1
|
Faber JE. Collateral blood vessels in stroke and ischemic disease: Formation, physiology, rarefaction, remodeling. J Cereb Blood Flow Metab 2025:271678X251322378. [PMID: 40072222 PMCID: PMC11904929 DOI: 10.1177/0271678x251322378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Collateral blood vessels are unique, naturally occurring endogenous bypass vessels that provide alternative pathways for oxygen delivery in obstructive arterial conditions and diseases. Surprisingly however, the capacity of the collateral circulation to provide protection varies greatly among individuals, resulting in a significant fraction having poor collateral circulation in their tissues. We recently reviewed evidence that the presence of naturally-occurring polymorphisms in genes that determine the number and diameter of collaterals that form during development (ie, genetic background), is a major contributor to this variation. The purpose of this review is to summarize current understanding of the other determinants of collateral blood flow, drawing on both animal and human studies. These include the level of smooth muscle tone in collaterals, hemodynamic forces, how collaterals form during development (collaterogenesis), de novo formation of additional new collaterals during adulthood, loss of collaterals with aging and cardiovascular risk factor presence (rarefaction), and collateral remodeling (structural lumen enlargement). We also review emerging evidence that collaterals not only provide protection in ischemic conditions but may also serve a physiological function in healthy individuals. Primary focus is on studies conducted in brain, however relevant findings in other tissues are also reviewed, as are questions for future investigation.
Collapse
Affiliation(s)
- James E Faber
- Department of Cell Biology and Physiology, Curriculum in Neuroscience, McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
2
|
Smadja DM, Berkane Y, Bentounes NK, Rancic J, Cras A, Pinault C, Ouarne M, Paucod E, Rachidi W, Lellouch AG, Jeljeli M. Immune-privileged cord blood-derived endothelial colony-forming cells: advancing immunomodulation and vascular regeneration. Angiogenesis 2025; 28:19. [PMID: 40047974 PMCID: PMC11885380 DOI: 10.1007/s10456-025-09973-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/25/2025] [Indexed: 03/09/2025]
Abstract
Cord blood-derived endothelial colony-forming cells (CB-ECFCs) hold significant promise for regenerative medicine due to their unique vasculogenic and immunomodulatory properties. These cells exhibit a superior proliferative capacity, robust ability to form vascular networks, and lower immunogenicity compared to adult and embryonic stem cell-derived counterparts. The immune-privileged characteristics of CB-ECFCs, including reduced expression of pro-inflammatory mediators and tolerance-inducing molecules such as HLA-G, further enhance their therapeutic potential. Their low immunogenicity minimizes the risk of immune rejection, making them suitable for allogenic cell therapies. Their application extends to complex tissue engineering and organ revascularization, where their ability to integrate into three-dimensional scaffolds and support vascular tree formation represents a significant advancement. Moreover, CB-ECFCs' capability to adapt to inflammatory stimuli and retain immunological memory highlights their functional versatility in dynamic microenvironments. This review highlights the remarkable ontogeny of ECFCs while unveiling the unparalleled potential of CB-ECFCs in revolutionizing regenerative medicine. From pre-vascularizing engineered tissues and organoids to pioneering cell-based therapies for cardiovascular, dermatological, and degenerative diseases, CB-ECFCs stand at the forefront of cutting-edge biomedical advancements, offering unprecedented opportunities for therapeutic innovation. By leveraging their vasculogenic, immune-regulatory, and regenerative capacities, CB-ECFCs offer a robust alternative for addressing the challenges of vascular repair and organ engineering. Future research should focus on unraveling their transcriptomic and functional profiles to optimize clinical applications and advance the field of regenerative medicine.
Collapse
Affiliation(s)
- David M Smadja
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France.
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Paris, F-75015, France.
| | - Yanis Berkane
- Department of Plastic, Reconstructive and Aesthetic Surgery, Hôpital Sud, CHU Rennes, University of Rennes, Rennes, France
- SITI Laboratory, UMR INSERM 1236, Rennes University Hospital, Rennes, France
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Nun K Bentounes
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Paris, F-75015, France
| | - Jeanne Rancic
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Paris, F-75015, France
| | - Audrey Cras
- Cell Therapy Department, AP-HP, Saint-Louis Hospital, Paris, F-75010, France
| | - Cécile Pinault
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Paris, F-75015, France
| | - Marie Ouarne
- Univ. Grenoble Alpes, CEA, INSERM, IRIG-BGE UA13, Grenoble, 38000, France
| | - Elise Paucod
- Univ. Grenoble Alpes, CEA, INSERM, IRIG-BGE UA13, Grenoble, 38000, France
| | - Walid Rachidi
- Univ. Grenoble Alpes, CEA, INSERM, IRIG-BGE UA13, Grenoble, 38000, France
| | - Alexandre G Lellouch
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Paris, F-75015, France
- Department of Plastic, Reconstructive and Aesthetic Surgery, Cedars Sinai Hospital, Los Angeles, USA
| | - Maxime Jeljeli
- Department of Plastic, Reconstructive and Aesthetic Surgery, Cedars Sinai Hospital, Los Angeles, USA
| |
Collapse
|
3
|
Lin YC, Juan YH, Al Deek NF, Tsun-Ching Chang BS, Yu-Jr Lin BS, Chang CJ, Wei FC. Fate of the Flexor Hallucis Longus Muscle at the Donor Site After Fibula Flap Harvest: Assessing Muscle Viability Using Novel MRI Techniques - A Cohort Study. Microsurgery 2025; 45:e70047. [PMID: 40129170 DOI: 10.1002/micr.70047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 01/12/2025] [Accepted: 02/21/2025] [Indexed: 03/26/2025]
Abstract
BACKGROUND Fibula flap dissection sacrifices the blood vessels to the flexor hallucis longus (FHL) and other calf muscles. This novel MRI study investigates perfusion and fibrosis of the FHL muscle after fibula flap harvest. MATERIAL AND METHODS A cohort study from September 2018 to January 2021, we prospectively recruited head and neck cancer patients who were planned to receive fibula flap transfer for jaw reconstruction. All participants received MRI before and one year after the operation. Muscle fibrosis was quantified via extracellular volume matrix fraction (ECV); muscle perfusion via time to peak (TTP), T2* change, and T2* slope by BOLD sequence. Muscle fibrosis and perfusion were compared before and after the operation. RESULTS Total of 18 patients completed the study (mean age: 48.83 years). Significantly increased fibrosis was seen in the postoperative FHL muscle via elevated ECV (11.23%-32.54%, p < 0.001). Despite prolonged TTP in postoperative FHL (38.17-51.83 s, p = 0.343), increased T2* change (8.43%-9.53%, p = 0.369), and increased T2* slope (0.22%-0.23%/s, p = 0.766), these muscle perfusion changes were not statistically significant. Postoperative complications, such as great toe clawing and donor site infection, were not observed. CONCLUSIONS Despite severe fibrosis within the FHL muscle after extensive dissection during fibula harvest and peroneal arterial ligation, the FHL muscle perfusion could be retained from an MRI perspective.
Collapse
Affiliation(s)
- Yu-Ching Lin
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou and Taoyuan, Institute for Radiological Research, Taoyuan, Taiwan
- College of Medicine, Chang Gung Univeristy, Taoyuan, Taiwan
| | - Yu-Hsiang Juan
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou and Taoyuan, Institute for Radiological Research, Taoyuan, Taiwan
- College of Medicine, Chang Gung Univeristy, Taoyuan, Taiwan
| | - Nidal F Al Deek
- College of Medicine, Chang Gung Univeristy, Taoyuan, Taiwan
- Division of Reconstructive Microsurgery, Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- University Hospitals Cleveland Medical Center & Case Western Reserve School of Medicine, Cleveland, Ohio, USA
| | - B S Tsun-Ching Chang
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Keelung, Keelung, Taiwan
| | - B S Yu-Jr Lin
- Research Services Center for Health Information, Chang Gung University, Taoyuan, Taiwan
| | - Chee-Jen Chang
- College of Medicine, Chang Gung Univeristy, Taoyuan, Taiwan
- Research Services Center for Health Information, Chang Gung University, Taoyuan, Taiwan
| | - Fu-Chan Wei
- College of Medicine, Chang Gung Univeristy, Taoyuan, Taiwan
- Division of Reconstructive Microsurgery, Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| |
Collapse
|
4
|
Smadja DM, Mauge L, Rancic J, Gaussem P, Feraud O, Oudrhiri N, Bennaceur-Griscelli A. Comparative Evaluation of Endothelial Colony-Forming Cells from Cord and Adult Blood vs. Human Embryonic Stem Cell-Derived Endothelial Cells: Insights into Therapeutic Angiogenesis Potential. Stem Cell Rev Rep 2025; 21:581-588. [PMID: 39612122 DOI: 10.1007/s12015-024-10830-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
The discovery of endothelial progenitor cells has revolutionized our understanding of postnatal blood vessel formation, with endothelial colony-forming cells (ECFCs) emerging as key players in vasculogenesis. Among various ECFC sources, cord blood-derived ECFCs (CB-ECFCs) are of particular interest due to their superior proliferative and clonogenic potential and their ability to promote vascular network formation. Human embryonic stem cell-derived endothelial cells (hESC-ECs) have also shown potential in regenerative medicine, though their vasculogenic efficacy remains unclear compared to CB- and adult blood-derived ECFCs (AB-ECFCs). This study aimed to directly compare the angiogenic and vasculogenic capabilities of CB-ECFCs, AB-ECFCs, and hESC-ECs in vitro and in vivo. Our results demonstrated that CB-ECFCs had a significantly higher proliferation rate than both AB-ECFCs and hESC-ECs (p < 0.01). In tube formation assays, CB-ECFCs exhibited superior ability to form capillary-like structures compared to hESC-ECs (p < 0.0001) and AB-ECFCs (p < 0.01). In vivo, CB-ECFCs significantly improved blood flow recovery in ischemic tissue (p < 0.01), outperforming both AB-ECFCs and hESC-ECs, with no significant recovery observed in the latter two groups. These findings suggest that CB-ECFCs represent a more effective cell source for therapeutic angiogenesis, while further optimization is needed to enhance the efficacy of hESC-ECs for clinical applications. Future research should explore the molecular mechanisms underlying the superior regenerative potential of CB-ECFCs and focus on improving the stability and functionality of stem cell-derived ECs for therapeutic use.
Collapse
Affiliation(s)
- David M Smadja
- Université Paris Cité, INSERM, Innovative Therapies in Hemostasis, Paris, F-75006, France.
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Paris, F-75015, France.
| | - Laetitia Mauge
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Paris, F-75015, France
- Université Paris Cité, INSERM, PARCC, Paris, F-75015, France
| | - Jeanne Rancic
- Université Paris Cité, INSERM, Innovative Therapies in Hemostasis, Paris, F-75006, France
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Paris, F-75015, France
| | - Pascale Gaussem
- Université Paris Cité, INSERM, Innovative Therapies in Hemostasis, Paris, F-75006, France
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Paris, F-75015, France
| | - Olivier Feraud
- INSERM U935/U1310 ESTeam Paris Sud Human Pluripotent Stem Cell Core Facility, Villejuif, France
| | - Noufissa Oudrhiri
- INSERM U935/U1310 ESTeam Paris Sud Human Pluripotent Stem Cell Core Facility, Villejuif, France
- CITHERA UMS45, Infrastructure-INGESTEM, INSERM, Université Paris-Saclay, Faculté de Médecine, Kremlin Bicêtre, France
- Hematology Department, AP-HP, Hôpital Universitaire Paris Sud-Kremlin Bicêtre, Kremlin-Bicêtre, France
| | - Annelise Bennaceur-Griscelli
- INSERM U935/U1310 ESTeam Paris Sud Human Pluripotent Stem Cell Core Facility, Villejuif, France.
- CITHERA UMS45, Infrastructure-INGESTEM, INSERM, Université Paris-Saclay, Faculté de Médecine, Kremlin Bicêtre, France.
- Hematology Department, AP-HP, Hôpital Universitaire Paris Sud-Kremlin Bicêtre, Kremlin-Bicêtre, France.
| |
Collapse
|
5
|
Chen B, Chen J, Shen Z, Wang W, Li J, Liu S, Cai H, Lu S. The Inhibition of γ-Aminobutyric Acid B1 Receptor Regulates Angiogenesis via the Hippo/YAP Signaling Pathway. Ann Vasc Surg 2024; 109:370-381. [PMID: 39025214 DOI: 10.1016/j.avsg.2024.05.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 05/07/2024] [Accepted: 05/18/2024] [Indexed: 07/20/2024]
Abstract
Promoting the establishment of collateral circulation is essential for chronic lower extremity ischemia. However, no effective therapeutic drugs have yet been developed. Recent studies discovered that in the peripheral arteries, there are γ-aminobutyric acid B1 (GABAB1) receptors expressed in endothelial cells and smooth muscle cells, these receptors may have some effects in regulating vascular functions, but the precise mechanism is not yet clear. This study explores the effect of GABAB1 receptor inhibition on angiogenesis and its regulatory mechanism. The expression of GABAB1 in human umbilical vein endothelial cells (HUVECs) was knocked down using shRNA transfection, and effects on HUVECs' proliferation, migration, and tube formation ability were detected. Western blot and RT-PCR were used to verify the signal pathway. The murine hind limb ischemia model was used to verify the effect of CGP35348, an antagonist of GABAB1R, on the recovery of blood flow perfusion and angiogenesis in ischemic tissues. Cell proliferation, migration, and tube formation ability were improved after GABAB1 receptor knockdown in HUVECs. The phosphorylation of the HIPPO/Yes-associated protein (YAP) pathway decreased, while the effect of promoting angiogenesis increased. After treating the ischemic hindlimbs of mice with GABAB1 receptor antagonists, the blood flow perfusion recovered and the angiogenesis increased. These findings demonstrate the effect of GABAB1 receptor inhibition on the HIPPO/YAP pathway in regulating angiogenesis, suggesting that inhibiting GABAB1 receptor levels might be a novel approach for chronic lower extremity ischemia diseases.
Collapse
MESH Headings
- Animals
- Humans
- Signal Transduction
- Hindlimb
- Cell Proliferation/drug effects
- Protein Serine-Threonine Kinases/metabolism
- Protein Serine-Threonine Kinases/genetics
- Neovascularization, Physiologic/drug effects
- Human Umbilical Vein Endothelial Cells/metabolism
- Hippo Signaling Pathway
- Cell Movement
- Ischemia/physiopathology
- Ischemia/metabolism
- Ischemia/genetics
- Disease Models, Animal
- YAP-Signaling Proteins/metabolism
- Receptors, GABA-B/metabolism
- Receptors, GABA-B/genetics
- Mice, Inbred C57BL
- Adaptor Proteins, Signal Transducing/metabolism
- Adaptor Proteins, Signal Transducing/genetics
- Cells, Cultured
- Phosphorylation
- GABA-B Receptor Antagonists/pharmacology
- Male
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Regional Blood Flow
- Collateral Circulation
- Cell Cycle Proteins/metabolism
- Cell Cycle Proteins/genetics
- Muscle, Skeletal/blood supply
- Muscle, Skeletal/metabolism
- Angiogenesis
Collapse
Affiliation(s)
- Bingyi Chen
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jinxing Chen
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zekun Shen
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Weiyi Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiayan Li
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shuang Liu
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hui Cai
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Shaoying Lu
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
6
|
Smadja DM. Extracellular Microvesicles vs. Mitochondria: Competing for the Top Spot in Cardiovascular Regenerative Medicine. Stem Cell Rev Rep 2024; 20:1813-1818. [PMID: 38976143 DOI: 10.1007/s12015-024-10758-8] [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] [Accepted: 06/25/2024] [Indexed: 07/09/2024]
Abstract
Regenerative medicine aims to restore, replace, and regenerate human cells, tissues, and organs. Despite significant advancements, many cell therapy trials for cardiovascular diseases face challenges like cell survival and immune compatibility, with benefits largely stemming from paracrine effects. Two promising therapeutic tools have been recently emerged in cardiovascular diseases: extracellular vesicles (EVs) and mitochondrial transfer. Concerning EVs, the first pivotal study with EV-enriched secretome derived from cardiovascular progenitor cells has been done treating heart failure. This first in man demonstrated the safety and feasibility of repeated intravenous infusions and highlighted significant clinical improvements, including enhanced cardiac function and reduced symptoms in heart failure patients. The second study uncovered a novel mechanism of endothelial regeneration through mitochondrial transfer via tunneling nanotubes (TNTs). This research showed that mesenchymal stromal cells (MSCs) transfer mitochondria to endothelial cells, significantly enhancing their bioenergetics and vessel-forming capabilities. This mitochondrial transfer was crucial for endothelial cell engraftment and function, offering a new strategy for vascular regeneration without the need for additional cell types. Combining EV and mitochondrial strategies presents new clinical opportunities. These approaches could revolutionize regenerative medicine, offering new hope for treating cardiovascular and other degenerative diseases. Continued research and clinical trials will be crucial in optimizing these therapies, potentially leading to personalized medicine approaches that enhance patient outcomes.
Collapse
Affiliation(s)
- David M Smadja
- Université Paris Cité, INSERM, Innovative Therapies in Hemostasis, Paris, F-75006, France.
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Inserm UMR-S1140, 56 rue Leblanc, Paris, F-75015, France.
| |
Collapse
|
7
|
Quax PHA, Deindl E. The Intriguing World of Vascular Remodeling, Angiogenesis, and Arteriogenesis. Int J Mol Sci 2024; 25:6376. [PMID: 38928082 PMCID: PMC11204171 DOI: 10.3390/ijms25126376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Vascular remodeling is a very general feature related to angiogenesis and arteriogenesis, which are involved in neovascularization processes [...].
Collapse
Affiliation(s)
- Paul H. A. Quax
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Elisabeth Deindl
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität, 81377 Munich, Germany
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, Planegg-Martinsried, 82152 Munich, Germany
| |
Collapse
|
8
|
Yan J, Tie G, Tutto A, Messina LM. Hypercholesterolemia impairs collateral artery enlargement by ten-eleven translocation 1-dependent hematopoietic stem cell autonomous mechanism in a murine model of limb ischemia. JVS Vasc Sci 2024; 5:100203. [PMID: 38774713 PMCID: PMC11106542 DOI: 10.1016/j.jvssci.2024.100203] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 03/22/2024] [Indexed: 05/24/2024] Open
Abstract
Objective The extent of collateral artery enlargement determines the risk of limb loss due to peripheral arterial disease. Hypercholesterolemia impairs collateral artery enlargement, but the underlying mechanism remains poorly characterized. This study tests the hypothesis that hypercholesterolemia impairs collateral artery enlargement through a ten-eleven translocation 1 (Tet1)-dependent hematopoietic stem cell (HSC)-autonomous mechanism that increases their differentiation into proinflammatory Ly6Chi monocytes and restricts their conversion into proangiogenic Ly6Clow monocytes. Methods To test our hypothesis, we induced limb ischemia and generated chimeric mouse models by transplanting HSCs from either wild-type (WT) mice or hypercholesterolemic mice into lethally irradiated WT recipient mice. Results We found that the lethally irradiated WT recipient mice reconstituted with HSCs from hypercholesterolemic mice displayed lower blood flow recovery and collateral artery enlargement that was nearly identical to that observed in hypercholesterolemic mice, despite the absence of hypercholesterolemia and consistent with an HSC-autonomous mechanism. We showed that hypercholesterolemia impairs collateral artery enlargement by a Tet1-dependent mechanism that increases HSC differentiation toward proinflammatory Ly6Chi monocytes and restricts the conversion of Ly6Chi monocytes into proangiogenic Ly6Clow monocytes. Moreover, Tet1 epigenetically reprograms monocyte gene expression within the HSCs. Restoration of Tet1 expression in HSCs of hypercholesterolemic mice restores WT collateral artery enlargement and blood flow recovery after induction of hindlimb ischemia. Conclusions These results show that hypercholesterolemia impairs collateral artery enlargement by a novel Tet1-dependent HSC-autonomous mechanism that epigenetically reprograms monocyte gene expression within the HSCs.
Collapse
Affiliation(s)
- Jinglian Yan
- Division of Vascular and Endovascular Surgery, University of Massachusetts Medical School, Worcester, MA
| | - Guodong Tie
- Division of Vascular and Endovascular Surgery, University of Massachusetts Medical School, Worcester, MA
| | - Amanda Tutto
- Division of Vascular and Endovascular Surgery, University of Massachusetts Medical School, Worcester, MA
| | - Louis M. Messina
- Division of Vascular and Endovascular Surgery, University of Massachusetts Medical School, Worcester, MA
| |
Collapse
|
9
|
Cheng B, Fu X. The Role of Stem Cell on Wound Healing After Revascularization-Healing Following Revascularization-Unlocking Skin Potential. INT J LOW EXTR WOUND 2024; 23:63-69. [PMID: 37899578 DOI: 10.1177/15347346231210709] [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: 10/31/2023]
Abstract
Wound healing is a complex and dynamic process involving a series of cellular and molecular events. Revascularization, the restoration of blood flow to ischemic or damaged tissue, is a key step in wound healing. Adequate vascularization has been recognized as a necessary factor for successful tissue regeneration. In the later stage of revascularization and tissue remodeling in wound healing, stem cells regulate other repair cells and matrix formation by influencing the maturation of blood vessels. The reductive oxidation (REDOX) state may be a key mechanism through stem/progenitor cells to influence endothelial cells to mature blood vessels and improve the quality of healing. Mitochondria may play an important role in this process.
Collapse
Affiliation(s)
- Biao Cheng
- Department of Burns and Plastic Surgery, General Hospital of Southern Theater Command of PLA, Guangzhou, China
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College; Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, P. R. China
| |
Collapse
|
10
|
Smadja DM, Rossi E, Haviari S, Bieche I, Cras A, Gaussem P. Thrombin receptor PAR1 silencing in endothelial colony-forming cells modifies stemness and vasculogenic properties. J Thromb Haemost 2023; 21:3640-3648. [PMID: 37678550 DOI: 10.1016/j.jtha.2023.08.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/09/2023] [Accepted: 08/28/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND The involvement of thrombin receptor PAR1 in blood vessel development has been largely demonstrated in knockout mice; however, its implication in adult mouse angiogenesis seems very moderate. OBJECTIVES We aimed to explore the potential relationships between PAR1, stemness, and angiogenic properties of human endothelial colony-forming cells (ECFCs). METHODS AND RESULTS PAR1 activation on ECFCs using the selective PAR1-activating peptide induced a significant decrease in CD133 expression (RTQ-PCR analysis). In line, silencing of PAR1 gene expression with siRNA increased CD133 mRNA as well as intracellular CD133 protein expression. To confirm the link between CD133 and PAR1, we explored the association between PAR1 and CD133 levels in fast and slow fibroblasts prone to reprogramming. An imbalance between PAR1 and CD133 levels was evidenced, with a decreased expression of PAR1 in fast reprogramming fibroblasts expressing a high CD133 level. Regarding in vitro ECFC angiogenic properties, PAR1 silencing with specific siRNA induced cell proliferation evidenced by the overexpression of Ki67. However, it did not impact migration properties nor ECFC adhesion on smooth muscle cells or human arterial endothelial cells. In a mouse model of hind-limb ischemia, PAR1 silencing in ECFCs significantly increased postischemic revascularization compared to siCtrl-ECFCs along with a significant increase in cutaneous blood flows (P < .0001), microvessel density (P = .02), myofiber regeneration (P < .0001), and human endothelial cell incorporation in muscle (P < .0001). CONCLUSION In conclusion, our work describes for the first time a link between PAR1, stemness, and vasculogenesis in human ECFCs.
Collapse
Affiliation(s)
- David M Smadja
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Paris, France; Université Paris-Cité, INSERM UMR-S 1140, Innovative Therapies in Haemostasis, Paris, France.
| | - Elisa Rossi
- Université Paris-Cité, INSERM UMR-S 1140, Innovative Therapies in Haemostasis, Paris, France
| | - Skerdi Haviari
- Université Paris-Cité, INSERM UMR-S 1137 (IAME), Paris, France; Département Épidémiologie Biostatistiques et Recherche Clinique, AP-HP, Hôpital Bichat, Paris, France
| | | | - Audrey Cras
- Université Paris-Cité, INSERM UMR-S 1140, Innovative Therapies in Haemostasis, Paris, France; Cell Therapy Unit, AP-HP, Saint Louis Hospital, Paris, France
| | - Pascale Gaussem
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Paris, France; Université Paris-Cité, INSERM UMR-S 1140, Innovative Therapies in Haemostasis, Paris, France
| |
Collapse
|
11
|
Sun Z, Yang L, Kiram A, Yang J, Yang Z, Xiao L, Yin Y, Liu J, Mao Y, Zhou D, Yu H, Zhou Z, Xu D, Jia Y, Ding C, Guo Q, Wang H, Li Y, Wang L, Fu T, Hu S, Gan Z. FNIP1 abrogation promotes functional revascularization of ischemic skeletal muscle by driving macrophage recruitment. Nat Commun 2023; 14:7136. [PMID: 37932296 PMCID: PMC10628247 DOI: 10.1038/s41467-023-42690-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 10/18/2023] [Indexed: 11/08/2023] Open
Abstract
Ischaemia of the heart and limbs attributable to compromised blood supply is a major cause of mortality and morbidity. The mechanisms of functional angiogenesis remain poorly understood, however. Here we show that FNIP1 plays a critical role in controlling skeletal muscle functional angiogenesis, a process pivotal for muscle revascularization during ischemia. Muscle FNIP1 expression is down-regulated by exercise. Genetic overexpression of FNIP1 in myofiber causes limited angiogenesis in mice, whereas its myofiber-specific ablation markedly promotes the formation of functional blood vessels. Interestingly, the increased muscle angiogenesis is independent of AMPK but due to enhanced macrophage recruitment in FNIP1-depleted muscles. Mechanistically, myofiber FNIP1 deficiency induces PGC-1α to activate chemokine gene transcription, thereby driving macrophage recruitment and muscle angiogenesis program. Furthermore, in a mouse hindlimb ischemia model of peripheral artery disease, the loss of myofiber FNIP1 significantly improved the recovery of blood flow. Thus, these results reveal a pivotal role of FNIP1 as a negative regulator of functional angiogenesis in muscle, offering insight into potential therapeutic strategies for ischemic diseases.
Collapse
Affiliation(s)
- Zongchao Sun
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Likun Yang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Abdukahar Kiram
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Jing Yang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Zhuangzhuang Yang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Liwei Xiao
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Yujing Yin
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Jing Liu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Yan Mao
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Danxia Zhou
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Hao Yu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Zheng Zhou
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Dengqiu Xu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Yuhuan Jia
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Chenyun Ding
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Qiqi Guo
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Hongwei Wang
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Yan Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Li Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Tingting Fu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China.
| | - Shijun Hu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China.
| | - Zhenji Gan
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China.
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing University, Nanjing, China.
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China.
| |
Collapse
|
12
|
Caradonna E, Mormone E, Centritto EM, Mazzanti A, Papini S, Fanelli M, Petrella L, Petruzziello A, Farina MA, Farina E, Amato B, De Filippo CM, Vanoli E. Different methods of bone marrow harvesting influence cell characteristics and purity, affecting clinical outcomes. JVS Vasc Sci 2023; 4:100130. [PMID: 38058747 PMCID: PMC10696233 DOI: 10.1016/j.jvssci.2023.100130] [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: 07/14/2023] [Accepted: 09/17/2023] [Indexed: 12/08/2023] Open
Abstract
Background Bone marrow (BM)-derived stem cells were implanted to induce angiogenesis in patients with no-option critical limb-threatening ischemia. Considering the potential for this therapy, conflicting results related to BM harvesting methods have been reported that could affect stem cell concentrations and quality. Methods A total of 75 patients with no-option critical limb-threatening ischemia were treated with BM implantation. For 58 patients, BM was harvested using a BM aspirate concentrate system (Harvest Technologies; group HT) with a standard aspiration needle, followed by an automated centrifugation process, to produce BM aspirate concentrate. For 17 patients, BM was harvested using the Marrow Cellution system (Aspire Medical Innovation; group MC). CD34+ cells/mL, CD117+ cells/mL, CD133+ cells/mL, CD309+ cells/mL, hematocrit, and BM purity were compared between the two BM preparations. Results The retrospective analysis of a subset group after adjustment for age shows that the quality of BM obtained using the Marrow Cellution system is better, in terms of purity, than the classic harvesting method before centrifugation. Harvested BM before centrifugation is characterized by a higher percentage of CD133+ cells compared with BM after centrifugation. In contrast, the MC aspirate had a larger amount of very small embryonic-like cells, as indicated by the higher percentage of CD133+, CD34+, and CD45- cells. These differences translated into an increased occurrence of leg amputations in group HT than in group MC and an increase in transcutaneous oxygen pressure in patients treated with BM aspirated using MC. Conclusions BM manipulation, such as centrifugation, affects the quality and number of stem cells, with detrimental consequences on clinical outcomes, as reflected by the different amputation rates between the two groups.
Collapse
Affiliation(s)
| | - Elisabetta Mormone
- Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies, Fondazione IRCCS Casa Sollievo della Sofferenza, Foggia, Italy
| | | | - Andrea Mazzanti
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Unit of Molecular Cardiology, ICS Maugeri, Pavia, Italy
| | - Stefano Papini
- Clinical and Research Laboratory, Gemelli Molise S.p.A., Campobasso, Italy
| | - Mara Fanelli
- Laboratorio di Diagnostica Molecolare, Gemelli Molise S.p.A., Campobasso, Italy
| | - Lella Petrella
- Laboratorio di Diagnostica Molecolare, Gemelli Molise S.p.A., Campobasso, Italy
| | - Arnolfo Petruzziello
- UOC Patologia Clinica, Dipartimento dei Servizi Sanitari, AORN CASERTA, Caserta, Italy
| | | | | | - Bruno Amato
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | | | - Emilio Vanoli
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Cardiology Unit, Sacra Famiglia Hospital, Erba, Italy
| |
Collapse
|
13
|
Sacco AM, Castaldo C, Di Meglio FD, Nurzynska D, Palermi S, Spera R, Gnasso R, Zinno G, Romano V, Belviso I. The Long and Winding Road to Cardiac Regeneration. APPLIED SCIENCES 2023; 13:9432. [DOI: 10.3390/app13169432] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Cardiac regeneration is a critical endeavor in the treatment of heart diseases, aimed at repairing and enhancing the structure and function of damaged myocardium. This review offers a comprehensive overview of current advancements and strategies in cardiac regeneration, with a specific focus on regenerative medicine and tissue engineering-based approaches. Stem cell-based therapies, which involve the utilization of adult stem cells and pluripotent stem cells hold immense potential for replenishing lost cardiomyocytes and facilitating cardiac tissue repair and regeneration. Tissue engineering also plays a prominent role employing synthetic or natural biomaterials, engineering cardiac patches and grafts with suitable properties, and fabricating upscale bioreactors to create functional constructs for cardiac recovery. These constructs can be transplanted into the heart to provide mechanical support and facilitate tissue healing. Additionally, the production of organoids and chips that accurately replicate the structure and function of the whole organ is an area of extensive research. Despite significant progress, several challenges persist in the field of cardiac regeneration. These include enhancing cell survival and engraftment, achieving proper vascularization, and ensuring the long-term functionality of engineered constructs. Overcoming these obstacles and offering effective therapies to restore cardiac function could improve the quality of life for individuals with heart diseases.
Collapse
Affiliation(s)
- Anna Maria Sacco
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| | - Clotilde Castaldo
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| | - Franca Di Di Meglio
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| | - Daria Nurzynska
- Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, 84081 Baronissi, Italy
| | - Stefano Palermi
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| | - Rocco Spera
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| | - Rossana Gnasso
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| | - Giorgio Zinno
- Department of Clinical and Surgical Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Veronica Romano
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| | - Immacolata Belviso
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
14
|
Wirth G, Juusola G, Tarvainen S, Laakkonen JP, Korpisalo P, Ylä-Herttuala S. Capillary Dynamics Regulate Post-Ischemic Muscle Damage and Regeneration in Experimental Hindlimb Ischemia. Cells 2023; 12:2060. [PMID: 37626870 PMCID: PMC10453415 DOI: 10.3390/cells12162060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/20/2023] [Accepted: 07/23/2023] [Indexed: 08/27/2023] Open
Abstract
This study aimed to show the significance of capillary function in post-ischemic recovery from the perspective of physiological parameters, such as blood flow, hemoglobin oxygenation and tissue regeneration. Muscle-level microvascular alterations of blood flow and hemoglobin oxygenation, and post-ischemic myofiber and capillary responses were analyzed in aged, healthy C57Bl/6J mice (n = 48) and aged, hyperlipidemic LDLR-/-ApoB100/100 mice (n = 69) after the induction of acute hindlimb ischemia using contrast ultrasound, photoacoustic imaging and histological analyses, respectively. The capillary responses that led to successful post-ischemic muscle repair in C57Bl/6J mice included an early capillary dilation phase, preceding the return of arterial driving pressure, followed by an increase in capillary density that further supported satellite cell-induced muscle regeneration. Initial capillary enlargement was absent in the LDLR-/-ApoB100/100 mice with lifelong moderate hypercholesterolemia and led to an inability to recover arterial driving pressure, with a resulting increase in distal necrosis, chronic tissue damage and a delay in the overall recovery after ischemia. To conclude, this manuscript highlights, beyond arterial collateralization, the importance of the proper function of the capillary endothelium in post-ischemic recovery and displays how post-ischemic capillary dynamics associate beyond tissue blood flow to both hemoglobin oxygenation and tissue regeneration.
Collapse
Affiliation(s)
- Galina Wirth
- Heart Center, Kuopio University Hospital, FI-70200 Kuopio, Finland (P.K.)
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Greta Juusola
- Heart Center, Kuopio University Hospital, FI-70200 Kuopio, Finland (P.K.)
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Santeri Tarvainen
- Heart Center, Kuopio University Hospital, FI-70200 Kuopio, Finland (P.K.)
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Johanna P. Laakkonen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Petra Korpisalo
- Heart Center, Kuopio University Hospital, FI-70200 Kuopio, Finland (P.K.)
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Seppo Ylä-Herttuala
- Heart Center, Kuopio University Hospital, FI-70200 Kuopio, Finland (P.K.)
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland
| |
Collapse
|
15
|
Berkeley B, Tang MNH, Brittan M. Mechanisms regulating vascular and lymphatic regeneration in the heart after myocardial infarction. J Pathol 2023; 260:666-678. [PMID: 37272582 PMCID: PMC10953458 DOI: 10.1002/path.6093] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/14/2023] [Accepted: 04/27/2023] [Indexed: 06/06/2023]
Abstract
Myocardial infarction, caused by a thrombus or coronary vascular occlusion, leads to irreversible ischaemic injury. Advances in early reperfusion strategies have significantly reduced short-term mortality after myocardial infarction. However, survivors have an increased risk of developing heart failure, which confers a high risk of death at 1 year. The capacity of the injured neonatal mammalian heart to regenerate has stimulated extensive research into whether recapitulation of developmental regeneration programmes may be beneficial in adult cardiovascular disease. Restoration of functional blood and lymphatic vascular networks in the infarct and border regions via neovascularisation and lymphangiogenesis, respectively, is a key requirement to facilitate myocardial regeneration. An improved understanding of the endogenous mechanisms regulating coronary vascular and lymphatic expansion and function in development and in adult patients after myocardial infarction may inform future therapeutic strategies and improve translation from pre-clinical studies. In this review, we explore the underpinning research and key findings in the field of cardiovascular regeneration, with a focus on neovascularisation and lymphangiogenesis, and discuss the outcomes of therapeutic strategies employed to date. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Bronwyn Berkeley
- Centre for Cardiovascular Science, The Queen's Medical Research InstituteUniversity of EdinburghEdinburghUK
| | - Michelle Nga Huen Tang
- Centre for Cardiovascular Science, The Queen's Medical Research InstituteUniversity of EdinburghEdinburghUK
| | - Mairi Brittan
- Centre for Cardiovascular Science, The Queen's Medical Research InstituteUniversity of EdinburghEdinburghUK
| |
Collapse
|
16
|
Wang Y, Zheng Y, Qi B, Liu Y, Cheng X, Feng J, Gao W, Li T. α-Lipoic acid alleviates myocardial injury and induces M2b macrophage polarization after myocardial infarction via HMGB1/NF-kB signaling pathway. Int Immunopharmacol 2023; 121:110435. [PMID: 37320869 DOI: 10.1016/j.intimp.2023.110435] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Myocardial infarction (MI) is a serious cardiovascular disease with a poor prognosis. Macrophages are the predominant immune cells in patients with MI and macrophage regulation during the different phases of MI has important consequences for cardiac recovery. Alpha-lipoic acid (ALA) plays a critical role in MI by modulating the number of cardiomyocytes and macrophages. METHODS MI mice were generated by ligating the left anterior descending coronary artery. Macrophages were exposed to hypoxia to establish a hypoxia model and M1 polarization was induced by LPS and IFN-γ. Different groups of macrophages and MI mice were treated with ALA. The cardiomyocytes were treated with various macrophage supernatants and the cardiac function, cytokine levels, and pathology were also analyzed. Factors related to apoptosis, autophagy, reactive oxygen species (ROS), and the mitochondrial membrane potential (MMP) were assessed. Finally, the HMGB1/NF-κB pathway was identified. RESULTS ALA promoted M2b polarization in normal cells and suppressed inflammatory cytokines during hypoxia. ALA inhibited ROS and MMP production in vitro. Supernatants containing ALA inhibited apoptosis and autophagy in hypoxic cardiomyocytes. Moreover, ALA suppressed the HMGB1/NF-κB pathway in macrophages, which may be a potential mechanism for attenuating MI. CONCLUSION ALA alleviates MI and induces M2b polarization via the HMGB1/NF-κB pathway, impeding inflammation, oxidation, apoptosis, and autophagy, and might be a potential strategy for MI treatment.
Collapse
Affiliation(s)
- Yuchao Wang
- School of Medicine, Nankai University, Tianjin 300071, China; Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China.
| | - Yue Zheng
- School of Medicine, Nankai University, Tianjin 300071, China; Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Bingcai Qi
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Yanwu Liu
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Xuan Cheng
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Jianyu Feng
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Wenqing Gao
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China.
| | - Tong Li
- School of Medicine, Nankai University, Tianjin 300071, China; Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China.
| |
Collapse
|
17
|
Mangiardi M, Bonura A, Iaccarino G, Alessiani M, Bravi MC, Crupi D, Pezzella FR, Fabiano S, Pampana E, Stilo F, Alfano G, Anticoli S. The Pathophysiology of Collateral Circulation in Acute Ischemic Stroke. Diagnostics (Basel) 2023; 13:2425. [PMID: 37510169 PMCID: PMC10378392 DOI: 10.3390/diagnostics13142425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/08/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Cerebral collateral circulation is a network of blood vessels which stabilizes blood flow and maintains cerebral perfusion whenever the main arteries fail to provide an adequate blood supply, as happens in ischemic stroke. These arterial networks are able to divert blood flow to hypoperfused cerebral areas. The extent of the collateral circulation determines the volume of the salvageable tissue, the so-called "penumbra". Clinically, this is associated with greater efficacy of reperfusion therapies (thrombolysis and thrombectomy) in terms of better short- and long-term functional outcomes, lower incidence of hemorrhagic transformation and of malignant oedema, and smaller cerebral infarctions. Recent advancements in brain imaging techniques (CT and MRI) allow us to study these anastomotic networks in detail and increase the likelihood of making effective therapeutic choices. In this narrative review we will investigate the pathophysiology, the clinical aspects, and the possible diagnostic and therapeutic role of collateral circulation in acute ischemic stroke.
Collapse
Affiliation(s)
- Marilena Mangiardi
- Department of Stroke Unit, San Camillo-Forlanini Hospital, 00152 Rome, Italy
| | - Adriano Bonura
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Campus Bio-Medico University, 00128 Rome, Italy
| | - Gianmarco Iaccarino
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Campus Bio-Medico University, 00128 Rome, Italy
| | - Michele Alessiani
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Campus Bio-Medico University, 00128 Rome, Italy
| | - Maria Cristina Bravi
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Campus Bio-Medico University, 00128 Rome, Italy
| | - Domenica Crupi
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Campus Bio-Medico University, 00128 Rome, Italy
| | - Francesca Romana Pezzella
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Campus Bio-Medico University, 00128 Rome, Italy
| | - Sebastiano Fabiano
- Department of Neuroradiology and Interventional Neuroradiology, San Camillo-Forlanini Hospital, 00152 Rome, Italy
| | - Enrico Pampana
- Department of Neuroradiology and Interventional Neuroradiology, San Camillo-Forlanini Hospital, 00152 Rome, Italy
| | - Francesco Stilo
- Unit of Vascular Surgery, Campus Bio-Medico University, 00128 Rome, Italy
| | - Guido Alfano
- Department of Radiology and Interventional Radiology, M.G. Vannini Hospital, 00177 Rome, Italy
| | - Sabrina Anticoli
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Campus Bio-Medico University, 00128 Rome, Italy
| |
Collapse
|
18
|
Sun L, Li X, Luo H, Guo H, Zhang J, Chen Z, Lin F, Zhao G. EZH2 can be used as a therapeutic agent for inhibiting endothelial dysfunction. Biochem Pharmacol 2023; 213:115594. [PMID: 37207700 DOI: 10.1016/j.bcp.2023.115594] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 05/01/2023] [Accepted: 05/04/2023] [Indexed: 05/21/2023]
Abstract
Enhancer of zeste homolog 2 (EZH2) is a catalytic subunit of polycomb repressor complex 2 and plays important roles in endothelial cell homeostasis. EZH2 functionally methylates lysine 27 of histone H3 and represses gene expression through chromatin compaction. EZH2 mediates the effects of environmental stimuli by regulating endothelial functions, such as angiogenesis, endothelial barrier integrity, inflammatory signaling, and endothelial mesenchymal transition. Numerous studies have been conducted to determine the significance of EZH2 in endothelial function. The aim of this review is to provide a concise summary of the roles EZH2 plays in endothelial function and elucidate its therapeutic potential in cardiovascular diseases.
Collapse
Affiliation(s)
- Li Sun
- Cardiovascular Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China; Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| | - Xuefang Li
- Cardiovascular Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China; Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| | - Hui Luo
- Cardiovascular Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China; Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| | - Huige Guo
- Cardiovascular Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China; Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| | - Jie Zhang
- Cardiovascular Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China; Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| | - Zhigang Chen
- Cardiovascular Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China; Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| | - Fei Lin
- Cardiovascular Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China; Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China.
| | - Guoan Zhao
- Cardiovascular Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China; Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China.
| |
Collapse
|
19
|
Silvestre JS. Prostaglandins bet on regulatory T cells to promote therapeutic revascularization. Cardiovasc Res 2023; 119:1097-1099. [PMID: 37114987 DOI: 10.1093/cvr/cvad051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Indexed: 04/29/2023] Open
Affiliation(s)
- Jean-Sébastien Silvestre
- Paris Cardiovascular Research Center, Université Paris Cité, INSERM UMRS 970, 56 rue Leblanc, 75737 Paris Cedex 15, France
| |
Collapse
|
20
|
Im GB, Kim YG, Yoo TY, Kim YH, Kim K, Hyun J, Soh M, Hyeon T, Bhang SH. Ceria Nanoparticles as Copper Chaperones that Activate SOD1 for Synergistic Antioxidant Therapy to Treat Ischemic Vascular Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2208989. [PMID: 36706357 DOI: 10.1002/adma.202208989] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/20/2023] [Indexed: 06/18/2023]
Abstract
All exogenous nanomaterials undergo rapid biotransformation once injected into the body and fall short of executing the intended purpose. Here, it is reported that copper-deposited ceria nanoparticles (CuCe NPs) exhibit enhanced antioxidant effects over pristine ceria nanoparticles, as the released copper buffers the depletion of glutathione while providing the bioavailable copper as a cofactor for the antioxidant enzyme, superoxide dismutase 1. The upregulated intracellular antioxidants along with the ceria nanoparticles synergistically scavenge reactive oxygen species and promote anti-inflammation and M2 polarization of macrophages by modulating signal transducer and activator of transcription 1 and 6 (STAT1 and STAT6). The therapeutic effect of CuCe NPs is demonstrated in ischemic vascular diseases (i.e., murine models of hindlimb ischemia and myocardial infarction) in which the copper-deposition affords increased perfusion and alleviation in tissue damage. The results provide rationale that metal oxide nanomaterials can be designed in a way to induce the upregulation of specific biological factors for optimal therapeutic performance.
Collapse
Affiliation(s)
- Gwang-Bum Im
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Young Geon Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Tae Yong Yoo
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yeong Hwan Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Kang Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jiyu Hyun
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Min Soh
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Taeghwan Hyeon
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
21
|
Sun J, Ge Y, Chao T, Bai R, Wang C. The Role of miRNA in the Regulation of Angiogenesis in Ischemic Heart Disease. Curr Probl Cardiol 2023; 48:101637. [PMID: 36773949 DOI: 10.1016/j.cpcardiol.2023.101637] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 02/04/2023] [Indexed: 02/12/2023]
Abstract
Despite continued improvements in primary prevention and treatment, ischemic heart disease (IHD) is the most common cause of mortality in both developing and developed countries. Promoting angiogenesis and reconstructing vascular network in ischemic myocardium are critical process of postischemic tissue repair. Effective strategies to promote survival and avoid apoptosis of endothelial cells in the ischemic myocardium can help to achieve long-term cardiac angiogenesis. Therefore, it is of great importance to investigate the molecular pathophysiology of angiogenesis in-depth and to find the key targets that promote angiogenesis. Recently years, many studies have found that microRNAs play important regulatory roles in almost all process of angiogenesis, including vascular sprouting, proliferation, survival and migration of vascular endothelial cells, recruitment of vascular progenitor cells, and control of angiopoietin expression. This review presents detailed information about the regulatory role of miRNAs in the angiogenesis of IHD in recent years, and provides new therapeutic ideas for the treatment of IHD.
Collapse
Affiliation(s)
- Jinghui Sun
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yaru Ge
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tiantian Chao
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ruina Bai
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Chenglong Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
22
|
HuMSC-EV induce monocyte/macrophage mobilization to orchestrate neovascularization in wound healing process following radiation injury. Cell Death Dis 2023; 9:38. [PMID: 36725841 PMCID: PMC9892506 DOI: 10.1038/s41420-023-01335-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 01/06/2023] [Accepted: 01/12/2023] [Indexed: 02/03/2023]
Abstract
This study aims to investigate the mechanisms of human mesenchymal stem cell-derived extracellular vesicles (HuMSC-EV)-induced proangiogenic paracrine effects after radiation injury. HuMSC-EV were locally administered in mice hindlimb following 80-Gy X-ray irradiation and animals were monitored at different time points. HuMSC-EV improved neovascularization of the irradiated tissue, by stimulating angiogenesis, normalizing cutaneous blood perfusion, and increasing capillary density and production of proangiogenic factors. HuMSC-EV also stimulated vasculogenesis by promoting the recruitment and differentiation of bone marrow progenitors. Moreover, HuMSC-EV improved arteriogenesis by increasing the mobilization of monocytes from the spleen and the bone marrow and their recruitment into the muscle, with a pro-inflammatory potential. Importantly, monocyte depletion by clodronate treatment abolished the proangiogenic effect of HuMSC-EV. The critical role of Ly6C(hi) monocyte subset in HuMSC-EV-induced neovascularization process was further confirmed using Ccr2-/- mice. This study demonstrates that HuMSC-derived EV enhances the neovascularization process in the irradiated tissue by increasing the production of proangiogenic factors, promoting the recruitment of vascular progenitor cells, and the mobilization of innate cells to the injured site. These results support the concept that HuMSC-EV might represent a suitable alternative to stem cells for therapeutic neovascularization in tissue repair.
Collapse
|
23
|
Li Y, An D, Xie X, Dong Y. The relationship between neutrophil-to-lymphocyte ratio and cerebral collateral circulation in patients with symptomatic severe intracranial artery stenosis or occlusion. J Clin Neurosci 2023; 108:13-18. [PMID: 36565522 DOI: 10.1016/j.jocn.2022.10.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 10/11/2022] [Accepted: 10/24/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND The neutrophil/lymphocyte ratio (NLR) has been considered a prognostic indicator for determining the systemic inflammatory response and atherosclerosis. We aimed to determine the relationship between NLR and the development of cerebral collateral circulation in patients with symptomatic severe stenosis or occlusion of intracranial arteries. METHODS All patients underwent digital subtraction angiography (DSA) within 14 days of admission and were divided into a group with good collateral circulation (77 patients) and a group with poor collateral circulation (86 patients) according to the DSA collateral compensation grading method. Apo B, total cholesterol, LDL, and Neutrophil count in the poor side branch group were significantly higher than in the good side branch group. Multifactorial analysis showed that high NLR levels were a valid predictor of poor collateral circulation in patients with symptomatic severe intracranial artery stenosis or occlusion. Spearman correlation analysis showed that the size of the collateral branch score was negatively correlated with NLR (r = -0.509, P < 0.001) and cholesterol content (r = -0.249, P = 0.002). NLR predicted poor collateral circulation with an AUC of 0.620 (sensitivity 66.7 %, specificity 61.3 %, 95 % CI = 0.517-0.723,P < 0.05). CONCLUSION We demonstrate a correlation between NLR levels and the development of collateral circulation in the brain in patients with symptomatic severe stenosis or occlusion of the intracranial arteries.
Collapse
Affiliation(s)
- Yao Li
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China; Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, China
| | - Dongxia An
- Department of Neurointervention, Beijing Fengtai Youanmen Hospital, Beijing, China; Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, China
| | - Xiaohua Xie
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China; Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, China
| | - Yanhong Dong
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China; Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, China.
| |
Collapse
|
24
|
Zhi Z, Sun Q, Tang W. Research advances and challenges in tissue-derived extracellular vesicles. Front Mol Biosci 2022; 9:1036746. [PMID: 36589228 PMCID: PMC9797684 DOI: 10.3389/fmolb.2022.1036746] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Extracellular vesicles (EV) are vesicular vesicles with phospholipid bilayer, which are present in biological fluids and extracellular microenvironment. Extracellular vesicles serve as pivotal mediators in intercellular communication by delivering lipids, proteins, and RNAs to the recipient cells. Different from extracellular vesicles derived from biofluids and that originate from cell culture, the tissue derived extracellular vesicles (Ti-EVs) send us more enriched and accurate information of tissue microenvironment. Notably, tissue derived extracellular vesicles directly participate in the crosstalk between numerous cell types within microenvironment. Current research mainly focused on the extracellular vesicles present in biological fluids and cell culture supernatant, yet the studies on tissue derived extracellular vesicles are increasing due to the tissue derived extracellular vesicles are promising agents to reflect the occurrence and development of human diseases more accurately. In this review, we aimed to clarify the characteristics of tissue derived extracellular vesicles, specify the isolation methods and the roles of tissue derived extracellular vesicles in various diseases, including tumors. Moreover, we summarized the advances and challenges of tissue derived extracellular vesicles research.
Collapse
|
25
|
GPR174 knockdown enhances blood flow recovery in hindlimb ischemia mice model by upregulating AREG expression. Nat Commun 2022; 13:7519. [PMID: 36473866 PMCID: PMC9727025 DOI: 10.1038/s41467-022-35159-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Regulatory T cells (Tregs) are critically involved in neovascularization, an important compensatory mechanism in peripheral artery disease. The contribution of G protein coupled receptor 174 (GPR174), which is a regulator of Treg function and development, in neovascularization remains elusive. Here, we show that genetic deletion of GPR174 in Tregs potentiated blood flow recovery in mice after hindlimb ischemia. GPR174 deficiency upregulates amphiregulin (AREG) expression in Tregs, thereby enhancing endothelial cell functions and reducing pro-inflammatory macrophage polarization and endothelial cell apoptosis. Mechanically, GPR174 regulates AREG expression by inhibiting the nuclear accumulation of early growth response protein 1 (EGR1) via Gαs/cAMP/PKA signal pathway activation. Collectively, these findings demonstrate that GPR174 negatively regulates angiogenesis and vascular remodeling in response to ischemic injury and that GPR174 may be a potential molecular target for therapeutic interventions of ischemic vascular diseases.
Collapse
|
26
|
Mameli E, Martello A, Caporali A. Autophagy at the interface of endothelial cell homeostasis and vascular disease. FEBS J 2022; 289:2976-2991. [PMID: 33934518 DOI: 10.1111/febs.15873] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 03/16/2021] [Accepted: 04/09/2021] [Indexed: 12/19/2022]
Abstract
Autophagy is an essential intracellular process for cellular quality control. It enables cell homeostasis through the selective degradation of harmful protein aggregates and damaged organelles. Autophagy is essential for recycling nutrients, generating energy to maintain cell viability in most tissues and during adverse conditions such as hypoxia/ischaemia. The progressive understanding of the mechanisms modulating autophagy in the vasculature has recently led numerous studies to link intact autophagic responses with endothelial cell (EC) homeostasis and function. Preserved autophagic flux within the ECs has an essential role in maintaining their physiological characteristics, whereas defective autophagy can promote endothelial pro-inflammatory and atherogenic phenotype. However, we still lack a good knowledge of the complete molecular repertoire controlling various aspects of endothelial autophagy and how this is associated with vascular diseases. Here, we provide an overview of the current state of the art of autophagy in ECs. We review the discoveries that have so far defined autophagy as an essential mechanism in vascular biology and analyse how autophagy influences ECs behaviour in vascular disease. Finally, we emphasise opportunities for compounds to regulate autophagy in ECs and discuss the challenges of exploiting them to resolve vascular disease.
Collapse
Affiliation(s)
- Eleonora Mameli
- University/BHF Centre for Cardiovascular Science, QMRI, University of Edinburgh, UK
| | | | - Andrea Caporali
- University/BHF Centre for Cardiovascular Science, QMRI, University of Edinburgh, UK
| |
Collapse
|
27
|
Madonna R. Angiocrine endothelium: From physiology to atherosclerosis and cardiac repair. Vascul Pharmacol 2022; 144:106993. [DOI: 10.1016/j.vph.2022.106993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 03/25/2022] [Accepted: 03/30/2022] [Indexed: 02/08/2023]
|
28
|
Spadaccio C, Nenna A, Rose D, Piccirillo F, Nusca A, Grigioni F, Chello M, Vlahakes GJ. The Role of Angiogenesis and Arteriogenesisin Myocardial Infarction and Coronary Revascularization. J Cardiovasc Transl Res 2022; 15:1024-1048. [PMID: 35357670 DOI: 10.1007/s12265-022-10241-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 03/18/2022] [Indexed: 12/25/2022]
Abstract
Surgical myocardial revascularization is associated with long-term survival benefit in patients with multivessel coronary artery disease. However, the exact biological mechanisms underlying the clinical benefits of myocardial revascularization have not been elucidated yet. Angiogenesis and arteriogenesis biologically leading to vascular collateralization are considered one of the endogenous mechanisms to preserve myocardial viability during ischemia, and the presence of coronary collateralization has been regarded as one of the predictors of long-term survival in patients with coronary artery disease (CAD). Some experimental studies and indirect clinical evidence on chronic CAD confirmed an angiogenetic response induced by myocardial revascularization and suggested that revascularization procedures could constitute an angiogenetic trigger per se. In this review, the clinical and basic science evidence regarding arteriogenesis and angiogenesis in both CAD and coronary revascularization is analyzed with the aim to better elucidate their significance in the clinical arena and potential therapeutic use.
Collapse
Affiliation(s)
- Cristiano Spadaccio
- Cardiac Surgery, Massachusetts General Hospital & Harvard Medical School, Boston, USA. .,Cardiac Surgery, Golden Jubilee National Hospital & University of Glasgow, Glasgow, UK.
| | - Antonio Nenna
- Cardiac Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - David Rose
- Cardiac Surgery, Lancashire Cardiac Centre, Blackpool Victoria Hospital, Blackpool, UK
| | | | | | | | - Massimo Chello
- Cardiac Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Gus J Vlahakes
- Cardiac Surgery, Massachusetts General Hospital & Harvard Medical School, Boston, USA
| |
Collapse
|
29
|
Tian Y, Fopiano KA, Buncha V, Lang L, Rudic RD, Filosa JA, Dou H, Bagi Z. Aging-induced impaired endothelial wall shear stress mechanosensing causes arterial remodeling via JAM-A/F11R shedding by ADAM17. GeroScience 2022; 44:349-369. [PMID: 34718985 PMCID: PMC8810930 DOI: 10.1007/s11357-021-00476-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 10/08/2021] [Indexed: 11/25/2022] Open
Abstract
Physiological and pathological vascular remodeling is uniquely driven by mechanical forces from blood flow in which wall shear stress (WSS) mechanosensing by the vascular endothelium plays a pivotal role. This study aimed to determine the novel role for a disintegrin and metalloproteinase 17 (ADAM17) in impaired WSS mechanosensing, which was hypothesized to contribute to aging-associated abnormal vascular remodeling. Without changes in arterial blood pressure and blood flow rate, skeletal muscle resistance arteries of aged mice (30-month-old vs. 12-week-old) exhibited impaired WSS mechanosensing and displayed inward hypertrophic arterial remodeling. These vascular changes were recapitulated by in vivo confined, AAV9-mediated overexpression of ADAM17 in the resistance arteries of young mice. An aging-related increase in ADAM17 expression reduced the endothelial junction level of its cleavage substrate, junctional adhesion molecule-A/F11 receptor (JAM-A/F11R). In cultured endothelial cells subjected to steady WSS ADAM17 activation or JAM-A/F11R knockdown inhibited WSS mechanosensing. The ADAM17-activation induced, impaired WSS mechanosensing was normalized by overexpression of ADAM17 cleavage resistant, mutated JAM-AV232Y both in cultured endothelial cells and in resistance arteries of aged mice, in vivo. These data demonstrate a novel role for ADAM17 in JAM-A/F11R cleavage-mediated impaired endothelial WSS mechanosensing and subsequently developed abnormal arterial remodeling in aging. ADAM17 could prove to be a key regulator of WSS mechanosensing, whereby it can also play a role in pathological vascular remodeling in diseases.
Collapse
Affiliation(s)
- Yanna Tian
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Katie Anne Fopiano
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Vadym Buncha
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Liwei Lang
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - R Daniel Rudic
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Jessica A Filosa
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Huijuan Dou
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
- Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Zsolt Bagi
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
30
|
Zhang Z, Xu Y, Cao C, Wang B, Guo J, Qin Z, Lu Y, Zhang J, Zhang L, Wang W, Zhang J, Tang J. Exosomes as a messager to regulate the crosstalk between macrophages and cardiomyocytes under hypoxia conditions. J Cell Mol Med 2022; 26:1486-1500. [PMID: 35088943 PMCID: PMC8899199 DOI: 10.1111/jcmm.17162] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/01/2021] [Accepted: 12/19/2021] [Indexed: 12/12/2022] Open
Abstract
Recent studies have confirmed that cardiomyocyte‐derived exosomes have many pivotal biological functions, like influencing the progress of coronary artery disease via modulating macrophage phenotypes. However, the mechanisms underlying the crosstalk between cardiomyocytes and macrophages have not been fully characterized. Hence, this study aimed to observe the interaction between cardiomyocytes under hypoxia and macrophages through exosome communication and further evaluate the ability of exosomes derived from cardiomyocytes cultured under hypoxic conditions (Hypo‐Exo) to polarize macrophages, and the effect of alternatively activated macrophages (M2) on hypoxic cardiomyocytes. Our results revealed that hypoxia facilitated the production of transforming growth factor‐beta (TGF‐β) in H9c2 cell‐derived exosomes. Moreover, exosomes derived from cardiomyocytes cultured under normal conditions (Nor‐Exo) and Hypo‐Exo could induce RAW264.7 cells into classically activated macrophages (M1) and M2 macrophages respectively. Likewise, macrophage activation was induced by circulating exosomes isolated from normal human controls (hNor‐Exo) or patients with acute myocardial infarction (hAMI‐Exo). Thus, our findings support that the profiles of hAMI‐Exo have been changed, which could regulate the polarization of macrophages and subsequently the polarized M2 macrophages reduced the apoptosis of cardiomyocytes in return. Based on our findings, we speculate that exosomes have emerged as important inflammatory response modulators regulating cardiac oxidative stress injury.
Collapse
Affiliation(s)
- Zenglei Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China.,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Yanyan Xu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China.,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Chang Cao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China.,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Bo Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China.,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Jiacheng Guo
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China.,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Zhen Qin
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China.,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Yongzheng Lu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China.,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Jianchao Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China.,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Li Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China.,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Wei Wang
- Henan Medical Association, Zhengzhou, China
| | - Jinying Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China.,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Junnan Tang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China.,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| |
Collapse
|
31
|
Mehanna RA, Essawy MM, Barkat MA, Awaad AK, Thabet EH, Hamed HA, Elkafrawy H, Khalil NA, Sallam A, Kholief MA, Ibrahim SS, Mourad GM. Cardiac stem cells: Current knowledge and future prospects. World J Stem Cells 2022; 14:1-40. [PMID: 35126826 PMCID: PMC8788183 DOI: 10.4252/wjsc.v14.i1.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 07/02/2021] [Accepted: 01/06/2022] [Indexed: 02/06/2023] Open
Abstract
Regenerative medicine is the field concerned with the repair and restoration of the integrity of damaged human tissues as well as whole organs. Since the inception of the field several decades ago, regenerative medicine therapies, namely stem cells, have received significant attention in preclinical studies and clinical trials. Apart from their known potential for differentiation into the various body cells, stem cells enhance the organ's intrinsic regenerative capacity by altering its environment, whether by exogenous injection or introducing their products that modulate endogenous stem cell function and fate for the sake of regeneration. Recently, research in cardiology has highlighted the evidence for the existence of cardiac stem and progenitor cells (CSCs/CPCs). The global burden of cardiovascular diseases' morbidity and mortality has demanded an in-depth understanding of the biology of CSCs/CPCs aiming at improving the outcome for an innovative therapeutic strategy. This review will discuss the nature of each of the CSCs/CPCs, their environment, their interplay with other cells, and their metabolism. In addition, important issues are tackled concerning the potency of CSCs/CPCs in relation to their secretome for mediating the ability to influence other cells. Moreover, the review will throw the light on the clinical trials and the preclinical studies using CSCs/CPCs and combined therapy for cardiac regeneration. Finally, the novel role of nanotechnology in cardiac regeneration will be explored.
Collapse
Affiliation(s)
- Radwa A Mehanna
- Medical Physiology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Marwa M Essawy
- Oral Pathology Department, Faculty of Dentistry/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Mona A Barkat
- Human Anatomy and Embryology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Ashraf K Awaad
- Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Eman H Thabet
- Medical Physiology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Heba A Hamed
- Histology and Cell Biology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Hagar Elkafrawy
- Medical Biochemistry Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Nehal A Khalil
- Medical Biochemistry Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Abeer Sallam
- Medical Physiology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Marwa A Kholief
- Forensic Medicine and Clinical toxicology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Samar S Ibrahim
- Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Ghada M Mourad
- Histology and Cell Biology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt.
| |
Collapse
|
32
|
Mehanna RA, Essawy MM, Barkat MA, Awaad AK, Thabet EH, Hamed HA, Elkafrawy H, Khalil NA, Sallam A, Kholief MA, Ibrahim SS, Mourad GM. Cardiac stem cells: Current knowledge and future prospects. World J Stem Cells 2022. [PMID: 35126826 DOI: 10.4252/wjsc.v14.i1.1]] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Regenerative medicine is the field concerned with the repair and restoration of the integrity of damaged human tissues as well as whole organs. Since the inception of the field several decades ago, regenerative medicine therapies, namely stem cells, have received significant attention in preclinical studies and clinical trials. Apart from their known potential for differentiation into the various body cells, stem cells enhance the organ's intrinsic regenerative capacity by altering its environment, whether by exogenous injection or introducing their products that modulate endogenous stem cell function and fate for the sake of regeneration. Recently, research in cardiology has highlighted the evidence for the existence of cardiac stem and progenitor cells (CSCs/CPCs). The global burden of cardiovascular diseases' morbidity and mortality has demanded an in-depth understanding of the biology of CSCs/CPCs aiming at improving the outcome for an innovative therapeutic strategy. This review will discuss the nature of each of the CSCs/CPCs, their environment, their interplay with other cells, and their metabolism. In addition, important issues are tackled concerning the potency of CSCs/CPCs in relation to their secretome for mediating the ability to influence other cells. Moreover, the review will throw the light on the clinical trials and the preclinical studies using CSCs/CPCs and combined therapy for cardiac regeneration. Finally, the novel role of nanotechnology in cardiac regeneration will be explored.
Collapse
Affiliation(s)
- Radwa A Mehanna
- Medical Physiology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Marwa M Essawy
- Oral Pathology Department, Faculty of Dentistry/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Mona A Barkat
- Human Anatomy and Embryology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Ashraf K Awaad
- Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Eman H Thabet
- Medical Physiology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Heba A Hamed
- Histology and Cell Biology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Hagar Elkafrawy
- Medical Biochemistry Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Nehal A Khalil
- Medical Biochemistry Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Abeer Sallam
- Medical Physiology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Marwa A Kholief
- Forensic Medicine and Clinical toxicology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Samar S Ibrahim
- Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Ghada M Mourad
- Histology and Cell Biology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt.
| |
Collapse
|
33
|
Gui L, Chen Y, Diao Y, Chen Z, Duan J, Liang X, Li H, Liu K, Miao Y, Gao Q, Li Z, Yang J, Li Y. ROS-responsive nanoparticle-mediated delivery of CYP2J2 gene for therapeutic angiogenesis in severe hindlimb ischemia. Mater Today Bio 2022; 13:100192. [PMID: 34988419 PMCID: PMC8695365 DOI: 10.1016/j.mtbio.2021.100192] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/07/2021] [Accepted: 12/17/2021] [Indexed: 11/21/2022] Open
Abstract
With critical limb ischemia (CLI) being a multi-factorial disease, it is becoming evident that gene therapy with a multiple bio-functional growth factor could achieve better therapeutic outcomes. Cytochrome P450 epoxygenase-2J2 (CYP2J2) and its catalytic products epoxyeicosatrienoic acids (EETs) exhibit pleiotropic biological activities, including pro-angiogenic, anti-inflammatory and cardiovascular protective effects, which are considerably beneficial for reversing ischemia and restoring local blood flow in CLI. Here, we designed a nanoparticle-based pcDNA3.1-CYP2J2 plasmid DNA (pDNA) delivery system (nanoparticle/pDNA complex) composed of a novel three-arm star block copolymer (3S-PLGA-po-PEG), which was achieved by conjugating three-armed PLGA to PEG via the peroxalate ester bond. Considering the multiple bio-functions of CYP2J2-EETs and the sensitivity of the peroxalate ester bond to H2O2, this nanoparticle-based gene delivery system is expected to exhibit excellent pro-angiogenic effects while improving the high oxidative stress and inflammatory micro-environment in ischemic hindlimb. Our study reports the first application of CYP2J2 in the field of therapeutic angiogenesis for CLI treatment and our findings demonstrated good biocompatibility, stability and sustained release properties of the CYP2J2 nano-delivery system. In addition, this nanoparticle-based gene delivery system showed high transfection efficiency and efficient VEGF expression in vitro and in vivo. Intramuscular injection of nanoparticle/pDNA complexes into mice with hindlimb ischemia resulted in significant rapid blood flow recovery and improved muscle repair compared to mice treated with naked pDNA. In summary, 3S-PLGA-po-PEG/CYP2J2-pDNA complexes have tremendous potential and provide a practical strategy for the treatment of limb ischemia. Moreover, 3S-PLGA-po-PEG nanoparticles might be useful as a potential non-viral carrier for other gene delivery applications. Cytochrome P450 epoxygenase-2J2 (CYP2J2) was first applied in the field of therapeutic angiogenesis for critical limb ischemia treatment. The ROS-responsive three-arm star block copolymer (3S-PLGA-po-PEG) was synthesized with peroxalate ester as H2O2-responsive linkages through the esterification reaction of oxalyl chloride and hydroxyl group. The CYP2J2 nano-delivery system achieved high transfection efficiency and significant therapeutic angiogenesis effect.
Collapse
Affiliation(s)
- Liang Gui
- Department of Vascular Surgery, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, PR China.,Graduate School of Peking Union Medical College, Beijing, 100730, PR China.,Department of Vascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, PR China
| | - Youlu Chen
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, PR China
| | - Yongpeng Diao
- Department of Vascular Surgery, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, PR China
| | - Zuoguan Chen
- Department of Vascular Surgery, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, PR China
| | - Jianwei Duan
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, PR China
| | - Xiaoyu Liang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, PR China
| | - Huiyang Li
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, PR China
| | - Kaijing Liu
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, PR China
| | - Yuqing Miao
- Department of Vascular Surgery, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, PR China
| | - Qing Gao
- Department of Vascular Surgery, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, PR China
| | - Zhichao Li
- Department of Vascular Surgery, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, PR China
| | - Jing Yang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, PR China
| | - Yongjun Li
- Department of Vascular Surgery, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, PR China
| |
Collapse
|
34
|
Plasma and genetic determinants of soluble TREM-1 and major adverse cardiovascular events in a prospective cohort of acute myocardial infarction patients. Results from the FAST-MI 2010 study. Int J Cardiol 2021; 344:213-219. [PMID: 34534607 DOI: 10.1016/j.ijcard.2021.09.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 09/01/2021] [Accepted: 09/09/2021] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Triggering receptor expressing on myeloid cells (TREM)-1 is involved in the pathophysiology of ischemic heart disease. Plasma soluble TREM-1 levels (sTREM-1) has been associated with increased risk of major adverse cardiovascular events (MACE) in acute myocardial infarction (AMI) patients. However, the causative link between TREM-1 and MACE remains unknown and requires further investigation before developing potential therapeutic approaches. METHODS AND RESULTS Using the serum and DNA data bank from the prospective, nationwide French registry of Acute ST-elevation and non-ST-elevation Myocardial Infarction (FAST-MI 2010, N = 1293), we studied the association of plasma levels of sTREM-1 with 9 common genetic variants at the TREM1 locus and their relationship with recurrent MACE over a 3-year follow up. Plasma levels of sTREM-1 were associated with an increased risk of MACEs (death, recurrent MI or stroke) (adjusted HR = 1.86, 95%CI = 1.06-3.26 and HR = 1.11, 95%CI = 0.61-2.02 respectively for tertiles 3 and 2 versus tertile 1, P < 0.001). The study of common variants identified two major genetic determinants of sTREM-1 (rs4714449: beta = -0.11, Padd = 7.85 × 10-5 and rs3804276: beta = 0.18, Padd = 2.65 × 10-11) with a potential role on maintenance and/or differentiation of hematopoietic stem cells. However, associated variants only explained 4% of sTREM-1 variance (P = 2.74 × 10-14). Moreover, the rs4714449 variant, individually and in haplotype, was not significantly associated with MACE (HR = 0.61, 95%CI: 0.35-1.05, P = 0.07). CONCLUSIONS Despite its relationship with increased risk of death, recurrent MI and stroke, genetic determinants of plasma levels of sTREM-1 were not found to be causal prognostic factors in patients with acute myocardial infarction.
Collapse
|
35
|
Zhu J, Tan J, Zhang C, Jia J, Leng H, Xu Y, Song C. Single Intraosseous Simvastatin Application Induces Endothelial Progenitor Cell Mobilization and Therapeutic Angiogenesis in a Diabetic Hindlimb Ischemia Rat Model. Plast Reconstr Surg 2021; 148:936e-945e. [PMID: 34644264 DOI: 10.1097/prs.0000000000008526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Endothelial progenitor cells have shown the ability to enhance neovascularization. In this study, the authors tested whether intraosseous delivery of simvastatin could mobilize endothelial progenitor cells and enhance recovery in a hindlimb ischemia model. METHODS There are eight groups of rats in this study: normal control; type 1 diabetes mellitus control group control without drug intervention; and type 1 diabetes mellitus rats that randomly received intraosseous simvastatin (0, 0.5, or 1 mg) or oral simvastatin administration (0, 20, or 400 mg). All type 1 diabetes mellitus rats had induced hindlimb ischemia. The number of endothelial progenitor cells in peripheral blood, and serum markers, were detected. The recovery of blood flow at 21 days after treatment was used as the main outcome. RESULTS The authors demonstrated that endothelial progenitor cell mobilization was increased in the simvastatin 0.5- and 1-mg groups compared with the type 1 diabetes mellitus control and simvastatin 0-mg groups at 1, 2, and 3 weeks. Serum vascular endothelial growth factor levels were significantly increased at 2 weeks in the simvastatin 0.5- and 1-mg groups, in addition to the increase of the blood flow and the gastrocnemius weight at 3 weeks. Similar increase can also been seen in simvastatin 400 mg orally but not in simvastatin 20 mg orally. CONCLUSION These findings demonstrate that a single intraosseous administration of simvastatin mobilized endothelial progenitor cells at a dose one-hundredth of the required daily oral dose in rats, and this potent mobilization of endothelial progenitor cells markedly improved diabetic limb ischemia by means of neovascularization.
Collapse
Affiliation(s)
- Junxiong Zhu
- From the Departments of Orthopedics and Neurology, Peking University Third Hospital; and Beijing Key Laboratory of Spinal Diseases
| | - Jie Tan
- From the Departments of Orthopedics and Neurology, Peking University Third Hospital; and Beijing Key Laboratory of Spinal Diseases
| | - Chenggui Zhang
- From the Departments of Orthopedics and Neurology, Peking University Third Hospital; and Beijing Key Laboratory of Spinal Diseases
| | - Jialin Jia
- From the Departments of Orthopedics and Neurology, Peking University Third Hospital; and Beijing Key Laboratory of Spinal Diseases
| | - Huijie Leng
- From the Departments of Orthopedics and Neurology, Peking University Third Hospital; and Beijing Key Laboratory of Spinal Diseases
| | - Yingsheng Xu
- From the Departments of Orthopedics and Neurology, Peking University Third Hospital; and Beijing Key Laboratory of Spinal Diseases
| | - ChunLi Song
- From the Departments of Orthopedics and Neurology, Peking University Third Hospital; and Beijing Key Laboratory of Spinal Diseases
| |
Collapse
|
36
|
Ge L, Xun C, Li W, Jin S, Liu Z, Zhuo Y, Duan D, Hu Z, Chen P, Lu M. Extracellular vesicles derived from hypoxia-preconditioned olfactory mucosa mesenchymal stem cells enhance angiogenesis via miR-612. J Nanobiotechnology 2021; 19:380. [PMID: 34802444 PMCID: PMC8607643 DOI: 10.1186/s12951-021-01126-6] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/05/2021] [Indexed: 12/21/2022] Open
Abstract
Mesenchymal stem cells (MSCs) play important roles in tissue repair and regeneration, such as the induction of angiogenesis, particularly under hypoxic conditions. However, the molecular mechanisms underlying hypoxic MSC activation remain largely unknown. MSC-derived extracellular vesicles (EVs) are vital mediators of cell-to-cell communication and can be directly utilized as therapeutic agents for tissue repair and regeneration. Here, we explored the effects of EVs from human hypoxic olfactory mucosa MSCs (OM-MSCs) on angiogenesis and its underlying mechanism. EVs were isolated from normoxic (N) OM-MSCs (N-EVs) and hypoxic (H) OM-MSCs (H-EVs) using differential centrifugation and identified by transmission electron microscopy and flow cytometry. In vitro and in vivo, both types of OM-MSC-EVs promoted the proliferation, migration, and angiogenic activities of human brain microvascular endothelial cells (HBMECs). In addition, angiogenesis-stimulatory activity in the H-EV group was significantly enhanced compared to the N-EV group. MicroRNA profiling revealed a higher abundance of miR-612 in H-EVs than in N-EVs, while miR-612 inactivation abolished the N-EV treatment benefit. To explore the roles of miR-612, overexpression and knock-down experiments were performed using a mimic and inhibitor or agomir and antagomir of miR-612. The miR-612 target genes were confirmed using the luciferase reporter assay. Gain- and loss-of-function studies allowed the validation of miR-612 (enriched in hypoxic OM-MSC-EVs) as a functional messenger that stimulates angiogenesis and represses the expression of TP53 by targeting its 3′-untranslated region. Further functional assays showed that hypoxic OM-MSC-EVs promote paracrine Hypoxia-inducible factor 1-alpha (HIF-1α)-Vascular endothelial growth factor (VEGF) signaling in HBMECs via the exosomal miR-612-TP53-HIF-1α-VEGF axis. These findings suggest that hypoxic OM-MSC-EVs may represent a promising strategy for ischemic disease by promoting angiogenesis via miR-612 transfer. ![]()
Collapse
Affiliation(s)
- Lite Ge
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China.,The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, People's Republic of China.,Hunan Provincical Key Laboratory of Neurorestoratology, The Second Affiliated Hospital, Hunan Normal University, Changsha, 410003, People's Republic of China
| | - Chengfeng Xun
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, People's Republic of China.,Hunan Provincical Key Laboratory of Neurorestoratology, The Second Affiliated Hospital, Hunan Normal University, Changsha, 410003, People's Republic of China
| | - Wenshui Li
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, People's Republic of China.,Hunan Provincical Key Laboratory of Neurorestoratology, The Second Affiliated Hospital, Hunan Normal University, Changsha, 410003, People's Republic of China
| | - Shengyu Jin
- Hunan Provincical Key Laboratory of Neurorestoratology, The Second Affiliated Hospital, Hunan Normal University, Changsha, 410003, People's Republic of China
| | - Zuo Liu
- Hunan Provincical Key Laboratory of Neurorestoratology, The Second Affiliated Hospital, Hunan Normal University, Changsha, 410003, People's Republic of China
| | - Yi Zhuo
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, People's Republic of China.,Hunan Provincical Key Laboratory of Neurorestoratology, The Second Affiliated Hospital, Hunan Normal University, Changsha, 410003, People's Republic of China
| | - Da Duan
- Hunan Provincical Key Laboratory of Neurorestoratology, The Second Affiliated Hospital, Hunan Normal University, Changsha, 410003, People's Republic of China
| | - Zhiping Hu
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China.
| | - Ping Chen
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, People's Republic of China.
| | - Ming Lu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, People's Republic of China. .,Hunan Provincical Key Laboratory of Neurorestoratology, The Second Affiliated Hospital, Hunan Normal University, Changsha, 410003, People's Republic of China. .,Department of Neurosurgery, The Second Affiliated Hospital of Hunan Normal University, Changsha, 410003, People's Republic of China.
| |
Collapse
|
37
|
Grenier C, Caillon A, Munier M, Grimaud L, Champin T, Toutain B, Fassot C, Blanc-Brude O, Loufrani L. Dual Role of Thrombospondin-1 in Flow-Induced Remodeling. Int J Mol Sci 2021; 22:12086. [PMID: 34769516 PMCID: PMC8584526 DOI: 10.3390/ijms222112086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/13/2021] [Accepted: 10/29/2021] [Indexed: 11/21/2022] Open
Abstract
(1) Background: Chronic increases in blood flow, as in cardiovascular diseases, induce outward arterial remodeling. Thrombospondin-1 (TSP-1) is known to interact with matrix proteins and immune cell-surface receptors, but its contribution to flow-mediated remodeling in the microcirculation remains unknown. (2) Methods: Mesenteric arteries were ligated in vivo to generate high- (HF) and normal-flow (NF) arteries in wild-type (WT) and TSP-1-deleted mice (TSP-1-/-). After 7 days, arteries were isolated and studied ex vivo. (3) Results: Chronic increases in blood flow induced outward remodeling in WT mice (increasing diameter from 221 ± 10 to 280 ± 10 µm with 75 mmHg intraluminal pressure) without significant effect in TSP-1-/- (296 ± 18 to 303 ± 14 µm), neutropenic or adoptive bone marrow transfer mice. Four days after ligature, pro inflammatory gene expression levels (CD68, Cox2, Gp91phox, p47phox and p22phox) increased in WT HF arteries but not in TSP-1-/- mice. Perivascular neutrophil accumulation at day 4 was significantly lower in TSP-1-/- than in WT mice. (4) Conclusions: TSP-1 origin is important; indeed, circulating TSP-1 participates in vasodilation, whereas both circulating and tissue TSP-1 are involved in arterial wall thickness and diameter expansion.
Collapse
Affiliation(s)
- Céline Grenier
- UMR CNRS 6015, 49100 Angers, France; (C.G.); (A.C.); (M.M.); (L.G.); (T.C.); (B.T.); (C.F.)
- INSERM U1083, 49100 Angers, France
- MITOVASC Institute, University of Angers, 49100 Angers, France
| | - Antoine Caillon
- UMR CNRS 6015, 49100 Angers, France; (C.G.); (A.C.); (M.M.); (L.G.); (T.C.); (B.T.); (C.F.)
- INSERM U1083, 49100 Angers, France
- MITOVASC Institute, University of Angers, 49100 Angers, France
| | - Mathilde Munier
- UMR CNRS 6015, 49100 Angers, France; (C.G.); (A.C.); (M.M.); (L.G.); (T.C.); (B.T.); (C.F.)
- INSERM U1083, 49100 Angers, France
- MITOVASC Institute, University of Angers, 49100 Angers, France
| | - Linda Grimaud
- UMR CNRS 6015, 49100 Angers, France; (C.G.); (A.C.); (M.M.); (L.G.); (T.C.); (B.T.); (C.F.)
- INSERM U1083, 49100 Angers, France
- MITOVASC Institute, University of Angers, 49100 Angers, France
| | - Tristan Champin
- UMR CNRS 6015, 49100 Angers, France; (C.G.); (A.C.); (M.M.); (L.G.); (T.C.); (B.T.); (C.F.)
- INSERM U1083, 49100 Angers, France
- MITOVASC Institute, University of Angers, 49100 Angers, France
| | - Bertrand Toutain
- UMR CNRS 6015, 49100 Angers, France; (C.G.); (A.C.); (M.M.); (L.G.); (T.C.); (B.T.); (C.F.)
- INSERM U1083, 49100 Angers, France
- MITOVASC Institute, University of Angers, 49100 Angers, France
| | - Céline Fassot
- UMR CNRS 6015, 49100 Angers, France; (C.G.); (A.C.); (M.M.); (L.G.); (T.C.); (B.T.); (C.F.)
- INSERM U1083, 49100 Angers, France
- MITOVASC Institute, University of Angers, 49100 Angers, France
| | | | - Laurent Loufrani
- UMR CNRS 6015, 49100 Angers, France; (C.G.); (A.C.); (M.M.); (L.G.); (T.C.); (B.T.); (C.F.)
- INSERM U1083, 49100 Angers, France
- MITOVASC Institute, University of Angers, 49100 Angers, France
| |
Collapse
|
38
|
Bovine Milk Exosomes Alleviate Cardiac Fibrosis via Enhancing Angiogenesis In Vivo and In Vitro. J Cardiovasc Transl Res 2021; 15:560-570. [PMID: 34599486 DOI: 10.1007/s12265-021-10174-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/13/2021] [Indexed: 12/20/2022]
Abstract
Cardiac fibrosis is a difficult clinical puzzle without effective therapy. Exosomes play an important role in alleviating cardiac fibrosis via angiogenesis. This research aimed to assess the effect of bovine milk on cardiac fibrosis. The proangiogenic effect of bovine milk exosomes was analyzed both in isoproterenol (ISO)-induced cardiac fibrosis rats in vivo and in human umbilical vein endothelial cells (HUVECs) after oxygen and glucose deprivation (OGD) in vitro. Results indicated that bovine milk exosomes alleviated the extracellular matrix (ECM) deposition and enhanced the cardiac function in cardiac fibrosis rat. The proangiogenic growth factors were significantly enhanced in rats accepted bovine milk exosomes. Meanwhile, bovine milk exosomes ameliorated the motility, migration, and tube-forming ability of HUVECs after OGD in vitro. Bovine milk exosomes alleviate cardiac fibrosis and enhance cardiac function in cardiac fibrosis rats via enhancing angiogenesis. Bovine milk exosomes may represent a potential strategy for the treatment of cardiac fibrosis.
Collapse
|
39
|
Primikiris P, Hadjigeorgiou G, Tsamopoulou M, Biondi A, Iosif C. Review on the current treatment status of vein of Galen malformations and future directions in research and treatment. Expert Rev Med Devices 2021; 18:933-954. [PMID: 34424109 DOI: 10.1080/17434440.2021.1970527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Vein of Galen malformations (VOGMs) represent a rare pathologic entity with often catastrophic natural history. The advances in endovascular treatment in recent years have allowed for a paradigm shift in the treatment and outcome of these high-flow shunts, even though their pathogenetic mechanisms and evolution remain in part obscure. AREAS COVERED The overall management of VOGMs requires a tailored case-to-case approach, starting with in utero detection and reserving endovascular treatment for indicated cases. Lately, the advances in translational research with whole-genome sequencing and the coupling with cellular-level hemodynamics attempt to shed more light in the pathogenesis and evolution of these lesions. At the same time the advances in endovascular techniques allow for more safety and tailored technical strategy planning. Furthermore, the advances in MRI techniques allow a better understanding of their vascular anatomy. In view of these recent advances and by performing a PUBMED literature review of the last 15 years, we attempt a review of the evolutions in the imaging, management, endovascular treatment and understanding of underlying mechanisms for VOGMs. EXPERT OPINION The progress in the fields detailed in this review appears very promising in better understanding VOGMs and expanding the available therapeutic arsenal.
Collapse
Affiliation(s)
- Panagiotis Primikiris
- Department of Interventional Neuroradiology, Jean Minjoz University Hospital, Besancon, France
| | | | - Maria Tsamopoulou
- School of Medicine, National Kapodistrian University of Athens, Greece
| | - Alessandra Biondi
- Department of Interventional Neuroradiology, Jean Minjoz University Hospital, Besancon, France
| | - Christina Iosif
- School of Medicine, European University of Cyprus, Nicosia, Cyprus.,Department of Interventional Neuroradiology, Henry Dunant Hospital, Athens, Greece
| |
Collapse
|
40
|
Magenta A, Florio MC, Ruggeri M, Furgiuele S. Autologous cell therapy in diabetes‑associated critical limb ischemia: From basic studies to clinical outcomes (Review). Int J Mol Med 2021; 48:173. [PMID: 34278463 DOI: 10.3892/ijmm.2021.5006] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/30/2020] [Indexed: 01/13/2023] Open
Abstract
Cell therapy is becoming an attractive alternative for the treatment of patients with no‑option critical limb ischemia (CLI). The main benefits of cell therapy are the induction of therapeutic angiogenesis and neovascularization that lead to an increase in blood flow in the ischemic limb and tissue regeneration in non‑healing cutaneous trophic lesions. In the present review, the current state of the art of strategies in the cell therapy field are summarized, focusing on intra‑operative autologous cell concentrates in diabetic patients with CLI, examining different sources of cell concentrates and their mechanisms of action. The present study underlined the detrimental effects of the diabetic condition on different sources of autologous cells used in cell therapy, and also in delaying wound healing capacity. Moreover, relevant clinical trials and critical issues arising from cell therapy trials are discussed. Finally, the new concept of cell therapy as an adjuvant therapy to increase wound healing in revascularized diabetic patients is introduced.
Collapse
Affiliation(s)
| | - Maria Cristina Florio
- Laboratory of Cardiovascular Science, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, MD 21224, USA
| | - Massimo Ruggeri
- Department of Vascular Surgery, San Camillo de Lellis Hospital, I‑02100 Rieti, Italy
| | | |
Collapse
|
41
|
Raposo L, Lourenço AP, Nascimento DS, Cerqueira R, Cardim N, Leite-Moreira A. Human umbilical cord tissue-derived mesenchymal stromal cells as adjuvant therapy for myocardial infarction: a review of current evidence focusing on pre-clinical large animal models and early human trials. Cytotherapy 2021; 23:974-979. [PMID: 34112613 DOI: 10.1016/j.jcyt.2021.05.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/25/2021] [Accepted: 05/06/2021] [Indexed: 12/28/2022]
Abstract
Although biologically appealing, the concept of tissue regeneration underlying first- and second-generation cell therapies has failed to translate into consistent results in clinical trials. Several types of cells from different origins have been tested in pre-clinical models and in patients with acute myocardial infarction (AMI). Mesenchymal stromal cells (MSCs) have gained attention because of their potential for immune modulation and ability to promote endogenous tissue repair, mainly through their secretome. MSCs can be easily obtained from several human tissues, the umbilical cord being the most abundant source, and further expanded in culture, making them attractive as an allogeneic "of-the-shelf" cell product, suitable for the AMI setting. The available evidence concerning umbilical cord-derived MSCs in AMI is reviewed, focusing on large animal pre-clinical studies and early human trials. Molecular and cellular mechanisms as well as current limitations and possible translational solutions are also discussed.
Collapse
Affiliation(s)
- Luís Raposo
- Cardiology Department, Santa Cruz Hospital, West Lisbon Hospital Center, Lisbon, Portugal; Hospital da Luz Lisboa, Luz Saúde, Lisbon, Portugal; Nova Medical School, Lisbon, Portugal.
| | - André P Lourenço
- Department of Cardiac Surgery, University Hospital Centre São João, Porto, Portugal; Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Diana S Nascimento
- Institute for Research and Innovation in Health, University of Porto, Portugal; Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Portugal; Instituto Nacional de Engenharia Biomédica, University of Porto, Portugal
| | - Rui Cerqueira
- Department of Cardiac Surgery, University Hospital Centre São João, Porto, Portugal; Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Nuno Cardim
- Hospital da Luz Lisboa, Luz Saúde, Lisbon, Portugal; Nova Medical School, Lisbon, Portugal
| | - Adelino Leite-Moreira
- Department of Cardiac Surgery, University Hospital Centre São João, Porto, Portugal; Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
42
|
Valproic Acid Decreases Endothelial Colony Forming Cells Differentiation and Induces Endothelial-to-Mesenchymal Transition-like Process. Stem Cell Rev Rep 2021; 16:357-368. [PMID: 31898801 DOI: 10.1007/s12015-019-09950-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Valproic acid (VPA), a histone deacetylase (HDAC) inhibitor is a widely used anticonvulsant drug. VPA is also under clinical evaluation to be employed in anticancer therapy, as an antithrombotic agent or a molecule to be used in the stem cells expansion protocols. Since endothelial colony forming cells (ECFC) has been identified as the human postnatal vasculogenic cells involved in thrombotic disorders and serve as a promising source of immature cell for vascular repair, objectives of the present study were to determine how VPA contributes to ECFC commitment and their angiogenic properties. We examined the effect of VPA on ECFC obtained from cord blood by evaluating colony number, proliferation, migration and their sprouting ability in vitro, as well as their in vivo vasculogenic properties. VPA inhibited endothelial differentiation potential from of cord blood derived stem cells associated with decreased proliferation and sprouting activity of cultured ECFC. VPA treatment significantly decreased the vessel-forming ability of ECFC transplanted together with mesenchymal stem cells (MSC) in Matrigel implants in nude mice model. Surprisingly, a microscopic evaluation revealed that VPA induces marked morphological changes from a cobblestone-like EC morphology to enlarged spindle shaped morphology of ECFC. RT-qPCR and a CD31/CD90 flow cytometry analysis confirmed a phenotypic switch of VPA-treated ECFC to mesenchymal-like phenotype. In conclusion, the pan-HDAC inhibitor VPA described for expansion of hematopoietic stem cells and very small embryonic like stem cells cannot be successfully employed for differentiation of endothelial lineage committed ECFC into functional endothelial cells. Our data also suggest that VPA based therapeutics may induce endothelial dysfunction associated with fibrosis that might induce thrombosis recurrence or venous insufficiency.
Collapse
|
43
|
Cheng Z, Naga Srikanth Garikipati V, Truongcao MM, Cimini M, Huang G, Wang C, Benedict C, Gonzalez C, Mallaredy V, Goukassian DA, Verma SK, Kishore R. Serum-Derived Small Extracellular Vesicles From Diabetic Mice Impair Angiogenic Property of Microvascular Endothelial Cells: Role of EZH2. J Am Heart Assoc 2021; 10:e019755. [PMID: 33988033 PMCID: PMC8200714 DOI: 10.1161/jaha.120.019755] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Impaired angiogenic abilities of the microvascular endothelial cell (MVEC) play a crucial role in diabetes mellitus–impaired ischemic tissue repair. However, the underlying mechanisms of diabetes mellitus–impaired MVEC function remain unclear. We studied the role of serum‐derived small extracellular vesicles (ssEVs) in diabetes mellitus–impaired MVEC function. Methods and Results ssEVs were isolated from 8‐week‐old male db/db and db/+ mice by ultracentrifugation and size/number were determined by the Nano‐sight tracking system. Diabetic ssEVs significantly impaired tube formation and migration abilities of human MVECs. Furthermore, local transplantation of diabetic ssEVs strikingly reduced blood perfusion and capillary/arteriole density in ischemic hind limb of wildtype C57BL/6J mice. Diabetic ssEVs decreased secretion/expression of several pro‐angiogenic factors in human MVECs. Mechanistically, expression of enhancer of zest homolog 2 (EZH2), the major methyltransferase responsible for catalyzing H3K27me3 (a transcription repressive maker), and H3K27me3 was increased in MVECs from db/db mice. Diabetic ssEVs increased EZH2 and H3K27me3 expression/activity in human MVECs. Expression of EZH2 mRNA was increased in diabetic ssEVs. EZH2‐specific inhibitor significantly reversed diabetic ssEVs‐enhanced expression of EZH2 and H3K27me3, impaired expression of angiogenic factors, and improved blood perfusion and vessel density in ischemic hind limb of C57BL/6J mice. Finally, EZH2 inactivation repressed diabetic ssEVs‐induced H3K27me3 expression at promoter of pro‐angiogenic genes. Conclusions Diabetic ssEVs impair the angiogenic property of MVECs via, at least partially, transferring EZH2 mRNA to MVECs, thus inducing the epigenetic mechanism involving EZH2‐enhanced expression of H3K27me3 and consequent silencing of pro‐angiogenic genes. Our findings unravel the cellular mechanism and expand the scope of bloodborne substances that impair MVEC function in diabetes mellitus.
Collapse
Affiliation(s)
- Zhongjian Cheng
- Center for Translational Medicine Lewis Katz School of Medicine Temple University Philadelphia PA
| | - Venkata Naga Srikanth Garikipati
- Department of Emergency Medicine Dorothy M. Davis Heart Lung and Research InstituteThe Ohio State University Wexner Medical Center Columbus OH
| | - May M Truongcao
- Center for Translational Medicine Lewis Katz School of Medicine Temple University Philadelphia PA
| | - Maria Cimini
- Center for Translational Medicine Lewis Katz School of Medicine Temple University Philadelphia PA
| | - Grace Huang
- Center for Translational Medicine Lewis Katz School of Medicine Temple University Philadelphia PA
| | - Chunlin Wang
- Center for Translational Medicine Lewis Katz School of Medicine Temple University Philadelphia PA
| | - Cindy Benedict
- Center for Translational Medicine Lewis Katz School of Medicine Temple University Philadelphia PA
| | - Carolina Gonzalez
- Center for Translational Medicine Lewis Katz School of Medicine Temple University Philadelphia PA
| | - Vandana Mallaredy
- Center for Translational Medicine Lewis Katz School of Medicine Temple University Philadelphia PA
| | - David A Goukassian
- Cardiovascular Research CenterIcahn School of Medicine at Mount Sinai New York NY
| | - Suresh K Verma
- Department of Medicine-Cardiovascular Disease The University of Alabama at Birmingham Birmingham AL
| | - Raj Kishore
- Center for Translational Medicine Lewis Katz School of Medicine Temple University Philadelphia PA.,Department of Pharmacology Lewis Katz School of Medicine Temple University Philadelphia PA
| |
Collapse
|
44
|
Alfì E, Thairi C, Femminò S, Alloatti G, Moccia F, Brizzi MF, Pagliaro P, Penna C. Extracellular vesicles (EVs) in ischemic conditioning and angiogenesis: Focus on endothelial derived EVs. Vascul Pharmacol 2021; 140:106873. [PMID: 33992781 DOI: 10.1016/j.vph.2021.106873] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 01/08/2023]
Abstract
During myocardial ischemia, timely reperfusion is critical to limit infarct area and the overall loss of cardiac contractile function. However, reperfusion further exacerbates the damage of the ischemic heart. This type of injury is known as ischemia-reperfusion injury (IRI). Ischemic conditioning is a procedure which consists of brief cycles of ischemia and reperfusion in order to protect the myocardium against IRI. Remote ischemic conditioning (RIC), namely transient brief episodes of ischemia at a remote site before a subsequent damaging ischemia/reperfusion procedure of the target organ (e.g., the heart), protects against IRI. However, how the stimulus of RIC is transduced from the remote organ to the ischemic heart is still unknown. Recently, extracellular vesicles (EVs) have been proposed to have a role in the RIC procedure. The endothelium releases EVs and is also one of the tissues mostly exposed to EVs during their journey to the target organ. Moreover, EVs may have important roles in angiogenesis and, therefore, in the remodeling of post-ischemic organs. Here we analyze how EVs may contribute to the overall cardioprotective effect and the implication of the endothelium and its EVs in RIC mediated acute cardioprotection as well as in angiogenesis.
Collapse
Affiliation(s)
- Edoardo Alfì
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, TO, Italy
| | - Cecilia Thairi
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, TO, Italy
| | - Saveria Femminò
- Department of Medical Sciences, University of Turin, Corso Dogliotti 14, 10126 Turin, Italy
| | - Giuseppe Alloatti
- Uni-Astiss, Polo Universitario Rita Levi Montalcini, 14100 Asti, Italy
| | - Francesco Moccia
- Department of Biology and Biotechnology, University of Pavia, Laboratory of General Physiology, 27100 Pavia, Italy
| | - Maria F Brizzi
- Department of Medical Sciences, University of Turin, Corso Dogliotti 14, 10126 Turin, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, TO, Italy.
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, TO, Italy
| |
Collapse
|
45
|
Ma Y, Jia L, Wang Y, Ji Y, Chen J, Ma H, Lin X, Zhang Y, Li W, Ni H, Xie L, Xie Y, Xiang M. Heme Oxygenase-1 in Macrophages Impairs the Perfusion Recovery After Hindlimb Ischemia by Suppressing Autolysosome-Dependent Degradation of NLRP3. Arterioscler Thromb Vasc Biol 2021; 41:1710-1723. [PMID: 33761761 DOI: 10.1161/atvbaha.121.315805] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Databases, Genetic
- Disease Models, Animal
- Heme Oxygenase-1/genetics
- Heme Oxygenase-1/metabolism
- Hindlimb
- Humans
- Inflammasomes/genetics
- Inflammasomes/metabolism
- Inflammation Mediators/metabolism
- Ischemia/enzymology
- Ischemia/genetics
- Ischemia/physiopathology
- Lysosomes/enzymology
- Macrophages/enzymology
- Male
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Skeletal/blood supply
- Muscle, Skeletal/enzymology
- Muscle, Skeletal/physiopathology
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- Neovascularization, Physiologic
- Proteolysis
- Recovery of Function
- Regional Blood Flow
- Mice
Collapse
Affiliation(s)
- Yuankun Ma
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liangliang Jia
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yidong Wang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongli Ji
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Chen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Ma
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoping Lin
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuhao Zhang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wudi Li
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Ni
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lan Xie
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Xie
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
46
|
Augmentation of Perforator Flap Blood Supply with Vascular Supercharge or Flap Prefabrication: Evaluation in a Rat Model. Plast Reconstr Surg 2021; 147:1105-1115. [PMID: 33890892 DOI: 10.1097/prs.0000000000007893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Vascular supercharge and flap prefabrication are two surgical maneuvers to improve flap blood supply. Although these techniques have been studied intensively, few studies have focused on the differences between supercharge and prefabricated flaps regarding their flap survival areas, vasculatures, and hemodynamics. METHODS In this study, 21 male Sprague-Dawley rats were divided into three groups as follows: group A, single perforator flap; group B, supercharge flap; and group C, prefabricated flap. Flap survival was measured 1 week after flap elevation. Indocyanine green angiography was applied to visualize flap vascularity and to analyze flap hemodynamics. Von Willebrand factor immunohistochemical staining was applied to assess the number of microvessels in the choke zone of the abdominal wall. RESULTS The flap survival areas were expanded significantly in the arteriovenous supercharge group and the vascular bundle prefabricated group compared with that in the single-perforator group (81.34 ± 8.12 percent and 75.51 ± 8.08 percent versus 46.27 ± 10.01 percent, respectively; p < 0.05). Hemodynamic analysis suggested that although a significant increase in arterial infusion could be achieved with flap prefabrication, the venous effusion of the prefabricated flap was the worst among the three groups, indicating greater susceptibility to compromised venous return. Active neovascularization was confirmed by an increased number of microvessels in group C. Specifically, the dilatation of choke vessels and the newly formed vessels of the prefabricated pedicle could be appreciated by indocyanine green angiographic mapping. CONCLUSIONS Both vascular supercharge and flap prefabrication can augment the blood supply of the perforator flap but by means of different mechanisms. Because a supercharge flap is less susceptible to venous compromise, it is suggested to first consider the use of vascular supercharging when feasible.
Collapse
|
47
|
Marsico G, Martin‐Saldaña S, Pandit A. Therapeutic Biomaterial Approaches to Alleviate Chronic Limb Threatening Ischemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003119. [PMID: 33854887 PMCID: PMC8025020 DOI: 10.1002/advs.202003119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/24/2020] [Indexed: 05/14/2023]
Abstract
Chronic limb threatening ischemia (CLTI) is a severe condition defined by the blockage of arteries in the lower extremities that leads to the degeneration of blood vessels and is characterized by the formation of non-healing ulcers and necrosis. The gold standard therapies such as bypass and endovascular surgery aim at the removal of the blockage. These therapies are not suitable for the so-called "no option patients" which present multiple artery occlusions with a likelihood of significant limb amputation. Therefore, CLTI represents a significant clinical challenge, and the efforts of developing new treatments have been focused on stimulating angiogenesis in the ischemic muscle. The delivery of pro-angiogenic nucleic acid, protein, and stem cell-based interventions have limited efficacy due to their short survival. Engineered biomaterials have emerged as a promising method to improve the effectiveness of these latter strategies. Several synthetic and natural biomaterials are tested in different formulations aiming to incorporate nucleic acid, proteins, stem cells, macrophages, or endothelial cells in supportive matrices. In this review, an overview of the biomaterials used alone and in combination with growth factors, nucleic acid, and cells in preclinical models is provided and their potential to induce revascularization and regeneration for CLTI applications is discussed.
Collapse
Affiliation(s)
- Grazia Marsico
- CÚRAM SFI Research Centre for Medical DevicesNational University of IrelandGalwayIreland
| | - Sergio Martin‐Saldaña
- CÚRAM SFI Research Centre for Medical DevicesNational University of IrelandGalwayIreland
| | - Abhay Pandit
- CÚRAM SFI Research Centre for Medical DevicesNational University of IrelandGalwayIreland
| |
Collapse
|
48
|
Detriche G, Guerin CL, Gendron N, Mirault T, Smadja DM. Do Endothelial Colony-forming Cells Come From Bone Marrow or Vessels/VSELs? Stem Cell Rev Rep 2021; 17:1500-1502. [PMID: 33651335 DOI: 10.1007/s12015-021-10140-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2021] [Indexed: 10/22/2022]
Affiliation(s)
- Grégoire Detriche
- Innovative Therapies in Hemostasis, Université de Paris, INSERM, F-75006, Paris, France.,Vascular Medicine Department, Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP- CUP), F-75015, Paris, France
| | - Coralie L Guerin
- Innovative Therapies in Hemostasis, Université de Paris, INSERM, F-75006, Paris, France.,Institut Curie, 75006, Paris, France
| | - Nicolas Gendron
- Innovative Therapies in Hemostasis, Université de Paris, INSERM, F-75006, Paris, France.,Hematology Department and Biosurgical Research Lab (Carpentier Foundation), Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), F-75015, Paris, France
| | - Tristan Mirault
- Vascular Medicine Department, Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP- CUP), F-75015, Paris, France.,Université de Paris, PARCC, INSERM, F-75015, Paris, France
| | - David M Smadja
- Innovative Therapies in Hemostasis, Université de Paris, INSERM, F-75006, Paris, France. .,Hematology Department and Biosurgical Research Lab (Carpentier Foundation), Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), F-75015, Paris, France.
| |
Collapse
|
49
|
Li C, Kitzerow O, Nie F, Dai J, Liu X, Carlson MA, Casale GP, Pipinos II, Li X. Bioengineering strategies for the treatment of peripheral arterial disease. Bioact Mater 2021; 6:684-696. [PMID: 33005831 PMCID: PMC7511653 DOI: 10.1016/j.bioactmat.2020.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/12/2020] [Accepted: 09/12/2020] [Indexed: 12/21/2022] Open
Abstract
Peripheral arterial disease (PAD) is a progressive atherosclerotic disorder characterized by narrowing and occlusion of arteries supplying the lower extremities. Approximately 200 million people worldwide are affected by PAD. The current standard of operative care is open or endovascular revascularization in which blood flow restoration is the goal. However, many patients are not appropriate candidates for these treatments and are subject to continuous ischemia of their lower limbs. Current research in the therapy of PAD involves developing modalities that induce angiogenesis, but the results of simple cell transplantation or growth factor delivery have been found to be relatively poor mainly due to difficulties in stem cell retention and survival and rapid diffusion and enzymolysis of growth factors following injection of these agents in the affected tissues. Biomaterials, including hydrogels, have the capability to protect stem cells during injection and to support cell survival. Hydrogels can also provide a sustained release of growth factors at the injection site. This review will focus on biomaterial systems currently being investigated as carriers for cell and growth factor delivery, and will also discuss biomaterials as a potential stand-alone method for the treatment of PAD. Finally, the challenges of development and use of biomaterials systems for PAD treatment will be reviewed.
Collapse
Affiliation(s)
- Cui Li
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Oliver Kitzerow
- Department of Genetics Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Fujiao Nie
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Jingxuan Dai
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Xiaoyan Liu
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Mark A. Carlson
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, United States
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, 68198, United States
- Omaha VA Medical Center, Omaha, NE, 68105, United States
| | - George P. Casale
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Iraklis I. Pipinos
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Xiaowei Li
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| |
Collapse
|
50
|
Wang DP, Lin Q, Kang K, Wu YF, Su SH, Hai J. Preservation of spatial memory and neuroprotection by the fatty acid amide hydrolase inhibitor URB597 in a rat model of vascular dementia. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:228. [PMID: 33708855 PMCID: PMC7940933 DOI: 10.21037/atm-20-4431] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Chronic cerebral hypoperfusion (CCH) is a major risk factor for vascular dementia (VaD). There are currently no broadly effective prevention or treatment strategies for VaD, but recent studies have reported promising results following vascular bypass surgery and pharmacomodulation of the brain endocannabinoid system (ECS). In this study, early effects of encephalomyosynangiosis (EMS) bypass surgery and augmented endocannabinoid signaling on CCH-induced cognitive dysfunction and neuronal damage were investigated. Methods An animal model of VaD was established by bilateral common carotid artery occlusion (BCCAO). Cannabinoid signaling was upregulated by treatment with the fatty acid amide hydrolase inhibitor URB597 (URB). Spatial learning and memory, cerebral blood flow (CBF), revascularization, brain-derived neurotrophic factor (BDNF)-tropomyosin receptor kinase B (TrkB) signaling, and apoptosis were compared among Sham, BCCAO, BCCAO + EMS, BCCAO + URB, and BCCAO + URB + EMS groups. Spatial learning and memory were evaluated using the Morris water maze (MWM). The CBF in cortex and hippocampus was evaluated by 3-dimensional arterial spin labeling. The neovascularization was visualized by CD34 immunofluorescence staining, and BDNF-TrkB signaling protein expression levels were assessed by Western blotting. Results Treatment with URB597 but not EMS alone reversed the spatial learning and memory deficits induced by BCCAO. Neovascularization was enhanced after EMS surgery but not by URB597. Alternatively, there were no significant differences in CBF among treatment groups. Expression levels of BDNF and TrkB were significantly reduced by CCH compared to Sham treatment, and downregulation of both proteins was reversed by URB597 treatment but not EMS. BCCAO enhanced neuronal apoptosis, which was also reversed by URB597. Conclusions Augmentation of endogenous cannabinoid signaling but not EMS protects against CCH-induced neurodegeneration and preserves spatial learning and memory, possibly by activating BDNF-TrkB signaling.
Collapse
Affiliation(s)
- Da-Peng Wang
- Department of Neurosurgery, Tong Ji Hospital, Tong Ji University School of Medicine, Shanghai, China.,Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
| | - Qi Lin
- Department of Pharmacy, Institutes of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kai Kang
- Department of Research and Surveillance Evaluation, Shanghai Center for Health Promotion, Shanghai, China
| | - Yi-Fang Wu
- Department of Neurosurgery, Tong Ji Hospital, Tong Ji University School of Medicine, Shanghai, China
| | - Shao-Hua Su
- Department of Neurosurgery, Tong Ji Hospital, Tong Ji University School of Medicine, Shanghai, China
| | - Jian Hai
- Department of Neurosurgery, Tong Ji Hospital, Tong Ji University School of Medicine, Shanghai, China
| |
Collapse
|