101
|
Gantenbein B, Tang S, Guerrero J, Higuita-Castro N, Salazar-Puerta AI, Croft AS, Gazdhar A, Purmessur D. Non-viral Gene Delivery Methods for Bone and Joints. Front Bioeng Biotechnol 2020; 8:598466. [PMID: 33330428 PMCID: PMC7711090 DOI: 10.3389/fbioe.2020.598466] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/26/2020] [Indexed: 12/12/2022] Open
Abstract
Viral carrier transport efficiency of gene delivery is high, depending on the type of vector. However, viral delivery poses significant safety concerns such as inefficient/unpredictable reprogramming outcomes, genomic integration, as well as unwarranted immune responses and toxicity. Thus, non-viral gene delivery methods are more feasible for translation as these allow safer delivery of genes and can modulate gene expression transiently both in vivo, ex vivo, and in vitro. Based on current studies, the efficiency of these technologies appears to be more limited, but they are appealing for clinical translation. This review presents a summary of recent advancements in orthopedics, where primarily bone and joints from the musculoskeletal apparatus were targeted. In connective tissues, which are known to have a poor healing capacity, and have a relatively low cell-density, i.e., articular cartilage, bone, and the intervertebral disk (IVD) several approaches have recently been undertaken. We provide a brief overview of the existing technologies, using nano-spheres/engineered vesicles, lipofection, and in vivo electroporation. Here, delivery for microRNA (miRNA), and silencing RNA (siRNA) and DNA plasmids will be discussed. Recent studies will be summarized that aimed to improve regeneration of these tissues, involving the delivery of bone morphogenic proteins (BMPs), such as BMP2 for improvement of bone healing. For articular cartilage/osteochondral junction, non-viral methods concentrate on targeted delivery to chondrocytes or MSCs for tissue engineering-based approaches. For the IVD, growth factors such as GDF5 or GDF6 or developmental transcription factors such as Brachyury or FOXF1 seem to be of high clinical interest. However, the most efficient method of gene transfer is still elusive, as several preclinical studies have reported many different non-viral methods and clinical translation of these techniques still needs to be validated. Here we discuss the non-viral methods applied for bone and joint and propose methods that can be promising in clinical use.
Collapse
Affiliation(s)
- Benjamin Gantenbein
- Tissue Engineering for Orthopaedics and Mechanobiology, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, Bern, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Shirley Tang
- Department of Biomedical Engineering and Department of Orthopaedics, Spine Research Institute Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Julien Guerrero
- Tissue Engineering for Orthopaedics and Mechanobiology, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, Bern, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Natalia Higuita-Castro
- Department of Biomedical Engineering and Department of Surgery, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Ana I Salazar-Puerta
- Department of Biomedical Engineering and Department of Surgery, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Andreas S Croft
- Tissue Engineering for Orthopaedics and Mechanobiology, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, Bern, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Amiq Gazdhar
- Department of Pulmonary Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland
| | - Devina Purmessur
- Department of Biomedical Engineering and Department of Orthopaedics, Spine Research Institute Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
102
|
Zinger A, Brozovich A, Pasto A, Sushnitha M, Martinez JO, Evangelopoulos M, Boada C, Tasciotti E, Taraballi F. Bioinspired Extracellular Vesicles: Lessons Learned From Nature for Biomedicine and Bioengineering. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E2172. [PMID: 33143238 PMCID: PMC7693812 DOI: 10.3390/nano10112172] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/23/2020] [Accepted: 10/23/2020] [Indexed: 12/14/2022]
Abstract
Efficient communication is essential in all layers of the biological chain. Cells exchange information using a variety of signaling moieties, such as small molecules, proteins, and nucleic acids. Cells carefully package these messages into lipid complexes, collectively named extracellular vesicles (EVs). In this work, we discuss the nature of these cell carriers, categorize them by their origin, explore their role in the homeostasis of healthy tissues, and examine how they regulate the pathophysiology of several diseases. This review will also address the limitations of using EVs for clinical applications and discuss novel methods to engineer nanoparticles to mimic the structure, function, and features of EVs. Using lessons learned from nature and understanding how cells use EVs to communicate across distant sites, we can develop a better understanding of how to tailor the fundamental features of drug delivery carriers to encapsulate various cargos and target specific sites for biomedicine and bioengineering.
Collapse
Affiliation(s)
- Assaf Zinger
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Ava Brozovich
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
- Texas A&M College of Medicine, Bryan, TX 77807, USA
| | - Anna Pasto
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Inflammation and Immunology, Humanitas Clinical and Research Center, 20089 Rozzano, Italy
| | - Manuela Sushnitha
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Jonathan O. Martinez
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Michael Evangelopoulos
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Christian Boada
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Ennio Tasciotti
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
- Biotechnology Program, San Raffaele University, Via di Val Cannuta, 247, 00166 Roma RM, Italy
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| |
Collapse
|
103
|
Kaddour H, Panzner TD, Welch JL, Shouman N, Mohan M, Stapleton JT, Okeoma CM. Electrostatic Surface Properties of Blood and Semen Extracellular Vesicles: Implications of Sialylation and HIV-Induced Changes on EV Internalization. Viruses 2020; 12:E1117. [PMID: 33019624 PMCID: PMC7601085 DOI: 10.3390/v12101117] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 09/04/2020] [Accepted: 09/24/2020] [Indexed: 12/14/2022] Open
Abstract
Although extracellular vesicle (EV) surface electrostatic properties (measured as zeta potential, ζ-potential) have been reported by many investigators, the biophysical implications of charge and EV origin remains uncertain. Here, we compared the ζ-potential of human blood EVs (BEVs) and semen EVs (SEVs) from 26 donors that were HIV-infected (HIV+, n = 13) or HIV uninfected (HIV-, n = 13). We found that, compared to BEVs that bear neutral surface charge, SEVs were significantly more negatively charged, even when BEVs and SEVs were from the same individual. Comparison of BEVs and SEVs from HIV- and HIV+ groups revealed subtle HIV-induced alteration in the ζ-potential of EVs, with the effect being more significant in SEVs (∆ζ-potential = -8.82 mV, p-value = 0.0062) than BEVs (∆ζ-potential = -1.4 mV, p-value = 0.0462). These observations were validated by differences in the isoelectric point (IEP) of EVs, which was in the order of HIV + SEV ≤ HIV-SEV ≪ HIV + BEV ≤ HIV-BEV. Functionally, the rate and efficiency of SEV internalization by the human cervical epithelial cell line, primary peripheral blood lymphocytes, and primary blood-derived monocytes were significantly higher than those of BEVs. Mechanistically, removal of sialic acids from the surface of EVs using neuraminidase treatment significantly decreased SEV's surface charge, concomitant with a substantial reduction in SEV's internalization. The neuraminidase effect was independent of HIV infection and insignificant for BEVs. Finally, these results were corroborated by enrichment of glycoproteins in SEVs versus BEVs. Taken together, these findings uncover fundamental tissue-specific differences in surface electrostatic properties of EVs and highlight the critical role of surface charge in EV/target cell interactions.
Collapse
Affiliation(s)
- Hussein Kaddour
- Department of Pharmacology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY 11794, USA; (H.K.); (T.D.P.); (N.S.)
| | - Tyler D. Panzner
- Department of Pharmacology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY 11794, USA; (H.K.); (T.D.P.); (N.S.)
| | - Jennifer L. Welch
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (J.L.W.); (J.T.S.)
- Medical Service, Iowa City Veterans Affairs Medical Center, Iowa City, IA 52246, USA
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Nadia Shouman
- Department of Pharmacology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY 11794, USA; (H.K.); (T.D.P.); (N.S.)
| | - Mahesh Mohan
- Host Pathogen Interaction Program, Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA;
| | - Jack T. Stapleton
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (J.L.W.); (J.T.S.)
- Medical Service, Iowa City Veterans Affairs Medical Center, Iowa City, IA 52246, USA
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Chioma M. Okeoma
- Department of Pharmacology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY 11794, USA; (H.K.); (T.D.P.); (N.S.)
| |
Collapse
|
104
|
Yin K, Liu X. CircMMP1
promotes the progression of glioma through
miR
‐433/
HMGB3
axis in vitro and in vivo. IUBMB Life 2020; 72:2508-2524. [PMID: 32918539 DOI: 10.1002/iub.2383] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/11/2020] [Accepted: 08/25/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Kai Yin
- Department of Neurosurgery the First Affiliated Hospital of Zhengzhou University Zhengzhou China
| | - Xianzhi Liu
- Department of Neurosurgery the First Affiliated Hospital of Zhengzhou University Zhengzhou China
| |
Collapse
|
105
|
Sun J, Shen H, Shao L, Teng X, Chen Y, Liu X, Yang Z, Shen Z. HIF-1α overexpression in mesenchymal stem cell-derived exosomes mediates cardioprotection in myocardial infarction by enhanced angiogenesis. Stem Cell Res Ther 2020; 11:373. [PMID: 32859268 PMCID: PMC7455909 DOI: 10.1186/s13287-020-01881-7] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 06/26/2020] [Accepted: 08/09/2020] [Indexed: 12/12/2022] Open
Abstract
Background Myocardial infarction (MI) is a severe disease that often associated with dysfunction of angiogenesis. Cell-based therapies for MI using mesenchymal stem cell (MSC)-derived exosomes have been well studied due to their strong proangiogenic effect. Genetic modification is one of the most common methods to enhance exosome therapy. This study investigated the proangiogenic and cardioprotective effect of exosomes derived from hypoxia-inducible factor 1-alpha (HIF-1α)-modified MSCs. Methods Lentivirus containing HIF-1α overexpressing vector was packaged and used to infect MSCs. Exosomes were isolated from MSC-conditioned medium by ultracentrifugation. Human umbilical vein endothelial cells (HUVECs) were treated under hypoxia condition for 48 h co-cultured with PBS, control exosomes, or HIF-1α-overexpressed exosomes, respectively. Then the preconditioned HUVECs were subjected to tube formation assay, Transwell assay, and EdU assay to evaluate the protective effect of exosomes. Meanwhile, mRNA and secretion levels of proangiogenic factors were measured by RT-qPCR and ELISA assays. In vivo assays were conducted using the rat myocardial infarction model. PBS, control exosomes, or HIF-1α-overexpressed exosomes were injected through tail vein after MI surgery. Heart function was assessed by echocardiography at days 3, 14, and 28. At day 7, mRNA and protein expression levels of proangiogenic factors in the peri-infarction area and circulation were evaluated, respectively. At day 28, hearts were collected and subjected to H&E staining, Masson’s trichrome staining, and immunofluorescent staining. Results HIF-1α-overexpressed exosomes rescued the impaired angiogenic ability, migratory function, and proliferation of hypoxia-injured HUVECs. Simultaneously, HIF-1α-overexpressed exosomes preserved heart function by promoting neovessel formation and inhibiting fibrosis in the rat MI model. In addition, both in vitro and in vivo proangiogenic factors mRNA and protein expression levels were elevated after HIF-1α-overexpressed exosome application. Conclusion HIF-1α-overexpressed exosomes could rescue the impaired angiogenic ability, migration, and proliferation of hypoxia-pretreated HUVECs in vitro and mediate cardioprotection by upregulating proangiogenic factors and enhancing neovessel formation.
Collapse
Affiliation(s)
- Jiacheng Sun
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, No.899, Pinghai Road, Suzhou, 215006, China
| | - Han Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, No.899, Pinghai Road, Suzhou, 215006, China
| | - Lianbo Shao
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, No.899, Pinghai Road, Suzhou, 215006, China
| | - Xiaomei Teng
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, No.899, Pinghai Road, Suzhou, 215006, China
| | - Yueqiu Chen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, No.899, Pinghai Road, Suzhou, 215006, China
| | - Xuan Liu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, No.899, Pinghai Road, Suzhou, 215006, China
| | - Ziying Yang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, No.899, Pinghai Road, Suzhou, 215006, China.
| | - Zhenya Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, No.899, Pinghai Road, Suzhou, 215006, China.
| |
Collapse
|
106
|
Critical Roles of Tumor Extracellular Vesicles in the Microenvironment of Thoracic Cancers. Int J Mol Sci 2020; 21:ijms21176024. [PMID: 32825667 PMCID: PMC7504491 DOI: 10.3390/ijms21176024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 12/17/2022] Open
Abstract
Extracellular vesicles (EVs), such as exosomes, are critical mediators of intercellular communication between tumor cells and other cells located in the microenvironment but also in more distant sites. Exosomes are small EVs that can carry a variety of molecules, such as lipids, proteins, and non-coding RNA, especially microRNAs (miRNAs). In thoracic cancers, including lung cancers and malignant pleural mesothelioma, EVs contribute to the immune-suppressive tumor microenvironment and to tumor growth and metastasis. In this review, we discuss the recent understanding of how exosomes behave in thoracic cancers and how and why they are promising liquid biomarkers for diagnosis, prognosis, and therapy, with a special focus on exosomal miRNAs.
Collapse
|
107
|
Yan HC, Yu TT, Li J, Qiao YQ, Wang LC, Zhang T, Li Q, Zhou YH, Liu DW. The Delivery of Extracellular Vesicles Loaded in Biomaterial Scaffolds for Bone Regeneration. Front Bioeng Biotechnol 2020; 8:1015. [PMID: 32974327 PMCID: PMC7466762 DOI: 10.3389/fbioe.2020.01015] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/03/2020] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) are heterogeneous nanoparticles actively released by cells that comprise highly conserved and efficient systems of intercellular communication. In recent years, numerous studies have proven that EVs play an important role in the field of bone tissue engineering (BTE) due to several advantages, such as good biosafety, stability and efficient delivery. However, the application of EVs therapies in bone regeneration has not been widely used. One of the major challenges for the application of EVs is the lack of sufficient scaffolds to load and control the release of EVs. Thus, in this review, we describe the most advanced current strategies for delivering EVs with various biomaterials for the use in bone regeneration, the role of EVs in bone regeneration, the distribution of EVs mediated by biomaterials and common methods of promoting EVs delivery efficacy with a focus on biomaterial properties.
Collapse
Affiliation(s)
- Hui-Chun Yan
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Ting-Ting Yu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Jing Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Yi-Qiang Qiao
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lin-Chuan Wang
- Eastman Institute for Oral Health, University of Rochester, Rochester, NY, United States
| | - Ting Zhang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Qian Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Yan-Heng Zhou
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Da-Wei Liu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Beijing, China
| |
Collapse
|
108
|
Biomaterials and extracellular vesicles in cell-free therapy for bone repair and regeneration: Future line of treatment in regenerative medicine. MATERIALIA 2020. [DOI: 10.1016/j.mtla.2020.100736] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
109
|
Can A, Coskun H. The rationale of using mesenchymal stem cells in patients with COVID-19-related acute respiratory distress syndrome: What to expect. Stem Cells Transl Med 2020; 9:1287-1302. [PMID: 32779878 PMCID: PMC7404450 DOI: 10.1002/sctm.20-0164] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/06/2020] [Accepted: 06/23/2020] [Indexed: 02/06/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2)‐caused coronavirus disease 2019 (COVID‐19) pandemic has become a global health crisis with an extremely rapid progress resulting in thousands of patients who may develop acute respiratory distress syndrome (ARDS) requiring intensive care unit (ICU) treatment. So far, no specific antiviral therapeutic agent has been demonstrated to be effective for COVID‐19; therefore, the clinical management is largely supportive and depends on the patients' immune response leading to a cytokine storm followed by lung edema, dysfunction of air exchange, and ARDS, which could lead to multiorgan failure and death. Given that human mesenchymal stem cells (MSCs) from various tissue sources have revealed successful clinical outcomes in many immunocompromised disorders by inhibiting the overactivation of the immune system and promoting endogenous repair by improving the microenvironment, there is a growing demand for MSC infusions in patients with COVID‐19‐related ARDS in the ICU. In this review, we have documented the rationale and possible outcomes of compassionate use of MSCs, particularly in patients with SARS‐CoV‐2 infections, toward proving or disproving the efficacy of this approach in the near future. Many centers have registered and approved, and some already started, single‐case or phase I/II trials primarily aiming to rescue their critical patients when no other therapeutic approach responds. On the other hand, it is also very important to mention that there is a good deal of concern about clinics offering unproven stem cell treatments for COVID‐19. The reviewers and oversight bodies will be looking for a balanced but critical appraisal of current trials.
Collapse
Affiliation(s)
- Alp Can
- Laboratory for Stem Cells and Reproductive Cell Biology, Department of Histology and Embryology, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Hakan Coskun
- Harvard Medical School, Boston, Massachusetts, USA.,Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
110
|
Sonoda S, Murata S, Nishida K, Kato H, Uehara N, Kyumoto YN, Yamaza H, Takahashi I, Kukita T, Yamaza T. Extracellular vesicles from deciduous pulp stem cells recover bone loss by regulating telomerase activity in an osteoporosis mouse model. Stem Cell Res Ther 2020; 11:296. [PMID: 32680564 PMCID: PMC7367365 DOI: 10.1186/s13287-020-01818-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/23/2020] [Accepted: 07/08/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Systemic transplantation of stem cells from human exfoliated deciduous teeth (SHED) recovers bone loss in animal models of osteoporosis; however, the mechanisms underlying this remain unclear. Here, we hypothesized that trophic factors within SHED-releasing extracellular vesicles (SHED-EVs) rescue osteoporotic phenotype. METHODS EVs were isolated from culture supernatant of SHED. SHED-EVs were treated with or without ribonuclease and systemically administrated into ovariectomized mice, followed by the function of recipient bone marrow mesenchymal stem cells (BMMSCs) including telomerase activity, osteoblast differentiation, and sepmaphorine-3A (SEMA3A) secretion. Subsequently, human BMMSCs were stimulated by SHED-EVs with or without ribonuclease treatment, and then human BMMSCs were examined regarding the function of telomerase activity, osteoblast differentiation, and SEMA3A secretion. Furthermore, SHED-EV-treated human BMMSCs were subcutaneously transplanted into the dorsal skin of immunocompromised mice with hydroxyapatite tricalcium phosphate (HA/TCP) careers and analyzed the de novo bone-forming ability. RESULTS We revealed that systemic SHED-EV-infusion recovered bone volume in ovariectomized mice and improved the function of recipient BMMSCs by rescuing the mRNA levels of Tert and telomerase activity, osteoblast differentiation, and SEMA3A secretion. Ribonuclease treatment depleted RNAs, including microRNAs, within SHED-EVs, and these RNA-depleted SHED-EVs attenuated SHED-EV-rescued function of recipient BMMSCs in the ovariectomized mice. These findings were supported by in vitro assays using human BMMSCs incubated with SHED-EVs. CONCLUSION Collectively, our findings suggest that SHED-secreted RNAs, such as microRNAs, play a crucial role in treating postmenopausal osteoporosis by targeting the telomerase activity of recipient BMMSCs.
Collapse
Affiliation(s)
- Soichiro Sonoda
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Sara Murata
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
- Section of Orthodontics and Dentofacial Orthopedics, Division of Oral Health, Growth & Development, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Kento Nishida
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hiroki Kato
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Norihisa Uehara
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yukari N Kyumoto
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Haruyoshi Yamaza
- Department of Pediatric Dentistry, Division of Oral Health, Growth & Development, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Ichiro Takahashi
- Section of Orthodontics and Dentofacial Orthopedics, Division of Oral Health, Growth & Development, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Toshio Kukita
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takayoshi Yamaza
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
111
|
Sengupta V, Sengupta S, Lazo A, Woods P, Nolan A, Bremer N. Exosomes Derived from Bone Marrow Mesenchymal Stem Cells as Treatment for Severe COVID-19. Stem Cells Dev 2020; 29:747-754. [PMID: 32380908 PMCID: PMC7310206 DOI: 10.1089/scd.2020.0080] [Citation(s) in RCA: 472] [Impact Index Per Article: 94.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 05/07/2020] [Indexed: 01/08/2023] Open
Abstract
This prospective nonrandomized open-label cohort study addresses the safety and efficacy of exosomes (ExoFlo™) derived from allogeneic bone marrow mesenchymal stem cells as treatment for severe COVID-19. During April 2020, ExoFlo was provided to 24 SARS-CoV-2 polymerase chain reaction-positive patients at a single hospital center, all of whom met criteria for severe COVID-19 as well as moderate-to-severe acute respiratory distress syndrome. Patients received a single 15 mL intravenous dose of ExoFlo and were evaluated for both safety and efficacy from days 1 to 14 post-treatment. All safety endpoints were met with no adverse events observed within 72 h of ExoFlo administration. A survival rate of 83% was observed. In total, 17 of 24 (71%) patients recovered, 3 of 24 (13%) patients remained critically ill though stable, and 4 of 24 (16%) patients expired for reasons unrelated to the treatment. Overall, after one treatment, patients' clinical status and oxygenation improved with an average pressure of arterial oxygen to fraction of inspired oxygen ratio (PaO2/FiO2) increase of 192% (P < 0.001). Laboratory values revealed significant improvements in absolute neutrophil count [mean reduction 32% (P value <0.001)] and lymphopenia with average CD3+, CD4+, and CD8+ lymphocyte counts increasing by 46% (P < 0.05), 45% (P < 0.05), and 46% (P < 0.001), respectively. Likewise, acute phase reactants declined, with mean C-reactive protein, ferritin, and D-dimer reduction of 77% (P < 0.001), 43% (P < 0.001), and 42% (P < 0.05), respectively. In conclusion, owing to its safety profile, capacity to restore oxygenation, downregulate cytokine storm, and reconstitute immunity, ExoFlo is a promising therapeutic candidate for severe COVID-19. Future randomized controlled trials (RCTs) are needed to determine ExoFlo therapeutic potential.
Collapse
Affiliation(s)
- Vikram Sengupta
- Department of Internal Medicine, NYU Grossman School of Medicine, New York, New York, USA
- Thrivewell Infusion, LLC, Brooklyn, New York, USA
| | - Sascha Sengupta
- Thrivewell Infusion, LLC, Brooklyn, New York, USA
- Department of Anesthesiology, Mount Sinai Hospital-Brooklyn, Brooklyn, New York, USA
| | - Angel Lazo
- Department of Internal Medicine, Englewood Hospital, Englewood, New Jersey, USA
| | - Peter Woods
- Department of Internal Medicine, CarePoint Health System–Christ Hospital, Jersey City, New Jersey, USA
| | - Anna Nolan
- Departments of Internal Medicine and Environmental Medicine, NYU School of Medicine, New York, New York, USA
| | - Nicholas Bremer
- Department of Anesthesiology, NYC Health + Hospitals/Bellevue, New York, New York, USA
| |
Collapse
|
112
|
Rodriguez-Quijada C, Dahl JB. Non-contact microfluidic mechanical property measurements of single apoptotic bodies. Biochim Biophys Acta Gen Subj 2020; 1865:129657. [PMID: 32512171 DOI: 10.1016/j.bbagen.2020.129657] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 03/18/2020] [Accepted: 06/02/2020] [Indexed: 01/29/2023]
Abstract
BACKGROUND Cells exchange information by secreting micro- and nanosized extracellular vesicles (EVs), ranging from exosomes (30-100 nm) to apoptotic bodies (ABs, 1-5 μm). There is still much to understand about fundamental EV biological, physical, and chemical properties before clinical applications can be developed. EV mechanical properties have only been measured with atomic force microscopy (AFM) with its problematic adhesion and hard substrate effects. To understand EV mechanical behavior in less extreme mechanical conditions relevant to blood flow and many soft tissue environments, a non-contact measurement technique is needed. METHODS We measured the mechanical properties of single microscale ABs derived from human blood plasma using non-contact microfluidics. EVs were gently stretched in extensional flow, similar to a traditional tensile test, and a linear mechanical model was applied to estimate mechanical stiffnesses from the observed stretching. RESULTS The effective shear elastic modulus of ABs in non-contact flow conditions is approximately 5.6 ± 0.5 Pa, 7 orders of magnitude lower than previously reported AFM-measured biological exosome stiffnesses and 200 times smaller than suspended cells. CONCLUSIONS Apoptotic bodies are very soft in fluid environments and exhibit lower effective stiffnesses than suspended cells. By measuring ABs in a natural fluid environment and low-force regime without hard probes and surfaces, we achieved closer agreement with linear mechanical theory and therefore more accurate stiffness measurements. GENERAL SIGNIFICANCE AFM manufacturers and users should consider implementing new mechanical models to interpret AFM force indentation curves so that accurate extracellular vesicle mechanical properties can be extracted.
Collapse
Affiliation(s)
| | - Joanna B Dahl
- Engineering Department, University of Massachusetts Boston, Boston, MA 02125, United States of America.
| |
Collapse
|
113
|
de Pontes LG, Altei WF, Galan A, Bilić P, Guillemin N, Kuleš J, Horvatić A, Ribeiro LNDM, de Paula E, Pereira VBR, Lucheis SB, Mrljak V, Eckersall PD, Ferreira RS, Dos Santos LD. Extracellular vesicles in infectious diseases caused by protozoan parasites in buffaloes. J Venom Anim Toxins Incl Trop Dis 2020; 26:e20190067. [PMID: 32528536 PMCID: PMC7262785 DOI: 10.1590/1678-9199-jvatitd-2019-0067] [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: 09/11/2019] [Accepted: 05/01/2020] [Indexed: 11/22/2022] Open
Abstract
Background Extracellular vesicles (EVs) are small membrane-bound vesicles of growing interest in vetetinary parasitology. The aim of the present report was to provide the first isolation, quantification and protein characterization of EVs from buffalo (Bubalus bubalis) sera infected with Theileria spp. Methods Infected animals were identified through optical microscopy and PCR. EVs were isolated from buffalo sera by size-exclusion chromatography and characterized using western blotting analysis, nanoparticle tracking analysis and transmission electron microscopy. Subsequently, the proteins from isolated vesicles were characterized by mass spectrometry. Results EVs from buffalo sera have shown sizes in the 124-140 nm range and 306 proteins were characterized. The protein-protein interaction analysis has evidenced biological processes and molecular function associated with signal transduction, binding, regulation of metabolic processes, transport, catalytic activity and response to acute stress. Five proteins have been shown to be differentially expressed between the control group and that infected with Theileria spp., all acting in the oxidative stress pathway. Conclusions EVs from buffaloes infected with Theileria spp. were successfully isolated and characterized. This is an advance in the knowledge of host-parasite relationship that contributes to the understanding of host immune response and theileriosis evasion mechanisms. These findings may pave the way for searching new EVs candidate-markers for a better production of safe biological products derived from buffaloes.
Collapse
Affiliation(s)
- Leticia Gomes de Pontes
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Wanessa Fernanda Altei
- Laboratory of Biochemistry and Molecular Biology, Department of Physiological Sciences, Federal University of São Carlos (UFSCar), São Carlos, SP, Brazil
| | - Asier Galan
- ERA Chair Team (VetMedZg), Clinic for Internal Diseases, Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Petra Bilić
- ERA Chair Team (VetMedZg), Clinic for Internal Diseases, Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Nicolas Guillemin
- ERA Chair Team (VetMedZg), Clinic for Internal Diseases, Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Josipa Kuleš
- ERA Chair Team (VetMedZg), Clinic for Internal Diseases, Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Anita Horvatić
- ERA Chair Team (VetMedZg), Clinic for Internal Diseases, Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Lígia Nunes de Morais Ribeiro
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Eneida de Paula
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | | | | | - Vladimir Mrljak
- ERA Chair Team (VetMedZg), Clinic for Internal Diseases, Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Peter David Eckersall
- ERA Chair Team (VetMedZg), Clinic for Internal Diseases, Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia.,Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, United Kingdom, UK
| | - Rui Seabra Ferreira
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo State University (UNESP), Botucatu, SP, Brazil.,Graduate Program in Clinical Research, Botucatu Medical School (FMB), São Paulo State University (UNESP), Botucatu, SP, Brazil.,Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Lucilene Delazari Dos Santos
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo State University (UNESP), Botucatu, SP, Brazil.,Graduate Program in Clinical Research, Botucatu Medical School (FMB), São Paulo State University (UNESP), Botucatu, SP, Brazil.,Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP), Botucatu, SP, Brazil
| |
Collapse
|
114
|
Zhao X, Wu D, Ma X, Wang J, Hou W, Zhang W. Exosomes as drug carriers for cancer therapy and challenges regarding exosome uptake. Biomed Pharmacother 2020; 128:110237. [PMID: 32470747 DOI: 10.1016/j.biopha.2020.110237] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/30/2020] [Accepted: 05/05/2020] [Indexed: 12/20/2022] Open
Abstract
With the development of biomedicine, exosomes are rapidly developing as a new therapy for tumors. As biological carriers, exosomes possess biological activity and can transport their contents between cells. The contents are natural or artificially loaded with biomolecules or chemical drugs. Exosomes deliver biomolecules or chemical drugs into the pathological sites of recipient, which can effectively inhibit the progression of tumors. However, the treatments of tumors through the delivery of exosomes are not sufficiently accurate or efficient, and various challenges need to be overcome. Exosomes from different cell sources possess different characteristics, as well as different specificity for various cells. In the future, for the promotion and application of exosomes, it is of great significance to understand how to select appropriate exosomes loaded with biomolecules or chemical drugs for different tumors types, and how to deliver exosomes to recipient cells accurately and efficiently. This review introduces the application and challenges of exosomes as delivery carriers in tumors.
Collapse
Affiliation(s)
- Xiaoyin Zhao
- Lab of Chemical Biology and Molecular Drug Design, Institute of Drug Development & Chemical Biology, College of Pharmaceutical Science, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, 310014, China.
| | - Dongliang Wu
- Lab of Chemical Biology and Molecular Drug Design, Institute of Drug Development & Chemical Biology, College of Pharmaceutical Science, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, 310014, China
| | - Xudong Ma
- Lab of Chemical Biology and Molecular Drug Design, Institute of Drug Development & Chemical Biology, College of Pharmaceutical Science, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, 310014, China
| | - Jiale Wang
- Lab of Chemical Biology and Molecular Drug Design, Institute of Drug Development & Chemical Biology, College of Pharmaceutical Science, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, 310014, China
| | - Wenjun Hou
- Lab of Chemical Biology and Molecular Drug Design, Institute of Drug Development & Chemical Biology, College of Pharmaceutical Science, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, 310014, China
| | - Wen Zhang
- Lab of Chemical Biology and Molecular Drug Design, Institute of Drug Development & Chemical Biology, College of Pharmaceutical Science, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, 310014, China.
| |
Collapse
|
115
|
Wang X, Ao J, Lu H, Zhao Q, Ma Y, Zhang J, Ren H, Zhang Y. Osteoimmune Modulation and Guided Osteogenesis Promoted by Barrier Membranes Incorporated with S-Nitrosoglutathione (GSNO) and Mesenchymal Stem Cell-Derived Exosomes. Int J Nanomedicine 2020; 15:3483-3496. [PMID: 32523344 PMCID: PMC7237116 DOI: 10.2147/ijn.s248741] [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: 02/07/2020] [Accepted: 05/05/2020] [Indexed: 12/11/2022] Open
Abstract
Background The use of polycaprolactone (PCL) for bone defects in a clinical setting is limited due to a lack of bioactivity. Exosomes derived from mesenchymal stem cells (MSCs) have an important immunoregulatory potential and together with S-nitrosoglutathione (GSNO) they possess therapeutic potential for bone regeneration. Materials and Methods In this study, PCL was modified with GSNO and MSC-derived exosomes and the impact on macrophages and osteogenes is evaluated. Results MSC-derived exosomes exhibited a cup-shaped morphology and were internalized by macrophages and human bone marrow-derived mesenchymal stromal cells (hBMSCs). The pattern of internalization of scaffold-immobilized exosomes was similar in RAW264.7 cells and hBMSCs after 4h and 24h of co-culture. Assessment of macrophage morphology under inflammatory conditions by scanning electronic microscopy (SEM) and confocal microscopy demonstrated macrophages were significantly elongated and expression of pro-inflammatory genes markedly decreased when co-cultured with PCL/PDA + GSNO + exosome scaffolds. Furthermore, this scaffold modification significantly enhanced osteogenic differentiation of hBMSCs. Discussion This study demonstrated the possibility of using a GSNO- and exosome-based strategy to adapt barrier membrane scaffolds. PCL/PDA + GSNO + exosome scaffolds may serve as an important barrier membrane for osteogenesis and tissue regeneration.
Collapse
Affiliation(s)
- Xin Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, People's Republic of China.,Joint Orthopaedic Research Center of Zunyi Medical University & University of Rochester Medical Center (JCMR-ZMU & URMC), Zunyi Medical University, Zunyi 563000, Guizhou, People's Republic of China
| | - Jun Ao
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, People's Republic of China
| | - Haiping Lu
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, People's Republic of China
| | - Qingyu Zhao
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, People's Republic of China
| | - Yaping Ma
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, People's Republic of China.,Joint Orthopaedic Research Center of Zunyi Medical University & University of Rochester Medical Center (JCMR-ZMU & URMC), Zunyi Medical University, Zunyi 563000, Guizhou, People's Republic of China
| | - Jun Zhang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, People's Republic of China
| | - Hao Ren
- Shenzhen Institute for Innovation and Translational Medicine, Shenzhen International Biological Valley-Life Science Industrial Park, Shenzhen, Guangdong 518119, People's Republic of China
| | - Yi Zhang
- Joint Orthopaedic Research Center of Zunyi Medical University & University of Rochester Medical Center (JCMR-ZMU & URMC), Zunyi Medical University, Zunyi 563000, Guizhou, People's Republic of China.,Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University, Zunyi 563000, Guizhou, People's Republic of China
| |
Collapse
|
116
|
Yamada Y, Nakamura-Yamada S, Konoki R, Baba S. Promising advances in clinical trials of dental tissue-derived cell-based regenerative medicine. Stem Cell Res Ther 2020; 11:175. [PMID: 32398041 PMCID: PMC7218566 DOI: 10.1186/s13287-020-01683-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/08/2020] [Accepted: 04/17/2020] [Indexed: 12/11/2022] Open
Abstract
Background Advances in regenerative medicine with stem cells have led to clinical trials. Dental/oral tissues are emerging as promising cellular sources of human mesenchymal stem cells. Recently, dental tissue-derived cells have been used clinically due to their great potential, easy accessibility, and ability to be obtained via methods with low invasiveness. The aim of this study is to systematically assess the clinical effectiveness of dental cell-mediated therapies compared to current evidence-based methods in human patients. Methods The electronic databases MEDLINE, Cochrane Central Register of Controlled Trials (CENTRAL), and ClinicalTrials.gov were searched up to December 2019 for clinical trials. Clinical trials with any intervention using stem cells/cells derived from dental tissue were included. Results A total of 815 studies were identified by the electronic search, and 38 articles qualified for full-text evaluation. Finally, 20 studies (10 clinical trials using dental pulp-derived cells, 3 clinical trials using periodontal ligament-derived cells, and 7 studies using gingiva-derived cells) were included in this review. No clinical trials using dental follicle- or apical papilla-derived cells were selected in this review. Dental pulp-derived cells were used in clinical trials for bone regeneration, periodontitis, and dental pulp regeneration. All clinical trials using periodontal ligament-derived cells and gingiva-derived cells were conducted for periodontal disease treatment and gingival augmentation, respectively. Among the 20 selected studies, 16 showed clinical benefits of cell transplantation therapies. In addition, no study reported adverse events that may have been associated with cell transplantation. Conclusions These findings indicate that dental tissue-derived cells would be useful for cell-based regenerative medicine for various diseases.
Collapse
Affiliation(s)
- Yoichi Yamada
- Department of Oral Implantology, Osaka Dental University, 1-5-17 Otemae Chuoku, Osaka, 540-0008, Japan. .,Department of Infectious Diseases and Applied Immunology, IMSUT Hospital of The Institute of Medical Science, The University of Tokyo, 4-6-1 Shiroganedai, Minato-ku, Tokyo, 108-8639, Japan.
| | - Sayaka Nakamura-Yamada
- Department of Oral Implantology, Osaka Dental University, 1-5-17 Otemae Chuoku, Osaka, 540-0008, Japan
| | - Ryutaro Konoki
- Department of Oral Implantology, Osaka Dental University, 1-5-17 Otemae Chuoku, Osaka, 540-0008, Japan
| | - Shunsuke Baba
- Department of Oral Implantology, Osaka Dental University, 1-5-17 Otemae Chuoku, Osaka, 540-0008, Japan
| |
Collapse
|
117
|
New concepts in regenerative medicine approaches to the treatment of female stress urinary incontinence. Curr Opin Urol 2020; 29:380-384. [PMID: 30855380 DOI: 10.1097/mou.0000000000000617] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW Update on recent regenerative medicine approaches to the treatment of stress urinary incontinence (SUI) caused by intrinsic sphincter deficiency (ISD). RECENT FINDINGS In the treatment of female SUI/ISD, results using different types of cellular therapy have been disappointing, and new approaches are desirable. To advance our regenerative medicine approaches to SUI/ISD, it is critical to utilize animal models that best parallel the pathophysiology of this disease in women. Many current animal models mimic acute SUI/ISD. However, SUI/ISD in women is usually a chronic condition resulting from previous muscle and nerve sphincter damage during parturition or muscle loss during aging. Similar to women, a nonhuman primate (NHP) model of chronic SUI/ISD has demonstrated only modest response to cell therapy. However, treatment with stromal cell-derived factor 1 (SDF1), also known as C-X-C motif chemokine 12 (CXCL12) restored continence in this model. SUMMARY As a potential therapeutic approach, the use of a well characterized chemokine, such as CXCL12, may by-pass the lengthy and expensive process of cell isolation, expansion, and injection. Recent findings in this new NHP model of chronic SUI/ISD may open up the field for noncell-based treatments.
Collapse
|
118
|
Xu T, Luo Y, Wang J, Zhang N, Gu C, Li L, Qian D, Cai W, Fan J, Yin G. Exosomal miRNA-128-3p from mesenchymal stem cells of aged rats regulates osteogenesis and bone fracture healing by targeting Smad5. J Nanobiotechnology 2020; 18:47. [PMID: 32178675 PMCID: PMC7077029 DOI: 10.1186/s12951-020-00601-w] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 03/05/2020] [Indexed: 02/08/2023] Open
Abstract
Transplantation of mesenchymal stem cells (MSCs) has been considered an effective therapeutic treatment for a variety of diseases including bone fracture. However, there are associated complications along with MSCs transplantation. There is evidence to show that exosomes (Exos) derived from MSCs exert a similar paracrine function. In addition, repair capabilities of MSCs decline with age. Hence, this study aims to confirm whether the Exos protective function on osteogenic differentiation and fracture healing from aged MSCs was attenuated. This information was used in order to investigate the underlying mechanism. MSCs were successfully isolated and identified from young and aged rats, and Exos were then obtained. Aged-Exos exhibited significantly attenuated effects on MSCs osteogenic differentiation in vitro and facture healing in vivo. Using miRNA array analysis, it was shown that miR-128-3p was markedly upregulated in Aged-Exos. In vitro experiments confirmed that Smad5 is a direct downstream target of miR-128-3p, and was inhibited by overexpressed miR-128-3p. A series gain- and loss- function experiment indicated that miR-128-3P serves a suppressor role in the process of fracture healing. Furthermore, effects caused by miR-128-3P mimic/inhibitor were reversed by the application of Smad5/siSmad5. Taken together, these results suggest that the therapeutic effects of MSCs-derived Exos may vary according to differential expression of miRNAs. Exosomal miR-128-3P antagomir may act as a promising therapeutic strategy for bone fracture healing, especially for the elderly.
Collapse
Affiliation(s)
- Tao Xu
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Yongjun Luo
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jiaxing Wang
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Ning Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Changjiang Gu
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Linwei Li
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Dingfei Qian
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Weihua Cai
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Jin Fan
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Guoyong Yin
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
119
|
Floriano JF, Willis G, Catapano F, de Lima PR, Reis FVDS, Barbosa AMP, Rudge MVC, Emanueli C. Exosomes Could Offer New Options to Combat the Long-Term Complications Inflicted by Gestational Diabetes Mellitus. Cells 2020; 9:E675. [PMID: 32164322 PMCID: PMC7140615 DOI: 10.3390/cells9030675] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 02/20/2020] [Accepted: 02/29/2020] [Indexed: 02/08/2023] Open
Abstract
Gestational diabetes Mellitus (GDM) is a complex clinical condition that promotes pelvic floor myopathy, thus predisposing sufferers to urinary incontinence (UI). GDM usually regresses after birth. Nonetheless, a GDM history is associated with higher risk of subsequently developing type 2 diabetes, cardiovascular diseases (CVD) and UI. Some aspects of the pathophysiology of GDM remain unclear and the associated pathologies (outcomes) are poorly addressed, simultaneously raising public health costs and diminishing women's quality of life. Exosomes are small extracellular vesicles produced and actively secreted by cells as part of their intercellular communication system. Exosomes are heterogenous in their cargo and depending on the cell sources and environment, they can mediate both pathogenetic and therapeutic functions. With the advancement in knowledge of exosomes, new perspectives have emerged to support the mechanistic understanding, prediction/diagnosis and ultimately, treatment of the post-GMD outcomes. Here, we will review recent advances in knowledge of the role of exosomes in GDM and related areas and discuss the possibilities for translating exosomes as therapeutic agents in the GDM clinical setting.
Collapse
Affiliation(s)
- Juliana Ferreira Floriano
- Botucatu Medical School, Sao Paulo State University, 18618687 Botucatu, Brazil; (J.F.F.); (P.R.d.L.); (F.V.D.S.R.); (A.M.P.B.)
| | - Gareth Willis
- Division of Newborn Medicine/Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Francesco Catapano
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK;
| | - Patrícia Rodrigues de Lima
- Botucatu Medical School, Sao Paulo State University, 18618687 Botucatu, Brazil; (J.F.F.); (P.R.d.L.); (F.V.D.S.R.); (A.M.P.B.)
| | | | - Angélica Mercia Pascon Barbosa
- Botucatu Medical School, Sao Paulo State University, 18618687 Botucatu, Brazil; (J.F.F.); (P.R.d.L.); (F.V.D.S.R.); (A.M.P.B.)
| | - Marilza Vieira Cunha Rudge
- Botucatu Medical School, Sao Paulo State University, 18618687 Botucatu, Brazil; (J.F.F.); (P.R.d.L.); (F.V.D.S.R.); (A.M.P.B.)
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK;
| |
Collapse
|
120
|
de Jong OG, Murphy DE, Mäger I, Willms E, Garcia-Guerra A, Gitz-Francois JJ, Lefferts J, Gupta D, Steenbeek SC, van Rheenen J, El Andaloussi S, Schiffelers RM, Wood MJA, Vader P. A CRISPR-Cas9-based reporter system for single-cell detection of extracellular vesicle-mediated functional transfer of RNA. Nat Commun 2020; 11:1113. [PMID: 32111843 PMCID: PMC7048928 DOI: 10.1038/s41467-020-14977-8] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 02/12/2020] [Indexed: 01/08/2023] Open
Abstract
Extracellular vesicles (EVs) form an endogenous transport system for intercellular transfer of biological cargo, including RNA, that plays a pivotal role in physiological and pathological processes. Unfortunately, whereas biological effects of EV-mediated RNA transfer are abundantly studied, regulatory pathways and mechanisms remain poorly defined due to a lack of suitable readout systems. Here, we describe a highly-sensitive CRISPR-Cas9-based reporter system that allows direct functional study of EV-mediated transfer of small non-coding RNA molecules at single-cell resolution. Using this CRISPR operated stoplight system for functional intercellular RNA exchange (CROSS-FIRE) we uncover various genes involved in EV subtype biogenesis that play a regulatory role in RNA transfer. Moreover we identify multiple genes involved in endocytosis and intracellular membrane trafficking that strongly regulate EV-mediated functional RNA delivery. Altogether, this approach allows the elucidation of regulatory mechanisms in EV-mediated RNA transfer at the level of EV biogenesis, endocytosis, intracellular trafficking, and RNA delivery.
Collapse
Affiliation(s)
- Olivier G de Jong
- Laboratory of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Daniel E Murphy
- Laboratory of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Imre Mäger
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Eduard Willms
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Antonio Garcia-Guerra
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- Department of Physics, University of Oxford, Oxford, United Kingdom
| | - Jerney J Gitz-Francois
- Laboratory of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Juliet Lefferts
- Pediatric Pulmonology and Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dhanu Gupta
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Huddinge, Sweden
| | - Sander C Steenbeek
- Department of Molecular Pathology, Oncode Institute, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jacco van Rheenen
- Department of Molecular Pathology, Oncode Institute, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Samir El Andaloussi
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Huddinge, Sweden
| | - Raymond M Schiffelers
- Laboratory of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Matthew J A Wood
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Pieter Vader
- Laboratory of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands.
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
121
|
Perspectives for Future Use of Extracellular Vesicles from Umbilical Cord- and Adipose Tissue-Derived Mesenchymal Stem/Stromal Cells in Regenerative Therapies-Synthetic Review. Int J Mol Sci 2020; 21:ijms21030799. [PMID: 31991836 PMCID: PMC7036930 DOI: 10.3390/ijms21030799] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/18/2020] [Accepted: 01/23/2020] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem/ stromal cells (MSCs) represent progenitor cells of various origin with multiple differentiation potential, representing the most studied population of stem cells in both in vivo pre-clinical and clinical studies. MSCs may be found in many tissue sources including extensively studied adipose tissue (ADSCs) and umbilical cord Wharton’s jelly (UC-MSCs). Most of sanative effects of MSCs are due to their paracrine activity, which includes also release of extracellular vesicles (EVs). EVs are small, round cellular derivatives carrying lipids, proteins, and nucleic acids including various classes of RNAs. Due to several advantages of EVs when compare to their parental cells, MSC-derived EVs are currently drawing attention of several laboratories as potential new tools in tissue repair. This review focuses on pro-regenerative properties of EVs derived from ADSCs and UC-MSCs. We provide a synthetic summary of research conducted in vitro and in vivo by employing animal models and within initial clinical trials focusing on neurological, cardiovascular, liver, kidney, and skin diseases. The summarized studies provide encouraging evidence about MSC-EVs pro-regenerative capacity in various models of diseases, mediated by several mechanisms. Although, direct molecular mechanisms of MSC-EV action are still under investigation, the current growing data strongly indicates their potential future usefulness for tissue repair.
Collapse
|
122
|
Zhu Z, Kalyan BS, Chen L. Therapeutic potential role of exosomes for ischemic stroke. BRAIN SCIENCE ADVANCES 2020. [DOI: 10.1177/2096595820902588] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Exosomes are extracellular vesicles with a diameter of 30–100 nm, which are released into the extracellular space by fusion of multivesicular and plasma membranes. These vesicles actually play a distinct role in cell communication, although they were considered as membrane debris in the past. The endosomal sorting complex required for transport (ESCRT)-dependent and ESCRT-independent mechanisms are currently considered to be involved in the sorting of exosomes, and the release of exosomes is related to the members of Rab protein family and SNARE family. In recent years, the therapeutic potential of exosomes has become apparent. For example, via the direct transplantation of exosomes, the ischemic area after stroke is reduced, and the neurological function is improved significantly. Furthermore, they can be used as effective drug delivery vehicles due to their unique characteristics such as low immunogenicity and nanometer size. In conclusion, exosomes provide a cell-free treatment for ischemic stroke.
Collapse
Affiliation(s)
- Zhihan Zhu
- Department of Neurosurgery, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Bikram Shah Kalyan
- Department of Neurosurgery, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Lukui Chen
- Department of Neurosurgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510310, Guangdong, China
| |
Collapse
|
123
|
Hu Y, Zhang Y, Ni CY, Chen CY, Rao SS, Yin H, Huang J, Tan YJ, Wang ZX, Cao J, Liu ZZ, Xie PL, Wu B, Luo J, Xie H. Human umbilical cord mesenchymal stromal cells-derived extracellular vesicles exert potent bone protective effects by CLEC11A-mediated regulation of bone metabolism. Am J Cancer Res 2020; 10:2293-2308. [PMID: 32089743 PMCID: PMC7019162 DOI: 10.7150/thno.39238] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/30/2019] [Indexed: 12/11/2022] Open
Abstract
Osteoporosis and osteoporotic fractures severely compromise quality of life in elderly people and lead to early death. Human umbilical cord mesenchymal stromal cell (MSC)-derived extracellular vesicles (hucMSC-EVs) possess considerable therapeutic effects in tissue repair and regeneration. Thus, in the present study, we investigated the effects of hucMSC-EVs on primary and secondary osteoporosis and explored the underlying mechanisms. Methods: hucMSCs were isolated and cultured. EVs were obtained from the conditioned medium of hucMSCs and determined by using transmission electron microscopy, dynamic light scattering and Western Blot analyses. The effects of hucMSC-EVs on ovariectomy-induced postmenopausal osteoporosis and tail suspension-induced hindlimb disuse osteoporosis in mouse models were assessed by using microcomputed tomography, biomechanical, histochemical and immunohistochemical, as well as histomorphometric analyses. Proteomic analysis was applied between hucMSC-EVs and hucMSCs to screen the candidate proteins that mediate hucMSC-EVs function. The effects of hucMSC-EVs on osteogenic and adipogenic differentiation of bone marrow mesenchymal stromal cells (BMSCs), and osteoclastogenesis of the macrophage cell line RAW264.7 in vitro were determined by using cytochemical staining and quantitative real-time PCR analysis. Subsequently, the roles of the key protein in hucMSC-EVs-induced regulation on BMSCs and RAW264.7 cells were evaluated. Results: hucMSCs were able to differentiate into osteoblasts, adipocytes or chondrocytes and positively expressed CD29, CD44, CD73 and CD90, but negatively expressed CD34 and CD45. The morphological assessment revealed the typical cup- or sphere-shaped morphology of hucMSC-EVs with diameters predominantly ranging from 60 nm to 150 nm and expressed CD9, CD63, CD81 and TSG101. The systemic administration of hucMSC-EVs prevented bone loss and maintained bone strength in osteoporotic mice by enhancing bone formation, reducing marrow fat accumulation and decreasing bone resorption. Proteomic analysis showed that the potently pro-osteogenic protein, CLEC11A (C-type lectin domain family 11, member A) was very highly enriched in hucMSC-EVs. In addition, hucMSC-EVs enhanced the shift from adipogenic to osteogenic differentiation of BMSCs via delivering CLEC11A in vitro. Moreover, CLEC11A was required for the inhibitory effects of hucMSC-EVs on osteoclast formation. Conclusion: Our results suggest that hucMSC-EVs serve as a critical regulator of bone metabolism by transferring CLEC11A and may represent a potential agent for prevention and treatment of osteoporosis.
Collapse
|
124
|
Zhong D, Cao Y, Li CJ, Li M, Rong ZJ, Jiang L, Guo Z, Lu HB, Hu JZ. Neural stem cell-derived exosomes facilitate spinal cord functional recovery after injury by promoting angiogenesis. Exp Biol Med (Maywood) 2020; 245:54-65. [PMID: 31903774 DOI: 10.1177/1535370219895491] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Acute traumatic spinal cord injury is a devastating event without effective therapeutic approach. The feeble plasticity of spinal cord microvascular endothelial cells (SCMECs) after trauma is one of the major causes for the exacerbation of spinal cord injury. Therefore, improving the plasticity and regeneration of SCMECs is crucial to promote recovery after spinal cord injury. For the present study, we explored the influence of exosomes derived from neural stem cells (NSCs-Exos) on the spinal cord microvascular regeneration after spinal cord injury and determined the underlying mechanisms. After the primary NSCs and SCMECs were extracted, exosomes were isolated from NSCs conditioned medium and used to co-incubated with the SCMECs in vitro, and then the effect of exosomes on the angiogenic activities of SCMECs was measured. The candidate molecules involved in the NSCs-Exos-mediated angiogenesis were screened using Western blotting. The effect of NSCs-Exos on angiogenesis and spinal cord functional recovery after injury in vivo was analyzed. The results demonstrated that NSCs-Exos could enhance the angiogenic activities of SCMECs, and were highly enriched in VEGF-A. The level of VEGF-A was downregulated in NSCsshVEGF-A-Exos and the pro-angiogenic effects on cocultured SCMECs were inhibited. Furthermore, NSCs-Exos significantly accelerated the microvascular regeneration, reduced the spinal cord cavity, and improved the Basso mouse scale scores in spinal cord injury mice. This work provides the evidence of the underlying mechanism of NSCs-Exos-mediated angiogenesis and suggests a novel therapeutic target for spinal cord injury. Impact statement The feeble plasticity of SCMECs after trauma is one of the major causes for the exacerbation of SCI. Therefore, improving the regeneration ability of SCMECs is crucial to promote spinal cord functional recovery after injury. Our current study uncovered that NSCs-Exos could promote SCMECs migration, tube formation and proliferation in vitro, and further identified that exosomal VEGF-A mediated the pro-angiogenic effect. Furthermore, we observed a remarkable microvascular density increase, spinal cord cavity shrinkage, and motor function recovery in SCI mice treated with NSCs-Exos, which confirmed the therapeutic effects of NSCs-Exos to alleviate SCI. Downregulating VEGF-A partially abolished these effects of NSCs-Exos. This is the first study to reveal that NSCs-Exos has the pro-angiogenic effect on SCMECs by transferring VEGF-A and promote microvascular regeneration and tissue healing, indicating that NSCs-Exos can become a promising therapeutic bioagent for facilitating the functional recovery of SCI.
Collapse
Affiliation(s)
- Dong Zhong
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha 410008, PR China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha 410008, PR China
| | - Yong Cao
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha 410008, PR China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha 410008, PR China
| | - Cheng-Jun Li
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha 410008, PR China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha 410008, PR China
| | - Miao Li
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha 410008, PR China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha 410008, PR China
| | - Zi-Jie Rong
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha 410008, PR China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha 410008, PR China
| | - Liyuan Jiang
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha 410008, PR China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha 410008, PR China
| | - Zhu Guo
- Department of Spine Surgery, Affiliated Hospital of Qingdao University, Qingdao, 266003, PR China
| | - Hong-Bin Lu
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha 410008, PR China.,Department of Sports Medicine, Research Center of Sports Medicine, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Jian-Zhong Hu
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha 410008, PR China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha 410008, PR China
| |
Collapse
|
125
|
Alcaraz MJ, Compañ A, Guillén MI. Extracellular Vesicles from Mesenchymal Stem Cells as Novel Treatments for Musculoskeletal Diseases. Cells 2019; 9:cells9010098. [PMID: 31906087 PMCID: PMC7017209 DOI: 10.3390/cells9010098] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/23/2019] [Accepted: 12/28/2019] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) represent a promising therapy for musculoskeletal diseases. There is compelling evidence indicating that MSC effects are mainly mediated by paracrine mechanisms and in particular by the secretion of extracellular vesicles (EVs). Many studies have thus suggested that EVs may be an alternative to cell therapy with MSCs in tissue repair. In this review, we summarize the current understanding of MSC EVs actions in preclinical studies of (1) immune regulation and rheumatoid arthritis, (2) bone repair and bone diseases, (3) cartilage repair and osteoarthritis, (4) intervertebral disk degeneration and (5) skeletal muscle and tendon repair. We also discuss the mechanisms underlying these actions and the perspectives of MSC EVs-based strategies for future treatments of musculoskeletal disorders.
Collapse
Affiliation(s)
- María José Alcaraz
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Av. Vicent A. Estellés s/n, 46100 Burjasot, Valencia, Spain
- Correspondence:
| | - Alvaro Compañ
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Av. Vicent A. Estellés s/n, 46100 Burjasot, Valencia, Spain
| | - María Isabel Guillén
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Av. Vicent A. Estellés s/n, 46100 Burjasot, Valencia, Spain
- Department of Pharmacy, Cardenal Herrera-CEU University, Ed. Ciencias de la Salud, 46115 Alfara, Valencia, Spain
| |
Collapse
|
126
|
Ramasubramanian L, Kumar P, Wang A. Engineering Extracellular Vesicles as Nanotherapeutics for Regenerative Medicine. Biomolecules 2019; 10:E48. [PMID: 31905611 PMCID: PMC7023093 DOI: 10.3390/biom10010048] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 12/24/2019] [Accepted: 12/26/2019] [Indexed: 01/01/2023] Open
Abstract
Long thought of to be vesicles that primarily recycled waste biomolecules from cells, extracellular vesicles (EVs) have now emerged as a new class of nanotherapeutics for regenerative medicine. Recent studies have proven their potential as mediators of cell proliferation, immunomodulation, extracellular matrix organization and angiogenesis, and are currently being used as treatments for a variety of diseases and injuries. They are now being used in combination with a variety of more traditional biomaterials and tissue engineering strategies to stimulate tissue repair and wound healing. However, the clinical translation of EVs has been greatly slowed due to difficulties in EV isolation and purification, as well as their limited yields and functional heterogeneity. Thus, a field of EV engineering has emerged in order to augment the natural properties of EVs and to recapitulate their function in semi-synthetic and synthetic EVs. Here, we have reviewed current technologies and techniques in this growing field of EV engineering while highlighting possible future applications for regenerative medicine.
Collapse
Affiliation(s)
- Lalithasri Ramasubramanian
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California–Davis, Sacramento, CA 95817, USA (P.K.)
- Department of Biomedical Engineering, University of California–Davis, Davis, CA 95616, USA
| | - Priyadarsini Kumar
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California–Davis, Sacramento, CA 95817, USA (P.K.)
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children–Northern California, Sacramento, CA 95817, USA
| | - Aijun Wang
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California–Davis, Sacramento, CA 95817, USA (P.K.)
- Department of Biomedical Engineering, University of California–Davis, Davis, CA 95616, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children–Northern California, Sacramento, CA 95817, USA
| |
Collapse
|
127
|
Paracrine Proangiogenic Function of Human Bone Marrow-Derived Mesenchymal Stem Cells Is Not Affected by Chronic Kidney Disease. Stem Cells Int 2019; 2019:1232810. [PMID: 31933648 PMCID: PMC6942892 DOI: 10.1155/2019/1232810] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/20/2019] [Accepted: 11/26/2019] [Indexed: 12/18/2022] Open
Abstract
Background Cell-based therapies are being developed to meet the need for curative therapy in chronic kidney disease (CKD). Bone marrow- (BM-) derived mesenchymal stromal cells (MSCs) enhance tissue repair and induce neoangiogenesis through paracrine action of secreted proteins and extracellular vesicles (EVs). Administration of allogeneic BM MSCs is less desirable in a patient population likely to require a kidney transplant, but potency of autologous MSCs should be confirmed, given previous indications that CKD-induced dysfunction is present. While the immunomodulatory capacity of CKD BM MSCs has been established, it is unknown whether CKD affects wound healing and angiogenic potential of MSC-derived CM and EVs. Methods MSCs were cultured from BM obtained from kidney transplant recipients (N = 15) or kidney donors (N = 17). Passage 3 BM MSCs and BM MSC-conditioned medium (CM) were used for experiments. EVs were isolated from CM by differential ultracentrifugation. BM MSC differentiation capacity, proliferation, and senescence-associated β-galactosidase activity was assessed. In vitro promigratory and proangiogenic capacity of BM MSC-derived CM and EVs was assessed using an in vitro scratch wound assay and Matrigel angiogenesis assay. Results Healthy and CKD BM MSCs exhibited similar differentiation capacity, proliferation, and senescence-associated β-galactosidase activity. Scratch wound migration was not significantly different between healthy and CKD MSCs (P = 0.18). Healthy and CKD BM MSC-derived CM induced similar tubule formation (P = 0.21). There was also no difference in paracrine regenerative function of EVs (scratch wound: P = 0.6; tubulogenesis: P = 0.46). Conclusions Our results indicate that MSCs have an intrinsic capacity to produce proangiogenic paracrine factors, including EVs, which is not affected by donor health status regarding CKD. This suggests that autologous MSC-based therapy is a viable option in CKD.
Collapse
|
128
|
Ranjan P, Kumari R, Verma SK. Cardiac Fibroblasts and Cardiac Fibrosis: Precise Role of Exosomes. Front Cell Dev Biol 2019; 7:318. [PMID: 31867328 PMCID: PMC6904280 DOI: 10.3389/fcell.2019.00318] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/20/2019] [Indexed: 12/12/2022] Open
Abstract
Exosomes are a group of extracellular microvesicles that deliver biologically active RNAs, proteins, lipids and other signaling molecules to recipient cells. Classically, exosomes act as a vehicle by which cells or organs communicate with each other to maintain cellular/tissue homeostasis and to respond to pathological stress. Most multicellular systems, including the cardiovascular system, use exosomes for intercellular communication. In heart, endogenous exosomes from cardiac cells or stem cells aid in regulation of cell survival, cell proliferation and cell death; and thus tightly regulate cardiac biology and repair processes. Pathological stimulus in heart alters secretion and molecular composition of exosomes, thus influencing the above processes. The past decade has yielded increasing interest in the role of exosomes in the cardiovascular system and significant contribution of cardiac fibroblast (CF) and mediated cardiac fibrosis in heart failure, in this review we had overviewed the relevant literatures about fibroblast exosomes, its effect in the cardiovascular biology and its impact on cardiovascular disease (CVD). This review briefly describes the communication between fibroblasts and other cardiac cells via exosomes, the influence of such on myocardial fibrosis and remodeling, and the possibilities to use exosomes as biomarkers for acute and chronic heart diseases.
Collapse
Affiliation(s)
- Prabhat Ranjan
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rajesh Kumari
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Suresh Kumar Verma
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
129
|
Thankam FG, Chandra I, Diaz C, Dilisio MF, Fleegel J, Gross RM, Agrawal DK. Matrix regeneration proteins in the hypoxia-triggered exosomes of shoulder tenocytes and adipose-derived mesenchymal stem cells. Mol Cell Biochem 2019; 465:75-87. [PMID: 31797254 DOI: 10.1007/s11010-019-03669-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/30/2019] [Indexed: 12/14/2022]
Abstract
Regenerative functions of exosomes rely on their contents which are influenced by pathological stimuli, including hypoxia, in rotator cuff tendon injuries (RCTI). The hypoxic environment triggers tenocytes and adjacent adipose-derived mesenchymal stem cells (ADMSCs) to release regenerative mediators to the ECM via the exosomes which elicit autocrine/paracrine responses to protect the tendon matrix from injury. We investigated the exosomal protein contents from tenocytes and subcutaneous ADMSCs from the shoulder of Yucatan microswine cultured under hypoxic conditions (2% O2). The exosomal proteins were detected using high-resolution mass spectrometry nano-LC-MS/MS Tribrid system and were compiled using 'Scaffold' software. Hypoxic exosomes from tenocytes and ADMSCs carried 199 and 65 proteins, respectively. The key proteins identified by mass spectrometry and associated with ECM homeostasis from hypoxic ADMSCs included MMP2, COL6A, CTSD and TN-C and those from hypoxic tenocytes were THSB1, NSEP1, ITIH4 and TN-C. These findings were confirmed at the mRNA and protein level in the hypoxic ADMSCs and tenocytes. These proteins are involved in multiple signaling pathways of ECM repair/regeneration. This warrants further investigations for their translational significance in the management of RCTI.
Collapse
Affiliation(s)
- Finosh G Thankam
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Isaiah Chandra
- Departments of Clinical & Translational Science and Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - Connor Diaz
- Departments of Clinical & Translational Science and Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - Matthew F Dilisio
- Departments of Clinical & Translational Science and Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - Jonathan Fleegel
- Departments of Clinical & Translational Science and Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - R Michael Gross
- Departments of Clinical & Translational Science and Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA.
| |
Collapse
|
130
|
Coalson E, Bishop E, Liu W, Feng Y, Spezia M, Liu B, Shen Y, Wu D, Du S, Li AJ, Ye Z, Zhao L, Cao D, Li A, Hagag O, Deng A, Liu W, Li M, Haydon RC, Shi L, Athiviraham A, Lee MJ, Wolf JM, Ameer GA, He TC, Reid RR. Stem cell therapy for chronic skin wounds in the era of personalized medicine: From bench to bedside. Genes Dis 2019; 6:342-358. [PMID: 31832514 PMCID: PMC6888708 DOI: 10.1016/j.gendis.2019.09.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 09/07/2019] [Accepted: 09/09/2019] [Indexed: 02/06/2023] Open
Abstract
With the significant financial burden of chronic cutaneous wounds on the healthcare system, not to the personal burden mention on those individuals afflicted, it has become increasingly essential to improve our clinical treatments. This requires the translation of the most recent benchtop approaches to clinical wound repair as our current treatment modalities have proven insufficient. The most promising potential treatment options rely on stem cell-based therapies. Stem cell proliferation and signaling play crucial roles in every phase of the wound healing process and chronic wounds are often associated with impaired stem cell function. Clinical approaches involving stem cells could thus be utilized in some cases to improve a body's inhibited healing capacity. We aim to present the laboratory research behind the mechanisms and effects of this technology as well as current clinical trials which showcase their therapeutic potential. Given the current problems and complications presented by chronic wounds, we hope to show that developing the clinical applications of stem cell therapies is the rational next step in improving wound care.
Collapse
Affiliation(s)
- Elam Coalson
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Elliot Bishop
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Surgery, Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Wei Liu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Key Laboratory of Diagnostic Medicine (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Yixiao Feng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Key Laboratory of Diagnostic Medicine (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Mia Spezia
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Bo Liu
- Department of Surgery, Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Key Laboratory of Diagnostic Medicine (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Yi Shen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, Xiangya Second Hospital of Central South University, Changsha 410011, China
| | - Di Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Scott Du
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Student Inquiry Research Program, Illinois Mathematics and Science Academy (IMSA), Aurora, IL 60506, USA
| | - Alexander J. Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Zhenyu Ye
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Ling Zhao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Key Laboratory of Diagnostic Medicine (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Daigui Cao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, Chongqing General Hospital, Chongqing 400013, China
| | - Alissa Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Ofir Hagag
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Alison Deng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Student Inquiry Research Program, Illinois Mathematics and Science Academy (IMSA), Aurora, IL 60506, USA
| | - Winny Liu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Student Inquiry Research Program, Illinois Mathematics and Science Academy (IMSA), Aurora, IL 60506, USA
| | - Mingyang Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Student Inquiry Research Program, Illinois Mathematics and Science Academy (IMSA), Aurora, IL 60506, USA
| | - Rex C. Haydon
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Lewis Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Aravind Athiviraham
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Michael J. Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jennifer Moriatis Wolf
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Guillermo A. Ameer
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60616, USA
- Center for Advanced Regenerative Engineering (CARE), Evanston, IL 60208, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Center for Advanced Regenerative Engineering (CARE), Evanston, IL 60208, USA
| | - Russell R. Reid
- Department of Surgery, Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Center for Advanced Regenerative Engineering (CARE), Evanston, IL 60208, USA
| |
Collapse
|
131
|
Tian J, Popal MS, Zhao Y, Liu Y, Chen K, Liu Y. Interplay between Exosomes and Autophagy in Cardiovascular Diseases: Novel Promising Target for Diagnostic and Therapeutic Application. Aging Dis 2019; 10:1302-1310. [PMID: 31788341 PMCID: PMC6844582 DOI: 10.14336/ad.2018.1020] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/20/2018] [Indexed: 12/12/2022] Open
Abstract
Exosome, is identified as a nature nanocarrier and intercellular messenger that regulates cell to cell communication. Autophagy is critical in maintenance of protein homeostasis by degradation of damaged proteins and organelles. Autophagy and exosomes take pivotal roles in cellular homeostasis and cardiovascular disease. Currently, the coordinated mechanisms for exosomes and autophagy in the maintenance of cellular fitness are now garnering much attention. In the present review, we discussed the interplay of exosomes and autophagy in the context of physiology and pathology of the heart, which might provide novel insights for diagnostic and therapeutic application of cardiovascular diseases.
Collapse
Affiliation(s)
- Jinfan Tian
- 1Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,2Cardiovascular disease center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mohammad Sharif Popal
- 1Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yingke Zhao
- 3Li Ka Shing Faculty of Medicine, The University of HongKong, Pokfulam, Hong Kong
| | - Yanfei Liu
- 4Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Keji Chen
- 2Cardiovascular disease center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yue Liu
- 2Cardiovascular disease center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
132
|
Ullah M, Qiao Y, Concepcion W, Thakor AS. Stem cell-derived extracellular vesicles: role in oncogenic processes, bioengineering potential, and technical challenges. Stem Cell Res Ther 2019; 10:347. [PMID: 31771657 PMCID: PMC6880555 DOI: 10.1186/s13287-019-1468-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 10/21/2019] [Accepted: 10/23/2019] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) are cellular-derived versatile transporters with a specialized property for trafficking a variety of cargo, including metabolites, growth factors, cytokines, proteins, lipids, and nucleic acids, throughout the microenvironment. EVs can act in a paracrine manner to facilitate communication between cells as well as modulate immune, inflammatory, regenerative, and remodeling processes. Of particular interest is the emerging association between EVs and stem cells, given their ability to integrate complex inputs for facilitating cellular migration to the sites of tissue injury. Additionally, stem cell-derived EVs can also act in an autocrine manner to influence stem cell proliferation, mobilization, differentiation, and self-renewal. Hence, it has been postulated that stem cells and EVs may work synergistically in the process of tissue repair and that dysregulation of EVs may cause a loss of homeostasis in the microenvironment leading to disease. By harnessing the property of EVs for delivery of small molecules, stem cell-derived EVs possess significant potential as a platform for developing bioengineering approaches for next-generation cancer therapies and targeted drug delivery methods. Although one of the main challenges of clinical cancer treatment remains a lack of specificity for the delivery of effective treatment options, EVs can be modified via genetic, biochemical, or synthetic methods for enhanced targeting ability of chemotherapeutic agents in promoting tumor regression. Here, we summarize recent research on the bioengineering potential of EV-based cancer therapies. A comprehensive understanding of EV modification may provide a novel strategy for cancer therapy and for the utilization of EVs in the targeting of oncogenic processes. Furthermore, innovative and emerging new technologies are shifting the paradigm and playing pivotal roles by continually expanding novel methods and materials for synthetic processes involved in the bioengineering of EVs for enhanced precision therapeutics.
Collapse
Affiliation(s)
- Mujib Ullah
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, 3155 Porter Dr., Stanford, CA, 94304, USA.
| | - Yang Qiao
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, 3155 Porter Dr., Stanford, CA, 94304, USA.,Texas A&M University College of Medicine, 8447 Riverside Pkwy, Bryan, TX, 77807, USA.,Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.,Department of Surgery, Columbia University Irving Medical Center, 177 Fort Washington Ave, New York, NY, 10032, USA
| | - Waldo Concepcion
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, 3155 Porter Dr., Stanford, CA, 94304, USA
| | - Avnesh S Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, 3155 Porter Dr., Stanford, CA, 94304, USA
| |
Collapse
|
133
|
Stem Cell-Derived Extracellular Vesicles and Kidney Regeneration. Cells 2019; 8:cells8101240. [PMID: 31614642 PMCID: PMC6830104 DOI: 10.3390/cells8101240] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) are membranous vesicles containing active proteins, lipids, and different types of genetic material such as miRNAs, mRNAs, and DNAs related to the characteristics of the originating cell. They possess a distinctive capacity to communicate over long distances. EVs have been involved in the modulation of several pathophysiological conditions and, more importantly, stem cell-derived EVs appear as a new promising therapeutic option. In fact, several reports provide convincing evidence of the regenerative potential of EVs released by stem cells and, in particular, mesenchymal stromal cells (MSCs) in different kidney injury models. Described mechanisms involve the reprogramming of injured cells, cell proliferation and angiogenesis, and inhibition of cell apoptosis and inflammation. Besides, the therapeutic use of MSC-EVs in clinical trials is under investigation. This review will focus on MSC-EV applications in preclinical models of acute and chronic renal damage including recent data on their use in kidney transplant conditioning. Moreover, ongoing clinical trials are described. Finally, new strategies to broaden and enhance EV therapeutic efficacy by engineering are discussed.
Collapse
|
134
|
Deng C, Xie Y, Zhang C, Ouyang B, Chen H, Lv L, Yao J, Liang X, Zhang Y, Sun X, Deng C, Liu G. Urine-Derived Stem Cells Facilitate Endogenous Spermatogenesis Restoration of Busulfan-Induced Nonobstructive Azoospermic Mice by Paracrine Exosomes. Stem Cells Dev 2019; 28:1322-1333. [PMID: 31311428 DOI: 10.1089/scd.2019.0026] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Nonobstructive azoospermia (NOA) is a severe form of male infertility, with limited effective treatments. Urine-derived stem cells (USCs) possess multipotent differentiation capacity and paracrine effects, and participate in tissue repair and regeneration. The aim of this study is to investigate whether the transplantation of USCs or USC exosomes (USC-exos) could promote endogenous spermatogenesis restoration in a busulfan-induced NOA mice model. USCs were cultured and characterized by flow cytometry. High-density USCs were cultured in a hollow fiber bioreactor for exosomes collection. USC-exos were isolated from USCs conditional media and identified by transmission electron microscopy, western blotting, and Flow NanoAnalyzer analysis. USC-exos exhibited sphere- or cup-shaped morphology with a mean diameter of 66.5 ± 16.0 nm, and expressed CD63 and CD9. USCs and USC-exos were transplanted into the interstitial space in the testes of NOA mice per the following groups: normal group; groups treated with no injection, phosphate-buffered saline (PBS), USCs or USC-exos on days 3 and 36 after busulfan administration, respectively. Thirty days after USCs and USC-exos transplantation, spermatogenesis was restored by both USCs and USC-exos in NOA mice 36 days after busulfan treatment as confirmed by immunofluorescence staining and hematoxylin and eosin staining. Moreover, spermatogenic genes (Pou5f1, Prm1, SYCP3, and DAZL) and the spermatogenic protein UCHL1 were significantly increased in both the USCs 36 and USC-exos36 groups compared with the PBS group, as demonstrated using quantitative real-time polymerase chain reaction and western blot analysis. However, the transplantation of USCs or USC-exos at day 3 after busulfan treatment did not improve spermatogenesis in NOA mice. Our study demonstrated that USCs could facilitate endogenous spermatogenesis restoration of busulfan-induced NOA mice through paracrine exosomes but could not protect the mouse testicles at the early stage of destruction caused by busulfan. This study provides a novel insight into the treatment of NOA.
Collapse
Affiliation(s)
- Cuncan Deng
- Reproductive Medicine Center, The Sixth Affiliate Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yun Xie
- Department of Andrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, China
| | - Chi Zhang
- Department of Andrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bin Ouyang
- Department of Andrology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Haicheng Chen
- Department of Andrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Linyan Lv
- Reproductive Medicine Center, The Sixth Affiliate Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiahui Yao
- Reproductive Medicine Center, The Sixth Affiliate Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoyan Liang
- Reproductive Medicine Center, The Sixth Affiliate Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina
| | - Xiangzhou Sun
- Department of Andrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chunhua Deng
- Department of Andrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Guihua Liu
- Reproductive Medicine Center, The Sixth Affiliate Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
135
|
Blood manufacturing methods affect red blood cell product characteristics and immunomodulatory activity. Blood Adv 2019; 2:2296-2306. [PMID: 30217795 DOI: 10.1182/bloodadvances.2018021931] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 07/26/2018] [Indexed: 12/14/2022] Open
Abstract
Transfusion of red cell concentrates (RCCs) is associated with increased risk of adverse outcomes that may be affected by different blood manufacturing methods and the presence of extracellular vesicles (EVs). We investigated the effect of different manufacturing methods on hemolysis, residual cells, cell-derived EVs, and immunomodulatory effects on monocyte activity. Thirty-two RCC units produced using whole blood filtration (WBF), red cell filtration (RCF), apheresis-derived (AD), and whole blood-derived (WBD) methods were examined (n = 8 per method). Residual platelet and white blood cells (WBCs) and the concentration, cell of origin, and characterization of EVs in RCC supernatants were assessed in fresh and stored supernatants. Immunomodulatory activity of RCC supernatants was assessed by quantifying monocyte cytokine production capacity in an in vitro transfusion model. RCF units yielded the lowest number of platelet and WBC-derived EVs, whereas the highest number of platelet EVs was in AD (day 5) and in WBD (day 42). The number of small EVs (<200 nm) was greater than large EVs (≥200 nm) in all tested supernatants, and the highest level of small EVs were in AD units. Immunomodulatory activity was mixed, with evidence of both inflammatory and immunosuppressive effects. Monocytes produced more inflammatory interleukin-8 after exposure to fresh WBF or expired WBD supernatants. Exposure to supernatants from AD and WBD RCC suppressed monocyte lipopolysaccharide-induced cytokine production. Manufacturing methods significantly affect RCC unit EV characteristics and are associated with an immunomodulatory effect of RCC supernatants, which may affect the quality and safety of RCCs.
Collapse
|
136
|
Kim C, Lee K. Polydiacetylene (PDA) Liposome-Based Immunosensor for the Detection of Exosomes. Biomacromolecules 2019; 20:3392-3398. [PMID: 31385692 DOI: 10.1021/acs.biomac.9b00641] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Exosomes are extracellular vesicles (EVs) that have attracted attention because of their important biological roles in intercellular communication and transportation of various biomolecules, including proteins and genetic materials. However, due to difficulties in their selective capture and detection, further application of exosomes remains challenging. To detect EVs, we fabricated a liposomal biosensor based on polydiacetylene (PDA), a conjugate polymer that has been widely used in sensing applications derived from its unique optical properties. To confer selectivity and sensitivity to the sensory material, antibodies targeting CD63, a membrane protein exclusively found in exosomes, were attached to the PDA liposomes and phospholipid molecules were incorporated into the PDA vesicles. Signal analysis derived from PDA liposomes for exosome detection and quantification was performed by observing colorimetric changes triggered by the ligand-receptor interaction of PDA vesicles. Visual, UV-visible, and fluorescence spectroscopic methods were used to obtain signals from the PDA lipid immunosensor, which achieved a detection limit of 3 × 108 vesicles/mL, the minimum concentration that can be used in practical applications. The strategies used in the system have the potential to expand into the field of dealing with exosomes.
Collapse
Affiliation(s)
- Changheon Kim
- Program in Nanoscience and Technology, Graduate School of Convergence Science and Technology , Seoul National University , Seoul 08826 , Republic of Korea
| | - Kangwon Lee
- Program in Nanoscience and Technology, Graduate School of Convergence Science and Technology , Seoul National University , Seoul 08826 , Republic of Korea
| |
Collapse
|
137
|
Biomaterial-based delivery systems of nucleic acid for regenerative research and regenerative therapy. Regen Ther 2019; 11:123-130. [PMID: 31338391 PMCID: PMC6626072 DOI: 10.1016/j.reth.2019.06.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/10/2019] [Accepted: 06/25/2019] [Indexed: 12/22/2022] Open
Abstract
Regenerative medicine is a new and promising medical method aiming at treating patients with defective or dysfunctional tissues by maintaining or enhancing the biological activity of cells. The development of biomaterial-based technologies, such as cell scaffolds and carriers for drug delivery system, are highly required to promote the regenerative research and regenerative therapy. Nucleic acids are one of the most feasible factors to efficiently modify the biological activity of cells. The effective and stable delivery of nucleic acids into cells is highly required to succeed in the modification. Biomaterials-based non-viral carriers or biological carriers, like exosomes, play an important role in the efficient delivery of nucleic acids. This review introduces the examples of regenerative research and regenerative therapy based on the delivery of nucleic acids with biomaterials technologies and emphasizes their importance to accomplish regenerative medicine. Modifying the activity of cells is important for regenerative medicine. Various nucleic acids regulate gene expression to modify the activity of cells. Intracellular delivery system is vital to the nucleic acids-based modification. Biomaterials are useful for the intracellular delivery of nucleic acids.
Collapse
Key Words
- Biomaterials
- CRISPR, clustered regularly interspaced short palindromic repeats
- Cas, CRISPR-associated systems
- Cell scaffold
- DDS, drug delivery system
- Drug delivery system
- ECM, extracellular matrix
- MSC, mesenchymal stem cells
- Nucleic acids
- PEG, polyethylene glycol
- PLGA, poly(d,l-lactic acid-co-glycolic acid)
- RISC, RNA-induced silencing complex
- RNAi, RNA interferince
- Regenerative research
- Regenerative therapy
- TALEN, transcription activator-like effector nuclease
- ZFN, zinc finger nucleases
- lncRNA, long non-coding RNA
- mRNA, messenger RNA
- miRNA, microRNA
- siRNA, small interfering RNA
Collapse
|
138
|
Gorgulho CM, Romagnoli GG, Bharthi R, Lotze MT. Johnny on the Spot-Chronic Inflammation Is Driven by HMGB1. Front Immunol 2019; 10:1561. [PMID: 31379812 PMCID: PMC6660267 DOI: 10.3389/fimmu.2019.01561] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 06/24/2019] [Indexed: 12/24/2022] Open
Abstract
Although much has been made of the role of HMGB1 acting as an acute damage associated molecular pattern (DAMP) molecule, prompting the response to tissue damage or injury, it is also released at sites of chronic inflammation including sites of infection, autoimmunity, and cancer. As such, the biology is distinguished from homeostasis and acute inflammation by the recruitment and persistence of myeloid derived suppressor cells, T regulatory cells, fibrosis and/or exuberant angiogenesis depending on the antecedents and the other individual inflammatory partners that HMGB1 binds and focuses, including IL-1β, CXCL12/SDF1, LPS, DNA, RNA, and sRAGE. High levels of HMGB1 released into the extracellular milieu and its persistence in the microenvironment can contribute to the pathogenesis of many if not all autoimmune disorders and is a key factor that drives inflammation further and worsens symptoms. HMGB1 is also pivotal in the maintenance of chronic inflammation and a “wound healing” type of immune response that ultimately contributes to the onset of carcinogenesis and tumor progression. Exosomes carrying HMGB1 and other instructive molecules are released and shape the response of various cells in the chronic inflammatory environment. Understanding the defining roles of REDOX, DAMPs and PAMPs, and the host response in chronic inflammation requires an alternative means for positing HMGB1's central role in limiting and focusing inflammation, distinguishing chronic from acute inflammation.
Collapse
Affiliation(s)
- Carolina M Gorgulho
- Tumor Immunology Laboratory, Department of Microbiology and Immunology, Botucatu Institute of Biosciences, São Paulo State University, Botucatu, Brazil.,DAMP Laboratory, Department of Surgery, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Graziela G Romagnoli
- Tumor Immunology Laboratory, Department of Microbiology and Immunology, Botucatu Institute of Biosciences, São Paulo State University, Botucatu, Brazil
| | - Rosh Bharthi
- DAMP Laboratory, Department of Surgery, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michael T Lotze
- DAMP Laboratory, Department of Surgery, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Immunology, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
139
|
Tan GZ, Zhou Y. Electrospinning of biomimetic fibrous scaffolds for tissue engineering: a review. INT J POLYM MATER PO 2019. [DOI: 10.1080/00914037.2019.1636248] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- George Z. Tan
- Department of Industrial, Manufacturing and Systems Engineering, Texas Tech University, Lubbock, TX, USA
| | - Yingge Zhou
- Department of Industrial, Manufacturing and Systems Engineering, Texas Tech University, Lubbock, TX, USA
| |
Collapse
|
140
|
Dinda S, Das PK. Metal Ion (Fe2+ and Co2+) Induced Morphological Transformation of Self-Aggregates of Cholesterol-Tethered Bipyridine Amphiphiles: Selective Cancer Cell Killing by Pro-Drug Activation. ACS APPLIED BIO MATERIALS 2019; 2:3737-3747. [DOI: 10.1021/acsabm.9b00340] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Soumik Dinda
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700 032, India
| | - Prasanta Kumar Das
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700 032, India
| |
Collapse
|
141
|
Surface-structured bacterial cellulose loaded with hUSCs accelerate skin wound healing by promoting angiogenesis in rats. Biochem Biophys Res Commun 2019; 516:1167-1174. [PMID: 31284954 DOI: 10.1016/j.bbrc.2019.06.161] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 06/29/2019] [Indexed: 01/26/2023]
Abstract
Promotion of wound healing is one of the most important fields in clinical medical research. This study aimed to evaluate the potential use of a new surface-structured bacterial cellulose(S-BC) biomaterial with human urine-derived stem cells (hUSCs) for wound healing. In vitro, EA.hy926 were inoculated on structured/non-structured bacterial cellulose, and the growth of EA.hy926 on bacterial cellulose in medium with/without conditioned medium of the hUSCs were observed to explore the effect of bacterial cellulose's surface structure and hUSCs-CM on vascular endothelial cell growth. In vivo, we covered wound surface with various BC materials and/or injected the hUSCs into the wound site on group BC, group S-BC, group hUSCs, group BC + hUSCs, group S-BC + hUSCs to evaluate the effect of S-BC and hUSCs on wound healing in rat full-thickness skin defect model. In vitro study, surface structure of S-BC could promote the growth and survival of EA.hy926, and the hUSCs-CM could further promote the proliferation of EA.hy926 on S-BC. In vivo study, wound healing rate of the group BC, group S-BC, group hUSCs was significantly accelerated, accompanied by faster re-epithelialization, collagen production and neovascularization than control group. It is note worthy that the effect of S-BC on wound healing was better than BC, the effect of S-BC + hUSCs on wound healing was better than BC + hUSCs. Moreover, the effect of S-BC combined with hUSCs on wound is better than treated with S-BC or hUSCs alone. All the findings suggest that the combination of S-BC and hUSCs could facilitate skin wound healing by promoting angiogenesis. This combination of the role of stem cells and biomaterial surface structures may provide a new way to address clinical wound healing problems.
Collapse
|
142
|
Hu H, Jiang C, Li R, Zhao J. Comparison of endothelial cell- and endothelial progenitor cell-derived exosomes in promoting vascular endothelial cell repair. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:2793-2800. [PMID: 31934115 PMCID: PMC6949553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/24/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Timely endothelial repair after intervention-associated vascular injury is critical to prevent restenosis and thrombosis. Compared to living cell therapies, exosomes may be a better alternative. Thus, we aimed to compare the role of exosomes derived from human umbilical vein endothelial cells (HUVECs) versus those derived from endothelial progenitor cells (EPCs) in vascular endothelial repair. MATERIAL/METHODS Exosomes secreted from HUVECs and EPCs were isolated and characterized respectively. In vitro, the effects of the two types of exosomes on migration and proliferation of endothelial cells were studied. In vivo, rats were systemically treated with the two exosome groups respectively after carotid artery endothelial injury induced by a balloon. The efficacy in promoting re-endothelialization was measured by Evans blue dye and histological examination. RESULTS Both types of exosomes, sized from 30 nm to 100 nm, had sphere-shaped or cupped morphology, and expressed CD63, CD9, and CD81; but the total yield of exosomes (particles/mL) based on number of cells, was 4.2 times higher from EPCs than HUVECs. Compared with control treatment, both exosome treated groups manifested significant enhancement of migration and proliferation in vitro and vascular recovery at an early stage in vivo. The two exosome-treated groups, however, did not statistically significantly differ. CONCLUSION In conclusion, our results indicated that exosomes derived from HUVECs and EPCs had similar morphology, size distributions and characteristics, but those derived from EPCs were more abundant with comparable biologic activity. Therefore, EPCs may be a robust source of exosomes to promote vascular repair.
Collapse
Affiliation(s)
- Hui Hu
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai, China
| | - Chunyu Jiang
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai, China
| | - Ruiting Li
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai, China
| | - Jungong Zhao
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai, China
| |
Collapse
|
143
|
Extracellular vesicles from human urine-derived stem cells prevent osteoporosis by transferring CTHRC1 and OPG. Bone Res 2019; 7:18. [PMID: 31263627 PMCID: PMC6594995 DOI: 10.1038/s41413-019-0056-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 04/19/2019] [Accepted: 05/04/2019] [Indexed: 12/19/2022] Open
Abstract
Osteoporosis is a debilitating bone disease affecting millions of people. Here, we used human urine-derived stem cells (USCs), which were noninvasively harvested from unlimited and easily available urine, as a “factory” to obtain extracellular vesicles (USC-EVs) and demonstrated that the systemic injection of USC-EVs effectively alleviates bone loss and maintains bone strength in osteoporotic mice by enhancing osteoblastic bone formation and suppressing osteoclastic bone resorption. More importantly, the anti-osteoporotic properties of USC-EVs are not notably disrupted by the age, gender, or health condition (with or without osteoporosis) of the USC donor. Mechanistic studies determined that collagen triple-helix repeat containing 1 (CTHRC1) and osteoprotegerin (OPG) proteins are enriched in USC-EVs and required for USC-EV-induced pro-osteogenic and anti-osteoclastic effects. Our results suggest that autologous USC-EVs represent a promising novel therapeutic agent for osteoporosis by promoting osteogenesis and inhibiting osteoclastogenesis by transferring CTHRC1 and OPG.
Collapse
|
144
|
Ramazzotti G, Ratti S, Fiume R, Follo MY, Billi AM, Rusciano I, Owusu Obeng E, Manzoli L, Cocco L, Faenza I. Phosphoinositide 3 Kinase Signaling in Human Stem Cells from Reprogramming to Differentiation: A Tale in Cytoplasmic and Nuclear Compartments. Int J Mol Sci 2019; 20:ijms20082026. [PMID: 31022972 PMCID: PMC6514809 DOI: 10.3390/ijms20082026] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 04/19/2019] [Accepted: 04/21/2019] [Indexed: 12/11/2022] Open
Abstract
Stem cells are undifferentiated cells that can give rise to several different cell types and can self-renew. Given their ability to differentiate into different lineages, stem cells retain huge therapeutic potential for regenerative medicine. Therefore, the understanding of the signaling pathways involved in stem cell pluripotency maintenance and differentiation has a paramount importance in order to understand these biological processes and to develop therapeutic strategies. In this review, we focus on phosphoinositide 3 kinase (PI3K) since its signaling pathway regulates many cellular processes, such as cell growth, proliferation, survival, and cellular transformation. Precisely, in human stem cells, the PI3K cascade is involved in different processes from pluripotency and induced pluripotent stem cell (iPSC) reprogramming to mesenchymal and oral mesenchymal differentiation, through different and interconnected mechanisms.
Collapse
Affiliation(s)
- Giulia Ramazzotti
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Stefano Ratti
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Roberta Fiume
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Matilde Yung Follo
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Anna Maria Billi
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Isabella Rusciano
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Eric Owusu Obeng
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Lucia Manzoli
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Lucio Cocco
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Irene Faenza
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| |
Collapse
|
145
|
Kwack MH, Seo CH, Gangadaran P, Ahn B, Kim MK, Kim JC, Sung YK. Exosomes derived from human dermal papilla cells promote hair growth in cultured human hair follicles and augment the hair‐inductive capacity of cultured dermal papilla spheres. Exp Dermatol 2019; 28:854-857. [PMID: 30924959 DOI: 10.1111/exd.13927] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 03/04/2019] [Accepted: 03/18/2019] [Indexed: 01/13/2023]
Affiliation(s)
- Mi H. Kwack
- Department of Immunology School of Medicine Kyungpook National University Daegu Korea
| | - Chang H. Seo
- New Drug Development Center Daegu‐Gyeongbuk Medical Innovation Foundation Daegu Korea
| | - Prakash Gangadaran
- Department of Nuclear Medicine School of Medicine Kyungpook National University Daegu Korea
| | - Byeong‐Cheol Ahn
- Department of Nuclear Medicine School of Medicine Kyungpook National University Daegu Korea
| | - Moon K. Kim
- Department of Immunology School of Medicine Kyungpook National University Daegu Korea
- Hair Transplantation Center Kyungpook National University Hospital Daegu Korea
| | - Jung C. Kim
- Department of Immunology School of Medicine Kyungpook National University Daegu Korea
- Hair Transplantation Center Kyungpook National University Hospital Daegu Korea
| | - Young K. Sung
- Department of Immunology School of Medicine Kyungpook National University Daegu Korea
| |
Collapse
|
146
|
Song Y, Zhang C, Zhang J, Jiao Z, Dong N, Wang G, Wang Z, Wang L. Localized injection of miRNA-21-enriched extracellular vesicles effectively restores cardiac function after myocardial infarction. Am J Cancer Res 2019; 9:2346-2360. [PMID: 31149048 PMCID: PMC6531307 DOI: 10.7150/thno.29945] [Citation(s) in RCA: 145] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 02/14/2019] [Indexed: 12/12/2022] Open
Abstract
Myocardial infarction (MI), a main cause of heart failure, leads to irreversible cardiomyocytes loss and cardiac function impairment. Current clinical treatments for MI are largely ineffective as they mostly aim to alleviate symptoms rather than repairing the injured myocardium. Thus, development of more effective therapies is compelling. This study aims to investigate whether the extracellular vesicles (EVs) carrying specific anti-apoptotic miRNA can be efficiently internalized into myocardium to achieve desired therapeutic outcomes. Methods: EVs were isolated from HEK293T cells overexpressing miRNA-21 (miR21-EVs) and identified. The RNase resistant rate of miR21-EVs was calculated by real-time PCR and compared with liposomes and polyethylenimine (PEI). Confocal laser scanning microscopy was used for visualizing the cellular internalization of miR21-EVs in primary cultured mouse neonatal cardiomyocytes (CMs), H9c2 rat cardiomyoblasts, and human umbilical vein endothelial cells (HUVECs). The effect of miR21-EVs on the expression of PDCD4, a pro-apoptotic protein that plays an important role in regulating myocardial apoptosis, was also evaluated in these three cell types by real-time PCR and Western blot analysis. In vivo, miR21-EVs was directly injected into the infarct zone following ligation of the left anterior descending of coronary artery in mice. The miR21-EVs distribution and blood vessel (capillary and arteriole) density were evaluated by immunofluorescence staining. Fluorescence in situ hybridization of miRNA-21 was also carried out to confirm the miR21-EVs distribution in vitro and in vivo. The protein level of PDCD4 in myocardium was assessed by immunohistochemical staining. The anti-apoptotic effect of miR21-EVs in cardiomyocytes and endothelial cells were measured using TUNEL staining. Four weeks after injection, the cardiac histological and functional recovery was evaluated by histochemistry staining and echocardiography, respectively. Results: In contrast to liposomes and PEI, EVs significantly inhibited miRNA-21 degradation. MiR21-EVs efficiently delivered miRNA-21 into cardiomyocytes and endothelial cells within 4 hours. Exogenous miRNA-21 in turn significantly reduced PDCD4 expression and attenuated cell apoptosis in vitro. Consistently and importantly, in a preclinical MI animal model, miRNA-21-loaded EVs effectively sent miRNA-21 into cardiomyocytes and endothelial cells, drastically inhibited cell apoptosis and led to significant cardiac function improvement. Conclusion: Our results suggest the cell-derived, genetically engineered EVs may be used therapeutically for the delivery of miRNAs for the rescue of MI and may benefit patients in the future.
Collapse
|
147
|
Yin H, Chen CY, Liu YW, Tan YJ, Deng ZL, Yang F, Huang FY, Wen C, Rao SS, Luo MJ, Hu XK, Liu ZZ, Wang ZX, Cao J, Liu HM, Liu JH, Yue T, Tang SY, Xie H. Synechococcus elongatus PCC7942 secretes extracellular vesicles to accelerate cutaneous wound healing by promoting angiogenesis. Am J Cancer Res 2019; 9:2678-2693. [PMID: 31131061 PMCID: PMC6525994 DOI: 10.7150/thno.31884] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 02/17/2019] [Indexed: 01/08/2023] Open
Abstract
Poor wound healing affects millions of people worldwide each year and needs better therapeutic strategies. Synechococcus elongatus PCC 7942 is a naturally occurring photoautotrophic cyanobacterium that can be easily obtained and large-scale expanded. Here, we investigated the therapeutic efficacy of this cyanobacterium in a mouse model of acute burn injury and whether the secretion of extracellular vesicles (EVs), important mediators of cell paracrine activity, is a key mechanism of the cyanobacterium-induced regulation of wound healing. Methods: The effects of Synechococcus elongatus PCC 7942 on burn wound healing in mice under light or dark conditions were evaluated by measuring wound closure rates, histological and immunofluorescence analyses. A series of assays in vivo and in vitro were conducted to assess the impact of the cyanobacterium on angiogenesis. GW4869 was used to interfere with the secretion of EVs by the cyanobacterium and the abilities of the GW4869-pretreated and untreated Synechococcus elongatus PCC 7942 to regulate endothelial angiogenesis were compared. The direct effects of the cyanobacterium-derived EVs (S. elongatus-EVs) on angiogenesis, wound healing and expressions of a class of pro-inflammatory factors that have regulatory roles in wound healing were also examined. Results: Synechococcus elongatus PCC 7942 treatment under light and dark conditions both significantly promoted angiogenesis and burn wound repair in mice. In vitro, the cyanobacterium enhanced angiogenic activities of endothelial cells, but the effects were markedly blocked by GW4869 pretreatment. S. elongatus-EVs were capable of augmenting endothelial angiogenesis in vitro, and stimulating new blood vessel formation and burn wound healing in mice. The expression of interleukin 6 (IL-6), which has an essential role in angiogenesis during skin wound repair, was induced in wound tissues and wound healing-related cells by S. elongatus-EVs and Synechococcus elongatus PCC 7942. Conclusion: Synechococcus elongatus PCC 7942 has the potential as a promising strategy for therapeutic angiogenesis and wound healing primarily by the delivery of functional EVs, not by its photosynthetic activity. The promotion of IL-6 expression may be a mechanism of the cyanobacterium and its EVs-induced pro-angiogenic and -wound healing effects.
Collapse
|
148
|
Intra-Vitreal Administration of Microvesicles Derived from Human Adipose-Derived Multipotent Stromal Cells Improves Retinal Functionality in Dogs with Retinal Degeneration. J Clin Med 2019; 8:jcm8040510. [PMID: 31013950 PMCID: PMC6518198 DOI: 10.3390/jcm8040510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/08/2019] [Accepted: 04/10/2019] [Indexed: 12/28/2022] Open
Abstract
This study was designed to determine the influence of microvesicles (MVs) derived from multipotent stromal cells isolated from human adipose tissue (hASCs) on retinal functionality in dogs with various types of retinal degeneration. The biological properties of hASC-MVs were first determined using an in vitro model of retinal Muller-like cells (CaMLCs). The in vitro assays included analysis of hASC-MVs influence on cell viability and metabolism. Brain-derived neurotrophic factor (BDNF) expression was also determined. Evaluation of the hASC-MVs was performed under normal and oxidative stress conditions. Preliminary clinical studies were performed on ten dogs with retinal degeneration. The clinical studies included behavioral tests, fundoscopy and electroretinography before and after hASC-MVs intra-vitreal injection. The in vitro study showed that CaMLCs treated with hASC-MVs were characterized by improved viability and mitochondrial potential, both under normal and oxidative stress conditions. Additionally, hASC-MVs under oxidative stress conditions reduced the number of senescence-associated markers, correlating with the increased expression of BDNF. The preliminary clinical study showed that the intra-vitreal administration of hASC-MVs significantly improved the dogs’ general behavior and tracking ability. Furthermore, fundoscopy demonstrated that the retinal blood vessels appeared to be less attenuated, and electroretinography using HMsERG demonstrated an increase in a- and b-wave amplitude after treatment. These results shed promising light on the application of cell-free therapies in veterinary medicine for retinal degenerative disorders treatment.
Collapse
|
149
|
Lange-Consiglio A, Stucchi L, Zucca E, Lavoie JP, Cremonesi F, Ferrucci F. Insights into animal models for cell-based therapies in translational studies of lung diseases: Is the horse with naturally occurring asthma the right choice? Cytotherapy 2019; 21:525-534. [PMID: 30929991 DOI: 10.1016/j.jcyt.2019.02.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 02/21/2019] [Accepted: 02/25/2019] [Indexed: 12/15/2022]
Abstract
Human asthma is a widespread disease associated with chronic inflammation of the airways, leading to loss of quality of life, disability and death. Corticosteroid administration is the mainstream treatment for asthmatic patients. Corticosteroids reduce airway obstruction and improve quality of life, although symptoms persist despite treatment in many patients. Moreover, available therapies failed to reverse the lung pathology present in asthma. Animal models, mostly rats and mice, in which the disease is experimentally induced, have been studied to identify new therapeutic targets for human asthma. Alternative animal models could include horses in which naturally occurring asthma could represent an important step to test therapies, potentially designed around mouse studies, before being translated to human testing. Horses naturally suffer from asthma, which has striking parallels with human asthma. Severe equine asthma (SEA) is characterized by reversible bronchospasms and neutrophil accumulation in the lungs immunologically mediated mainly by Th2. Moreover, the pulmonary remodelling that occurs in SEA closely resembles that of human asthma, making the equine model unique for investigation of tissue repair and new therapies. Cell therapy, consisting on mesenchymal stromal cells (MSCs) and derivatives (conditioned medium and extracellular vesicles), could represent a novel therapeutic contribution for tissue regeneration. Cell therapy may prove advantageous over conventional therapy in that it may repair or regenerate the site of injury and reduce the reaction to allergens, rather than simply modulating the inflammatory process.
Collapse
Affiliation(s)
- Anna Lange-Consiglio
- Department of Veterinary Medicine, Università degli Studi di Milano, Milano, Italy; Reproduction Unit, Centro Clinico-Veterinario e Zootecnico Sperimentale di Ateneo, Università degli Studi di Milano, Lodi, Italy.
| | - Luca Stucchi
- Equine Medicine Unit, Department of Health, Animal Science and Food Safety, Università degli Studi di Milano, Milano, Italy
| | - Enrica Zucca
- Equine Medicine Unit, Department of Health, Animal Science and Food Safety, Università degli Studi di Milano, Milano, Italy
| | - Jean Pierre Lavoie
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Université de Montréal, St-Hyacinthe, QC, Canada
| | - Fausto Cremonesi
- Department of Veterinary Medicine, Università degli Studi di Milano, Milano, Italy; Reproduction Unit, Centro Clinico-Veterinario e Zootecnico Sperimentale di Ateneo, Università degli Studi di Milano, Lodi, Italy
| | - Francesco Ferrucci
- Equine Medicine Unit, Department of Health, Animal Science and Food Safety, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
150
|
Queckbörner S, Davies LC, von Grothusen C, Santamaria X, Simón C, Gemzell-Danielsson K. Cellular therapies for the endometrium: An update. Acta Obstet Gynecol Scand 2019; 98:672-677. [PMID: 30815850 DOI: 10.1111/aogs.13598] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/22/2019] [Indexed: 12/14/2022]
Abstract
An update on the current state of endometrial cell therapies in terms of cell types, mechanisms of action, delivery, safety, regulatory frameworks and future perspectives. This review focuses on clinical trials using angiogenesis-promoting therapies and stromal therapies piloted in the last 10 years for alleviating Asherman's syndrome and long-term infertility. All studies present promising preliminary results, indicating increased endometrial thickness and resumed menstruation. Further characterization of individual cell products, their mode of action and larger clinical trials will be essential to establishing cell therapy as a viable option for the treatment of infertility and fertility preservation.
Collapse
Affiliation(s)
- Suzanna Queckbörner
- Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet, and Karolinska University Hospital, Solna, Sweden
| | - Lindsay C Davies
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Carolina von Grothusen
- Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet, and Karolinska University Hospital, Solna, Sweden
| | - Xavier Santamaria
- Department of Obstetrics and Gynecology, University of Valencia/INCLIVA, Valencia, Spain
| | - Carlos Simón
- Department of Obstetrics and Gynecology, University of Valencia/INCLIVA, Valencia, Spain
| | - Kristina Gemzell-Danielsson
- Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet, and Karolinska University Hospital, Solna, Sweden
| |
Collapse
|