1
|
Lee C, Kates SL, Graves ML, Jeray KJ, Javedan H, Firoozabadi R, Schemitsch E. Geriatric trauma: there is more to it than just the implant! OTA Int 2024; 7:e327. [PMID: 38487398 PMCID: PMC10936161 DOI: 10.1097/oi9.0000000000000327] [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: 11/27/2023] [Revised: 12/30/2023] [Accepted: 01/01/2024] [Indexed: 03/17/2024]
Abstract
Geriatric trauma continues to rise, corresponding with the continuing growth of the older population. These fractures continue to expand, demonstrated by the incidence of hip fractures having grown to 1.5 million adults worldwide per year. This patient population and their associated fracture patterns present unique challenges to the surgeon, as well as having a profound economic impact on the health care system. Pharmacologic treatment has focused on prevention, with aging adults having impaired fracture healing in addition to diminished bone mineral density. Intraoperatively, novel ideas to assess fracture reduction to facilitate decreased fracture collapse have recently been explored. Postoperatively, pharmacologic avenues have focused on future fracture prevention, while shared care models between geriatrics and orthopaedics have shown promise regarding decreasing mortality and length of stay. As geriatric trauma continues to grow, it is imperative that we look to optimize all phases of care, from preoperative to postoperative.
Collapse
Affiliation(s)
- Christopher Lee
- Department of Orthopaedic Surgery, University of California Los Angeles, Los Angeles, CA
| | - Stephen L. Kates
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA
| | - Matthew L. Graves
- Department of Orthopaedic Surgery, University of Mississippi, Jackson, MI
| | - Kyle J. Jeray
- Department of Orthopaedic Surgery, Univeristy of South Carolina School of Medicine, Greenville, SC
| | | | - Reza Firoozabadi
- Department of Orthopaedic Surgery, Univeristy of Washington, Seattle, WA
| | - Emil Schemitsch
- Department of Orthopaedic Surgery, Western University, Bone and Joint Institute, London, ON, Canada
| |
Collapse
|
2
|
Zhang X, Deng C, Qi S. Periosteum Containing Implicit Stem Cells: A Progressive Source of Inspiration for Bone Tissue Regeneration. Int J Mol Sci 2024; 25:2162. [PMID: 38396834 PMCID: PMC10889827 DOI: 10.3390/ijms25042162] [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: 12/15/2023] [Revised: 01/12/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
The periosteum is known as the thin connective tissue covering most bone surfaces. Its extrusive bone regeneration capacity was confirmed from the very first century-old studies. Recently, pluripotent stem cells in the periosteum with unique physiological properties were unveiled. Existing in dynamic contexts and regulated by complex molecular networks, periosteal stem cells emerge as having strong capabilities of proliferation and multipotential differentiation. Through continuous exploration of studies, we are now starting to acquire more insight into the great potential of the periosteum in bone formation and repair in situ or ectopically. It is undeniable that the periosteum is developing further into a more promising strategy to be harnessed in bone tissue regeneration. Here, we summarized the development and structure of the periosteum, cell markers, and the biological features of periosteal stem cells. Then, we reviewed their pivotal role in bone repair and the underlying molecular regulation. The understanding of periosteum-related cellular and molecular content will help enhance future research efforts and application transformation of the periosteum.
Collapse
Affiliation(s)
- Xinyuan Zhang
- Department of Prosthodontics, Shanghai Stomatological Hospital, School of Stomatology, Fudan University, Shanghai 200001, China;
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai 200001, China
| | - Chen Deng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China;
| | - Shengcai Qi
- Department of Prosthodontics, Shanghai Stomatological Hospital, School of Stomatology, Fudan University, Shanghai 200001, China;
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai 200001, China
| |
Collapse
|
3
|
Torres HM, Arnold KM, Oviedo M, Westendorf JJ, Weaver SR. Inflammatory Processes Affecting Bone Health and Repair. Curr Osteoporos Rep 2023; 21:842-853. [PMID: 37759135 PMCID: PMC10842967 DOI: 10.1007/s11914-023-00824-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2023] [Indexed: 09/29/2023]
Abstract
PURPOSE OF REVIEW The purpose of this article is to review the current understanding of inflammatory processes on bone, including direct impacts of inflammatory factors on bone cells, the effect of senescence on inflamed bone, and the critical role of inflammation in bone pain and healing. RECENT FINDINGS Advances in osteoimmunology have provided new perspectives on inflammatory bone loss in recent years. Characterization of so-called inflammatory osteoclasts has revealed insights into physiological and pathological bone loss. The identification of inflammation-associated senescent markers in bone cells indicates that therapies that reduce senescent cell burden may reverse bone loss caused by inflammatory processes. Finally, novel studies have refined the role of inflammation in bone healing, including cross talk between nerves and bone cells. Except for the initial stages of fracture healing, inflammation has predominately negative effects on bone and increases fracture risk. Eliminating senescent cells, priming the osteo-immune axis in bone cells, and alleviating pro-inflammatory cytokine burden may ameliorate the negative effects of inflammation on bone.
Collapse
Affiliation(s)
- Haydee M Torres
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Katherine M Arnold
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
- Biomedical Engineering and Physiology Track/Regenerative Sciences Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, 55905, USA
| | - Manuela Oviedo
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Jennifer J Westendorf
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Samantha R Weaver
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.
| |
Collapse
|
4
|
Kushioka J, Chow SKH, Toya M, Tsubosaka M, Shen H, Gao Q, Li X, Zhang N, Goodman SB. Bone regeneration in inflammation with aging and cell-based immunomodulatory therapy. Inflamm Regen 2023; 43:29. [PMID: 37231450 DOI: 10.1186/s41232-023-00279-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023] Open
Abstract
Aging of the global population increases the incidence of osteoporosis and associated fragility fractures, significantly impacting patient quality of life and healthcare costs. The acute inflammatory reaction is essential to initiate healing after injury. However, aging is associated with "inflammaging", referring to the presence of systemic low-level chronic inflammation. Chronic inflammation impairs the initiation of bone regeneration in elderly patients. This review examines current knowledge of the bone regeneration process and potential immunomodulatory therapies to facilitate bone healing in inflammaging.Aged macrophages show increased sensitivity and responsiveness to inflammatory signals. While M1 macrophages are activated during the acute inflammatory response, proper resolution of the inflammatory phase involves repolarizing pro-inflammatory M1 macrophages to an anti-inflammatory M2 phenotype associated with tissue regeneration. In aging, persistent chronic inflammation resulting from the failure of M1 to M2 repolarization leads to increased osteoclast activation and decreased osteoblast formation, thus increasing bone resorption and decreasing bone formation during healing.Inflammaging can impair the ability of stem cells to support bone regeneration and contributes to the decline in bone mass and strength that occurs with aging. Therefore, modulating inflammaging is a promising approach for improving bone health in the aging population. Mesenchymal stem cells (MSCs) possess immunomodulatory properties that may benefit bone regeneration in inflammation. Preconditioning MSCs with pro-inflammatory cytokines affects MSCs' secretory profile and osteogenic ability. MSCs cultured under hypoxic conditions show increased proliferation rates and secretion of growth factors. Resolution of inflammation via local delivery of anti-inflammatory cytokines is also a potential therapy for bone regeneration in inflammaging. Scaffolds containing anti-inflammatory cytokines, unaltered MSCs, and genetically modified MSCs can also have therapeutic potential. MSC exosomes can increase the migration of MSCs to the fracture site and enhance osteogenic differentiation and angiogenesis.In conclusion, inflammaging can impair the proper initiation of bone regeneration in the elderly. Modulating inflammaging is a promising approach for improving compromised bone healing in the aging population.
Collapse
Affiliation(s)
- Junichi Kushioka
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA.
| | - Simon Kwoon-Ho Chow
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Masakazu Toya
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Masanori Tsubosaka
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Huaishuang Shen
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Xueping Li
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
5
|
Saul D, Khosla S. Fracture Healing in the Setting of Endocrine Diseases, Aging, and Cellular Senescence. Endocr Rev 2022; 43:984-1002. [PMID: 35182420 PMCID: PMC9695115 DOI: 10.1210/endrev/bnac008] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Indexed: 11/19/2022]
Abstract
More than 2.1 million age-related fractures occur in the United States annually, resulting in an immense socioeconomic burden. Importantly, the age-related deterioration of bone structure is associated with impaired bone healing. Fracture healing is a dynamic process which can be divided into four stages. While the initial hematoma generates an inflammatory environment in which mesenchymal stem cells and macrophages orchestrate the framework for repair, angiogenesis and cartilage formation mark the second healing period. In the central region, endochondral ossification favors soft callus development while next to the fractured bony ends, intramembranous ossification directly forms woven bone. The third stage is characterized by removal and calcification of the endochondral cartilage. Finally, the chronic remodeling phase concludes the healing process. Impaired fracture healing due to aging is related to detrimental changes at the cellular level. Macrophages, osteocytes, and chondrocytes express markers of senescence, leading to reduced self-renewal and proliferative capacity. A prolonged phase of "inflammaging" results in an extended remodeling phase, characterized by a senescent microenvironment and deteriorating healing capacity. Although there is evidence that in the setting of injury, at least in some tissues, senescent cells may play a beneficial role in facilitating tissue repair, recent data demonstrate that clearing senescent cells enhances fracture repair. In this review, we summarize the physiological as well as pathological processes during fracture healing in endocrine disease and aging in order to establish a broad understanding of the biomechanical as well as molecular mechanisms involved in bone repair.
Collapse
Affiliation(s)
- Dominik Saul
- Kogod Center on Aging and Division of Endocrinology, Mayo Clinic, Rochester, Minnesota 55905, USA.,Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August-University of Goettingen, 37073 Goettingen, Germany
| | - Sundeep Khosla
- Kogod Center on Aging and Division of Endocrinology, Mayo Clinic, Rochester, Minnesota 55905, USA
| |
Collapse
|
6
|
Kukolj T, Lazarević J, Borojević A, Ralević U, Vujić D, Jauković A, Lazarević N, Bugarski D. A Single-Cell Raman Spectroscopy Analysis of Bone Marrow Mesenchymal Stem/Stromal Cells to Identify Inter-Individual Diversity. Int J Mol Sci 2022; 23:4915. [PMID: 35563306 PMCID: PMC9103070 DOI: 10.3390/ijms23094915] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 12/15/2022] Open
Abstract
The heterogeneity of stem cells represents the main challenge in regenerative medicine development. This issue is particularly pronounced when it comes to the use of primary mesenchymal stem/stromal cells (MSCs) due to a lack of identification markers. Considering the need for additional approaches in MSCs characterization, we applied Raman spectroscopy to investigate inter-individual differences between bone marrow MSCs (BM-MSCs). Based on standard biological tests, BM-MSCs of analyzed donors fulfill all conditions for their characterization, while no donor-related specifics were observed in terms of BM-MSCs morphology, phenotype, multilineage differentiation potential, colony-forming capacity, expression of pluripotency-associated markers or proliferative capacity. However, examination of BM-MSCs at a single-cell level by Raman spectroscopy revealed that despite similar biochemical background, fine differences in the Raman spectra of BM-MSCs of each donor can be detected. After extensive principal component analysis (PCA) of Raman spectra, our study revealed the possibility of this method to diversify BM-MSCs populations, whereby the grouping of cell populations was most prominent when cell populations were analyzed in pairs. These results indicate that Raman spectroscopy, as a label-free assay, could have a huge potential in understanding stem cell heterogeneity and sorting cell populations with a similar biochemical background that can be significant for the development of personalized therapy approaches.
Collapse
Affiliation(s)
- Tamara Kukolj
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia; (A.J.); (D.B.)
| | - Jasmina Lazarević
- Center for Solid State Physics and New Materials, Institute of Physics Belgrade, University of Belgrade, Pregrevica 118, 11080 Belgrade, Serbia; (J.L.); (U.R.); (N.L.)
| | - Ana Borojević
- Mother and Child Health Care Institute of Serbia ‘’Dr Vukan Čupić’’, 11000 Belgrade, Serbia; (A.B.); (D.V.)
| | - Uroš Ralević
- Center for Solid State Physics and New Materials, Institute of Physics Belgrade, University of Belgrade, Pregrevica 118, 11080 Belgrade, Serbia; (J.L.); (U.R.); (N.L.)
| | - Dragana Vujić
- Mother and Child Health Care Institute of Serbia ‘’Dr Vukan Čupić’’, 11000 Belgrade, Serbia; (A.B.); (D.V.)
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Aleksandra Jauković
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia; (A.J.); (D.B.)
| | - Nenad Lazarević
- Center for Solid State Physics and New Materials, Institute of Physics Belgrade, University of Belgrade, Pregrevica 118, 11080 Belgrade, Serbia; (J.L.); (U.R.); (N.L.)
| | - Diana Bugarski
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia; (A.J.); (D.B.)
| |
Collapse
|
7
|
Kapetanos K, Asimakopoulos D, Christodoulou N, Vogt A, Khan W. Chronological Age Affects MSC Senescence In Vitro-A Systematic Review. Int J Mol Sci 2021; 22:ijms22157945. [PMID: 34360725 PMCID: PMC8348192 DOI: 10.3390/ijms22157945] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 01/10/2023] Open
Abstract
The use of mesenchymal stromal cells (MSCs) in regenerative medicine and tissue engineering is well established, given their properties of self-renewal and differentiation. However, several studies have shown that these properties diminish with age, and understanding the pathways involved are important to provide regenerative therapies in an ageing population. In this PRISMA systematic review, we investigated the effects of chronological donor ageing on the senescence of MSCs. We identified 3023 studies after searching four databases including PubMed, Web of Science, Cochrane, and Medline. Nine studies met the inclusion and exclusion criteria and were included in the final analyses. These studies showed an increase in the expression of p21, p53, p16, ROS, and NF-κB with chronological age. This implies an activated DNA damage response (DDR), as well as increased levels of stress and inflammation in the MSCs of older donors. Additionally, highlighting the effects of an activated DDR in cells from older donors, a decrease in the expression of proliferative markers including Ki67, MAPK pathway elements, and Wnt/β-catenin pathway elements was observed. Furthermore, we found an increase in the levels of SA-β-galactosidase, a specific marker of cellular senescence. Together, these findings support an association between chronological age and MSC senescence. The precise threshold for chronological age where the reported changes become significant is yet to be defined and should form the basis for further scientific investigations. The outcomes of this review should direct further investigations into reversing the biological effects of chronological age on the MSC senescence phenotype.
Collapse
Affiliation(s)
- Konstantinos Kapetanos
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 2SP, UK; (D.A.); (N.C.)
- Correspondence: ; Tel.: +44-357-9664-9787
| | | | | | - Antonia Vogt
- Division of Trauma & Orthopaedic Surgery, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0QQ, UK; (A.V.); (W.K.)
| | - Wasim Khan
- Division of Trauma & Orthopaedic Surgery, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0QQ, UK; (A.V.); (W.K.)
| |
Collapse
|
8
|
Influence of Age on Calvarial Critical Size Defect Dimensions: A Radiographic and Histological Study. J Craniofac Surg 2021; 32:2896-2900. [PMID: 33867513 PMCID: PMC8549460 DOI: 10.1097/scs.0000000000007690] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Calvarial critical-size defect has been used to assess techniques and materials in the bone regeneration field. Previous studies utilized young adult rats with 3 months of age, which might not reflect the geriatric conditions. This study aimed to assess the dimensions of the calvarial critical-size defect in aged rats. Seventy-two rats in a randomized block design were allocated into a control young adult (11–12 weeks), and a test old group (22–24 months). Both groups were divided according to bone defect's size: 3 mm, 5 mm, and 7 mm defects, which were surgically created and followed for 4 and 8 weeks. Radiographic and histologic analyses were performed. Based on the results, additional groups with 4 mm defect size were added following the same protocols. Young groups yielded higher bone volumes, defect closure percentages, and density of newly formed bone. Closure of cranial defects was only observed in 3 mm defects in both age groups after 8 weeks; however, the 4 mm defect group demonstrated bony bridging after 8 weeks in young but not old rats. Results confirmed that 5-mm defect is considered a critical size for calvarial bone defects in young adult rats; however, 4 mm defect might be considered critical size for the aged rats after 8 weeks.
Collapse
|
9
|
MicroRNA Profiling in Mesenchymal Stromal Cells: the Tissue Source as the Missing Piece in the Puzzle of Ageing. Stem Cell Rev Rep 2021; 17:1014-1026. [PMID: 33405068 DOI: 10.1007/s12015-020-10095-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2020] [Indexed: 10/22/2022]
Abstract
Ageing is among the main risk factors for human disease onset and the identification of the hallmarks of senescence remains a challenge for the development of appropriate therapeutic target in the elderly. Here, we compare senescence-related changes in two cell populations of mesenchymal stromal cells by analysing their miRNA profiling: Human Dental Pulp Stromal Cells (hDPSCs) and human Periosteum-Derived Progenitor Cells (hPDPCs). After these cells were harvested, total RNA extraction and whole genome miRNA profiling was performed, and DIANA-miRPath analysis was applied to find the target/pathways. Only 69 microRNAs showed a significant differential expression between dental pulp and periosteum progenitor cells. Among these, 24 were up regulated, and 45 were downregulated in hDPSCs compared to hPDPCs. Our attention was centered on miRNAs (22 upregulated and 34 downregulated) involved in common pathways for cell senescence (i.e. p53, mTOR pathways), autophagy (i.e. mTOR and MAPK pathways) and cell cycle (i.e. MAPK pathway). The p53, mTOR and MAPK signaling pathways comprised 43, 37 and 112 genes targeted by all selected miRNAs, respectively. Our finding is consistent with the idea that the embryological origin influences cell behavior and the ageing process. Our study strengthens the hypothesis that ageing is driven by numerous mediators interacting through an intricate molecular network, which affects adult stem cells self-renewal capability. Graphical abstract.
Collapse
|
10
|
Jin Y, Sun X, Pei F, Zhao Z, Mao J. Wnt16 signaling promotes osteoblast differentiation of periosteal derived cells in vitro and in vivo. PeerJ 2020; 8:e10374. [PMID: 33282557 PMCID: PMC7694570 DOI: 10.7717/peerj.10374] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/26/2020] [Indexed: 02/05/2023] Open
Abstract
Background Periosteum plays critical roles in de novo bone formation and fracture repair. Wnt16 has been regarded as a key regulator in periosteum bone formation. However, the role of Wnt16 in periosteum derived cells (PDCs) osteogenic differentiation remains unclear. The study goal is to uncover whether and how Wnt16 acts on the osteogenesis of PDCs. Methods We detected the variation of Wnt16 mRNA expression in PDCs, which were isolated from mouse femur and identified by flow cytometry, cultured in osteogenic medium for 14 days, then knocked down and over-expressed Wnt16 in PDCs to analysis its effects in osteogenesis. Further, we seeded PDCs (Wnt16 over-expressed/vector) in β-tricalcium phosphate cubes, and transplanted this complex into a critical size calvarial defect. Lastly, we used immunofluorescence, Topflash and NFAT luciferase reporter assay to study the possible downstream signaling pathway of Wnt16. Results Wnt16 mRNA expression showed an increasing trend in PDCs under osteogenic induction for 14 days. Wnt16 shRNA reduced mRNA expression of Runx2, collage type I (Col-1) and osteocalcin (OCN) after 7 days of osteogenic induction, as well as alizarin red staining intensity after 21days. Wnt16 also increased the mRNA expression of Runx2 and OCN and the protein production of Runx2 and Col-1 after 2 days of osteogenic stimulation. In the orthotopic transplantation assay, more bone volume, trabecula number and less trabecula space were found in Wnt16 over-expressed group. Besides, in the newly formed tissue Brdu positive area was smaller and Col-1 was larger in Wnt16 over-expressed group compared to the control group. Finally, Wnt16 upregulated CTNNB1/β-catenin expression and its nuclear translocation in PDCs, also increased Topflash reporter luciferase activity. By contrast, Wnt16 failed to increase NFAT reporter luciferase activity. Conclusion Together, Wnt16 plays a positive role in regulating PDCs osteogenesis, and Wnt16 may have a potential use in improving bone regeneration.
Collapse
Affiliation(s)
- Ying Jin
- Department of Orthodontics, State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Xiaoyan Sun
- Stomatological Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fang Pei
- Department of Orthodontics, State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Zhihe Zhao
- Department of Orthodontics, State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Jeremy Mao
- Columbia University, Center for Craniofacial Regeneration, New York, NY, United States of America
| |
Collapse
|
11
|
Mao H, Yang A, Pan Y, Li H, Lei L. Displacement in root apex and changes in incisor inclination affect alveolar bone remodeling in adult bimaxillary protrusion patients: a retrospective study. Head Face Med 2020; 16:29. [PMID: 33213460 PMCID: PMC7678166 DOI: 10.1186/s13005-020-00242-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 11/09/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Periodontal health is of great concern for periodontists and orthodontists in the inter-disciplinary management of patients with bimaxillary protrusion. The aim of present study is to investigate changes in the alveolar bone in the maxillary incisor region and to explore its relationship with displacement of root apex as well as changes in the inclination of maxillary incisors during incisor retraction. METHODS Samples in this retrospective study consisted of 38 patients with bimaxillary protrusion. Cone-beam computed tomography (CBCT) images was taken before(T0) and after (T1) treatment. Alveolar bone thickness (ABT), height (ABH) and area (ABA) were utilized to evaluate changes in the alveolar bone, while incisor inclination and apex displacement were used to assess changes in the position of maxillary central and lateral incisors. Correlations between alveolar bone remodeling and apex displacement as well as changes in the inclination were investigated. RESULTS The labial ABT of central and lateral incisors at the mid-root third was increased. In contrast, the palatal ABT at crestal, mid-root and apical third level were consistently decreased. ABH was not altered on the labial side, while significantly decreased on the palatal side. ABA was not significantly increased on the labial side, but significantly decreased on the palatal side, leading to a significantly reduced total ABA. Orthodontic treatment significantly reduced inclination of upper incisors. Changes in the amount (T1-T0) of ABA was remarkably correlated with apex displacement and changes of inclination (T1-T0); in addition, using the multivariate linear regression analysis, changes of ABA on the palatal side (T1-T0) can be described by following equation: Changes of palatal ABA (T1-T0) = - 3.258- 0.139× changes of inclination (T1-T0) + 2.533 × apex displacement (T1-T0). CONCLUSIONS Retraction of incisors in bimaxillary protrusion patients may compromise periodontal bone support on the palatal side. An equation that incorporated the displacement of root apex and change in the incisor inclination may enable periodontist-orthodontist interdisciplinary coordination in assessing treatment risks and developing an individualized treatment plan for adult patients with bimaxillary protrusion. Moreover, the equation in predicating area of alveolar bone may reduce the risks of placing the teeth out of the bone boundary during 3D digital setups.
Collapse
Affiliation(s)
- Huimin Mao
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Andi Yang
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yue Pan
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Houxuan Li
- Department of Periodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.
| | - Lang Lei
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|
12
|
Park JH, Kang YH, Hwang SC, Oh SH, Byun JH. Parthenolide Has Negative Effects on In Vitro Enhanced Osteogenic Phenotypes by Inflammatory Cytokine TNF-α via Inhibiting JNK Signaling. Int J Mol Sci 2020; 21:ijms21155433. [PMID: 32751648 DOI: 10.3390/ijms21155433] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/14/2022] Open
Abstract
Nuclear factor kappa B (NF-κB) regulates inflammatory gene expression and represents a likely target for novel disease treatment approaches, including skeletal disorders. Several plant-derived sesquiterpene lactones can inhibit the activation of NF-κB. Parthenolide (PTL) is an abundant sesquiterpene lactone, found in Mexican Indian Asteraceae family plants, with reported anti-inflammatory activity, through the inhibition of a common step in the NF-κB activation pathway. This study examined the effects of PTL on the enhanced, in vitro, osteogenic phenotypes of human periosteum-derived cells (hPDCs), mediated by the inflammatory cytokine tumor necrosis factor (TNF)-α. PTL had no significant effects on hPDC viability or osteoblastic activities, whereas TNF-α had positive effects on the in vitro osteoblastic differentiation of hPDCs. c-Jun N-terminal kinase (JNK) signaling played an important role in the enhanced osteoblastic differentiation of TNF-α-treated hPDCs. Treatment with 1 µM PTL did not affect TNF-α-treated hPDCs; however, 5 and 10 µM PTL treatment decreased the histochemical detection and activity of alkaline phosphatase (ALP), alizarin red-positive mineralization, and the expression of ALP and osteocalcin mRNA. JNK phosphorylation decreased significantly in TNF-α-treated hPDCs pretreated with PTL. These results suggested that PTL exerts negative effects on the increased osteoblastic differentiation of TNF-α-treated hPDCs by inhibiting JNK signaling.
Collapse
Affiliation(s)
- Jin-Ho Park
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University, Jinju 52727, Korea
| | - Young-Hoon Kang
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University, Jinju 52727, Korea
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Changwon Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University, Jinju 52727, Korea
| | - Sun-Chul Hwang
- Department of Orthopaedic Surgery, Institute of Health Sciences, School of Medicine, Gyeongsang National University, Jinju 52727, Korea
| | - Se Heang Oh
- Department of Pharmaceutical Engineering, Dankook University, Cheonan 31116, Korea
| | - June-Ho Byun
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University, Jinju 52727, Korea
| |
Collapse
|
13
|
Mantripragada VP, Piuzzi NS, Bova WA, Boehm C, Obuchowski NA, Lefebvre V, Midura RJ, Muschler GF. Donor-matched comparison of chondrogenic progenitors resident in human infrapatellar fat pad, synovium, and periosteum - implications for cartilage repair. Connect Tissue Res 2019; 60:597-610. [PMID: 31020864 DOI: 10.1080/03008207.2019.1611795] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Purpose: There is a clinical need to better characterize tissue sources being used for stem cell therapies. This study focuses on comparison of cells and connective tissue progenitors (CTPs) derived from native human infrapatellar fatpad (IPFP), synovium (SYN), and periosteum (PERI). Materials and Methods: IPFP, SYN, PERI were harvested from twenty-eight patients undergoing arthroplasty. CTPs were quantitatively characterized using automated colony-forming-unit assay to compare total nucleated cell concentration-[Cell], cells/mg; prevalence-(PCTP), CTPs/million nucleated cells; CTP concentration-[CTP], CTPs/mg; proliferation and differentiation potential; and correlate outcomes with patient's age and gender. Results: [Cell] did not differ between IPFP, SYN, and PERI. PCTP was influenced by age and gender: patients >60 years, IPFP and SYN had higher PCTP than PERI (p < 0.001) and females had higher PCTP in IPFP (p < 0.001) and SYN (p = 0.001) than PERI. [CTP] was influenced by age: patients <50 years, SYN (p = 0.0165) and PERI (p < 0.001) had higher [CTP] than IPFP; patients between 60 and 69 years, SYN (p < 0.001) had higher [CTP] than PERI; patients >70 years, IPFP (p = 0.006) had higher [CTP] than PERI. In patients >60 years, proliferation potential of CTPs differed significantly (SYN>IPFP>PERI); however, differentiation potentials were comparable between all three tissue sources. Conclusion: SYN and IPFP may serve as a preferred tissue source for patients >60 years, and PERI along with SYN and IPFP may serve as a preferred tissue source for patients <60 years for cartilage repair. However, the heterogeneity among the CTPs in any given tissue source suggests performance-based selection might be useful to optimize cell-sourcing strategies to improve efficacy of cellular therapies for cartilage repair.
Collapse
Affiliation(s)
- V P Mantripragada
- Department of Biomedical Engineering, Lerner Research Institute , Cleveland , OH , USA
| | - N S Piuzzi
- Department of Biomedical Engineering, Lerner Research Institute , Cleveland , OH , USA.,Department of Orthopedic Surgery, Cleveland Clinic , Cleveland , OH , USA.,Department of Orthopaedic Surgery, Instituto Universitario del Hospital Italiano de Buenos Aires , Buenos Aires , Argentina
| | - W A Bova
- Department of Biomedical Engineering, Lerner Research Institute , Cleveland , OH , USA
| | - C Boehm
- Department of Biomedical Engineering, Lerner Research Institute , Cleveland , OH , USA
| | - N A Obuchowski
- Department of Quantitative Health Science, Cleveland Clinic , Cleveland , OH , USA
| | - V Lefebvre
- Department of Cellular and Molecular Medicine, Cleveland Clinic , Cleveland , OH , USA
| | - R J Midura
- Department of Biomedical Engineering, Lerner Research Institute , Cleveland , OH , USA
| | - G F Muschler
- Department of Biomedical Engineering, Lerner Research Institute , Cleveland , OH , USA.,Department of Orthopedic Surgery, Cleveland Clinic , Cleveland , OH , USA
| |
Collapse
|
14
|
Synovium-derived stromal cell-induced osteoclastogenesis: a potential osteoarthritis trigger. Clin Sci (Lond) 2019; 133:1813-1824. [DOI: 10.1042/cs20190169] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 08/06/2019] [Accepted: 08/14/2019] [Indexed: 12/20/2022]
Abstract
Abstract
Purpose: To shed light on the idea that mesenchymal stem/stromal cells (MSCs) recruited in synovium (SM) (i.e. Synovium-Derived Stromal Cells, SDSCs) could be involved in Osteoarthritis (OA) pathophysiology. Attention was also paid to a further stromal cell type with a peculiar ultrastructure called telocytes (TCs), whose role is far from clarified. Methods: In the present in vitro study, we compared SDSCs isolated from healthy and OA subjects in terms of phenotype, morphology and differentiation potential as well as in their capability to activate normal Peripheral Blood Mononuclear Cells (PBMCs). Histological, immunohistochemical and ultrastructural analyses were integrated by qRT-PCR and functional resorbing assays. Results: Our data demonstrated that both SDSC populations stimulated the formation of osteoclasts from PBMCs: the osteoclast-like cells generated by healthy-SDSCs via transwell co-cultures were inactive, while OA-derived SDSCs have a much greater effectiveness. Moreover, the presence of TCs was more evident in cultures obtained from OA subjects and suggests a possible involvement of these cells in OA. Conclusions: Osteoclastogenic differentiation capability of PBMCs from OA subjects, also induced by B synoviocytes has been already documented. Here we hypothesized that SDSCs, generally considered for their regenerative potential in cartilage lesions, have also a role in the onset/maintenance of OA. Clinical relevance: Our observations may represent an interesting opportunity for the development of a holistic approach for OA treatment, that considers the multifaceted capability of MSCs in relation to the environment.
Collapse
|
15
|
Meinberg EG, Clark D, Miclau KR, Marcucio R, Miclau T. Fracture repair in the elderly: Clinical and experimental considerations. Injury 2019; 50 Suppl 1:S62-S65. [PMID: 31130210 PMCID: PMC7021229 DOI: 10.1016/j.injury.2019.05.005] [Citation(s) in RCA: 17] [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: 02/28/2019] [Revised: 05/01/2019] [Accepted: 05/08/2019] [Indexed: 02/07/2023]
Abstract
Fractures in the elderly represent a significant and rising socioeconomic problem. Although aging has been associated with delays in healing, there is little direct clinical data isolating the effects of aging on bone healing from the associated comorbidities that are frequently present in elderly populations. Basic research has demonstrated that all of the components of fracture repair-cells, extracellular matrix, blood supply, and molecules and their receptors-are negatively impacted by the aging process, which likely explains poorer clinical outcomes. Improved understanding of age-related fracture healing should aid in the development of novel treatment strategies, technologies, and therapies to improve bone repair in elderly patients.
Collapse
Affiliation(s)
- E G Meinberg
- UCSF/ZSFG Orthopaedic Trauma Institute, UCSF Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA
| | - D Clark
- UCSF/ZSFG Orthopaedic Trauma Institute, UCSF Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA
| | - K R Miclau
- UCSF/ZSFG Orthopaedic Trauma Institute, UCSF Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA
| | - R Marcucio
- UCSF/ZSFG Orthopaedic Trauma Institute, UCSF Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA
| | - T Miclau
- UCSF/ZSFG Orthopaedic Trauma Institute, UCSF Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA.
| |
Collapse
|
16
|
Gao B, Deng R, Chai Y, Chen H, Hu B, Wang X, Zhu S, Cao Y, Ni S, Wan M, Yang L, Luo Z, Cao X. Macrophage-lineage TRAP+ cells recruit periosteum-derived cells for periosteal osteogenesis and regeneration. J Clin Invest 2019; 129:2578-2594. [PMID: 30946695 DOI: 10.1172/jci98857] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The periosteum, a thin tissue that covers almost the entire bone surface, accounts for more than 80% of human bone mass and is essential for bone regeneration. Its osteogenic and bone regenerative abilities are well studied, but much is unknown about the periosteum. In this study, we found that macrophage-lineage cells recruit periosteum-derived cells (PDCs) for cortical bone formation. Knockout of colony stimulating factor-1 eliminated macrophage-lineage cells and resulted in loss of PDCs with impaired periosteal bone formation. Moreover, macrophage-lineage TRAP+ cells induced transcriptional expression of periostin and recruitment of PDCs to the periosteal surface through secretion of platelet-derived growth factor-BB (PDGF-BB), where the recruited PDCs underwent osteoblast differentiation coupled with type H vessel formation. We also found that subsets of Nestin+ and LepR+ PDCs possess multipotent and self-renewal abilities and contribute to cortical bone formation. Nestin+ PDCs are found primarily during bone development, whereas LepR+ PDCs are essential for bone homeostasis in adult mice. Importantly, conditional knockout of Pdgfrβ (platelet-derived growth factor receptor beta) in LepR+ cells impaired periosteal bone formation and regeneration. These findings uncover the essential role of periosteal macrophage-lineage cells in regulating periosteum homeostasis and regeneration.
Collapse
Affiliation(s)
- Bo Gao
- Department of Orthopaedic Surgery, Institute of Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Institute of Orthopaedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Ruoxian Deng
- Department of Orthopaedic Surgery, Institute of Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yu Chai
- Department of Orthopaedic Surgery, Institute of Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hao Chen
- Department of Orthopaedic Surgery, Institute of Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Bo Hu
- Department of Orthopaedic Surgery, Institute of Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xiao Wang
- Department of Orthopaedic Surgery, Institute of Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shouan Zhu
- Department of Orthopaedic Surgery, Institute of Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yong Cao
- Department of Orthopaedic Surgery, Institute of Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shuangfei Ni
- Department of Orthopaedic Surgery, Institute of Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mei Wan
- Department of Orthopaedic Surgery, Institute of Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Liu Yang
- Institute of Orthopaedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zhuojing Luo
- Institute of Orthopaedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xu Cao
- Department of Orthopaedic Surgery, Institute of Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
17
|
Duchamp de Lageneste O, Colnot C. Periostin in Bone Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1132:49-61. [PMID: 31037624 DOI: 10.1007/978-981-13-6657-4_6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Bone regeneration is an efficient regenerative process depending on the recruitment and activation of skeletal stem cells that allow cartilage and bone formation leading to fracture consolidation. Periosteum, the tissue located at the outer surface of bone is now recognized as an essential player in the bone repair process and contains skeletal stem cells with high regenerative potential. The matrix composition of the periosteum defines its roles in bone growth, in cortical bone modeling and remodeling in response to mechanical strain, and in bone repair. Periostin is a key extracellular matrix component of the periosteum involved in periosteum functions. In this chapter, we summarize the current knowledge on the bone regeneration process, the role of the periosteum and skeletal stem cells, and Periostin functions in this context. The matricellular protein Periostin has several roles through all stages of bone repair: in the early days of repair during the initial activation of stem cells within periosteum, in the active phase of cartilage and bone deposition in the facture callus, and in the final phase of bone bridging and reconstitution of the stem cell pool within periosteum.
Collapse
Affiliation(s)
| | - Céline Colnot
- INSERM UMR1163, Imagine Institute, Paris Descartes University, Paris, France.
| |
Collapse
|
18
|
Detecting senescent fate in mesenchymal stem cells: a combined cytofluorimetric and ultrastructural approach. Biogerontology 2018; 19:401-414. [PMID: 30101381 DOI: 10.1007/s10522-018-9766-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 08/06/2018] [Indexed: 02/08/2023]
Abstract
Senescence can impair the therapeutic potential of stem cells. In this study, senescence-associated morphofunctional changes in periosteum-derived progenitor cells (PDPCs) from old and young individuals were investigated by combining cytofluorimetry, immunohistochemistry, and transmission electron microscopy. Cell cycle analysis demonstrated a large number of G0/G1 phase cells in PDPCs from old subjects and a progressive accumulation of G0/G1 cells during passaging in cultures from young subjects. Cytofluorimetry documented significant changes in light scattering parameters and closely correlated with the ultrastructural features, especially changes in mitochondrial shape and autophagy, which are consistent with the mitochondrial-lysosomal axis theory of ageing. The combined morphological, biofunctional, and ultrastructural approach enhanced the flow cytometric study of PDPC ageing. We speculate that impaired autophagy, documented in replicative senescent and old PDPCs, reflect a switch from quiescence to senescence. Its demonstration in a tissue with limited turnover-like the cambium layer of the periosteum, where reversible quiescence is the normal stem cell state throughout life-adds a new piece to the regenerative medicine jigsaw in an ageing society.
Collapse
|
19
|
Treatment of intrabony defects with modified perforated membranes in aggressive periodontitis: a 12-month randomized controlled trial. Clin Oral Investig 2018; 22:2819-2828. [PMID: 29411112 DOI: 10.1007/s00784-018-2368-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 01/25/2018] [Indexed: 12/27/2022]
Abstract
OBJECTIVE The aim of this study was to compare the clinical and radiographic efficacy of guided tissue regeneration with a modified perforated collagen membrane (MPM) or standard collagen membrane (CM) in the treatment of intrabony defects in patients with aggressive periodontitis (AgP). MATERIALS AND METHODS Fifteen AgP patients were included in the study. Two single intrabony defects of at least 3 mm depth with ≥ 6 mm probing pocket depth (PPD) from each patient were randomly assigned to either xenogenic graft plus MPM (test group) or xenogenic graft plus CM (control group). PPD, clinical attachment level (CAL), and gingival recession (GR) were recorded at baseline and at 12 months. The radiographic assessments included the measurements of defect depth (DD), change in alveolar crest position (ACP), linear defect fill (LDF), and percentage defect fill (%DF). RESULTS After treatment, PPD, CAL, DD, and ACP values improved significantly in both groups, without statistical differences between them. However, with respect to LDF and %DF, the 12-month radiographic analysis at MPM-treated sites showed a significant improvement compared to the 6-month outcomes, that was not observed at control sites (additional LDF of 0.4 ± 0.5 mm, p = 0.010 and %DF of 6.4 ± 7.6%, p = 0.025). CONCLUSIONS Both strategies proved effective in the treatment of intrabony defects in patients with AgP. Nonetheless, enhanced LDF and %DF 12 months postoperatively at MPM-treated sites may stem from cellular and molecular migration from the periosteum and overlying gingival connective tissue through barrier's pores. CLINICAL RELEVANCE Modification of CM may have positive ramifications on periodontal regeneration.
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW This review summarizes research on the physiological changes that occur with aging and the resulting effects on fracture healing. RECENT FINDINGS Aging affects the inflammatory response during fracture healing through senescence of the immune response and increased systemic pro-inflammatory status. Important cells of the inflammatory response, macrophages, T cells, mesenchymal stem cells, have demonstrated intrinsic age-related changes that could impact fracture healing. Additionally, vascularization and angiogenesis are impaired in fracture healing of the elderly. Finally, osteochondral cells and their progenitors demonstrate decreased activity and quantity within the callus. Age-related changes affect many of the biologic processes involved in fracture healing. However, the contributions of such changes do not fully explain the poorer healing outcomes and increased morbidity reported in elderly patients. Future research should address this gap in understanding in order to provide improved and more directed treatment options for the elderly population.
Collapse
Affiliation(s)
- Dan Clark
- Department of Orthopaedic Surgery, University of California at San Francisco, 513 Parnassus Ave., San Francisco, CA, 94143, USA
- Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital and Trauma Center, 2550 23rd St, Building 9, San Francisco, CA, 94110, USA
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of California at San Francisco, 513 Parnassus Ave., Rm. S-619A, San Francisco, CA, 94143, USA
| | - Mary Nakamura
- Department of Medicine, University of California at San Francisco, San Francisco, CA, 94143-0451, USA
- Department of Pathology, VA Medical Center, University of California San Francisco & Pathology Service, San Francisco, CA, 94121, USA
| | - Ted Miclau
- Department of Orthopaedic Surgery, University of California at San Francisco, 513 Parnassus Ave., San Francisco, CA, 94143, USA
- Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital and Trauma Center, 2550 23rd St, Building 9, San Francisco, CA, 94110, USA
| | - Ralph Marcucio
- Department of Orthopaedic Surgery, University of California at San Francisco, 513 Parnassus Ave., San Francisco, CA, 94143, USA.
- Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital and Trauma Center, 2550 23rd St, Building 9, San Francisco, CA, 94110, USA.
| |
Collapse
|
21
|
Abstract
Bone healing involves complex biological pathways and interactions among various cell types and microenvironments. Among them, the monocyte–macrophage–osteoclast lineage and the mesenchymal stem cell–osteoblast lineage are critical, in addition to an initial inflammatory microenvironment. These cellular interactions induce the necessary inflammatory milieu and provide the cells for bone regeneration and immune modulation. Increasing age is accompanied with a rise in the basal state of inflammation, potentially impairing osteogenesis. The translational potential of this article: Translational research has shown multiple interactions between inflammation, ageing, and bone regeneration. This review presents recent, relevant considerations regarding the effects of inflammation and ageing on bone healing.
Collapse
Affiliation(s)
- Emmanuel Gibon
- Corresponding author. Department of Orthopaedic Surgery, Stanford University, 300 Pasteur Drive, Edwards Building R116, Stanford, CA 94305, USA.Department of Orthopaedic SurgeryStanford University300 Pasteur DriveEdwards Building R116StanfordCA94305USA
| | | | | | | |
Collapse
|
22
|
Orciani M, Fini M, Di Primio R, Mattioli-Belmonte M. Biofabrication and Bone Tissue Regeneration: Cell Source, Approaches, and Challenges. Front Bioeng Biotechnol 2017; 5:17. [PMID: 28386538 PMCID: PMC5362636 DOI: 10.3389/fbioe.2017.00017] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 02/22/2017] [Indexed: 01/06/2023] Open
Abstract
The growing occurrence of bone disorders and the increase in aging population have resulted in the need for more effective therapies to meet this request. Bone tissue engineering strategies, by combining biomaterials, cells, and signaling factors, are seen as alternatives to conventional bone grafts for repairing or rebuilding bone defects. Indeed, skeletal tissue engineering has not yet achieved full translation into clinical practice because of several challenges. Bone biofabrication by additive manufacturing techniques may represent a possible solution, with its intrinsic capability for accuracy, reproducibility, and customization of scaffolds as well as cell and signaling molecule delivery. This review examines the existing research in bone biofabrication and the appropriate cells and factors selection for successful bone regeneration as well as limitations affecting these approaches. Challenges that need to be tackled with the highest priority are the obtainment of appropriate vascularized scaffolds with an accurate spatiotemporal biochemical and mechanical stimuli release, in order to improve osseointegration as well as osteogenesis.
Collapse
Affiliation(s)
- Monia Orciani
- Department of Molecular and Clinical Sciences, Università Politenica delle Marche , Ancona , Italy
| | - Milena Fini
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopedic Institute , Bologna , Italy
| | - Roberto Di Primio
- Department of Molecular and Clinical Sciences, Università Politenica delle Marche , Ancona , Italy
| | - Monica Mattioli-Belmonte
- Department of Molecular and Clinical Sciences, Università Politenica delle Marche , Ancona , Italy
| |
Collapse
|
23
|
Najar M, Lagneaux L. Foreskin as a source of immunotherapeutic mesenchymal stromal cells. Immunotherapy 2017; 9:207-217. [PMID: 28128711 DOI: 10.2217/imt-2016-0093] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 01/10/2017] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have well-characterized properties and thus represent an attractive cell population for use in several therapeutic applications. Due to the limitations and inconveniences associated with classical sources of MSCs, the identification and characterization of alternative sources are required for safe and efficient cell therapy. The skin tissue is currently referred to as a reservoir of cells with therapeutically relevant functions. Historically considered biological waste, foreskin (FSK) is increasingly used to provide immunotherapeutic MSCs for medicinal products. This review discusses for the first time the nature and profile of MSCs within the foreskin tissue and, in particular, their immunobiology. A better immunological characterization and understanding of foreskin-derived cells will be critical for improving MSC-based cellular strategies for immunotherapeutic applications.
Collapse
Affiliation(s)
- Mehdi Najar
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Bâtiment de Transfusion (Level +1), Route de Lennik n° 808, 1070 Brussels, Belgium
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Bâtiment de Transfusion (Level +1), Route de Lennik n° 808, 1070 Brussels, Belgium
| |
Collapse
|
24
|
Vozzi G, Lucarini G, Dicarlo M, Andreoni C, Salvolini E, Ferretti C, Mattioli-Belmonte M. In vitro lifespan and senescent behaviour of human periosteal derived stem cells. Bone 2016; 88:1-12. [PMID: 27102545 DOI: 10.1016/j.bone.2016.04.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 03/08/2016] [Accepted: 04/06/2016] [Indexed: 01/26/2023]
Abstract
Periosteum derived progenitor cells (PDPCs) represent promising mesenchymal stem cells (MSCs) for skeletal regeneration and to test bone cell based tissue engineering strategies. Most of regenerative medicine approaches based on MSCs require a noteworthy amount of cells that must be expanded in vitro prior to their use. As culture expansion method may impact on cell behaviour, we assessed the replicative and metabolic capacity (nitric oxide production and glucose consumption), senescence hallmarks of PDPCs serially passaged as well as the expression of selected genes specifically related to early osteoblastic differentiation, bone remodelling and stemness during PDPC sequential passaging. We also scouted a Systems Biology approach to examine and elucidate the experimental results through mathematical modelling and in silico simulations. PDPC subculture led to a progressive proliferative decline but, despite this, PDPCs maintained almost constant their metabolic activity. In vitro, senescent PDPCs displayed the typical "replicative senescence" features, involving p16 and not p53 in the regulation of this phenomenon. Gene expression analysis evidenced the tendency of sub-cultured PDPCs to increase the expression of genes involved in bone resorption. The mathematical analysis of the experimental results showed a strict similarity between replicative senescence and age-related changes, enabling the definition of an in silico model mimicking PDPC behaviour in terms of nitric oxide (NO) production. The relationship between NO production and subculture passages could represent a cutting edge "replicative senescence index". Overall, our findings suggest the possibility to use early-passage PDPCs for bone regenerative approaches based on the local recruitment of stem cells, whilst the later cell passages could be a suitable in vitro tool to validate scaffolds intended for bone regeneration in elderly subjects.
Collapse
Affiliation(s)
- Giovanni Vozzi
- Research Centre "E. Piaggio", Faculty of Engineering, University of Pisa, Largo Lucio Lazzarino, 2, 56126 Pisa, Italy
| | - Guendalina Lucarini
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/a, 60126 Ancona, Italy
| | - Manuela Dicarlo
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/a, 60126 Ancona, Italy
| | - Chiara Andreoni
- Research Centre "E. Piaggio", Faculty of Engineering, University of Pisa, Largo Lucio Lazzarino, 2, 56126 Pisa, Italy
| | - Eleonora Salvolini
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/a, 60126 Ancona, Italy
| | - Concetta Ferretti
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/a, 60126 Ancona, Italy
| | - Monica Mattioli-Belmonte
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/a, 60126 Ancona, Italy.
| |
Collapse
|
25
|
Decoupling the role of stiffness from other hydroxyapatite signalling cues in periosteal derived stem cell differentiation. Sci Rep 2015; 5:10778. [PMID: 26035412 PMCID: PMC4451686 DOI: 10.1038/srep10778] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 05/05/2015] [Indexed: 12/15/2022] Open
Abstract
Bone extracellular matrix (ECM) is a natural composite made of collagen and mineral hydroxyapatite (HA). Dynamic cell-ECM interactions play a critical role in regulating cell differentiation and function. Understanding the principal ECM cues promoting osteogenic differentiation would be pivotal for both bone tissue engineering and regenerative medicine. Altering the mineral content generally modifies the stiffness as well as other physicochemical cues provided by composite materials, complicating the “cause-effect” analysis of resultant cell behaviour. To isolate the contribution of mechanical cues from other HA-derived signals, we developed and characterised composite HA/gelatin scaffolds with different mineral contents along with a set of stiffness-matched HA-free gelatin scaffolds. Samples were seeded with human periosteal derived progenitor cells (PDPCs) and cultured over 7 days, analysing their resultant morphology and gene expression. Our results show that both stiffness and HA contribute to directing PDPC osteogenic differentiation, highlighting the role of stiffness in triggering the expression of osteogenic genes and of HA in accelerating the process, particularly at high concentrations.
Collapse
|