1
|
Kaloss AM, Theus MH. Leptomeningeal anastomoses: Mechanisms of pial collateral remodeling in ischemic stroke. WIREs Mech Dis 2022; 14:e1553. [PMID: 35118835 PMCID: PMC9283306 DOI: 10.1002/wsbm.1553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/09/2022] [Accepted: 01/11/2022] [Indexed: 12/13/2022]
Abstract
Arterial collateralization, as determined by leptomeningeal anastomoses or pial collateral vessels, is a well‐established vital player in cerebral blood flow restoration and neurological recovery from ischemic stroke. A secondary network of cerebral collateral circulation apart from the Circle of Willis, exist as remnants of arteriole development that connect the distal arteries in the pia mater. Recent interest lies in understanding the cellular and molecular adaptations that control the growth and remodeling, or arteriogenesis, of these pre‐existing collateral vessels. New findings from both animal models and human studies of ischemic stroke suggest a multi‐factorial and complex, temporospatial interplay of endothelium, immune and vessel‐associated cell interactions may work in concert to facilitate or thwart arteriogenesis. These valuable reports may provide critical insight into potential predictors of the pial collateral response in patients with large vessel occlusion and may aid in therapeutics to enhance collateral function and improve recovery from stroke. This article is categorized under:Neurological Diseases > Molecular and Cellular Physiology
Collapse
Affiliation(s)
- Alexandra M Kaloss
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Michelle H Theus
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA.,School of Neuroscience, Virginia Tech, Blacksburg, Virginia, USA.,Center for Regenerative Medicine, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
2
|
Vakhrushev IV, Nezhurina EK, Karalkin PA, Tsvetkova AV, Sergeeva NS, Majouga AG, Yarygin KN. Heterotypic Multicellular Spheroids as Experimental and Preclinical Models of Sprouting Angiogenesis. BIOLOGY 2021; 11:18. [PMID: 35053016 PMCID: PMC8772844 DOI: 10.3390/biology11010018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022]
Abstract
Sprouting angiogenesis is the common response of live tissues to physiological and pathological angiogenic stimuli. Its accurate evaluation is of utmost importance for basic research and practical medicine and pharmacology and requires adequate experimental models. A variety of assays for angiogenesis were developed, none of them perfect. In vitro approaches are generally less physiologically relevant due to the omission of essential components regulating the process. However, only in vitro models can be entirely non-xenogeneic. The limitations of the in vitro angiogenesis assays can be partially overcome using 3D models mimicking tissue O2 and nutrient gradients, the influence of the extracellular matrix (ECM), and enabling cell-cell interactions. Here we present a review of the existing models of sprouting angiogenesis that are based on the use of endothelial cells (ECs) co-cultured with perivascular or other stromal cells. This approach provides an excellent in vitro platform for further decoding of the cellular and molecular mechanisms of sprouting angiogenesis under conditions close to the in vivo conditions, as well as for preclinical drug testing and preclinical research in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Igor V. Vakhrushev
- Laboratory of Cell Biology, Institute of Biomedical Chemistry, 119121 Moscow, Russia;
| | - Elizaveta K. Nezhurina
- P.A. Hertsen Moscow Oncology Research Center, National Medical Research Radiological Center, 125284 Moscow, Russia;
| | - Pavel A. Karalkin
- Institute for Cluster Oncology, Sechenov University, 119435 Moscow, Russia;
| | | | - Nataliya S. Sergeeva
- Department of Biology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia;
| | - Alexander G. Majouga
- Faculty of Chemical and Pharmaceutical Technologies and Biomedical Products, D. Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia;
| | - Konstantin N. Yarygin
- Laboratory of Cell Biology, Institute of Biomedical Chemistry, 119121 Moscow, Russia;
| |
Collapse
|
3
|
Ravindranath MH, El Hilali F, Filippone EJ. The Impact of Inflammation on the Immune Responses to Transplantation: Tolerance or Rejection? Front Immunol 2021; 12:667834. [PMID: 34880853 PMCID: PMC8647190 DOI: 10.3389/fimmu.2021.667834] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 10/11/2021] [Indexed: 12/21/2022] Open
Abstract
Transplantation (Tx) remains the optimal therapy for end-stage disease (ESD) of various solid organs. Although alloimmune events remain the leading cause of long-term allograft loss, many patients develop innate and adaptive immune responses leading to graft tolerance. The focus of this review is to provide an overview of selected aspects of the effects of inflammation on this delicate balance following solid organ transplantation. Initially, we discuss the inflammatory mediators detectable in an ESD patient. Then, the specific inflammatory mediators found post-Tx are elucidated. We examine the reciprocal relationship between donor-derived passenger leukocytes (PLs) and those of the recipient, with additional emphasis on extracellular vesicles, specifically exosomes, and we examine their role in determining the balance between tolerance and rejection. The concept of recipient antigen-presenting cell "cross-dressing" by donor exosomes is detailed. Immunological consequences of the changes undergone by cell surface antigens, including HLA molecules in donor and host immune cells activated by proinflammatory cytokines, are examined. Inflammation-mediated donor endothelial cell (EC) activation is discussed along with the effect of donor-recipient EC chimerism. Finally, as an example of a specific inflammatory mediator, a detailed analysis is provided on the dynamic role of Interleukin-6 (IL-6) and its receptor post-Tx, especially given the potential for therapeutic interdiction of this axis with monoclonal antibodies. We aim to provide a holistic as well as a reductionist perspective of the inflammation-impacted immune events that precede and follow Tx. The objective is to differentiate tolerogenic inflammation from that enhancing rejection, for potential therapeutic modifications. (Words 247).
Collapse
Affiliation(s)
- Mepur H. Ravindranath
- Department of Hematology and Oncology, Children’s Hospital, Los Angeles, CA, United States
- Terasaki Foundation Laboratory, Santa Monica, CA, United States
| | | | - Edward J. Filippone
- Division of Nephrology, Department of Medicine, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
4
|
Giménez CS, Castillo MG, Simonin JA, Núñez Pedrozo CN, Pascuali N, Bauzá MDR, Locatelli P, López AE, Belaich MN, Mendiz AO, Crottogini AJ, Cuniberti LA, Olea FD. Effect of intramuscular baculovirus encoding mutant hypoxia-inducible factor 1-alpha on neovasculogenesis and ischemic muscle protection in rabbits with peripheral arterial disease. Cytotherapy 2020; 22:563-572. [PMID: 32723595 DOI: 10.1016/j.jcyt.2020.06.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 06/07/2020] [Accepted: 06/26/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND AIMS Peripheral arterial disease (PAD) is a progressive, disabling ailment for which no effective treatment exists. Gene therapy-mediated neovascularization has emerged as a potentially useful strategy. We tested the angiogenic and arteriogenic efficacy and safety of a baculovirus (BV) encoding mutant, oxygen-resistant hypoxia-inducible factor 1-alpha (mHIF-1α), in rabbits with PAD. METHODS After assessing the transfection efficiency of the BV.mHIF-1α vector and its tubulogenesis potential in vitro, we randomized rabbits with experimental PAD to receive 1 × 109 copies of BV.mHIF-1α or BV.null (n = 6 per group) 7 days after surgery. Two weeks post-treatment, collateralization (digital angiography) and capillary and arteriolar densities (immunohistochemistry) were measured in the posterior limbs. Ischemic damage was evaluated in adductor and gastrocnemius muscle samples. Tracking of viral DNA in injected zones and remote tissues at different time points was performed in additional rabbits using a BV encoding GFP. RESULTS Angiographically visible collaterals were more numerous in BV.mHIF-1α-treated rabbits (8.12 ± 0.42 vs 6.13 ± 1.15 collaterals/cm2, P < 0.05). The same occurred with arteriolar (27.9 ± 7.0 vs 15.3 ± 4.0 arterioles/mm2) and capillary (341.8 ± 109.9 vs 208.8 ± 87.7 capillaries/mm2, P < 0.05) densities. BV.mHIF-1α-treated rabbits displayed less ischemic muscle damage than BV.null-treated animals. Viral DNA and GFP mRNA were detectable only at 3 and 7 days after injection in hind limbs. Neither the virus nor GFP mRNA was detected in remote tissues. CONCLUSIONS In rabbits with PAD, BV.mHIF-1α induced neovascularization and reduced ischemic damage, exhibiting a good safety profile at 14 days post-treatment. Complementary studies to evaluate its potential usefulness in the clinic are needed.
Collapse
Affiliation(s)
- Carlos S Giménez
- Laboratorio de Medicina Regenerativa Cardiovascular, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Favaloro, Buenos Aires, Argentina
| | - Martha G Castillo
- Laboratorio de Medicina Regenerativa Cardiovascular, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Favaloro, Buenos Aires, Argentina
| | - Jorge A Simonin
- Laboratorio de Ingeniería Genética y Biología Celular y Molecular (LIGBCM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Quilmes, Bernal, Argentina
| | - Cristian N Núñez Pedrozo
- Laboratorio de Medicina Regenerativa Cardiovascular, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Favaloro, Buenos Aires, Argentina
| | - Natalia Pascuali
- Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María Del Rosario Bauzá
- Laboratorio de Medicina Regenerativa Cardiovascular, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Favaloro, Buenos Aires, Argentina
| | - Paola Locatelli
- Laboratorio de Medicina Regenerativa Cardiovascular, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Favaloro, Buenos Aires, Argentina
| | - Ayelén E López
- Laboratorio de Medicina Regenerativa Cardiovascular, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Favaloro, Buenos Aires, Argentina
| | - Mariano N Belaich
- Laboratorio de Ingeniería Genética y Biología Celular y Molecular (LIGBCM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Quilmes, Bernal, Argentina
| | - Alfredo O Mendiz
- Hospital Universitario de la Fundación Favaloro, Buenos Aires, Argentina
| | - Alberto J Crottogini
- Laboratorio de Medicina Regenerativa Cardiovascular, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Favaloro, Buenos Aires, Argentina
| | - Luis A Cuniberti
- Laboratorio de Medicina Regenerativa Cardiovascular, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Favaloro, Buenos Aires, Argentina
| | - Fernanda D Olea
- Laboratorio de Medicina Regenerativa Cardiovascular, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Favaloro, Buenos Aires, Argentina.
| |
Collapse
|
5
|
Tanaka R, Umeyama Y, Hagiwara H, Ito-Hirano R, Fujimura S, Mizuno H, Ogawa R. Keloid patients have higher peripheral blood endothelial progenitor cell counts and CD34 + cells with normal vasculogenic and angiogenic function that overexpress vascular endothelial growth factor and interleukin-8. Int J Dermatol 2019; 58:1398-1405. [PMID: 31290139 DOI: 10.1111/ijd.14575] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 06/04/2019] [Accepted: 06/10/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND One suggested reason for aberrant wound healing in keloid scars is chronic inflammation of the dermis. We hypothesized that excessive blood vessel formation and high capillary density in keloid tissue is caused by dysfunction of endothelial progenitor cells. METHODS We compared the number of circulating endothelial progenitor cells and vasculogenic and angiogenic capacity, as well as secretory function, of circulating CD34+ cells in keloid patients and healthy individuals. RESULTS Compared to mononuclear cell cultures from healthy donors, cultures of peripheral blood mononuclear cells obtained from keloid patients showed a more than twofold increase in the number of peripheral blood EPCs (fibronectin-adhering cells that phagocytized acetylated low-density lipoprotein and bound Ulex europaeus agglutinin-I lectin). However, there was no difference in colony-forming ability and participation in in vitro angiogenesis between circulating CD34+ cells isolated from keloid patients and healthy individuals. This means that circulating CD34+ /endothelial progenitor cells in keloid patients have normal vasculogenic and angiogenic function. However, CD34+ cells derived from keloid patients demonstrated a more than sevenfold expression of the interleukin-8 gene and a more than fivefold expression of the vascular endothelial growth factor gene than CD34+ cells derived from healthy individuals. CONCLUSIONS These results support the role of vascular endothelial growth factor and interleukin-8 in increased recruitment of endothelial progenitor cells in keloid patients.
Collapse
Affiliation(s)
- Rica Tanaka
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Yuri Umeyama
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Hiroko Hagiwara
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Rie Ito-Hirano
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Satoshi Fujimura
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Hiroshi Mizuno
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Rei Ogawa
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
6
|
Barć P, Antkiewicz M, Śliwa B, Baczyńska D, Witkiewicz W, Skóra JP. Treatment of Critical Limb Ischemia by pIRES/VEGF165/HGF Administration. Ann Vasc Surg 2019; 60:346-354. [PMID: 31200059 DOI: 10.1016/j.avsg.2019.03.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 03/03/2019] [Accepted: 03/11/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND Prognosis of peripheral artery disease (PAD), especially critical limb ischemia (CLI), is very poor despite the development of endovascular therapy and bypass surgery. Many patients result in having leg amputation. We decided to investigate the safety and efficacy of plasmid of internal ribosome entry site/vascular endothelial growth factor (VEGF) 165/hepatocyte growth factor (HGF) gene therapy (GT) in patients suffered from CLI. METHODS Administration of plasmid of internal ribosome entry site/VEGF165/HGF was performed in 12 limbs of 12 patients with rest pain and ischemic ulcers due to CLI. Plasmid was injected into the muscles of the ischemic limbs. The levels of VEGF in serum and the ankle-brachial index (ABI) were measured before and after treatment. RESULTS Mean (±SD) plasma levels of VEGF increased nonsignificantly from 258 ± 81 pg/L to 489 ± 96 pg/L (P > 0.05) 2 weeks after therapy, and the ABI improved significantly from 0.27 ± 0.20 to 0.50 ± 0.22 (P < 0.001) 3 months after therapy. Ischemic ulcers healed in 9 limbs. Amputation was performed in 3 patients because of advanced necrosis and wound infection. However, the level of amputations was lowered below knee in these cases. Complications were limited to transient leg edema in 3 patients and fever in 2 patients. CONCLUSIONS Intramuscular administration of plasmid of internal ribosome entry site/VEGF165/HGF is safe, feasible, and effective for patients with critical leg ischemia.
Collapse
Affiliation(s)
- Piotr Barć
- Department and Clinic of Vascular, General and Transplantation Surgery, Jan Mikulicz-Radecki Medical University Hospital, Wroclaw Medical University, Wroclaw, Poland
| | - Maciej Antkiewicz
- Department and Clinic of Vascular, General and Transplantation Surgery, Jan Mikulicz-Radecki Medical University Hospital, Wroclaw Medical University, Wroclaw, Poland.
| | - Barbara Śliwa
- Department and Clinic of Vascular, General and Transplantation Surgery, Jan Mikulicz-Radecki Medical University Hospital, Wroclaw Medical University, Wroclaw, Poland
| | - Dagmara Baczyńska
- Molecular Techniques Unit, Wroclaw Medical University, Wroclaw, Poland
| | - Wojciech Witkiewicz
- Regional Specialized Hospital in Wroclaw, Research and Development Center, Wroclaw, Poland
| | - Jan Paweł Skóra
- Department and Clinic of Vascular, General and Transplantation Surgery, Jan Mikulicz-Radecki Medical University Hospital, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
7
|
Genetically engineered human muscle transplant enhances murine host neovascularization and myogenesis. Commun Biol 2018; 1:161. [PMID: 30320229 PMCID: PMC6172230 DOI: 10.1038/s42003-018-0161-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 08/24/2018] [Indexed: 11/30/2022] Open
Abstract
Engineered tissues are a promising tool for addressing the growing need for tissues and organs in surgical reconstructions. Prevascularization of implanted tissues is expected to enhance survival prospects post transplantation and minimize deficiencies and/or hypoxia deeper in the tissue. Here, we fabricate a three-dimensional, prevascularized engineered muscle containing human myoblasts, genetically modified endothelial cells secreting angiopoietin 1 (ANGPT1) and genetically modified smooth muscle cells secreting vascular endothelial growth factor (VEGF). The genetically engineered human muscle shows enhanced host neovascularization and myogenesis following transplantation into a mouse host, compared to the non-secreting control. The vascular, genetically modified cells have been cleared for clinical trials and can be used to construct autologous vascularized tissues. Therefore, the described genetically engineered vascularized muscle has the potential to be fully translated to the clinical setting to overcome autologous tissue shortage and to accelerate host neovascularization and integration of engineered grafts following transplantation. Luba Perry et al. report transplantation of engineered prevascularized human muscle into mice to repair an abdominal muscle defect. They show that genetically engineering smooth muscle cells to secrete VEGF and endothelial cells to secrete ANGPT1 significantly improves host neovascularization and myogenesis.
Collapse
|
8
|
Spheroids as vascularization units: From angiogenesis research to tissue engineering applications. Biotechnol Adv 2017; 35:782-791. [DOI: 10.1016/j.biotechadv.2017.07.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 07/03/2017] [Accepted: 07/05/2017] [Indexed: 02/08/2023]
|
9
|
Tevis KM, Colson YL, Grinstaff MW. Embedded Spheroids as Models of the Cancer Microenvironment. ADVANCED BIOSYSTEMS 2017; 1:1700083. [PMID: 30221187 PMCID: PMC6135264 DOI: 10.1002/adbi.201700083] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
To more accurately study the complex mechanisms behind cancer invasion, progression, and response to treatment, researchers require models that replicate both the multicellular nature and 3D stromal environment present in an in vivo tumor. Multicellular aggregates (i.e., spheroids) embedded in an extracellular matrix mimic are a prevalent model. Recently, quantitative metrics that fully utilize the capability of spheroids are described along with conventional experiments, such as invasion into a matrix, to provide additional details and insights into the underlying cancer biology. The review begins with a discussion of the salient features of the tumor microenvironment, introduces the early work on non-embedded spheroids as tumor models, and then concentrates on the successes achieved with the study of embedded spheroids. Examples of studies include cell movement, drug response, tumor cellular heterogeneity, stromal effects, and cancer progression. Additionally, new methodologies and those borrowed from other research fields (e.g., vascularization and tissue engineering) are highlighted that expand the capability of spheroids to aid future users in designing their cancer-related experiments. The convergence of spheroid research among the various fields catalyzes new applications and leads to a natural synergy. Finally, the review concludes with a reflection and future perspectives for cancer spheroid research.
Collapse
Affiliation(s)
- Kristie M. Tevis
- Departments of Biomedical Engineering, Chemistry, and Medicine, Metcalf Center for Science and Engineering, Boston University, Boston, MA 02215
| | - Yolonda L. Colson
- Division of Thoracic Surgery, Department of Surgery, Brigham and Women’s Hospital, Boston, MA 02215
| | - Mark W. Grinstaff
- Departments of Biomedical Engineering, Chemistry, and Medicine, Metcalf Center for Science and Engineering, Boston University, Boston, MA 02215
| |
Collapse
|
10
|
Perry L, Flugelman MY, Levenberg S. Elderly Patient-Derived Endothelial Cells for Vascularization of Engineered Muscle. Mol Ther 2017; 25:935-948. [PMID: 28279644 DOI: 10.1016/j.ymthe.2017.02.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/06/2017] [Accepted: 02/10/2017] [Indexed: 11/29/2022] Open
Abstract
In vitro prevascularization of engineered tissue constructs promises to enhance their clinical applicability. We hypothesize that adult endothelial cells (ECs), isolated from limb veins of elderly patients, bear the vasculogenic properties required to form vascular networks in vitro that can later integrate with the host vasculature upon implantation. Here, we show that adult ECs formed vessel networks that were more developed and complex than those formed by human umbilical vein endothelial cells (HUVECs) seeded with various supporting cells on three-dimensional (3D) biodegradable polymer scaffolds. In parallel, secreted levels of key proangiogenic cytokines were significantly higher in adult EC-bearing scaffolds as compared to HUVEC scaffolds. As a proof of concept for applicability of this model, adult ECs were co-seeded with human myoblasts as well as supporting cells and successfully formed a branched network, which was surrounded by aligned human myotubes. The vascularized engineered muscle tissue implanted into a full-thickness defect in immunodeficient mice remained viable and anastomosed with the host vasculature within 9 days of implantation. Functional "chimeric" blood vessels and various types of anastomosis were observed. These findings provide strong evidence of the applicability of adult ECs in construction of clinically relevant autologous vascularized tissue.
Collapse
Affiliation(s)
- Luba Perry
- Biomedical Engineering Department, Technion-Israel Institute of Technology, Haifa 32000, Israel; Inter-departmental Program in Biotechnology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Moshe Y Flugelman
- Department of Cardiovascular Medicine, Lady Davis Carmel Medical Center, Haifa 32000, Israel
| | - Shulamit Levenberg
- Biomedical Engineering Department, Technion-Israel Institute of Technology, Haifa 32000, Israel.
| |
Collapse
|
11
|
Flugelman MY, Halak M, Yoffe B, Schneiderman J, Rubinstein C, Bloom AI, Weinmann E, Goldin I, Ginzburg V, Mayzler O, Hoffman A, Koren B, Gershtein D, Inbar M, Hutoran M, Tsaba A. Phase Ib Safety, Two-Dose Study of MultiGeneAngio in Patients with Chronic Critical Limb Ischemia. Mol Ther 2017; 25:816-825. [PMID: 28143739 DOI: 10.1016/j.ymthe.2016.12.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/10/2016] [Accepted: 12/15/2016] [Indexed: 01/22/2023] Open
Abstract
Critical limb ischemia (CLI) is the most severe presentation of peripheral arterial disease. We developed cell-based therapy entailing intra-arterial injection of autologous venous endothelial cells (ECs) modified to express angiopoietin 1, combined with autologous venous smooth muscle cells (SMCs) modified to express vascular endothelial growth factor. This combination promoted arteriogenesis in animal models and was safe in patients with limiting claudication. In an open-label, phase Ib study, we assessed the safety and efficacy of this therapy in CLI patients who failed or were unsuitable for surgery or intravascular intervention. Of 23 patients enrolled, 18 with rest pain or non-healing ulcers (Rutherford categories 4 and 5) were treated according to protocol, and 5 with significant tissue loss (Rutherford 6) were treated under compassionate treatment. Patients were assigned randomly to receive 1 × 107 or 5 × 107 (EC-to-SMC ratio, 1:1) of the cell combination. One-year amputation-free survival rate was 72% (13/18) for Rutherford 4 and 5 patients; all 5 patients with Rutherford 6 underwent amputation. Of the 12 with unhealing ulcers at dosing, 6 had complete healing and 2 others had >66% reduction in ulcer size. Outcomes did not differ between the dose groups. No severe adverse events were observed related to the therapy.
Collapse
Affiliation(s)
- Moshe Y Flugelman
- Department of Cardiovascular Medicine, Lady Davis Carmel Medical Center, Haifa 3436212, Israel; Rappaport Faculty of Medicine, Technion IIT, Haifa 3200003, Israel; VESSL Therapeutics Ltd., Haifa 3436212, Israel.
| | - Moshe Halak
- Department of Vascular Surgery, Chaim Sheba Medical Center, Ramat Gan 5265601, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Boris Yoffe
- Department of General and Vascular Surgery, Barzilai Medical Center, Ashkelon 7830604, Israel; The Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8499000, Israel
| | - Jacob Schneiderman
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Chen Rubinstein
- Departments of Vascular Surgery and Radiology, Hadassah University Hospital, Jerusalem 91120, Israel
| | - Allan-Isaac Bloom
- Departments of Vascular Surgery and Radiology, Hadassah University Hospital, Jerusalem 91120, Israel
| | - Eran Weinmann
- Department of Vascular Surgery, Kaplan Medical Center, Rehovot 76100, Israel
| | - Ilya Goldin
- Department of Vascular Surgery, Shaare Zedek Medical Center, Jerusalem 9103102, Israel
| | - Victor Ginzburg
- The Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8499000, Israel; Department of Vascular Surgery, Soroka Medical Center, Beer-Sheva 8410101, Israel
| | - Olga Mayzler
- The Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8499000, Israel; Department of Vascular Surgery, Soroka Medical Center, Beer-Sheva 8410101, Israel
| | - Aaron Hoffman
- Rappaport Faculty of Medicine, Technion IIT, Haifa 3200003, Israel; Department of Vascular Surgery, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Belly Koren
- VESSL Therapeutics Ltd., Haifa 3436212, Israel
| | | | | | | | - Adili Tsaba
- Rappaport Faculty of Medicine, Technion IIT, Haifa 3200003, Israel; VESSL Therapeutics Ltd., Haifa 3436212, Israel
| |
Collapse
|
12
|
Preis M, Schneiderman J, Koren B, Ben-Yosef Y, Levin-Ashkenazi D, Shapiro S, Cohen T, Blich M, Israeli-Amit M, Sarnatzki Y, Gershtein D, Shofti R, Lewis BS, Shaul Y, Flugelman MY. Co-expression of fibulin-5 and VEGF165 increases long-term patency of synthetic vascular grafts seeded with autologous endothelial cells. Gene Ther 2015; 23:237-46. [PMID: 26588709 DOI: 10.1038/gt.2015.104] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Revised: 10/12/2015] [Accepted: 10/15/2015] [Indexed: 12/30/2022]
Abstract
Small caliber synthetic vascular grafts are commonly used for bypass surgery and dialysis access sites but have high failure rates because of neointima formation and thrombosis. Seeding synthetic grafts with endothelial cells (ECs) provides a biocompatible surface that may prevent graft failure. However, EC detachment following exposure to blood flow still remains a major obstacle in the development of biosynthetic grafts. We tested the hypothesis that induced expression by the seeded EC, of vascular endothelial growth factor165 (VEGF165) and of fibulin-5, an extracellular matrix glycoprotein that has a crucial role in elastin fiber organization and increase EC adherence to surfaces, may improve long-term graft patency. Autologous ECs were isolated from venous segments, and were transduced with retroviral vectors expressing fibulin-5 and VEGF165. The modified cells were seeded on expanded polytetrafluoroethylene (ePTFE) grafts and implanted in a large animal model. Three months after transplantation, all grafts seeded with modified EC were patent on a selective angiography, whereas only a third of the control grafts were patent. Similar results were shown at 6 months. Thus, seeding ePTFE vascular grafts with genetically modified EC improved long-term small caliber graft patency. The biosynthetic grafts may provide a novel therapeutic modality for patients with peripheral vascular disease and patients requiring vascular access for hemodialysis.
Collapse
Affiliation(s)
- M Preis
- Institute of Hematology, Lady Davis Carmel Medical Center, Haifa, Israel
| | - J Schneiderman
- Department of Vascular Surgery, Sheba Medical Center, Tel Hashomer and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - B Koren
- Department of Cardiovascular Medicine, Lady Davis Carmel Medical Center, Haifa, Israel.,MultiGene Vascular Systems Ltd, Haifa, Israel
| | - Y Ben-Yosef
- Department of Cardiovascular Medicine, Lady Davis Carmel Medical Center, Haifa, Israel.,MultiGene Vascular Systems Ltd, Haifa, Israel
| | - D Levin-Ashkenazi
- The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - S Shapiro
- Immunology Research Unit, Lady Davis Carmel Medical Center, Haifa, Israel
| | - T Cohen
- Department of Cardiovascular Medicine, Lady Davis Carmel Medical Center, Haifa, Israel.,MultiGene Vascular Systems Ltd, Haifa, Israel
| | - M Blich
- Department of Cardiovascular Medicine, Lady Davis Carmel Medical Center, Haifa, Israel
| | - M Israeli-Amit
- Department of Cardiovascular Medicine, Lady Davis Carmel Medical Center, Haifa, Israel.,MultiGene Vascular Systems Ltd, Haifa, Israel
| | - Y Sarnatzki
- Department of Cardiovascular Medicine, Lady Davis Carmel Medical Center, Haifa, Israel.,MultiGene Vascular Systems Ltd, Haifa, Israel
| | - D Gershtein
- Department of Cardiovascular Medicine, Lady Davis Carmel Medical Center, Haifa, Israel.,MultiGene Vascular Systems Ltd, Haifa, Israel
| | - R Shofti
- The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - B S Lewis
- Department of Cardiovascular Medicine, Lady Davis Carmel Medical Center, Haifa, Israel.,MultiGene Vascular Systems Ltd, Haifa, Israel.,The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Y Shaul
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - M Y Flugelman
- Department of Cardiovascular Medicine, Lady Davis Carmel Medical Center, Haifa, Israel.,MultiGene Vascular Systems Ltd, Haifa, Israel.,The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
13
|
Grossman PM, Mohler ER, Roessler BJ, Wilensky RL, Levine BL, Woo EY, Upchurch GR, Schneiderman J, Koren B, Hutoran M, Gershstein D, Flugelman MY. Phase I study of multi-gene cell therapy in patients with peripheral artery disease. Vasc Med 2015; 21:21-32. [PMID: 26584888 DOI: 10.1177/1358863x15612148] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
UNLABELLED Alternative treatment strategies for claudication are needed and cell-based therapies designed to induce angiogenesis are promising. The purpose of this report was to conduct a Phase I safety, dose-escalating, non-randomized, open-label study of autologous, fully differentiated venous endothelial and smooth muscle cells called MultiGeneAngio (MGA) for claudication due to peripheral artery disease. Twelve subjects, at two centers, received a single intra-arterial infusion of a suspension of equal amounts of transduced autologous venous smooth muscle cells expressing vascular endothelial growth factor (VEGF165) and endothelial cells expressing angiopoietin-1 (Ang-1) (Cohort 1: 1 × 10(7), Cohort 2: 2 × 10(7), Cohort 3: 5 × 10(7), Cohort 4: 7 × 10(7)). The treatment was given unblinded and in the more symptomatic lower extremity. Transduced cells were tested for in vitro doubling time, telomerase activity, and gene expression. The main outcomes were clinical safety and tolerability. Other safety measures included ankle-brachial index (ABI) and walking time on a treadmill. All subjects were male (mean age 60 ± 5 years) including 25% with diabetes mellitus. At 1-year follow-up, there was one serious adverse event possibly related to MGA. Safety endpoints including VEGF and Ang-1 plasma protein levels were within normal ranges in all subjects. The mean maximal walking time increased from baseline to 1 year and the index limb ABI was unchanged, indicating no safety concerns. MGA, an autologous, transduced, cell-based therapy was well tolerated and safe in this Phase I study. Further evaluation is warranted in randomized human studies. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT00390767.
Collapse
Affiliation(s)
- P Michael Grossman
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Emile R Mohler
- Department of Medicine, Cardiovascular Division, Vascular Medicine Section, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Blake J Roessler
- Michigan Institute for Clinical and Health Research, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Robert L Wilensky
- Department of Medicine, Cardiovascular Division, Vascular Medicine Section, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Bruce L Levine
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Edward Y Woo
- Department of Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Gilbert R Upchurch
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Jacob Schneiderman
- Department of Vascular Surgery, Sheba Medical Center, Tel Hashomer, Israel
| | - Belly Koren
- Department of Cardiovascular Medicine, Lady Davis Carmel Medical Center, Bruce Rappaport Faculty of Medicine, Technion IIT, Haifa, Israel MultiGene Vascular Systems Ltd, Haifa, Israel
| | - Marina Hutoran
- Department of Cardiovascular Medicine, Lady Davis Carmel Medical Center, Bruce Rappaport Faculty of Medicine, Technion IIT, Haifa, Israel MultiGene Vascular Systems Ltd, Haifa, Israel
| | - Diana Gershstein
- Department of Cardiovascular Medicine, Lady Davis Carmel Medical Center, Bruce Rappaport Faculty of Medicine, Technion IIT, Haifa, Israel MultiGene Vascular Systems Ltd, Haifa, Israel
| | - Moshe Y Flugelman
- Department of Cardiovascular Medicine, Lady Davis Carmel Medical Center, Bruce Rappaport Faculty of Medicine, Technion IIT, Haifa, Israel MultiGene Vascular Systems Ltd, Haifa, Israel
| |
Collapse
|
14
|
Masuda H, Tanaka R, Fujimura S, Ishikawa M, Akimaru H, Shizuno T, Sato A, Okada Y, Iida Y, Itoh J, Itoh Y, Kamiguchi H, Kawamoto A, Asahara T. Vasculogenic conditioning of peripheral blood mononuclear cells promotes endothelial progenitor cell expansion and phenotype transition of anti-inflammatory macrophage and T lymphocyte to cells with regenerative potential. J Am Heart Assoc 2014; 3:e000743. [PMID: 24965023 PMCID: PMC4309104 DOI: 10.1161/jaha.113.000743] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Background Cell‐based therapies involving mononuclear cells (MNCs) have been developed for vascular regeneration to treat ischemic diseases; however, quality control of therapeutic MNCs has not been evaluated. We investigated the therapeutic potential of peripheral blood (PB) MNCs, operated by recently developed quality and quantity (QQ) culture of endothelial progenitor cells (EPCs). Methods and Results PBs were collected from healthy volunteers; peripheral blood mononuclear cells (PBMNCs) isolated from these PBs were subjected to QQ culture for 7 days with medium containing stem cell factor, thrombopoietin, Flt‐3 ligand, vascular endothelial growth factor, and interleukin‐6. The resulting cells (QQMNCs) in EPC colony‐forming assay generated significantly more definitive EPC colonies than PBMNCs. In flow cytometry, macrophages and helper T lymphocytes of QQMNCs became phenotypically polarized into angiogenic, anti‐inflammatory, and regenerative subsets: classical M1 to alternative M2; T helper (Th)1 to Th2; angiogenic or regulatory T‐cell expansion. Quantitative real‐time polymerase chain reaction (qRT‐PCR) assay revealed the predominant proangiogenic gene expressions in QQMNCs versus PBMNCs. Using murine ischemic hindlimb models, the efficacy of QQMNC intramuscular transplantation (Tx) was compared to that of PBMNCTx, cultured “early EPC” Tx (eEPCTx), and granulocyte colony‐stimulating factor mobilized CD34+ cell Tx (GmCD34Tx). Laser Doppler imaging revealed the blood perfusion recovery in ischemic hindlimbs after QQMNCTx superior to after PBMNCTx and eEPCTx, but also earlier than after GmCD34Tx. Histological evaluations and qRT‐PCR assays in ischemic hindlimbs demonstrated that QQMNCTx, similarly to GmCD34Tx, enhanced angiovasculogenesis and myogenesis, whereas it preponderantly inhibited inflammation and fibrosis versus PBMNCTx and eEPCTx. Conclusions QQ culture potentiates the ability of PBMNCs to promote regeneration of injured tissue; considering the feasible cell preparation, QQ culture‐treated PBMNCs may provide a promising therapeutic option for ischemic diseases. Clinical Trial Registration URL: irb.med.u-tokai.ac.jp/d/2/monthly/2010.html; IRB No.: 10R‐020. URL: irb.med.u-tokai.ac.jp/d/2/monthly/201312.html; IRB No.: 13R228.
Collapse
Affiliation(s)
- Haruchika Masuda
- Department of Regenerative Medicine Science, Tokai University School of Medicine, Isehara, Japan (H.M., T.S., A.S., T.A.)
| | - Rica Tanaka
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan (R.T., S.F.)
| | - Satoshi Fujimura
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan (R.T., S.F.)
| | - Masakazu Ishikawa
- Department of Orthopedic Surgery, Graduate School of Biomedical Science, Hiroshima University, Hiroshima, Japan (M.I.)
| | - Hiroshi Akimaru
- Vascular Regeneration Research Group, Institute of Biomedical Research and Innovation IBRI, Kobe, Japan (H.A., A.K., T.A.)
| | - Tomoko Shizuno
- Department of Regenerative Medicine Science, Tokai University School of Medicine, Isehara, Japan (H.M., T.S., A.S., T.A.)
| | - Atsuko Sato
- Department of Regenerative Medicine Science, Tokai University School of Medicine, Isehara, Japan (H.M., T.S., A.S., T.A.)
| | - Yoshinori Okada
- Teaching and Research Support Core Center, Tokai University School of Medicine, Isehara, Japan (Y.O., Y.I., J.I., Y.I., H.K.)
| | - Yumi Iida
- Teaching and Research Support Core Center, Tokai University School of Medicine, Isehara, Japan (Y.O., Y.I., J.I., Y.I., H.K.)
| | - Jobu Itoh
- Teaching and Research Support Core Center, Tokai University School of Medicine, Isehara, Japan (Y.O., Y.I., J.I., Y.I., H.K.)
| | - Yoshiko Itoh
- Teaching and Research Support Core Center, Tokai University School of Medicine, Isehara, Japan (Y.O., Y.I., J.I., Y.I., H.K.)
| | - Hiroshi Kamiguchi
- Teaching and Research Support Core Center, Tokai University School of Medicine, Isehara, Japan (Y.O., Y.I., J.I., Y.I., H.K.)
| | - Atsuhiko Kawamoto
- Vascular Regeneration Research Group, Institute of Biomedical Research and Innovation IBRI, Kobe, Japan (H.A., A.K., T.A.)
| | - Takayuki Asahara
- Department of Regenerative Medicine Science, Tokai University School of Medicine, Isehara, Japan (H.M., T.S., A.S., T.A.) Vascular Regeneration Research Group, Institute of Biomedical Research and Innovation IBRI, Kobe, Japan (H.A., A.K., T.A.)
| |
Collapse
|
15
|
Silvestre JS, Smadja DM, Lévy BI. Postischemic revascularization: from cellular and molecular mechanisms to clinical applications. Physiol Rev 2013; 93:1743-802. [PMID: 24137021 DOI: 10.1152/physrev.00006.2013] [Citation(s) in RCA: 171] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
After the onset of ischemia, cardiac or skeletal muscle undergoes a continuum of molecular, cellular, and extracellular responses that determine the function and the remodeling of the ischemic tissue. Hypoxia-related pathways, immunoinflammatory balance, circulating or local vascular progenitor cells, as well as changes in hemodynamical forces within vascular wall trigger all the processes regulating vascular homeostasis, including vasculogenesis, angiogenesis, arteriogenesis, and collateral growth, which act in concert to establish a functional vascular network in ischemic zones. In patients with ischemic diseases, most of the cellular (mainly those involving bone marrow-derived cells and local stem/progenitor cells) and molecular mechanisms involved in the activation of vessel growth and vascular remodeling are markedly impaired by the deleterious microenvironment characterized by fibrosis, inflammation, hypoperfusion, and inhibition of endogenous angiogenic and regenerative programs. Furthermore, cardiovascular risk factors, including diabetes, hypercholesterolemia, hypertension, diabetes, and aging, constitute a deleterious macroenvironment that participates to the abrogation of postischemic revascularization and tissue regeneration observed in these patient populations. Thus stimulation of vessel growth and/or remodeling has emerged as a new therapeutic option in patients with ischemic diseases. Many strategies of therapeutic revascularization, based on the administration of growth factors or stem/progenitor cells from diverse sources, have been proposed and are currently tested in patients with peripheral arterial disease or cardiac diseases. This review provides an overview from our current knowledge regarding molecular and cellular mechanisms involved in postischemic revascularization, as well as advances in the clinical application of such strategies of therapeutic revascularization.
Collapse
|
16
|
|
17
|
Staudacher DL, Sela Y, Itskovitz-Eldor J, Flugelman MY. Intra-arterial injection of human embryonic stem cells in athymic rat hind limb ischemia model leads to arteriogenesis. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2011; 12:228-34. [PMID: 21367671 DOI: 10.1016/j.carrev.2010.11.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Revised: 11/20/2010] [Accepted: 11/23/2010] [Indexed: 01/16/2023]
Abstract
UNLABELLED Shear stress can enhance differentiation of human embryonic stem cells (hESC) to vascular cells. We tested the hypothesis that intra-arterial hESC injection will lead to arteriogenesis while intramuscular injection will have no effect on vascularization. METHODS AND RESULTS The superficial femoral arteries were excised on both hind limbs in athymic rats. hESC (2×10(6)) were injected intra-arterially (shear stress) or intramuscular (no shear stress) in one limb after arterial excision. Blood flow, muscle perfusion, and number of arteries/mm(2) muscle were studied at 10 and 21 days after injection. Blood flow in the common iliac artery improved significantly at 10 days after intra-arterial injection of hESC (22±9%, P<.02), and tight muscle perfusion improved significantly both at 10 and 21 days (9±2%, 16±5% respectively, both P<.02). In comparison, intramuscular injection of hESC did not affect blood flow at 10 and 21 days (-3±10% and 4±6%, respectively), while perfusion showed no significant effect of hESC injection after 10 days (1±8%) and was increased 21 days after hESC injection (11±5%, P=.03). Arterial density did not improve after intra-arterial hESC injection at 10 days (15±13%, P=.15) and significantly improved at 21 days (13±4%, P<.05). No significant change was demonstrated after intramuscular injection. SUMMARY Intra-arterial injection of hESC resulted in moderate improvement of flow and perfusion and increased number of arteries in the ischemic hind limb. No consistent change in perfusion, flow, and number of arteries was observed after intramuscular injection.
Collapse
Affiliation(s)
- Dawid L Staudacher
- Department of Cardiovascular Medicine, Lady Davis Carmel Medical Center, Haifa, Israel
| | | | | | | |
Collapse
|
18
|
Zacharek A, Shehadah A, Chen J, Cui X, Roberts C, Lu M, Chopp M. Comparison of bone marrow stromal cells derived from stroke and normal rats for stroke treatment. Stroke 2010; 41:524-30. [PMID: 20056925 DOI: 10.1161/strokeaha.109.568881] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND PURPOSE We compared the effect of treatment of stroke with bone marrow stromal cells from stroke rats (Isch-BMSC) and normal rats (Nor-BMSC) on functional outcome. METHODS Isch-BMSCs and Nor-BMSCs were intravenously injected into rats 24 hours after middle cerebral artery occlusion. To test the mechanism of Isch-BMSC-enhanced neurorestoration, Isch-BMSC and Nor-BMSC cultures were used. RESULTS Isch-BMSC significantly promoted functional outcome and enhanced angiogenesis, arterial density, and axonal regeneration compared with Nor-BMSC treatment animals. Isch-BMSCs exhibited increased Angiopoietin-1, Tie2, basic fibroblast growth factor, glial cell-derived neurotrophic factor, vascular endothelial growth factor, and Flk1 gene expression compared with Nor-BMSC. Using transwell coculture of BMSCs with brain-derived endothelial cells, Isch-BMSCs increased phosphorylated-Tie2 activity in brain-derived endothelial cells and enhanced brain-derived endothelial cells capillary tube formation compared with Nor-BMSCs. Inhibition of Tie2 gene expression in brain-derived endothelial cells using siRNA significantly attenuated BMSC-induced capillary tube formation. CONCLUSIONS These data suggest that Isch-BMSCs are superior to Nor-BMSCs for the neurorestorative treatment of stroke, which may be mediated by the enhanced trophic factor and angiogenic characteristics of Isch-BMSCs.
Collapse
Affiliation(s)
- Alex Zacharek
- Department of Neurology, Henry Ford Health Sciences Center, Detroit, Mich 48202, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Following an arterial occlusion outward remodeling of pre-existent inter-connecting arterioles occurs by proliferation of vascular smooth muscle and endothelial cells. This is initiated by deformation of the endothelial cells through increased pulsatile fluid shear stress (FSS) caused by the steep pressure gradient between the high pre-occlusive and the very low post-occlusive pressure regions that are interconnected by collateral vessels. Shear stress leads to the activation and expression of all NOS isoforms and NO production, followed by endothelial VEGF secretion, which induces MCP-1 synthesis in endothelium and in the smooth muscle of the media. This leads to attraction and activation of monocytes and T-cells into the adventitial space (peripheral collateral vessels) or attachment of these cells to the endothelium (coronary collaterals). Mononuclear cells produce proteases and growth factors to digest the extra-cellular scaffold and allow motility and provide space for the new cells. They also produce NO from iNOS, which is essential for arteriogenesis. The bulk of new tissue production is carried by the smooth muscles of the media, which transform their phenotype from a contractile into a synthetic and proliferative one. Important roles are played by actin binding proteins like ABRA, cofilin, and thymosin beta 4 which determine actin polymerization and maturation. Integrins and connexins are markedly up-regulated. A key role in this concerted action which leads to a 2-to-20 fold increase in vascular diameter, depending on species size (mouse versus human) are the transcription factors AP-1, egr-1, carp, ets, by the Rho pathway and by the Mitogen Activated Kinases ERK-1 and -2. In spite of the enormous increase in tissue mass (up to 50-fold) the degree of functional restoration of blood flow capacity is incomplete and ends at 30% of maximal conductance (coronary) and 40% in the vascular periphery. The process of arteriogenesis can be drastically stimulated by increases in FSS (arterio-venous fistulas) and can be completely blocked by inhibition of NO production, by pharmacological blockade of VEGF-A and by the inhibition of the Rho-pathway. Pharmacological stimulation of arteriogenesis, important for the treatment of arterial occlusive diseases, seems feasible with NO donors.
Collapse
|
20
|
Sela S, Itin A, Natanson-Yaron S, Greenfield C, Goldman-Wohl D, Yagel S, Keshet E. A Novel Human-Specific Soluble Vascular Endothelial Growth Factor Receptor 1. Circ Res 2008; 102:1566-74. [DOI: 10.1161/circresaha.108.171504] [Citation(s) in RCA: 164] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
A human-specific splicing variant of vascular endothelial growth factor (VEGF) receptor 1 (Flt1) was discovered, producing a soluble receptor (designated sFlt1-14) that is qualitatively different from the previously described soluble receptor (sFlt1) and functioning as a potent VEGF inhibitor. sFlt1-14 is generated in a cell type-specific fashion, primarily in nonendothelial cells. Notably, in vascular smooth muscle cells, all Flt1 messenger RNA is converted to sFlt1-14, whereas endothelial cells of the same human vessel express sFlt1. sFlt1-14 expression by vascular smooth muscle cells is dynamically regulated as evidenced by its upregulation on coculture with endothelial cells or by direct exposure to VEGF. Increased production of soluble VEGF receptors during pregnancy is entirely attributable to induced expression of placental sFlt1-14 starting by the end of the first trimester. Expression is dramatically elevated in the placenta of women with preeclampsia, specifically induced in abnormal clusters of degenerative syncytiotrophoblasts known as syncytial knots, where it may undergo further messenger RNA editing. sFlt1-14 is the predominant VEGF-inhibiting protein produced by the preeclamptic placenta, accumulates in the circulation, and hence is capable of neutralizing VEGF in distant organs affected in preeclampsia. Together, these findings revealed a new natural VEGF inhibitor that has evolved in humans, possibly to protect nonendothelial cells from adverse VEGF signaling. Furthermore, the study uncovered the identity of a VEGF-blocking protein implicated in preeclampsia.
Collapse
Affiliation(s)
- Shay Sela
- From the Department of Molecular Biology (S.S., A.I., E.K.), the Hebrew University-Hadassah Medical School; and Department of Obstetrics and Gynecology (S.N.-Y., C.G., D.G.-W., S.Y.), Hadassah University Hospital-Mount Scopus, Jerusalem, Israel
| | - Ahuva Itin
- From the Department of Molecular Biology (S.S., A.I., E.K.), the Hebrew University-Hadassah Medical School; and Department of Obstetrics and Gynecology (S.N.-Y., C.G., D.G.-W., S.Y.), Hadassah University Hospital-Mount Scopus, Jerusalem, Israel
| | - Shira Natanson-Yaron
- From the Department of Molecular Biology (S.S., A.I., E.K.), the Hebrew University-Hadassah Medical School; and Department of Obstetrics and Gynecology (S.N.-Y., C.G., D.G.-W., S.Y.), Hadassah University Hospital-Mount Scopus, Jerusalem, Israel
| | - Caryn Greenfield
- From the Department of Molecular Biology (S.S., A.I., E.K.), the Hebrew University-Hadassah Medical School; and Department of Obstetrics and Gynecology (S.N.-Y., C.G., D.G.-W., S.Y.), Hadassah University Hospital-Mount Scopus, Jerusalem, Israel
| | - Debra Goldman-Wohl
- From the Department of Molecular Biology (S.S., A.I., E.K.), the Hebrew University-Hadassah Medical School; and Department of Obstetrics and Gynecology (S.N.-Y., C.G., D.G.-W., S.Y.), Hadassah University Hospital-Mount Scopus, Jerusalem, Israel
| | - Simcha Yagel
- From the Department of Molecular Biology (S.S., A.I., E.K.), the Hebrew University-Hadassah Medical School; and Department of Obstetrics and Gynecology (S.N.-Y., C.G., D.G.-W., S.Y.), Hadassah University Hospital-Mount Scopus, Jerusalem, Israel
| | - Eli Keshet
- From the Department of Molecular Biology (S.S., A.I., E.K.), the Hebrew University-Hadassah Medical School; and Department of Obstetrics and Gynecology (S.N.-Y., C.G., D.G.-W., S.Y.), Hadassah University Hospital-Mount Scopus, Jerusalem, Israel
| |
Collapse
|