1
|
Sun Z, Fukui M, Taketani S, Kako A, Kunieda S, Kakudo N. Predominant control of PDGF/PDGF receptor signaling in the migration and proliferation of human adipose‑derived stem cells under culture conditions with a combination of growth factors. Exp Ther Med 2024; 27:156. [PMID: 38476902 PMCID: PMC10928992 DOI: 10.3892/etm.2024.12444] [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: 09/25/2023] [Accepted: 01/19/2024] [Indexed: 03/14/2024] Open
Abstract
Human adipose-derived stem cells (hASCs) play important roles in regenerative medicine and tissue engineering. However, their clinical applications are limited because of their instability during cell culture. Platelet lysates (PLTs) contain large amounts of growth factors that are useful for manufacturing cellular products. Platelet-derived growth factor (PDGF) is a major growth factor in PLTs and a potent mitogen in hASCs. To optimize growth conditions, the effects of a combination of growth factors on the promotion of hASC proliferation were investigated. Moreover, PDGF-BB combined with vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF) markedly enhanced the viability of hASCs compared with the effects of PDGF-BB alone. Neither VEGF nor HGF had any effect alone. All growth factor receptor inhibitors inhibited cell proliferation. Wound healing assays revealed that VEGF and HGF stimulated PDGF-dependent cell migration. The effects of these growth factors on the activation of their cognate receptors and signaling enzymes were assessed using immunoblotting. Phosphorylation of PDGF receptor (PDGFR)β, VEGF receptor (VEGFR)2 and MET proto-oncogene and receptor tyrosine kinase was induced by PDGF-BB treatment, and was further increased by treatment with PDGF-BB/VEGF and PDGF-BB/HGF. The levels of phospho-ERK1/2 and phospho-p38MAPK were increased by these treatments in parallel. Furthermore, the expression levels of SRY-box transcription factor 2 and peroxisome proliferator-activated receptor g were increased in PDGF-BB-treated cells, and PDGF-BB played a dominant role in spheroid formation. The findings of the present study highlighted that PDGF/PDGFR signaling played a predominant role in the proliferation and migration of hASCs, and suggested that PDGF was responsible for the efficacy of other growth factors when hASCs were cultured with PLTs.
Collapse
Affiliation(s)
- Zhongxin Sun
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Michika Fukui
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Shigeru Taketani
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Ayako Kako
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Sakurako Kunieda
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Natsuko Kakudo
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| |
Collapse
|
2
|
Hu S, Yang B, Shu S, He X, Sang H, Fan X, Zhang H. Targeting Pericytes for Functional Recovery in Ischemic Stroke. Neuromolecular Med 2023; 25:457-470. [PMID: 37166748 DOI: 10.1007/s12017-023-08748-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/03/2023] [Indexed: 05/12/2023]
Abstract
Pericytes surrounding endothelial cells in the capillaries are emerging as an attractive cell resource, which can show a large variety of functions in ischemic stroke, including preservation of the blood-brain barrier, regulation of immune function, and support for cerebral vasculature. These functions have been fully elucidated in previous studies. However, in recent years, increasing evidence has shown that pericytes play an important role in neurological recovery after ischemic stroke due to their regenerative function which can be summarized in two aspects according to current discoveries, one is that pericytes are thought to be multipotential themselves, and the other is that pericytes can promote the differentiation of oligodendrocyte progenitor cells (OPCs). Considering the neuroprotective treatment for stroke has not been much progressed in recent years, new therapies targeting pericytes may be a future direction. Here, we will review the beneficial effects of pericytes in ischemic stroke from two directions: the barrier and vascular functions and the regenerative functions of pericytes.
Collapse
Affiliation(s)
- Shuqi Hu
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Bingjie Yang
- Department of Neurology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Song Shu
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Xudong He
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Hongfei Sang
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Xuemei Fan
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Hao Zhang
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China.
- Department of Neurology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|
3
|
Fu J, Liang H, Yuan P, Wei Z, Zhong P. Brain pericyte biology: from physiopathological mechanisms to potential therapeutic applications in ischemic stroke. Front Cell Neurosci 2023; 17:1267785. [PMID: 37780206 PMCID: PMC10536258 DOI: 10.3389/fncel.2023.1267785] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 08/30/2023] [Indexed: 10/03/2023] Open
Abstract
Pericytes play an indispensable role in various organs and biological processes, such as promoting angiogenesis, regulating microvascular blood flow, and participating in immune responses. Therefore, in this review, we will first introduce the discovery and development of pericytes, identification methods and functional characteristics, then focus on brain pericytes, on the one hand, to summarize the functions of brain pericytes under physiological conditions, mainly discussing from the aspects of stem cell characteristics, contractile characteristics and paracrine characteristics; on the other hand, to summarize the role of brain pericytes under pathological conditions, mainly taking ischemic stroke as an example. Finally, we will discuss and analyze the application and development of pericytes as therapeutic targets, providing the research basis and direction for future microvascular diseases, especially ischemic stroke treatment.
Collapse
Affiliation(s)
- Jiaqi Fu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- Department of Neurology, Shidong Hospital, Yangpu District, Shanghai, China
| | - Huazheng Liang
- Monash Suzhou Research Institute, Suzhou, Jiangsu, China
| | - Ping Yuan
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhenyu Wei
- Department of Neurology, Shidong Hospital, Yangpu District, Shanghai, China
| | - Ping Zhong
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- Department of Neurology, Shidong Hospital, Yangpu District, Shanghai, China
| |
Collapse
|
4
|
Yamaguchi S, Yoshida M, Horie N, Satoh K, Fukuda Y, Ishizaka S, Ogawa K, Morofuji Y, Hiu T, Izumo T, Kawakami S, Nishida N, Matsuo T. Stem Cell Therapy for Acute/Subacute Ischemic Stroke with a Focus on Intraarterial Stem Cell Transplantation: From Basic Research to Clinical Trials. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 10:bioengineering10010033. [PMID: 36671605 PMCID: PMC9854681 DOI: 10.3390/bioengineering10010033] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022]
Abstract
Stem cell therapy for ischemic stroke holds great promise for the treatment of neurological impairment and has moved from the laboratory into early clinical trials. The mechanism of action of stem cell therapy includes the bystander effect and cell replacement. The bystander effect plays an important role in the acute to subacute phase, and cell replacement plays an important role in the subacute to chronic phase. Intraarterial (IA) transplantation is less invasive than intraparenchymal transplantation and can provide more cells in the affected brain region than intravenous transplantation. However, transplanted cell migration was reported to be insufficient, and few transplanted cells were retained in the brain for an extended period. Therefore, the bystander effect was considered the main mechanism of action of IA stem cell transplantation. In most clinical trials, IA transplantation was performed during the acute and subacute phases. Although clinical trials of IA transplantation demonstrated safety, they did not demonstrate satisfactory efficacy in improving patient outcomes. To increase efficacy, increased migration of transplanted cells and production of long surviving and effective stem cells would be crucial. Given the lack of knowledge on this subject, we review and summarize the mechanisms of action of transplanted stem cells and recent advancements in preclinical and clinical studies to provide information and guidance for further advancement of acute/subacute phase IA stem cell transplantation therapy for ischemic stroke.
Collapse
Affiliation(s)
- Susumu Yamaguchi
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
- Department of Neurosurgery, Sasebo General Hospital, Nagasaki 857-8511, Japan
- Correspondence: ; Tel.: +81-095-819-7375
| | - Michiharu Yoshida
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
- Department of Neurosurgery, Sasebo General Hospital, Nagasaki 857-8511, Japan
| | - Nobutaka Horie
- Department of Neurosurgery, Hiroshima University, Hiroshima 734-8551, Japan
| | - Katsuya Satoh
- Department of Occupational Therapy Sciences, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| | - Yuutaka Fukuda
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| | - Shunsuke Ishizaka
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| | - Koki Ogawa
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan
| | - Yoichi Morofuji
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| | - Takeshi Hiu
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| | - Tsuyoshi Izumo
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| | - Shigeru Kawakami
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan
| | - Noriyuki Nishida
- Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan
| | - Takayuki Matsuo
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| |
Collapse
|
5
|
Zhang Y, Warden AR, Ahmad KZ, Liu Y, He X, Zheng M, Huo X, Zhi X, Ke Y, Li H, Yan S, Su W, Cai D, Ding X. Single-Cell Microwell Platform Reveals Circulating Neural Cells as a Clinical Indicator for Patients with Blood-Brain Barrier Breakdown. RESEARCH 2021; 2021:9873545. [PMID: 34327332 PMCID: PMC8285994 DOI: 10.34133/2021/9873545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 06/01/2021] [Indexed: 12/21/2022]
Abstract
Central nervous system diseases commonly occur with the destruction of the blood-brain barrier. As a primary cause of morbidity and mortality, stroke remains unpredictable and lacks cellular biomarkers that accurately quantify its occurrence and development. Here, we identify NeuN+/CD45−/DAPI+ phenotype nonblood cells in the peripheral blood of mice subjected to middle cerebral artery occlusion (MCAO) and stroke patients. Since NeuN is a specific marker of neural cells, we term these newly identified cells as circulating neural cells (CNCs). We find that the enumeration of CNCs in the blood is significantly associated with the severity of brain damage in MCAO mice (p < 0.05). Meanwhile, the number of CNCs is significantly higher in stroke patients than in negative subjects (p < 0.0001). These findings suggest that the amount of CNCs in circulation may serve as a clinical indicator for the real-time prognosis and progression monitor of the occurrence and development of ischemic stroke and other nervous system disease.
Collapse
Affiliation(s)
- Yu Zhang
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Antony R Warden
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Khan Zara Ahmad
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Yanlei Liu
- Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Xijun He
- Department of Neurosurgery, Wenling Hospital Affiliated to Wenzhou Medical University, Chuan'an Nan Road, Chengxi Subdistrict, Wenling, 317500 Zhejiang, China
| | - Minqiao Zheng
- Central Laboratory, Wenling Hospital Affiliated to Wenzhou Medical University, Chuan'an Nan Road, Chengxi Subdistrict, Wenling, 317500 Zhejiang, China
| | - Xinlong Huo
- Department of Neurology, Wenling Hospital Affiliated to Wenzhou Medical University, Chuan'an Nan Road, Chengxi Subdistrict, Wenling, 317500 Zhejiang, China
| | - Xiao Zhi
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Yuqing Ke
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Hongxia Li
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Sijia Yan
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Wenqiong Su
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Deng Cai
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Road, Shanghai 200030, China
| | - Xianting Ding
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| |
Collapse
|
6
|
Okada T, Suzuki H, Travis ZD, Zhang JH. The Stroke-Induced Blood-Brain Barrier Disruption: Current Progress of Inspection Technique, Mechanism, and Therapeutic Target. Curr Neuropharmacol 2020; 18:1187-1212. [PMID: 32484111 PMCID: PMC7770643 DOI: 10.2174/1570159x18666200528143301] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/23/2020] [Accepted: 05/23/2020] [Indexed: 02/07/2023] Open
Abstract
Stroke is one of the leading causes of mortality and morbidity worldwide. The blood-brain barrier (BBB) is a characteristic structure of microvessel within the brain. Under normal physiological conditions, the BBB plays a role in the prevention of harmful substances entering into the brain parenchyma within the central nervous system. However, stroke stimuli induce the breakdown of BBB leading to the influx of cytotoxic substances, vasogenic brain edema, and hemorrhagic transformation. Therefore, BBB disruption is a major complication, which needs to be addressed in order to improve clinical outcomes in stroke. In this review, we first discuss the structure and function of the BBB. Next, we discuss the progress of the techniques utilized to study BBB breakdown in in-vitro and in-vivo studies, along with biomarkers and imaging techniques in clinical settings. Lastly, we highlight the mechanisms of stroke-induced neuroinflammation and apoptotic process of endothelial cells causing BBB breakdown, and the potential therapeutic targets to protect BBB integrity after stroke. Secondary products arising from stroke-induced tissue damage provide transformation of myeloid cells such as microglia and macrophages to pro-inflammatory phenotype followed by further BBB disruption via neuroinflammation and apoptosis of endothelial cells. In contrast, these myeloid cells are also polarized to anti-inflammatory phenotype, repairing compromised BBB. Therefore, therapeutic strategies to induce anti-inflammatory phenotypes of the myeloid cells may protect BBB in order to improve clinical outcomes of stroke patients.
Collapse
Affiliation(s)
- Takeshi Okada
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA, Risley Hall, Room 219,
11041 Campus St, Loma Linda, CA 92354, USA,Department of Neurosurgery, Mie University Graduate School of Medicine, Mie, Japan, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Hidenori Suzuki
- Department of Neurosurgery, Mie University Graduate School of Medicine, Mie, Japan, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Zachary D Travis
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA, Risley Hall, Room 219,
11041 Campus St, Loma Linda, CA 92354, USA,Department of Earth and Biological Sciences, Loma Linda University, Loma Linda, CA, USA , Risley Hall, Room 219, 11041 Campus St, Loma Linda, CA 92354, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA, Risley Hall, Room 219,
11041 Campus St, Loma Linda, CA 92354, USA,Department of Anesthesiology, Loma Linda University, Loma Linda, CA, USA, Risley Hall, Room 219, 11041 Campus St, Loma Linda, CA 92354, USA,Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA, Risley Hall, Room 219, 11041 Campus St, Loma Linda, CA 92354, USA
| |
Collapse
|
7
|
Caporarello N, D’Angeli F, Cambria MT, Candido S, Giallongo C, Salmeri M, Lombardo C, Longo A, Giurdanella G, Anfuso CD, Lupo G. Pericytes in Microvessels: From "Mural" Function to Brain and Retina Regeneration. Int J Mol Sci 2019; 20:ijms20246351. [PMID: 31861092 PMCID: PMC6940987 DOI: 10.3390/ijms20246351] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/13/2019] [Accepted: 12/14/2019] [Indexed: 12/13/2022] Open
Abstract
Pericytes are branched cells located in the wall of capillary blood vessels that are found throughout the body, embedded within the microvascular basement membrane and wrapping endothelial cells, with which they establish a strong physical contact. Pericytes regulate angiogenesis, vessel stabilization, and contribute to the formation of both the blood-brain and blood-retina barriers by Angiopoietin-1/Tie-2, platelet derived growth factor (PDGF) and transforming growth factor (TGF) signaling pathways, regulating pericyte-endothelial cell communication. Human pericytes that have been cultured for a long period give rise to multilineage progenitor cells and exhibit mesenchymal stem cell (MSC) features. We focused our attention on the roles of pericytes in brain and ocular diseases. In particular, pericyte involvement in brain ischemia, brain tumors, diabetic retinopathy, and uveal melanoma is described. Several molecules, such as adenosine and nitric oxide, are responsible for pericyte shrinkage during ischemia-reperfusion. Anti-inflammatory molecules, such as IL-10, TGFβ, and MHC-II, which are increased in glioblastoma-activated pericytes, are responsible for tumor growth. As regards the eye, pericytes play a role not only in ocular vessel stabilization, but also as a stem cell niche that contributes to regenerative processes in diabetic retinopathy. Moreover, pericytes participate in melanoma cell extravasation and the genetic ablation of the PDGF receptor reduces the number of pericytes and aberrant tumor microvessel formation with important implications for therapy efficacy. Thanks to their MSC features, pericytes could be considered excellent candidates to promote nervous tissue repair and for regenerative medicine.
Collapse
Affiliation(s)
- Nunzia Caporarello
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA;
| | - Floriana D’Angeli
- Section of Medical Biochemistry, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (F.D.); (M.T.C.); (A.L.); (G.G.)
| | - Maria Teresa Cambria
- Section of Medical Biochemistry, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (F.D.); (M.T.C.); (A.L.); (G.G.)
| | - Saverio Candido
- Section of General and Clinical Pathology and Oncology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy;
| | - Cesarina Giallongo
- Section of Haematology, Department of General Surgery and Medical-Surgical Specialties, University of Catania, 95123 Catania, Italy;
| | - Mario Salmeri
- Section of Microbiology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (M.S.); (C.L.)
| | - Cinzia Lombardo
- Section of Microbiology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (M.S.); (C.L.)
| | - Anna Longo
- Section of Medical Biochemistry, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (F.D.); (M.T.C.); (A.L.); (G.G.)
| | - Giovanni Giurdanella
- Section of Medical Biochemistry, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (F.D.); (M.T.C.); (A.L.); (G.G.)
| | - Carmelina Daniela Anfuso
- Section of Medical Biochemistry, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (F.D.); (M.T.C.); (A.L.); (G.G.)
- Correspondence: (G.L.); (C.D.A.); Tel.: +39-095-4781158 (G.L.); +39-095-4781170 (C.D.A.)
| | - Gabriella Lupo
- Section of Medical Biochemistry, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (F.D.); (M.T.C.); (A.L.); (G.G.)
- Correspondence: (G.L.); (C.D.A.); Tel.: +39-095-4781158 (G.L.); +39-095-4781170 (C.D.A.)
| |
Collapse
|
8
|
Geranmayeh MH, Rahbarghazi R, Farhoudi M. Targeting pericytes for neurovascular regeneration. Cell Commun Signal 2019; 17:26. [PMID: 30894190 PMCID: PMC6425710 DOI: 10.1186/s12964-019-0340-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 03/13/2019] [Indexed: 02/06/2023] Open
Abstract
Pericytes, as a key cellular part of the blood-brain barrier, play an important role in the maintenance of brain neurovascular unit. These cells participate in brain homeostasis by regulating vascular development and integrity mainly through secreting various factors. Pericytes per se show different restorative properties after blood-brain barrier injury. Upon the occurrence of brain acute and chronic diseases, pericytes provoke immune cells to regulate neuro-inflammatory conditions. Loss of pericytes in distinct neurologic disorders intensifies blood-brain barrier permeability and leads to vascular dementia. The therapeutic potential of pericytes is originated from the unique morphological shape, location, and their ability in providing vast paracrine and juxtacrine interactions. A subset of pericytes possesses multipotentiality and exhibit trans-differentiation capacity in the context of damaged tissue. This review article aimed to highlight the critical role of pericytes in restoration of the blood-brain barrier after injury by focusing on the dynamics of pericytes and cross-talk with other cell types.
Collapse
Affiliation(s)
- Mohammad Hossein Geranmayeh
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Neurosciences Research Center (NSRC), Imam Reza Medical Center, Tabriz University of Medical Sciences, Golgasht St., Azadi Ave, Tabriz, 5166614756, Iran
| | - Reza Rahbarghazi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mehdi Farhoudi
- Neurosciences Research Center (NSRC), Imam Reza Medical Center, Tabriz University of Medical Sciences, Golgasht St., Azadi Ave, Tabriz, 5166614756, Iran.
| |
Collapse
|
9
|
Dias DO, Göritz C. Fibrotic scarring following lesions to the central nervous system. Matrix Biol 2018; 68-69:561-570. [PMID: 29428230 DOI: 10.1016/j.matbio.2018.02.009] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/05/2018] [Accepted: 02/05/2018] [Indexed: 10/18/2022]
Abstract
Following lesions to the central nervous system, scar tissue forms at the lesion site. Injury often severs axons and scar tissue is thought to block axonal regeneration, resulting in permanent functional deficits. While scar-forming astrocytes have been extensively studied, much less attention has been given to the fibrotic, non-glial component of the scar. We here review recent progress in understanding fibrotic scar formation following different lesions to the brain and spinal cord. We specifically highlight recent evidence for pericyte-derived fibrotic scar tissue formation, discussing the origin, recruitment, function and therapeutic relevance of fibrotic scarring.
Collapse
Affiliation(s)
- David Oliveira Dias
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Christian Göritz
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| |
Collapse
|
10
|
Davies JE, Walker JT, Keating A. Concise Review: Wharton's Jelly: The Rich, but Enigmatic, Source of Mesenchymal Stromal Cells. Stem Cells Transl Med 2017; 6:1620-1630. [PMID: 28488282 PMCID: PMC5689772 DOI: 10.1002/sctm.16-0492] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 02/03/2017] [Accepted: 02/24/2017] [Indexed: 12/21/2022] Open
Abstract
The umbilical cord has become an increasingly used source of mesenchymal stromal cells for preclinical and, more recently, clinical studies. Despite the increased activity, several aspects of this cell population have been under‐appreciated. Key issues are that consensus on the anatomical structures within the cord is lacking, and potentially different populations are identified as arising from a single source. To help address these points, we propose a histologically based nomenclature for cord structures and provide an analysis of their developmental origins and composition. Methods of cell isolation from Wharton's jelly are discussed and the immunophenotypic and clonal characteristics of the cells are evaluated. The perivascular origin of the cells is also addressed. Finally, clinical trials with umbilical cord cells are briefly reviewed. Interpreting the outcomes of the many clinical studies that have been undertaken with mesenchymal stromal cells from different tissue sources has been challenging, for many reasons. It is, therefore, particularly important that as umbilical cord cells are increasingly deployed therapeutically, we strive to better understand the derivation and functional characteristics of the cells from this important tissue source. Stem Cells Translational Medicine2017;6:1620–1630
Collapse
Affiliation(s)
- John E Davies
- Institute of Biomaterials and Biomedical Engineering, Toronto, Ontario, Canada.,Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - John T Walker
- Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Armand Keating
- Institute of Biomaterials and Biomedical Engineering, Toronto, Ontario, Canada.,Cell Therapy Program, Arthritis Program, Krembil Research Institute, and Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| |
Collapse
|
11
|
Rustenhoven J, Jansson D, Smyth LC, Dragunow M. Brain Pericytes As Mediators of Neuroinflammation. Trends Pharmacol Sci 2017; 38:291-304. [DOI: 10.1016/j.tips.2016.12.001] [Citation(s) in RCA: 156] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 12/01/2016] [Accepted: 12/01/2016] [Indexed: 01/03/2023]
|
12
|
Pericytes: The Role of Multipotent Stem Cells in Vascular Maintenance and Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1079:69-86. [PMID: 29282647 DOI: 10.1007/5584_2017_138] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Blood vessels consist of an inner endothelial cell layer lining the vessel wall and perivascular pericytes, also known as mural cells, which envelop the vascular tube surface. Pericytes have recently been recognized for their central role in blood vessel formation. Pericytes are multipotent cells that are heterogeneous in their origin, function, morphology and surface markers. Similar to other types of stem cells, pericytes act as a repair system in response to injury by maintaining the structural integrity of blood vessels. Several studies have shown that blood vessels lacking pericytes become hyperdilated and haemorrhagic, leading to vascular complications ranging from diabetic retinopathy to embryonic death. The role of pericytes is not restricted to the formation and development of the vasculature: they have been shown to possess stem cell-like characteristics and may differentiate into cell types from different lineages. Recent discoveries regarding the contribution of pericytes to tumour metastasis and the maintenance of tumour vascular supply and angiogenesis have led researchers to propose targeting pericytes with anti-angiogenic therapies. In this review, we will examine the different physiological roles of pericytes, their differentiation potential, and how they interact with surrounding cells to ensure the integrity of blood vessel formation and maintenance.
Collapse
|
13
|
Absolute circulating pericyte progenitor cell counts in mice by flow cytometry: comparison of 2 single-platform technologies. Int J Biol Markers 2015; 30:e434-8. [PMID: 26109365 DOI: 10.5301/jbm.5000156] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2015] [Indexed: 11/20/2022]
Abstract
BACKGROUND Accumulating evidence indicates that circulating pericyte progenitor cells (CPPCs) may be angiogenic biomarkers in cancer and diabetes. Their validity as biomarkers depends on the accuracy of techniques used for enumeration. In this report, absolute CPPC counts were performed by 2 single-platform technologies. The reliability of the 2 methods, including retest reliability and intraobserver and interobserver variability, was assessed according to the intraclass correlation coefficient (ICC). The linear correlation and agreement among both methods were assessed, and the stability of CPPC numbers in blood samples was analyzed. METHODS The blood samples were obtained from ICR mice. The samples were processed through a no-lyse, 1-wash procedure, and Syto16+CD45-CD31-CD140b+ CPPCs were analyzed by exclusion of dead cells and by fluorescence-minus-one control. CPPCs were enumerated by 2 methods: bead-based 123count eBeads count (eBioscience) and direct volume-based Accuri C6 Flow Cytometer count (BD). The cells were measured immediately and after storage of blood samples for 24 and 48 hours. RESULTS There were excellent retest correlations and intraobserver and interobserver agreement in both methods. The 2 methods showed a high linear correlation (R2 = 0.923) and with a high level of agreement (0.986). It was demonstrated that CPPCs are unstable in blood samples. CONCLUSIONS In this study, 2 reproducible protocols for CPPC quantification were established. These protocols should facilitate future studies to further define the role of CPPCs as cellular biomarkers.
Collapse
|
14
|
Occurring of In Vitro Functional Vasculogenic Pericytes from Human Circulating Early Endothelial Precursor Cell Culture. Stem Cells Int 2015; 2015:943671. [PMID: 26064139 PMCID: PMC4438190 DOI: 10.1155/2015/943671] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 04/10/2015] [Accepted: 04/21/2015] [Indexed: 12/21/2022] Open
Abstract
Pericytes are periendothelial cells of the microcirculation which contribute to tissue homeostasis and hemostasis by regulating microvascular morphogenesis and stability. Because of their multipotential ex vivo differentiation capabilities, pericytes are becoming very interesting in regenerative medicine field. Several studies address this issue by attempting to isolate pericyte/mesenchymal-like cells from peripheral blood; however the origin of these cells and their culture conditions are still debated. Here we showed that early Endothelial Progenitor Cells (EPCs) expressing CD45+/CD146+/CD31+ can be a source of cells with pericyte/mesenchymal phenotype and function, identified as human Progenitor Perivascular Cells (hPPCs). We provided evidence that hPPCs have an immunophenotype consistent with Mesenchymal Stem Cells (MSCs) from human adipose tissue (hASCs) and fetal membranes of term placenta (FM-hMSCs). In addition, hPPCs can be subcultured and exhibit expression of pluripotent genes (OCT-4, KLF-4, and NANOG) as well as a remarkable vasculogenic potential. Our findings could be helpful to develop innovative cell-based therapies for future clinical applications with distinct therapeutic purposes.
Collapse
|
15
|
Kelly-Goss MR, Sweat RS, Stapor PC, Peirce SM, Murfee WL. Targeting pericytes for angiogenic therapies. Microcirculation 2015; 21:345-57. [PMID: 24267154 DOI: 10.1111/micc.12107] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 11/19/2013] [Indexed: 12/18/2022]
Abstract
In pathological scenarios, such as tumor growth and diabetic retinopathy, blocking angiogenesis would be beneficial. In others, such as myocardial infarction and hypertension, promoting angiogenesis might be desirable. Due to their putative influence on endothelial cells, vascular pericytes have become a topic of growing interest and are increasingly being evaluated as a potential target for angioregulatory therapies. The strategy of manipulating pericyte recruitment to capillaries could result in anti- or proangiogenic effects. Our current understanding of pericytes, however, is limited by knowledge gaps regarding pericyte identity and lineage. To use a music analogy, this review is a "mash-up" that attempts to integrate what we know about pericyte functionality and expression with what is beginning to be elucidated regarding their regenerative potential. We explore the lingering questions regarding pericyte phenotypic identity and lineage. The expression of different pericyte markers (e.g., SMA, Desmin, NG2, and PDGFR-β) varies for different subpopulations and tissues. Previous use of these markers to identify pericytes has suggested potential phenotypic overlaps and plasticity toward other cell phenotypes. Our review chronicles the state of the literature, identifies critical unanswered questions, and motivates future research aimed at understanding this intriguing cell type and harnessing its therapeutic potential.
Collapse
Affiliation(s)
- Molly R Kelly-Goss
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | | | | | | | | |
Collapse
|
16
|
Birbrair A, Zhang T, Wang ZM, Messi ML, Mintz A, Delbono O. Pericytes at the intersection between tissue regeneration and pathology. Clin Sci (Lond) 2015; 128:81-93. [PMID: 25236972 PMCID: PMC4200531 DOI: 10.1042/cs20140278] [Citation(s) in RCA: 173] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Perivascular multipotent cells, pericytes, contribute to the generation and repair of various tissues in response to injury. They are heterogeneous in their morphology, distribution, origin and markers, and elucidating their molecular and cellular differences may inform novel treatments for disorders in which tissue regeneration is either impaired or excessive. Moreover, these discoveries offer novel cellular targets for therapeutic approaches to many diseases. This review discusses recent studies that support the concept that pericyte subtypes play a distinctive role in myogenesis, neurogenesis, adipogenesis, fibrogenesis and angiogenesis.
Collapse
Affiliation(s)
- Alexander Birbrair
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
- Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Tan Zhang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Zhong-Min Wang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Maria Laura Messi
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Akiva Mintz
- Department of Neurosurgery, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Osvaldo Delbono
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
- Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| |
Collapse
|
17
|
Makihara N, Arimura K, Ago T, Tachibana M, Nishimura A, Nakamura K, Matsuo R, Wakisaka Y, Kuroda J, Sugimori H, Kamouchi M, Kitazono T. Involvement of platelet-derived growth factor receptor β in fibrosis through extracellular matrix protein production after ischemic stroke. Exp Neurol 2014; 264:127-34. [PMID: 25510317 DOI: 10.1016/j.expneurol.2014.12.007] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 12/04/2014] [Accepted: 12/07/2014] [Indexed: 02/06/2023]
Abstract
Fibrosis is concomitant with repair processes following injuries in the central nervous system (CNS). Pericytes are considered as an origin of fibrosis-forming cells in the CNS. Here, we examined whether platelet-derived growth factor receptor β (PDGFRβ), a well-known indispensable molecule for migration, proliferation, and survival of pericytes, was involved in the production of extracellular matrix proteins, fibronectin and collagen type I, which is crucial for fibrosis after ischemic stroke. Immunohistochemistry demonstrated induction of PDGFRβ expression in vascular cells of peri-infarct areas at 3-7days in a mouse stroke model. The PDGFRβ-expressing cells extended from peri-infarct areas toward the ischemic core after day 7 while expressing fibronectin and collagen type I in the infarct areas. In contrast, desmin and α-smooth muscle actin, markers of pericytes, were only expressed in vascular cells. In PDGFRβ heterozygous knockout mice, the expression of fibronectin and collagen type I was attenuated at both mRNA and protein levels with an enlargement of the infarct volume after ischemic stroke compared with that in wild-type littermates. In cultured brain pericytes, the expression of PDGF-B, PDGFRβ, fibronectin, and collagen type I, but not desmin, was significantly increased by serum depletion (SD). The SD-induced upregulation of fibronectin and collagen type I was suppressed by SU11652, an inhibitor of PDGFRβ, while PDGF-B further increased the SD-induced upregulation. In conclusion, the expression level of PDGFRβ may be a crucial determinant of fibrosis after ischemic stroke. Moreover, PDGFRβ signaling participates in the production of fibronectin and collagen type I after ischemic stroke.
Collapse
Affiliation(s)
- Noriko Makihara
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Koichi Arimura
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Japan; Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Tetsuro Ago
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Japan.
| | - Masaki Tachibana
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Ataru Nishimura
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Japan; Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Kuniyuki Nakamura
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Ryu Matsuo
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Japan; Department of Health Care Administration and Management, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Yoshinobu Wakisaka
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Junya Kuroda
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Hiroshi Sugimori
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Masahiro Kamouchi
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Japan; Department of Health Care Administration and Management, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Japan
| |
Collapse
|
18
|
Birbrair A, Zhang T, Wang ZM, Messi ML, Mintz A, Delbono O. Pericytes: multitasking cells in the regeneration of injured, diseased, and aged skeletal muscle. Front Aging Neurosci 2014; 6:245. [PMID: 25278877 PMCID: PMC4166895 DOI: 10.3389/fnagi.2014.00245] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 08/29/2014] [Indexed: 12/16/2022] Open
Abstract
Pericytes are perivascular cells that envelop and make intimate connections with adjacent capillary endothelial cells. Recent studies show that they may have a profound impact in skeletal muscle regeneration, innervation, vessel formation, fibrosis, fat accumulation, and ectopic bone formation throughout life. In this review, we summarize and evaluate recent advances in our understanding of pericytes' influence on adult skeletal muscle pathophysiology. We also discuss how further elucidating their biology may offer new approaches to the treatment of conditions characterized by muscle wasting.
Collapse
Affiliation(s)
- Alexander Birbrair
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine Winston-Salem, NC, USA ; Neuroscience Program, Wake Forest School of Medicine Winston-Salem, NC, USA
| | - Tan Zhang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine Winston-Salem, NC, USA
| | - Zhong-Min Wang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine Winston-Salem, NC, USA
| | - Maria L Messi
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine Winston-Salem, NC, USA
| | - Akiva Mintz
- Department of Neurosurgery, Wake Forest School of Medicine Winston-Salem, NC, USA
| | - Osvaldo Delbono
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine Winston-Salem, NC, USA ; Neuroscience Program, Wake Forest School of Medicine Winston-Salem, NC, USA
| |
Collapse
|
19
|
Bazarek S, Peterson DA. Prospects for engineering neurons from local neocortical cell populations as cell-mediated therapy for neurological disorders. J Comp Neurol 2014; 522:2857-76. [PMID: 24756774 PMCID: PMC4729289 DOI: 10.1002/cne.23618] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 04/18/2014] [Accepted: 04/20/2014] [Indexed: 12/21/2022]
Abstract
There is little cell replacement following neurological injury, limiting the regenerative response of the CNS. Progress in understanding the biology of neural stem cells has raised interest in using stem cells for replacing neurons lost to injury or to disease. Stem cell therapy may also have a role in rebuilding deficient neural circuitry underlying mood disorders, epilepsy, and pain modulation among other roles. In vitro expansion of stem cells with directed differentiation prior to transplantation is one approach to stem cell therapy. Emerging evidence suggests that it may be possible to convert in vivo endogenous neural cells to a neuronal fate directly, providing an alternative strategy for stem cell therapy to the CNS. This review assesses the evidence for engineering a subtype-specific neuronal fate of endogenous neural cells in the cerebral cortex as a function of initial cell lineage, reactive response to injury, conversion factors, and environmental context. We conclude with a discussion of some of the challenges that must be overcome to move this alternative in vivo engineered conversion process toward becoming a viable therapeutic option.
Collapse
Affiliation(s)
- Stanley Bazarek
- Center for Stem Cell and Regenerative Medicine, Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, 60064
| | | |
Collapse
|
20
|
Youn SW, Jung KH, Chu K, Lee JY, Lee ST, Bahn JJ, Park DK, Yu JS, Kim SY, Kim M, Lee SK, Han MH, Roh JK. Feasibility and Safety of Intra-arterial Pericyte Progenitor Cell Delivery Following Mannitol-Induced Transient Blood-Brain Barrier Opening in a Canine Model. Cell Transplant 2014; 24:1469-79. [PMID: 24932854 DOI: 10.3727/096368914x682413] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Stem cell therapy is currently being studied with a view to rescuing various neurological diseases. Such studies require not only the discovery of potent candidate cells but also the development of methods that allow optimal delivery of those candidates to the brain tissues. Given that the blood-brain barrier (BBB) precludes cells from entering the brain, the present study was designed to test whether hyperosmolar mannitol securely opens the BBB and enhances intra-arterial cell delivery. A noninjured normal canine model in which the BBB was presumed to be closed was used to evaluate the feasibility and safety of the tested protocol. Autologous adipose tissue-derived pericytes with platelet-derived growth factor receptor β positivity were utilized. Cells were administered 5 min after mannitol pretreatment using one of following techniques: (1) bolus injection of a concentrated suspension, (2) continuous infusion of a diluted suspension, or (3) bolus injection of a concentrated suspension that had been shaken by repeated syringe pumping. Animals administered a concentrated cell suspension without mannitol pretreatment served as a control group. Vital signs, blood parameters, neurologic status, and major artery patency were kept stable throughout the experiment and the 1-month posttreatment period. Although ischemic lesions were noted on magnetic resonance imaging in several mongrel dogs with concentrated cell suspension, the injection technique using repeated syringe shaking could avert this complication. The cells were detected in both ipsilateral and contralateral cortices and were more frequent at the ipsilateral and frontal locations, whereas very few cells were observed anywhere in the brain when mannitol was not preinjected. These data suggest that intra-arterial cell infusion with mannitol pretreatment is a feasible and safe therapeutic approach in stable brain diseases such as chronic stroke.
Collapse
Affiliation(s)
- Sung Won Youn
- Department of Neuroradiology, Catholic University of Daegu Medical Center, School of Medicine, Catholic University of Daegu, Daegu, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Lopatina T, Bruno S, Tetta C, Kalinina N, Porta M, Camussi G. Platelet-derived growth factor regulates the secretion of extracellular vesicles by adipose mesenchymal stem cells and enhances their angiogenic potential. Cell Commun Signal 2014; 12:26. [PMID: 24725987 PMCID: PMC4022079 DOI: 10.1186/1478-811x-12-26] [Citation(s) in RCA: 232] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 04/04/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Several studies demonstrate the role of adipose mesenchymal stem cells (ASCs) in angiogenesis. The angiogenic mechanism has been ascribed to paracrine factors since these cells secrete a plenty of signal molecules and growth factors. Recently it has been suggested that besides soluble factors, extracellular vesicles (EVs) that include exosomes and microvesicles may play a major role in cell-to-cell communication. It has been shown that EVs are implicated in the angiogenic process. RESULTS Herein we studied whether EVs released by ASCs may mediate the angiogenic activity of these cells. Our results demonstrated that ASC-derived EVs induced in vitro vessel-like structure formation by human microvascular endothelial cells (HMEC). EV-stimulated HMEC when injected subcutaneously within Matrigel in SCID mice formed vessels. Treatment of ASCs with platelet-derived growth factor (PDGF) stimulated the secretion of EVs, changed their protein composition and enhanced the angiogenic potential. At variance of EVs released in basal conditions, PDGF-EVs carried c-kit and SCF that played a role in angiogenesis as specific blocking antibodies inhibited in vitro vessel-like structure formation. The enhanced content of matrix metalloproteinases in PDGF-EVs may also account for their angiogenic activity. CONCLUSIONS Our findings indicate that EVs released by ASCs may contribute to the ASC-induced angiogenesis and suggest that PDGF may trigger the release of EVs with an enhanced angiogenic potential.
Collapse
Affiliation(s)
| | | | | | | | | | - Giovanni Camussi
- Department of Medical Sciences and Molecular Biotechnology Center, University of Torino, Corso Dogliotti 14, 10126, Torino, Italy.
| |
Collapse
|
22
|
Birbrair A, Zhang T, Wang ZM, Messi ML, Enikolopov GN, Mintz A, Delbono O. Role of pericytes in skeletal muscle regeneration and fat accumulation. Stem Cells Dev 2013; 22:2298-314. [PMID: 23517218 PMCID: PMC3730538 DOI: 10.1089/scd.2012.0647] [Citation(s) in RCA: 203] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Accepted: 03/20/2013] [Indexed: 02/06/2023] Open
Abstract
Stem cells ensure tissue regeneration, while overgrowth of adipogenic cells may compromise organ recovery and impair function. In myopathies and muscle atrophy associated with aging, fat accumulation increases dysfunction, and after chronic injury, the process of fatty degeneration, in which muscle is replaced by white adipocytes, further compromises tissue function and environment. Some studies suggest that pericytes may contribute to muscle regeneration as well as fat formation. This work reports the presence of two pericyte subpopulations in the skeletal muscle and characterizes their specific roles. Skeletal muscle from Nestin-GFP/NG2-DsRed mice show two types of pericytes, Nestin-GFP-/NG2-DsRed+ (type-1) and Nestin-GFP+/NG2-DsRed+ (type-2), in close proximity to endothelial cells. We also found that both Nestin-GFP-/NG2-DsRed+ and Nestin-GFP+/NG2-DsRed+ cells colocalize with staining of two pericyte markers, PDGFRβ and CD146, but only type-1 pericyte express the adipogenic progenitor marker PDGFRα. Type-2 pericytes participate in muscle regeneration, while type-1 contribute to fat accumulation. Transplantation studies indicate that type-1 pericytes do not form muscle in vivo, but contribute to fat deposition in the skeletal muscle, while type-2 pericytes contribute only to the new muscle formation after injury, but not to the fat accumulation. Our results suggest that type-1 and type-2 pericytes contribute to successful muscle regeneration which results from a balance of myogenic and nonmyogenic cells activation.
Collapse
MESH Headings
- Adipogenesis/genetics
- Animals
- Antigens/genetics
- Antigens/metabolism
- CD146 Antigen/genetics
- CD146 Antigen/metabolism
- Cell Lineage/genetics
- Endothelial Cells/cytology
- Female
- Gene Expression
- Genes, Reporter
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Luminescent Proteins/genetics
- Luminescent Proteins/metabolism
- Male
- Mice
- Mice, Nude
- Mice, Transgenic
- Muscle, Skeletal/cytology
- Muscle, Skeletal/injuries
- Muscle, Skeletal/metabolism
- Nestin/genetics
- Nestin/metabolism
- Pericytes/cytology
- Pericytes/metabolism
- Pericytes/transplantation
- Proteoglycans/genetics
- Proteoglycans/metabolism
- Receptor, Platelet-Derived Growth Factor alpha/genetics
- Receptor, Platelet-Derived Growth Factor alpha/metabolism
- Receptor, Platelet-Derived Growth Factor beta/genetics
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Regeneration/genetics
- Red Fluorescent Protein
Collapse
Affiliation(s)
- Alexander Birbrair
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Department of Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Tan Zhang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Zhong-Min Wang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Maria Laura Messi
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Grigori N. Enikolopov
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
- NBIC, Moscow Institute of Physics and Technology, Moscow, Russia
| | - Akiva Mintz
- Department of Neurosurgery, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Osvaldo Delbono
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Department of Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
23
|
Brain pericyte plasticity as a potential drug target in CNS repair. Drug Discov Today 2012; 18:456-63. [PMID: 23266366 DOI: 10.1016/j.drudis.2012.12.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 11/23/2012] [Accepted: 12/11/2012] [Indexed: 01/09/2023]
Abstract
Brain pericytes (BrPCs) are essential cellular components of the central nervous system neurovascular unit involved in the regulation of blood flow, blood-brain barrier function, as well as in the stabilization of the vessel architecture. More recently, it became evident that BrPCs, besides their regulatory activities in brain vessel function and homeostasis, have pleiotropic functions in the adult CNS ranging from stromal and regeneration promoting activities to stem cell properties. This special characteristic confers BrPC cell plasticity, being able to display features of other cells within the organism. BrPCs might also be causally involved in certain brain diseases. Due to these properties BrPCs might be potential drug targets for future therapies of neurological disorders. This review summarizes BrPC properties, disorders in which this cell type might be involved, and provides suggestions for future therapeutic developments targeting BrPCs.
Collapse
|
24
|
Skeletal muscle pericyte subtypes differ in their differentiation potential. Stem Cell Res 2012; 10:67-84. [PMID: 23128780 DOI: 10.1016/j.scr.2012.09.003] [Citation(s) in RCA: 153] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 09/10/2012] [Accepted: 09/12/2012] [Indexed: 12/16/2022] Open
Abstract
Neural progenitor cells have been proposed as a therapy for central nervous system disorders, including neurodegenerative diseases and trauma injuries, however their accessibility is a major limitation. We recently isolated Tuj1+ cells from skeletal muscle culture of Nestin-GFP transgenic mice however whether they form functional neurons in the brain is not yet known. Additionally, their isolation from nontransgenic species and identification of their ancestors is unknown. This gap of knowledge precludes us from studying their role as a valuable alternative to neural progenitors. Here, we identified two pericyte subtypes, type-1 and type-2, using a double transgenic Nestin-GFP/NG2-DsRed mouse and demonstrated that Nestin-GFP+/Tuj1+ cells derive from type-2 Nestin-GFP+/NG2-DsRed+/CD146+ pericytes located in the skeletal muscle interstitium. These cells are bipotential as they generate either Tuj1+ cells when cultured with muscle cells or become "classical" α-SMA+pericytes when cultured alone. In contrast, type-1 Nestin-GFP-/NG2-DsRed+/CD146+ pericytes generate α-SMA+pericytes but not Tuj1+ cells. Interestingly, type-2 pericyte derived Tuj1+ cells retain some pericytic markers (CD146+/PDGFRβ+/NG2+). Given the potential application of Nestin-GFP+/NG2-DsRed+/Tuj1+ cells for cell therapy, we found a surface marker, the nerve growth factor receptor, which is expressed exclusively in these cells and can be used to identify and isolate them from mixed cell populations in nontransgenic species for clinical purposes.
Collapse
|
25
|
Kamouchi M, Ago T, Kuroda J, Kitazono T. The possible roles of brain pericytes in brain ischemia and stroke. Cell Mol Neurobiol 2012; 32:159-65. [PMID: 21830084 DOI: 10.1007/s10571-011-9747-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2011] [Accepted: 07/28/2011] [Indexed: 02/06/2023]
Abstract
Brain pericytes regulate a variety of functions, such as microcirculation, angiogenesis, and the blood brain barrier in the brain. Recent studies have also shown that they are pluripotent in a manner similar to mesenchymal stem cells. Since, brain pericytes actively control these functions, these cells probably play an important role not only during brain ischemia, but also in the post-stroke period.
Collapse
Affiliation(s)
- Masahiro Kamouchi
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan.
| | | | | | | |
Collapse
|
26
|
Current World Literature. Curr Opin Nephrol Hypertens 2012; 21:106-18. [DOI: 10.1097/mnh.0b013e32834ee42b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
27
|
Kolonin MG, Evans KW, Mani SA, Gomer RH. Alternative origins of stroma in normal organs and disease. Stem Cell Res 2011; 8:312-23. [PMID: 22209011 DOI: 10.1016/j.scr.2011.11.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 11/07/2011] [Accepted: 11/24/2011] [Indexed: 01/07/2023] Open
Abstract
Stromal fibroblasts are a new prospective drug target. Mesenchymal stromal cells (MSCs) and monocyte-derived stromal cells, also known as fibrocytes, are distinct fibroblastic populations derived from separate lineages. Mesenchymal and myeloid fibroblast progenitors are multipotent, serve as progenitor cells in animal models, and are implicated in several diseases. In addition, epithelial-mesenchymal transition (EMT) has been established as a mechanism for generation of stromal cells. Organ sources, relative contributions, and functions of these populations in normal development and pathology are not well understood. Innovative approaches are needed to identify markers that can distinguish these stromal populations.
Collapse
Affiliation(s)
- Mikhail G Kolonin
- The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, 1825 Pressler st., Houston, TX 77030, USA
| | | | | | | |
Collapse
|
28
|
Nakagomi T, Molnár Z, Nakano-Doi A, Taguchi A, Saino O, Kubo S, Clausen M, Yoshikawa H, Nakagomi N, Matsuyama T. Ischemia-induced neural stem/progenitor cells in the pia mater following cortical infarction. Stem Cells Dev 2011; 20:2037-51. [PMID: 21838536 DOI: 10.1089/scd.2011.0279] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Increasing evidence shows that neural stem/progenitor cells (NSPCs) can be activated in the nonconventional neurogenic zones such as the cortex following ischemic stroke. However, the precise origin, identity, and subtypes of the ischemia-induced NSPCs (iNSPCs), which can contribute to cortical neurogenesis, is currently still unclear. In our present study, using an adult mouse cortical infarction model, we found that the leptomeninges (pia mater), which is widely distributed within and closely associated with blood vessels as microvascular pericytes/perivascular cells throughout central nervous system (CNS), have NSPC activity in response to ischemia and can generate neurons. These observations indicate that microvascular pericytes residing near blood vessels that are distributed from the leptomeninges to the cortex are potential sources of iNSPCs for neurogenesis following cortical infarction. In addition, our results propose a novel concept that the leptomeninges, which cover the entire brain, have an important role in CNS restoration following brain injury such as stroke.
Collapse
Affiliation(s)
- Takayuki Nakagomi
- Institute for Advanced Medical Sciences, Hyogo College of Medicine, Hyogo, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Andres RH, Choi R, Pendharkar AV, Gaeta X, Wang N, Nathan JK, Chua JY, Lee SW, Palmer TD, Steinberg GK, Guzman R. The CCR2/CCL2 interaction mediates the transendothelial recruitment of intravascularly delivered neural stem cells to the ischemic brain. Stroke 2011; 42:2923-31. [PMID: 21836091 DOI: 10.1161/strokeaha.110.606368] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND PURPOSE The inflammatory response is a critical component of ischemic stroke. In addition to its physiological role, the mechanisms behind transendothelial recruitment of immune cells also offer a unique therapeutic opportunity for translational stem cell therapies. Recent reports have demonstrated homing of neural stem cells (NSC) into the injured brain areas after intravascular delivery. However, the mechanisms underlying the process of transendothelial recruitment remain largely unknown. Here we describe the critical role of the chemokine CCL2 and its receptor CCR2 in targeted homing of NSC after ischemia. METHODS Twenty-four hours after induction of stroke using the hypoxia-ischemia model in mice CCR2+/+ and CCR2-/- reporter NSC were intra-arterially delivered. Histology and bioluminescence imaging were used to investigate NSC homing to the ischemic brain. Functional outcome was assessed with the horizontal ladder test. RESULTS Using NSC isolated from CCR2+/+ and CCR2-/- mice, we show that receptor deficiency significantly impaired transendothelial diapedesis specifically in response to CCL2. Accordingly, wild-type NSC injected into CCL2-/- mice exhibited significantly decreased homing. Bioluminescence imaging showed robust recruitment of CCR2+/+ cells within 6 hours after transplantation in contrast to CCR2-/- cells. Mice receiving CCR2+/+ grafts after ischemic injury showed a significantly improved recovery of neurological deficits as compared to animals with transplantation of CCR2-/- NSC. CONCLUSIONS The CCL2/CCR2 interaction is critical for transendothelial recruitment of intravascularly delivered NSC in response to ischemic injury. This finding could have significant implications in advancing minimally invasive intravascular therapeutics for regenerative medicine or cell-based drug delivery systems for central nervous system diseases.
Collapse
Affiliation(s)
- Robert H Andres
- Department of Neurosurgery, Stanford Stroke Center, Stanford University, School of Medicine, 300 Pasteur Drive R211, Stanford, CA 94305-5327, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|