1
|
Wang J, Xiong T, Wu Q, Qin X. Integrated Strategies for Targeting Arteriogenesis and Angiogenesis After Stroke. Transl Stroke Res 2024:10.1007/s12975-024-01291-4. [PMID: 39225878 DOI: 10.1007/s12975-024-01291-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/29/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024]
Abstract
The interdependence between arteriogenesis and angiogenesis is crucial for enhancing perfusion by synchronously improving leptomeningeal collaterals (LMCs) and microvascular networks after stroke. However, current approaches often focus on promoting arteriogenesis and angiogenesis separately, neglecting the potential synergistic benefits of targeting both processes simultaneously. Therefore, it is imperative to consider both arteriogenesis and angiogenesis as integral and complementary strategies for post-stroke revascularization. To gain a deeper understanding of their relationships after stroke and to facilitate the development of targeted revascularization strategies, we compared them based on their timescale, space, and pathophysiology. The temporal differences in the occurrence of arteriogenesis and angiogenesis allow them to restore blood flow at different stages after stroke. The spatial differences in the effects of arteriogenesis and angiogenesis enable them to specifically target the ischemic penumbra and core infarct region. Additionally, the endothelial cell, as the primary effector cell in their pathophysiological processes, is promising target for enhancing both. Therefore, we provide an overview of key signals that regulate endothelium-mediated arteriogenesis and angiogenesis. Finally, we summarize current therapeutic strategies that involve these signals to promote both processes after stroke, with the aim of inspiring future therapeutic advances in revascularization.
Collapse
Affiliation(s)
- Jing Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Taoying Xiong
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Qisi Wu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Xinyue Qin
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
2
|
Panda SK, Torsiello M, Rehman A, Desiderio V, Del Vecchio V. Mitochondrial Transfer Between Mesenchymal Stem Cells and Cancer Cells. Methods Mol Biol 2024; 2835:39-48. [PMID: 39105904 DOI: 10.1007/978-1-0716-3995-5_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
Mitochondrial transfer (MT) is a biological process that allows a donor cell to horizontally share its own mitochondria with a recipient cell. Mitochondria are highly dynamic membrane-bound sub-cellular organelles prominently involved in the regulation of the cell energy balance, calcium homeostasis, and apoptotic machinery activation. They physiologically undergo fusion and fission processes in response to the cell requirement, with a continuous morphological re-arrangement. This structural and functional plasticity is at the basis of the MT, described in tissue regeneration, cardiac and neurological diseases, as well as in cancer. Here, the MT has been observed in the tumor micro-environment (TME) from the adipose-derived stem cells (ASCs) to the cancer cells, eventually reverting the lack of the mitochondria respiration function, or enhancing their motility and drug resistance. In this chapter, we outline some key protocols for evaluating this exciting phenomenon of MT. These methodological and technical approaches are very important, considering all the limitations that scientists constantly face, especially in this field of the research.
Collapse
Affiliation(s)
- S Kumar Panda
- Department of Experimental Medicine, Histology and Embryology Section, University of Campania "L. Vanvitelli", Naples, Italy
| | - M Torsiello
- Department of Experimental Medicine, Histology and Embryology Section, University of Campania "L. Vanvitelli", Naples, Italy
| | - A Rehman
- Department of Experimental Medicine, Histology and Embryology Section, University of Campania "L. Vanvitelli", Naples, Italy
| | - Vincenzo Desiderio
- Department of Experimental Medicine, Histology and Embryology Section, University of Campania "L. Vanvitelli", Naples, Italy
| | - V Del Vecchio
- Department of Experimental Medicine, Histology and Embryology Section, University of Campania "L. Vanvitelli", Naples, Italy.
| |
Collapse
|
3
|
Rudnicka-Drożak E, Drożak P, Mizerski G, Drożak M. Endothelial Progenitor Cells in Neurovascular Disorders—A Comprehensive Overview of the Current State of Knowledge. Biomedicines 2022; 10:biomedicines10102616. [PMID: 36289878 PMCID: PMC9599182 DOI: 10.3390/biomedicines10102616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/11/2022] [Accepted: 10/15/2022] [Indexed: 11/16/2022] Open
Abstract
Endothelial progenitor cells (EPCs) are a population of cells that circulate in the blood looking for areas of endothelial or vascular injury in order to repair them. Endothelial dysfunction is an important component of disorders with neurovascular involvement. Thus, the subject of involvement of EPCs in such conditions has been gaining increasing scientific interest in recent years. Overall, decreased levels of EPCs are associated with worse disease outcome. Moreover, their functionalities appear to decline with severity of disease. These findings inspired the application of EPCs as therapeutic targets and agents. So far, EPCs appear safe and promising based on the results of pre-clinical studies conducted on their use in the treatment of Alzheimer’s disease and ischemic stroke. In the case of the latter, human clinical trials have recently started to be performed in this subject and provided optimistic results thus far. Whereas in the case of migraine, existing findings pave the way for testing EPCs in in vitro studies. This review aims to thoroughly summarize current knowledge on the role EPCs in four disorders with neurovascular involvement, which are Alzheimer’s disease, cerebral small vessel disease, ischemic stroke and migraine, with a particular focus on the potential practical use of these cells as a treatment remedy.
Collapse
Affiliation(s)
- Ewa Rudnicka-Drożak
- Department of Family Medicine, Medical University of Lublin, Langiewicza 6a, 20-035 Lublin, Poland
| | - Paulina Drożak
- Student Scientific Society, Department of Family Medicine, Medical University of Lublin, Langiewicza 6a, 20-035 Lublin, Poland
- Correspondence: ; Tel.: +48-669-084-455
| | - Grzegorz Mizerski
- Department of Family Medicine, Medical University of Lublin, Langiewicza 6a, 20-035 Lublin, Poland
| | - Martyna Drożak
- Student Scientific Society, Department of Family Medicine, Medical University of Lublin, Langiewicza 6a, 20-035 Lublin, Poland
| |
Collapse
|
4
|
Oxidative stress-induced endothelial cells-derived exosomes accelerate skin flap survival through Lnc NEAT1-mediated promotion of endothelial progenitor cell function. Stem Cell Res Ther 2022; 13:325. [PMID: 35850692 PMCID: PMC9290268 DOI: 10.1186/s13287-022-03013-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 06/08/2022] [Indexed: 12/14/2022] Open
Abstract
Background Flap transplantation is commonly used in reconstructive surgery. A prerequisite for skin flap survival is sufficient blood supply. However, such approaches remain unclear. This study aimed to explore the underlying mechanisms of exosomes derived from human umbilical vascular endothelial cells (HUVECs) exposed to oxidative stress on endothelial progenitor cells (EPCs) and their subsequent influence on the survival of skin flaps. Methods HUVECs were treated with various concentrations of H2O2 to establish an oxidative stress model. To investigate the effects of H2O2-HUVEC-Exos and HUVEC-Exos, Cell Counting Kit-8, tube formation, invasion assays, and quantitative real-time polymerase chain reaction (qRT-PCR) were performed in EPCs. Microarray analysis was used to reveal the differentially expressed long non-coding RNAs (lncRNAs) in the H2O2-HUVEC-Exos and HUVEC-Exos. In addition, gene silencing and western blotting were employed to determine the mechanism behind lncRNA nuclear enrichment enriched transcript 1 (Lnc NEAT1) in EPCs. Further, a rat skin flap model was used to determine the role of the exosomes in skin flap survival in vivo. Results HUVECs were stimulated with 100 μmol/L H2O2 for 12 h to establish an oxidative stress model. H2O2-HUVEC-Exos promoted the proliferation, tube formation, and invasion of EPCs and remarkably increased skin flap survival compared to the HUVEC-Exos and control groups. Sequencing of exosome RNAs revealed that the Lnc NEAT1 level was dramatically increased in the H2O2-HUVEC-Exos, leading to activation of the Wnt/β-catenin signaling pathway. Comparatively, knockdown of Lnc NEAT1 in HUVEC-Exos and H2O2-HUVEC-Exos significantly inhibits the angiogenic capacity of EPCs, reduced the survival area of skin flap and downregulated the expression levels of Wnt/β-catenin signaling pathway proteins, whereas Wnt agonist partly reversed the negative effect of NEAT1 downregulation on EPCs through the Wnt/β-catenin signaling pathway. Conclusions Exosomes derived from HUVECs stimulated by oxidative stress significantly promoted the pro-angiogenic ability of EPCs through the Wnt/β-catenin signaling pathway mediated by Lnc NEAT1 and hence enhanced random flap survival in vivo. Therefore, the application of H2O2-HUVEC-Exos may serve as an alternative therapy for improving random skin flap survival. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03013-9.
Collapse
|
5
|
Intercellular mitochondrial transfer as a means of tissue revitalization. Signal Transduct Target Ther 2021; 6:65. [PMID: 33589598 PMCID: PMC7884415 DOI: 10.1038/s41392-020-00440-z] [Citation(s) in RCA: 154] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 11/04/2020] [Accepted: 11/24/2020] [Indexed: 01/31/2023] Open
Abstract
As the crucial powerhouse for cell metabolism and tissue survival, the mitochondrion frequently undergoes morphological or positional changes when responding to various stresses and energy demands. In addition to intracellular changes, mitochondria can also be transferred intercellularly. Besides restoring stressed cells and damaged tissues due to mitochondrial dysfunction, the intercellular mitochondrial transfer also occurs under physiological conditions. In this review, the phenomenon of mitochondrial transfer is described according to its function under both physiological and pathological conditions, including tissue homeostasis, damaged tissue repair, tumor progression, and immunoregulation. Then, the mechanisms that contribute to this process are summarized, such as the trigger factors and transfer routes. Furthermore, various perspectives are explored to better understand the mysteries of cell-cell mitochondrial trafficking. In addition, potential therapeutic strategies for mitochondria-targeted application to rescue tissue damage and degeneration, as well as the inhibition of tumor progression, are discussed.
Collapse
|
6
|
Díaz del Moral S, Barrena S, Muñoz-Chápuli R, Carmona R. Embryonic circulating endothelial progenitor cells. Angiogenesis 2020; 23:531-541. [DOI: 10.1007/s10456-020-09732-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 06/22/2020] [Indexed: 12/26/2022]
|
7
|
Czabanka M, Petrilli LL, Elvers-Hornung S, Bieback K, Albert Imhof B, Vajkoczy P, Vinci M. Junctional Adhesion Molecule-C Mediates the Recruitment of Embryonic-Endothelial Progenitor Cells to the Perivascular Niche during Tumor Angiogenesis. Int J Mol Sci 2020; 21:ijms21041209. [PMID: 32054130 PMCID: PMC7072851 DOI: 10.3390/ijms21041209] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 02/07/2020] [Indexed: 11/29/2022] Open
Abstract
The homing of Endothelial Progenitor Cells (EPCs) to tumor angiogenic sites has been described as a multistep process, involving adhesion, migration, incorporation and sprouting, for which the underlying molecular and cellular mechanisms are yet to be fully defined. Here, we studied the expression of Junctional Adhesion Molecule-C (JAM-C) by EPCs and its role in EPC homing to tumor angiogenic vessels. For this, we used mouse embryonic-Endothelial Progenitor Cells (e-EPCs), intravital multi-fluorescence microscopy techniques and the dorsal skin-fold chamber model. JAM-C was found to be expressed by e-EPCs and endothelial cells. Blocking JAM-C did not affect adhesion of e-EPCs to endothelial monolayers in vitro but, interestingly, it did reduce their adhesion to tumor endothelium in vivo. The most striking effect of JAM-C blocking was on tube formation on matrigel in vitro and the incorporation and sprouting of e-EPCs to tumor endothelium in vivo. Our results demonstrate that JAM-C mediates e-EPC recruitment to tumor angiogenic sites, i.e., coordinated homing of EPCs to the perivascular niche, where they cluster and interact with tumor blood vessels. This suggests that JAM-C plays a critical role in the process of vascular assembly and may represent a potential therapeutic target to control tumor angiogenesis.
Collapse
Affiliation(s)
- Marcus Czabanka
- Department of Neurosurgery, Universitätsmedizin Charitè, 10117 Berlin, Germany;
- Department of Neurosurgery Medical Faculty of the University of Heidelberg, 68167 Mannheim, Germany;
| | - Lucia Lisa Petrilli
- Department of Onco-haematology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital – IRCCS, 00146 Rome, Italy;
| | - Susanne Elvers-Hornung
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Fred Cross Blood Donor Service Baden-Württemberg – Hessen, 68167 Mannheim, Germany (K.B.)
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Fred Cross Blood Donor Service Baden-Württemberg – Hessen, 68167 Mannheim, Germany (K.B.)
| | - Beat Albert Imhof
- Department of Pathology and Immunology, Medical Faculty, Centre Medical Universitaire (CMU), University of Geneva, 1206 Geneva, Switzerland;
| | - Peter Vajkoczy
- Department of Neurosurgery, Universitätsmedizin Charitè, 10117 Berlin, Germany;
- Department of Neurosurgery Medical Faculty of the University of Heidelberg, 68167 Mannheim, Germany;
- Correspondence: ; Tel.: +49-30450560-002
| | - Maria Vinci
- Department of Neurosurgery Medical Faculty of the University of Heidelberg, 68167 Mannheim, Germany;
- Department of Onco-haematology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital – IRCCS, 00146 Rome, Italy;
| |
Collapse
|
8
|
Sadanandan N, Di Santo S, Widmer HR. Another win for endothelial progenitor cells: Endothelial progenitor cell-derived conditioned medium promotes proliferation and exerts neuroprotection in cultured neuronal progenitor cells. Brain Circ 2019; 5:106-111. [PMID: 31620656 PMCID: PMC6785943 DOI: 10.4103/bc.bc_41_19] [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: 06/02/2019] [Revised: 09/10/2019] [Accepted: 09/11/2019] [Indexed: 01/08/2023] Open
Abstract
Progress in stem cell research demonstrates stem cells' potential for treating neurodegenerative diseases. Stem cells have proliferative/differentiative properties and produce a variety of paracrine factors that can potentially be used to regenerate nervous tissue. Previous studies have shown the positive regenerative effects of endothelial progenitor cells (EPCs), and thus, they may be used as a tool for regeneration. A study by Di Santo et al. explored whether EPC-derived conditioned medium (EPC-CM) promotes the survival of cultured striatal progenitor cells and attempted to find the paracrine factors and signaling pathways involved with EPC-CM's effects. The neuronal progenitor cells that were cultured with EPC-CM had much higher densities of GABA-immunoreactive (GABA-ir) neurons. It was shown that phosphatidylinositol-3-kinase/AKT and mitogen-activated protein kinase/ERK signaling pathways are involved in the proliferation of GABAergic neurons, as inhibition of these pathways decreased GABAergic densities. In addition, the results suggest that paracrine factors from EPC, both proteinaceous and lipidic, significantly elevated the viability and/or differentiation in the cultures. Importantly, it was found that EPC-CM provided neuroprotection against toxins from 3-nitropropionic acid. In sum, EPC-CM engendered proliferation and regeneration of the cultured striatal cells through paracrine factors and imparted neuroprotection. Furthermore, the effects of EPC-CM may generate a cell-free therapeutic strategy to address neurodegeneration.
Collapse
Affiliation(s)
- Nadia Sadanandan
- Department of Neurosurgery and Brain Repair, College of Medicine, University of South Florida Morsani, Tampa, FL, USA
| | - Stefano Di Santo
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Hans Rudolf Widmer
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
9
|
Patschan S, Vogt M, Bakhtiari D, Bramlage CP, Henze E, Muller GA, Krause A, Patschan D. Humoral and Cellular Patterns of Early Endothelial Progenitor Cells in Relation to the Cardiovascular Risk in Axial Spondylarthritis. J Clin Med Res 2019; 11:391-400. [PMID: 31143305 PMCID: PMC6522236 DOI: 10.14740/jocmr3441w] [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: 04/10/2018] [Accepted: 04/30/2018] [Indexed: 11/21/2022] Open
Abstract
Background Spondylarthritis (SpA) significantly affects sacroiliac, intervertebral and peripheral joints. Patients with SpA suffer from increased cardiovascular risk (CVR). The endothelial progenitor cell (EPC) system critically perpetuates vascular repair. The aim of the study was to evaluate circulating EPCs in axial (ax)SpA with special attention on parameters of disease activity and CVR. Methods Disease activity and functional impairment were quantified in 50 axSpA patients by using standardized parameters (Bath ankylosing spondylitis disease activity index (BASDAI), C-reactive protein (CRP), finger-floor distance (FFD) and Ott’ sign). Circulating EPCs and EPC regeneration were analyzed (fluorescence-activated cell sorting (FACS) and colony-forming unit (CFU) assay). Serum vasomodulatory mediators were quantified by enzyme-linked immunosorbent assay (ELISA). Results EPC colony numbers were lower in axSpA as compared to controls. Females displayed more colonies than males. In addition, fewer colonies were observed in smokers, in patients with a BASDAI of below 4 and in hypertension. Circulating CD133+/KDR+ cells did not differ between the groups. Follow-up analysis (33 months later) did not show any differences in gender, colony formation, CD133+/KDR+ cells or serum levels of vasomodulatory mediators if related to the categories of BASDAI, Ott’ sign or FFD. Conclusions EPC colony formation is significantly affected in axSpA with particularly low levels in males. EPC-related parameters do not allow predicting disease activity-related or functional parameters nor are they useful for CVR assessment in SpA.
Collapse
Affiliation(s)
- Susann Patschan
- Clinic of Nephrology and Rheumatology, University Hospital of Gottingen, Gottingen, Germany.,Department of Cardiology, Pulmonology, Angiology and Nephrology, Brandenburg Medical School, University Hospital Brandenburg, Brandenburg, Germany
| | - Maria Vogt
- Clinic of Nephrology and Rheumatology, University Hospital of Gottingen, Gottingen, Germany
| | - Donia Bakhtiari
- Clinic of Nephrology and Rheumatology, University Hospital of Gottingen, Gottingen, Germany
| | - Carsten Peter Bramlage
- Clinic of Nephrology and Rheumatology, University Hospital of Gottingen, Gottingen, Germany
| | - Elvira Henze
- Clinic of Nephrology and Rheumatology, University Hospital of Gottingen, Gottingen, Germany
| | - Gerhard Anton Muller
- Clinic of Nephrology and Rheumatology, University Hospital of Gottingen, Gottingen, Germany
| | - Andreas Krause
- Rheumatology and Clinical Immunology, Immanuel-Krankenhaus Berlin, Berlin, Germany
| | - Daniel Patschan
- Department of Cardiology, Pulmonology, Angiology and Nephrology, Brandenburg Medical School, University Hospital Brandenburg, Brandenburg, Germany
| |
Collapse
|
10
|
Santo SD, Seiler S, Andres R, Widmer HR. Endothelial Progenitor Cells Conditioned Medium Supports Number of GABAergic Neurons and Exerts Neuroprotection in Cultured Striatal Neuronal Progenitor Cells. Cell Transplant 2019; 28:367-378. [PMID: 31017468 PMCID: PMC6628568 DOI: 10.1177/0963689719835192] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
There is growing evidence that stem and progenitor cells exert regenerative actions by means of paracrine factors. In line with these notions, we recently demonstrated that endothelial progenitor cell (EPC)-derived conditioned medium (EPC-CM) substantially increased viability of brain microvascular cells. In the present study, we aimed at investigating whether EPC-CM supports cell survival of cultured striatal progenitor cells. For that purpose, primary cultures from fetal rat embryonic (E14) ganglionic eminence were prepared and grown for 7 days in vitro (DIV). EPC-CM was administered from DIV5–7. Treatment of the striatal cultures with EPC-CM resulted in significantly increased densities of GABA-immunoreactive (-ir) neurons. Inhibition of mitogen-activated protein kinase and phosphatidylinositol-3-kinase, but not of the ROCK pathway, significantly attenuated the EPC-CM induced increase in GABA-ir cell densities. Similar results were observed when EPC-CM was subjected to proteolytic digestion and lipid extraction. Furthermore, inhibition of translation abolished the EPC-CM induced effects. Importantly, EPC-CM displayed neuroprotection against 3-nitropropionic acid induced toxicity. These findings demonstrate that EPC-derived paracrine factors substantially promote survival and/or differentiation of cultured striatal progenitor cells involving both proteinaceous factors and lipidic factors. In sum, EPC-CM constituents might lead to a novel cell-free therapeutic strategy to challenge neuronal degeneration.
Collapse
Affiliation(s)
- Stefano Di Santo
- 1 Department of Neurosurgery, Bern University Hospital, Switzerland
| | - Stefanie Seiler
- 1 Department of Neurosurgery, Bern University Hospital, Switzerland
| | - Robert Andres
- 1 Department of Neurosurgery, Bern University Hospital, Switzerland
| | | |
Collapse
|
11
|
Factors related to improvement of cerebrovascular reserve after superficial temporal artery to middle cerebral artery anastomosis for patients with atherosclerotic steno-occlusive disease. Acta Neurochir (Wien) 2019; 161:799-805. [PMID: 30778681 DOI: 10.1007/s00701-019-03841-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 02/08/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND This study aimed to investigate factors related to improvement of hemodynamics and evaluated the usefulness of intraoperative Doppler for predicting postoperative hemodynamics in patients with cerebrovascular atherosclerotic steno-occlusive disease (CASD) of the internal carotid artery (ICA) or middle cerebral artery (MCA) who were treated with extracranial-intracranial (EC-IC) bypass surgery. METHOD Forty-eight patients with CASD of the ICA or MCA who were treated by superficial temporal artery to middle cerebral artery bypass with a follow-up longer than 12 months were enrolled. Repeated transient ischemic attack or completed ischemic stroke was observed under optimal medical therapy in all patients. Intraoperative blood flow velocity of the MCA was evaluated by a Doppler flowmeter. Cerebral blood flow and cerebrovascular reserve (CVR) were evaluated using N-isopropyl-[123I] p-iodoamphetamine (IMP) single photon emission computed tomography (SPECT) preoperatively and 3 months after surgery. Imaging and clinical data were retrospectively reviewed. RESULTS CVR was significantly increased postoperatively (p = 0.03). One year after the operation, two (4.2%) patients developed cerebral infarction. The change in MCA flow velocity just after anastomosis compared with pre-anastomosis proximal and distal of the anastomosis site was a median of 3.0 and 2.6 times, respectively. However, there was no significant association between changes in intraoperative MCA flow velocity and postoperative CVR. Multivariate analysis showed that the presence of a lower estimated glomerular filtration rate (eGFR) was an independent risk factor for a decrease in CVR (p = 0.036). CONCLUSIONS A higher eGFR might have prognostic value for improvement in CVR after EC-IC bypass surgery in patients with CASD and misery perfusion.
Collapse
|
12
|
Choi SA, Chong S, Kwak PA, Moon YJ, Jangra A, Phi JH, Lee JY, Park SH, Kim SK. Impaired functional recovery of endothelial colony-forming cells from moyamoya disease in a chronic cerebral hypoperfusion rat model. J Neurosurg Pediatr 2019; 23:204-213. [PMID: 30668528 DOI: 10.3171/2018.8.peds1883] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 08/06/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVEEndothelial colony-forming cells (ECFCs) isolated from pediatric patients with moyamoya disease (MMD) have demonstrated decreased numbers and defective functioning in in vitro experiments. However, the function of ECFCs has not been evaluated using in vivo animal models. In this study, the authors compared normal and MMD ECFCs using a chronic cerebral hypoperfusion (CCH) rat model.METHODSA CCH rat model was made via ligation of the bilateral common carotid arteries (2-vessel occlusion [2-VO]). The rats were divided into three experimental groups: vehicle-treated (n = 8), normal ECFC-treated (n = 8), and MMD ECFC-treated (n = 8). ECFCs were injected into the cisterna magna. A laser Doppler flowmeter was used to evaluate cerebral blood flow, and a radial arm maze test was used to examine cognitive function. Neuropathological examinations of the hippocampus and agranular cortex were performed using hematoxylin and eosin and Luxol fast blue staining in addition to immunofluorescence with CD31, von Willebrand factor, NeuN, myelin basic protein, glial fibrillary acidic protein, and cleaved caspase-3 antibodies.RESULTSThe normal ECFC-treated group exhibited improvement in the restoration of cerebral perfusion and in behavior compared with the vehicle-treated and MMD ECFC-treated groups at the 12-week follow-up after the 2-VO surgery. The normal ECFC-treated group showed a greater amount of neovasculogenesis and neurogenesis, with less apoptosis, than the other groups.CONCLUSIONSThese results support the impaired functional recovery of MMD ECFCs compared with normal ECFCs in a CCH rat model. This in vivo study suggests the functional role of ECFCs in the pathogenesis of MMD.
Collapse
Affiliation(s)
- Seung Ah Choi
- 1Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital
- 2Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine
| | - Sangjoon Chong
- 1Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital
- 2Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine
| | - Pil Ae Kwak
- 1Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital
- 2Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine
| | - Youn Joo Moon
- 1Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital
- 2Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine
| | - Anshika Jangra
- 1Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital
- 2Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine
| | - Ji Hoon Phi
- 1Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital
- 2Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine
| | - Ji Yeoun Lee
- 1Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital
- 2Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine
- 3Department of Anatomy, Seoul National University College of Medicine; and
| | - Sung-Hye Park
- 4Department of Pathology, Seoul National University Hospital, Seoul, Korea
| | - Seung-Ki Kim
- 1Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital
- 2Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine
| |
Collapse
|
13
|
Nakayama T, Nagata E, Masuda H, Asahara T, Takizawa S. Regeneration-associated cell transplantation contributes to tissue recovery in mice with acute ischemic stroke. PLoS One 2019; 14:e0210198. [PMID: 30682162 PMCID: PMC6347160 DOI: 10.1371/journal.pone.0210198] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 12/18/2018] [Indexed: 01/05/2023] Open
Abstract
Various cell-based therapeutic strategies have been investigated for vascular and tissue regeneration after ischemic stroke. We have developed a novel cell population, called regeneration-associated cells (RACs), by quality- and quantity-controlled culture of unfractionated mononuclear cells. RACs were trans-arterially injected into 10-week-old syngeneic male mice at 1, 3, 5 or 7 days after permanent middle cerebral artery occlusion (MCAO) to determine the optimal timing for administration in terms of outcome at day 21. Next, we examined the effects of RACs injection at day 1 after MCAO on neurological deficits, infarct volume, and mediators of vascular regeneration and anti-inflammation at days 7 and 21. Infarct volume at day 21 was significantly reduced by transplantation of RACs at day 1 or 3. RACs injected at day 1 reduced the infarct volume at day 7 and 21. Angiogenesis and anti-inflammatory mediators, VEGF and IL-10, were increased at day 7, and VEGF was still upregulated at day 21. We also observed significantly enhanced ink perfusion in vivo, tube formation in vitro, and definitive endothelial progenitor cell colonies in colony assay. These results suggest that RAC transplantation in MCAO models promoted significant recovery of neural tissues through intensified anti-inflammatory and angiogenic effects.
Collapse
Affiliation(s)
- Taira Nakayama
- Department of Neurology, Tokai University School of Medicine, Isehara, Japan
| | - Eiichiro Nagata
- Department of Neurology, Tokai University School of Medicine, Isehara, Japan
| | - Haruchika Masuda
- Department of Physiology, Tokai University School of Medicine, Isehara, Japan
| | - Takayuki Asahara
- Department of Regenerative Medicine Science, Tokai University School of Medicine, Isehara, Japan
| | - Shunya Takizawa
- Department of Neurology, Tokai University School of Medicine, Isehara, Japan
- * E-mail:
| |
Collapse
|
14
|
Esquiva G, Grayston A, Rosell A. Revascularization and endothelial progenitor cells in stroke. Am J Physiol Cell Physiol 2018; 315:C664-C674. [PMID: 30133323 DOI: 10.1152/ajpcell.00200.2018] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Stroke is one of the leading causes of death and disability worldwide. Tremendous improvements have been achieved in the acute care of stroke patients with the implementation of stroke units, thrombolytic drugs, and endovascular trombectomies. However, stroke survivors with neurological deficits require long periods of neurorehabilitation, which is the only approved therapy for poststroke recovery. With this scenario, more treatments are urgently needed, and only the understanding of the mechanisms of brain recovery might contribute to identify new therapeutic agents. Fortunately, brain injury after stroke is counteracted by the birth and migration of several populations of progenitor cells towards the injured areas, where angiogenesis and vascular remodeling play a key role providing trophic support and guidance during neurorepair. Endothelial progenitor cells (EPCs) constitute a pool of circulating bone-marrow derived cells that mobilize after an ischemic injury with the potential to incorporate into the damaged endothelium, to form new vessels, or to secrete trophic factors stimulating vessel remodeling. The circulating levels of EPCs are altered after stroke, and several subpopulations have proved to boost brain neurorepair in preclinical models of cerebral ischemia. The goal of this review is to discuss the current state of the neuroreparative actions of EPCs, focusing on their paracrine signaling mechanisms thorough their secretome and released extracellular vesicles.
Collapse
Affiliation(s)
- Gema Esquiva
- Neurovascular Research Laboratory and Neurology Department, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - Alba Grayston
- Neurovascular Research Laboratory and Neurology Department, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - Anna Rosell
- Neurovascular Research Laboratory and Neurology Department, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona , Barcelona , Spain
| |
Collapse
|
15
|
Hayakawa K, Chan SJ, Mandeville ET, Park JH, Bruzzese M, Montaner J, Arai K, Rosell A, Lo EH. Protective Effects of Endothelial Progenitor Cell-Derived Extracellular Mitochondria in Brain Endothelium. Stem Cells 2018; 36:1404-1410. [PMID: 29781122 DOI: 10.1002/stem.2856] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/10/2018] [Accepted: 05/01/2018] [Indexed: 12/15/2022]
Abstract
Endothelial progenitor cells (EPCs) have been pursued as a potential cellular therapy for stroke and central nervous system injury. However, their underlying mechanisms remain to be fully defined. Recent experimental studies suggest that mitochondria may be released and transferred between cells. In this proof-of-concept study, we asked whether beneficial effects of EPCs may partly involve a mitochondrial phenomenon as well. First, EPC-derived conditioned medium was collected and divided into supernatant and particle fractions after centrifugation. Electron microscopy, Western blots, and flow cytometry showed that EPCs were able to release mitochondria. ATP and oxygen consumption assays suggested that these extracellular mitochondria may still be functionally viable. Confocal microscopy confirmed that EPC-derived extracellular mitochondria can be incorporated into normal brain endothelial cells. Adding EPC particles to brain endothelial cells promoted angiogenesis and decreased the permeability of brain endothelial cells. Next, we asked whether EPC-derived mitochondria may be protective. As expected, oxygen-glucose deprivation (OGD) increased brain endothelial permeability. Adding EPC-derived mitochondria particles to the damaged brain endothelium increased levels of mitochondrial protein TOM40, mitochondrial DNA copy number, and intracellular ATP. Along with these indirect markers of mitochondrial transfer, endothelial tightness was also restored after OGD. Taken together, these findings suggest that EPCs may support brain endothelial energetics, barrier integrity, and angiogenic function partly through extracellular mitochondrial transfer. Stem Cells 2018;36:1404-1410.
Collapse
Affiliation(s)
- Kazuhide Hayakawa
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Su Jing Chan
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Emiri T Mandeville
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Ji Hyun Park
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Morgan Bruzzese
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Anna Rosell
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| |
Collapse
|
16
|
Zhang N, Song C, Zhao B, Xing M, Luo L, Gordon ML, Cheng Y. Neovascularization and Synaptic Function Regulation with Memantine and Rosuvastatin in a Rat Model of Chronic Cerebral Hypoperfusion. J Mol Neurosci 2017; 63:223-232. [PMID: 28920182 DOI: 10.1007/s12031-017-0974-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 09/08/2017] [Indexed: 12/16/2022]
Abstract
Cerebral hypoperfusion is an important factor in the pathogenesis of cerebrovascular diseases and neurodegenerative disorders. We investigated the effects of memantine and rosuvastatin on both neovascularization and synaptic function in a rat model of chronic cerebral hypoperfusion, which was established by the bilateral common carotid occlusion (2VO) method. We tested learning and memory ability, synaptic function, circulating endothelial progenitor cell (EPC) number, expression of neurotrophic factors, and markers of neovasculogenesis and cell proliferation after memantine and/or rosuvastatin treatment. Rats treated with memantine and/or rosuvastatin showed significant improvement in Morris water maze task and long-term potentiation (LTP) in the hippocampus, compared with untreated 2VO model rats. Circulating EPCs, expression of brain-derived neurotrophic factor, and vascular endothelial growth factor, markers of microvessel density were increased by each of the three interventions. Rosuvastatin also increased cell proliferation in the hippocampus. Combined treatment with memantine and rosuvastatin showed greater effect on enhancement of LTP and expression of neurotrophic factors than either single medication treatment alone. Both memantine and rosuvastatin improved learning and memory, enhanced neovascularization and synaptic function, and upregulated neurotrophic factors in a rat model of chronic cerebral hypoperfusion.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, 154, Anshan Road, Tianjin, 300052, China.
- The Litwin-Zucker Research Center, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
| | - Chenchen Song
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, 154, Anshan Road, Tianjin, 300052, China
- Department of Neurology, No.254 Hospital of the PLA, Tianjin, China
| | - Baomin Zhao
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, 154, Anshan Road, Tianjin, 300052, China
- Department of Neurology, Yidu Central Hospital of Weifang, Qingzhou, China
| | - Mengya Xing
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, 154, Anshan Road, Tianjin, 300052, China
| | - Lanlan Luo
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, 154, Anshan Road, Tianjin, 300052, China
| | - Marc L Gordon
- The Litwin-Zucker Research Center, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
| | - Yan Cheng
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, 154, Anshan Road, Tianjin, 300052, China
| |
Collapse
|
17
|
Guo XB, Deng X, Wei Y. Homing of Cultured Endothelial Progenitor Cells and Their Effect on Traumatic Brain Injury in Rat Model. Sci Rep 2017. [PMID: 28646184 PMCID: PMC5482798 DOI: 10.1038/s41598-017-04153-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Transplanted endothelial progenitor cells (EPCs) may play an important role in reestablishing the endothelial integrity of the vessels after brain injury, and contribute to neurogenesis. We, therefore, tested the homing of ex vivo cultured peripheral blood-derived EPCs and their effect on injured brain tissue after intravenous administration. To track the homing of implanted EPCs in injured brain tissues, EPCs were labeled with DAPI and BrdU in vitro before transplantation. EPCs were transplanted into the host animal through peripheral administration through the femoral vein, and homing of EPCs was evaluated. The integration of intravenously injected EPCs into the injured brain tissue was demonstrated. Immunohistochemical staining showed that microvessel density in the perifocal region of EPCs-transplanted rats was significantly increased, and the numbers of BrdU+ cells in the DG of subventricular zone were increased in EPCs-transplanted rats as compared to the control group. Transplanted EPCs may play an important role in reestablishing the endothelial integrity in the vessels after brain injury and further contribute to neurogenesis. EPCs enhanced recovery following brain injury in a rat model of TBI.
Collapse
Affiliation(s)
- Xin-Bin Guo
- Department of Neuro-interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, Zhengzhou, 450052, China.
| | - Xin Deng
- Department of Neuro-interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, Zhengzhou, 450052, China
| | - Ying Wei
- Department of Neuro-interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, Zhengzhou, 450052, China
| |
Collapse
|
18
|
Lu D, Wu Y, Qu Y, Shi F, Hu J, Gao B, Wang B, Gao G, He S, Zhao T. A modified method to reduce variable outcomes in a rat model of four-vessel arterial occlusion. Neurol Res 2016; 38:1102-1110. [PMID: 27796195 DOI: 10.1080/01616412.2016.1249996] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Dan Lu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Yingxi Wu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Yan Qu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Fei Shi
- Department of Aerospace Biodynamics, The Fourth Military Medical University, Xi’an, China
| | - Jun Hu
- Department of Neurology, Chinese PLA NO. 451 Hospital, Xi’an, Shaanxi, China
| | - Bo Gao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Bao Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Guodong Gao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Shiming He
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Tianzhi Zhao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| |
Collapse
|
19
|
Du SQ, Wang XR, Xiao LY, Tu JF, Zhu W, He T, Liu CZ. Molecular Mechanisms of Vascular Dementia: What Can Be Learned from Animal Models of Chronic Cerebral Hypoperfusion? Mol Neurobiol 2016; 54:3670-3682. [PMID: 27206432 DOI: 10.1007/s12035-016-9915-1] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 05/03/2016] [Indexed: 01/06/2023]
Abstract
Vascular dementia (VD) is defined as a progressive neurodegenerative disease of cognitive decline, attributable to cerebrovascular factors. Numerous studies have demonstrated that chronic cerebral hypoperfusion (CCH) is associated with the initiation and progression of VD and Alzheimer's disease (AD). Suitable animal models were established to replicate such pathological condition in experimental research, which contributes largely to comprehending causal relationships between CCH and cognitive impairment. The most widely used experimental model of VD and CCH is permanent bilateral common carotid artery occlusion in rats. In CCH models, changes of learning and memory, cerebral blood flow (CBF), energy metabolism, and neuropathology initiated by ischemia were revealed. However, in order to achieve potential therapeutic targets, particular mechanisms in cognitive and neuropathological changes from CCH to dementia should be investigated. Recent studies have shown that hypoperfusion resulted in a chain of disruption of homeostatic interactions, including oxidative stress, neuroinflammation, neurotransmitter system dysfunction, mitochondrial dysfunction, disturbance of lipid metabolism, and alterations of growth factors. Evidence from experimental studies that elucidate the damaging effects of such imbalances suggests their critical roles in the pathogenesis of VD. The present review provides a summary of the achievements in mechanisms made with the CCH models, permits an understanding of the causative role played by CCH in VD, and highlights preventative and therapeutic prospects.
Collapse
Affiliation(s)
- Si-Qi Du
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing, 100010, China
| | - Xue-Rui Wang
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing, 100010, China
| | - Ling-Yong Xiao
- Beijing University of Chinese Medicine, 11 Beisanhuan East Road, Chaoyang District, Beijing, 100029, China
| | - Jian-Feng Tu
- Beijing University of Chinese Medicine, 11 Beisanhuan East Road, Chaoyang District, Beijing, 100029, China
| | - Wen Zhu
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing, 100010, China
| | - Tian He
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing, 100010, China
| | - Cun-Zhi Liu
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing, 100010, China.
| |
Collapse
|
20
|
Takizawa S, Nagata E, Nakayama T, Masuda H, Asahara T. Recent Progress in Endothelial Progenitor Cell Culture Systems: Potential for Stroke Therapy. Neurol Med Chir (Tokyo) 2016; 56:302-9. [PMID: 27041632 PMCID: PMC4908073 DOI: 10.2176/nmc.ra.2016-0027] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Endothelial progenitor cells (EPCs) participate in endothelial repair and angiogenesis due to their abilities to differentiate into endothelial cells and to secrete protective cytokines and growth factors. Consequently, there is considerable interest in cell therapy with EPCs isolated from peripheral blood to treat various ischemic injuries. Quality and quantity-controlled culture systems to obtain mononuclear cells enriched in EPCs with well-defined angiogenic and anti-inflammatory phenotypes have recently been developed, and increasing evidence from animal models and clinical trials supports the idea that transplantation of EPCs contributes to the regenerative process in ischemic organs and is effective for the therapy of ischemic cerebral injury. Here, we briefly describe the general characteristics of EPCs, and we review recent developments in culture systems and applications of EPCs and EPC-enriched cell populations to treat ischemic stroke.
Collapse
Affiliation(s)
- Shunya Takizawa
- Department of Neurology, Tokai University School of Medicine
| | | | | | | | | |
Collapse
|
21
|
Preconditioning with VEGF Enhances Angiogenic and Neuroprotective Effects of Bone Marrow Mononuclear Cell Transplantation in a Rat Model of Chronic Cerebral Hypoperfusion. Mol Neurobiol 2015; 53:6057-6068. [PMID: 26530694 DOI: 10.1007/s12035-015-9512-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 10/26/2015] [Indexed: 01/01/2023]
Abstract
Bone marrow mononuclear cell (BMMNC) transplantation is a promising therapy for brain ischemia. However, BMMNCs are few in number, and a limited time window is available during which they can penetrate the blood-brain barrier (BBB) and migrate to the brain. We investigated whether vascular endothelial growth factor (VEGF) can facilitate BMMNC migration into the ischemic brain and enhance their therapeutic effect in a rat model of chronic cerebral hypoperfusion. First, we assessed the impact of VEGF on the BBB of rats that had undergone permanent bilateral occlusion of the common carotid arteries (2VO). Then, we transplanted BMMNCs into 2VO rats pretreated with intracerebroventricular VEGF or vehicle. We examined cognitive function with the Morris water maze test, BMMNC migration by immunofluorescence analysis, and cytokine levels in the peripheral blood by enzyme-linked immunosorbent assay (ELISA). Angiogenesis and neural degeneration were evaluated by staining tissue with Ki67/lectin or Fluoro-Jade B. We found that at a dose of 0.2 μg/rat, VEGF significantly increased BBB permeability without causing brain edema in 2VO rats. VEGF + BMMNC-treated rats had more BMMNC migration in the ischemic brain, better learning and memory, greater proliferation of vessels, and fewer degenerating neurons than did BMMNC-treated rats. Pretreatment with VEGF receptor inhibitor SU5416 significantly decreased BMMNC migration and abolished the therapeutic effect of BMMNC transplantation. We conclude that preconditioning with an appropriate dose of VEGF can enhance the therapeutic efficacy of BMMNC transplantation in 2VO rats, possibly by facilitating BMMNC migration into the ischemic brain.
Collapse
|
22
|
Martynov MY, Gusev EI. Current knowledge on the neuroprotective and neuroregenerative properties of citicoline in acute ischemic stroke. J Exp Pharmacol 2015; 7:17-28. [PMID: 27186142 PMCID: PMC4863531 DOI: 10.2147/jep.s63544] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Ischemic stroke is one of the leading causes of long-lasting disability and death. Two main strategies have been proposed for the treatment of ischemic stroke: restoration of blood flow by thrombolysis or mechanical thrombus extraction during the first few hours of ischemic stroke, which is one of the most effective treatments and leads to a better functional and clinical outcome. The other direction of treatment, which is potentially applicable to most of the patients with ischemic stroke, is neuroprotection. Initially, neuroprotection was mainly targeted at protecting gray matter, but during the past few years there has been a transition from a neuron-oriented approach toward salvaging the whole neurovascular unit using multimodal drugs. Citicoline is a multimodal drug that exhibits neuroprotective and neuroregenerative effects in a variety of experimental and clinical disorders of the central nervous system, including acute and chronic cerebral ischemia, intracerebral hemorrhage, and global cerebral hypoxia. Citicoline has a prolonged therapeutic window and is active at various temporal and biochemical stages of the ischemic cascade. In acute ischemic stroke, citicoline provides neuroprotection by attenuating glutamate exitotoxicity, oxidative stress, apoptosis, and blood–brain barrier dysfunction. In the subacute and chronic phases of ischemic stroke, citicoline exhibits neuroregenerative effects and activates neurogenesis, synaptogenesis, and angiogenesis and enhances neurotransmitter metabolism. Acute and long-term treatment with citicoline is safe and in most clinical studies is effective and improves functional outcome.
Collapse
Affiliation(s)
- Mikhail Yu Martynov
- Department of Neurology, Neurosurgery and Medical Genetics, Russian National Research Medical University, Moscow, Russia
| | - Eugeny I Gusev
- Department of Neurology, Neurosurgery and Medical Genetics, Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
23
|
Particle Radiation-Induced Nontargeted Effects in Bone-Marrow-Derived Endothelial Progenitor Cells. Stem Cells Int 2015; 2015:496512. [PMID: 26074973 PMCID: PMC4436457 DOI: 10.1155/2015/496512] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 02/23/2015] [Accepted: 02/24/2015] [Indexed: 01/17/2023] Open
Abstract
Bone-marrow- (BM-) derived endothelial progenitor cells (EPCs) are critical for endothelial cell maintenance and repair. During future space exploration missions astronauts will be exposed to space irradiation (IR) composed of a spectrum of low-fluence protons ((1)H) and high charge and energy (HZE) nuclei (e.g., iron-(56)Fe) for extended time. How the space-type IR affects BM-EPCs is limited. In media transfer experiments in vitro we studied nontargeted effects induced by (1)H- and (56)Fe-IR conditioned medium (CM), which showed significant increase in the number of p-H2AX foci in nonirradiated EPCs between 2 and 24 h. A 2-15-fold increase in the levels of various cytokines and chemokines was observed in both types of IR-CM at 24 h. Ex vivo analysis of BM-EPCs from single, low-dose, full-body (1)H- and (56)Fe-IR mice demonstrated a cyclical (early 5-24 h and delayed 28 days) increase in apoptosis. This early increase in BM-EPC apoptosis may be the effect of direct IR exposure, whereas late increase in apoptosis could be a result of nontargeted effects (NTE) in the cells that were not traversed by IR directly. Identifying the role of specific cytokines responsible for IR-induced NTE and inhibiting such NTE may prevent long-term and cyclical loss of stem and progenitors cells in the BM milieu.
Collapse
|