1
|
Getachew H, Mehrotra S, Kaur T, Fernandez-Godino R, Pierce EA, Garita-Hernandez M. The RNA content of extracellular vesicles from gene-edited PRPF31 +/- hiPSC-RPE show potential as biomarkers of retinal degeneration. Mol Ther Methods Clin Dev 2025; 33:101452. [PMID: 40231248 PMCID: PMC11995067 DOI: 10.1016/j.omtm.2025.101452] [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: 10/01/2024] [Accepted: 03/13/2025] [Indexed: 04/16/2025]
Abstract
Retinitis pigmentosa (RP) is the most common inherited retinal degeneration (IRD), causing vision loss via the dysfunction and death of photoreceptors and retinal pigment epithelium (RPE). Mutations in the PRPF31 gene are associated with autosomal dominant RP, impairing RPE function. While adeno-associated virus (AAV)-mediated gene therapy shows promise for treating IRDs, the slow progression of these diseases often makes timely measurement of clinical efficacy challenging. Extracellular vesicles (EVs) are lipid enclosed vesicles secreted by cells, and their RNA contents are being explored as circulating biomarkers for other diseases. We hypothesize that EV RNAs could serve as biomarkers of the health status of the neural retina and RPE. To test this, we used PRPF31 +/+ and PRPF31 +/- human induced pluripotent stem cell (hiPSC)-derived RPE (hi-RPE) to investigate the RNAs contained in RPE-derived EVs and how they change in disease. We also compared the RNA contents of RPE-EVs with the RNAs of the hi-RPE cells themselves. We found that EVs from mutant PRPF31 hi-RPE cells have distinct RNA profiles compared to those from control cells, suggesting that EV RNA contents change during disease. Additionally, we identified 18 miRNAs and 865 poly(A) RNAs enriched in EVs from PRPF31 +/- hi-RPE, which could serve as biomarkers for RPE degeneration.
Collapse
Affiliation(s)
- Heran Getachew
- Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Sudeep Mehrotra
- Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Tarandeep Kaur
- Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Rosario Fernandez-Godino
- Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Eric A. Pierce
- Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Marcela Garita-Hernandez
- Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
2
|
Emami A, Arabpour Z, Izadi E. Extracellular vesicles: essential agents in critical bone defect repair and therapeutic enhancement. Mol Biol Rep 2025; 52:113. [PMID: 39798011 DOI: 10.1007/s11033-024-10209-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025]
Abstract
Bone serves as a fundamental structural component in the body, playing pivotal roles in support, protection, mineral supply, and hormonal regulation. However, critical-sized bone injuries have become increasingly prevalent, necessitating extensive medical interventions due to limitations in the body's capacity for self-repair. Traditional approaches, such as autografts, allografts, and xenografts, have yielded unsatisfactory results. Stem cell therapy emerges as a promising avenue, but challenges like immune rejection and low cell survival rates hinder its widespread clinical implementation. Extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) have garnered attention for their regenerative capabilities, which surpass those of MSCs themselves. EVs offer advantages such as reduced immunogenicity, enhanced stability, and simplified storage, positioning them as a promising tool in stem cell-based therapies. This review explores the potential of EV-based therapy in bone tissue regeneration, delving into their biological characteristics, communication mechanisms, and preclinical applications across various physiological and pathological conditions. The mechanisms underlying EV-mediated bone regeneration, including angiogenesis, osteoblast proliferation, mineralization, and immunomodulation, are elucidated. Preclinical studies demonstrate the efficacy of EVs in promoting bone repair and neovascularization, even in pathological conditions like osteoporosis. EVs hold promise as a potential alternative for regenerating bone tissue, particularly in the context of critical-sized bone defects, offering new avenues for effective bone defect repair and management.
Collapse
Affiliation(s)
- Asrin Emami
- Iranian Tissue Bank and Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Zohreh Arabpour
- Department of Ophthalmology and Visual Science and University of Illinois, Chicago, IL, 60612, USA
| | - Elaheh Izadi
- Pediatric Cell, and Gene Therapy Research Center Gene, Cell and Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Zhu M, Yea JH, Li Z, Qin Q, Xu M, Xing X, Negri S, Archer M, Mittal M, Levi B, James AW. Pharmacologic or genetic targeting of peripheral nerves prevents peri-articular traumatic heterotopic ossification. Bone Res 2024; 12:54. [PMID: 39327413 PMCID: PMC11427465 DOI: 10.1038/s41413-024-00358-0] [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] [Revised: 07/08/2024] [Accepted: 07/16/2024] [Indexed: 09/28/2024] Open
Abstract
Heterotopic ossification (HO) is a pathological process that commonly arises following severe polytrauma, characterized by the anomalous differentiation of mesenchymal progenitor cells and resulting in the formation of ectopic bone in non-skeletal tissues. This abnormal bone growth contributes to pain and reduced mobility, especially when adjacent to a joint. Our prior observations suggested an essential role of NGF (Nerve Growth Factor)-responsive TrkA (Tropomyosin Receptor Kinase A)-expressing peripheral nerves in regulating abnormal osteochondral differentiation following tendon injury. Here, we utilized a recently developed mouse model of hip arthroplasty-induced HO to further validate the role of peripheral nerve regulation of traumatic HO. Nerve ingrowth was either modulated using a knockin transgenic animals with point mutation in TrkA, or local treatment with an FDA-approved formulation of long acting Bupivacaine which prevents peripheral nerve growth. Results demonstrate exuberant sensory and sympathetic nerve growth within the peri-articular HO site, and that both methods to reduce local innervation significantly reduced heterotopic bone formation. TrkA inhibition led to a 34% reduction in bone volume, while bupivacaine treatment resulted in a 50% decrease. Mechanistically, alterations in TGFβ and FGF signaling activation accompanied both methods of local denervation, and a shift in macrophages from M1 to M2 phenotypes was observed. In sum, these studies reinforce the observations that peripheral nerves play a role in the etiopathogenesis of HO, and that targeting local nerves represents a potential therapeutic approach for disease prevention.
Collapse
Affiliation(s)
- Manyu Zhu
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Ji-Hye Yea
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Zhao Li
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Qizhi Qin
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Mingxin Xu
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Xin Xing
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Stefano Negri
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
- Orthopedic Unit, University of Verona, Verona, Italy
| | - Mary Archer
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Monisha Mittal
- Department of Surgery, Center for Organogenesis and Trauma, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Benjamin Levi
- Department of Surgery, Center for Organogenesis and Trauma, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA.
| |
Collapse
|
4
|
Ren J, Huang P, Wang F. IGSF8 is a potential target for the treatment of gliomas. Asian J Surg 2024; 47:3883-3891. [PMID: 38453613 DOI: 10.1016/j.asjsur.2024.02.118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/14/2024] [Accepted: 02/22/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND Immunoglobulin superfamily member 8, or IGSF8, is a member of the recently identified immunoglobulin family of proteins. It is mostly produced on cell membranes and has a unique transmembrane structure. It has recently been demonstrated that there is a strong correlation between the expression variation of IGSF8 and the growth of gliomas. Therefore, we used data from the TCGA and CGGA databases to evaluate the function of IGSF8. METHODS The TCGA and GTEx data sets' RNA-seq data were utilized to examine IGSF8 expression. The Gene Cards database was utilized to get IGSF8 protein data. The Cluster Profiler data package was used to carry out the IGSF8 enrichment study. The GO and KEGG databases were used to examine the relationship between IGSF8 and cellular physiological and biochemical processes. The TCGA immune cell infiltration scores were obtained from online databases and published studies. Clinical survival data from TCGA and CGGA were used to investigate the predictive significance of IGSF8. RESULTS TGGA revealed that the majority of cancers had differential expression of IGSF8. IGSF8 was discovered to be enriched in numerous significant pathways in tumor cells by GO and KEGG. Moreover, a strong correlation was seen between the expression of IGSF8 and the immunomodulatory interactions that occur between non-lymphocytes and lymphocytes. T-cell infiltration, immunological checkpoints, immune-activating and immune-suppressive genes, chemokines, and chemokine receptors were all strongly correlated with IGSF8 expression. Lastly, the TCGA and CGGA databases showed a strong correlation between IGSF8 and the grade and prognosis of gliomas. CONCLUSION According to our findings, IGSF8 may be a glioma marker. In order to control the immunological microenvironment, IGSF8 may cooperate with a number of immune checkpoints. This information may be utilized to create novel targeted immunotherapy medications.
Collapse
Affiliation(s)
- Jiaxing Ren
- Inner Mongolia Medical University Affiliated Hospital, Inner Mongolia, China.
| | - Ping Huang
- Inner Mongolia Medical University Affiliated Hospital, Inner Mongolia, China.
| | - Fei Wang
- Inner Mongolia Medical University Affiliated Hospital, Inner Mongolia, China.
| |
Collapse
|
5
|
Xing X, Li Z, Xu J, Chen AZ, Archer M, Wang Y, Xu M, Wang Z, Zhu M, Qin Q, Thottappillil N, Zhou M, James AW. Requirement of Pdgfrα+ cells for calvarial bone repair. Stem Cells Transl Med 2024; 13:791-802. [PMID: 38986535 PMCID: PMC11328938 DOI: 10.1093/stcltm/szae041] [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: 02/29/2024] [Accepted: 05/19/2024] [Indexed: 07/12/2024] Open
Abstract
Platelet-derived growth factor receptor α (PDGFRα) is often considered as a general marker of mesenchymal cells and fibroblasts, but also shows expression in a portion of osteoprogenitor cells. Within the skeleton, Pdgfrα+ mesenchymal cells have been identified in bone marrow and periosteum of long bones, where they play a crucial role in participating in fracture repair. A similar examination of Pdgfrα+ cells in calvarial bone healing has not been examined. Here, we utilize Pdgfrα-CreERTM;mT/mG reporter animals to examine the contribution of Pdgfrα+ mesenchymal cells to calvarial bone repair through histology and single-cell RNA sequencing (scRNA-Seq). Results showed that Pdgfrα+ mesenchymal cells are present in several cell clusters by scRNA-Seq, and by histology a dramatic increase in Pdgfrα+ cells populated the defect site at early timepoints to give rise to healed bone tissue overtime. Notably, diphtheria toxin-mediated ablation of Pdgfrα reporter+ cells resulted in significantly impaired calvarial bone healing. Our findings suggest that Pdgfrα-expressing cells within the calvarial niche play a critical role in the process of calvarial bone repair.
Collapse
Affiliation(s)
- Xin Xing
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Zhao Li
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Jiajia Xu
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Austin Z Chen
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Mary Archer
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Yiyun Wang
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Mingxin Xu
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Ziyi Wang
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Manyu Zhu
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Qizhi Qin
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Neelima Thottappillil
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Myles Zhou
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| |
Collapse
|
6
|
Deluca A, Wagner A, Heimel P, Deininger C, Wichlas F, Redl H, Rohde E, Tempfer H, Gimona M, Traweger A. Synergistic effect of umbilical cord extracellular vesicles and rhBMP-2 to enhance the regeneration of a metaphyseal femoral defect in osteoporotic rats. Stem Cell Res Ther 2024; 15:144. [PMID: 38764077 PMCID: PMC11103988 DOI: 10.1186/s13287-024-03755-8] [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: 11/03/2023] [Accepted: 05/07/2024] [Indexed: 05/21/2024] Open
Abstract
BACKGROUND The aim of this study was to evaluate potential synergistic effects of a single, local application of human umbilical cord MSC-derived sEVs in combination with a low dose of recombinant human rhBMP-2 to promote the regeneration of a metaphyseal femoral defect in an osteoporotic rat model. METHODS 6 weeks after induction of osteoporosis by bilateral ventral ovariectomy and administration of a special diet, a total of 64 rats underwent a distal femoral metaphyseal osteotomy using a manual Gigli wire saw. Defects were stabilized with an adapted Y-shaped mini-locking plate and were subsequently treated with alginate only, or alginate loaded with hUC-MSC-sEVs (2 × 109), rhBMP-2 (1.5 µg), or a combination of sEVs and rhBMP-2 (n = 16 for each group). 6 weeks post-surgery, femora were evaluated by µCT, descriptive histology, and biomechanical testing. RESULTS Native radiographs and µCT analysis confirmed superior bony union with callus formation after treatment with hUC-MSC-sEVs in combination with a low dose of rhBMP-2. This finding was further substantiated by histology, showing robust defect consolidation 6 weeks after treatment. Torsion testing of the explanted femora revealed increased stiffness after application of both, rhBMP-2 alone, or in combination with sEVs, whereas torque was only significantly increased after treatment with rhBMP-2 together with sEVs. CONCLUSION The present study demonstrates that the co-application of hUC-MSC-sEVs can improve the efficacy of rhBMP-2 to promote the regeneration of osteoporotic bone defects.
Collapse
Affiliation(s)
- Amelie Deluca
- Institute of Tendon and Bone Regeneration, Salzburg, 5020, Austria.
- Department of Traumatology, KABEG-Klinikum Klagenfurt am Woerthersee, Klagenfurt, 9020, Austria.
| | - Andrea Wagner
- Institute of Tendon and Bone Regeneration, Salzburg, 5020, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, 1200, Austria
| | - Patrick Heimel
- Austrian Cluster for Tissue Regeneration, Vienna, 1200, Austria
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Vienna, 1200, Austria
| | - Christian Deininger
- Institute of Tendon and Bone Regeneration, Salzburg, 5020, Austria
- Department of Orthopedics and Traumatology, Salzburg University Hospital, Paracelsus Medical University, Salzburg, 5020, Austria
| | - Florian Wichlas
- Department of Orthopedics and Traumatology, Salzburg University Hospital, Paracelsus Medical University, Salzburg, 5020, Austria
| | - Heinz Redl
- Austrian Cluster for Tissue Regeneration, Vienna, 1200, Austria
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Vienna, 1200, Austria
| | - Eva Rohde
- GMP Unit, Spinal Cord Injury and Tissue Regeneration Centre Salzburg, Paracelsus Medical University, Salzburg, Austria
- Department of Transfusion Medicine, Salzburger Landeskliniken GesmbH, Paracelsus Medical University, Salzburg, Austria
| | - Herbert Tempfer
- Institute of Tendon and Bone Regeneration, Salzburg, 5020, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, 1200, Austria
| | - Mario Gimona
- GMP Unit, Spinal Cord Injury and Tissue Regeneration Centre Salzburg, Paracelsus Medical University, Salzburg, Austria
- Research Program "Nanovesicular Therapies", Paracelsus Medical University, Salzburg, Austria
| | - Andreas Traweger
- Institute of Tendon and Bone Regeneration, Salzburg, 5020, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, 1200, Austria
| |
Collapse
|
7
|
Wang P, Shao W, Li Z, Wang B, Lv X, Huang Y, Feng Y. Non-bone-derived exosomes: a new perspective on regulators of bone homeostasis. Cell Commun Signal 2024; 22:70. [PMID: 38273356 PMCID: PMC10811851 DOI: 10.1186/s12964-023-01431-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 12/09/2023] [Indexed: 01/27/2024] Open
Abstract
Accumulating evidence indicates that exosomes help to regulate bone homeostasis. The roles of bone-derived exosomes have been well-described; however, recent studies have shown that some non-bone-derived exosomes have better bone targeting ability than bone-derived exosomes and that their performance as a drug delivery vehicle for regulating bone homeostasis may be better than that of bone-derived exosomes, and the sources of non-bone-derived exosomes are more extensive and can thus be better for clinical needs. Here, we sort non-bone-derived exosomes and describe their composition and biogenesis. Their roles and specific mechanisms in bone homeostasis and bone-related diseases are also discussed. Furthermore, we reveal obstacles to current research and future challenges in the practical application of exosomes, and we provide potential strategies for more effective application of exosomes for the regulation of bone homeostasis and the treatment of bone-related diseases. Video Abstract.
Collapse
Affiliation(s)
- Ping Wang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wenkai Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zilin Li
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bo Wang
- Department of Rehabilitation, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao Lv
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yiyao Huang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Yong Feng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
8
|
Zheng X, Zhao D, Liu Y, Jin Y, Liu T, Li H, Liu D. Regeneration and anti-inflammatory effects of stem cells and their extracellular vesicles in gynecological diseases. Biomed Pharmacother 2023; 168:115739. [PMID: 37862976 DOI: 10.1016/j.biopha.2023.115739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/10/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023] Open
Abstract
There are many gynecological diseases, among which breast cancer (BC), cervical cancer (CC), endometriosis (EMs), and polycystic ovary syndrome (PCOS) are common and difficult to cure. Stem cells (SCs) are a focus of regenerative medicine. They are commonly used to treat organ damage and difficult diseases because of their potential for self-renewal and multidirectional differentiation. SCs are also commonly used for difficult-to-treat gynecological diseases because of their strong directional differentiation ability with unlimited possibilities, their tendency to adhere to the diseased tissue site, and their use as carriers for drug delivery. SCs can produce exosomes in a paracrine manner. Exosomes can be produced in large quantities and have the advantage of easy storage. Their safety and efficacy are superior to those of SCs, which have considerable potential in gynecological treatment, such as inhibiting endometrial senescence, promoting vascular reconstruction, and improving anti-inflammatory and immune functions. In this paper, we review the mechanisms of the regenerative and anti-inflammatory capacity of SCs and exosomes in incurable gynecological diseases and the current progress in their application in genetic engineering to provide a foundation for further research.
Collapse
Affiliation(s)
- Xu Zheng
- Changchun University of Chinese Medicine, Changchun 130117, China
| | - Dan Zhao
- Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun 130000, China
| | - Yang Liu
- Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun 130000, China
| | - Ye Jin
- Changchun University of Chinese Medicine, Changchun 130117, China
| | - Tianjia Liu
- Changchun University of Chinese Medicine, Changchun 130117, China; Baicheng Medical College, Baicheng 137000, China.
| | - Huijing Li
- Changchun University of Chinese Medicine, Changchun 130117, China.
| | - Da Liu
- Changchun University of Chinese Medicine, Changchun 130117, China.
| |
Collapse
|
9
|
Ball JR, Shelby T, Hernandez F, Mayfield CK, Lieberman JR. Delivery of Growth Factors to Enhance Bone Repair. Bioengineering (Basel) 2023; 10:1252. [PMID: 38002376 PMCID: PMC10669014 DOI: 10.3390/bioengineering10111252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/20/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
The management of critical-sized bone defects caused by nonunion, trauma, infection, malignancy, pseudoarthrosis, and osteolysis poses complex reconstruction challenges for orthopedic surgeons. Current treatment modalities, including autograft, allograft, and distraction osteogenesis, are insufficient for the diverse range of pathology encountered in clinical practice, with significant complications associated with each. Therefore, there is significant interest in the development of delivery vehicles for growth factors to aid in bone repair in these settings. This article reviews innovative strategies for the management of critical-sized bone loss, including novel scaffolds designed for controlled release of rhBMP, bioengineered extracellular vesicles for delivery of intracellular signaling molecules, and advances in regional gene therapy for sustained signaling strategies. Improvement in the delivery of growth factors to areas of significant bone loss has the potential to revolutionize current treatment for this complex clinical challenge.
Collapse
Affiliation(s)
- Jacob R. Ball
- Department of Orthopaedic Surgery, University of Southern California Keck School of Medicine, 1500 San Pablo St., Los Angeles, CA 90033, USA
| | | | | | | | | |
Collapse
|
10
|
Lee E, Moon JY, Ko JY, Park SY, Im GI. GSTT1 as a Predictive Marker and Enhancer for Osteogenic Potential of Human Adipose-Derived Stromal/Stem Cells. J Bone Miner Res 2023; 38:1480-1496. [PMID: 37537994 DOI: 10.1002/jbmr.4893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 07/23/2023] [Accepted: 08/01/2023] [Indexed: 08/05/2023]
Abstract
Adipose-derived stromal/stem cells (ASCs) have been extensively studied as cell sources for regenerative medicine for bone because of their excellent proliferative capacity and the ability to obtain a large number of cells with minimal donor morbidity. On the other hand, the differentiation potential of ASCs is generally lower than that of bone marrow-derived stromal/stem cells and varies greatly depending on donors. In this study, we mined a marker that can predict the osteogenic potential of ASC clones and also investigated the usefulness of the molecule as the enhancer of osteogenic differentiation of ASCs as well as its mechanism of action. Through RNA-seq gene analysis, we discovered that GSTT1 (Glutathione S-transferase theta-1) was the most distinguished gene marker between highly osteogenic and poorly osteogenic ASC clones. Knockdown of GSTT1 in high osteogenic ASCs by siGSTT1 treatment reduced mineralized matrix formation. On the other hand, GSTT1 overexpression by GSTT1 transfection or GSTT1 recombinant protein treatment enhanced osteogenic differentiation of low osteogenic ASCs. Metabolomic analysis confirmed significant changes of metabolites related to bone differentiation in ASCs transfected with GSTT1. A high total antioxidant capacity, low levels of cellular reactive oxygen species, and increased GSH/GSSG ratios were also detected in GSTT1-transfected ASCs. When the in vivo effect of GSTT1-transfected ASCs on bone regeneration was investigated with segmental long-bone defect model in rats, bone regeneration was significantly better after implantation of GSTT1-transfected ASCs compared with that of control vector-transfected ASCs. In conclusion, GSTT1 can be a useful marker to screen the highly osteogenic ASC clones and also a therapeutic factor to enhance the osteogenic differentiation of poorly osteogenic ASC clones. © 2023 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Eugene Lee
- Research Institute for Integrative Regenerative Biomedical Engineering, Dongguk University, Goyang, Republic of Korea
| | - Jae-Yeon Moon
- Research Institute for Integrative Regenerative Biomedical Engineering, Dongguk University, Goyang, Republic of Korea
| | - Ji-Yun Ko
- Research Institute for Integrative Regenerative Biomedical Engineering, Dongguk University, Goyang, Republic of Korea
| | - Seo-Young Park
- Research Institute for Integrative Regenerative Biomedical Engineering, Dongguk University, Goyang, Republic of Korea
| | - Gun-Il Im
- Research Institute for Integrative Regenerative Biomedical Engineering, Dongguk University, Goyang, Republic of Korea
- Department of Orthopedics, Dongguk University Ilsan Hospital, Goyang, Republic of Korea
| |
Collapse
|
11
|
Skelton AM, Cohen DJ, Boyan BD, Schwartz Z. Osteoblast-Derived Matrix Vesicles Exhibit Exosomal Traits and a Unique Subset of microRNA: Their Caveolae-Dependent Endocytosis Results in Reduced Osteogenic Differentiation. Int J Mol Sci 2023; 24:12770. [PMID: 37628952 PMCID: PMC10454939 DOI: 10.3390/ijms241612770] [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: 06/19/2023] [Revised: 08/08/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Matrix vesicles (MVs) are nano-sized extracellular vesicles that are anchored in the extracellular matrix (ECM). In addition to playing a role in biomineralization, osteoblast-derived MVs were recently suggested to have regulatory duties. The aims of this study were to establish the characteristics of osteoblast-derived MVs in the context of extracellular vesicles like exosomes, assess their role in modulating osteoblast differentiation, and examine their mechanism of uptake. MVs were isolated from the ECM of MG63 human osteoblast-like cell cultures and characterized via enzyme activity, transmission electron microscopy, nanoparticle tracking analysis, Western blot, and small RNA sequencing. Osteoblasts were treated with MVs from two different culture conditions (growth media [GM]; osteogenic media [OM]) to evaluate their effects on the differentiation and production of inflammatory markers and on macrophage polarization. MV endocytosis was assessed using a lipophilic, fluorescent dye and confocal microscopy with the role of caveolae determined using methyl-β-cyclodextrin. MVs exhibited a four-fold enrichment in alkaline phosphatase specific activity compared to plasma membranes; were 50-150 nm in diameter; possessed exosomal markers CD63, CD81, and CD9 and endosomal markers ALIX, TSG101, and HSP70; and were selectively enriched in microRNA linked to an anti-osteogenic effect and to M2 macrophage polarization. Treatment with GM or OM MVs decreased osteoblast differentiation. Osteoblasts endocytosed MVs using a mechanism that involves caveolae. These results support the hypothesis that osteoblasts produce MVs that participate in the regulation of osteogenesis.
Collapse
Affiliation(s)
- Anne M. Skelton
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (A.M.S.); (B.D.B.)
| | - D. Joshua Cohen
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | - Barbara D. Boyan
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (A.M.S.); (B.D.B.)
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA;
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Zvi Schwartz
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA;
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
12
|
Torrecillas-Baena B, Pulido-Escribano V, Dorado G, Gálvez-Moreno MÁ, Camacho-Cardenosa M, Casado-Díaz A. Clinical Potential of Mesenchymal Stem Cell-Derived Exosomes in Bone Regeneration. J Clin Med 2023; 12:4385. [PMID: 37445420 DOI: 10.3390/jcm12134385] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Bone metabolism is regulated by osteoblasts, osteoclasts, osteocytes, and stem cells. Pathologies such as osteoporosis, osteoarthritis, osteonecrosis, and traumatic fractures require effective treatments that favor bone formation and regeneration. Among these, cell therapy based on mesenchymal stem cells (MSC) has been proposed. MSC are osteoprogenitors, but their regenerative activity depends in part on their paracrine properties. These are mainly mediated by extracellular vesicle (EV) secretion. EV modulates regenerative processes such as inflammation, angiogenesis, cell proliferation, migration, and differentiation. Thus, MSC-EV are currently an important tool for the development of cell-free therapies in regenerative medicine. This review describes the current knowledge of the effects of MSC-EV in the different phases of bone regeneration. MSC-EV has been used by intravenous injection, directly or in combination with different types of biomaterials, in preclinical models of bone diseases. They have shown great clinical potential in regenerative medicine applied to bone. These findings should be confirmed through standardization of protocols, a better understanding of the mechanisms of action, and appropriate clinical trials. All that will allow the translation of such cell-free therapy to human clinic applications.
Collapse
Affiliation(s)
- Bárbara Torrecillas-Baena
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain
| | - Victoria Pulido-Escribano
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain
| | - Gabriel Dorado
- Department Bioquímica y Biología Molecular, Campus Rabanales C6-1-E17, Campus de Excelencia Internacional Agroalimentario (ceiA3), Universidad de Córdoba, 14071 Córdoba, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), 14004 Córdoba, Spain
| | - María Ángeles Gálvez-Moreno
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain
| | - Marta Camacho-Cardenosa
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain
| | - Antonio Casado-Díaz
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), 14004 Córdoba, Spain
| |
Collapse
|
13
|
Lu Y, Zhao L, Mao J, Liu W, Ma W, Zhao B. Rab27a-mediated extracellular vesicle secretion contributes to osteogenesis in periodontal ligament-bone niche communication. Sci Rep 2023; 13:8479. [PMID: 37231020 DOI: 10.1038/s41598-023-35172-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/14/2023] [Indexed: 05/27/2023] Open
Abstract
Periodontitis, an infectious and common disease worldwide, leads to the destruction of the periodontal ligament-alveolar bone complex. Within the bone metabolic niche, communication between periodontal ligament stem cells (PDLSCs) and bone marrow mesenchymal stem cells (BMMSCs) has been considered a major contributor to osteogenesis. PDLSC-derived extracellular vesicles (P-EVs) have shown great potential for bone regeneration. However, the secretion and uptake mechanisms of P-EVs remain elusive. Herein, the biogenesis of extracellular vesicles (EVs) from PDLSCs was observed using scanning and transmission electron microscopy. PDLSCs were transduced with Ras-associated protein 27a (Rab27a) siRNA (PDLSCsiRab27a) to inhibit EV secretion. The effect of P-EVs on BMMSCs was evaluated using a non-contact transwell co-culture system. We observed that Rab27a knockdown decreased EV secretion, and PDLSCsiRab27a remarkably attenuated co-culture-enhanced osteogenesis of BMMSCs. Isolated PDLSC-derived EVs enhanced osteogenic differentiation of BMMSCs in vitro and induced bone regeneration in a calvarial defect model in vivo. PDLSC-derived EVs were rapidly endocytosed by BMMSCs via the lipid raft/cholesterol endocytosis pathway and triggered the phosphorylation of extracellular signal-regulated kinase 1/2. In conclusion, PDLSCs contribute to the osteogenesis of BMMSCs through Rab27a-mediated EV secretion, thereby providing a potential cell-free approach for bone regeneration.
Collapse
Affiliation(s)
- Yun Lu
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, China
| | - Liru Zhao
- Department of Orthodontics, Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Jiaqi Mao
- Department of Orthodontics, Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Wen Liu
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, China
| | - Wensheng Ma
- Department of Orthodontics, Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, China.
| | - Bingjiao Zhao
- Department of Orthodontics, 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
|
14
|
Davies OG. Extracellular vesicles: From bone development to regenerative orthopedics. Mol Ther 2023; 31:1251-1274. [PMID: 36869588 PMCID: PMC10188641 DOI: 10.1016/j.ymthe.2023.02.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/31/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023] Open
Abstract
Regenerative medicine aims to promote the replacement of tissues lost to damage or disease. While positive outcomes have been observed experimentally, challenges remain in their clinical translation. This has led to growing interest in applying extracellular vesicles (EVs) to augment or even replace existing approaches. Through the engineering of culture environments or direct/indirect manipulation of EVs themselves, multiple avenues have emerged to modulate EV production, targeting, and therapeutic potency. Drives to modulate release using material systems or functionalize implants for improved osseointegration have also led to outcomes that could have real-world impact. The purpose of this review is to highlight advantages in applying EVs for the treatment of skeletal defects, outlining the current state of the art in the field and emphasizing avenues for further investigation. Notably, the review identifies inconsistencies in EV nomenclature and outstanding challenges in defining a reproducible therapeutic dose. Challenges also remain in the scalable manufacture of a therapeutically potent and pure EV product, with a need to address scalable cell sources and optimal culture environments. Addressing these issues will be critical if we are to develop regenerative EV therapies that meet the demands of regulators and can be translated from bench to bedside.
Collapse
Affiliation(s)
- Owen G Davies
- School of Sport, Exercise, and Health Sciences, Loughborough University, Epinal Way, Loughborough, Leicestershire LE11 3TU, UK.
| |
Collapse
|
15
|
Ren S, Lin Y, Liu W, Yang L, Zhao M. MSC-Exos: Important active factor of bone regeneration. Front Bioeng Biotechnol 2023; 11:1136453. [PMID: 36814713 PMCID: PMC9939647 DOI: 10.3389/fbioe.2023.1136453] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 01/24/2023] [Indexed: 02/08/2023] Open
Abstract
Bone defect and repair is a common but difficult problem in restorative and reconstructive surgery. Bone tissue defects of different sizes caused by different reasons bring functional limitations and cosmetic deformities to patients. Mesenchymal stem cells (MSC), a major hotspot in the field of regeneration in recent years, have been widely used in various studies on bone tissue regeneration. Numerous studies have shown that the bone regenerative effects of MSC can be achieved through exosome-delivered messages. Although its osteogenic mechanism is still unclear, it is clear that MSC-Exos can directly or indirectly support the action of bone regeneration. It can act directly on various cells associated with osteogenesis, or by carrying substances that affect cellular activators or the local internal environment in target cells, or it can achieve activation of the osteogenic framework by binding to materials. Therefore, this review aims to summarize the types and content of effective contents of MSC-Exos in bone regeneration, as well as recent advances in the currently commonly used methods to enable the binding of MSC-Exos to the framework and to conclude that MSC-Exos is effective in promoting osteogenesis.
Collapse
Affiliation(s)
- Sihang Ren
- Department of Plastic Surgery, The Second Hospital of Dalian Medical University, Dalian, China,Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China,NHC Key Laboratory of Reproductive Health and Medical Genetics (China Medical University), Liaoning Research Institute of Family Planning (The Affiliated Reproductive Hospital of China Medical University), Shenyang, China
| | - Yuyang Lin
- Department of Plastic Surgery, The Second Hospital of Dalian Medical University, Dalian, China
| | - Wenyue Liu
- Department of Plastic Surgery, The Second Hospital of Dalian Medical University, Dalian, China
| | - Liqun Yang
- NHC Key Laboratory of Reproductive Health and Medical Genetics (China Medical University), Liaoning Research Institute of Family Planning (The Affiliated Reproductive Hospital of China Medical University), Shenyang, China,Department of Biomaterials, Shengjing Hospital of China Medical University, Shenyang, China,*Correspondence: Liqun Yang, ; Muxin Zhao,
| | - Muxin Zhao
- Department of Plastic Surgery, The Second Hospital of Dalian Medical University, Dalian, China,*Correspondence: Liqun Yang, ; Muxin Zhao,
| |
Collapse
|
16
|
Effect of ozone therapy on the modulation of inflammation and on new bone formation in critical defects of rat calvaria filled with autogenous graft. JOURNAL OF STOMATOLOGY, ORAL AND MAXILLOFACIAL SURGERY 2023; 124:101292. [PMID: 36150689 DOI: 10.1016/j.jormas.2022.09.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/19/2022] [Accepted: 09/19/2022] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The aim of this study was to evaluate the effect of ozone therapy on new bone formation and inflammation modulation in defects of rat calvaria filled with autogenous bone. MATERIAL AND METHODS Critical size defects were created in the calvaria of 24 male Wistar rats. The animals were randomly divided into four groups according to the treatment: G1: clot; G2: clot and covered with xenogenic membrane; G3: particulate autogenous bone graft; G4: autogenous bone graft and application of 3 mL O2/O3 gas mixture (10 µg/ml). The defects were filled immediately after surgery with a bilateral retroauricular application, in the region immediately above the incision. After 21 days, the animals were euthanized, and the samples were processed for morphometric evaluations designed to measure both the intensity of the inflammatory infiltrate, and the presence of new bone formation in the defect. RESULTS The results showed a lower inflammation score and higher mean of newly formed bone in the region of the defect for the group associated with ozone therapy (G4). The bone formed in the region of the defect could be observed as being more lamellar and mineralized in the case of associated ozone therapy. CONCLUSION Ozone therapy represents a promising adjuvant therapy to accelerate tissue regeneration.
Collapse
|
17
|
Exosomes in Cerebral Ischemia-Reperfusion Injury: Current Perspectives and Future Challenges. Brain Sci 2022; 12:brainsci12121657. [PMID: 36552117 PMCID: PMC9776031 DOI: 10.3390/brainsci12121657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/18/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022] Open
Abstract
Cerebral ischemia impedes the functional or metabolic demands of the central nervous system (CNS), which subsequently leads to irreversible brain damage. While recanalization of blocked vessels recovers cerebral blood flow, it can also aggravate brain injury, termed as ischemia/reperfusion (I/R) injury. Exosomes, nanometric membrane vesicles, attracted wide attention as carriers of biological macromolecules. In the brain, exosomes can be secreted by almost all types of cells, and their contents can be altered during the pathological and clinical processes of cerebral I/R injury. Herein, we will review the current literature on the possible role of cargos derived from exosomes and exosomes-mediated intercellular communication in cerebral I/R injury. The PubMed and Web of Science databases were searched through January 2015. The studies published in English were identified using search terms including "exosomes", "cerebral ischemia-reperfusion injury", "brain ischemia-reperfusion injury", and "stroke". We will also focus on the potential therapeutic effects of stem cell-derived exosomes and underlying mechanisms in cerebral I/R injury. Meanwhile, with the advantages of low immunogenicity and cytotoxicity, high bioavailability, and the capacity to pass through the blood-brain barrier, exosomes also attract more attention as therapeutic modalities for the treatment of cerebral I/R injury.
Collapse
|
18
|
Paré A, Charbonnier B, Veziers J, Vignes C, Dutilleul M, De Pinieux G, Laure B, Bossard A, Saucet-Zerbib A, Touzot-Jourde G, Weiss P, Corre P, Gauthier O, Marchat D. Standardized and axially vascularized calcium phosphate-based implants for segmental mandibular defects: A promising proof of concept. Acta Biomater 2022; 154:626-640. [PMID: 36210043 DOI: 10.1016/j.actbio.2022.09.071] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 09/09/2022] [Accepted: 09/28/2022] [Indexed: 12/14/2022]
Abstract
The reconstruction of massive segmental mandibular bone defects (SMDs) remains challenging even today; the current gold standard in human clinics being vascularized bone transplantation (VBT). As alternative to this onerous approach, bone tissue engineering strategies have been widely investigated. However, they displayed limited clinical success, particularly in failing to address the essential problem of quick vascularization of the implant. Although routinely used in clinics, the insertion of intrinsic vascularization in bioengineered constructs for the rapid formation of a feeding angiosome remains uncommon. In a clinically relevant model (sheep), a custom calcium phosphate-based bioceramic soaked with autologous bone marrow and perfused by an arteriovenous loop was tested to regenerate a massive SMD and was compared to VBT (clinical standard). Animals did not support well the VBT treatment, and the study was aborted 2 weeks after surgery due to ethical and animal welfare considerations. SMD regeneration was successful with the custom vascularized bone construct. Implants were well osseointegrated and vascularized after only 3 months of implantation and totally entrapped in lamellar bone after 12 months; a healthy yellow bone marrow filled the remaining space. STATEMENT OF SIGNIFICANCE: Regenerative medicine struggles with the generation of large functional bone volume. Among them segmental mandibular defects are particularly challenging to restore. The standard of care, based on bone free flaps, still displays ethical and technical drawbacks (e.g., donor site morbidity). Modern engineering technologies (e.g., 3D printing, digital chain) were combined to relevant surgical techniques to provide a pre-clinical proof of concept, investigating for the benefits of such a strategy in bone-related regenerative field. Results proved that a synthetic-biologics-free approach is able to regenerate a critical size segmental mandibular defect of 15 cm3 in a relevant preclinical model, mimicking real life scenarii of segmental mandibular defect, with a full physiological regeneration of the defect after 12 months.
Collapse
Affiliation(s)
- Arnaud Paré
- INSERM, U 1229, Laboratory of Regenerative Medicine and Skeleton, RMeS, Nantes Université, 1 Place Alexis Ricordeau, Nantes 44042, France; Department of Maxillofacial and Plastic surgery, Burn Unit, University Hospital of Tours, Trousseau Hospital, Avenue de la République, Chambray lès Tours 37170, France
| | - Baptiste Charbonnier
- INSERM, U 1229, Laboratory of Regenerative Medicine and Skeleton, RMeS, Nantes Université, 1 Place Alexis Ricordeau, Nantes 44042, France; Mines Saint-Étienne, Univ Jean Monnet, INSERM, U 1059 Sainbiose, 42023, Saint-Étienne, France
| | - Joëlle Veziers
- INSERM, U 1229, Laboratory of Regenerative Medicine and Skeleton, RMeS, Nantes Université, 1 Place Alexis Ricordeau, Nantes 44042, France
| | - Caroline Vignes
- INSERM, U 1229, Laboratory of Regenerative Medicine and Skeleton, RMeS, Nantes Université, 1 Place Alexis Ricordeau, Nantes 44042, France
| | - Maeva Dutilleul
- INSERM, U 1229, Laboratory of Regenerative Medicine and Skeleton, RMeS, Nantes Université, 1 Place Alexis Ricordeau, Nantes 44042, France
| | - Gonzague De Pinieux
- Department of Pathology, University Hospital of Tours, Trousseau Hospital, Avenue de la République, Chambray lès Tours 37170, France
| | - Boris Laure
- Department of Maxillofacial and Plastic surgery, Burn Unit, University Hospital of Tours, Trousseau Hospital, Avenue de la République, Chambray lès Tours 37170, France
| | - Adeline Bossard
- ONIRIS Nantes-Atlantic College of Veterinary Medicine, Research Center of Preclinical Invesitagtion (CRIP), Site de la Chantrerie, 101 route de Gachet, Nantes 44307, France
| | - Annaëlle Saucet-Zerbib
- ONIRIS Nantes-Atlantic College of Veterinary Medicine, Research Center of Preclinical Invesitagtion (CRIP), Site de la Chantrerie, 101 route de Gachet, Nantes 44307, France
| | - Gwenola Touzot-Jourde
- INSERM, U 1229, Laboratory of Regenerative Medicine and Skeleton, RMeS, Nantes Université, 1 Place Alexis Ricordeau, Nantes 44042, France; ONIRIS Nantes-Atlantic College of Veterinary Medicine, Research Center of Preclinical Invesitagtion (CRIP), Site de la Chantrerie, 101 route de Gachet, Nantes 44307, France
| | - Pierre Weiss
- INSERM, U 1229, Laboratory of Regenerative Medicine and Skeleton, RMeS, Nantes Université, 1 Place Alexis Ricordeau, Nantes 44042, France
| | - Pierre Corre
- INSERM, U 1229, Laboratory of Regenerative Medicine and Skeleton, RMeS, Nantes Université, 1 Place Alexis Ricordeau, Nantes 44042, France; Clinique de Stomatologie et Chirurgie Maxillo-Faciale, Nantes University Hospital, 1 Place Alexis Ricordeau, Nantes 44042, France
| | - Olivier Gauthier
- INSERM, U 1229, Laboratory of Regenerative Medicine and Skeleton, RMeS, Nantes Université, 1 Place Alexis Ricordeau, Nantes 44042, France; ONIRIS Nantes-Atlantic College of Veterinary Medicine, Research Center of Preclinical Invesitagtion (CRIP), Site de la Chantrerie, 101 route de Gachet, Nantes 44307, France
| | - David Marchat
- Mines Saint-Étienne, Univ Jean Monnet, INSERM, U 1059 Sainbiose, 42023, Saint-Étienne, France.
| |
Collapse
|
19
|
The Use of Mesenchymal Stem Cells in the Complex Treatment of Kidney Tuberculosis (Experimental Study). Biomedicines 2022; 10:biomedicines10123062. [PMID: 36551818 PMCID: PMC9775022 DOI: 10.3390/biomedicines10123062] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/07/2022] [Accepted: 11/18/2022] [Indexed: 11/30/2022] Open
Abstract
In recent years, the application of mesenchymal stem cells (MSCs) has been recognized as a promising method for treatment of different diseases associated with inflammation and sclerosis, which include nephrotuberculosis. The aim of our study is to investigate the effectiveness of MSCs in the complex therapy of experimental rabbit kidney tuberculosis and to evaluate the effect of cell therapy on the reparative processes. Methods: To simulate kidney tuberculosis, a suspension of the standard strain Mycobacterium tuberculosis H37Rv (106 CFU) was used, which was injected into the cortical layer of the lower pole parenchyma of the left kidney under ultrasound control in rabbits. Anti-tuberculosis therapy (aTBT) was started on the 18th day after infection. MSCs (5 × 107 cells) were transplanted intravenously after the start of aTBT. Results: 2.5 months after infection, all animals showed renal failure. Conducted aTBT significantly reduced the level of albumin, ceruloplasmin, elastase and the severity of disorders in the proteinase/inhibitor system and increased the productive nature of inflammation. A month after MSC transplantation, the level of inflammatory reaction activity proteins decreased, the area of specific and destructive inflammation in kidneys decreased and the formation of mature connective tissue was noted, which indicates the reparative reaction activation.
Collapse
|
20
|
Invasiveness of a Growth-Migration System in a Two-dimensional Percolation cluster: A Stochastic Mathematical Approach. Bull Math Biol 2022; 84:104. [DOI: 10.1007/s11538-022-01056-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 07/20/2022] [Indexed: 11/02/2022]
|
21
|
Alejo AL, McDermott S, Khalil Y, Ball HC, Robinson GT, Solorzano E, Alejo AM, Douglas J, Samson TK, Young JW, Safadi FF. A Pre-clinical Standard Operating Procedure for Evaluating Orthobiologics in an In Vivo Rat Spinal Fusion Model. JOURNAL OF ORTHOPAEDICS AND SPORTS MEDICINE 2022; 4:224-240. [PMID: 36203492 PMCID: PMC9534599 DOI: 10.26502/josm.511500060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The rat animal model is a cost effective and reliable model used in spinal pre-clinical research. Complications from various surgical procedures in humans often arise that were based on these pre-clinical animal models. Therefore safe and efficacious pre-clinical animal models are needed to establish continuity into clinical trials. A Standard Operating Procedure (SOP) is a validated method that allows researchers to safely and carefully replicate previously successful surgical techniques. Thus, the aim of this study is to describe in detail the procedures involved in a common rat bilateral posterolateral intertransverse spinal fusion SOP used to test the efficacy and safety different orthobiologics using a collagen-soaked sponge as an orthobiologic carrier. Only two orthobiologics are currently FDA approved for spinal fusion surgery which include recombinant bone morphogenetic protein 2 (rhBMP-2), and I-FACTOR. While there are many additional orthobiologics currently being tested, one way to show their safety profile and gain FDA approval, is to use well established pre-clinical animal models. A preoperative, intraoperative, and postoperative surgical setup including specific anesthesia and euthanasia protocols are outlined. Furthermore, we describe different postoperative methods used to validate the spinal fusion SOP, which include μCT analysis, histopathology, biomechanical testing, and blood analysis. This SOP can help increase validity, transparency, efficacy, and reproducibly in future rat spinal fusion surgery procedures.
Collapse
Affiliation(s)
- Andrew L Alejo
- College of Medicine, Northeast Ohio Medical University (NEOMED), Rootstown, OH, USA
- Musculoskeletal Research Group, NEOMED, Rootstown, OH, USA
| | - Scott McDermott
- Roper St. Francis Physician Partners Orthopaedics, Summerville, SC, USA
| | - Yusuf Khalil
- College of Medicine, Northeast Ohio Medical University (NEOMED), Rootstown, OH, USA
- Musculoskeletal Research Group, NEOMED, Rootstown, OH, USA
| | - Hope C Ball
- Musculoskeletal Research Group, NEOMED, Rootstown, OH, USA
| | - Gabrielle T Robinson
- College of Graduate Studies, NEOMED, Rootstown, OH, USA
- Musculoskeletal Research Group, NEOMED, Rootstown, OH, USA
| | - Ernesto Solorzano
- College of Graduate Studies, NEOMED, Rootstown, OH, USA
- Musculoskeletal Research Group, NEOMED, Rootstown, OH, USA
| | - Amanda M Alejo
- College of Medicine, Northeast Ohio Medical University (NEOMED), Rootstown, OH, USA
- Musculoskeletal Research Group, NEOMED, Rootstown, OH, USA
| | - Jacob Douglas
- College of Medicine, Northeast Ohio Medical University (NEOMED), Rootstown, OH, USA
- Musculoskeletal Research Group, NEOMED, Rootstown, OH, USA
| | - Trinity K Samson
- College of Medicine, Northeast Ohio Medical University (NEOMED), Rootstown, OH, USA
- College of Graduate Studies, NEOMED, Rootstown, OH, USA
- Musculoskeletal Research Group, NEOMED, Rootstown, OH, USA
| | - Jesse W Young
- College of Medicine, Northeast Ohio Medical University (NEOMED), Rootstown, OH, USA
- College of Graduate Studies, NEOMED, Rootstown, OH, USA
- Musculoskeletal Research Group, NEOMED, Rootstown, OH, USA
| | - Fayez F Safadi
- College of Medicine, Northeast Ohio Medical University (NEOMED), Rootstown, OH, USA
- College of Graduate Studies, NEOMED, Rootstown, OH, USA
- Musculoskeletal Research Group, NEOMED, Rootstown, OH, USA
- Rebecca D. Considine Research Institute, Akron Children's Hospital, Akron, OH, USA
- GPN Therapeutics Inc., Redi Zone NEOMED, Rootstown, OH, USA
| |
Collapse
|
22
|
Xu J, Zhang Z, Zhao J, Meyers CA, Lee S, Qin Q, James AW. Interaction between the nervous and skeletal systems. Front Cell Dev Biol 2022; 10:976736. [PMID: 36111341 PMCID: PMC9468661 DOI: 10.3389/fcell.2022.976736] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/08/2022] [Indexed: 11/14/2022] Open
Abstract
The skeleton is one of the largest organ systems in the body and is richly innervated by the network of nerves. Peripheral nerves in the skeleton include sensory and sympathetic nerves. Crosstalk between bones and nerves is a hot topic of current research, yet it is not well understood. In this review, we will explore the role of nerves in bone repair and remodeling, as well as summarize the molecular mechanisms by which neurotransmitters regulate osteogenic differentiation. Furthermore, we discuss the skeleton’s role as an endocrine organ that regulates the innervation and function of nerves by secreting bone-derived factors. An understanding of the interactions between nerves and bone can help to prevent and treat bone diseases caused by abnormal innervation or nerve function, develop new strategies for clinical bone regeneration, and improve patient outcomes.
Collapse
Affiliation(s)
- Jiajia Xu
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Academy of Orthopedics, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zhongmin Zhang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junjie Zhao
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Carolyn A. Meyers
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Seungyong Lee
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
- Department of Physical Education, Incheon National University, Incheon, South Korea
| | - Qizhi Qin
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Aaron W. James
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
- *Correspondence: Aaron W. James,
| |
Collapse
|
23
|
Cheng P, Cao T, Zhao X, Lu W, Miao S, Ning F, Wang D, Gao Y, Wang L, Pei G, Yang L. Nidogen1-enriched extracellular vesicles accelerate angiogenesis and bone regeneration by targeting Myosin-10 to regulate endothelial cell adhesion. Bioact Mater 2022; 12:185-197. [PMID: 35310379 PMCID: PMC8897190 DOI: 10.1016/j.bioactmat.2021.10.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/15/2021] [Accepted: 10/18/2021] [Indexed: 12/17/2022] Open
Abstract
The technique bottleneck of repairing large bone defects with tissue engineered bone is the vascularization of tissue engineered grafts. Although some studies have shown that extracellular vesicles (EVs) derived from bone marrow mesenchymal stem cells (BMSCs) promote bone healing and repair by accelerating angiogenesis, the effector molecules and the mechanism remain unclear, which fail to provide ideas for the future research and development of cell-free interventions. Here, we found that Nidogen1-enriched EV (EV-NID1) derived from BMSCs interferes with the formation and assembly of focal adhesions (FAs) by targeting myosin-10, thereby reducing the adhesion strength of rat arterial endothelial cells (RAECs) to the extracellular matrix (ECM), and enhancing the migration and angiogenesis potential of RAECs. Moreover, by delivery with composite hydrogel, EV-NID1 is demonstrated to promote angiogenesis and bone regeneration in rat femoral defects. This study identifies the intracellular binding target of EV-NID1 and further elucidates a novel approach and mechanism, thereby providing a cell-free construction strategy with precise targets for the development of vascularized tissue engineering products. Nidogen1 is enriched in extracellular vesicles (EV-NID1) derived from BMSCs. EV-NID1 interferes with the formation and assembly of focal adhesions (FAs). Myosin-10 was identified as the intracellular binding target of EV-NID1. The composite hydrogel loaded with EV-NID1 promotes the repair of bone defects by accelerating angiogenesis.
Collapse
Affiliation(s)
- Pengzhen Cheng
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
- College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Tianqing Cao
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xueyi Zhao
- College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Weiguang Lu
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Sheng Miao
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Fenru Ning
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Dong Wang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yi Gao
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Long Wang
- Department of Orthopaedics, Chinese PLA General Hospital, Beijing, 100853, China
| | - Guoxian Pei
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
- Corresponding author.
| | - Liu Yang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
- Corresponding author.
| |
Collapse
|
24
|
Zhou X, Liu J, Zheng Y, Zhang Z, Wu Y, Yang W, Liu J, Huang Y, Yi Y, Zhao Z, Xiao H, Mo X, Wang J. SM22α-lineage niche cells regulate intramembranous bone regeneration via PDGFRβ-triggered hydrogen sulfide production. Cell Rep 2022; 39:110750. [PMID: 35508129 DOI: 10.1016/j.celrep.2022.110750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 02/02/2022] [Accepted: 04/06/2022] [Indexed: 02/08/2023] Open
Abstract
Bone stromal cells are critical for bone homeostasis and regeneration. Growing evidence suggests that non-stem bone niche cells support bone homeostasis and regeneration via paracrine mechanisms, which remain to be elucidated. Here, we show that physiologically quiescent SM22α-lineage stromal cells expand after bone injury to regulate diverse processes of intramembranous bone regeneration. The majority of SM22α-lineage cells neither act as stem cells in vivo nor show their expression patterns. Dysfunction of SM22α-lineage niche cells induced by loss of platelet-derived growth factor receptor β (PDGFRβ) impairs bone repair. We further show that PDGFRβ-triggered hydrogen sulfide (H2S) generation in SM22α-lineage niche cells facilitates osteogenesis and angiogenesis and suppresses overactive osteoclastogenesis. Collectively, these data demonstrate that non-stem SM22α-lineage niche cells support the niche for bone regeneration with a PDGFRβ/H2S-dependent regulatory mechanism. Our findings provide further insight into non-stem bone stromal niche cell populations and niche-regulation strategy for bone repair.
Collapse
Affiliation(s)
- Xueman Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China; Laboratory of Aging Research, State Key Laboratory of Biotherapy & National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jin Liu
- Laboratory of Aging Research, State Key Laboratory of Biotherapy & National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Yingcheng Zheng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China; Laboratory of Aging Research, State Key Laboratory of Biotherapy & National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenzhen Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China; Laboratory of Aging Research, State Key Laboratory of Biotherapy & National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yange Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wenke Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiaqi Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yanmei Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yating Yi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hengyi Xiao
- Laboratory of Aging Research, State Key Laboratory of Biotherapy & National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xianming Mo
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Jun Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
25
|
Ginini L, Billan S, Fridman E, Gil Z. Insight into Extracellular Vesicle-Cell Communication: From Cell Recognition to Intracellular Fate. Cells 2022; 11:1375. [PMID: 35563681 PMCID: PMC9101098 DOI: 10.3390/cells11091375] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 01/27/2023] Open
Abstract
Extracellular vesicles (EVs) are heterogamous lipid bilayer-enclosed membranous structures secreted by cells. They are comprised of apoptotic bodies, microvesicles, and exosomes, and carry a range of nucleic acids and proteins that are necessary for cell-to-cell communication via interaction on the cells surface. They initiate intracellular signaling pathways or the transference of cargo molecules, which elicit pleiotropic responses in recipient cells in physiological processes, as well as pathological processes, such as cancer. It is therefore important to understand the molecular means by which EVs are taken up into cells. Accordingly, this review summarizes the underlying mechanisms involved in EV targeting and uptake. The primary method of entry by EVs appears to be endocytosis, where clathrin-mediated, caveolae-dependent, macropinocytotic, phagocytotic, and lipid raft-mediated uptake have been variously described as being prevalent. EV uptake mechanisms may depend on proteins and lipids found on the surfaces of both vesicles and target cells. As EVs have been shown to contribute to cancer growth and progression, further exploration and targeting of the gateways utilized by EVs to internalize into tumor cells may assist in the prevention or deceleration of cancer pathogenesis.
Collapse
Affiliation(s)
- Lana Ginini
- Rappaport Family Institute for Research in the Medical Sciences, Technion–Israel Institute of Technology, Haifa 31096, Israel; (L.G.); (E.F.)
| | - Salem Billan
- Head and Neck Institute, The Holy Family Hospital Nazareth, Nazareth 1641100, Israel;
- Medical Oncology and Radiation Therapy Program, Oncology Section, Rambam Health Care Campus, HaAliya HaShniya Street 8, Haifa 3109601, Israel
| | - Eran Fridman
- Rappaport Family Institute for Research in the Medical Sciences, Technion–Israel Institute of Technology, Haifa 31096, Israel; (L.G.); (E.F.)
| | - Ziv Gil
- Head and Neck Institute, The Holy Family Hospital Nazareth, Nazareth 1641100, Israel;
| |
Collapse
|
26
|
Meng F, Xue X, Yin Z, Gao F, Wang X, Geng Z. Research Progress of Exosomes in Bone Diseases: Mechanism, Diagnosis and Therapy. Front Bioeng Biotechnol 2022; 10:866627. [PMID: 35497358 PMCID: PMC9039039 DOI: 10.3389/fbioe.2022.866627] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/15/2022] [Indexed: 11/13/2022] Open
Abstract
With the global escalation of the aging process, the number of patients with bone diseases is increasing year by year. Currently, there are limited effective treatments for bone diseases. Exosome, as a vital medium in cell-cell communication, can mediate tissue metabolism through the paracrine transmission of various cargos (proteins, nucleic acids, lipids, etc.) carried by itself. Recently, an increasing number of researchers have proven that exosomes play essential roles in the formation, metabolism, and pathological changes of bone and cartilage. Because exosomes have the advantages of small size, rich sources, and low immunogenicity, they can be used not only as substitutes for the traditional treatment of bone diseases, but also as biomarkers for the diagnosis of bone diseases. This paper reviews the research progress of several kinds of cells derived-exosomes in bone diseases and provides a theoretical basis for further research and clinical application of exosomes in bone diseases in the future.
Collapse
Affiliation(s)
- Fanying Meng
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Xu Xue
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Zhifeng Yin
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, China
| | - Fei Gao
- Institute of Translational Medicine, Shanghai University, Shanghai, China,*Correspondence: Fei Gao, ; Xiuhui Wang, ; Zhen Geng,
| | - Xiuhui Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, China,*Correspondence: Fei Gao, ; Xiuhui Wang, ; Zhen Geng,
| | - Zhen Geng
- Institute of Translational Medicine, Shanghai University, Shanghai, China,*Correspondence: Fei Gao, ; Xiuhui Wang, ; Zhen Geng,
| |
Collapse
|
27
|
Xu J, Li Z, Tower RJ, Negri S, Wang Y, Meyers CA, Sono T, Qin Q, Lu A, Xing X, McCarthy EF, Clemens TL, James AW. NGF-p75 signaling coordinates skeletal cell migration during bone repair. SCIENCE ADVANCES 2022; 8:eabl5716. [PMID: 35302859 PMCID: PMC8932666 DOI: 10.1126/sciadv.abl5716] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 01/26/2022] [Indexed: 05/26/2023]
Abstract
Bone regeneration following injury is initiated by inflammatory signals and occurs in association with infiltration by sensory nerve fibers. Together, these events are believed to coordinate angiogenesis and tissue reprogramming, but the mechanism of coupling immune signals to reinnervation and osteogenesis is unknown. Here, we found that nerve growth factor (NGF) is expressed following cranial bone injury and signals via p75 in resident mesenchymal osteogenic precursors to affect their migration into the damaged tissue. Mice lacking Ngf in myeloid cells demonstrated reduced migration of osteogenic precursors to the injury site with consequently delayed bone healing. These features were phenocopied by mice lacking p75 in Pdgfra+ osteoblast precursors. Single-cell transcriptomics identified mesenchymal subpopulations with potential roles in cell migration and immune response, altered in the context of p75 deletion. Together, these results identify the role of p75 signaling pathway in coordinating skeletal cell migration during early bone repair.
Collapse
Affiliation(s)
- Jiajia Xu
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Zhao Li
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Robert J. Tower
- Department of Orthopaedics, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Stefano Negri
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Orthopaedics and Traumatology, University of Verona, Verona 37129, Italy
| | - Yiyun Wang
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Carolyn A. Meyers
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Takashi Sono
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Qizhi Qin
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Amy Lu
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Xin Xing
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Edward F. McCarthy
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Thomas L. Clemens
- Department of Orthopaedics, Johns Hopkins University, Baltimore, MD 21205, USA
- Baltimore Veterans Administration Medical Center, Baltimore, MD 21201, USA
| | - Aaron W. James
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
28
|
Brown SV, Dewitt S, Clayton A, Waddington RJ. Identifying the Efficacy of Extracellular Vesicles in Osteogenic Differentiation: An EV-Lution in Regenerative Medicine. FRONTIERS IN DENTAL MEDICINE 2022. [DOI: 10.3389/fdmed.2022.849724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have long been the focus for regenerative medicine and the restoration of damaged or aging cells throughout the body. However, the efficacy of MSCs in cell-based therapy still remains unpredictable and carries with it enumerable risks. It is estimated that only 3-10% of MSCs survive transplantation, and there remains undefined and highly variable heterogeneous biological potency within these administered cell populations. The mode of action points to secreted factors produced by MSCs rather than the reliance on engraftment. Hence harnessing such secreted elements as a replacement for live-cell therapies is attractive. Extracellular vesicles (EVs) are heterogenous lipid bounded structures, secreted by cells. They comprise a complex repertoire of molecules including RNA, proteins and other factors that facilitate cell-to-cell communication. Described as protected signaling centers, EVs can modify the cellular activity of recipient cells and are emerging as a credible alternative to cell-based therapies. EV therapeutics demonstrate beneficial roles for wound healing by preventing apoptosis, moderating immune responses, and stimulating angiogenesis, in addition to promoting cell proliferation and differentiation required for tissue matrix synthesis. Significantly, EVs maintain their signaling function following transplantation, circumventing the issues related to cell-based therapies. However, EV research is still in its infancy in terms of their utility as medicinal agents, with many questions still surrounding mechanistic understanding, optimal sourcing, and isolation of EVs for regenerative medicine. This review will consider the efficacy of using cell-derived EVs compared to traditional cell-based therapies for bone repair and regeneration. We discuss the factors to consider in developing productive lines of inquiry and establishment of standardized protocols so that EVs can be harnessed from optimal secretome production, to deliver reproducible and effective therapies.
Collapse
|
29
|
Educating EVs to Improve Bone Regeneration: Getting Closer to the Clinic. Int J Mol Sci 2022; 23:ijms23031865. [PMID: 35163787 PMCID: PMC8836395 DOI: 10.3390/ijms23031865] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/28/2022] [Accepted: 02/05/2022] [Indexed: 12/11/2022] Open
Abstract
The incidence of bone-related disorders is continuously growing as the aging of the population in developing countries continues to increase. Although therapeutic interventions for bone regeneration exist, their effectiveness is questioned, especially under certain circumstances, such as critical size defects. This gap of curative options has led to the search for new and more effective therapeutic approaches for bone regeneration; among them, the possibility of using extracellular vesicles (EVs) is gaining ground. EVs are secreted, biocompatible, nano-sized vesicles that play a pivotal role as messengers between donor and target cells, mediated by their specific cargo. Evidence shows that bone-relevant cells secrete osteoanabolic EVs, whose functionality can be further improved by several strategies. This, together with the low immunogenicity of EVs and their storage advantages, make them attractive candidates for clinical prospects in bone regeneration. However, before EVs reach clinical translation, a number of concerns should be addressed. Unraveling the EVs’ mode of action in bone regeneration is one of them; the molecular mediators driving their osteoanabolic effects in acceptor cells are now beginning to be uncovered. Increasing the functional and bone targeting abilities of EVs are also matters of intense research. Here, we summarize the cell sources offering osteoanabolic EVs, and the current knowledge about the molecular cargos that mediate bone regeneration. Moreover, we discuss strategies under development to improve the osteoanabolic and bone-targeting potential of EVs.
Collapse
|
30
|
Vermeulen S, Birgani ZT, Habibovic P. Biomaterial-induced pathway modulation for bone regeneration. Biomaterials 2022; 283:121431. [DOI: 10.1016/j.biomaterials.2022.121431] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/28/2022] [Accepted: 02/17/2022] [Indexed: 12/18/2022]
|
31
|
Zhao B, Chen Q, Zhao L, Mao J, Huang W, Han X, Liu Y. Periodontal Ligament Stem Cell-Derived Small Extracellular Vesicles Embedded in Matrigel Enhance Bone Repair Through the Adenosine Receptor Signaling Pathway. Int J Nanomedicine 2022; 17:519-536. [PMID: 35140462 PMCID: PMC8819539 DOI: 10.2147/ijn.s346755] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
Purpose Small extracellular vesicles (sEVs) are natural biocarriers for biomolecule transfer between cells and promising therapeutic strategies for bone defect repair. In this study, human periodontal ligament stem cell (PDLSC)-derived sEVs (P-EVs) were immobilized in Matrigel to establish a topical cell-free transplantation strategy for bone repair. Methods PDLSCs were cultured and P-EVs were isolated from the culture supernatant. In a rat bilateral calvarial defect model, P-EV/Matrigel was plugged into one defect and PBS/Matrigel was applied to the other. Bone repair in vivo was assessed by micro-computed tomography, histomorphometry, and immunohistochemical staining. In vitro, we investigated the effects of P-EVs on the proliferation and migration capabilities of bone marrow mesenchymal stem cells (BMMSCs) and explored the potential mechanism of action. Results The in vivo study showed that P-EV/Matrigel accelerated bone tissue repair by increasing cell infiltration when compared with the control. In vitro, P-EVs enhanced proliferation and migration of BMMSCs via increased phosphorylation of AKT and extracellular signal-regulated kinase 1/2 (ERK1/2). The role of P-EV-induced adenosine receptor signaling in AKT and ERK1/2 phosphorylation was a key mediator during enhanced BMMSC migration. Conclusion These results are the first to demonstrate that P-EVs accelerated the repair of bone defects, partially through promoting cell proliferation and migration. P-EV/Matrigel, which combines topical EV-implantation and extracellular matrix scaffolds, provides a new cell-free strategy for bone tissue repair.
Collapse
Affiliation(s)
- Bingjiao Zhao
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200001, People’s Republic of China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, People’s Republic of China
| | - Qingqing Chen
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, People’s Republic of China
| | - Liru Zhao
- Department of Orthodontics, School of Stomatology, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Jiaqi Mao
- Department of Orthodontics, School of Stomatology, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Wei Huang
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, People’s Republic of China
| | - Xinxin Han
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, People’s Republic of China
| | - Yuehua Liu
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200001, People’s Republic of China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, People’s Republic of China
- Correspondence: Yuehua Liu, Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, 356 East Beijing Road, Shanghai, 200001, People’s Republic of China, Tel +86-63298475, Fax +86-63614515, Email
| |
Collapse
|
32
|
Hong P, Yu M, Tian W. Diverse RNAs in adipose-derived extracellular vesicles and their therapeutic potential. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:665-677. [PMID: 34703651 PMCID: PMC8516999 DOI: 10.1016/j.omtn.2021.08.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Adipose tissue, which is considered an energy storage and active endocrine organ, produces and secretes a large amount of adipokines to regulate distant targets through blood circulation, especially extracellular vesicles (EVs). As cell-derived, membranous nanoparticles, EVs have recently garnered great attention as novel mediators in establishing intercellular communications as well as in accelerating interorgan crosstalk. Studies have revealed that the RNAs, including coding RNAs (messenger RNAs) and noncoding RNAs (long noncoding RNAs, microRNAs, and circular RNAs) are key bioactive cargoes of EV functions in various pathophysiological processes, such as cell differentiation, metabolic homeostasis, immune signal transduction, and cancer. Moreover, certain EV-contained RNAs have gradually been recognized as novel biomarkers, prognostic indicators, or even therapeutic nanodrugs of diseases. Therefore, in this review, we comprehensively summarize different classes of RNAs presented in adipose-derived EVs and discuss their therapeutic potential according to the latest research progress to provide valuable knowledge in this area.
Collapse
Affiliation(s)
- Pengyu Hong
- State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Diseases West China School of Stomatology, Sichuan University, Chengdu, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, China.,Engineering Research Center of Oral Translational Medicine, Ministry of Education, Sichuan University, Chengdu, China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Mei Yu
- State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Diseases West China School of Stomatology, Sichuan University, Chengdu, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, China.,Engineering Research Center of Oral Translational Medicine, Ministry of Education, Sichuan University, Chengdu, China
| | - Weidong Tian
- State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Diseases West China School of Stomatology, Sichuan University, Chengdu, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, China.,Engineering Research Center of Oral Translational Medicine, Ministry of Education, Sichuan University, Chengdu, China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
33
|
Hsu GCY, Cherief M, Sono T, Wang Y, Negri S, Xu J, Peault B, James AW. Divergent effects of distinct perivascular cell subsets for intra-articular cell therapy in posttraumatic osteoarthritis. J Orthop Res 2021; 39:2388-2397. [PMID: 33512030 PMCID: PMC8319216 DOI: 10.1002/jor.24997] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/30/2020] [Accepted: 01/24/2021] [Indexed: 02/04/2023]
Abstract
Intra-articular injection of mesenchymal stem cells has shown benefit for the treatment of osteoarthritis (OA). However, mesenchymal stem/stromal cells at the origin of these clinical results are heterogenous cell populations with limited cellular characterization. Here, two transgenic reporter mice were used to examine the differential effects of two precisely defined perivascular cell populations (Pdgfrα+ and Pdgfrβ+ cells) from white adipose tissue for alleviation of OA. Perivascular mesenchymal cells were isolated from transgenic Pdgfrα-and Pdgfrβ-CreERT2 reporter animals and delivered as a one-time intra-articular dose to C57BL/6J mice after destabilization of the medial meniscus (DMM). Both Pdgfrα+ and Pdgfrβ+ cell preparations improved metrics of cartilage degradation and reduced markers of chondrocyte hypertrophy. While some similarities in cell distribution were identified within the synovial and perivascular spaces, injected Pdgfrα+ cells remained in the superficial layers of articular cartilage, while Pdgfrβ+ cells were more widely dispersed. Pdgfrβ+ cell therapy prevented subchondral sclerosis induced by DMM, while Pdgfrα+ cell therapy had no effect. In summary, while both cell therapies showed beneficial effects in the DMM model, important differences in cell incorporation, persistence, and subchondral sclerosis were identified.
Collapse
Affiliation(s)
- Ginny Ching-Yun Hsu
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Masnsen Cherief
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Takashi Sono
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States;,Department of Orthopedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yiyun Wang
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Stefano Negri
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Jiajia Xu
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Bruno Peault
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, 90095;,Center For Cardiovascular Science and Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Aaron W. James
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| |
Collapse
|
34
|
Guo Y, Wu D, Zhang X, Zhang K, Luo Y. Biomolecules in cell-derived extracellular vesicle chariots as warriors to repair damaged tissues. NANOSCALE 2021; 13:16017-16033. [PMID: 34570853 DOI: 10.1039/d1nr04999b] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
In this review, we highlight the innovative applications of biomolecules from parent cell-derived extracellular vesicles (EVs) for tissue repair that have been developed in recent years. We evaluate the underlying mechanisms and therapeutic efficacy of each therapy. In previous literature reviews, it was most common to classify the use of EVs in tissue repair by disease type. This article reviews the role of three biomolecules in EVs in tissue repair. This review first summarizes the definitions and classifications of EVs. Then, the importance and significance of treating tissue damage with EVs are discussed. In particular, EV contents for tissue repair are three main types of biomolecules: proteins, RNAs and cell growth factors. The therapeutic and repair mechanisms of the biomolecules are discussed respectively. Finally, the development prospect and potential challenges of EV contents from highly differentiated cells as specific agents for tissue repair are summarized. When EVs are used to treat diseases such as tissue or organ damage, EVs play a role in delivery, and the real repair effect is effected by the various biomolecules carried by EVs. We believe that EV biomolecules have unparalleled advantages and clinical transformation potential for tissue repair and expect this review to inspire more intensive research work in this field.
Collapse
Affiliation(s)
- Yingshu Guo
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
| | - Di Wu
- School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Xu Zhang
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
| | - Kaixiang Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Yang Luo
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing 400044, P.R. China.
| |
Collapse
|
35
|
Kirkham AM, Bailey AJM, Tieu A, Maganti HB, Montroy J, Shorr R, Campbell TM, Fergusson DA, Lalu MM, Elmoazzen H, Allan DS. MSC-Derived Extracellular Vesicles in Preclinical Animal Models of Bone Injury: A Systematic Review and Meta-Analysis. Stem Cell Rev Rep 2021; 18:1054-1066. [PMID: 34313927 DOI: 10.1007/s12015-021-10208-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND OBJECTIVE Mesenchymal stromal cell-derived extracellular vesicles (MSC-EVs) are a promising treatment for bone injuries, although studies remain preclinical. A systematic review and meta-analysis can assess the efficacy of MSC-EVs and identify treatment aspects associated with enhanced bone repair. METHODS English language, preclinical, controlled, in vivo studies identified in our systematic search (up to May 8, 2020) examining the use of MSC-EVs in bone healing were included. Risk of bias (ROB) was assessed using the SYRCLE tool. Aggregate Data Meta-Analysis was performed to determine the effect of MSC-EVs on Bone Volume/Total Volume (BV/TV) and New Bone Formation (NBF). RESULTS Thirteen studies were included. Twelve reported either BV/TV or NBF and were included in meta-analysis. ROB was unclear in all studies. Overall, MSC-EVs displayed benefit in terms of bone healing for both BV/TV (22.2% mean difference (MD); 95% CI: 15.8-28.5%, p < 0.001) and NBF (26.1% MD; 10.3-41.8%, p = 0.001) versus controls. Substantial heterogeneity, however, was observed between studies. MSC-EVs were reported to activate multiple signaling pathways including mTOR/AKT, AMPK and BMP2. Subgroup analysis indicated no significant difference in the improvement of BV/TV when using modified EVs isolated after gene transfection, preconditioning (p = 0.61), or using EVs in combination with a tissue scaffold and/or hydrogel versus other delivery methods (p = 0.20). CONCLUSION Use of MSC-EVs to promote healing of bone injury appears promising, however, heterogeneity between studies and the potential for reporting bias limits confidence in the extent of benefit. Reducing bias between studies and addressing aspects of potential reporting bias should augment confidence in future meta-analyses and propel the field towards clinical studies. Forest Plot analysis assessing the percentage change in bone volume (BV) / total volume (TV) in the presence (experimental) or absence (control) of MSC-EVs.
Collapse
Affiliation(s)
- Aidan M Kirkham
- Stem Cells, Canadian Blood Services, Ottawa, ON, Canada.,Clinical Epidemiology, Ottawa Hospital Research Institute, 501 Smyth Rd, Box 704, Ottawa, ON, K1H 8L6, Canada
| | - Adrian J M Bailey
- Stem Cells, Canadian Blood Services, Ottawa, ON, Canada.,Clinical Epidemiology, Ottawa Hospital Research Institute, 501 Smyth Rd, Box 704, Ottawa, ON, K1H 8L6, Canada
| | - Alvin Tieu
- Clinical Epidemiology, Ottawa Hospital Research Institute, 501 Smyth Rd, Box 704, Ottawa, ON, K1H 8L6, Canada.,Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Medicine, The Ottawa Hospital, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Harinad B Maganti
- Stem Cells, Canadian Blood Services, Ottawa, ON, Canada.,Clinical Epidemiology, Ottawa Hospital Research Institute, 501 Smyth Rd, Box 704, Ottawa, ON, K1H 8L6, Canada
| | - Joshua Montroy
- Clinical Epidemiology, Ottawa Hospital Research Institute, 501 Smyth Rd, Box 704, Ottawa, ON, K1H 8L6, Canada
| | - Risa Shorr
- Medical Information and Education Services, The Ottawa Hospital, Ottawa, ON, Canada
| | - T Mark Campbell
- Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Medicine, The Ottawa Hospital, Ottawa, ON, Canada.,Department of Physical Medicine and Rehabilitation, Elisabeth Bruyère Hospital, Ottawa, ON, Canada
| | - Dean A Fergusson
- Clinical Epidemiology, Ottawa Hospital Research Institute, 501 Smyth Rd, Box 704, Ottawa, ON, K1H 8L6, Canada.,Medicine, The Ottawa Hospital, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Manoj M Lalu
- Clinical Epidemiology, Ottawa Hospital Research Institute, 501 Smyth Rd, Box 704, Ottawa, ON, K1H 8L6, Canada.,Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Departments of Anesthesiology and Pain Medicine, The Ottawa Hospital, Ottawa, ON, Canada.,School of Public Health and Epidemiology, University of Ottawa, Ottawa, ON, Canada
| | | | - David S Allan
- Stem Cells, Canadian Blood Services, Ottawa, ON, Canada. .,Clinical Epidemiology, Ottawa Hospital Research Institute, 501 Smyth Rd, Box 704, Ottawa, ON, K1H 8L6, Canada. .,Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, ON, Canada. .,Medicine, The Ottawa Hospital, Ottawa, ON, Canada. .,Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
36
|
Xu J, Wang Y, Gomez-Salazar MA, Hsu GCY, Negri S, Li Z, Hardy W, Ding L, Peault B, James AW. Bone-forming perivascular cells: Cellular heterogeneity and use for tissue repair. STEM CELLS (DAYTON, OHIO) 2021; 39:1427-1434. [PMID: 34252260 DOI: 10.1002/stem.3436] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/31/2021] [Accepted: 06/25/2021] [Indexed: 11/11/2022]
Abstract
Mesenchymal progenitor cells are broadly distributed across perivascular niches-an observation conserved between species. One common histologic zone with a high frequency of mesenchymal progenitor cells within mammalian tissues is the tunica adventitia, the outer layer of blood vessel walls populated by cells with a fibroblastic morphology. The diversity and functions of (re)generative cells present in this outermost perivascular niche are under intense investigation; we have reviewed herein our current knowledge of adventitial cell potential with a somewhat narrow focus on bone formation. Antigens of interest to functionally segregate adventicytes are discussed, including CD10, CD107a, aldehyde dehydrogenase isoforms, and CD140a among others. Purified adventicytes (such as CD10+ , CD107alow , and CD140a+ cells) have stronger osteogenic potential and promote bone formation in vivo. Recent bone tissue engineering applications of adventitial cells are also presented. A better understanding of perivascular progenitor cell subsets may represent a beneficial advance for future efforts in tissue repair and bioengineering.
Collapse
Affiliation(s)
- Jiajia Xu
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Yiyun Wang
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | | - Stefano Negri
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Zhao Li
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Winters Hardy
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, California, USA
| | - Lijun Ding
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Drum Tower Hospital of Nanjing University Medical School, Nanjing, People's Republic of China.,Clinical Center for Stem Cell Research, Drum Tower Hospital of Nanjing University Medical School, Nanjing, People's Republic of China.,Center For Cardiovascular Sciences, University of Edinburgh, Edinburgh, UK
| | - Bruno Peault
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, California, USA.,Center For Cardiovascular Sciences, University of Edinburgh, Edinburgh, UK
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
37
|
Man K, Brunet MY, Fernandez‐Rhodes M, Williams S, Heaney LM, Gethings LA, Federici A, Davies OG, Hoey D, Cox SC. Epigenetic reprogramming enhances the therapeutic efficacy of osteoblast-derived extracellular vesicles to promote human bone marrow stem cell osteogenic differentiation. J Extracell Vesicles 2021; 10:e12118. [PMID: 34262674 PMCID: PMC8263905 DOI: 10.1002/jev2.12118] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 05/18/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) are emerging in tissue engineering as promising acellular tools, circumventing many of the limitations associated with cell-based therapies. Epigenetic regulation through histone deacetylase (HDAC) inhibition has been shown to increase differentiation capacity. Therefore, this study aimed to investigate the potential of augmenting osteoblast epigenetic functionality using the HDAC inhibitor Trichostatin A (TSA) to enhance the therapeutic efficacy of osteoblast-derived EVs for bone regeneration. TSA was found to substantially alter osteoblast epigenetic function through reduced HDAC activity and increased histone acetylation. Treatment with TSA also significantly enhanced osteoblast alkaline phosphatase activity (1.35-fold), collagen production (2.8-fold) and calcium deposition (1.55-fold) during osteogenic culture (P ≤ 0.001). EVs derived from TSA-treated osteoblasts (TSA-EVs) exhibited reduced particle size (1-05-fold) (P > 0.05), concentration (1.4-fold) (P > 0.05) and protein content (1.16-fold) (P ≤ 0.001) when compared to untreated EVs. TSA-EVs significantly enhanced the proliferation (1.13-fold) and migration (1.3-fold) of human bone marrow stem cells (hBMSCs) when compared to untreated EVs (P ≤ 0.05). Moreover, TSA-EVs upregulated hBMSCs osteoblast-related gene and protein expression (ALP, Col1a, BSP1 and OCN) when compared to cells cultured with untreated EVs. Importantly, TSA-EVs elicited a time-dose dependent increase in hBMSCs extracellular matrix mineralisation. MicroRNA profiling revealed a set of differentially expressed microRNAs from TSA-EVs, which were osteogenic-related. Target prediction demonstrated these microRNAs were involved in regulating pathways such as 'endocytosis' and 'Wnt signalling pathway'. Moreover, proteomics analysis identified the enrichment of proteins involved in transcriptional regulation within TSA-EVs. Taken together, our findings suggest that altering osteoblasts' epigenome accelerates their mineralisation and promotes the osteoinductive potency of secreted EVs partly due to the delivery of pro-osteogenic microRNAs and transcriptional regulating proteins. As such, for the first time we demonstrate the potential to harness epigenetic regulation as a novel engineering approach to enhance EVs therapeutic efficacy for bone repair.
Collapse
Affiliation(s)
- Kenny Man
- School of Chemical EngineeringUniversity of BirminghamBirminghamUK
| | | | | | - Soraya Williams
- School of Sport, Exercise and Health SciencesLoughborough UniversityLoughboroughUK
| | - Liam M. Heaney
- School of Sport, Exercise and Health SciencesLoughborough UniversityLoughboroughUK
| | - Lee A. Gethings
- Waters CorporationStamford AvenueWilmslowUK
- Division of Infection, Immunity and Respiratory MedicineFaculty of Biology, Medicine and HealthManchester Institute of BiotechnologyUniversity of ManchesterManchesterUK
| | - Angelica Federici
- Trinity Biomedical Sciences InstituteTrinity CollegeTrinity Centre for Biomedical EngineeringDublinIreland
- Department of Mechanical, Manufacturing, and Biomedical EngineeringSchool of EngineeringTrinity College DublinIreland
- Trinity College Dublin & RCSIAdvanced Materials and Bioengineering Research CentreDublinIreland
| | - Owen G. Davies
- School of Sport, Exercise and Health SciencesLoughborough UniversityLoughboroughUK
| | - David Hoey
- Trinity Biomedical Sciences InstituteTrinity CollegeTrinity Centre for Biomedical EngineeringDublinIreland
- Department of Mechanical, Manufacturing, and Biomedical EngineeringSchool of EngineeringTrinity College DublinIreland
- Trinity College Dublin & RCSIAdvanced Materials and Bioengineering Research CentreDublinIreland
| | - Sophie C. Cox
- School of Chemical EngineeringUniversity of BirminghamBirminghamUK
| |
Collapse
|
38
|
Zhu X, Yu F, Yan G, Hu Y, Sun H, Ding L. Human endometrial perivascular stem cells exhibit a limited potential to regenerate endometrium after xenotransplantation. Hum Reprod 2021; 36:145-159. [PMID: 33283858 DOI: 10.1093/humrep/deaa261] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 09/02/2020] [Indexed: 12/15/2022] Open
Abstract
STUDY QUESTION What are the localization, characteristics and potential for tissue regeneration of two perivascular stem cells, namely CD34+ adventitial cells and CD146+ pericytes, in human endometrium? SUMMARY ANSWER Human endometrial CD34+ adventitial cells (located in the outermost layer of blood vessels and mainly in the basal layer) and CD146+ pericytes showed mesenchymal stem cell (MSC) phenotypes in in vitro culture, but presented limited potential to regenerate endometrium. WHAT IS KNOWN ALREADY Periodic endometrial regeneration is considered to be maintained by MSCs. Blood vessel wall, regarded as stem cell niche, harbors a large reserve of progenitor cells that may be integral to the origin of MSCs. However, a lack of validated markers has hampered the isolation of putative endometrial MSCs. Currently, CD146+ pericytes and Sushi Domain Containing 2 (SUSD2) positive cells have been identified in the endometrial perivascular region as sharing MSCs characteristics. STUDY DESIGN, SIZE, DURATION The locations of adventitial cells and pericytes in the human endometrium were identified by immunofluorescence staining (n = 4). After CD34+CD146-CD45-CD56-CD144- adventitial cells and CD146+CD34-CD45-CD56-CD144- pericytes were isolated from the endometrium of normal women (n = 6) by fluorescence-activated cell sorting, their characteristics were investigated in culture. Adventitial cells and pericytes were induced to differentiate, respectively, into vascular endothelial-like cells or endometrial stromal-like cells in vitro, with their potential explored by in vivo xenotransplantation (n = 2 in each group) and eutopic transplantation (n = 2 in each group). PARTICIPANTS/MATERIALS, SETTING, METHODS CD34+ adventitial cells and CD146+ pericytes were cultured in the inducing medium to differentiate into endothelial-like cells in vitro, and then analyzed for CD31, von Willebrand factor immunofluorescent staining and tube formation. They were also cultured to differentiate into endometrial stromal cells in vitro, with the expression of vimentin and CD13 being detected by western blot before and after induction, and the expression of prolactin and insulin-like growth factor-binding protein 1 being determined as well. Single dispersed CD34+ adventitial cells and CD146+ pericytes were respectively transplanted under the kidney capsule of NOG mice to investigate their differentiation potential in vivo. A eutopic transplantation model was constructed by grafting recellularized uterine matrix loaded up with CM-Dil labeled adventitial cells or pericytes into the injury region of nude rat's uterus. MAIN RESULTS AND THE ROLE OF CHANCE CD34+ adventitial cells were mainly located at the outmost layer of endometrial large vessels, while CD146+ pericytes were found surrounding the inner endothelial cells of microvessels. A small proportion of CD34+ adventitial cells expressed SUSD2. The number of adventitial cells was ∼40 times higher than that of pericytes in the endometrium. Both adventitial cells and pericytes showed MSC phenotypes after in vitro culture. After in vitro induction into endometrial endothelial-like cells and stromal-like cells, adventitial cells showed higher plasticity than pericytes and a closer correlation with stromal-like cells. In the mouse xenotransplantation model, vimentin+ cells, CD31+ endothelial-like cells and CD146+ pericyte-like cells could be observed after adventitial cells were transplanted. CM-Dil-labeled adventitial cells or pericytes could survive in the immunocompromised nude rats after eutopic transplantation, and vimentin+ cells were detected. In addition, CM-Dil-labeled adventitial cells or pericytes did not express α-smooth muscle actin or E-cadherin after transplantation. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION CD34 was chosen as a novel marker to isolate adventitial cells from human endometrium according to previous literature. The association of endometrial CD34+ adventitial cells and SUSD2+ MSCs should be further investigated. WIDER IMPLICATIONS OF THE FINDINGS The decellularized uterine matrix model might be useful in endometrial stem cell therapy. STUDY FUNDING/COMPETING INTEREST(S) L.D. is supported by grants from National Key Research and Development Program of China (2018YFC1004700), Nature Science Foundation of China (81871128, 81571391) and Nanjing Medical Science Development Project (ZKX16042). H.S. is supported by a grant from Jiangsu Province Social Development Project (BE2018602). X.Z. was supported by grants from the Postgraduate Innovative Project of Jiangsu Province (KYCX19-1177). The authors declare no conflict of interest.
Collapse
Affiliation(s)
- Xinxin Zhu
- Center for Reproductive Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fei Yu
- Center for Experimental Animal, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Guijun Yan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yali Hu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Haixiang Sun
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Lijun Ding
- Center for Reproductive Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Clinical Center for Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
39
|
A Neurotrophic Mechanism Directs Sensory Nerve Transit in Cranial Bone. Cell Rep 2021; 31:107696. [PMID: 32460020 PMCID: PMC7335423 DOI: 10.1016/j.celrep.2020.107696] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 03/17/2020] [Accepted: 05/06/2020] [Indexed: 11/21/2022] Open
Abstract
The flat bones of the skull are densely innervated during development, but little is known regarding their role during repair. We describe a neurotrophic mechanism that directs sensory nerve transit in the mouse calvaria. Patent cranial suture mesenchyme represents an NGF (nerve growth factor)-rich domain, in which sensory nerves transit. Experimental calvarial injury upregulates Ngf in an IL-1β/TNF-α-rich defect niche, with consequent axonal ingrowth. In calvarial osteoblasts, IL-1β and TNF-α stimulate Ngf and downstream NF-κB signaling. Locoregional deletion of Ngf delays defect site re-innervation and blunted repair. Genetic disruption of Ngf among LysM-expressing macrophages phenocopies these observations, whereas conditional knockout of Ngf among Pdgfra-expressing cells does not. Finally, inhibition of TrkA catalytic activity similarly delays re-innervation and repair. These results demonstrate an essential role of NGF-TrkA signaling in bone healing and implicate macrophage-derived NGF-induced ingrowth of skeletal sensory nerves as an important mediator of this repair.
Collapse
|
40
|
Murali VP, Holmes CA. Mesenchymal stromal cell-derived extracellular vesicles for bone regeneration therapy. Bone Rep 2021; 14:101093. [PMID: 34095360 PMCID: PMC8166743 DOI: 10.1016/j.bonr.2021.101093] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 12/24/2022] Open
Abstract
Purpose To analyze preclinical bone regeneration studies employing mesenchymal stromal cell (MSC)- derived extracellular vesicles (EVs) and highlight any commonalities in EV biomarker expression, miRNA cargo(s) or pathway activation that will aid in understanding the underlying therapeutic mechanisms. Methods Articles employing EVs derived from either MSCs or MSC-like osteogenic stromal cells in preclinical bone regeneration studies are included in this review. Results EVs derived from a variety of MSC types were able to successfully induce bone formation in preclinical models. Many studies failed to perform in-depth EV characterization. The studies with detailed EV characterization data report very different miRNA cargos, even in EVs isolated from the same species and cell types. Few preclinical studies have analyzed the underlying mechanisms of MSC-EV therapeutic action. Conclusion There is a critical need for mechanistic preclinical studies with thorough EV characterization to determine the best therapeutic MSC-EV source for bone regeneration therapies. Issues including controlled EV delivery, large scale production, and proper storage also need to be addressed before EV-based bone regeneration therapies can be translated for clinical bone repair. EVs from different MSC sources successfully regenerate bone in preclinical models. Studies were reviewed to find commonalities in EV cargo(s)/pathways activated in MSC-EV-based bone regeneration therapies. Issues that need to be overcome to enable clinical translation of EV-based therapies were addressed.
Collapse
Affiliation(s)
- Vishnu Priya Murali
- Department of Chemical and Biomedical Engineering, College of Engineering, Florida A&M University-Florida State University, 2525 Pottsdamer Street, Room A131, Tallahassee, FL 32310, USA
| | - Christina A Holmes
- Department of Chemical and Biomedical Engineering, College of Engineering, Florida A&M University-Florida State University, 2525 Pottsdamer Street, Room A131, Tallahassee, FL 32310, USA
| |
Collapse
|
41
|
Srivatsav AT, Kapoor S. The Emerging World of Membrane Vesicles: Functional Relevance, Theranostic Avenues and Tools for Investigating Membrane Function. Front Mol Biosci 2021; 8:640355. [PMID: 33968983 PMCID: PMC8101706 DOI: 10.3389/fmolb.2021.640355] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 01/26/2021] [Indexed: 12/12/2022] Open
Abstract
Lipids are essential components of cell membranes and govern various membrane functions. Lipid organization within membrane plane dictates recruitment of specific proteins and lipids into distinct nanoclusters that initiate cellular signaling while modulating protein and lipid functions. In addition, one of the most versatile function of lipids is the formation of diverse lipid membrane vesicles for regulating various cellular processes including intracellular trafficking of molecular cargo. In this review, we focus on the various kinds of membrane vesicles in eukaryotes and bacteria, their biogenesis, and their multifaceted functional roles in cellular communication, host-pathogen interactions and biotechnological applications. We elaborate on how their distinct lipid composition of membrane vesicles compared to parent cells enables early and non-invasive diagnosis of cancer and tuberculosis, while inspiring vaccine development and drug delivery platforms. Finally, we discuss the use of membrane vesicles as excellent tools for investigating membrane lateral organization and protein sorting, which is otherwise challenging but extremely crucial for normal cellular functioning. We present current limitations in this field and how the same could be addressed to propel a fundamental and technology-oriented future for extracellular membrane vesicles.
Collapse
Affiliation(s)
- Aswin T. Srivatsav
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, India
| | - Shobhna Kapoor
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, India
- Wadhwani Research Center of Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| |
Collapse
|
42
|
Chen W, Foo SS, Hong E, Wu C, Lee WS, Lee SA, Evseenko D, Lopes Moreira ME, García-Sastre A, Cheng G, Nielsen-Saines K, Brasil P, Avvad-Portari E, Jung JU. Zika virus NS3 protease induces bone morphogenetic protein-dependent brain calcification in human fetuses. Nat Microbiol 2021; 6:455-466. [PMID: 33510473 PMCID: PMC8012254 DOI: 10.1038/s41564-020-00850-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 12/11/2020] [Indexed: 01/29/2023]
Abstract
The most frequent fetal birth defect associated with prenatal Zika virus (ZIKV) infection is brain calcification, which in turn may potentially affect neurological development in infants. Understanding the mechanism could inform the development of potential therapies against prenatal ZIKV brain calcification. In perivascular cells, bone morphogenetic protein (BMP) is an osteogenic factor that undergoes maturation to activate osteogenesis and calcification. Here, we show that ZIKV infection of cultivated primary human brain pericytes triggers BMP2 maturation, leading to osteogenic gene expression and calcification. We observed extensive calcification near ZIKV+ pericytes of fetal human brain specimens and in vertically transmitted ZIKV+ human signal transducer and activator of transcription 2-knockin mouse pup brains. ZIKV infection of primary pericytes stimulated BMP2 maturation, inducing osteogenic gene expression and calcification that were completely blocked by anti-BMP2/4 neutralizing antibody. Not only did ZIKV NS3 expression alone induce BMP2 maturation, osteogenic gene expression and calcification, but purified NS3 protease also effectively cleaved pro-BMP2 in vitro to generate biologically active mature BMP2. These findings highlight ZIKV-induced calcification where the NS3 protease subverts the BMP2-mediated osteogenic signalling pathway to trigger brain calcification.
Collapse
Affiliation(s)
- Weiqiang Chen
- Department of Cancer Biology and Global Center for Pathogens Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Suan-Sin Foo
- Department of Cancer Biology and Global Center for Pathogens Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Eunjin Hong
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Christine Wu
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Wai-Suet Lee
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Shin-Ae Lee
- Department of Cancer Biology and Global Center for Pathogens Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Denis Evseenko
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Maria Elisabeth Lopes Moreira
- Clinical Research Unit, Fernandes Figueira Institute-FioCruz, Avenida Rui Barbosa, 716, Flamengo, Rio De Janeiro, RJ CEP 22250-020, Brazil
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA;,Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA;,Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA;,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Genhong Cheng
- Department of Microbiology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Marion Davies Children’s Health Center, 10833 LeConte Avenue, Los Angeles, CA 90095, USA
| | - Karin Nielsen-Saines
- Division of Pediatric Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Marion Davies Children’s Health Center, 10833 LeConte Avenue, Los Angeles, CA 90095, USA
| | - Patrícia Brasil
- Laboratório de Pesquisa Clínica em Doenças Febris Agudas, Instituto Nacional de Infectologia Evandro Chagas, FioCruz, 4365 Avenida Brasil, Rio de Janeiro – RJ, 21040-360, Brazil
| | - Elyzabeth Avvad-Portari
- Department of Pathology, Fernandes Figueira Institute-FioCruz, Avenida Rui Barbosa, 716, Flamengo, Rio De Janeiro, RJ CEP 22250-020, Brazil
| | - Jae U. Jung
- Department of Cancer Biology and Global Center for Pathogens Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA;,Correspondence: (Jae U. Jung, PhD)
| |
Collapse
|
43
|
Myronovych A, Peck BCE, An M, Zhu J, Warm A, Kupe A, Lubman DM, Seeley RJ. Intestinal extracellular vesicles are altered by vertical sleeve gastrectomy. Am J Physiol Gastrointest Liver Physiol 2021; 320:G153-G165. [PMID: 33175569 PMCID: PMC7864234 DOI: 10.1152/ajpgi.00224.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Bariatric surgery is the most effective treatment for obesity and its comorbidities. However, our understanding of the molecular mechanisms behind its beneficial effects is limited. Extracellular vesicles (EVs) comprise an important mode of intercellular communication. They carry nucleic acids, hormones, and signaling molecules and regulate multiple processes. Our aim was to test the role of EVs in the effects of vertical sleeve gastrectomy (VSG) using a mouse model. Small intestinal EVs were obtained from the mice that underwent VSG or control surgery and were on chow or high-fat diet or diet-restricted, and then they were subjected to the proteomic analysis. Enteroid and bacterial cultures were treated with EVs to evaluate their survival effect. A mouse cohort received intraduodenal administration of EVs from VSG or Sham mice for 10 days. Body weight, glucose metabolism, and intestinal morphology were evaluated. EVs were enriched in the intestinal lumen and mucus of VSG compared with Sham mice. Protein composition of VSG and Sham-derived EVs was highly distinct. When introduced into culture, VSG EVs decreased survival of intestinal enteroids and, conversely, promoted proliferation of bacteria. Mice administered with EVs obtained from VSG and Sham groups did not show differences in body weight, food intake, or glucose metabolism. Intestinal morphology was altered, as VSG EVs caused reduction of ileal villi length and decreased epithelial proliferation in the jejunum and ileum. VSG causes remodeling of intestinal EVs, which results in unique protein composition. VSG-derived EVs exhibit cytotoxic effects on epithelial cells and reduce proliferation of intestinal progenitor cells in mice.NEW & NOTEWORTHY This is the first study that investigates the impact of bariatric surgery on protein composition of intestinal extracellular vesicles. Extracellular vesicle composition is greatly altered after vertical sleeve gastrectomy and may potentially modulate various signaling pathways. In our study, extracellular vesicles from vertical sleeve gastrectomy-treated mice promote bacterial proliferation but exhibit cytotoxic effect on epithelial cells and reduce proliferation of intestinal progenitor cells in mice.
Collapse
Affiliation(s)
| | | | - Mingrui An
- 1Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Jianhui Zhu
- 1Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | | | - Aleksander Kupe
- 1Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - David M. Lubman
- 1Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Randy J. Seeley
- 1Department of Surgery, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
44
|
Marquez J, Dong J, Dong C, Tian C, Serrero G. Identification of Prostaglandin F2 Receptor Negative Regulator (PTGFRN) as an internalizable target in cancer cells for antibody-drug conjugate development. PLoS One 2021; 16:e0246197. [PMID: 33503070 PMCID: PMC7840024 DOI: 10.1371/journal.pone.0246197] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 01/14/2021] [Indexed: 11/19/2022] Open
Abstract
Antibody-drug conjugates (ADC) are effective antibody-based therapeutics for hematopoietic and lymphoid tumors. However, there is need to identify new targets for ADCs, particularly for solid tumors and cancers with unmet needs. From a hybridoma library developed against cancer cells, we selected the mouse monoclonal antibody 33B7, which was able to bind to, and internalize, cancer cell lines. This antibody was used for identification of the target by immunoprecipitation and mass spectrometric analysis, followed by target validation. After target validation, 33B7 binding and target positivity were tested by flow cytometry and western blot analysis in several cancer cell lines. The ability of 33B7 conjugated to saporin to inhibit in vitro proliferation of PTFRN positive cell lines was investigated, as well as the 33B7 ADC in vivo effect on tumor growth in athymic mice. All flow cytometry and in vitro internalization assays were analyzed for statistical significance using a Welsh's T-test. Animal studies were analyzed using Two-Way Analysis of Variance (ANOVA) utilizing post-hoc Bonferroni analysis, and/or Mixed Effects analysis. The 33B7 cell surface target was identified as Prostaglandin F2 Receptor Negative Regulator (PTGFRN), a transmembrane protein in the Tetraspanin family. This target was confirmed by showing that PTGFRN-expressing cells bound and internalized 33B7, compared to PTGFRN negative cells. Cells able to bind 33B7 were PTGFRN-positive by Western blot analysis. In vitro treatment PTGFRN-positive cancer cell lines with the 33B7-saporin ADC inhibited their proliferation in a dose-dependent fashion. 33B7 conjugated to saporin was also able to block tumor growth in vivo in mouse xenografts when compared to a control ADC. These findings show that screening antibody libraries for internalizing antibodies in cancer cell lines is a good approach to identify new cancer targets for ADC development. These results suggest PTGFRN is a possible therapeutic target via antibody-based approach for certain cancers.
Collapse
Affiliation(s)
- Jorge Marquez
- Target Discovery Division, A&G Pharmaceutical, Inc., Columbia, Maryland, United States of America
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore School of Pharmacy, Baltimore, Maryland, United States of America
| | - Jianping Dong
- Target Discovery Division, A&G Pharmaceutical, Inc., Columbia, Maryland, United States of America
| | - Chun Dong
- Precision Antibody Division, A&G Pharmaceutical, Inc., Columbia, Maryland, United States of America
| | - Changsheng Tian
- Target Discovery Division, A&G Pharmaceutical, Inc., Columbia, Maryland, United States of America
| | - Ginette Serrero
- Target Discovery Division, A&G Pharmaceutical, Inc., Columbia, Maryland, United States of America
- Precision Antibody Division, A&G Pharmaceutical, Inc., Columbia, Maryland, United States of America
| |
Collapse
|
45
|
Negri S, Wang Y, Sono T, Qin Q, Hsu GCY, Cherief M, Xu J, Lee S, Tower RJ, Yu V, Piplani A, Meyers CA, Broderick K, Lee M, James AW. Systemic DKK1 neutralization enhances human adipose-derived stem cell mediated bone repair. Stem Cells Transl Med 2020; 10:610-622. [PMID: 33377628 PMCID: PMC7980212 DOI: 10.1002/sctm.20-0293] [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: 07/02/2020] [Revised: 10/26/2020] [Accepted: 11/15/2020] [Indexed: 12/15/2022] Open
Abstract
Progenitor cells from adipose tissue are able to induce bone repair; however, inconsistent or unreliable efficacy has been reported across preclinical and clinical studies. Soluble inhibitory factors, such as the secreted Wnt signaling antagonists Dickkopf-1 (DKK1), are expressed to variable degrees in human adipose-derived stem cells (ASCs), and may represent a targetable "molecular brake" on ASC mediated bone repair. Here, anti-DKK1 neutralizing antibodies were observed to increase the osteogenic differentiation of human ASCs in vitro, accompanied by increased canonical Wnt signaling. Human ASCs were next engrafted into a femoral segmental bone defect in NOD-Scid mice, with animals subsequently treated with systemic anti-DKK1 or isotype control during the repair process. Human ASCs alone induced significant but modest bone repair. However, systemic anti-DKK1 induced an increase in human ASC engraftment and survival, an increase in vascular ingrowth, and ultimately improved bone repair outcomes. In summary, anti-DKK1 can be used as a method to augment cell-mediated bone regeneration, and could be particularly valuable in the contexts of impaired bone healing such as osteoporotic bone repair.
Collapse
Affiliation(s)
- Stefano Negri
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA.,Orthopaedic and Trauma Surgery Unit, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, Verona, Italy
| | - Yiyun Wang
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Takashi Sono
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Qizhi Qin
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Masnsen Cherief
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jiajia Xu
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Seungyong Lee
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Robert J Tower
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Victoria Yu
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Abhi Piplani
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Carolyn A Meyers
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Kristen Broderick
- Department of Plastic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Min Lee
- School of Dentistry, University of California Los Angeles, Los Angeles, California, USA
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
46
|
Frühbeis C, Kuo-Elsner WP, Müller C, Barth K, Peris L, Tenzer S, Möbius W, Werner HB, Nave KA, Fröhlich D, Krämer-Albers EM. Oligodendrocytes support axonal transport and maintenance via exosome secretion. PLoS Biol 2020; 18:e3000621. [PMID: 33351792 PMCID: PMC7787684 DOI: 10.1371/journal.pbio.3000621] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/06/2021] [Accepted: 12/10/2020] [Indexed: 12/28/2022] Open
Abstract
Neurons extend long axons that require maintenance and are susceptible to degeneration. Long-term integrity of axons depends on intrinsic mechanisms including axonal transport and extrinsic support from adjacent glial cells. The mechanisms of support provided by myelinating oligodendrocytes to underlying axons are only partly understood. Oligodendrocytes release extracellular vesicles (EVs) with properties of exosomes, which upon delivery to neurons improve neuronal viability in vitro. Here, we show that oligodendroglial exosome secretion is impaired in 2 mouse mutants exhibiting secondary axonal degeneration due to oligodendrocyte-specific gene defects. Wild-type oligodendroglial exosomes support neurons by improving the metabolic state and promoting axonal transport in nutrient-deprived neurons. Mutant oligodendrocytes release fewer exosomes, which share a common signature of underrepresented proteins. Notably, mutant exosomes lack the ability to support nutrient-deprived neurons and to promote axonal transport. Together, these findings indicate that glia-to-neuron exosome transfer promotes neuronal long-term maintenance by facilitating axonal transport, providing a novel mechanistic link between myelin diseases and secondary loss of axonal integrity. The long-term integrity of neuronal axons depends on intrinsic mechanisms such as axonal transport and on extrinsic support from adjacent glial cells. This study shows that genetic defects in glia that affect axonal integrity impair the secretion of oligodendrocyte exosomes and their ability to support nutrient-deprived neurons and promote axonal transport.
Collapse
Affiliation(s)
- Carsten Frühbeis
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Wen Ping Kuo-Elsner
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University of Mainz, Mainz, Germany
- Focus Program Translational Neuroscience, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Christina Müller
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Kerstin Barth
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Leticia Peris
- Grenoble Institut des Neurosciences, Université Grenoble Alpes, Inserm, U1216, Grenoble, France
| | - Stefan Tenzer
- Institute of Immunology, University Medical Center, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Hauke B. Werner
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Dominik Fröhlich
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University of Mainz, Mainz, Germany
- Translational Neuroscience Facility and Department of Physiology, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Eva-Maria Krämer-Albers
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University of Mainz, Mainz, Germany
- Focus Program Translational Neuroscience, Johannes Gutenberg University of Mainz, Mainz, Germany
- * E-mail:
| |
Collapse
|
47
|
Leucht P, Einhorn TA. What's New in Musculoskeletal Basic Science. J Bone Joint Surg Am 2020; 102:2017-2021. [PMID: 33079894 DOI: 10.2106/jbjs.20.01701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Philipp Leucht
- Departments of Orthopaedic Surgery (P.L. and T.A.E.) and Cell Biology (P.L.), NYU Grossman School of Medicine, New York, NY
| | | |
Collapse
|
48
|
Negri S, Wang Y, Sono T, Lee S, Hsu GC, Xu J, Meyers CA, Qin Q, Broderick K, Witwer KW, Peault B, James AW. Human perivascular stem cells prevent bone graft resorption in osteoporotic contexts by inhibiting osteoclast formation. Stem Cells Transl Med 2020; 9:1617-1630. [PMID: 32697440 PMCID: PMC7695633 DOI: 10.1002/sctm.20-0152] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/24/2020] [Accepted: 06/15/2020] [Indexed: 12/15/2022] Open
Abstract
The vascular wall stores mesenchymal progenitor cells which are able to induce bone regeneration, via direct and paracrine mechanisms. Although much is known regarding perivascular cell regulation of osteoblasts, their regulation of osteoclasts, and by extension utility in states of high bone resorption, is not known. Here, human perivascular stem cells (PSCs) were used as a means to prevent autograft resorption in a gonadectomy-induced osteoporotic spine fusion model. Furthermore, the paracrine regulation by PSCs of osteoclast formation was evaluated, using coculture, conditioned medium, and purified extracellular vesicles. Results showed that PSCs when mixed with autograft bone induce an increase in osteoblast:osteoclast ratio, promote bone matrix formation, and prevent bone graft resorption. The confluence of these factors resulted in high rates of fusion in an ovariectomized rat lumbar spine fusion model. Application of PSCs was superior across metrics to either the use of unpurified, culture-defined adipose-derived stromal cells or autograft bone alone. Under coculture conditions, PSCs negatively regulated osteoclast formation and did so via secreted, nonvesicular paracrine factors. Total RNA sequencing identified secreted factors overexpressed by PSCs which may explain their negative regulation of graft resorption. In summary, PSCs reduce osteoclast formation and prevent bone graft resorption in high turnover states such as gonadectomy-induced osteoporosis.
Collapse
Affiliation(s)
- Stefano Negri
- Department of PathologyJohns Hopkins UniversityBaltimoreMarylandUSA
- Orthopaedic and Trauma Surgery Unit, Department of Surgery, DentistryPaediatrics and Gynaecology of the University of VeronaVeronaItaly
| | - Yiyun Wang
- Department of PathologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Takashi Sono
- Department of PathologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Seungyong Lee
- Department of PathologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | | | - Jiajia Xu
- Department of PathologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | | | - Qizhi Qin
- Department of PathologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Kristen Broderick
- Department of Plastic SurgeryJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Kenneth W. Witwer
- Departments of Molecular and Comparative Pathobiology and NeurologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Bruno Peault
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research CenterLos AngelesCaliforniaUSA
- Center for Cardiovascular Science and MRC Center for Regenerative MedicineUniversity of EdinburghEdinburghUK
| | - Aaron W. James
- Department of PathologyJohns Hopkins UniversityBaltimoreMarylandUSA
| |
Collapse
|
49
|
Recent Advances in Understandings Towards Pathogenesis and Treatment for Intrauterine Adhesion and Disruptive Insights from Single-Cell Analysis. Reprod Sci 2020; 28:1812-1826. [PMID: 33125685 PMCID: PMC8189970 DOI: 10.1007/s43032-020-00343-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/01/2020] [Indexed: 12/22/2022]
Abstract
Intrauterine adhesion is a major cause of menstrual irregularities, infertility, and recurrent pregnancy losses and the progress towards its amelioration and therapy is slow and unsatisfactory. We aim to summarize and evaluate the current treatment progress and research methods for intrauterine adhesion. We conducted literature review in January 2020 by searching articles at PubMed on prevention and treatment, pathogenesis, the repair of other tissues/organs, cell plasticity, and the stem cell–related therapies for intrauterine adhesion. A total of 110 articles were selected for review. Uterine cell heterogeneity, expression profile, and cell-cell interaction were investigated based on scRNA-seq of uterus provided by Human Cell Landscape (HCL) project. Previous knowledge on intrauterine adhesion (IUA) pathogenesis was mostly derived from correlation studies by differentially expressed genes between endometrial tissue of intrauterine adhesion patients/animal models and normal endometrial tissue. Although the TGF-β1/SMAD pathway was suggested as the key driver for IUA pathogenesis, uterine cell heterogeneity and distinct expression profile among different cell types highlighted the importance of single-cell investigations. Cell-cell interaction in the uterus revealed the central hub of endothelial cells interacting with other cells, with endothelial cells in endothelial to mesenchymal transition and fibroblasts as the strongest interaction partners. The potential of stem cell–related therapies appeared promising, yet suffers from largely animal studies and nonstandard study design. The need to dissect the roles of endometrial cells, endothelial cells, and fibroblasts and their interaction is evident in order to elucidate the molecular and cellular mechanisms in both intrauterine adhesion pathogenesis and treatment.
Collapse
|
50
|
Xu J, Wang Y, Hsu CY, Negri S, Tower RJ, Gao Y, Tian Y, Sono T, Meyers CA, Hardy WR, Chang L, Hu S, Kahn N, Broderick K, Péault B, James AW. Lysosomal protein surface expression discriminates fat- from bone-forming human mesenchymal precursor cells. eLife 2020; 9:e58990. [PMID: 33044169 PMCID: PMC7550188 DOI: 10.7554/elife.58990] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/25/2020] [Indexed: 12/25/2022] Open
Abstract
Tissue resident mesenchymal stem/stromal cells (MSCs) occupy perivascular spaces. Profiling human adipose perivascular mesenchyme with antibody arrays identified 16 novel surface antigens, including endolysosomal protein CD107a. Surface CD107a expression segregates MSCs into functionally distinct subsets. In culture, CD107alow cells demonstrate high colony formation, osteoprogenitor cell frequency, and osteogenic potential. Conversely, CD107ahigh cells include almost exclusively adipocyte progenitor cells. Accordingly, human CD107alow cells drove dramatic bone formation after intramuscular transplantation in mice, and induced spine fusion in rats, whereas CD107ahigh cells did not. CD107a protein trafficking to the cell surface is associated with exocytosis during early adipogenic differentiation. RNA sequencing also suggested that CD107alow cells are precursors of CD107ahigh cells. These results document the molecular and functional diversity of perivascular regenerative cells, and show that relocation to cell surface of a lysosomal protein marks the transition from osteo- to adipogenic potential in native human MSCs, a population of substantial therapeutic interest.
Collapse
Affiliation(s)
- Jiajia Xu
- Departments of Pathology, Johns Hopkins UniversityBaltimoreUnited States
| | - Yiyun Wang
- Departments of Pathology, Johns Hopkins UniversityBaltimoreUnited States
| | - Ching-Yun Hsu
- Departments of Pathology, Johns Hopkins UniversityBaltimoreUnited States
| | - Stefano Negri
- Departments of Pathology, Johns Hopkins UniversityBaltimoreUnited States
| | - Robert J Tower
- Departments of Pathology, Johns Hopkins UniversityBaltimoreUnited States
- Departments of Orthopaedics, Johns Hopkins UniversityBaltimoreUnited States
| | - Yongxing Gao
- Departments of Pathology, Johns Hopkins UniversityBaltimoreUnited States
| | - Ye Tian
- Departments of Pathology, Johns Hopkins UniversityBaltimoreUnited States
- Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical UniversityShenyangChina
| | - Takashi Sono
- Departments of Pathology, Johns Hopkins UniversityBaltimoreUnited States
| | - Carolyn A Meyers
- Departments of Pathology, Johns Hopkins UniversityBaltimoreUnited States
| | - Winters R Hardy
- Departments of Pathology, Johns Hopkins UniversityBaltimoreUnited States
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research CenterLos AngelesUnited States
| | - Leslie Chang
- Departments of Pathology, Johns Hopkins UniversityBaltimoreUnited States
| | - Shuaishuai Hu
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research CenterLos AngelesUnited States
| | - Nusrat Kahn
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research CenterLos AngelesUnited States
| | - Kristen Broderick
- Departments of Plastic Surgery, Johns Hopkins UniversityBaltimoreUnited States
| | - Bruno Péault
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research CenterLos AngelesUnited States
- Center For Cardiovascular Science and Center for Regenerative Medicine, University of EdinburghEdinburghUnited Kingdom
| | - Aaron W James
- Departments of Pathology, Johns Hopkins UniversityBaltimoreUnited States
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research CenterLos AngelesUnited States
| |
Collapse
|