1
|
Negri S, Reyff Z, Troyano-Rodriguez E, Milan M, Ihuoma J, Tavakol S, Shi H, Patai R, Jiang R, Mohon J, Boma-Iyaye J, Ungvari Z, Csiszar A, Yabluchanskiy A, Moccia F, Tarantini S. Endothelial Colony-Forming Cells (ECFCs) in cerebrovascular aging: Focus on the pathogenesis of Vascular Cognitive Impairment and Dementia (VCID), and treatment prospects. Ageing Res Rev 2025; 104:102672. [PMID: 39884362 DOI: 10.1016/j.arr.2025.102672] [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: 11/01/2024] [Revised: 01/21/2025] [Accepted: 01/24/2025] [Indexed: 02/01/2025]
Abstract
Endothelial colony-forming cells (ECFCs), a unique endothelial progenitor subset, are essential for vascular integrity and repair, providing significant regenerative potential. Recent studies highlight their role in cerebrovascular aging, particularly in the pathogenesis of vascular cognitive impairment and dementia (VCID). Aging disrupts ECFC functionality through mechanisms such as oxidative stress, chronic inflammation, and cellular senescence, leading to compromised vascular repair and reduced neurovascular resilience. ECFCs influence key cerebrovascular processes, including neurovascular coupling (NVC), blood-brain barrier (BBB) integrity, and vascular regeneration, which are critical for cognitive health. Age-related decline in ECFC quantity and functionality contributes to vascular rarefaction, diminished cerebral blood flow (CBF), and BBB permeability-processes that collectively exacerbate cognitive decline. This review delves into the multifaceted role of ECFCs in cerebrovascular aging and underscores their potential as therapeutic targets in addressing age-related vascular dysfunctions, presenting new directions for mitigating the effects of aging on brain health.
Collapse
Affiliation(s)
- Sharon Negri
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zeke Reyff
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Eva Troyano-Rodriguez
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Madison Milan
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jennifer Ihuoma
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Sherwin Tavakol
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Helen Shi
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Roland Patai
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Raymond Jiang
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Casady School, Oklahoma City, OK, USA
| | - Jonah Mohon
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Oklahoma School of Science and Mathematics, OK, USA
| | - Jed Boma-Iyaye
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Oklahoma School of Science and Mathematics, OK, USA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral College, Health Sciences Program/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary; Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Francesco Moccia
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso 86100, Italy
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Dept. of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral College, Health Sciences Program/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary; Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
2
|
Madonna R, Barachini S, Ghelardoni S, Lu L, Shen WF, De Caterina R. Vasostatins: new molecular targets for atherosclerosis, post-ischaemic angiogenesis, and arteriogenesis. Cardiovasc Res 2024; 120:132-139. [PMID: 38242632 DOI: 10.1093/cvr/cvae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/25/2023] [Accepted: 11/30/2023] [Indexed: 01/21/2024] Open
Abstract
The chromogranin-secretogranin secretory proteins-granins-are acidic proteins localized in granules of endocrine cells and neurons. The chromogranin family includes chromogranins A (CgA) and B, as well as secretogranin II (once called chromogranin C). Members of this family undergo catalytic proteolysis to produce active peptides. The CgA-derived peptides vasostatin-1 and vasostatin-2, in particular, appear to protect against atherosclerosis, suppressing the expression of vascular cell adhesion molecule-1 and intercellular adhesion molecule-1, as well as exerting vasodilatory effects by enhancing nitric oxide bioavailability. Vasostatin-1 also suppresses vasoconstriction and abnormal angiogenesis. Vasostatin-1 and vasostatin-2 may be novel therapeutic targets for atherosclerosis and coronary heart disease, also protecting the myocardium against ischaemic damage.
Collapse
Affiliation(s)
- Rosalinda Madonna
- Department of Surgical, Medical, and Molecular Pathology and Critical Care, University of Pisa and Pisa University Hospital, Via Paradisa 2, 56124 Pisa, Italy
| | - Serena Barachini
- Department of Clinical and Experimental Medicine, Laboratory for Cell Therapy, University of Pisa, Pisa, Italy
| | - Sandra Ghelardoni
- Department of Pathology, Laboratory of Biochemistry, University of Pisa, Pisa, Italy
| | - Lin Lu
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiaotong University School of Medicine, 197 Rui Jin Road II, Shanghai 200025, People's Republic of China
| | - Wei-Feng Shen
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiaotong University School of Medicine, 197 Rui Jin Road II, Shanghai 200025, People's Republic of China
| | - Raffaele De Caterina
- Department of Surgical, Medical, and Molecular Pathology and Critical Care, University of Pisa and Pisa University Hospital, Via Paradisa 2, 56124 Pisa, Italy
| |
Collapse
|
3
|
Kawaguchi Y, Kuwana M. Pathogenesis of vasculopathy in systemic sclerosis and its contribution to fibrosis. Curr Opin Rheumatol 2023; 35:309-316. [PMID: 37490353 DOI: 10.1097/bor.0000000000000959] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
PURPOSE OF REVIEW In patients with systemic sclerosis (SSc), vascular manifestations precede skin and organ fibrosis. There is increasing evidence demonstrating a pathogenic link between early vascular injury and subsequent development of tissue fibrosis. RECENT FINDINGS Our knowledge of cellular and molecular mechanisms underlying a unique relationship between SSc-related vasculopathy and fibrosis has changed over the last few years. There is increasing evidence showing viral infection as a potential trigger elucidating vascular injury. Due to defective vascular repair machinery, this initial event results in endothelial cell activation and apoptosis as well as the recruitment of inflammatory/immune cells, leading to endothelial-to-mesenchymal transition. This sequential process induces destructive vasculopathy in capillaries, fibroproliferative vascular lesions in arteries, and excessive fibrosis in the surrounding tissue. A variety of molecular mechanisms and pathways involved in vascular remodeling linked to subsequent excessive fibrosis have been identified and serve as attractive therapeutic targets for SSc. SUMMARY Endothelial injury may play a central role in connecting three features that characterize SSc pathogenesis: vasculopathy, chronic inflammation, and fibrosis. Our understanding of the processes responsible for myofibroblast differentiation triggered by vascular injury will provide the rationale for novel targeted therapies for SSc.
Collapse
Affiliation(s)
- Yasushi Kawaguchi
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine
| | - Masataka Kuwana
- Department of Allergy and Rheumatology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
4
|
Kim KM, An HJ, Kim SH, Kim J, Sim C, Lee J, Park SH, Lee HI, Jang I, Lee S. Therapeutic Effect of Pericytes for Diabetic Wound Healing. Front Cardiovasc Med 2022; 9:868600. [PMID: 35647064 PMCID: PMC9135971 DOI: 10.3389/fcvm.2022.868600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/12/2022] [Indexed: 12/28/2022] Open
Abstract
Objective Numerous attempts have been made to devise treatments for ischemic foot ulcer (IFU), which is one of the most severe and fatal consequences of diabetes mellitus (DM). Pericytes, which are perivascular multipotent cells, are of interest as a treatment option for IFU because they play a critical role in forming and repairing various tissues. In this study, we want to clarify the angiogenic potential of pericytes in DM-induced wounds. Methods We evaluated pericyte stimulation capability for tube formation, angiogenesis, and wound healing (cell migration) in human umbilical vein endothelial cells (HUVECs) with in-vivo and in-vitro models of high glucose conditions. Results When HUVECs were co-cultured with pericytes, their tube-forming capacity and cell migration were enhanced. Our diabetic mouse model showed that pericytes promote wound healing via increased vascularization. Conclusion The findings of this study indicate that pericytes may enhance wound healing in high glucose conditions, consequently making pericyte transplantation suitable for treating IFUs.
Collapse
Affiliation(s)
- Kyeong Mi Kim
- Department of Laboratory Medicine, CHA Ilsan Medical Center, CHA University School of Medicine, Goyang-si, South Korea
| | - Hyun-Ju An
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, South Korea
| | - Sang-Hoon Kim
- Department of Cardiology, CHA Bundang Medical Center, CHA University School of Medicine, Gyeonggi-do, South Korea
| | - JuHee Kim
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, South Korea
| | - Changgon Sim
- CHA Graduate School of Medicine, Gyeonggi-do, South Korea
| | - Jaemin Lee
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, South Korea
| | - Sin Hyung Park
- Department of Orthopaedic Surgery, Bucheon Hospital, Soonchunhyang University School of Medicine, Gyeonggi-do, South Korea
| | - Hyun Il Lee
- Department of Orthopedic Surgery, Ilsan Paik Hospital, Inje University, Gyeonggi-do, South Korea
| | - Inseok Jang
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, South Korea
| | - Soonchul Lee
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, South Korea
- *Correspondence: Soonchul Lee
| |
Collapse
|
5
|
Advances of research of Fc-fusion protein that activate NK cells for tumor immunotherapy. Int Immunopharmacol 2022; 109:108783. [PMID: 35561479 DOI: 10.1016/j.intimp.2022.108783] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/02/2022] [Accepted: 04/14/2022] [Indexed: 12/21/2022]
Abstract
The rapid development of bioengineering technology has introduced Fc-fusion proteins, representing a novel kind of recombinant protein, as promising biopharmaceutical products in tumor therapy. Numerous related anti-tumor Fc-fusion proteins have been investigated and are in different stages of development. Fc-fusion proteins are constructed by fusing the Fc-region of the antibody with functional proteins or peptides. They retain the bioactivity of the latter and partial properties of the former. This structural and functional advantage makes Fc-fusion proteins an effective tool in tumor immunotherapy, especially for the recruitment and activation of natural killer (NK) cells, which play a critical role in tumor immunotherapy. Even though tumor cells have developed mechanisms to circumvent the cytotoxic effect of NK cells or induce defective NK cells, Fc-fusion proteins have been proven to effectively activate NK cells to kill tumor cells in different ways, such as antibody-dependent cell-mediated cytotoxicity (ADCC), activate NK cells in different ways in order to promote killing of tumor cells. In this review, we focus on NK cell-based immunity for cancers and current research progress of the Fc-fusion proteins for anti-tumor therapy by activating NK cells.
Collapse
|
6
|
Watterston C, Halabi R, McFarlane S, Childs SJ. Endothelial Semaphorin 3fb regulates Vegf pathway-mediated angiogenic sprouting. PLoS Genet 2021; 17:e1009769. [PMID: 34424892 PMCID: PMC8412281 DOI: 10.1371/journal.pgen.1009769] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 09/02/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022] Open
Abstract
Vessel growth integrates diverse extrinsic signals with intrinsic signaling cascades to coordinate cell migration and sprouting morphogenesis. The pro-angiogenic effects of Vascular Endothelial Growth Factor (VEGF) are carefully controlled during sprouting to generate an efficiently patterned vascular network. We identify crosstalk between VEGF signaling and that of the secreted ligand Semaphorin 3fb (Sema3fb), one of two zebrafish paralogs of mammalian Sema3F. The sema3fb gene is expressed by endothelial cells in actively sprouting vessels. Loss of sema3fb results in abnormally wide and stunted intersegmental vessel artery sprouts. Although the sprouts initiate at the correct developmental time, they have a reduced migration speed. These sprouts have persistent filopodia and abnormally spaced nuclei suggesting dysregulated control of actin assembly. sema3fb mutants show simultaneously higher expression of pro-angiogenic (VEGF receptor 2 (vegfr2) and delta-like 4 (dll4)) and anti-angiogenic (soluble VEGF receptor 1 (svegfr1)/ soluble Fms Related Receptor Tyrosine Kinase 1 (sflt1)) pathway components. We show increased phospho-ERK staining in migrating angioblasts, consistent with enhanced Vegf activity. Reducing Vegfr2 kinase activity in sema3fb mutants rescues angiogenic sprouting. Our data suggest that Sema3fb plays a critical role in promoting endothelial sprouting through modulating the VEGF signaling pathway, acting as an autocrine cue that modulates intrinsic growth factor signaling.
Collapse
Affiliation(s)
- Charlene Watterston
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Rami Halabi
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Sarah McFarlane
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Canada
| | - Sarah J. Childs
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
| |
Collapse
|
7
|
Store-operated Ca 2+ entry as a key oncogenic Ca 2+ signaling driving tumor invasion-metastasis cascade and its translational potential. Cancer Lett 2021; 516:64-72. [PMID: 34089807 DOI: 10.1016/j.canlet.2021.05.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/12/2021] [Accepted: 05/26/2021] [Indexed: 12/25/2022]
Abstract
Tumor metastasis is the primary cause of treatment failure and cancer-related deaths. Store-operated Ca2+ entry (SOCE), which is mediated by stromal interaction molecules (STIM) and ORAI proteins, has been implicated in the tumor invasion-metastasis cascade. Epithelial-mesenchymal transition (EMT) is a cellular program that enables tumor cells to acquire the capacities needed for migration and invasion and the formation of distal metastases. Tumor-associated angiogenesis contributes to metastasis because aberrantly developed vessels offer a path for tumor cell dissemination as well as supply sufficient nutrients for the metastatic colony to develop into metastasis. Recently, increasing evidence has indicated that SOCE alterations actively participate in the multi-step process of tumor metastasis. In addition, the dysregulated expression of STIM/ORAI has been reported to be a predictor of poor prognosis. Herein, we review the latest advances about the critical role of SOCE in the tumor metastasis cascade and the underlying regulatory mechanisms. We emphasize the contributions of SOCE to the EMT program, tumor cell migration and invasion, and angiogenesis. We further discuss the possibility of modulating SOCE or intervening in the downstream signaling pathways as a feasible targeting therapy for cancer treatment.
Collapse
|
8
|
Nguyen HC, Qadura M, Singh KK. Role of the Fatty Acid Binding Proteins in Cardiovascular Diseases: A Systematic Review. J Clin Med 2020; 9:E3390. [PMID: 33105856 PMCID: PMC7690604 DOI: 10.3390/jcm9113390] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/19/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases (CVD) remain a global pandemic and leading cause of deaths worldwide. While several guidelines have been developed to control the development of CVDs, its prevalence keeps on increasing until this day. Cardiovascular risk factors, such as reduced exercises and high fat or glucose diets, culminate in the development of the metabolic syndrome and eventually atherosclerosis, which is driven by high blood lipid and cholesterol levels, and by endothelial dysfunction. Late complications of atherosclerosis give rise to serious clinical cardiovascular manifestations such as myocardial infarction and hypertension. Therefore, endothelial functions and the lipid metabolism play critical roles in the pathogenesis of CVDs. Fatty acid-binding proteins are a family of intracellular proteins expressed in many cell types known mainly for their interaction with and trafficking of cellular lipids. The roles of a number of isoforms in this family have been implicated in lipid metabolic homeostasis, but their influence on endothelial function and vascular homeostasis remain largely unknown. This review's purpose is to update fundamentals about the connection between cardiovascular disease, metabolism, endothelial function, and mainly the roles of fatty acid-binding proteins.
Collapse
Affiliation(s)
- Hien C. Nguyen
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada;
| | - Mohammad Qadura
- Vascular Surgery, Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada;
| | - Krishna K. Singh
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada;
- Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
- Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A1, Canada
| |
Collapse
|
9
|
Negri S, Faris P, Rosti V, Antognazza MR, Lodola F, Moccia F. Endothelial TRPV1 as an Emerging Molecular Target to Promote Therapeutic Angiogenesis. Cells 2020; 9:cells9061341. [PMID: 32471282 PMCID: PMC7349285 DOI: 10.3390/cells9061341] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/26/2020] [Accepted: 05/26/2020] [Indexed: 02/06/2023] Open
Abstract
Therapeutic angiogenesis represents an emerging strategy to treat ischemic diseases by stimulating blood vessel growth to rescue local blood perfusion. Therefore, injured microvasculature may be repaired by stimulating resident endothelial cells or circulating endothelial colony forming cells (ECFCs) or by autologous cell-based therapy. Endothelial Ca2+ signals represent a crucial player in angiogenesis and vasculogenesis; indeed, several angiogenic stimuli induce neovessel formation through an increase in intracellular Ca2+ concentration. Several members of the Transient Receptor Potential (TRP) channel superfamily are expressed and mediate Ca2+-dependent functions in vascular endothelial cells and in ECFCs, the only known truly endothelial precursor. TRP Vanilloid 1 (TRPV1), a polymodal cation channel, is emerging as an important player in endothelial cell migration, proliferation, and tubulogenesis, through the integration of several chemical stimuli. Herein, we first summarize TRPV1 structure and gating mechanisms. Next, we illustrate the physiological roles of TRPV1 in vascular endothelium, focusing our attention on how endothelial TRPV1 promotes angiogenesis. In particular, we describe a recent strategy to stimulate TRPV1-mediated pro-angiogenic activity in ECFCs, in the presence of a photosensitive conjugated polymer. Taken together, these observations suggest that TRPV1 represents a useful target in the treatment of ischemic diseases.
Collapse
Affiliation(s)
- Sharon Negri
- Laboratory of General Physiology, Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (S.N.); (P.F.)
| | - Pawan Faris
- Laboratory of General Physiology, Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (S.N.); (P.F.)
| | - Vittorio Rosti
- Center for the Study of Myelofibrosis, Laboratory of Biochemistry, Biotechnology and Advanced Diagnosis, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy;
| | - Maria Rosa Antognazza
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia, via Pascoli 70/3, 20133 Milano, Italy; (M.R.A.); (F.L.)
| | - Francesco Lodola
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia, via Pascoli 70/3, 20133 Milano, Italy; (M.R.A.); (F.L.)
| | - Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (S.N.); (P.F.)
- Correspondence:
| |
Collapse
|
10
|
Zaric B, Obradovic M, Trpkovic A, Banach M, Mikhailidis DP, Isenovic ER. Endothelial Dysfunction in Dyslipidaemia: Molecular Mechanisms and Clinical Implications. Curr Med Chem 2020; 27:1021-1040. [DOI: 10.2174/0929867326666190903112146] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/23/2019] [Accepted: 08/23/2019] [Indexed: 12/13/2022]
Abstract
The endothelium consists of a monolayer of Endothelial Cells (ECs) which form
the inner cellular lining of veins, arteries, capillaries and lymphatic vessels. ECs interact with
the blood and lymph. The endothelium fulfils functions such as vasodilatation, regulation of
adhesion, infiltration of leukocytes, inhibition of platelet adhesion, vessel remodeling and
lipoprotein metabolism. ECs synthesize and release compounds such as Nitric Oxide (NO),
metabolites of arachidonic acid, Reactive Oxygen Species (ROS) and enzymes that degrade
the extracellular matrix. Endothelial dysfunction represents a phenotype prone to atherogenesis
and may be used as a marker of atherosclerotic risk. Such dysfunction includes impaired
synthesis and availability of NO and an imbalance in the relative contribution of endothelialderived
relaxing factors and contracting factors such as endothelin-1 and angiotensin. This
dysfunction appears before the earliest anatomic evidence of atherosclerosis and could be an
important initial step in further development of atherosclerosis. Endothelial dysfunction was
historically treated with vitamin C supplementation and L-arginine supplementation. Short
term improvement of the expression of adhesion molecule and endothelial function during
antioxidant therapy has been observed. Statins are used in the treatment of hyperlipidaemia, a
risk factor for cardiovascular disease. Future studies should focus on identifying the mechanisms
involved in the beneficial effects of statins on the endothelium. This may help develop
drugs specifically aimed at endothelial dysfunction.
Collapse
Affiliation(s)
- Bozidarka Zaric
- Laboratory of Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia
| | - Milan Obradovic
- Laboratory of Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia
| | - Andreja Trpkovic
- Laboratory of Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia
| | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Lodz, Poland
| | - Dimitri P. Mikhailidis
- Department of Clinical Biochemistry, Royal Free Campus, University College London Medical School, University College London (UCL), London, United Kingdom
| | - Esma R. Isenovic
- Laboratory of Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia
| |
Collapse
|
11
|
Obradovic M, Essack M, Zafirovic S, Sudar‐Milovanovic E, Bajic VP, Van Neste C, Trpkovic A, Stanimirovic J, Bajic VB, Isenovic ER. Redox control of vascular biology. Biofactors 2020; 46:246-262. [PMID: 31483915 PMCID: PMC7187163 DOI: 10.1002/biof.1559] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 08/14/2019] [Indexed: 12/12/2022]
Abstract
Redox control is lost when the antioxidant defense system cannot remove abnormally high concentrations of signaling molecules, such as reactive oxygen species (ROS). Chronically elevated levels of ROS cause oxidative stress that may eventually lead to cancer and cardiovascular and neurodegenerative diseases. In this review, we focus on redox effects in the vascular system. We pay close attention to the subcompartments of the vascular system (endothelium, smooth muscle cell layer) and give an overview of how redox changes influence those different compartments. We also review the core aspects of redox biology, cardiovascular physiology, and pathophysiology. Moreover, the topic-specific knowledgebase DES-RedoxVasc was used to develop two case studies, one focused on endothelial cells and the other on the vascular smooth muscle cells, as a starting point to possibly extend our knowledge of redox control in vascular biology.
Collapse
Affiliation(s)
- Milan Obradovic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| | - Magbubah Essack
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE)ThuwalKingdom of Saudi Arabia
| | - Sonja Zafirovic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| | - Emina Sudar‐Milovanovic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| | - Vladan P. Bajic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| | - Christophe Van Neste
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE)ThuwalKingdom of Saudi Arabia
| | - Andreja Trpkovic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| | - Julijana Stanimirovic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| | - Vladimir B. Bajic
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE)ThuwalKingdom of Saudi Arabia
| | - Esma R. Isenovic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| |
Collapse
|
12
|
Negri S, Faris P, Berra-Romani R, Guerra G, Moccia F. Endothelial Transient Receptor Potential Channels and Vascular Remodeling: Extracellular Ca 2 + Entry for Angiogenesis, Arteriogenesis and Vasculogenesis. Front Physiol 2020; 10:1618. [PMID: 32038296 PMCID: PMC6985578 DOI: 10.3389/fphys.2019.01618] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 12/23/2019] [Indexed: 12/13/2022] Open
Abstract
Vasculogenesis, angiogenesis and arteriogenesis represent three crucial mechanisms involved in the formation and maintenance of the vascular network in embryonal and post-natal life. It has long been known that endothelial Ca2+ signals are key players in vascular remodeling; indeed, multiple pro-angiogenic factors, including vascular endothelial growth factor, regulate endothelial cell fate through an increase in intracellular Ca2+ concentration. Transient Receptor Potential (TRP) channel consist in a superfamily of non-selective cation channels that are widely expressed within vascular endothelial cells. In addition, TRP channels are present in the two main endothelial progenitor cell (EPC) populations, i.e., myeloid angiogenic cells (MACs) and endothelial colony forming cells (ECFCs). TRP channels are polymodal channels that can assemble in homo- and heteromeric complexes and may be sensitive to both pro-angiogenic cues and subtle changes in local microenvironment. These features render TRP channels the most versatile Ca2+ entry pathway in vascular endothelial cells and in EPCs. Herein, we describe how endothelial TRP channels stimulate vascular remodeling by promoting angiogenesis, arteriogenesis and vasculogenesis through the integration of multiple environmental, e.g., extracellular growth factors and chemokines, and intracellular, e.g., reactive oxygen species, a decrease in Mg2+ levels, or hypercholesterolemia, stimuli. In addition, we illustrate how endothelial TRP channels induce neovascularization in response to synthetic agonists and small molecule drugs. We focus the attention on TRPC1, TRPC3, TRPC4, TRPC5, TRPC6, TRPV1, TRPV4, TRPM2, TRPM4, TRPM7, TRPA1, that were shown to be involved in angiogenesis, arteriogenesis and vasculogenesis. Finally, we discuss the role of endothelial TRP channels in aberrant tumor vascularization by focusing on TRPC1, TRPC3, TRPV2, TRPV4, TRPM8, and TRPA1. These observations suggest that endothelial TRP channels represent potential therapeutic targets in multiple disorders featured by abnormal vascularization, including cancer, ischemic disorders, retinal degeneration and neurodegeneration.
Collapse
Affiliation(s)
- Sharon Negri
- Laboratory of General Physiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Pawan Faris
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Iraq
| | - Roberto Berra-Romani
- Department of Biomedicine, School of Medicine, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Germano Guerra
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| |
Collapse
|
13
|
Ota Y, Kuwana M. Endothelial cells and endothelial progenitor cells in the pathogenesis of systemic sclerosis. Eur J Rheumatol 2019; 7:S139-S146. [PMID: 31922471 DOI: 10.5152/eurjrheum.2019.19158] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 11/25/2019] [Indexed: 12/27/2022] Open
Abstract
Systemic sclerosis (SSc) is a connective tissue disease characterized by excessive fibrosis, microvasculopathy, and autoimmunity. Endothelial cell (EC) injury and subsequent endothelial cell dysfunction is believed to be an initial event that eventually leads to a vicious pathogenic cycle. This process is further enhanced by defective angiogenesis and vasculogenesis, as the vascular repair machinery does not work properly. Endothelial progenitor cells (EPCs) are functionally and quantitatively insufficient to recover the endothelium in SSc patients. The dysfunctional ECs and EPCs not only trigger the formation of typical vascular lesions, such as progressive intimal fibrosis in small arteries and the loss of capillaries, but also promote a series of inflammatory and profibrotic processes, such as endothelial-mesenchymal transition and recruitment and accumulation of monocytic EPCs with profibrotic properties. These processes together contribute to the accumulation of extracellular matrix in the affected tissue. This review features current insights into the roles of ECs and EPCs in the pathogenesis of SSc.
Collapse
Affiliation(s)
- Yuko Ota
- Department of Allergy and Rheumatology, Nippon Medical School Graduate School of Medicine, Tokyo, Japan
| | - Masataka Kuwana
- Department of Allergy and Rheumatology, Nippon Medical School Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
14
|
Circulating Metabolites Originating from Gut Microbiota Control Endothelial Cell Function. Molecules 2019; 24:molecules24213992. [PMID: 31694161 PMCID: PMC6864778 DOI: 10.3390/molecules24213992] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/15/2019] [Accepted: 11/03/2019] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular functionality strictly depends on endothelial cell trophism and proper biochemical function. Any condition (environmental, pharmacological/toxicological, physical, or neuro-humoral) that changes the vascular endothelium has great consequences for the organism’s wellness and on the outcome and evolution of severe cardiovascular pathologies. Thus, knowledge of the mechanisms, both endogenous and external, that affect endothelial dysfunction is pivotal to preventing and treating these disorders. In recent decades, significant attention has been focused on gut microbiota and how these symbiotic microorganisms can influence host health and disease development. Indeed, dysbiosis has been reported to be at the base of a range of different pathologies, including pathologies of the cardiovascular system. The study of the mechanism underlying this relationship has led to the identification of a series of metabolites (released by gut bacteria) that exert different effects on all the components of the vascular system, and in particular on endothelial cells. The imbalance of factors promoting or blunting endothelial cell viability and function and angiogenesis seems to be a potential target for the development of new therapeutic interventions. This review highlights the circulating factors identified to date, either directly produced by gut microbes or resulting from the metabolism of diet derivatives as polyphenols.
Collapse
|
15
|
Meyers CA, Xu J, Zhang L, Chang L, Wang Y, Asatrian G, Ding C, Yan N, Zou E, Broderick K, Lee M, Peault B, James AW. Skeletogenic Capacity of Human Perivascular Stem Cells Obtained Via Magnetic-Activated Cell Sorting. Tissue Eng Part A 2019; 25:1658-1666. [PMID: 31020920 DOI: 10.1089/ten.tea.2019.0031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Human perivascular stem/stromal cells (PSC) are a multipotent mesenchymal progenitor cell population defined by their perivascular residence. PSC are increasingly studied for their application in skeletal regenerative medicine. PSC from subcutaneous white adipose tissue are most commonly isolated via fluorescence-activated cell sorting (FACS), and defined as a bipartite population of CD146+CD34-CD31-CD45- pericytes and CD34+CD146-CD31-CD45- adventitial cells. FACS poses several challenges for clinical translation, including requirements for facilities, equipment, and personnel. The purpose of this study is to identify if magnetic-activated cell sorting (MACS) is a feasible method to derive PSC, and to determine if MACS-derived PSC are comparable to our previous experience with FACS-derived PSC. In brief, CD146+ pericytes and CD34+ adventitial cells were enriched from human lipoaspirate using a multistep column approach. Next, cell identity and purity were analyzed by flow cytometry. In vitro multilineage differentiation studies were performed with MACS-defined PSC subsets. Finally, in vivo application was performed in nonhealing calvarial bone defects in Scid mice. Results showed that human CD146+ pericytes and CD34+ adventitial cells may be enriched by MACS, with defined purity, anticipated cell surface marker expression, and capacity for multilineage differentiation. In vivo, MACS-derived PSC induce ossification of bone defects. These data document the feasibility of a MACS approach for the enrichment and application of PSC in the field of tissue engineering and regenerative medicine. Impact Statement Our findings suggest that perivascular stem/stromal cells, and in particular adventitial cells, may be isolated by magnetic-activated cell sorting and applied as an uncultured autologous stem cell therapy in a same-day setting for bone defect repair.
Collapse
Affiliation(s)
- Carolyn A Meyers
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Jiajia Xu
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Leititia Zhang
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland.,Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, Shenyang, Liaoning Province, P.R. China
| | - Leslie Chang
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Yiyun Wang
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Greg Asatrian
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, California
| | - Catherine Ding
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, California
| | - Noah Yan
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Erin Zou
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Kristen Broderick
- Department of Surgery, Johns Hopkins University, Baltimore, Maryland
| | - Min Lee
- School of Dentistry, University of California, Los Angeles, California
| | - Bruno Peault
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, California.,Center For Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland.,UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, California
| |
Collapse
|
16
|
Endothelial Ca 2+ Signaling, Angiogenesis and Vasculogenesis: just What It Takes to Make a Blood Vessel. Int J Mol Sci 2019; 20:ijms20163962. [PMID: 31416282 PMCID: PMC6721072 DOI: 10.3390/ijms20163962] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/09/2019] [Accepted: 08/13/2019] [Indexed: 12/13/2022] Open
Abstract
It has long been known that endothelial Ca2+ signals drive angiogenesis by recruiting multiple Ca2+-sensitive decoders in response to pro-angiogenic cues, such as vascular endothelial growth factor, basic fibroblast growth factor, stromal derived factor-1α and angiopoietins. Recently, it was shown that intracellular Ca2+ signaling also drives vasculogenesis by stimulation proliferation, tube formation and neovessel formation in endothelial progenitor cells. Herein, we survey how growth factors, chemokines and angiogenic modulators use endothelial Ca2+ signaling to regulate angiogenesis and vasculogenesis. The endothelial Ca2+ response to pro-angiogenic cues may adopt different waveforms, ranging from Ca2+ transients or biphasic Ca2+ signals to repetitive Ca2+ oscillations, and is mainly driven by endogenous Ca2+ release through inositol-1,4,5-trisphosphate receptors and by store-operated Ca2+ entry through Orai1 channels. Lysosomal Ca2+ release through nicotinic acid adenine dinucleotide phosphate-gated two-pore channels is, however, emerging as a crucial pro-angiogenic pathway, which sustains intracellular Ca2+ mobilization. Understanding how endothelial Ca2+ signaling regulates angiogenesis and vasculogenesis could shed light on alternative strategies to induce therapeutic angiogenesis or interfere with the aberrant vascularization featuring cancer and intraocular disorders.
Collapse
|
17
|
Carretero-Ortega J, Chhangawala Z, Hunt S, Narvaez C, Menéndez-González J, Gay CM, Zygmunt T, Li X, Torres-Vázquez J. GIPC proteins negatively modulate Plexind1 signaling during vascular development. eLife 2019; 8:e30454. [PMID: 31050647 PMCID: PMC6499541 DOI: 10.7554/elife.30454] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 04/15/2019] [Indexed: 12/18/2022] Open
Abstract
Semaphorins (SEMAs) and their Plexin (PLXN) receptors are central regulators of metazoan cellular communication. SEMA-PLXND1 signaling plays important roles in cardiovascular, nervous, and immune system development, and cancer biology. However, little is known about the molecular mechanisms that modulate SEMA-PLXND1 signaling. As PLXND1 associates with GIPC family endocytic adaptors, we evaluated the requirement for the molecular determinants of their association and PLXND1's vascular role. Zebrafish that endogenously express a Plxnd1 receptor with a predicted impairment in GIPC binding exhibit low penetrance angiogenesis deficits and antiangiogenic drug hypersensitivity. Moreover, gipc mutant fish show angiogenic impairments that are ameliorated by reducing Plxnd1 signaling. Finally, GIPC depletion potentiates SEMA-PLXND1 signaling in cultured endothelial cells. These findings expand the vascular roles of GIPCs beyond those of the Vascular Endothelial Growth Factor (VEGF)-dependent, proangiogenic GIPC1-Neuropilin 1 complex, recasting GIPCs as negative modulators of antiangiogenic PLXND1 signaling and suggest that PLXND1 trafficking shapes vascular development.
Collapse
Affiliation(s)
- Jorge Carretero-Ortega
- Department of Cell Biology, Skirball Institute of Biomolecular MedicineNew York University Langone Medical CenterNew YorkUnited States
| | - Zinal Chhangawala
- Department of Cell Biology, Skirball Institute of Biomolecular MedicineNew York University Langone Medical CenterNew YorkUnited States
| | - Shane Hunt
- Department of Cell Biology, Skirball Institute of Biomolecular MedicineNew York University Langone Medical CenterNew YorkUnited States
| | - Carlos Narvaez
- Department of Cell Biology, Skirball Institute of Biomolecular MedicineNew York University Langone Medical CenterNew YorkUnited States
| | - Javier Menéndez-González
- Department of Cell Biology, Skirball Institute of Biomolecular MedicineNew York University Langone Medical CenterNew YorkUnited States
| | - Carl M Gay
- Department of Cell Biology, Skirball Institute of Biomolecular MedicineNew York University Langone Medical CenterNew YorkUnited States
| | - Tomasz Zygmunt
- Department of Cell Biology, Skirball Institute of Biomolecular MedicineNew York University Langone Medical CenterNew YorkUnited States
| | - Xiaochun Li
- Department of Population HealthNew York University School of MedicineNew YorkUnited States
| | - Jesús Torres-Vázquez
- Department of Cell Biology, Skirball Institute of Biomolecular MedicineNew York University Langone Medical CenterNew YorkUnited States
| |
Collapse
|
18
|
Lyle AN, Taylor WR. The pathophysiological basis of vascular disease. J Transl Med 2019; 99:284-289. [PMID: 30755702 DOI: 10.1038/s41374-019-0192-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 01/08/2019] [Indexed: 12/12/2022] Open
Affiliation(s)
- Alicia N Lyle
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - W Robert Taylor
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA. .,Division of Cardiology, Atlanta Veterans Affairs Medical Center, Decatur, GA, USA. .,Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
19
|
Use of Nutraceuticals in Angiogenesis-Dependent Disorders. Molecules 2018; 23:molecules23102676. [PMID: 30340320 PMCID: PMC6222874 DOI: 10.3390/molecules23102676] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/11/2018] [Accepted: 10/16/2018] [Indexed: 12/13/2022] Open
Abstract
The term of angiogenesis refers to the growth of new vessels from pre-existing capillaries. The phenomenon is necessary for physiological growth, repair and functioning of our organs. When occurring in a not regulated manner, it concurs to pathological conditions as tumors, eye diseases, chronic degenerative disorders. On the contrary insufficient neovascularization or endothelial disfunction accompanies ischemic and metabolic disorders. In both the cases an inflammatory and oxidative condition exists in supporting angiogenesis deregulation and endothelial dysfunction. The use of nutraceuticals with antioxidant and anti-inflammatory activities can be a therapeutic option to maintain an adequate vascularization and endothelial cell proper functioning or to blunt aberrant angiogenesis. A revision of the updated literature reports on nutraceuticals to guide endothelial cell wellness and to restore physiological tissue vascularization is the objective of this paper. The critical aspects as well as lacking data for human use will be explored from a pharmacological perspective.
Collapse
|
20
|
Marsboom G, Rehman J. Hypoxia Signaling in Vascular Homeostasis. Physiology (Bethesda) 2018; 33:328-337. [PMID: 30109825 PMCID: PMC6230550 DOI: 10.1152/physiol.00018.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/09/2018] [Accepted: 07/09/2018] [Indexed: 01/10/2023] Open
Abstract
Hypoxia signaling in the vasculature controls vascular permeability, inflammation, vascular growth, and repair of vascular injury. In this review, we summarize recent insights in this burgeoning field and highlight the importance of studying the heterogeneity of hypoxia responses among individual patients, distinct vascular beds, and even individual vascular cells.
Collapse
Affiliation(s)
- Glenn Marsboom
- Department of Pharmacology, University of Illinois College of Medicine , Chicago, Illinois
| | - Jalees Rehman
- Department of Pharmacology, University of Illinois College of Medicine , Chicago, Illinois
- Department of Medicine, Section of Cardiology, University of Illinois College of Medicine , Chicago, Illinois
| |
Collapse
|
21
|
Zhang X, Tang X, Hamblin MH, Yin KJ. Long Non-Coding RNA Malat1 Regulates Angiogenesis in Hindlimb Ischemia. Int J Mol Sci 2018; 19:ijms19061723. [PMID: 29891768 PMCID: PMC6032369 DOI: 10.3390/ijms19061723] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 05/28/2018] [Accepted: 06/05/2018] [Indexed: 11/29/2022] Open
Abstract
Angiogenesis is a complex process that depends on the delicate regulation of gene expression. Dysregulation of transcription during angiogenesis often leads to various human diseases. Emerging evidence has recently begun to show that long non-coding RNAs (lncRNAs) may mediate angiogenesis in both physiological and pathological conditions; concurrently, underlying molecular mechanisms are largely unexplored. Previously, our lab identified metastasis associates lung adenocarcinoma transcript 1 (Malat1) as an oxygen-glucose deprivation (OGD)-responsive endothelial lncRNA. Here we reported that genetic deficiency of Malat1 leads to reduced blood vessel formation and local blood flow perfusion in mouse hind limbs at one to four weeks after hindlimb ischemia. Malat1 and vascular endothelial growth factor receptor 2 (VEGFR2) levels were found to be increased in both cultured mouse primary skeletal muscle microvascular endothelial cells (SMMECs) after 16 h OGD followed by 24 h reperfusion and in mouse gastrocnemius muscle that underwent hindlimb ischemia followed by 28 days of reperfusion. Moreover, Malat1 silencing by locked nucleic acid (LNA)-GapmeRs significantly reduced tube formation, cell migration, and cell proliferation in SMMEC cultures. Mechanistically, RNA subcellular isolation and RNA-immunoprecipitation experiments demonstrate that Malat1 directly targets VEGFR2 to facilitate angiogenesis. The results suggest that Malat1 regulates cell-autonomous angiogenesis through direct regulation of VEGFR2.
Collapse
Affiliation(s)
- Xuejing Zhang
- Pittsburgh Institute of Brain Disorders & Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | - Xuelian Tang
- Pittsburgh Institute of Brain Disorders & Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | - Milton H Hamblin
- Department of Pharmacology, Tulane University School of Medicine, 1430 Tulane Avenue SL83, New Orleans, LA 70112, USA.
| | - Ke-Jie Yin
- Pittsburgh Institute of Brain Disorders & Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
22
|
Borton AH, Benson BL, Neilson LE, Saunders A, Alaiti MA, Huang AY, Jain MK, Proweller A, Ramirez-Bergeron DL. Aryl Hydrocarbon Receptor Nuclear Translocator in Vascular Smooth Muscle Cells Is Required for Optimal Peripheral Perfusion Recovery. J Am Heart Assoc 2018; 7:e009205. [PMID: 29858371 PMCID: PMC6015385 DOI: 10.1161/jaha.118.009205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 05/02/2018] [Indexed: 11/23/2022]
Abstract
BACKGROUND Limb ischemia resulting from peripheral vascular disease is a common cause of morbidity. Vessel occlusion limits blood flow, creating a hypoxic environment that damages distal tissue, requiring therapeutic revascularization. Hypoxia-inducible factors (HIFs) are key transcriptional regulators of hypoxic vascular responses, including angiogenesis and arteriogenesis. Despite vascular smooth muscle cells' (VSMCs') importance in vessel integrity, little is known about their functional responses to hypoxia in peripheral vascular disease. This study investigated the role of VSMC HIF in mediating peripheral ischemic responses. METHODS AND RESULTS We used ArntSMKO mice with smooth muscle-specific deletion of aryl hydrocarbon receptor nuclear translocator (ARNT, HIF-1β), required for HIF transcriptional activity, in a femoral artery ligation model of peripheral vascular disease. ArntSMKO mice exhibit impaired perfusion recovery despite normal collateral vessel dilation and angiogenic capillary responses. Decreased blood flow manifests in extensive tissue damage and hypoxia in ligated limbs of ArntSMKO mice. Furthermore, loss of aryl hydrocarbon receptor nuclear translocator changes the proliferation, migration, and transcriptional profile of cultured VSMCs. ArntSMKO mice display disrupted VSMC morphologic features and wrapping around arterioles and increased vascular permeability linked to decreased local blood flow. CONCLUSIONS Our data demonstrate that traditional vascular remodeling responses are insufficient to provide robust peripheral tissue reperfusion in ArntSMKO mice. In all, this study highlights HIF responses to hypoxia in arteriole VSMCs critical for the phenotypic and functional stability of vessels that aid in the recovery of blood flow in ischemic peripheral tissues.
Collapse
MESH Headings
- Animals
- Aryl Hydrocarbon Receptor Nuclear Translocator/biosynthesis
- Aryl Hydrocarbon Receptor Nuclear Translocator/genetics
- Blotting, Western
- Cells, Cultured
- Disease Models, Animal
- Gene Expression Regulation
- Immunohistochemistry
- Ischemia/genetics
- Ischemia/metabolism
- Ischemia/pathology
- Lower Extremity/blood supply
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microscopy, Confocal
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Peripheral Vascular Diseases/genetics
- Peripheral Vascular Diseases/metabolism
- Peripheral Vascular Diseases/pathology
- RNA/genetics
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Anna Henry Borton
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH
- Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH
- Harrington Heart and Vascular Institute, University Hospitals, Cleveland, OH
| | - Bryan L Benson
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Lee E Neilson
- Neurological Institute, University Hospitals, Cleveland, OH
| | - Ashley Saunders
- Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH
- Harrington Heart and Vascular Institute, University Hospitals, Cleveland, OH
| | - M Amer Alaiti
- Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH
- Harrington Heart and Vascular Institute, University Hospitals, Cleveland, OH
| | - Alex Y Huang
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH
- Angie Fowler Adolescent and Young Adult Cancer Institute and University Hospitals Rainbow Babies and Children's Hospital University Hospitals, Cleveland, OH
| | - Mukesh K Jain
- Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH
- Harrington Heart and Vascular Institute, University Hospitals, Cleveland, OH
| | - Aaron Proweller
- Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH
- Harrington Heart and Vascular Institute, University Hospitals, Cleveland, OH
| | - Diana L Ramirez-Bergeron
- Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH
- Harrington Heart and Vascular Institute, University Hospitals, Cleveland, OH
| |
Collapse
|
23
|
Affiliation(s)
- Yoshihide Asano
- Department of Dermatology; University of Tokyo Graduate School of Medicine; Tokyo Japan
| |
Collapse
|
24
|
Rocca A, Tafuri D, Paccone M, Giuliani A, Zamboli AGI, Surfaro G, Paccone A, Compagna R, Amato M, Serra R, Amato B. Cell Based Therapeutic Approach in Vascular Surgery: Application and Review. Open Med (Wars) 2017; 12:308-322. [PMID: 29071303 PMCID: PMC5651406 DOI: 10.1515/med-2017-0045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 08/16/2017] [Indexed: 01/14/2023] Open
Abstract
Multipotent stem cells - such as mesenchymal stem/stromal cells and stem cells derived from different sources like vascular wall are intensely studied to try to rapidly translate their discovered features from bench to bedside. Vascular wall resident stem cells recruitment, differentiation, survival, proliferation, growth factor production, and signaling pathways transduced were analyzed. We studied biological properties of vascular resident stem cells and explored the relationship from several factors as Matrix Metalloproteinases (MMPs) and regulations of biological, translational and clinical features of these cells. In this review we described a translational and clinical approach to Adult Vascular Wall Resident Multipotent Vascular Stem Cells (VW-SCs) and reported their involvement in alternative clinical approach as cells based therapy in vascular disease like arterial aneurysms or peripheral arterial obstructive disease.
Collapse
Affiliation(s)
- Aldo Rocca
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, ItalyVia Sergio Pansini, 80131Naples, Italy
| | - Domenico Tafuri
- Department of Sport Sciences and Wellness, University of Naples “Parthenope”, Naples, Italy
| | - Marianna Paccone
- Department of Medicine and Health Sciences Vincenzo Tiberio, University of Molise, Campobasso, Italy
| | - Antonio Giuliani
- A.O.R.N. A. Cardarelli Hepatobiliary and Liver Transplatation Center, Naples, Italy
| | | | - Giuseppe Surfaro
- Antonio Cardarelli Hospital, General Surgery Unit, Campobasso, Italy
| | - Andrea Paccone
- Department of Medicine and Health Sciences Vincenzo Tiberio, University of Molise, Campobasso, Italy
| | - Rita Compagna
- Department of Translational Medical Sciences, University of Naples “Federico II”, Naples, Italy
| | - Maurizo Amato
- Department of Translational Medical Sciences, University of Naples “Federico II”, Naples, Italy
| | - Raffaele Serra
- Department of Medical and Surgical Sciences, University of Catanzaro, Catanzaro, Italy
| | - Bruno Amato
- Department of Translational Medical Sciences, University of Naples “Federico II”, Naples, Italy
| |
Collapse
|
25
|
Zhang M, Yu WZ, Shen XT, Xiang Q, Xu J, Yang JJ, Chen PP, Fan ZL, Xiao J, Zhao YZ, Lu CT. Advanced Interfere Treatment of Diabetic Cardiomyopathy Rats by aFGF-Loaded Heparin-Modified Microbubbles and UTMD Technique. Cardiovasc Drugs Ther 2017; 30:247-61. [PMID: 26947349 DOI: 10.1007/s10557-016-6639-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
This study aims to investigate the preclinical performance and mechanism of a novel strategy of aFGF-loaded heparin-modified microbubbles (aFGF-HMB) combined with ultrasound-targeted microbubble destruction (UTMD) technique for diabetic cardiomyopathy (DCM) prevention. Type 1 diabetic rats were induced by streptozotocin. Twelve weeks after intervention, indexes from transthoracic echocardiography and cardiac catheterization showed that the left ventricular function in the aFGF-HMB/UTMD group was significantly improved compared with diabetes control (DM). From Picrosirius Red staining and TUNEL staining, the aFGF-HMB/UTMD group showed significant difference from the other groups. The cardiac collagen volume fraction (CVF) and myocardial cell apoptosis index (AI) in aFGF-HMB/UTMD group decreased to 7.2 % and 7.11 % respectively, compared with the DM group (CVF = 24.5 % and AI =20.3 % respectively). The results of myocardial microvascular density (MCD) also proved the strongest inhibition of aFGF-HMB/UTMD group on DCM progress. CD31 staining of aFGF-HMB/UTMD group reached 22 n/hrp, much higher than that of DM group (9 n/hrp). These results confirmed that the abnormalities including left ventricular dysfunction, myocardial fibrosis, cardiomyocytes apoptosis and microvascular rarefaction could be suppressed by twice weekly aFGF treatments for 12 consecutive weeks (free aFGF or aFGF-HMB+/-UTMD), with the strongest improvements observed in the aFGF-HMB/UTMD group (P < 0.05 vs free aFGF or aFGF-HMB). Western blot analyses of heart tissue further revealed the highest aFGF, anti-apoptosis protein (Bcl-2), VEGF-C, pAkt, pFoxo-3a levels and strongest reduction in pro-apoptosis proteins (Bax) level in aFGF-HMB/UTMD group. Overall, aFGF-HMB combined with UTMD technique might be developed as an effective strategy to prevent DCM in future clinical therapy.
Collapse
Affiliation(s)
- Ming Zhang
- College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.,Department of Ultrasonography, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.,Department of Pharmacy, Ningbo Yinzhou Second Hospital, Ningbo, Zhejiang, 315100, China
| | - Wen-Ze Yu
- College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xiao-Tong Shen
- Department of Pharmacy, People's Hospital of Deqing County, Huzhou, Zhejiang, 313200, China
| | - Qi Xiang
- Biopharmaceutical R&D Center of Jinan University, Guangzhou, Guangdong, 510000, China
| | - Jie Xu
- College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jing-Jing Yang
- College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Pian-Pian Chen
- College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Zi-Liang Fan
- College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jian Xiao
- College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| | - Ying-Zheng Zhao
- College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| | - Cui-Tao Lu
- Department of Ultrasonography, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| |
Collapse
|
26
|
Wauman J, Zabeau L, Tavernier J. The Leptin Receptor Complex: Heavier Than Expected? Front Endocrinol (Lausanne) 2017; 8:30. [PMID: 28270795 PMCID: PMC5318964 DOI: 10.3389/fendo.2017.00030] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/01/2017] [Indexed: 12/31/2022] Open
Abstract
Under normal physiological conditions, leptin and the leptin receptor (ObR) regulate the body weight by balancing food intake and energy expenditure. However, this adipocyte-derived hormone also directs peripheral processes, including immunity, reproduction, and bone metabolism. Leptin, therefore, can act as a metabolic switch connecting the body's nutritional status to high energy consuming processes. We provide an extensive overview of current structural insights on the leptin-ObR interface and ObR activation, coupling to signaling pathways and their negative regulation, and leptin functioning under normal and pathophysiological conditions (obesity, autoimmunity, cancer, … ). We also discuss possible cross-talk with other receptor systems on the receptor (extracellular) and signaling cascade (intracellular) levels.
Collapse
Affiliation(s)
- Joris Wauman
- Cytokine Receptor Laboratory, Faculty of Medicine and Health Sciences, Department of Biochemistry, Ghent University, Ghent, Belgium
- VIB Medical Biotechnology Center, VIB, Ghent, Belgium
| | - Lennart Zabeau
- Cytokine Receptor Laboratory, Faculty of Medicine and Health Sciences, Department of Biochemistry, Ghent University, Ghent, Belgium
- VIB Medical Biotechnology Center, VIB, Ghent, Belgium
| | - Jan Tavernier
- Cytokine Receptor Laboratory, Faculty of Medicine and Health Sciences, Department of Biochemistry, Ghent University, Ghent, Belgium
- VIB Medical Biotechnology Center, VIB, Ghent, Belgium
- *Correspondence: Jan Tavernier,
| |
Collapse
|
27
|
Lanier V, Gillespie C, Leffers M, Daley-Brown D, Milner J, Lipsey C, Webb N, Anderson LM, Newman G, Waltenberger J, Gonzalez-Perez RR. Leptin-induced transphosphorylation of vascular endothelial growth factor receptor increases Notch and stimulates endothelial cell angiogenic transformation. Int J Biochem Cell Biol 2016; 79:139-150. [PMID: 27590851 DOI: 10.1016/j.biocel.2016.08.023] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 08/13/2016] [Accepted: 08/29/2016] [Indexed: 02/08/2023]
Abstract
Leptin increases vascular endothelial growth factor (VEGF), VEGF receptor-2 (VEGFR-2), and Notch expression in cancer cells, and transphosphorylates VEGFR-2 in endothelial cells. However, the mechanisms involved in leptin's actions in endothelial cells are not completely known. Here we investigated whether a leptin-VEGFR-Notch axis is involved in these leptin's actions. To this end, human umbilical vein and porcine aortic endothelial cells (wild type and genetically modified to overexpress VEGFR-1 or -2) were cultured in the absence of VEGF and treated with leptin and inhibitors of Notch (gamma-secretase inhibitors: DAPT and S2188, and silencing RNA), VEGFR (kinase inhibitor: SU5416, and silencing RNA) and leptin receptor, OB-R (pegylated leptin peptide receptor antagonist 2: PEG-LPrA2). Interestingly, in the absence of VEGF, leptin induced the expression of several components of Notch signaling pathway in endothelial cells. Inhibition of VEGFR and Notch signaling significantly decreased leptin-induced S-phase progression, proliferation, and tube formation in endothelial cells. Moreover, leptin/OB-R induced transphosphorylation of VEGFR-1 and VEGFR-2 was essential for leptin's effects. These results unveil for the first time a novel mechanism by which leptin could induce angiogenic features via upregulation/trans-activation of VEGFR and downstream expression/activation of Notch in endothelial cells. Thus, high levels of leptin found in overweight and obese patients might lead to increased angiogenesis by activating VEGFR-Notch signaling crosstalk in endothelial cells. These observations might be highly relevant for obese patients with cancer, where leptin/VEGFR/Notch crosstalk could play an important role in cancer growth, and could be a new target for the control of tumor angiogenesis.
Collapse
Affiliation(s)
- Viola Lanier
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, United States
| | - Corey Gillespie
- Atlanta Technical College, Bioscience Technology Program, Atlanta, GA 30310, United States
| | | | - Danielle Daley-Brown
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, United States
| | - Joy Milner
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, United States
| | - Crystal Lipsey
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, United States
| | - Nia Webb
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, United States
| | - Leonard M Anderson
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, United States; Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, GA 30310, United States
| | - Gale Newman
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, United States
| | | | - Ruben Rene Gonzalez-Perez
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, United States.
| |
Collapse
|
28
|
Lu WH, Shi YX, Ma ZL, Wang G, Liu L, Chuai M, Song X, Münsterberg A, Cao L, Yang X. Proper autophagy is indispensable for angiogenesis during chick embryo development. Cell Cycle 2016; 15:1742-54. [PMID: 27163719 DOI: 10.1080/15384101.2016.1184803] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
People have known that autophagy plays a very important role in many physiological and pathological events. But the role of autophagy on embryonic angiogenesis still remains obscure. In this study, we demonstrated that Atg7, Atg8 and Beclin1 were expressed in the plexus vessels of angiogenesis at chick yolk sac membrane and chorioallantoic membrane. Interfering in autophagy with autophagy inducer or inhibitor could restrict the angiogenesis in vivo, which might be driven by the disorder of angiogenesis-related gene expressions, and also lead to embryonic hemorrhage, which was due to imperfection cell junctions in endothelial cells including abnormal expressions of tight junction, adheren junction and desmosome genes. Using HUVECs, we revealed that cell viability and migration ability changed with the alteration of cell autophagy exposed to RAPA or 3-MA. Interestingly, tube formation assay showed that HUVECs ability of tube formation altered with the change of Atg5, Atg7 and Atg8 manipulated by the transfection of their corresponding siRNA or plasmids. Moreover, the lost cell polarity labeled by F-actin and the absenced β-catenin in RAPA-treated and 3-MA-treated cell membrane implied intracellular cytoskeleton alteration was induced by the activation and depression of autophagy. Taken together, our current experimental data reveal that autophagy is really involved in regulating angiogenesis during embryo development.
Collapse
Affiliation(s)
- Wen-Hui Lu
- a Division of Histology and Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College , Jinan University , Guangzhou , China
| | - Yu-Xun Shi
- a Division of Histology and Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College , Jinan University , Guangzhou , China
| | - Zheng-Lai Ma
- a Division of Histology and Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College , Jinan University , Guangzhou , China
| | - Guang Wang
- a Division of Histology and Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College , Jinan University , Guangzhou , China
| | - Langxia Liu
- b Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering , Jinan University , Guangzhou , China
| | - Manli Chuai
- c Division of Cell and Developmental Biology , University of Dundee , Dundee , UK
| | - Xiaoyu Song
- d Key Laboratory of Medical Cell Biology , China Medical University , Shenyang , China
| | - Andrea Münsterberg
- e School of Biological Sciences , University of East Anglia , Norwich , UK
| | - Liu Cao
- d Key Laboratory of Medical Cell Biology , China Medical University , Shenyang , China
| | - Xuesong Yang
- a Division of Histology and Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College , Jinan University , Guangzhou , China
| |
Collapse
|
29
|
Zhao YZ, Zhang M, Wong HL, Tian XQ, Zheng L, Yu XC, Tian FR, Mao KL, Fan ZL, Chen PP, Li XK, Lu CT. Prevent diabetic cardiomyopathy in diabetic rats by combined therapy of aFGF-loaded nanoparticles and ultrasound-targeted microbubble destruction technique. J Control Release 2016; 223:11-21. [DOI: 10.1016/j.jconrel.2015.12.030] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Revised: 12/10/2015] [Accepted: 12/16/2015] [Indexed: 01/01/2023]
|
30
|
Caolo V, Vries M, Zupancich J, Houben M, Mihov G, Wagenaar A, Swennen G, Nossent Y, Quax P, Suylen D, Dijkgraaf I, Molin D, Hackeng T, Post M. CXCL1 microspheres: a novel tool to stimulate arteriogenesis. Drug Deliv 2015; 23:2919-2926. [PMID: 26651867 DOI: 10.3109/10717544.2015.1120366] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
CONTEXT After arterial occlusion, diametrical growth of pre-existing natural bypasses around the obstruction, i.e. arteriogenesis, is the body's main coping mechanism. We have shown before that continuous infusion of chemokine (C-X-C motif) ligand 1 (CXCL1) promotes arteriogenesis in a rodent hind limb ischemia model. OBJECTIVE For clinical translation of these positive results, we developed a new administration strategy of local and sustained delivery. Here, we investigate the therapeutic potential of CXCL1 in a drug delivery system based on microspheres. MATERIALS AND METHODS We generated poly(ester amide) (PEA) microspheres loaded with CXCL1 and evaluated them in vitro for cellular toxicity and chemokine release characteristics. In vivo, murine femoral arteries were ligated and CXCL1 was administered either intra-arterially via osmopump or intramuscularly encapsulated in biodegradable microspheres. Perfusion recovery was measured with Laser-Doppler. RESULTS The developed microspheres were not cytotoxic and displayed a sustained chemokine release up to 28 d in vitro. The amount of released CXCL1 was 100-fold higher than levels in native ligated hind limb. Also, the CXCL1-loaded microspheres significantly enhanced perfusion recovery at day 7 after ligation compared with both saline and non-loaded conditions (55.4 ± 5.0% CXCL1-loaded microspheres versus 43.1 ± 4.5% non-loaded microspheres; n = 8-9; p < 0.05). On day 21 after ligation, the CXCL1-loaded microspheres performed even better than continuous CXCL1 administration (102.1 ± 4.4% CXCL1-loaded microspheres versus 85.7 ± 4.8% CXCL1 osmopump; n = 9; p < 0.05). CONCLUSION Our results demonstrate a proof of concept that sustained, local delivery of CXCL1 encapsulated in PEA microspheres provides a new tool to stimulate arteriogenesis in vivo.
Collapse
Affiliation(s)
- Vincenza Caolo
- a Department of Physiology , CARIM, Maastricht University , The Netherlands
| | - Mark Vries
- a Department of Physiology , CARIM, Maastricht University , The Netherlands
| | | | | | | | - Allard Wagenaar
- a Department of Physiology , CARIM, Maastricht University , The Netherlands
| | - Geertje Swennen
- a Department of Physiology , CARIM, Maastricht University , The Netherlands
| | - Yaël Nossent
- d Department of Surgery , Leiden University Medical Center , The Netherlands , and
| | - Paul Quax
- d Department of Surgery , Leiden University Medical Center , The Netherlands , and
| | - Dennis Suylen
- e Department of Biochemistry , CARIM, Maastricht University , The Netherlands
| | - Ingrid Dijkgraaf
- e Department of Biochemistry , CARIM, Maastricht University , The Netherlands
| | - Daniel Molin
- a Department of Physiology , CARIM, Maastricht University , The Netherlands
| | - Tilman Hackeng
- e Department of Biochemistry , CARIM, Maastricht University , The Netherlands
| | - Mark Post
- a Department of Physiology , CARIM, Maastricht University , The Netherlands
| |
Collapse
|
31
|
Haas TL, Nwadozi E. Regulation of skeletal muscle capillary growth in exercise and disease. Appl Physiol Nutr Metab 2015; 40:1221-32. [PMID: 26554747 DOI: 10.1139/apnm-2015-0336] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Capillaries, which are the smallest and most abundant type of blood vessel, form the primary site of gas, nutrient, and waste transfer between the vascular and tissue compartments. Skeletal muscle exhibits the capacity to generate new capillaries (angiogenesis) as an adaptation to exercise training, thus ensuring that the heightened metabolic demand of the active muscle is matched by an improved capacity for distribution of gases, nutrients, and waste products. This review summarizes the current understanding of the regulation of skeletal muscle capillary growth. The multi-step process of angiogenesis is coordinated through the integration of a diverse array of signals associated with hypoxic, metabolic, hemodynamic, and mechanical stresses within the active muscle. The contributions of metabolic and mechanical factors to the modulation of key pro- and anti-angiogenic molecules are discussed within the context of responses to a single aerobic exercise bout and short-term and long-term training. Finally, the paradoxical lack of angiogenesis in peripheral artery disease and diabetes and the implications for disease progression and muscle health are discussed. Future studies that emphasize an integrated analysis of the mechanisms that control skeletal muscle capillary growth will enable development of targeted exercise programs that effectively promote angiogenesis in healthy individuals and in patient populations.
Collapse
Affiliation(s)
- Tara L Haas
- Angiogenesis Research Group, York University, Toronto, ON M3J 1P3, Canada
- Angiogenesis Research Group, York University, Toronto, ON M3J 1P3, Canada
| | - Emmanuel Nwadozi
- Angiogenesis Research Group, York University, Toronto, ON M3J 1P3, Canada
- Angiogenesis Research Group, York University, Toronto, ON M3J 1P3, Canada
| |
Collapse
|
32
|
Interaction of Hydrogen Sulfide with Nitric Oxide in the Cardiovascular System. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:6904327. [PMID: 26640616 PMCID: PMC4657111 DOI: 10.1155/2016/6904327] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Accepted: 06/21/2015] [Indexed: 01/07/2023]
Abstract
Historically acknowledged as toxic gases, hydrogen sulfide (H2S) and nitric oxide (NO) are now recognized as the predominant members of a new family of signaling molecules, “gasotransmitters” in mammals. While H2S is biosynthesized by three constitutively expressed enzymes (CBS, CSE, and 3-MST) from L-cysteine and homocysteine, NO is generated endogenously from L-arginine by the action of various isoforms of NOS. Both gases have been transpired as the key and independent regulators of many physiological functions in mammalian cardiovascular, nervous, gastrointestinal, respiratory, and immune systems. The analogy between these two gasotransmitters is evident not only from their paracrine mode of signaling, but also from the identical and/or shared signaling transduction pathways. With the plethora of research in the pathophysiological role of gasotransmitters in various systems, the existence of interplay between these gases is being widely accepted. Chemical interaction between NO and H2S may generate nitroxyl (HNO), which plays a specific effective role within the cardiovascular system. In this review article, we have attempted to provide current understanding of the individual and interactive roles of H2S and NO signaling in mammalian cardiovascular system, focusing particularly on heart contractility, cardioprotection, vascular tone, angiogenesis, and oxidative stress.
Collapse
|
33
|
Moccia F, Guerra G. Ca2+Signalling in Endothelial Progenitor Cells: Friend or Foe? J Cell Physiol 2015; 231:314-27. [DOI: 10.1002/jcp.25126] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 08/04/2015] [Indexed: 01/06/2023]
Affiliation(s)
- Francesco Moccia
- Laboratory of General Physiology; Department of Biology and Biotechnology “Lazzaro Spallanzani”; University of Pavia; Pavia Italy
| | - Germano Guerra
- Department of Medicine and Health Sciences “Vincenzo Tiberio”; University of Molise; Campobasso Italy
| |
Collapse
|
34
|
Gahr S, Mayr C, Kiesslich T, Illig R, Neureiter D, Alinger B, Ganslmayer M, Wissniowski T, Fazio PD, Montalbano R, Ficker JH, Ocker M, Quint K. The pan-deacetylase inhibitor panobinostat affects angiogenesis in hepatocellular carcinoma models via modulation of CTGF expression. Int J Oncol 2015. [PMID: 26202945 DOI: 10.3892/ijo.2015.3087] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Post-translational modifications of chromatin components are significantly involved in the regulation of tumor suppressor gene and oncogene expression. Connective tissue growth factor (CTGF) is an epigenetically regulated growth factor with functions in angiogenesis and cell-matrix interactions and plays a pivotal role in hepatocellular carcinoma (HCC). The pharmacologic inhibition of histone and protein deacetylases represents a new approach to interfere with pathways of apoptosis and angiogenesis. We investigated the effect of the pan-deacetylase inhibitor panobinostat (LBH589) on human HCC cell lines HepG2 (p53wt) and Hep3B (p53null) and in a subcutaneous xenograft model and explored the influence on angiogenesis. Specimens were characterized by quantitative real-time PCR. Protein was separated for western blotting against CTGF, VEGF, VEGF receptor-1 (VEGFR-1/FLT-1), VEGF receptor-2 (VEGFR-2/KDR), MAPK and phospho-MAPK. In vivo, HepG2 cells were xenografted to NMRI mice and treated with daily i.p. injections of 10 mg/kg panobinostat. After 1, 7 and 28 days, real-time PCR was performed. Immunohistochemistry and western blotting were examined after 28 days. An increased significant expression of CTGF was only seen after 24 h treatment with 0.1 µM panobinostat in HepG2 cells and Hep3B cells, whereas after 72 h treatment CTGF expression clearly decreased. In the xenografts, treatment with panobinostat showed a minimal CTGF expression after 1 day and 4 weeks, respectively. In vitro as well as in vivo, VEGF was not affected by panobinostat treatment at any time. In conclusion, panobinostat influences extracellular signaling cascades via CTGF-dependent pathways.
Collapse
Affiliation(s)
- Susanne Gahr
- Department of Medicine 1, University Hospital Erlangen, Erlangen, Germany
| | - Christian Mayr
- Laboratory for Tumour Biology and Experimental Therapies, Paracelsus Medical University, Salzburg, Austria
| | - Tobias Kiesslich
- Laboratory for Tumour Biology and Experimental Therapies, Paracelsus Medical University, Salzburg, Austria
| | - Romana Illig
- Institute of Pathology, Salzburger Landeskliniken, Paracelsus Private Medical University, Salzburg, Austria
| | - Daniel Neureiter
- Institute of Pathology, Salzburger Landeskliniken, Paracelsus Private Medical University, Salzburg, Austria
| | - Beate Alinger
- Institute of Pathology, Salzburger Landeskliniken, Paracelsus Private Medical University, Salzburg, Austria
| | - Marion Ganslmayer
- Department of Medicine 1, University Hospital Erlangen, Erlangen, Germany
| | - Till Wissniowski
- Department of Medicine 1, University Hospital Erlangen, Erlangen, Germany
| | - Pietro Di Fazio
- Institute for Surgical Research, Phillips University Marburg, Marburg, Germany
| | - Roberta Montalbano
- Institute for Surgical Research, Phillips University Marburg, Marburg, Germany
| | - Joachim H Ficker
- Klinikum Nuernberg, Department of Respiratory Medicine, Allergology and Sleep Medicine, Nuremberg, Germany
| | - Matthias Ocker
- Department of Medicine 1, University Hospital Erlangen, Erlangen, Germany
| | - Karl Quint
- Department of Medicine 1, University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
35
|
Abstract
Systemic sclerosis (SSc) is a multisystem connective tissue disorder featured by vascular injury and fibrosis of the skin and various internal organs with autoimmune background. Although the pathogenesis of SSc still remains elusive, it is generally accepted that initial vascular injury due to autoimmunity and/or environmental factors causes structural and functional abnormalities of vasculature which eventually result in the constitutive activation of fibroblasts in various organs. Structural alterations consist of destructive vasculopathy (loss of small vessels) and proliferative obliterative vasculopathy (occlusion of arterioles and small arteries with fibro-proliferative change) caused by impaired compensatory vasculogenesis and angiogenesis. Impaired function of SSc vasculature includes the altered expression of cell adhesion molecules predominantly inducing Th2 and Th17 cell infiltration, endothelial dysfunction primarily due to the low availability of nitric oxide, the activated endothelial-to-mesenchymal transition leading to fibro-proliferative vascular change and tissue fibrosis, and the impaired coagulation/fibrinolysis system promoting the formation of intravascular fibrin deposits. Recent new insights into the therapeutic mechanisms of intravenous cyclophosphamide pulse and bosentan and the establishment of a new SSc animal model (Klf5 (+/-);Fli1 (+/-) mice) provide us useful clues to further understand the development of vascular alterations characteristic of SSc. This article overviewed the present understanding of the pathogenesis of SSc vasculopathy.
Collapse
|
36
|
Chistiakov DA, Orekhov AN, Bobryshev YV. Vascular smooth muscle cell in atherosclerosis. Acta Physiol (Oxf) 2015; 214:33-50. [PMID: 25677529 DOI: 10.1111/apha.12466] [Citation(s) in RCA: 294] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 02/02/2015] [Accepted: 02/09/2015] [Indexed: 12/30/2022]
Abstract
Vascular smooth muscle cells (VSMCs) exhibit phenotypic and functional plasticity in order to respond to vascular injury. In case of the vessel damage, VSMCs are able to switch from the quiescent 'contractile' phenotype to the 'proinflammatory' phenotype. This change is accompanied by decrease in expression of smooth muscle (SM)-specific markers responsible for SM contraction and production of proinflammatory mediators that modulate induction of proliferation and chemotaxis. Indeed, activated VSMCs could efficiently proliferate and migrate contributing to the vascular wall repair. However, in chronic inflammation that occurs in atherosclerosis, arterial VSMCs become aberrantly regulated and this leads to increased VSMC dedifferentiation and extracellular matrix formation in plaque areas. Proatherosclerotic switch in VSMC phenotype is a complex and multistep mechanism that may be induced by a variety of proinflammatory stimuli and hemodynamic alterations. Disturbances in hemodynamic forces could initiate the proinflammatory switch in VSMC phenotype even in pre-clinical stages of atherosclerosis. Proinflammatory signals play a crucial role in further dedifferentiation of VSMCs in affected vessels and propagation of pathological vascular remodelling.
Collapse
Affiliation(s)
- D. A. Chistiakov
- Research Center for Children's Health; Moscow Russia
- The Mount Sinai Community Clinical Oncology Program; Mount Sinai Comprehensive Cancer Center; Mount Sinai Medical Center; Miami Beach FL USA
| | - A. N. Orekhov
- Institute for Atherosclerosis; Skolkovo Innovative Center; Moscow Russia
- Laboratory of Angiopathology; Institute of General Pathology and Pathophysiology; Russian Academy of Sciences; Moscow Russia
- Department of Biophysics; Biological Faculty; Moscow State University; Moscow Russia
| | - Y. V. Bobryshev
- Institute for Atherosclerosis; Skolkovo Innovative Center; Moscow Russia
- Faculty of Medicine; School of Medical Sciences; University of New South Wales; Kensington Sydney NSW Australia
- School of Medicine; University of Western Sydney; Campbelltown NSW Australia
| |
Collapse
|
37
|
Combined effect of insulin-like growth factor-1 and CC chemokine ligand 2 on angiogenic events in endothelial cells. PLoS One 2015; 10:e0121249. [PMID: 25830234 PMCID: PMC4382320 DOI: 10.1371/journal.pone.0121249] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 01/29/2015] [Indexed: 12/31/2022] Open
Abstract
Therapeutic angiogenesis may be applied in medical conditions to promote stimulation of angiogenesis. Angiogenesis is a multistep process, which includes endothelial cell proliferation, migration, and tube formation, which is mediated by various angiogenic polypeptides. Thus, studies that elucidate the cellular mechanisms involved in these processes are necessary to develop novel therapeutic strategies. This study investigated the in vitro effects of the pro-angiogenic factors, insulin-like growth factor-1 (IGF-1) and/or chemokine (CC motif) ligand 2 (CCL2), on endothelial cells. Flow cytometry analysis showed that IGF-1 and CCL2 treatment did not interfere with IGF-1 receptor (IGF-1R) expression, but CCL2 treatment increased CCL2 receptor (CCR2) expression. Immunofluorescence analysis revealed that the IGF-1/CCL2 combination induced a greater increase in fibronectin deposition, but the treatments did not alter the expression of the fibronectin receptors, CD49e and CD44. The interaction of fibronectin with cytokines demonstrated that IGF-1/CCL2 promoted changes in intermediate F-actin remodeling that may result in increased endothelial cell adhesion and cell migration mediated by fibronectin. Furthermore, IGF-1/CCL2 stimulated endothelial cells, grown on fibronectin, to form capillary-like structures and intercellular lumina with greater luminal area. These data suggest that IGF-1/CCL2 combination and a fibronectin matrix may contribute to the angiogenesis process to stimulate adhesion, migration, and tube formation by endothelial cells as a result of F-actin remodeling.
Collapse
|
38
|
Amato B, Compagna R, Amato M, Grande R, Butrico L, Rossi A, Naso A, Ruggiero M, de Franciscis S, Serra R. Adult vascular wall resident multipotent vascular stem cells, matrix metalloproteinases, and arterial aneurysms. Stem Cells Int 2015; 2015:434962. [PMID: 25866513 PMCID: PMC4381852 DOI: 10.1155/2015/434962] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Revised: 02/23/2015] [Accepted: 03/06/2015] [Indexed: 12/20/2022] Open
Abstract
Evidences have shown the presence of multipotent stem cells (SCs) at sites of arterial aneurysms: they can differentiate into smooth muscle cells (SMCs) and are activated after residing in a quiescent state in the vascular wall. Recent studies have implicated the role of matrix metalloproteinases in the pathogenesis of arterial aneurysms: in fact the increased synthesis of MMPs by arterial SMCs is thought to be a pivotal mechanism in aneurysm formation. The factors and signaling pathways involved in regulating wall resident SC recruitment, survival, proliferation, growth factor production, and differentiation may be also related to selective expression of different MMPs. This review explores the relationship between adult vascular wall resident multipotent vascular SCs, MMPs, and arterial aneurysms.
Collapse
Affiliation(s)
- Bruno Amato
- Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology, Magna Graecia University of Catanzaro, Viale Europa, 88100 Catanzaro, Italy ; Department of Clinical Medicine and Surgery, University of Naples "Federico II", 80100 Naples, Italy
| | - Rita Compagna
- Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology, Magna Graecia University of Catanzaro, Viale Europa, 88100 Catanzaro, Italy ; Department of Clinical Medicine and Surgery, University of Naples "Federico II", 80100 Naples, Italy
| | - Maurizio Amato
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", 80100 Naples, Italy
| | - Raffaele Grande
- Department of Medical and Surgical Sciences, University of Catanzaro, 88100 Catanzaro, Italy
| | - Lucia Butrico
- Department of Medical and Surgical Sciences, University of Catanzaro, 88100 Catanzaro, Italy
| | - Alessio Rossi
- Department of Medicine and Health Sciences, University of Molise, 88100 Campobasso, Italy
| | - Agostino Naso
- Department of Medical and Surgical Sciences, University of Catanzaro, 88100 Catanzaro, Italy
| | - Michele Ruggiero
- Department of Medical and Surgical Sciences, University of Catanzaro, 88100 Catanzaro, Italy
| | - Stefano de Franciscis
- Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology, Magna Graecia University of Catanzaro, Viale Europa, 88100 Catanzaro, Italy ; Department of Medical and Surgical Sciences, University of Catanzaro, 88100 Catanzaro, Italy
| | - Raffaele Serra
- Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology, Magna Graecia University of Catanzaro, Viale Europa, 88100 Catanzaro, Italy ; Department of Medical and Surgical Sciences, University of Catanzaro, 88100 Catanzaro, Italy
| |
Collapse
|
39
|
Birbrair A, Zhang T, Wang ZM, Messi ML, Mintz A, Delbono O. Pericytes at the intersection between tissue regeneration and pathology. Clin Sci (Lond) 2015; 128:81-93. [PMID: 25236972 PMCID: PMC4200531 DOI: 10.1042/cs20140278] [Citation(s) in RCA: 173] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Perivascular multipotent cells, pericytes, contribute to the generation and repair of various tissues in response to injury. They are heterogeneous in their morphology, distribution, origin and markers, and elucidating their molecular and cellular differences may inform novel treatments for disorders in which tissue regeneration is either impaired or excessive. Moreover, these discoveries offer novel cellular targets for therapeutic approaches to many diseases. This review discusses recent studies that support the concept that pericyte subtypes play a distinctive role in myogenesis, neurogenesis, adipogenesis, fibrogenesis and angiogenesis.
Collapse
Affiliation(s)
- Alexander Birbrair
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
- Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Tan Zhang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Zhong-Min Wang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Maria Laura Messi
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Akiva Mintz
- Department of Neurosurgery, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Osvaldo Delbono
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
- Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| |
Collapse
|
40
|
Hasan A, Paul A, Vrana NE, Zhao X, Memic A, Hwang YS, Dokmeci MR, Khademhosseini A. Microfluidic techniques for development of 3D vascularized tissue. Biomaterials 2014; 35:7308-25. [PMID: 24906345 PMCID: PMC4118596 DOI: 10.1016/j.biomaterials.2014.04.091] [Citation(s) in RCA: 188] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 04/19/2014] [Indexed: 01/17/2023]
Abstract
Development of a vascularized tissue is one of the key challenges for the successful clinical application of tissue engineered constructs. Despite the significant efforts over the last few decades, establishing a gold standard to develop three dimensional (3D) vascularized tissues has still remained far from reality. Recent advances in the application of microfluidic platforms to the field of tissue engineering have greatly accelerated the progress toward the development of viable vascularized tissue constructs. Numerous techniques have emerged to induce the formation of vascular structure within tissues which can be broadly classified into two distinct categories, namely (1) prevascularization-based techniques and (2) vasculogenesis and angiogenesis-based techniques. This review presents an overview of the recent advancements in the vascularization techniques using both approaches for generating 3D vascular structure on microfluidic platforms.
Collapse
Affiliation(s)
- Anwarul Hasan
- Biomedical Engineering, and Department of Mechanical Engineering, American University of Beirut, Beirut 1107 2020, Lebanon; Biomaterials Innovation Research Center, Division of Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Arghya Paul
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Nihal E Vrana
- INSERM, UMR-S 1121, Biomatériaux et Bioingénierie, 11 rue Humann, F-67085 Strasbourg Cedex, France
| | - Xin Zhao
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Adnan Memic
- Center of Nanotechnology, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Yu-Shik Hwang
- Department of Maxillofacial Biomedical Engineering, School of Dentistry, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Mehmet R Dokmeci
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; World Premier International - Advanced Institute for Materials Research (WPI-AIMR), Tohoku University, Sendai 980-8577, Japan; Department of Physics, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
41
|
Chao CY, Lii CK, Ye SY, Li CC, Lu CY, Lin AH, Liu KL, Chen HW. Docosahexaenoic acid inhibits vascular endothelial growth factor (VEGF)-induced cell migration via the GPR120/PP2A/ERK1/2/eNOS signaling pathway in human umbilical vein endothelial cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2014; 62:4152-8. [PMID: 24734983 DOI: 10.1021/jf5007165] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Cell migration plays an important role in angiogenesis and wound repair. Vascular endothelial growth factor (VEGF) is an endothelial cell-specific mitogen that is essential for endothelial cell survival, proliferation, and migration. Docosahexaenoic acid (DHA), an n-3 polyunsaturated fatty acid, shows both anti-inflammatory and antioxidant activities in vitro and in vivo. This study investigated the molecular mechanism by which DHA down-regulates VEGF-induced cell migration. HUVECs were used as the study model, and the MTT assay, Western blot, wound-healing assay, and phosphatase activity assay were used to explore the effects of DHA on cell migration. GPR120 is the putative receptor for DHA action. The results showed that DHA, PD98059 (an ERK1/2 inhibitor), and GW9508 (a GPR120 agonist) inhibited VEGF-induced cell migration. In contrast, pretreatment with okadaic acid (OA, a PP2A inhibitor) and S-nitroso-N-acetyl-DL-penicillamine (an NO donor) reversed the inhibition of cell migration by DHA. VEGF-induced cell migration was accompanied by phosphorylation of ERK1/2 and eNOS. Treatment of HUVECs with DHA increased PP2A enzyme activity and decreased VEGF-induced phosphorylation of ERK1/2 and eNOS. However, pretreatment with OA significantly decreased DHA-induced PP2A enzyme activity and reversed the DHA inhibition of VEGF-induced ERK1/2 and eNOS phosphorylation. These results suggest that stimulation of PP2A activity and inhibition of the VEGF-induced ERK1/2/eNOS signaling pathway may be involved in the DHA suppression of VEGF-induced cell migration. Thus, the effect of DHA on angiogenesis and wound repair is at least partly by virtue of its attenuation of cell migration.
Collapse
Affiliation(s)
- Che-Yi Chao
- Department of Health and Nutrition Biotechnology, Asia University , Taichung, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Zhang H, van Olden C, Sweeney D, Martin-Rendon E. Blood vessel repair and regeneration in the ischaemic heart. Open Heart 2014; 1:e000016. [PMID: 25332783 PMCID: PMC4189230 DOI: 10.1136/openhrt-2013-000016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 12/11/2013] [Accepted: 12/15/2013] [Indexed: 12/19/2022] Open
Abstract
The term 'therapeutic angiogenesis' originated almost two decades ago, following evidence that factors that promote blood vessel formation could be delivered to ischaemic tissues and restore blood flow. Following this proof-of-principle, safety and efficacy of the best-studied angiogenic factors (eg, vascular endothelial growth factor) were demonstrated in early clinical studies. Promising results led to the development of larger controlled trials that, unfortunately, have failed to satisfy the initial expectations of therapeutic angiogenesis for ischaemic heart disease. As the quest to delay the progression to heart failure secondary to ischaemic heart disease continues, alternative therapies have emerged as potential novel treatments to improve myocardial reperfusion and long-term heart function. The disappointing results of the clinical studies using angiogenic factors were followed by mixed results from the cell therapy trials. This review reflects the current angiogenic strategies for the ischaemic heart, their limitations and discusses future perspectives in the light of recent scientific and clinical evidence. It is proposed that combination therapies may be a new direction to advance therapeutic repair and regeneration of blood vessels in the ischaemic heart.
Collapse
Affiliation(s)
- Huajun Zhang
- Nuffield Department of Surgical Sciences , University of Oxford , Oxford , UK ; Stem Cell Research Laboratory , NHS Blood and Transplant, John Radcliife Hospital , Oxford , UK
| | - Casper van Olden
- Nuffield Department of Surgical Sciences , University of Oxford , Oxford , UK ; Stem Cell Research Laboratory , NHS Blood and Transplant, John Radcliife Hospital , Oxford , UK
| | - Dominic Sweeney
- Stem Cell Research Laboratory , NHS Blood and Transplant, John Radcliife Hospital , Oxford , UK ; Radcliffe Department of Medicine , University of Oxford , Oxford , UK
| | - Enca Martin-Rendon
- Stem Cell Research Laboratory , NHS Blood and Transplant, John Radcliife Hospital , Oxford , UK ; Radcliffe Department of Medicine , University of Oxford , Oxford , UK
| |
Collapse
|
43
|
Wanjare M, Kusuma S, Gerecht S. Perivascular cells in blood vessel regeneration. Biotechnol J 2013; 8:434-47. [PMID: 23554249 DOI: 10.1002/biot.201200199] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 02/19/2013] [Accepted: 03/05/2013] [Indexed: 12/21/2022]
Abstract
Vascular engineering seeks to design and construct functional blood vessels comprising endothelial cells (ECs) and perivascular cells (PCs), with the ultimate goal of clinical translation. While EC behavior has been extensively investigated, PCs play an equally significant role in the development of novel regenerative strategies, providing functionality and stability to vessels. The two major classes of PCs are vascular smooth muscle cells (vSMCs) and pericytes; vSMCs can be further sub-classified as either contractile or synthetic. The inclusion of these cell types is crucial for successful regeneration of blood vessels. Furthermore, understanding distinctions between vSMCs and pericytes will enable improved therapeutics in a tissue-specific manner. Here we focus on the approaches and challenges facing the use of PCs in vascular regeneration, including their characteristics, stem cell sources, and interactions with ECs. Finally, we discuss biochemical and microRNA (miR) regulators of PC behavior and engineering approaches that mimic various cues affecting PC function.
Collapse
Affiliation(s)
- Maureen Wanjare
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center and Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | | | | |
Collapse
|
44
|
Abstract
INTRODUCTION Therapeutic angiogenesis is a strategy of inducing new collateral vessels and stimulating new capillaries that enhance tissue oxygen exchange in ischemic cardiovascular disorders, including acute myocardial infarction, chronic cardiac ischemia, peripheral artery disease and stroke. AREAS COVERED Over the last 10 years, promising results of early clinical trials have generated great expectation on the potential of therapeutic angiogenesis. However, even if large randomized placebo-controlled and double-blinded Phase II clinical trials have confirmed the feasibility, safety and potential effectiveness of therapeutic angiogenesis, they provided very limited evidence of its efficacy in terms of clinical benefit. EXPERT OPINION Results of the latest trials on therapeutic angiogenesis have not provided satisfactory results. Much is still unknown about the optimal delivery of angiogenic factors. Trials using alternative growth factors, dose regimens and methods of delivery are needed to enhance the treatment benefit of therapeutic angiogenesis.
Collapse
Affiliation(s)
- Domenico Ribatti
- University of Bari Medical School, National Cancer Institute, Giovanni Paolo II, Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Piazza G. Cesare, 11, Policlinico, 70124 Bari, Italy.
| | | |
Collapse
|
45
|
Haddad-Mashadrizeh A, Bahrami AR, Matin MM, Edalatmanesh MA, Zomorodipour A, Gardaneh M, Farshchian M, Momeni-Moghaddam M. Human adipose-derived mesenchymal stem cells can survive and integrate into the adult rat eye following xenotransplantation. Xenotransplantation 2013; 20:165-76. [PMID: 23679842 DOI: 10.1111/xen.12033] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 03/18/2013] [Indexed: 12/17/2022]
Abstract
BACKGROUND Novel threads of discovery provide the basis for optimism for the development of a stem-cell-based strategy for the treatment of retinal blindness. Accordingly, achievement to suitable cell source with potential-to-long-term survival and appropriate differentiation can be an effective step in this direction. METHODS After derivation of human adipose-derived mesenchymal stem cells (HAD-MSCs), they were stably transfected with a vector containing Turbo-green fluorescent protein (GFP) and JRed to be able to trace them after transplantation. Labeled HAD-MSCs were transplanted into the intact adult rat eye and their survival, integration, and migration during 6 months post-transplantation were assessed. RESULTS The transplanted cells were traceable in the rat vitreous humor (VH) up until 90 days after transplantation, with gradual reduction in numbers, their adhesion and expansion capacity after recovery. These cells were also integrated into the ocular tissues. Nonetheless, some of the implanted cells succeeded to cross the blood-retina barrier (BRB) and accumulate in the spleen with time. CONCLUSIONS The survival of the HAD-MSCs for a period of 90 days in VH and even longer period of up to 6 months in other eye tissues makes them a promising source to be considered in regenerative medicine of eye diseases. However, the potency of crossing the BRB by the implanted cells suggests that use of HAD-MSCs must be handled with extreme caution.
Collapse
Affiliation(s)
- Aliakbar Haddad-Mashadrizeh
- Cell and Molecular Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran; Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Lages E, Ipas H, Guttin A, Nesr H, Berger F, Issartel JP. MicroRNAs: molecular features and role in cancer. Front Biosci (Landmark Ed) 2012; 17:2508-40. [PMID: 22652795 DOI: 10.2741/4068] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
microRNAs (miRNAs) are small noncoding endogenously produced RNAs that play key roles in controlling the expression of many cellular proteins. Once they are recruited and incorporated into a ribonucleoprotein complex miRISC, they can target specific mRNAs in a miRNA sequence-dependent process and interfere in the translation into proteins of the targeted mRNAs via several mechanisms. Consequently, miRNAs can regulate many cellular pathways and processes. Dysregulation of their physiological roles may largely contribute to disease. In particular, in cancer, miRNAs can be involved in the deregulation of the expression of important genes that play key roles in tumorigenesis, tumor development, and angiogenesis and have oncogenic or tumor suppressor roles. This review focuses on the biogenesis and maturation of miRNAs, their mechanisms of gene regulation, and the way their expression is deregulated in cancer. The involvement of miRNAs in several oncogenic pathways such as angiogenesis and apoptosis, and in the inter-cellular dialog mediated by miRNA-loaded exosomes as well as the development of new therapeutical strategies based on miRNAs will be discussed.
Collapse
Affiliation(s)
- Elodie Lages
- INSERM, U836, Team7 Nanomedicine and Brain, BP 170, Grenoble, France
| | | | | | | | | | | |
Collapse
|
47
|
Scala G, Maruccio L. Angiogenesis of buffalo choroid plexuses: structural and immunocytochemical study. Microsc Res Tech 2012; 75:1104-12. [PMID: 22434550 DOI: 10.1002/jemt.22037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 02/14/2012] [Indexed: 01/05/2023]
Abstract
Mammalian choroid plexuses (CPs) are vascularized structures involved in numerous exchange processes that supply nutrients and hormones to the brain, and that remove deleterious compounds and metabolites from the brain. Studies in the adult Mediterranean buffalo have investigated the morphology of CPs using histochemical and immunohistochemical techniques. To date, however, there have been no studies conducted on ruminants regarding this removal process which serves to repair functional vascular damage in the CPs. Each of these vascular repair processes is a very complex and none of these has not yet been completely understood. Then, the aim of the present study is to investigate the morphological processes during angiogenesis in the CPs of healthy adult buffaloes, utilizing transmission electron microscopy (TEM), scanning electron microscopy (SEM), and immunogold-labeling SEM analysis (biomarkers: angiopoietin-2 [Ang-2], vascular endothelial growth factor receptor-3 [VEGFR-3], and CD133). At TEM, the inner surface of the blood capillaries sometimes showed pillar-like cells, which in contact with endothelial cells formed prominences, which in turn formed neo-blood capillaries. With immunogold-labeling SEM analysis, the CP blood capillaries showed Ang-2 and VEGF-3, respectively, in positive particles and spheroid formations. In addition, the external surface of the blood capillaries showed spheroid formations that originated from the neo-vascular capillaries whose terminals formed a capillary network, positive to CD133. On the basis of these results, the following hypothesis can be made, namely, that these CPs are vascular structures which play a fundamental role in maintaining brain homeostasis and self-repairing of functional vascular damage, independently of the presence of rete mirabile in this species.
Collapse
Affiliation(s)
- Gaetano Scala
- Department of Biological Structures, Functions and Technologies, University of Naples Federico II, Naples, Italy.
| | | |
Collapse
|
48
|
Abstract
Ischemic disease causes a large number of deaths and significant clinical problems worldwide. Therapeutic angiogenesis, strengthened by advances in growth-factor-based therapies, is a promising solution to ischemic pathologies. Major challenges in therapeutic angiogenesis are the lack of stability of native angiogenic proteins and also providing sustained delivery of biologically active proteins at the ischemic sites. This paper will discuss various protein engineering strategies to develop stabilized proangiogenic proteins and several biomaterial technologies used to amplify the angiogenic outcome by delivering biologically active growth factors in a sustained manner.
Collapse
Affiliation(s)
- Rituparna Sinha Roy
- Department of Biological and Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur Campus, P O BCKV Campus Mail Office, West Bengal 741252, India.
| | | | | |
Collapse
|
49
|
Persson AB, Buschmann IR. Vascular growth in health and disease. Front Mol Neurosci 2011; 4:14. [PMID: 21904523 PMCID: PMC3160751 DOI: 10.3389/fnmol.2011.00014] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Accepted: 07/18/2011] [Indexed: 12/21/2022] Open
Abstract
Vascular growth forms the first functional organ system during development, and continues into adult life, wherein it is often associated with disease states. Genetically determined vasculogenesis produces a primary vascular plexus during ontogenesis. Angiogenesis, occurring, e.g., in response to metabolic stress within hypoxic tissues, enhances tissue capillarization. Arteriogenesis denotes the adaptive outgrowth of pre-existent collateral arteries to bypass arterial stenoses in response to hemodynamic changes. It has been debated whether vasculogenesis occurs in the adult, and whether or not circulating progenitor cells structurally contribute to vessel regeneration. Secondly, the major determinants of vascular growth – genetic predisposition, metabolic factors (hypoxia), and hemodynamics – cannot be assigned in a mutually exclusive fashion to vasculogenesis, angiogenesis, and arteriogenesis, respectively; rather, mechanisms overlap. Lastly, all three mechanisms of vessel growth seem to contribute to physiological embryogenesis as well as adult adaptive vascularization as occurs in tumors or to circumvent arterial stenosis. Thus, much conceptual and terminological confusion has been created, while therapies targeting neovascularization have yielded promising results in the lab, but failed randomized studies when taken to the bedside. Therefore, this review article aims at providing an exact definition of the mechanisms of vascular growth and their contribution to embryonic development as well as adult adaptive revascularization. We have been looking for potential reasons for why clinical trials have failed, how vitally the application of appropriate methods of measuring and assessment influences study outcomes, and how relevant, e.g., results gained in models of vascular occlusive disease may be for antineoplastic strategies, advocating a reverse bedside-to-bench approach, which may hopefully yield successful approaches to therapeutically targeting vascular growth.
Collapse
Affiliation(s)
- Anja Bondke Persson
- Institut fuer Vegetative Physiologie, Campus Charité Mitte, Charité Universitaetsmedizin Berlin Berlin, Germany
| | | |
Collapse
|
50
|
Fiorio Pla A, Avanzato D, Munaron L, Ambudkar IS. Ion channels and transporters in cancer. 6. Vascularizing the tumor: TRP channels as molecular targets. Am J Physiol Cell Physiol 2011; 302:C9-15. [PMID: 21832241 DOI: 10.1152/ajpcell.00280.2011] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Tumor vascularization is a critical process that determines tumor growth and metastasis. In the last decade new experimental evidence obtained from in vitro and in vivo studies have challenged the classical angiogenesis model forcing us to consider new scenarios for tumor neovascularization. In particular, the genetic stability of tumor-derived endothelial cells (TECs) has been recently questioned in several studies, which show that TECs, as well as pericytes, differ significantly from their normal counterparts at genetic and functional levels. In addition to such an epigenetic action of tumor microenvironment on endothelial cells (ECs) commitment, the distinct characteristics of TECs could be due to differences in their origin compared with preexisting differentiated ECs. Intracellular Ca(2+) signals are involved at different critical phases in the regulation of the complex process of angiogenesis and tumor progression. These signals are generated by a wide variety of intrinsic and extrinsic factors. Several key components of Ca(2+) signaling including Ca(2+) channels in the plasma membrane, endoplasmic reticulum, calcium pumps, and mitochondria contribute to the generation, amplitude, and frequency of these Ca(2+) change. In particular, several members of the transient receptor potential (TRP) family of calcium-permeable channels have profound effects on the function of ECs. Because of its multifaceted role in the control of cell function, proliferation, and motility, TRP channels have been suggested as a potential molecular target for control of tumor neovascularization. Since plasma membrane Ca(2+) channels are easily and directly accessible via the bloodstream, they are potential targets for a number of pharmacological and antibody-targeted therapeutic strategies, with specificity being the main limitation. In this review we discuss recent advances in understanding the role of Ca(2+) channels, with specific reference to TRP channels, in tumor vascularization process.
Collapse
|