and their virulence potential on osteogenic differentiation is limited to few studies, particularly when there is no direct contact between prokaryotic and eukaryotic cells. SBF were produced by incubating biofilm from S. aureus and S. epidermidis in osteogenic media. Osteoblasts of seven donors were included in this study. Our results demonstrate that the detrimental effects of these pathogens do not require direct contact between prokaryotic and eukaryotic cells. SBF produced by S. aureus and S. epidermidis affect the metabolic activity of osteoblasts. However, the effect of SBF derived from S. aureus seems to be more pronounced compared to that of S. epidermidis. The influence of SBF of S. aureus and S. epidermidis on gene expression of COL1A1, ALPL, BGLAP, SPP1, RUNX2 is bacteria-, patient-, concentration-, and incubation time dependent. Mineralization was monitored by staining the calcium and phosphate deposition and revealed that the SBF of S. epidermidis markedly inhibits calcium deposition; however, S. aureus shows a less inhibitory effect. Therefore, these new findings support the hypotheses that soluble biofilm factors affect the osteogenic processes substantially, particularly when there is no direct interaction between bacteria and osteoblast.
Collapse
|
9
|
Xin W, Yuan S, Wang B, Qian Q, Chen Y. Hsa_circ_0066523 promotes the proliferation and osteogenic differentiation of bone mesenchymal stem cells by repressing PTEN. Bone Joint Res 2021; 10:526-535. [PMID: 34402627 PMCID: PMC8414438 DOI: 10.1302/2046-3758.108.bjr-2020-0127.r2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aims Circular RNAs (circRNAs) are a novel type of non-coding RNA that plays major roles in the development of diverse diseases including osteonecrosis of the femoral head (ONFH). Here, we explored the impact of hsa_circ_0066523 derived from forkhead box P1 (FOXP1) (also called circFOXP1) on bone mesenchymal stem cells (BMSCs), which is important for ONFH development. Methods RNA or protein expression in BMSCs was analyzed by quantitative real-time polymerase chain reaction (qRT-PCR) or western blot, respectively. Cell Counting Kit 8 (CCK8) and 5-ethynyl-2’-deoxyuridine (EdU) were used to analyze cell proliferation. Alkaline phosphatase (ALP) activity, ALP staining, and Alizarin Red S staining were employed to evaluate the osteoblastic differentiation. Chromatin immunoprecipitation (ChIP), luciferase reporter, RNA pull down, and RNA immunoprecipitation (RIP) assays were combined for exploring molecular associations. Results Circ_0066523 was upregulated in osteogenic induction process of BMSCs. Silencing circ_0066523 restrained the proliferation and osteogenic differentiation of BMSCs. Mechanistically, circ_0066523 activated phosphatidylinositol-4,5-bisphosphate 3-kinase / AKT serine/threonine kinase 1 (PI3K/AKT) pathway via recruiting lysine demethylase 5B (KDM5B) to epigenetically repress the transcription of phosphatase and tensin homolog (PTEN). Functionally, AKT signalling pathway agonist or PTEN knockdown counteracted the effects of silenced circ_0066523 on BMSC proliferation and differentiation. Conclusion Circ_0066523 promotes the proliferation and differentiation of BMSCs by epigenetically repressing PTEN and therefore activating AKT pathway. This finding might open new avenues for the identification of therapeutic targets for osteoblast differentiation related diseases such as ONFH. Cite this article: Bone Joint Res 2021;10(8):526–535.
Collapse
Affiliation(s)
- Wei Xin
- Department of Orthopedics, Changzheng Hospital, Shanghai, China
| | - Shuai Yuan
- Department of Orthopedics, Changzheng Hospital, Shanghai, China
| | - Bo Wang
- Department of Orthopedics, Changzheng Hospital, Shanghai, China
| | - Qirong Qian
- Department of Orthopedics, Changzheng Hospital, Shanghai, China
| | - Yi Chen
- Department of Orthopedics, Changzheng Hospital, Shanghai, China
| |
Collapse
|
10
|
Augustyniak D, Kramarska E, Mackiewicz P, Orczyk-Pawiłowicz M, Lundy FT. Mammalian Neuropeptides as Modulators of Microbial Infections: Their Dual Role in Defense versus Virulence and Pathogenesis. Int J Mol Sci 2021; 22:ijms22073658. [PMID: 33915818 PMCID: PMC8036953 DOI: 10.3390/ijms22073658] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/28/2021] [Accepted: 03/30/2021] [Indexed: 02/07/2023] Open
Abstract
The regulation of infection and inflammation by a variety of host peptides may represent an evolutionary failsafe in terms of functional degeneracy and it emphasizes the significance of host defense in survival. Neuropeptides have been demonstrated to have similar antimicrobial activities to conventional antimicrobial peptides with broad-spectrum action against a variety of microorganisms. Neuropeptides display indirect anti-infective capacity via enhancement of the host’s innate and adaptive immune defense mechanisms. However, more recently concerns have been raised that some neuropeptides may have the potential to augment microbial virulence. In this review we discuss the dual role of neuropeptides, perceived as a double-edged sword, with antimicrobial activity against bacteria, fungi, and protozoa but also capable of enhancing virulence and pathogenicity. We review the different ways by which neuropeptides modulate crucial stages of microbial pathogenesis such as adhesion, biofilm formation, invasion, intracellular lifestyle, dissemination, etc., including their anti-infective properties but also detrimental effects. Finally, we provide an overview of the efficacy and therapeutic potential of neuropeptides in murine models of infectious diseases and outline the intrinsic host factors as well as factors related to pathogen adaptation that may influence efficacy.
Collapse
Affiliation(s)
- Daria Augustyniak
- Department of Pathogen Biology and Immunology, Faculty of Biology, University of Wroclaw, 51-148 Wroclaw, Poland;
- Correspondence: ; Tel.: +48-71-375-6296
| | - Eliza Kramarska
- Department of Pathogen Biology and Immunology, Faculty of Biology, University of Wroclaw, 51-148 Wroclaw, Poland;
- Institute of Biostructures and Bioimaging, Consiglio Nazionale delle Ricerche, 80134 Napoli, Italy
| | - Paweł Mackiewicz
- Department of Bioinformatics and Genomics, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland;
| | | | - Fionnuala T. Lundy
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK;
| |
Collapse
|
11
|
Antich-Rosselló M, Forteza-Genestra MA, Calvo J, Gayà A, Monjo M, Ramis JM. Platelet-derived extracellular vesicles promote osteoinduction of mesenchymal stromal cells. Bone Joint Res 2020; 9:667-674. [PMID: 33101656 PMCID: PMC7563034 DOI: 10.1302/2046-3758.910.bjr-2020-0111.r2] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aims Platelet concentrates, like platelet-rich plasma (PRP) and platelet lysate (PL), are widely used in regenerative medicine, especially in bone regeneration. However, the lack of standard procedures and controls leads to high variability in the obtained results, limiting their regular clinical use. Here, we propose the use of platelet-derived extracellular vesicles (EVs) as an off-the-shelf alternative for PRP and PL for bone regeneration. In this article, we evaluate the effect of PL-derived EVs on the biocompatibility and differentiation of mesenchymal stromal cells (MSCs). Methods EVs were obtained first by ultracentrifugation (UC) and then by size exclusion chromatography (SEC) from non-activated PL. EVs were characterized by transmission electron microscopy, nanoparticle tracking analysis, and the expression of CD9 and CD63 markers by western blot. The effect of the obtained EVs on osteoinduction was evaluated in vitro on human umbilical cord MSCs by messenger RNA (mRNA) expression analysis of bone markers, alkaline phosphatase activity (ALP), and calcium (Ca2+) content. Results Osteogenic differentiation of MSCs was confirmed when treated with UC-isolated EVs. In order to disprove that the effect was due to co-isolated proteins, EVs were isolated by SEC. Purer EVs were obtained and proved to maintain the differentiation effect on MSCs and showed a dose-dependent response. Conclusion PL-derived EVs present an osteogenic capability comparable to PL treatments, emerging as an alternative able to overcome PL and PRP limitations. Cite this article: Bone Joint Res 2020;9(10):667–674.
Collapse
Affiliation(s)
- Miquel Antich-Rosselló
- Cell Therapy and Tissue Engineering Group, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Maria Antònia Forteza-Genestra
- Cell Therapy and Tissue Engineering Group, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Javier Calvo
- Cell Therapy and Tissue Engineering Group, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.,Fundació Banc de Sang i Teixits de les Illes Balears (FBSTIB), Palma, Spain
| | - Antoni Gayà
- Cell Therapy and Tissue Engineering Group, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.,Fundació Banc de Sang i Teixits de les Illes Balears (FBSTIB), Palma, Spain
| | - Marta Monjo
- Cell Therapy and Tissue Engineering Group, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.,Departament de Biologia Fonamental i Ciències de la Salut, University of the Balearic Islands, Palma, Spain
| | - Joana M Ramis
- Cell Therapy and Tissue Engineering Group, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.,Departament de Biologia Fonamental i Ciències de la Salut, University of the Balearic Islands, Palma, Spain
| |
Collapse
|