1
|
Arslan GD, Dogan L, Dogan Z, Kiziltoprak H. Relationship between choroidal structure and myocardial collateral flow regulation in acute and chronic coronary heart disease. Int Ophthalmol 2025; 45:132. [PMID: 40159520 DOI: 10.1007/s10792-025-03515-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 03/12/2025] [Indexed: 04/02/2025]
Abstract
PURPOSE This study aimed to evaluate the relationship between coronary collateral filling, collateral size, and choroidal parameters in patients with acute and chronic coronary heart disease (CHD). METHODS Thirty-eight patients with acute CHD and 38 with chronic CHD who underwent diagnostic angiography in a cardiology clinic were included in this observational cross-sectional study. The control group comprised 32 healthy participants, and we examined both eyes of all participants. Diagnostic coronary angiograms were used to score the coronary collaterals, and choroidal parameters were measured in patients with CHD. RESULTS Choroidal vascular index (CVI) and subfoveal choroidal thickness (SFCT) were significantly lower in the chronic CHD group than in the acute CHD and control groups (p < 0.05). In the multinominal logistic regression analysis, collateral size had a significant association with both CVI (OR, 0.751; 95% CI, 0.596-0.947), and SFCT (OR, 0.986; 95% CI, 0.976-0.996) in patients with chronic CHD (p < 0.05). However, in the acute CHD group, no significant relationship was observed among choroidal parameters, collateral size, and filling. CONCLUSION Patients with chronic CHD had the lowest mean CVI and SFCT among the three groups, and this may be helpful in indicating chronic myocardial ischaemia. Moreover, an association was observed between larger collateral size and reduced CVI and SFCT in patients with chronic CHD, which may potentially be triggered by decreased angiogenic factors.
Collapse
Affiliation(s)
- Gurcan Dogukan Arslan
- Department of Ophthalmology, Istanbul Medicine Hospital, Goztepe District, 2366th Street, Bagcilar, 34214, Istanbul, Turkey.
| | - Levent Dogan
- Department of Ophthalmology, Tatvan State Hospital, Bitlis, Turkey
| | - Zeki Dogan
- Department of Cardiology, Istanbul Medicine Hospital, Istanbul, Turkey
| | - Hasan Kiziltoprak
- Department of Ophthalmology, Istanbul Medicine Hospital, Goztepe District, 2366th Street, Bagcilar, 34214, Istanbul, Turkey
| |
Collapse
|
2
|
Alhusban S, Nofal M, Kovacs-Kasa A, Kress TC, Koseoglu MM, Zaied AA, Belin de Chantemele EJ, Annex BH. Glucosamine-Mediated Hexosamine Biosynthesis Pathway Activation Uses ATF4 to Promote "Exercise-Like" Angiogenesis and Perfusion Recovery in PAD. Circulation 2024; 150:1702-1719. [PMID: 39253813 PMCID: PMC11955094 DOI: 10.1161/circulationaha.124.069580] [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: 03/19/2024] [Accepted: 08/06/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND Endothelial cells (ECs) use glycolysis to produce energy. In preclinical models of peripheral arterial disease, further activation of EC glycolysis was ineffective or deleterious in promoting hypoxia-dependent angiogenesis, whereas pentose phosphate pathway activation was effective. Hexosamine biosynthesis pathway, pentose phosphate pathway, and glycolysis are closely linked. Glucosamine directly activates hexosamine biosynthesis pathway. METHODS Hind-limb ischemia in endothelial nitric oxide synthase knockout (eNOS-/-) and BALB/c mice was used. Glucosamine (600 μg/g per day) was injected intraperitoneally. Blood flow recovery was assessed using laser Doppler perfusion imaging and angiogenesis was studied by CD31 immunostaining. In vitro, human umbilical vein ECs and mouse microvascular ECs with glucosamine, L-glucose, or vascular endothelial growth factor (VEGF165a) were tested under hypoxia and serum starvation. Cell Counting Kit-8, tube formation, intracellular reactive oxygen species, electric cell-substrate impedance sensing, and fluorescein isothiocyanate dextran permeability were assessed. Glycolysis and oxidative phosphorylation were assessed by seahorse assay. Gene expression was assessed using RNA sequencing, real-time quantitative polymerase chain reaction, and Western blot. Human muscle biopsies from patients with peripheral arterial disease were assessed for EC O-GlcNAcylation before and after supervised exercise versus standard medical care. RESULTS On day 3 after hind-limb ischemia, glucosamine-treated versus control eNOS-/- mice had less necrosis (n=4 or 5 per group). Beginning on day 7 after hind-limb ischemia, glucosamine-treated versus control BALB/c mice had higher blood flow, which persisted to day 21, when ischemic muscles showed greater CD31 staining per muscle fiber (n=8 per group). In vitro, glucosamine versus L-glucose ECs showed improved survival (n=6 per group) and tube formation (n=6 per group). RNA sequencing of glucosamine versus L-glucose ECs showed increased amino acid metabolism (n=3 per group). That resulted in increased oxidative phosphorylation (n=8-12 per group) and serine biosynthesis pathway without an increase in glycolysis or pentose phosphate pathway genes (n=6 per group). This was associated with better barrier function (n=6-8 per group) and less reactive oxygen species (n=7 or 8 per group) compared with activating glycolysis by VEGF165a. These effects were mediated by activating transcription factor 4, a driver of exercise-induced angiogenesis. In muscle biopsies from humans with peripheral arterial disease, EC/O-GlcNAcylation was increased by 12 weeks of supervised exercise versus standard medical care (n=6 per group). CONCLUSIONS In cells, mice, and humans, activation of hexosamine biosynthesis pathway by glucosamine in peripheral arterial disease induces an "exercise-like" angiogenesis and offers a promising novel therapeutic pathway to treat this challenging disorder.
Collapse
Affiliation(s)
- Suhib Alhusban
- Vascular Biology Center (S.A., M.N., A.K.-K., T.C.K., M.M.K., A.A.Z., E.J.B.d.C., B.H.A.), Medical College of Georgia at Augusta University
| | - Mohamed Nofal
- Vascular Biology Center (S.A., M.N., A.K.-K., T.C.K., M.M.K., A.A.Z., E.J.B.d.C., B.H.A.), Medical College of Georgia at Augusta University
| | - Anita Kovacs-Kasa
- Vascular Biology Center (S.A., M.N., A.K.-K., T.C.K., M.M.K., A.A.Z., E.J.B.d.C., B.H.A.), Medical College of Georgia at Augusta University
| | - Taylor C Kress
- Vascular Biology Center (S.A., M.N., A.K.-K., T.C.K., M.M.K., A.A.Z., E.J.B.d.C., B.H.A.), Medical College of Georgia at Augusta University
| | - M Murat Koseoglu
- Vascular Biology Center (S.A., M.N., A.K.-K., T.C.K., M.M.K., A.A.Z., E.J.B.d.C., B.H.A.), Medical College of Georgia at Augusta University
| | - Abdelrahman A Zaied
- Vascular Biology Center (S.A., M.N., A.K.-K., T.C.K., M.M.K., A.A.Z., E.J.B.d.C., B.H.A.), Medical College of Georgia at Augusta University
- Department of Medicine (A.A.Z., B.H.A.), Medical College of Georgia at Augusta University
| | - Eric J Belin de Chantemele
- Vascular Biology Center (S.A., M.N., A.K.-K., T.C.K., M.M.K., A.A.Z., E.J.B.d.C., B.H.A.), Medical College of Georgia at Augusta University
| | - Brian H Annex
- Vascular Biology Center (S.A., M.N., A.K.-K., T.C.K., M.M.K., A.A.Z., E.J.B.d.C., B.H.A.), Medical College of Georgia at Augusta University
- Department of Medicine (A.A.Z., B.H.A.), Medical College of Georgia at Augusta University
| |
Collapse
|
3
|
Han C, Zhai C, Li A, Ma Y, Hallajzadeh J. Exercise mediates myocardial infarction via non-coding RNAs. Front Cardiovasc Med 2024; 11:1432468. [PMID: 39553846 PMCID: PMC11563808 DOI: 10.3389/fcvm.2024.1432468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/29/2024] [Indexed: 11/19/2024] Open
Abstract
Myocardial infarction (MI), a widespread cardiovascular issue, mainly occurs due to blood clot formation in the coronary arteries, which reduces blood flow to the heart muscle and leads to cell death. Incorporating exercise into a lifestyle can significantly benefit recovery and reduce the risk of future cardiac events for MI patients. Non-coding RNAs (ncRNAs) play various roles in the effects of exercise on myocardial infarction (MI). ncRNAs regulate gene expression, influence cardiac remodeling, angiogenesis, inflammation, oxidative stress, apoptosis, cardioprotection, and cardiac electrophysiology. The expression of specific ncRNAs is altered by exercise, leading to beneficial changes in heart structure, function, and recovery after MI. These ncRNAs modulate molecular pathways that contribute to improved cardiac health, including reducing inflammation, enhancing angiogenesis, promoting cell survival, and mitigating oxidative stress. Furthermore, they are involved in regulating changes in cardiac remodeling, such as hypertrophy and fibrosis, and can influence the electrical properties of the heart, thereby decreasing the risk of arrhythmias. Knowledge on MI has entered a new phase, with investigations of ncRNAs in physical exercise yielding invaluable insights into the impact of this therapeutic modality. This review compiled research on ncRNAs in MI, with an emphasis on their applicability to physical activity.
Collapse
Affiliation(s)
| | - Cuili Zhai
- College of Chinese Martial Arts, Beijing Sport University, Beijing, China
| | - Ailing Li
- City University of Malyasia, Kuala Lumpur, Malaysia
| | - Yongzhi Ma
- Division of Sports Science and Physical Education, Tsinghua University, Beijing, China
| | - Jamal Hallajzadeh
- Department of Biochemistry and Nutrition, Research Center for Evidence-Based Health Management, Maragheh University of Medical Sciences, Maragheh, Iran
| |
Collapse
|
4
|
Wang J, Xiong T, Wu Q, Qin X. Integrated Strategies for Targeting Arteriogenesis and Angiogenesis After Stroke. Transl Stroke Res 2024:10.1007/s12975-024-01291-4. [PMID: 39225878 DOI: 10.1007/s12975-024-01291-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/29/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024]
Abstract
The interdependence between arteriogenesis and angiogenesis is crucial for enhancing perfusion by synchronously improving leptomeningeal collaterals (LMCs) and microvascular networks after stroke. However, current approaches often focus on promoting arteriogenesis and angiogenesis separately, neglecting the potential synergistic benefits of targeting both processes simultaneously. Therefore, it is imperative to consider both arteriogenesis and angiogenesis as integral and complementary strategies for post-stroke revascularization. To gain a deeper understanding of their relationships after stroke and to facilitate the development of targeted revascularization strategies, we compared them based on their timescale, space, and pathophysiology. The temporal differences in the occurrence of arteriogenesis and angiogenesis allow them to restore blood flow at different stages after stroke. The spatial differences in the effects of arteriogenesis and angiogenesis enable them to specifically target the ischemic penumbra and core infarct region. Additionally, the endothelial cell, as the primary effector cell in their pathophysiological processes, is promising target for enhancing both. Therefore, we provide an overview of key signals that regulate endothelium-mediated arteriogenesis and angiogenesis. Finally, we summarize current therapeutic strategies that involve these signals to promote both processes after stroke, with the aim of inspiring future therapeutic advances in revascularization.
Collapse
Affiliation(s)
- Jing Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Taoying Xiong
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Qisi Wu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Xinyue Qin
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
5
|
Falero-Diaz G, Barboza CDA, Vazquez-Padron RI, Velazquez OC, Lassance-Soares RM. Loss of c-Kit in Endothelial Cells Protects against Hindlimb Ischemia. Biomedicines 2024; 12:1358. [PMID: 38927565 PMCID: PMC11201387 DOI: 10.3390/biomedicines12061358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Critical limb ischemia (CLI) is the end stage of peripheral artery disease (PAD), and around 30% of CLI patients are ineligible for current treatments. The angiogenic benefits of c-Kit have been reported in the ischemia scenario; however, the present study demonstrates the effects of specific endothelial c-Kit signaling in arteriogenesis during hindlimb ischemia. METHODS We created conditional knockout mouse models that decrease c-Kit (c-Kit VE-Cadherin CreERT2-c-Kit) or its ligand (SCF VE-Cadherin CreERT2-SCF) specifically in endothelial cells (ECs) after tamoxifen treatment. These mice and a control group (wild-type VE-Cadherin CreERT2-WT) were subjected to hindlimb ischemia or aortic crush to evaluate perfusion/arteriogenesis and endothelial barrier permeability, respectively. RESULTS Our data confirmed the lower gene expression of c-Kit and SCF in the ECs of c-Kit and SCF mice, respectively. In addition, we confirmed the lower percentage of ECs positive for c-Kit in c-Kit mice. Further, we found that c-Kit and SCF mice had better limb perfusion and arteriogenesis compared to WT mice. We also demonstrated that c-Kit and SCF mice had a preserved endothelial barrier after aortic crush compared to WT. CONCLUSIONS Our data demonstrate the deleterious effects of endothelial SCF/c-Kit signaling on arteriogenesis and endothelial barrier integrity.
Collapse
Affiliation(s)
- Gustavo Falero-Diaz
- Department of Surgery, Miller School of Medicine, University of Miami, 1600 NW 10th Ave, RMSB, Miami, FL 33136, USA; (G.F.-D.); (R.I.V.-P.); (O.C.V.)
| | - Catarina de A. Barboza
- Department of Medicine, Miller School of Medicine, University of Miami, 1580 NW 10th Ave, Batchelor Building, Miami, FL 33136, USA;
| | - Roberto I. Vazquez-Padron
- Department of Surgery, Miller School of Medicine, University of Miami, 1600 NW 10th Ave, RMSB, Miami, FL 33136, USA; (G.F.-D.); (R.I.V.-P.); (O.C.V.)
| | - Omaida C. Velazquez
- Department of Surgery, Miller School of Medicine, University of Miami, 1600 NW 10th Ave, RMSB, Miami, FL 33136, USA; (G.F.-D.); (R.I.V.-P.); (O.C.V.)
| | - Roberta M. Lassance-Soares
- Department of Surgery, Miller School of Medicine, University of Miami, 1600 NW 10th Ave, RMSB, Miami, FL 33136, USA; (G.F.-D.); (R.I.V.-P.); (O.C.V.)
| |
Collapse
|
6
|
Basaran MM, Ozgursoy SK, Arslan H, Kocaturk S. The effect of subperichondrial dissection on nasal vascularity in septorhinoplasty operations. Eur Arch Otorhinolaryngol 2024; 281:1827-1833. [PMID: 38052758 DOI: 10.1007/s00405-023-08356-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 11/13/2023] [Indexed: 12/07/2023]
Abstract
PURPOSE Nasal vascularization runs above the superficial musculoaponeurotic system (SMAS). Perichondrium covers the lower and upper lateral cartilages. In this study, nasal vascularization was compared between subperichondrial and supraperichondrial dissection in closed septorhinoplasty. METHODS 95 patients and 41 volunteers were included in this study. Supraperichondrial dissection was performed in 48 patients and subperichondrial dissection was performed in 47 patients. To measure blood stream, laser doppler flowmetry (LDF) was used and measurements were done preoperatively, on the postoperative first week; 3rd month and first year. RESULTS The nasal tip and dorsum measurements were similar between the preoperative and postoperative first year in both groups (p = 1.000). However, in the supraperichondrial dissection group, nasal tip measurements showed a significant increase between the preoperative and third postoperative months (p = 0.011). This increase was accompanied by an increase in the minimal blood stream (p = 0.014). CONCLUSION Both subperichondrial and supraperichondrial dissection techniques are physiological and result in fewer complications with minimal permanent vascular damage. We believe incision plays a critical role but keeping the perichondrium intact is important for short-term angiogenesis, where long-term results showed no difference in vascularization.
Collapse
Affiliation(s)
| | | | - Hande Arslan
- Department of Otorhinolaryngology, Samsun Research and Training Hospital, Samsun, Turkey
| | - Sinan Kocaturk
- Department of Otorhinolaryngology, Losante Hospital, Ankara, Turkey
| |
Collapse
|
7
|
French C, Robbins D, Gernigon M, Gordon D. The effects of lower limb ischaemic preconditioning: a systematic review. Front Physiol 2024; 14:1323310. [PMID: 38274048 PMCID: PMC10808809 DOI: 10.3389/fphys.2023.1323310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 12/28/2023] [Indexed: 01/27/2024] Open
Abstract
Ischaemic preconditioning (IPC) involves the use of repeated occlusions and reperfusions of the peripheral muscle blood supply at a limb. This systematic literature review examines the typical responses in response to the method of application during an IPC applied at the lower limb. This review focuses on the physiological responses for VO2max, haemoglobin, metabolic and genetic responses to various IPC interventions. The literature search was performed using four databases and assessed using the PRISMA search strategy and COSMIN to assess the quality of the articles. Seventeen articles were included in the review, with a total of 237 participants. While there is variation in the method of application, the average occlusion pressure was 222 ± 34 mmHg, ranging from 170 to 300 mmHg typically for 3 or 4 occlusion cycles. The distribution of this pressure is influenced by cuff width, although 8 studies failed to report cuff width. The majority of studies applies IPC at the proximal thigh with 16/17 studies applying an occlusion below this location. The results highlighted the disparities and conflicting findings in response to various IPC methods. While there is some agreement in certain aspects of the IPC manoeuvre such as the location of the occlusion during lower limb IPC, there is a lack of consensus in the optimal protocol to elicit the desired responses. This offers the opportunity for future research to refine the protocols, associated responses, and mechanisms responsible for these changes during the application of IPC.
Collapse
Affiliation(s)
- Chloe French
- Cambridge Centre for Sport and Exercise Sciences (CCSES), Faculty of Science and Engineering, Anglia Ruskin University, Cambridge, United Kingdom
- CIAMS, Université Paris-Saclay, Orsay Cedex, France
- CIAMS, Université d’Orléans, Orléans, France
| | - Dan Robbins
- Medical Technology Research Centre, Faculty of Health, Education, Medicine and Social Care, Anglia Ruskin University, Chelmsford, United Kingdom
| | - Marie Gernigon
- CIAMS, Université Paris-Saclay, Orsay Cedex, France
- CIAMS, Université d’Orléans, Orléans, France
| | - Dan Gordon
- Cambridge Centre for Sport and Exercise Sciences (CCSES), Faculty of Science and Engineering, Anglia Ruskin University, Cambridge, United Kingdom
| |
Collapse
|
8
|
Kumaraswami K, Arnholdt C, Deindl E, Lasch M. Rag1 Deficiency Impairs Arteriogenesis in Mice. Int J Mol Sci 2023; 24:12839. [PMID: 37629019 PMCID: PMC10454224 DOI: 10.3390/ijms241612839] [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: 07/05/2023] [Revised: 08/12/2023] [Accepted: 08/13/2023] [Indexed: 08/27/2023] Open
Abstract
Increasing evidence suggests that lymphocytes play distinct roles in inflammation-induced tissue remodeling and tissue damage. Arteriogenesis describes the growth of natural bypasses from pre-existing collateral arteries. This process compensates for the loss of artery function in occlusive arterial diseases. The role of innate immune cells is widely understood in the process of arteriogenesis, whereas the role of lymphocytes remains unclear and is the subject of the present study. To analyze the role of lymphocytes, we induced arteriogenesis in recombination activating gene-1 (Rag1) knockout (KO) mice by unilateral ligation of the femoral artery. The lack of functional lymphocytes in Rag1 KO mice resulted in reduced perfusion recovery as shown by laser Doppler imaging. Additionally, immunofluorescence staining revealed a reduced vascular cell proliferation along with a smaller inner luminal diameter in Rag1 KO mice. The perivascular macrophage polarization around the growing collateral arteries was shifted to more pro-inflammatory M1-like polarized macrophages. Together, these data suggest that lymphocytes are crucial for arteriogenesis by modulating perivascular macrophage polarization.
Collapse
Affiliation(s)
- Konda Kumaraswami
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (K.K.); (C.A.); (M.L.)
- Medical Clinic I, Department of Cardiology, University Hospital, Ludwig Maximilian University, 81377 Munich, Germany
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Christoph Arnholdt
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (K.K.); (C.A.); (M.L.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Elisabeth Deindl
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (K.K.); (C.A.); (M.L.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Manuel Lasch
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (K.K.); (C.A.); (M.L.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital, Ludwig Maximilian University, 81377 Munich, Germany
| |
Collapse
|
9
|
Apeldoorn C, Safaei S, Paton J, Maso Talou GD. Computational models for generating microvascular structures: Investigations beyond medical imaging resolution. WIREs Mech Dis 2023; 15:e1579. [PMID: 35880683 PMCID: PMC10077909 DOI: 10.1002/wsbm.1579] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 06/22/2022] [Accepted: 06/29/2022] [Indexed: 01/31/2023]
Abstract
Angiogenesis, arteriogenesis, and pruning are revascularization processes essential to our natural vascular development and adaptation, as well as central players in the onset and development of pathologies such as tumoral growth and stroke recovery. Computational modeling allows for repeatable experimentation and exploration of these complex biological processes. In this review, we provide an introduction to the biological understanding of the vascular adaptation processes of sprouting angiogenesis, intussusceptive angiogenesis, anastomosis, pruning, and arteriogenesis, discussing some of the more significant contributions made to the computational modeling of these processes. Each computational model represents a theoretical framework for how biology functions, and with rises in computing power and study of the problem these frameworks become more accurate and complete. We highlight physiological, pathological, and technological applications that can be benefit from the advances performed by these models, and we also identify which elements of the biology are underexplored in the current state-of-the-art computational models. This article is categorized under: Cancer > Computational Models Cardiovascular Diseases > Computational Models.
Collapse
Affiliation(s)
- Cameron Apeldoorn
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Soroush Safaei
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Julian Paton
- Cardiovascular Autonomic Research Cluster, Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Gonzalo D Maso Talou
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
10
|
Wang H, Dong D, Chen L, Yuan M, Liu C, Liu T. Methylene blue and carbon nanoparticles had no significant adverse effects on the survival and function of lymph nodes after non-vascularized transplantation in a mouse model. Transpl Immunol 2022; 72:101565. [DOI: 10.1016/j.trim.2022.101565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 02/27/2022] [Accepted: 02/27/2022] [Indexed: 11/26/2022]
|
11
|
Balakrishnan S, Senthil Kumar B. Correlation of serum Vascular Endothelial growth factor (VEGF) and cardiovascular risk factors on collateral formation in patients with acute coronary artery syndrome. Clin Anat 2022; 35:673-678. [PMID: 35451175 DOI: 10.1002/ca.23890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/11/2022] [Accepted: 04/18/2022] [Indexed: 11/10/2022]
Abstract
Coronary collaterals serve as an alternative source of blood flow in obstructive coronary heart disease. Coronary collateral development by releasing various angiogenic growth factors, including vascular endothelial growth factor-A (VEGF-A). Cardiovascular risk factors strongly associated with coronary artery disease include age, sex, elevated serum cholesterol, disturbed carbohydrate metabolism, and elevated blood pressure. A better understanding of the effects of these cardiovascular risk factors and serum VEGF-A level on collateral recruitment is necessary for a better prognosis in coronary artery diseases and new insight for further therapeutic promotion of coronary collaterals.220 consecutive patients undergoing coronary angiography with a mean age of 61 ± 9.83 were selected for the analysis. Two millilitres of blood were taken from the patients for analysis. The blood serum VEGF concentration was quantified via the ELISA method. Angiograms and other clinical reports were collected. Significant coronary artery disease was diagnosed in those with ≥ 70% of stenosis in at least one of the coronary arteries. The angiographic and clinical data were documented. The collateral grading was done according to the Rentrop Scoring system. The serum vascular endothelial growth factor level was correlated with the collateral score and cardiovascular risk factors like age, sex, type ll diabetes, blood pressure, and cholesterol level. An increase in the level of the collateral score was noted with an increase in the level of VEGF in blood serum. A significant association was founded between serum VEGF level and cardiovascular risk factors on collateral formation in patients with diabetes and hypertension. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Sheeja Balakrishnan
- Department of Anatomy, Government Medical College (Institute of Integrated Medical Sciences), Palakkad, Kerala, India
| | - B Senthil Kumar
- Department of Anatomy Vinayaka Mission's Kirupananda Variyar Medical College, Vinayaka Missions Research Foundation (DU)Salem-636308, Tamilnadu, India
| |
Collapse
|
12
|
Spadaccio C, Nenna A, Rose D, Piccirillo F, Nusca A, Grigioni F, Chello M, Vlahakes GJ. The Role of Angiogenesis and Arteriogenesisin Myocardial Infarction and Coronary Revascularization. J Cardiovasc Transl Res 2022; 15:1024-1048. [PMID: 35357670 DOI: 10.1007/s12265-022-10241-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 03/18/2022] [Indexed: 12/25/2022]
Abstract
Surgical myocardial revascularization is associated with long-term survival benefit in patients with multivessel coronary artery disease. However, the exact biological mechanisms underlying the clinical benefits of myocardial revascularization have not been elucidated yet. Angiogenesis and arteriogenesis biologically leading to vascular collateralization are considered one of the endogenous mechanisms to preserve myocardial viability during ischemia, and the presence of coronary collateralization has been regarded as one of the predictors of long-term survival in patients with coronary artery disease (CAD). Some experimental studies and indirect clinical evidence on chronic CAD confirmed an angiogenetic response induced by myocardial revascularization and suggested that revascularization procedures could constitute an angiogenetic trigger per se. In this review, the clinical and basic science evidence regarding arteriogenesis and angiogenesis in both CAD and coronary revascularization is analyzed with the aim to better elucidate their significance in the clinical arena and potential therapeutic use.
Collapse
Affiliation(s)
- Cristiano Spadaccio
- Cardiac Surgery, Massachusetts General Hospital & Harvard Medical School, Boston, USA. .,Cardiac Surgery, Golden Jubilee National Hospital & University of Glasgow, Glasgow, UK.
| | - Antonio Nenna
- Cardiac Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - David Rose
- Cardiac Surgery, Lancashire Cardiac Centre, Blackpool Victoria Hospital, Blackpool, UK
| | | | | | | | - Massimo Chello
- Cardiac Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Gus J Vlahakes
- Cardiac Surgery, Massachusetts General Hospital & Harvard Medical School, Boston, USA
| |
Collapse
|
13
|
Zhao G, Feng Y, Xue L, Cui M, Zhang Q, Xu F, Peng N, Jiang Z, Gao D, Zhang X. Anisotropic conductive reduced graphene oxide/silk matrices promote post-infarction myocardial function by restoring electrical integrity. Acta Biomater 2022; 139:190-203. [PMID: 33836222 DOI: 10.1016/j.actbio.2021.03.073] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 02/07/2023]
Abstract
Myocardial infarction (MI) remains the leading cause of death globally, often leading to impaired cardiac function and pathological myocardial microenvironment. Electrical conduction abnormalities of the infarcted myocardium not only induce adverse myocardial remodeling but also prevent tissue repair. Restoring the myocardial electrical integrity, particularly the anisotropic electrical signal propagation within the injured area after infarction is crucial for an effective function recovery. Herein, optimized reduced graphene oxide (rGO) functionalized electrospun silk fibroin (rGO/silk) biomaterials presenting anisotropic conductivity and enhanced suturablity were developed and investigated as cardiac patches for their potential in improving the post-MI myocardial function of rat models. The results show that the anisotropic conductive rGO/silk patches exhibit remarkable therapeutic effect on repairing the infarcted myocardium compared to the nonconductive silk and isotropic conductive rGO/silk patches as determined by the enhanced pumping function, reduced susceptibility to arrhythmias, thickened left ventricular walls and improved survival of functional cardiomyocytes. Their notable effect on promoting the angiogenesis of capillaries in the infarcted myocardium has also been demonstrated. This study highlights an effective and biomimetic reconstruction of the electrical myocardial microenvironment based on the anisotropic conductive rGO/silk biomaterials as a promising option for promoting the repair of infarcted myocardium. STATEMENT OF SIGNIFICANCE: The dysfunctional electrical microenvironment in the infarcted myocardium not only aggravates the adverse myocardial remodeling but also limits the effect of cardiac regenerative medicine. Although various conductive biomaterials have been employed to restore the electrical network in the infarcted myocardium in vivo, the anisotropic nature of the myocardial electrical microenvironment which enables directional electrical signal propagation were neglected. In this study, an anisotropic conductive rGO/silk biomaterial system is developed to improve the myocardial function post infarction by restoring the anisotropic electrical microenvironment in the infarcted myocardium. The promoted effects of anisotropic conductive grafts on repairing infarcted hearts are demonstrated with improved pumping function, cardiomyocyte survival, resistance to ventricular fibrillation, and angiogenesis of capillary network.
Collapse
Affiliation(s)
- Guoxu Zhao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Center for Mitochondrial Biology and Medicine, School of Life Science and Technology, International Joint Laboratory for Micro/Nano Manufacturing and Measurement Technology, Xi'an Key Laboratory for Biomedical Testing and High-end Equipment, Xi'an Jiaotong University, Xi'an 710049, Shannxi, PR China; School of Material Science and Chemical Engineering, Xi'an Technological University, Xi'an 710021, Shaanxi, PR China
| | - Yanjing Feng
- Department of Cardiology, The Second Affiliated Hospital, School of Medical, Xi'an Jiaotong University, Xi'an 710004, Shaanxi, PR China
| | - Li Xue
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Center for Mitochondrial Biology and Medicine, School of Life Science and Technology, International Joint Laboratory for Micro/Nano Manufacturing and Measurement Technology, Xi'an Key Laboratory for Biomedical Testing and High-end Equipment, Xi'an Jiaotong University, Xi'an 710049, Shannxi, PR China
| | - Mengjie Cui
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Center for Mitochondrial Biology and Medicine, School of Life Science and Technology, International Joint Laboratory for Micro/Nano Manufacturing and Measurement Technology, Xi'an Key Laboratory for Biomedical Testing and High-end Equipment, Xi'an Jiaotong University, Xi'an 710049, Shannxi, PR China
| | - Qi Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Center for Mitochondrial Biology and Medicine, School of Life Science and Technology, International Joint Laboratory for Micro/Nano Manufacturing and Measurement Technology, Xi'an Key Laboratory for Biomedical Testing and High-end Equipment, Xi'an Jiaotong University, Xi'an 710049, Shannxi, PR China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Bioinspired Engineering and Biomechanics Center (BEBC), School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, PR China
| | - Niancai Peng
- International Joint Laboratory for Micro/Nano Manufacturing and Measurement Technology, State Key Laboratory for Manufacturing Systems Engineering, Xi'an Key Laboratory for Biomedical Testing and High-end Equipment, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, PR China
| | - Zhuangde Jiang
- International Joint Laboratory for Micro/Nano Manufacturing and Measurement Technology, State Key Laboratory for Manufacturing Systems Engineering, Xi'an Key Laboratory for Biomedical Testing and High-end Equipment, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, PR China
| | - Dengfeng Gao
- Department of Cardiology, The Second Affiliated Hospital, School of Medical, Xi'an Jiaotong University, Xi'an 710004, Shaanxi, PR China.
| | - Xiaohui Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Center for Mitochondrial Biology and Medicine, School of Life Science and Technology, International Joint Laboratory for Micro/Nano Manufacturing and Measurement Technology, Xi'an Key Laboratory for Biomedical Testing and High-end Equipment, Xi'an Jiaotong University, Xi'an 710049, Shannxi, PR China.
| |
Collapse
|
14
|
Bhalla SR, Riu F, Machado MJC, Bates DO. Measurement of Revascularization in the Hind Limb After Experimental Ischemia in Mice. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2441:105-113. [PMID: 35099732 DOI: 10.1007/978-1-0716-2059-5_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Peripheral vascular disease is a major cause of morbidity and mortality, and is a consequence of impaired blood flow to the limbs. This arises due to the inability of the tissue to develop sufficiently functional collateral vessel circulation to overcome occluded arteries, or microvascular impairment. The mouse hind limb model of hind limb ischemia can be used to investigate the impact of different treatment modalities, behavioral changes, or genetic knockout. Here we described the model in detail, providing examples of adverse events, and details of ex vivo analysis of blood vessel density.
Collapse
Affiliation(s)
- Sohni Ria Bhalla
- Tumour and Vascular Biology Laboratories, Centre for Cancer Sciences, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Federica Riu
- Tumour and Vascular Biology Laboratories, Centre for Cancer Sciences, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Maria J C Machado
- Tumour and Vascular Biology Laboratories, Centre for Cancer Sciences, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - David O Bates
- Tumour and Vascular Biology Laboratories, Centre for Cancer Sciences, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK.
| |
Collapse
|
15
|
Zhang Y, Zhang Y, Wang Y, Xu X, Jin J, Zhang X, Zhang W, Wei W, Zhong C, Wu G. Effects of Enhanced External Counterpulsation With Different Sequential Levels on Lower Extremity Hemodynamics. Front Cardiovasc Med 2022; 8:795697. [PMID: 35004907 PMCID: PMC8739776 DOI: 10.3389/fcvm.2021.795697] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 11/29/2021] [Indexed: 11/23/2022] Open
Abstract
Objective: This study aimed to investigate acute hemodynamics of lower extremities during enhanced external counterpulsation with a three-level sequence at the hips, thighs, and calves (EECP-3), two-level sequence at the hips and thighs (EECP-2), and single leg three-level sequence (EECP-1). Methods: Twenty healthy volunteers were recruited in this study to receive a 45-min EECP intervention. Blood flow spectrums in the anterior tibial artery, posterior tibial artery, and dorsalis pedis artery were imaged by Color Doppler ultrasound. Mean flow rate (FR), area, pulsatility index (PI), peak systolic velocity (PSV), end-diastolic velocity (EDV), mean flow velocity (MV), and systolic maximum acceleration (CCAs) were sequentially measured and calculated at baseline during EECP-3, EECP-1, and EECP-2. Results: During EECP-3, PI, PSV, and MV in the anterior tibial artery were significantly higher, while EDV was markedly lower during EECP-1, EECP-2, and baseline (all P < 0.05). Additionally, ACCs were significantly elevated during EECP-3 compared with baseline. Moreover, FR in the anterior tibial artery was significantly increased during EECP-3 compared with baseline (P = 0.048). During EECP-2, PI and MV in the dorsalis pedis artery were significantly higher and lower than those at baseline, (both P < 0.05). In addition, FR was markedly reduced during EECP-2 compared with baseline (P = 0.028). During EECP-1, the area was significantly lower, while EDV was markedly higher in the posterior tibial artery than during EECP-1, EECP-2, and baseline (all P < 0.05). Meanwhile, FR of the posterior tibial artery was significantly reduced compared with baseline (P = 0.014). Conclusion: Enhanced external counterpulsation with three-level sequence (EECP-3), EECP-2, and EECP-1 induced different hemodynamic responses in the anterior tibial artery, dorsalis pedis artery, and posterior tibial artery, respectively. EECP-3 acutely improved the blood flow, blood flow velocity, and ACCs of the anterior tibial artery. In addition, EECP-1 and EECP-2 significantly increased the blood flow velocity and peripheral resistance of the inferior knee artery, whereas they markedly reduced blood flow in the posterior tibial artery.
Collapse
Affiliation(s)
- Yahui Zhang
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China.,Guangdong Innovative Engineering and Technology Research Center for Assisted Circulation, Shenzhen, China
| | - Yujia Zhang
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China.,Guangdong Innovative Engineering and Technology Research Center for Assisted Circulation, Shenzhen, China
| | - Yinfen Wang
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xiuli Xu
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China.,Guangdong Innovative Engineering and Technology Research Center for Assisted Circulation, Shenzhen, China
| | - Jing Jin
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xiaodong Zhang
- Department of Physical Education, Nanjing University of Finance and Economics, Nanjing, China
| | - Wei Zhang
- College of Computer, Jilin Normal University, Siping, China
| | - Wenbin Wei
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Chubin Zhong
- Department of Cardiac Ultrasound, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Guifu Wu
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China.,Guangdong Innovative Engineering and Technology Research Center for Assisted Circulation, Shenzhen, China
| |
Collapse
|
16
|
McEnaney RM, McCreary DD, Skirtich NO, Andraska EA, Sachdev U, Tzeng E. Elastic Laminar Reorganization Occurs with Outward Diameter Expansion during Collateral Artery Growth and Requires Lysyl Oxidase for Stabilization. Cells 2021; 11:7. [PMID: 35011567 PMCID: PMC8750335 DOI: 10.3390/cells11010007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/14/2021] [Accepted: 12/17/2021] [Indexed: 01/16/2023] Open
Abstract
When a large artery becomes occluded, hemodynamic changes stimulate remodeling of arterial networks to form collateral arteries in a process termed arteriogenesis. However, the structural changes necessary for collateral remodeling have not been defined. We hypothesize that deconstruction of the extracellular matrix is essential to remodel smaller arteries into effective collaterals. Using multiphoton microscopy, we analyzed collagen and elastin structure in maturing collateral arteries isolated from ischemic rat hindlimbs. Collateral arteries harvested at different timepoints showed progressive diameter expansion associated with striking rearrangement of internal elastic lamina (IEL) into a loose fibrous mesh, a pattern persisting at 8 weeks. Despite a 2.5-fold increase in luminal diameter, total elastin content remained unchanged in collaterals compared with control arteries. Among the collateral midzones, baseline elastic fiber content was low. Outward remodeling of these vessels with a 10-20 fold diameter increase was associated with fractures of the elastic fibers and evidence of increased wall tension, as demonstrated by the straightening of the adventitial collagen. Inhibition of lysyl oxidase (LOX) function with β-aminopropionitrile resulted in severe fragmentation or complete loss of continuity of the IEL in developing collaterals. Collateral artery development is associated with permanent redistribution of existing elastic fibers to accommodate diameter growth. We found no evidence of new elastic fiber formation. Stabilization of the arterial wall during outward remodeling is necessary and dependent on LOX activity.
Collapse
Affiliation(s)
- Ryan M. McEnaney
- VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA; (D.D.M.); (N.O.S.); (E.T.)
- Division of Vascular Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (E.A.A.); (U.S.)
| | - Dylan D. McCreary
- VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA; (D.D.M.); (N.O.S.); (E.T.)
| | - Nolan O. Skirtich
- VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA; (D.D.M.); (N.O.S.); (E.T.)
| | - Elizabeth A. Andraska
- Division of Vascular Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (E.A.A.); (U.S.)
| | - Ulka Sachdev
- Division of Vascular Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (E.A.A.); (U.S.)
| | - Edith Tzeng
- VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA; (D.D.M.); (N.O.S.); (E.T.)
- Division of Vascular Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (E.A.A.); (U.S.)
| |
Collapse
|
17
|
de Bortoli T, Boehm-Sturm P, Koch SP, Nieminen-Kelhä M, Wessels L, Mueller S, Ielacqua GD, Klohs J, Vajkoczy P, Hecht N. Three-Dimensional Iron Oxide Nanoparticle-Based Contrast-Enhanced Magnetic Resonance Imaging for Characterization of Cerebral Arteriogenesis in the Mouse Neocortex. Front Neurosci 2021; 15:756577. [PMID: 34899163 PMCID: PMC8662986 DOI: 10.3389/fnins.2021.756577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 11/02/2021] [Indexed: 11/13/2022] Open
Abstract
Purpose: Subsurface blood vessels in the cerebral cortex have been identified as a bottleneck in cerebral perfusion with the potential for collateral remodeling. However, valid techniques for non-invasive, longitudinal characterization of neocortical microvessels are still lacking. In this study, we validated contrast-enhanced magnetic resonance imaging (CE-MRI) for in vivo characterization of vascular changes in a model of spontaneous collateral outgrowth following chronic cerebral hypoperfusion. Methods: C57BL/6J mice were randomly assigned to unilateral internal carotid artery occlusion or sham surgery and after 21 days, CE-MRI based on T2*-weighted imaging was performed using ultra-small superparamagnetic iron oxide nanoparticles to obtain subtraction angiographies and steady-state cerebral blood volume (ss-CBV) maps. First pass dynamic susceptibility contrast MRI (DSC-MRI) was performed for internal validation of ss-CBV. Further validation at the histological level was provided by ex vivo serial two-photon tomography (STP). Results: Qualitatively, an increase in vessel density was observed on CE-MRI subtraction angiographies following occlusion; however, a quantitative vessel tracing analysis was prone to errors in our model. Measurements of ss-CBV reliably identified an increase in cortical vasculature, validated by DSC-MRI and STP. Conclusion: Iron oxide nanoparticle-based ss-CBV serves as a robust, non-invasive imaging surrogate marker for neocortical vessels, with the potential to reduce and refine preclinical models targeting the development and outgrowth of cerebral collateralization.
Collapse
Affiliation(s)
- Till de Bortoli
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Center for Stroke Research Berlin (CSB), Berlin, Germany
| | - Philipp Boehm-Sturm
- Center for Stroke Research Berlin (CSB), Berlin, Germany.,Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Stefan P Koch
- Center for Stroke Research Berlin (CSB), Berlin, Germany.,Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Melina Nieminen-Kelhä
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Center for Stroke Research Berlin (CSB), Berlin, Germany
| | - Lars Wessels
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Center for Stroke Research Berlin (CSB), Berlin, Germany
| | - Susanne Mueller
- Center for Stroke Research Berlin (CSB), Berlin, Germany.,Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Giovanna D Ielacqua
- Institute for Biomedical Engineering, University of Zurich and ETH Zürich, Zurich, Switzerland
| | - Jan Klohs
- Institute for Biomedical Engineering, University of Zurich and ETH Zürich, Zurich, Switzerland
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Center for Stroke Research Berlin (CSB), Berlin, Germany
| | - Nils Hecht
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Center for Stroke Research Berlin (CSB), Berlin, Germany
| |
Collapse
|
18
|
Zhang Y, Chen Z, Mai Z, Zhou W, Wang H, Zhang X, Wei W, Du J, Wu G. Acute Hemodynamic Responses to Enhanced External Counterpulsation in Patients With Coronary Artery Disease. Front Cardiovasc Med 2021; 8:721140. [PMID: 34869627 PMCID: PMC8632772 DOI: 10.3389/fcvm.2021.721140] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 10/12/2021] [Indexed: 11/16/2022] Open
Abstract
Purpose: Enhanced external counterpulsation is a non-invasive treatment that increases coronary flow in patients with coronary artery disease (CAD). However, the acute responses of vascular and blood flow characteristics in the conduit arteries during and immediately after enhanced external counterpulsation (EECP) need to be verified. Methods: Forty-two patients with CAD and 21 healthy controls were recruited into this study to receive 45 min-EECP. Both common carotid arteries (CCAs), namely, the left carotid (LC) and right carotid (RC), the right brachial (RB), and right femoral (RF) artery were imaged using a Color Doppler ultrasound. The peak systolic velocity (PSV), end-diastolic velocity (EDV), mean inner diameter (ID), resistance index (RI), and mean flow rate (FR) were measured and calculated before, during, and after the 45 min-EECP treatment. Results: During EECP, in the CCAs, the EDV was significantly decreased, while the RI was markedly increased in the two groups (both P < 0.01). However, immediately after EECP, the RI in the RC was significantly lower than that at the baseline in the patients with CAD (P = 0.039). The FR of the LC was markedly increased during EECP only in the CAD patients (P = 0.004). The PSV of the patients with CAD was also significantly reduced during EECP (P = 0.015) and immediately after EECP (P = 0.005) compared with the baseline. Moreover, the ID of the LC, RB, and RF was significantly higher immediately after EECP than that at the baseline (all P < 0.05) in the patients with CAD. In addition, they were also higher than that in the control groups (all P < 0.05). Furthermore, by the subgroup analysis, there were significant differences in the FR, PSV, and RI between females and males during and immediately after EECP (all P < 0.05). Conclusions: Enhanced external counterpulsation creates different responses of vascular and blood flow characteristics in carotid and peripheral arteries, with more significant effects in both the carotid arteries. Additionally, the beneficial effects in ID, blood flow velocity, RI, and FR after 45 min-EECP were shown only in the patients with CAD. More importantly, acute improvement of EECP in the FR of the brachial artery was showed in males, while the FR and RI of the carotid arteries changed in females.
Collapse
Affiliation(s)
- Yahui Zhang
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,National Health Commission (NHC) Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China.,Guangdong Innovative Engineering and Technology Research Center for Assisted Circulation, Shenzhen, China
| | - Ziqi Chen
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,National Health Commission (NHC) Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Zhouming Mai
- Department of Cardiology, Huizhou Third People's Hospital, Huizhou, China
| | - Wenjuan Zhou
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Hui Wang
- Department of Cardiac Ultrasound, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xiaodong Zhang
- Department of Physical Education, Nanjing University of Finance and Economics, Nanjing, China
| | - Wenbin Wei
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jianhang Du
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,National Health Commission (NHC) Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China.,Guangdong Innovative Engineering and Technology Research Center for Assisted Circulation, Shenzhen, China
| | - Guifu Wu
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,National Health Commission (NHC) Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China.,Guangdong Innovative Engineering and Technology Research Center for Assisted Circulation, Shenzhen, China
| |
Collapse
|
19
|
Human Induced Pluripotent Stem Cell-Derived Vascular Cells: Recent Progress and Future Directions. J Cardiovasc Dev Dis 2021; 8:jcdd8110148. [PMID: 34821701 PMCID: PMC8622843 DOI: 10.3390/jcdd8110148] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 12/12/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) hold great promise for cardiovascular regeneration following ischemic injury. Considerable effort has been made toward the development and optimization of methods to differentiate hiPSCs into vascular cells, such as endothelial and smooth muscle cells (ECs and SMCs). In particular, hiPSC-derived ECs have shown robust potential for promoting neovascularization in animal models of cardiovascular diseases, potentially achieving significant and sustained therapeutic benefits. However, the use of hiPSC-derived SMCs that possess high therapeutic relevance is a relatively new area of investigation, still in the earlier investigational stages. In this review, we first discuss different methodologies to derive vascular cells from hiPSCs with a particular emphasis on the role of key developmental signals. Furthermore, we propose a standardized framework for assessing and defining the EC and SMC identity that might be suitable for inducing tissue repair and regeneration. We then highlight the regenerative effects of hiPSC-derived vascular cells on animal models of myocardial infarction and hindlimb ischemia. Finally, we address several obstacles that need to be overcome to fully implement the use of hiPSC-derived vascular cells for clinical application.
Collapse
|
20
|
Blanco PJ, Bulant CA, Ares GD, Lemos PA, Feijóo RA. A simple coronary blood flow model to study the collateral flow index. Biomech Model Mechanobiol 2021; 20:1365-1382. [PMID: 33772676 DOI: 10.1007/s10237-021-01449-1] [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: 11/12/2020] [Accepted: 03/06/2021] [Indexed: 11/25/2022]
Abstract
In this work, we present a novel modeling framework to investigate the effects of collateral circulation into the coronary blood flow physiology. A prototypical model of the coronary tree, integrated with the concept of Collateral Flow Index (CFI), is employed to gain insight about the role of model parameters associated with the collateral circuitry, which results in physically-realizable solutions for specific CFI data. Then, we discuss the mathematical feasibility of pressure-derived CFI, anatomical implications and practical considerations involving the estimation of model parameters in collateral connections. A sensitivity analysis is carried out, and the investigation of the impact of the collateral circulation on FFR values is also addressed.
Collapse
Affiliation(s)
- Pablo J Blanco
- Laboratório Nacional de Computação Científica, Av. Getúlio Vargas 333, Petrópolis, 25651-075, Brazil.
- National Institute of Science and Technology in Medicine Assisted by Scientific Computing, INCT-MACC, Petrópolis, Brazil.
| | - Carlos A Bulant
- National University of the Center and National Scientific and Technical Research Council, CONICET, Tandil, Argentina
- National Institute of Science and Technology in Medicine Assisted by Scientific Computing, INCT-MACC, Petrópolis, Brazil
| | - Gonzalo D Ares
- National University of Mar del Plata, Mar del Plata, Argentina
| | - Pedro A Lemos
- Hospital Israelita Albert Einstein., São Paulo, Brazil
- National Institute of Science and Technology in Medicine Assisted by Scientific Computing, INCT-MACC, Petrópolis, Brazil
| | - Raúl A Feijóo
- Laboratório Nacional de Computação Científica, Av. Getúlio Vargas 333, Petrópolis, 25651-075, Brazil
- National Institute of Science and Technology in Medicine Assisted by Scientific Computing, INCT-MACC, Petrópolis, Brazil
| |
Collapse
|
21
|
Ferreira AK, Cristofaro B, Menezes MC, de Oliveira AK, Tashima AK, de Melo RL, Silva CCF, Rodriguez MGP, Carvalho DCDOS, de Azevedo RA, Junior PLDS, Mambelli LI, Portaro FV, Pardanaud L, Eichmann A, Sant'Anna OA, Faria M. Alphastatin-C a new inhibitor of endothelial cell activation is a pro-arteriogenic agent in vivo and retards B16-F10 melanoma growth in a preclinical model. Oncotarget 2020; 11:4770-4787. [PMID: 33473260 PMCID: PMC7771711 DOI: 10.18632/oncotarget.27839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 06/23/2018] [Indexed: 12/04/2022] Open
Abstract
Most characterized angiogenic modulators are proteolytic fragments of structural plasma and/or matrix components. Herein, we have identified a novel anti-angiogenic peptide generated by the in vitro hydrolysis of the C-terminal moiety of the fibrinogen alpha chain, produced by the snake venom metalloprotease bothropasin (SVMP), a hemorrhagic proteinase in Bothrops jararaca venom. The 14-amino acids peptide (alphastatin-C) is a potent antagonist of basic fibroblast growth factor, induced endothelial cell (HUVEC-CS) proliferation, migration and capillary tube formation in matrigel. It also inhibits cell adhesion to fibronectin. The basis of the antagonism between bFGF and alphastatin-C is elucidated by the inhibition of various bFGF induced signaling pathways and their molecular components modification, whenever the combination of the stimuli is provided, in comparison to the treatment with bFGF only. To corroborate to the potential therapeutic use of alphastatin-C, we have chosen to perform in vivo assays in two distinct angiogenic settings. In chick model, alphastatin-C inhibits chorioallantoic membrane angiogenesis. In mouse, it efficiently reduces tumor number and volume in a melanoma model, due to the impairment of tumor neovascularization in treated mice. In contrast, we show that the alphastatin-C peptide induces arteriogenesis, increasing pial collateral density in neonate mice. alphastatin-C is an efficient new antiangiogenic FGF-associated agent in vitro, it is an inhibitor of embryonic and tumor vascularization in vivo while, it is an arteriogenic agent. The results also suggest that SVMPs can be used as in vitro biochemical tools to process plasma and/or matrix macromolecular components unraveling new angiostatic peptides.
Collapse
Affiliation(s)
- Adilson Kleber Ferreira
- Department of Immunology, Laboratory of Tumor Immunology, Institute of Biomedical Science, University of Sao Paulo, Sao Paulo, SP, Brazil.,Alchemypet, Veterinary Dignostic Medicine, CIETEC/IPEN, Department of Oncology, University of Sao Paulo, Sao Paulo, Brazil
| | - Brunella Cristofaro
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, Paris, France
| | - Milene Cristina Menezes
- Special Laboratory of Applied Toxinology, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Butantan Institute, São Paulo, SP, Brazil
| | - Ana Karina de Oliveira
- Special Laboratory of Applied Toxinology, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Butantan Institute, São Paulo, SP, Brazil
| | - Alexandre Keiji Tashima
- Special Laboratory of Applied Toxinology, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Butantan Institute, São Paulo, SP, Brazil.,Department of Biochemistry, Escola Paulista de Medicina, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Robson Lopes de Melo
- Special Laboratory of Applied Toxinology, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Butantan Institute, São Paulo, SP, Brazil
| | | | | | | | | | | | - Lisley Inata Mambelli
- Department of Immunology, Laboratory of Tumor Immunology, Institute of Biomedical Science, University of Sao Paulo, Sao Paulo, SP, Brazil
| | | | - Luc Pardanaud
- Cardiovascular Research Center and the Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.,INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | - Anne Eichmann
- Cardiovascular Research Center and the Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.,INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | - Osvaldo Augusto Sant'Anna
- Special Laboratory of Applied Toxinology, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Butantan Institute, São Paulo, SP, Brazil
| | - Mxarcella Faria
- Special Laboratory of Applied Toxinology, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Butantan Institute, São Paulo, SP, Brazil
| |
Collapse
|
22
|
Recruitment and maturation of the coronary collateral circulation: Current understanding and perspectives in arteriogenesis. Microvasc Res 2020; 132:104058. [PMID: 32798552 DOI: 10.1016/j.mvr.2020.104058] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 06/09/2020] [Accepted: 08/11/2020] [Indexed: 12/13/2022]
Abstract
The coronary collateral circulation is a rich anastomotic network of primitive vessels which have the ability to augment in size and function through the process of arteriogenesis. In this review, we evaluate the current understandings of the molecular and cellular mechanisms by which this process occurs, specifically focussing on elevated fluid shear stress (FSS), inflammation, the redox state and gene expression along with the integrative, parallel and simultaneous process by which this occurs. The initiating step of arteriogenesis occurs following occlusion of an epicardial coronary artery, with an increase in FSS detected by mechanoreceptors within the endothelium. This must occur within a 'redox window' where an equilibrium of oxidative and reductive factors are present. These factors initially result in an inflammatory milieu, mediated by neutrophils as well as lymphocytes, with resultant activation of a number of downstream molecular pathways resulting in increased expression of proteins involved in monocyte attraction and adherence; namely vascular cell adhesion molecule 1 (VCAM-1), monocyte chemoattractant protein 1 (MCP-1) and transforming growth factor beta (TGF-β). Once monocytes and other inflammatory cells adhere to the endothelium they enter the extracellular matrix and differentiate into macrophages in an effort to create a favourable environment for vessel growth and development. Activated macrophages secrete inflammatory cytokines such as tumour necrosis factor-α (TNF-α), growth factors such as fibroblast growth factor-2 (FGF-2) and matrix metalloproteinases. Finally, vascular smooth muscle cells proliferate and switch to a contractile phenotype, resulting in an increased diameter and functionality of the collateral vessel, thereby allowing improved perfusion of the distal myocardium subtended by the occluded vessel. This simultaneously reduces FSS within the collateral vessel, inhibiting further vessel growth.
Collapse
|
23
|
Giménez CS, Castillo MG, Simonin JA, Núñez Pedrozo CN, Pascuali N, Bauzá MDR, Locatelli P, López AE, Belaich MN, Mendiz AO, Crottogini AJ, Cuniberti LA, Olea FD. Effect of intramuscular baculovirus encoding mutant hypoxia-inducible factor 1-alpha on neovasculogenesis and ischemic muscle protection in rabbits with peripheral arterial disease. Cytotherapy 2020; 22:563-572. [PMID: 32723595 DOI: 10.1016/j.jcyt.2020.06.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 06/07/2020] [Accepted: 06/26/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND AIMS Peripheral arterial disease (PAD) is a progressive, disabling ailment for which no effective treatment exists. Gene therapy-mediated neovascularization has emerged as a potentially useful strategy. We tested the angiogenic and arteriogenic efficacy and safety of a baculovirus (BV) encoding mutant, oxygen-resistant hypoxia-inducible factor 1-alpha (mHIF-1α), in rabbits with PAD. METHODS After assessing the transfection efficiency of the BV.mHIF-1α vector and its tubulogenesis potential in vitro, we randomized rabbits with experimental PAD to receive 1 × 109 copies of BV.mHIF-1α or BV.null (n = 6 per group) 7 days after surgery. Two weeks post-treatment, collateralization (digital angiography) and capillary and arteriolar densities (immunohistochemistry) were measured in the posterior limbs. Ischemic damage was evaluated in adductor and gastrocnemius muscle samples. Tracking of viral DNA in injected zones and remote tissues at different time points was performed in additional rabbits using a BV encoding GFP. RESULTS Angiographically visible collaterals were more numerous in BV.mHIF-1α-treated rabbits (8.12 ± 0.42 vs 6.13 ± 1.15 collaterals/cm2, P < 0.05). The same occurred with arteriolar (27.9 ± 7.0 vs 15.3 ± 4.0 arterioles/mm2) and capillary (341.8 ± 109.9 vs 208.8 ± 87.7 capillaries/mm2, P < 0.05) densities. BV.mHIF-1α-treated rabbits displayed less ischemic muscle damage than BV.null-treated animals. Viral DNA and GFP mRNA were detectable only at 3 and 7 days after injection in hind limbs. Neither the virus nor GFP mRNA was detected in remote tissues. CONCLUSIONS In rabbits with PAD, BV.mHIF-1α induced neovascularization and reduced ischemic damage, exhibiting a good safety profile at 14 days post-treatment. Complementary studies to evaluate its potential usefulness in the clinic are needed.
Collapse
Affiliation(s)
- Carlos S Giménez
- Laboratorio de Medicina Regenerativa Cardiovascular, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Favaloro, Buenos Aires, Argentina
| | - Martha G Castillo
- Laboratorio de Medicina Regenerativa Cardiovascular, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Favaloro, Buenos Aires, Argentina
| | - Jorge A Simonin
- Laboratorio de Ingeniería Genética y Biología Celular y Molecular (LIGBCM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Quilmes, Bernal, Argentina
| | - Cristian N Núñez Pedrozo
- Laboratorio de Medicina Regenerativa Cardiovascular, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Favaloro, Buenos Aires, Argentina
| | - Natalia Pascuali
- Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María Del Rosario Bauzá
- Laboratorio de Medicina Regenerativa Cardiovascular, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Favaloro, Buenos Aires, Argentina
| | - Paola Locatelli
- Laboratorio de Medicina Regenerativa Cardiovascular, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Favaloro, Buenos Aires, Argentina
| | - Ayelén E López
- Laboratorio de Medicina Regenerativa Cardiovascular, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Favaloro, Buenos Aires, Argentina
| | - Mariano N Belaich
- Laboratorio de Ingeniería Genética y Biología Celular y Molecular (LIGBCM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Quilmes, Bernal, Argentina
| | - Alfredo O Mendiz
- Hospital Universitario de la Fundación Favaloro, Buenos Aires, Argentina
| | - Alberto J Crottogini
- Laboratorio de Medicina Regenerativa Cardiovascular, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Favaloro, Buenos Aires, Argentina
| | - Luis A Cuniberti
- Laboratorio de Medicina Regenerativa Cardiovascular, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Favaloro, Buenos Aires, Argentina
| | - Fernanda D Olea
- Laboratorio de Medicina Regenerativa Cardiovascular, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Favaloro, Buenos Aires, Argentina.
| |
Collapse
|
24
|
Effect of Physical Exercise on the Release of Microparticles with Angiogenic Potential. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10144871] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cellular communication has a fundamental role in both human physiological and pathological states and various mechanisms are involved in the crosstalk between organs. Among these, microparticles (MPs) have an important involvement. MPs are a subtype of extracellular vesicles produced by a variety of cells following activation or apoptosis. They are normally present in physiological conditions, but their concentration varies in pathological states such as cardiovascular disease, diabetes mellitus, or cancer. Acute and chronic physical exercise are able to modify MPs amounts as well. Among various actions, exercise-responsive MPs affect angiogenesis, the process through which new blood vessels grow from pre-existing vessels. Usually, the neo vascular growth has functional role; but an aberrant neovascularization accompanies several oncogenic, ischemic, or inflammatory diseases. In addition, angiogenesis is one of the key adaptations to physical exercise and training. In the present review, we report evidence regarding the effect of various typologies of exercise on circulating MPs that are able to affect angiogenesis.
Collapse
|
25
|
Liu X, Dong H, Huang B, Miao H, Xu Z, Yuan Y, Qiu F, Chen J, Zhang H, Liu Z, Quan X, Zhu L, Zhang Z. Native Coronary Collateral Microcirculation Reserve in Rat Hearts. J Am Heart Assoc 2020; 8:e011220. [PMID: 30819021 PMCID: PMC6474916 DOI: 10.1161/jaha.118.011220] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background We occasionally noticed that native collateral blood flow showed a recessive trend in the early stages of acute myocardial infarction in rats, which greatly interferes with the accurate assessment of native collateral circulation levels. Here, we sought to recognize the coronary collateral circulation system in depth, especially the microcirculation part, on this basis. Methods and Results In this study, we detected native collateral flow with positron emission tomography perfusion imaging in rats and found that the native flow is relatively abundant when it is initially recruited. However, this flow is extremely unstable in the early stage of acute myocardial infarction and quickly fails. We used tracers to mark the collateral in an ischemic area and a massive preformed collateral network was labeled. The ultrastructures of these collateral microvessels are flawed, which contributes to extensive leakage and consequent interstitial edema in the ischemic region. Conclusions An unrecognized short-lived native coronary collateral microcirculation reserve is widely distributed in rat hearts. Recession of collateral blood flow transported by coronary collateral microcirculation reserve contributes to instability of native collateral blood flow in the early stage of acute myocardial infarction. The immature structure determines that these microvessels are short-lived and provide conditions for the development of early interstitial edema in acute myocardial infarction.
Collapse
Affiliation(s)
- Xiucheng Liu
- Department of Thoracic Cardiovascular SurgeryAffiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Hongyan Dong
- Morphological Research Experiment CenterXuzhou Medical UniversityXuzhouChina
| | - Bing Huang
- Department of Thoracic Cardiovascular SurgeryAffiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Haoran Miao
- Department of Thoracic Cardiovascular SurgeryAffiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Zhiwei Xu
- Department of Cardiovascular SurgeryShanghai Chest HospitalShanghai Jiaotong UniversityShanghaiChina
| | - Yanliang Yuan
- Department of Thoracic Cardiovascular SurgeryAffiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Fan Qiu
- Department of Thoracic Cardiovascular SurgeryAffiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Jiali Chen
- Department of Thoracic Cardiovascular SurgeryAffiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Hao Zhang
- Department of Thoracic Cardiovascular SurgeryAffiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Zhiwei Liu
- Morphological Research Experiment CenterXuzhou Medical UniversityXuzhouChina
| | - Xiaoyu Quan
- Department of Thoracic Cardiovascular SurgeryAffiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Lidong Zhu
- Department of Thoracic Cardiovascular SurgeryAffiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Zhongming Zhang
- Department of Thoracic Cardiovascular SurgeryAffiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| |
Collapse
|
26
|
Kumaraswami K, Salei N, Beck S, Rambichler S, Kluever AK, Lasch M, Richter L, Schraml BU, Deindl E. A Simple and Effective Flow Cytometry-Based Method for Identification and Quantification of Tissue Infiltrated Leukocyte Subpopulations in a Mouse Model of Peripheral Arterial Disease. Int J Mol Sci 2020; 21:ijms21103593. [PMID: 32438752 PMCID: PMC7279164 DOI: 10.3390/ijms21103593] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 05/16/2020] [Indexed: 01/25/2023] Open
Abstract
Arteriogenesis, the growth of a natural bypass from pre-existing arteriolar collaterals, is an endogenous mechanism to compensate for the loss of an artery. Mechanistically, this process relies on a locally and temporally restricted perivascular infiltration of leukocyte subpopulations, which mediate arteriogenesis by supplying growth factors and cytokines. Currently, the state-of-the-art method to identify and quantify these leukocyte subpopulations in mouse models is immunohistology. However, this is a time consuming procedure. Here, we aimed to develop an optimized protocol to identify and quantify leukocyte subpopulations by means of flow cytometry in adductor muscles containing growing collateral arteries. For that purpose, adductor muscles of murine hindlimbs were isolated at day one and three after induction of arteriogenesis, enzymatically digested, and infiltrated leukocyte subpopulations were identified and quantified by flow cytometry, as exemplary shown for neutrophils and macrophages (defined as CD45+/CD11b+/Ly6G+ and CD45+/CD11b+/F4/80+ cells, respectively). In summary, we show that flow cytometry is a suitable method to identify and quantify leukocyte subpopulations in muscle tissue, and provide a detailed protocol. Flow cytometry constitutes a timesaving tool compared to histology, which might be used in addition for precise localization of leukocytes in tissue samples.
Collapse
Affiliation(s)
- Konda Kumaraswami
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (K.K.); (N.S.); (S.B.); (S.R.); (A.-K.K.); (M.L.); (B.U.S.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, LMU Munich, 82152 Planegg-Martinsried, Germany
| | - Natallia Salei
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (K.K.); (N.S.); (S.B.); (S.R.); (A.-K.K.); (M.L.); (B.U.S.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, LMU Munich, 82152 Planegg-Martinsried, Germany
| | - Sebastian Beck
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (K.K.); (N.S.); (S.B.); (S.R.); (A.-K.K.); (M.L.); (B.U.S.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, LMU Munich, 82152 Planegg-Martinsried, Germany
| | - Stephan Rambichler
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (K.K.); (N.S.); (S.B.); (S.R.); (A.-K.K.); (M.L.); (B.U.S.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, LMU Munich, 82152 Planegg-Martinsried, Germany
| | - Anna-Kristina Kluever
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (K.K.); (N.S.); (S.B.); (S.R.); (A.-K.K.); (M.L.); (B.U.S.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, LMU Munich, 82152 Planegg-Martinsried, Germany
| | - Manuel Lasch
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (K.K.); (N.S.); (S.B.); (S.R.); (A.-K.K.); (M.L.); (B.U.S.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, LMU Munich, 82152 Planegg-Martinsried, Germany
- Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Lisa Richter
- Core Facility Flow Cytometry, Biomedical Centre, LMU Munich, 82152 Planegg-Martinsried, Germany;
| | - Barbara U. Schraml
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (K.K.); (N.S.); (S.B.); (S.R.); (A.-K.K.); (M.L.); (B.U.S.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, LMU Munich, 82152 Planegg-Martinsried, Germany
| | - Elisabeth Deindl
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (K.K.); (N.S.); (S.B.); (S.R.); (A.-K.K.); (M.L.); (B.U.S.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, LMU Munich, 82152 Planegg-Martinsried, Germany
- Correspondence: ; Tel.: +49-89-2180-76504
| |
Collapse
|
27
|
Zukerman H, Khoury M, Shammay Y, Sznitman J, Lotan N, Korin N. Targeting functionalized nanoparticles to activated endothelial cells under high wall shear stress. Bioeng Transl Med 2020; 5:e10151. [PMID: 32440559 PMCID: PMC7237145 DOI: 10.1002/btm2.10151] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/12/2019] [Accepted: 12/03/2019] [Indexed: 11/26/2022] Open
Abstract
Local inflammation of the endothelium is associated with a plethora of cardiovascular diseases. Vascular-targeted carriers (VTCs) have been advocated to provide focal effective therapeutics to these disease sites. Here, we examine the design of functionalized nanoparticles (NPs) as VTCs that can specifically localize at an inflamed vessel wall under pathological levels of high shear stress, associated for example with clinical (or in vivo) conditions of vascular narrowing and arteriogenesis. To test this, carboxylated fluorescent 200 nm polystyrene particles were functionalized with ligands to activated endothelium, that is, an E-selectin binding peptide (Esbp), an anti ICAM-1 antibody, or using a combination of both. The functionalized NPs were investigated in vitro using microfluidic models lined with inflamed (TNF-α stimulated) and control endothelial cells (EC). Specifically, their adhesion was monitored under different relevant wall shear stresses (i.e., 40-300 dyne/cm2) via real-time confocal microscopy. Experiments reveal a significantly higher specific adhesion of the examined functionalized NPs to activated EC for the window of examined wall shear stresses. Moreover, particle adhesion correlated with the surface coating density whereby under high surface coating (i.e., ~10,000 molecule/particle), shear-dependent particle adhesion increased significantly. Altogether, our results show that functionalized NPs can be designed to target inflamed endothelial cells under high shear stress. Such VTCs underscore the potential for attractive avenues in targeting drugs to vasoconstriction and arteriogenesis sites.
Collapse
Affiliation(s)
- Hila Zukerman
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Maria Khoury
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Yosi Shammay
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Josué Sznitman
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Noah Lotan
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Netanel Korin
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifaIsrael
| |
Collapse
|
28
|
Wieczór R, Kulwas A, Rość D. Implications of Hemostasis Disorders in Patients with Critical Limb Ischemia-An In-Depth Comparison of Selected Factors. J Clin Med 2020; 9:E659. [PMID: 32121363 PMCID: PMC7141251 DOI: 10.3390/jcm9030659] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Atherosclerosis is a systemic disease. Among patients with atherosclerosis, those suffering from peripheral arterial disease (PAD) represent a group of individuals with particularly high death risk, especially during the course of critical limb ischemia (CLI). In the pathogenesis of PAD/CLI complications, blood coagulation disorders play a significant role. The study aim was to examine the activation of the coagulation system depending on tissue factor (TF) in patients with CLI as compared with those with intermittent claudication (IC). METHODS Before initiating proper treatment (invasive or maintenance), blood samples were collected from 65 patients with CLI and 15 with IC to measure the following selected hemostasis parameters: concentrations and activation of tissue factor (TF Ag and TF Act) and tissue factor pathway inhibitor (TFPI Ag and TFPI Act), concentrations of thrombin-antithrombin complex (TAT Ag) and fibrinogen, platelet count (PLT), and concentrations of tissue-plasminogen activator (t-PA Ag), plasminogen activator inhibitor 1 (PAI-1), and D-dimer. The control group included 30 healthy volunteers (10 female/20 male). RESULTS The values of all analyzed parameters (except for lower TFPI Act) were significantly higher in the blood of PAD patients (with respect to PLT only in the CLI subgroup) in comparison with healthy subjects. The blood of patients with CLI as compared to the IC subgroup revealed much higher concentrations of TF Ag (p < 0.001), with slightly decreased TF Act, significantly lower concentrations of TFPI Ag (p < 0.001), slightly increased TFPI Act, and significantly higher levels of TAT Ag (p < 0.001), fibrinogen (p = 0.026), and D-dimer (p < 0.05). CONCLUSIONS In patients with CLI, we can observe coagulation activation and a shifting balance toward prothrombotic processes. Furthermore, increased concentrations of D-dimer suggest a secondary activation of fibrinolysis and confirm the phenomenon as a prothrombotic condition with heightened fibrinolysis.
Collapse
Affiliation(s)
- Radosław Wieczór
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University in Toruń, Ludwik Rydygier Collegium Medicum in Bydgoszcz, 85–094 Bydgoszcz, Poland; (A.K.); (D.R.)
- Clinic of Vascular and Internal Medicine, Dr Jan Biziel University Hospital No. 2 in Bydgoszcz, 85-168 Bydgoszcz, Poland
| | - Arleta Kulwas
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University in Toruń, Ludwik Rydygier Collegium Medicum in Bydgoszcz, 85–094 Bydgoszcz, Poland; (A.K.); (D.R.)
| | - Danuta Rość
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University in Toruń, Ludwik Rydygier Collegium Medicum in Bydgoszcz, 85–094 Bydgoszcz, Poland; (A.K.); (D.R.)
| |
Collapse
|
29
|
Abstract
Coronary heart disease (CHD) is the most common and serious illness in the world and has been researched for many years. However, there are still no real effective ways to prevent and save patients with this disease. When patients present with myocardial infarction, the most important step is to recover ischemic prefusion, which usually is accomplished by coronary artery bypass surgery, coronary artery intervention (PCI), or coronary artery bypass grafting (CABG). These are invasive procedures, and patients with extensive lesions cannot tolerate surgery. It is, therefore, extremely urgent to search for a noninvasive way to save ischemic myocardium. After suffering from ischemia, cardiac or skeletal muscle can partly recover blood flow through angiogenesis (de novo capillary) induced by hypoxia, arteriogenesis, or collateral growth (opening and remodeling of arterioles) triggered by dramatical increase of fluid shear stress (FSS). Evidence has shown that both of them are regulated by various crossed pathways, such as hypoxia-related pathways, cellular metabolism remodeling, inflammatory cells invasion and infiltration, or hemodynamical changes within the vascular wall, but still they do not find effective target for regulating revascularization at present. 5′-Adenosine monophosphate-activated protein kinase (AMPK), as a kinase, is not only an energy modulator but also a sensor of cellular oxygen-reduction substances, and many researches have suggested that AMPK plays an essential role in revascularization but the mechanism is not completely understood. Usually, AMPK can be activated by ADP or AMP, upstream kinases or other cytokines, and pharmacological agents, and then it phosphorylates key molecules that are involved in energy metabolism, autophagy, anti-inflammation, oxidative stress, and aging process to keep cellular homeostasis and finally keeps cell normal activity and function. This review makes a summary on the subunits, activation and downstream targets of AMPK, the mechanism of revascularization, the effects of AMPK in endothelial cells, angiogenesis, and arteriogenesis along with some prospects.
Collapse
|
30
|
Sun N, Ning B, Bruce AC, Cao R, Seaman SA, Wang T, Fritsche-Danielson R, Carlsson LG, Peirce SM, Hu S. In vivo imaging of hemodynamic redistribution and arteriogenesis across microvascular network. Microcirculation 2019; 27:e12598. [PMID: 31660674 DOI: 10.1111/micc.12598] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 10/17/2019] [Accepted: 10/21/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Arteriogenesis is an important mechanism that contributes to restoration of oxygen supply in chronically ischemic tissues, but remains incompletely understood due to technical limitations. This study presents a novel approach for comprehensive assessment of the remodeling pattern in a complex microvascular network containing multiple collateral microvessels. METHODS We have developed a hardware-software integrated platform for quantitative, longitudinal, and label-free imaging of network-wide hemodynamic changes and arteriogenesis at the single-vessel level. By ligating feeding arteries in the mouse ear, we induced network-wide hemodynamic redistribution and localized arteriogenesis. The utility of this technology was demonstrated by studying the influence of obesity on microvascular arteriogenesis. RESULTS Simultaneously monitoring the remodeling of competing collateral arterioles revealed a new, inverse relationship between initial vascular resistance and extent of arteriogenesis. Obese mice exhibited similar remodeling responses to lean mice through the first week, including diameter increase and flow upregulation in collateral arterioles. However, these gains were subsequently lost in obese mice. CONCLUSIONS Capable of label-free, comprehensive, and dynamic quantification of structural and functional changes in the microvascular network in vivo, this platform opens up new opportunities to study the mechanisms of microvascular arteriogenesis, its implications in diseases, and approaches to pharmacologically rectify microvascular dysfunction.
Collapse
Affiliation(s)
- Naidi Sun
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Bo Ning
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Anthony C Bruce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Rui Cao
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Scott A Seaman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Tianxiong Wang
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | | | - Leif G Carlsson
- Bioscience Heart Failure, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Shayn M Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Song Hu
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
31
|
Liu X, Liu Z, Chen J, Zhu L, Zhang H, Quan X, Yuan Y, Miao H, Huang B, Dong H, Zhang Z. Pigment Epithelium-Derived Factor Increases Native Collateral Blood Flow to Improve Cardiac Function and Induce Ventricular Remodeling After Acute Myocardial Infarction. J Am Heart Assoc 2019; 8:e013323. [PMID: 31718448 PMCID: PMC6915271 DOI: 10.1161/jaha.119.013323] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background We previously found that the structural defects of the coronary collateral microcirculation reserve (CCMR) prevent these preformed collateral vessels from continuously delivering the native collateral blood and supporting the ischemic myocardium in rats. Here, we tested whether these native collaterals can be remodeled by artificially increasing pigment epithelium–derived factor (PEDF) expression and demonstrated the mechanism for this stimulation. Methods and Results We performed intramyocardial gene delivery (PEDF‐lentivirus, 2×107 TU) along the left anterior descending coronary artery to artificially increase the expression of PEDF in the tissue of the region for 2 weeks. By blocking the left anterior descending coronary artery, we examined the effects of PEDF on native collateral blood flow and CCMR. The results of positron emission tomography perfusion imaging showed that PEDF increased the native collateral blood flow and significantly inhibited its decline during acute myocardial infarction. In addition, the number of CCMR vessels decreased and the size increased. Similar results were obtained from in vitro experiments. We tested whether PEDF induces CCMR remodeling in a fluid shear stress–like manner by detecting proteins and signaling pathways that are closely related to fluid shear stress. The nitric oxide pathway and the Notch‐1 pathway participated in the process of CCMR remodeling induced by PEDF. Conclusions PEDF treatment activates the nitric oxide pathway, and the Notch‐1 pathway enabled CCMR remodeling. Increasing the native collateral blood flow can promote the ventricular remodeling process and improve prognosis after acute myocardial infarction.
Collapse
Affiliation(s)
- Xiucheng Liu
- Department of Thoracic Cardiovascular SurgeryAffiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Zhiwei Liu
- Morphological Research Experiment CenterXuzhou Medical UniversityXuzhouChina
| | - Jiali Chen
- Department of Thoracic Cardiovascular SurgeryAffiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Lidong Zhu
- Department of Thoracic Cardiovascular SurgeryAffiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Hao Zhang
- Department of Thoracic Cardiovascular SurgeryAffiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Xiaoyu Quan
- Department of Thoracic Cardiovascular SurgeryAffiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Yanliang Yuan
- Department of Thoracic Cardiovascular SurgeryAffiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Haoran Miao
- Department of Thoracic Cardiovascular SurgeryAffiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Bing Huang
- Department of Thoracic Cardiovascular SurgeryAffiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Hongyan Dong
- Morphological Research Experiment CenterXuzhou Medical UniversityXuzhouChina
| | - Zhongming Zhang
- Department of Thoracic Cardiovascular SurgeryAffiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| |
Collapse
|
32
|
Fadini GP, Albiero M, Bonora BM, Avogaro A. Angiogenic Abnormalities in Diabetes Mellitus: Mechanistic and Clinical Aspects. J Clin Endocrinol Metab 2019; 104:5431-5444. [PMID: 31211371 DOI: 10.1210/jc.2019-00980] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 06/12/2019] [Indexed: 12/25/2022]
Abstract
CONTEXT Diabetes causes severe pathological changes to the microvasculature in many organs and tissues and is at the same time associated with an increased risk of coronary and peripheral macrovascular events. We herein review alterations in angiogenesis observed in human and experimental diabetes and how they contribute to diabetes onset and development of vascular complications. EVIDENCE ACQUISITION The English language medical literature was searched for articles reporting on angiogenesis/vasculogenesis abnormalities in diabetes and their clinical manifestations, mechanistic aspects, and possible therapeutic implications. EVIDENCE SYNTHESIS Angiogenesis is a complex process, driven by a multiplicity of molecular mechanisms and involved in several physiological and pathological conditions. Incompetent angiogenesis is pervasive in diabetic vascular complications, with both excessive and defective angiogenesis observed in various tissues. A striking different angiogenic response typically occurs in the retina vs the myocardium and peripheral circulation, but some commonalities in abnormal angiogenesis can explain the well-known association between microangiopathy and macroangiopathy. Impaired angiogenesis can also affect endocrine islet and adipose tissue function, providing a link to diabetes onset. Exposure to high glucose itself directly affects angiogenic/vasculogenic processes, and the mechanisms include defective responses to hypoxia and proangiogenic factors, impaired nitric oxide bioavailability, shortage of proangiogenic cells, and loss of pericytes. CONCLUSIONS Dissecting the molecular drivers of tissue-specific alterations of angiogenesis/vasculogenesis is an important challenge to devise new therapeutic approaches. Angiogenesis-modulating therapies should be carefully evaluated in view of their potential off-target effects. At present, glycemic control remains the most reasonable therapeutic strategy to normalize angiogenesis in diabetes.
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine, University of Padova, Padova, Italy
- Venetian Institute of Molecular Medicine, Padova, Italy
| | - Mattia Albiero
- Department of Medicine, University of Padova, Padova, Italy
- Venetian Institute of Molecular Medicine, Padova, Italy
| | - Benedetta Maria Bonora
- Department of Medicine, University of Padova, Padova, Italy
- Venetian Institute of Molecular Medicine, Padova, Italy
| | - Angelo Avogaro
- Department of Medicine, University of Padova, Padova, Italy
| |
Collapse
|
33
|
Khalfallah M, Draz E, Shalaby K, Hafez YM. Predictors of poorly developed coronary collateral circulation in patients with subclinical hypothyroidism suffered from chronic stable angina. Glob Cardiol Sci Pract 2019; 2019:e201910. [PMID: 31799285 PMCID: PMC6865308 DOI: 10.21542/gcsp.2019.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Background. The development of coronary collaterals is variable among patients with coronary artery disease and remains incompletely understood. We aimed to demonstrate the predictors of poorly developed coronary collateral circulation (CCC) in patients with subclinical hypothyroidism suffered from chronic stable angina. Methods. The study was conducted on 226 patients with subclinical hypothyroidism suffered from chronic stable angina, coronary angiography documented total occlusion at any major coronary artery or coronary artery lumen diameter stenosis >90%. Patients were divided into two groups according to grade of CCC, group A: 138 patients with (good collaterals) and group B: 88 patients with (poor collaterals). To classify CCC, we used Rentrop’s classification. Results. Multivariate regression analysis was performed and identified the independent predictors of poor coronary collaterals: N/L ratio (OR 0.413, CI 95% [0.172–0.993], p = 0.048), and TSH (OR 2.511, CI 95% [1.784–3.534], p = 0.001). The ROC analysis provided a cut-off value of >4.6 for N/L ratio, and >9 µIU/mL for TSH to predict poor coronary collaterals. Conclusion. An elevated level of N/L ratio >4.6 and TSH level >9 µIU/mL were the independent predictors of poorly developed CCC in patients with subclinical hypothyroidism suffered from chronic stable angina.
Collapse
Affiliation(s)
- Mohamed Khalfallah
- Department of Cardiovascular Medicine, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Enas Draz
- Department of Cardiovascular Medicine, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Khaled Shalaby
- Department of Internal Medicine, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Yasser Mostafa Hafez
- Department of Internal Medicine, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
34
|
Rajendran S, Shen X, Glawe J, Kolluru GK, Kevil CG. Nitric Oxide and Hydrogen Sulfide Regulation of Ischemic Vascular Growth and Remodeling. Compr Physiol 2019; 9:1213-1247. [PMID: 31187898 DOI: 10.1002/cphy.c180026] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ischemic vascular remodeling occurs in response to stenosis or arterial occlusion leading to a change in blood flow and tissue perfusion. Altered blood flow elicits a cascade of molecular and cellular physiological responses leading to vascular remodeling of the macro- and micro-circulation. Although cellular mechanisms of vascular remodeling such as arteriogenesis and angiogenesis have been studied, therapeutic approaches in these areas have had limited success due to the complexity and heterogeneous constellation of molecular signaling events regulating these processes. Understanding central molecular players of vascular remodeling should lead to a deeper understanding of this response and aid in the development of novel therapeutic strategies. Hydrogen sulfide (H2 S) and nitric oxide (NO) are gaseous signaling molecules that are critically involved in regulating fundamental biochemical and molecular responses necessary for vascular growth and remodeling. This review examines how NO and H2 S regulate pathophysiological mechanisms of angiogenesis and arteriogenesis, along with important chemical and experimental considerations revealed thus far. The importance of NO and H2 S bioavailability, their synthesis enzymes and cofactors, and genetic variations associated with cardiovascular risk factors suggest that they serve as pivotal regulators of vascular remodeling responses. © 2019 American Physiological Society. Compr Physiol 9:1213-1247, 2019.
Collapse
Affiliation(s)
| | - Xinggui Shen
- Departments of Pathology, LSU Health Sciences Center, Shreveport
| | - John Glawe
- Departments of Pathology, LSU Health Sciences Center, Shreveport
| | - Gopi K Kolluru
- Departments of Pathology, LSU Health Sciences Center, Shreveport
| | - Christopher G Kevil
- Departments of Pathology, LSU Health Sciences Center, Shreveport.,Departments of Cellular Biology and Anatomy, LSU Health Sciences Center, Shreveport.,Departments of Molecular and Cellular Physiology, LSU Health Sciences Center, Shreveport
| |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW Refractory angina (RA), which is characterized by tissue ischemia along with neurological, mitochondrial, and psychogenic dysfunction, is becoming a major cause of morbidity in patients with advanced coronary artery disease. In this review, we discuss in detail the invasive mechanical non-cell therapy-based options, the evidence behind these therapies, and future trends. RECENT FINDINGS There is extensive ongoing research in the areas of spinal-cord stimulation, transmyocardial laser revascularization, sympathectomy, angiogenesis, and other non-cell-based therapies to explore the best therapy for refractory angina. There is conflicting data in the literature suggesting subjective improvement in angina, but very few studies boast improvement in core objective parameters such as myocardial blood flow, survival, or rehospitalizations. Patients with refractory angina are a complex group of patients that need novel approaches to help alleviate their symptoms and reduce mortality. A carefully selected sequence of therapies may provide the best results in this patient population.
Collapse
Affiliation(s)
- Amod Amritphale
- Oklahoma University Health Sciences Center, Oklahoma City, OK, USA.
| | | |
Collapse
|
36
|
Jamaiyar A, Juguilon C, Dong F, Cumpston D, Enrick M, Chilian WM, Yin L. Cardioprotection during ischemia by coronary collateral growth. Am J Physiol Heart Circ Physiol 2018; 316:H1-H9. [PMID: 30379567 DOI: 10.1152/ajpheart.00145.2018] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Ischemic heart diseases (IHD) cause millions of deaths around the world annually. While surgical and pharmacological interventions are commonly used to treat patients with IHD, their efficacy varies from patient to patient and is limited by the severity of the disease. One promising, at least theoretically, approach for treating IHD is induction of coronary collateral growth (CCG). Coronary collaterals are arteriole-to-arteriole anastomoses that can undergo expansion and remodeling in the setting of coronary disease when the disease elicits myocardial ischemia and creates a pressure difference across the collateral vessel that creates unidirectional flow. Well-developed collaterals can restore blood flow in the ischemic area of the myocardium and protect the myocardium at risk. Moreover, such collaterals are correlated to reduced mortality and infarct size and better cardiac function during occlusion of coronary arteries. Therefore, understanding the process of CCG is highly important as a potentially viable treatment of IHD. While there are several excellent review articles on this topic, this review will provide a unified overview of the various aspects related to CCG as well as an update of the advancements in the field. We also call for more detailed studies with an interdisciplinary approach to advance our knowledge of CCG. In this review, we will describe growth of coronary collaterals, the various factors that contribute to CCG, animal models used to study CCG, and the cardioprotective effects of coronary collaterals during ischemia. We will also discuss the impairment of CCG in metabolic syndrome and the therapeutic potentials of CCG in IHD.
Collapse
Affiliation(s)
- Anurag Jamaiyar
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio.,School of Biomedical Sciences, Kent State University , Kent, Ohio
| | - Cody Juguilon
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio
| | - Feng Dong
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio
| | - Devan Cumpston
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio
| | - Molly Enrick
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio
| | - William M Chilian
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio
| | - Liya Yin
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio
| |
Collapse
|
37
|
Castro PR, Barbosa AS, Pereira JM, Ranfley H, Felipetto M, Gonçalves CAX, Paiva IR, Berg BB, Barcelos LS. Cellular and Molecular Heterogeneity Associated with Vessel Formation Processes. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6740408. [PMID: 30406137 PMCID: PMC6199857 DOI: 10.1155/2018/6740408] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 09/06/2018] [Indexed: 12/11/2022]
Abstract
The microvasculature heterogeneity is a complex subject in vascular biology. The difficulty of building a dynamic and interactive view among the microenvironments, the cellular and molecular heterogeneities, and the basic aspects of the vessel formation processes make the available knowledge largely fragmented. The neovascularisation processes, termed vasculogenesis, angiogenesis, arteriogenesis, and lymphangiogenesis, are important to the formation and proper functioning of organs and tissues both in the embryo and the postnatal period. These processes are intrinsically related to microvascular cells, such as endothelial and mural cells. These cells are able to adjust their activities in response to the metabolic and physiological requirements of the tissues, by displaying a broad plasticity that results in a significant cellular and molecular heterogeneity. In this review, we intend to approach the microvasculature heterogeneity in an integrated view considering the diversity of neovascularisation processes and the cellular and molecular heterogeneity that contribute to microcirculatory homeostasis. For that, we will cover their interactions in the different blood-organ barriers and discuss how they cooperate in an integrated regulatory network that is controlled by specific molecular signatures.
Collapse
Affiliation(s)
- Pollyana Ribeiro Castro
- Department of Physiology and Biophysics, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Alan Sales Barbosa
- Department of Physiology and Biophysics, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Jousie Michel Pereira
- Department of Physiology and Biophysics, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Hedden Ranfley
- Department of Physiology and Biophysics, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Mariane Felipetto
- Department of Physiology and Biophysics, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Carlos Alberto Xavier Gonçalves
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Isabela Ribeiro Paiva
- Department of Pharmacology, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Bárbara Betônico Berg
- Department of Pharmacology, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Luciola Silva Barcelos
- Department of Physiology and Biophysics, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Brazil
| |
Collapse
|
38
|
Gatzke N, Hillmeister P, Dülsner A, Güc N, Dawid R, Smith KH, Pagonas N, Bramlage P, Gorath M, Buschmann IR. Nitroglycerin application and coronary arteriogenesis. PLoS One 2018; 13:e0201597. [PMID: 30118486 PMCID: PMC6097676 DOI: 10.1371/journal.pone.0201597] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 07/18/2018] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND In the presence of a coronary occlusion, pre-existing small collateral vessels (arterioles) develop into much larger arteries (biological bypasses) that have the potential to allow a certain level of perfusion distal to the blockage. Termed arteriogenesis, this phenomenon proceeds via a complex combination of events, with nitric oxide (NO) playing an essential role. The aim of this study was to investigate the effects of supplemental administration of NO donors, i.e., short-acting nitroglycerin (NTG) or slow-release pelleted isosorbide dinitrate (ISDN), on collateral development in a repetitive coronary artery occlusion model in rats. METHODS Coronary collateral growth was induced via a repetitive occlusion protocol (ROP) of the left anterior descending coronary artery (LAD) in rats. The primary endpoints were the histological evaluation of rat heart infarct size and ST-segment elevation (ECG-analysis) upon final permanent occlusion of the LAD (experimentally induced myocardial infarction). The effects of NTG or ISDN were also evaluated by administration during 5 days of ROP. We additionally investigated whether concomitant application of NTG can compensate for the anti-arteriogenic effect of acetylsalicylic acid (ASA). RESULTS After 5 days of ROP, the mean infarct size and degree of ST-elevation were only slightly lower than those of the SHAM group; however, after 10 days of the protocol, the ROP group displayed significantly less severe infarct damage, indicating enhanced arteriogenesis. Intermittent NTG application greatly decreased the ST-elevation and infarct size. The ISDN also had a positive effect on arteriogenesis, but not to the same extent as the NTG. Administration of ASA increased the infarct severity; however, concomitant dosing with NTG somewhat attenuated this effect. CONCLUSION Intermittent treatment with the short-acting NTG decreased the size of an experimentally induced myocardial infarct by promoting coronary collateral development. These new insights are of great relevance for future clinical strategies for the treatment of occlusive vascular diseases.
Collapse
Affiliation(s)
- Nora Gatzke
- Department for Angiology, Brandenburg Medical School, Campus Brandenburg/Havel, Brandenburg/Havel, Germany
- Department of Cardiology, Charité University Hospital, Campus Virchow, Berlin, Germany
- Center for Cardiovascular Research (CCR) Charité University Hospital, Campus Mitte, Berlin, Germany
| | - Philipp Hillmeister
- Department for Angiology, Brandenburg Medical School, Campus Brandenburg/Havel, Brandenburg/Havel, Germany
- Department of Cardiology, Charité University Hospital, Campus Virchow, Berlin, Germany
- Center for Cardiovascular Research (CCR) Charité University Hospital, Campus Mitte, Berlin, Germany
| | - André Dülsner
- Department of Cardiology, Charité University Hospital, Campus Virchow, Berlin, Germany
- Center for Cardiovascular Research (CCR) Charité University Hospital, Campus Mitte, Berlin, Germany
| | - Nadija Güc
- Department of Cardiology, Charité University Hospital, Campus Virchow, Berlin, Germany
- Center for Cardiovascular Research (CCR) Charité University Hospital, Campus Mitte, Berlin, Germany
| | - Rica Dawid
- Department for Angiology, Brandenburg Medical School, Campus Brandenburg/Havel, Brandenburg/Havel, Germany
| | | | - Nikolaos Pagonas
- Department for Angiology, Brandenburg Medical School, Campus Brandenburg/Havel, Brandenburg/Havel, Germany
| | - Peter Bramlage
- Department for Angiology, Brandenburg Medical School, Campus Brandenburg/Havel, Brandenburg/Havel, Germany
- Institute for Pharmacology and Preventive Medicine, Mahlow, Germany
| | | | - Ivo R. Buschmann
- Department for Angiology, Brandenburg Medical School, Campus Brandenburg/Havel, Brandenburg/Havel, Germany
- Department of Cardiology, Charité University Hospital, Campus Virchow, Berlin, Germany
- Center for Cardiovascular Research (CCR) Charité University Hospital, Campus Mitte, Berlin, Germany
- * E-mail:
| |
Collapse
|
39
|
Broughton KM, Wang BJ, Firouzi F, Khalafalla F, Dimmeler S, Fernandez-Aviles F, Sussman MA. Mechanisms of Cardiac Repair and Regeneration. Circ Res 2018; 122:1151-1163. [PMID: 29650632 PMCID: PMC6191043 DOI: 10.1161/circresaha.117.312586] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiovascular regenerative therapies are pursued on both basic and translational levels. Although efficacy and value of cell therapy for myocardial regeneration can be debated, there is a consensus that profound deficits in mechanistic understanding limit advances, optimization, and implementation. In collaboration with the TACTICS (Transnational Alliance for Regenerative Therapies in Cardiovascular Syndromes), this review overviews several pivotal aspects of biological processes impinging on cardiac maintenance, repair, and regeneration. The goal of summarizing current mechanistic understanding is to prompt innovative directions for fundamental studies delineating cellular reparative and regenerative processes. Empowering myocardial regenerative interventions, whether dependent on endogenous processes or exogenously delivered repair agents, ultimately depends on mastering mechanisms and novel strategies that take advantage of rather than being limited by inherent myocardial biology.
Collapse
Affiliation(s)
- Kathleen M Broughton
- From the Department of Biology, San Diego State University Heart Institute and the Integrated Regenerative Research Institute, CA (K.M.B., B.J.W., F.F., F.K., M.A.S.); Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Frankfurt, Germany (S.D.); and Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), CIBERCV and Universidad Complutense de Madrid, Spain (F.F.-A.)
| | - Bingyan J Wang
- From the Department of Biology, San Diego State University Heart Institute and the Integrated Regenerative Research Institute, CA (K.M.B., B.J.W., F.F., F.K., M.A.S.); Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Frankfurt, Germany (S.D.); and Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), CIBERCV and Universidad Complutense de Madrid, Spain (F.F.-A.)
| | - Fareheh Firouzi
- From the Department of Biology, San Diego State University Heart Institute and the Integrated Regenerative Research Institute, CA (K.M.B., B.J.W., F.F., F.K., M.A.S.); Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Frankfurt, Germany (S.D.); and Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), CIBERCV and Universidad Complutense de Madrid, Spain (F.F.-A.)
| | - Farid Khalafalla
- From the Department of Biology, San Diego State University Heart Institute and the Integrated Regenerative Research Institute, CA (K.M.B., B.J.W., F.F., F.K., M.A.S.); Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Frankfurt, Germany (S.D.); and Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), CIBERCV and Universidad Complutense de Madrid, Spain (F.F.-A.)
| | - Stefanie Dimmeler
- From the Department of Biology, San Diego State University Heart Institute and the Integrated Regenerative Research Institute, CA (K.M.B., B.J.W., F.F., F.K., M.A.S.); Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Frankfurt, Germany (S.D.); and Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), CIBERCV and Universidad Complutense de Madrid, Spain (F.F.-A.)
| | - Francisco Fernandez-Aviles
- From the Department of Biology, San Diego State University Heart Institute and the Integrated Regenerative Research Institute, CA (K.M.B., B.J.W., F.F., F.K., M.A.S.); Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Frankfurt, Germany (S.D.); and Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), CIBERCV and Universidad Complutense de Madrid, Spain (F.F.-A.)
| | - Mark A Sussman
- From the Department of Biology, San Diego State University Heart Institute and the Integrated Regenerative Research Institute, CA (K.M.B., B.J.W., F.F., F.K., M.A.S.); Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Frankfurt, Germany (S.D.); and Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), CIBERCV and Universidad Complutense de Madrid, Spain (F.F.-A.).
| |
Collapse
|
40
|
McEnaney RM, McCreary D, Tzeng E. A modified rat model of hindlimb ischemia for augmentation and functional measurement of arteriogenesis. J Biol Methods 2018; 5:e89. [PMID: 31435496 PMCID: PMC6703558 DOI: 10.14440/jbm.2018.234] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Arteriogenesis (collateral artery development) is an adaptive pathway critical for salvage of tissue in the setting of arterial occlusion. Rodent models of arteriogenesis typically involve an experimental occlusion (ligation) of a hindlimb artery and then rely on indirect measures such as laser Doppler perfusion imaging to assess blood flow recovery. Unfortunately, the more commonly utilized measures of distal tissue perfusion at rest are unable to account for hemodynamic and vasoactive variables and thus provide an incomplete assessment of collateral network capacity. We provide a detailed description of modifications to the commonly used model of femoral artery ligation. These serve to alter and then directly assess collateral network's hemodynamic capacity. By incorporating an arteriovenous fistula distal to the arterial ligation, arterial growth is maximized. Hindlimb perfusion may be isolated to measure minimum resistance of flow around the arterial occlusion, which provides a direct measure of collateral network capacity. Our results reinforce that arteriogenesis is driven by hemodynamic variables, and it can be reliably augmented and measured in absolute terms. Using these modifications to a widely used model, functional arteriogenesis may be more directly studied.
Collapse
Affiliation(s)
- Ryan M McEnaney
- Department of Surgery, University of Pittsburgh School of Medicine.,Veterans Affairs Pittsburgh Healthcare System
| | | | - Edith Tzeng
- Department of Surgery, University of Pittsburgh School of Medicine.,Veterans Affairs Pittsburgh Healthcare System
| |
Collapse
|
41
|
Ribatti D. A new role of mast cells in arteriogenesis. Microvasc Res 2018; 118:57-60. [PMID: 29501537 DOI: 10.1016/j.mvr.2018.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 02/27/2018] [Accepted: 02/27/2018] [Indexed: 02/03/2023]
Abstract
Arteriogenesis is defined as the growth of functional collateral arteries from pre-existing arterio-arteriolar anastomoses. The role of mast cells in arteriogenesis is largely unexplored. Recent evidences suggest that mast cells together with other inflammatory cells, including monocytes-macrophages, lymphocytes, NK cells and endothelial precursor cells (EPCs) may be involved in this process. This review article analyzes the literature concerning this new aspect of biological activity of mast cells.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy; National Cancer Institute "Giovanni Paolo II", Bari, Italy.
| |
Collapse
|
42
|
Xiang W, Reglin B, Nitzsche B, Maibier M, Rong WW, Hoffmann B, Ruggeri A, Guimarães P, Secomb TW, Pries AR. Dynamic remodeling of arteriolar collaterals after acute occlusion in chick chorioallantoic membrane. Microcirculation 2018; 24. [PMID: 28075525 DOI: 10.1111/micc.12351] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 01/09/2017] [Indexed: 12/26/2022]
Abstract
OBJECTIVE After arteriolar occlusion, collaterals enlarge and initially elevated WSS normalizes. While most previous studies focused on endpoints of such adaptive changes in larger collaterals, the present investigation aimed to continuously determine the relation between WSS and diameter in microvascular collaterals during adaptive reactions. METHODS In Hamburger-Hamilton stage 40 CAMs, junction points between arteriolar segments were identified and the third upstream segment on one side was occluded. Intravital microscopy recordings were taken for 24 hours post-occlusion. Segment diameter and blood velocity were measured: WSS and capillary density were calculated. RESULTS After occlusion, vascular diameters exhibited an immediate decrease, then increased with a time constant of 2.5 ± 0.8 hours and reached a plateau of up to 60% above baseline after about 7 hours. Vascular tone showed no significant change. WSS exhibited an immediate increase post-occlusion and linearly returned to baseline after about 12 hours. Local WSS change and diameter change rate showed similar patterns during the initial but not the later phase of post-occlusive adaptation. CONCLUSIONS CAM collaterals undergo fast structural remodeling within 24 hours post-occlusion. This remodeling might be driven by local WSS and by other regulators within the vascular network.
Collapse
Affiliation(s)
- Weiwei Xiang
- Department of Physiology, Charité Berlin, Berlin, Germany
| | - Bettina Reglin
- Department of Physiology, Charité Berlin, Berlin, Germany
| | | | - Martin Maibier
- Department of Physiology, Charité Berlin, Berlin, Germany
| | - Wen Wei Rong
- Department of Physiology, Charité Berlin, Berlin, Germany
| | - Björn Hoffmann
- Department of Physiology, Charité Berlin, Berlin, Germany
| | - Alfredo Ruggeri
- Department of Information Engineering, University of Padua, Padua, Italy
| | - Pedro Guimarães
- Department of Information Engineering, University of Padua, Padua, Italy
| | - Timothy W Secomb
- Department of Physiology, University of Arizona, Tucson, AZ, USA
| | - Axel R Pries
- Department of Physiology, Charité Berlin, Berlin, Germany.,Deutsches Herzzentrum Berlin, Berlin, Germany
| |
Collapse
|
43
|
Okyere B, Creasey M, Lebovitz Y, Theus MH. Temporal remodeling of pial collaterals and functional deficits in a murine model of ischemic stroke. J Neurosci Methods 2018; 293:86-96. [PMID: 28935424 PMCID: PMC5749401 DOI: 10.1016/j.jneumeth.2017.09.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 09/14/2017] [Accepted: 09/16/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Leptomeningeal anastomoses play a critical role in regulating reperfusion following cerebrovascular obstruction; however, methods to evaluate their temporospatial remodeling remains under investigation. NEW METHOD We combined arteriole-specific vessel painting with histological evaluation to assess the density and diameter of inter-collateral vessels between the middle cerebral artery and anterior cerebral artery (MCA-ACA) or posterior cerebral artery (MCA-PCA) in a murine model of permanent middle cerebral artery occlusion (pMCAO). RESULTS While the overall density was not influenced by pMCAO, the size of MCA-ACA and MCA-PCA vessels had significantly increased 2days post-pMCAO and peaked by 4days compared to the un-injured hemisphere. Using a combination of vessel painting and immunofluorescence, we uniquely observed an induction of cellular division and a remodeling of the smooth muscle cells within the collateral niche following post-pMCAO on whole mount tissue sections. Vessel painting was also applied to pMCAO-injured Cx3cr1GFP mice, in order to identify the spatial relationship between Cx3cr1-positive peripheral-derived monocyte/macrophages and the vessel painted collaterals. Our histological findings were supplemented with analysis of cerebral blood flow using laser Doppler imaging and behavioral changes following pMCAO. COMPARISON WITH EXISTING METHODS Compared to polyurethane and latex methods for collateral labeling, this new method provides detailed cell-type specific analysis within the collateral niche at the microscopic level, which has previously been unavailable. CONCLUSIONS This simple and reproducible combination of techniques is the first to dissect the temporospatial remodeling of pial collateral arterioles. The method will advance investigations into the underlying mechanisms governing the intricate processes of arteriogenesis.
Collapse
Affiliation(s)
- Benjamin Okyere
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, 970 Washington St. SW, Blacksburg, VA, 24061, USA
| | - Miranda Creasey
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, 970 Washington St. SW, Blacksburg, VA, 24061, USA
| | - Yeonwoo Lebovitz
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, 970 Washington St. SW, Blacksburg, VA, 24061, USA
| | - Michelle H Theus
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, 970 Washington St. SW, Blacksburg, VA, 24061, USA.
| |
Collapse
|
44
|
Dhindsa DS, Khambhati J, Sandesara PB, Eapen DJ, Quyyumi AA. Biomarkers to Predict Cardiovascular Death. Card Electrophysiol Clin 2017; 9:651-664. [PMID: 29173408 DOI: 10.1016/j.ccep.2017.07.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
This article reviews biomarkers that have been shown to identify subjects at increased risk for cardiovascular death within the general population, in those with established coronary artery disease, and in those with heart failure. Use of biomarkers for risk stratification for sudden cardiac death continues to evolve. It seems that a multimarker strategy for risk stratification using simple measures of circulating proteins and usual clinical risk factors, particularly in patients with known coronary artery disease, can be used to identify patients at near-term risk of death. Whether similar strategies in the general population will prove to be cost-effective needs to be investigated.
Collapse
Affiliation(s)
- Devinder S Dhindsa
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road Northeast, Suite 507, Atlanta, GA 30322, USA
| | - Jay Khambhati
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road Northeast, Suite 507, Atlanta, GA 30322, USA
| | - Pratik B Sandesara
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road Northeast, Suite 507, Atlanta, GA 30322, USA
| | - Danny J Eapen
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road Northeast, Suite 507, Atlanta, GA 30322, USA
| | - Arshed A Quyyumi
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road Northeast, Suite 507, Atlanta, GA 30322, USA.
| |
Collapse
|
45
|
Ai J, Sun JH, Ma J, Yao K. Effects of lentivirus-mediated endostatin on endothelial progenitor cells. Oncotarget 2017; 8:94431-94439. [PMID: 29212239 PMCID: PMC5706885 DOI: 10.18632/oncotarget.21770] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 09/20/2017] [Indexed: 12/27/2022] Open
Abstract
Endothelial progenitor cells (EPCs) are candidates for gene therapies against retinal neovascularization (NV). The aim of present study was to investigate the effects of endostatin transfection on EPC function. In the present study, the EPCs were infected with lentivirus overexpressing endostatin. The transfection effects of endostatin overexpression on the proliferation, migratory, differentiation, apoptosis and the cell cycle of this cell line were determined. The real-time quantitative polymerase chain reaction (RT-qPCR) and western blot assays showed high expression levels of endostatin. A cell counting kit-8 assay showed that endostatin overexpression inhibited EPC proliferation. The transwell assay indicated that endostatin overexpression could suppress EPC migration. Furthermore, endostatin overexpression enhanced apoptosis (as showed by AnnexinV-FITC/propidiumiodide staining analysis), induced differentiation and blocked the cell cycle. As compared with negative control group, EPC viability significantly decreased in gene transfection group. In conclusion, present study determined the feasibility of lentivirus-mediated endostatin gene transfer, and indirectly proved the effect of endostatin secretion on EPCs. Also our study provided a new opportunity for the potential application of gene therapy in retinal NV.
Collapse
Affiliation(s)
- Jing Ai
- Eye Center, 2nd Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Jun-Hui Sun
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jian Ma
- Eye Center, 2nd Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Ke Yao
- Eye Center, 2nd Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou 310009, China
| |
Collapse
|
46
|
Fujita Y, Kawamoto A. Stem cell-based peripheral vascular regeneration. Adv Drug Deliv Rev 2017; 120:25-40. [PMID: 28912015 DOI: 10.1016/j.addr.2017.09.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/28/2017] [Accepted: 09/07/2017] [Indexed: 02/07/2023]
Abstract
Chronic critical limb ischemia (CLI) represents an end-stage manifestation of peripheral arterial disease (PAD). CLI patients are at very high risk of amputation and cardiovascular complications, leading to severe morbidity and mortality. Because many patients with CLI are ineligible for conventional revascularization procedures, it is urgently needed to explore alternative strategies to improve blood supply in the ischemic tissue. Although researchers initially focused on gene/protein therapy using proangiogenic growth factors/cytokines, recent discovery of somatic stem/progenitor cells including bone marrow (BM)-derived endothelial progenitor cells (EPCs) and mesenchymal stem cells (MSCs) has drastically developed the field of therapeutic angiogenesis for CLI. Overall, early phase clinical trials demonstrated that stem/progenitor cell therapies may be safe, feasible and potentially effective. However, only few late-phase clinical trials have been conducted. This review provides an overview of the preclinical and clinical reports to demonstrate the usefulness and the current limitations of the cell-based therapies.
Collapse
Affiliation(s)
- Yasuyuki Fujita
- Division of Vascular Regeneration, Unit of Regenerative Medicine, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation, Japan; Translational Research Informatics Center, Foundation for Biomedical Research and Innovation, Japan
| | - Atsuhiko Kawamoto
- Division of Vascular Regeneration, Unit of Regenerative Medicine, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation, Japan; Translational Research Informatics Center, Foundation for Biomedical Research and Innovation, Japan.
| |
Collapse
|
47
|
Harnoss JM, Krackhardt F, Ritter Z, Granzow S, Felsenberg D, Neumann K, Lerman LO, Riediger F, Hillmeister P, Bramlage P, Buschmann IR. Porcine arteriogenesis based on vasa vasorum in a novel semi-acute occlusion model using high-resolution imaging. Heart Vessels 2017; 32:1400-1409. [PMID: 28776069 DOI: 10.1007/s00380-017-1028-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 07/28/2017] [Indexed: 11/29/2022]
Abstract
Bridging collaterals (BC) develop in several chronic total artery occlusion diseases, and can prevent extensive myocardial necrosis. Yet, their origin, growth process, and histo-morphology are still unclear. Since vasa vasorum (VV) may take part in collateralization, we hypothesized that VV are the basis for BCs. To comprehensively investigate this arteriogenesis process, we used high-resolution imaging, including corrosion casts, post-mortem angiography with stereoscopy, micro-CT, and immunohistology, in combination with a novel semi-acute vessel occlusion model. This porcine model was produced by implanting a copper stent minimally invasively into the left anterior descending coronary artery. To define the kinetics of arteriogenesis, pigs (n = 11) were assigned to one of the five euthanasia timepoints: day 0.5 (D0.5, n = 2), D3 (n = 2), D5 (n = 1), D7 (n = 3), or D12 (n = 3) after stent implantation. We found that (1) BCs originate from longitudinally running type 1 VV, mainly VV interna, partially also from VV externa; (2) the growth of VV to BC is rapid, occurring within 7 days; and (3) porcine BCs are likely functionally relevant, considering an observed 102% increase in the number of smooth muscle cell layers in their vascular wall. High-resolution imaging in a minimally invasive non-acute vessel occlusion model is an innovative technique that allowed us to provide direct evidence that porcine BCs develop from the VV. These data may be crucial for further studies on the treatment of angina pectoris and thromboangiitis obliterans through therapeutic stimulation of BC development.
Collapse
Affiliation(s)
- Jonathan M Harnoss
- Department for Angiology, Center for Internal Medicine I, Medizinische Hochschule Brandenburg (MHB), Brandenburg Medical School, Hochstr. 29, 14770, Brandenburg, Germany.,Department of Cardiology, Charité University Hospital, Campus Virchow, Berlin, Germany
| | - Florian Krackhardt
- Department for Angiology, Center for Internal Medicine I, Medizinische Hochschule Brandenburg (MHB), Brandenburg Medical School, Hochstr. 29, 14770, Brandenburg, Germany.,Department of Cardiology, Charité University Hospital, Campus Virchow, Berlin, Germany
| | - Zully Ritter
- Center for Muscle and Bone Research (ZMK), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Susanne Granzow
- Department of Cardiology, Charité University Hospital, Campus Virchow, Berlin, Germany
| | - Dieter Felsenberg
- Center for Muscle and Bone Research (ZMK), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Konrad Neumann
- Institute for Biometry and Clinical Epidemiology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Fabian Riediger
- Department for Angiology, Center for Internal Medicine I, Medizinische Hochschule Brandenburg (MHB), Brandenburg Medical School, Hochstr. 29, 14770, Brandenburg, Germany
| | - Philipp Hillmeister
- Department for Angiology, Center for Internal Medicine I, Medizinische Hochschule Brandenburg (MHB), Brandenburg Medical School, Hochstr. 29, 14770, Brandenburg, Germany.,Department of Cardiology, Charité University Hospital, Campus Virchow, Berlin, Germany
| | - Peter Bramlage
- Department for Angiology, Center for Internal Medicine I, Medizinische Hochschule Brandenburg (MHB), Brandenburg Medical School, Hochstr. 29, 14770, Brandenburg, Germany.,Institute for Pharmacology and Preventive Medicine, Mahlow, Germany
| | - Ivo R Buschmann
- Department for Angiology, Center for Internal Medicine I, Medizinische Hochschule Brandenburg (MHB), Brandenburg Medical School, Hochstr. 29, 14770, Brandenburg, Germany. .,Department of Cardiology, Charité University Hospital, Campus Virchow, Berlin, Germany.
| |
Collapse
|
48
|
Seaman SA, Cao Y, Campbell CA, Peirce SM. Arteriogenesis in murine adipose tissue is contingent on CD68 + /CD206 + macrophages. Microcirculation 2017; 24:10.1111/micc.12341. [PMID: 27976451 PMCID: PMC5432396 DOI: 10.1111/micc.12341] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 12/05/2016] [Indexed: 12/28/2022]
Abstract
OBJECTIVE The surgical transfer of skin, fat, and/or muscle from a donor site to a recipient site within the same patient is a widely performed procedure in reconstructive surgeries. A surgical pretreatment strategy that is intended to increase perfusion in the flap, termed "flap delay," is a commonly employed technique by plastic surgeons prior to flap transplantation. Here, we explored whether CD68+ /CD206+ macrophages are required for arteriogenesis within the flap by performing gain-of-function and loss-of-function studies in a previously published flap delay murine model. METHODS AND RESULTS Local injection of M2-polarized macrophages into the flap resulted in an increase in collateral vessel diameter. Application of a thin biomaterial film loaded with a pharmacological agent (FTY720), which has been previously shown to recruit CD68+ /CD206+ macrophages to remodeling tissue, increased CD68+ /CD206+ cell recruitment and collateral vessel enlargement. Conversely, when local macrophage populations were depleted within the inguinal fat pad via clodronate liposome delivery, we observed fewer CD68+ cells accompanied by diminished collateral vessel enlargement. CONCLUSIONS Our study underscores the importance of macrophages during microvascular adaptations that are induced by flap delay. These studies suggest a mechanism for a translatable therapeutic target that may be used to enhance the clinical flap delay procedure.
Collapse
Affiliation(s)
- Scott A. Seaman
- Department of Biomedical Engineering, University of Virginia
| | - Yiqi Cao
- Department of Biomedical Engineering, University of Virginia
| | | | - Shayn M. Peirce
- Department of Biomedical Engineering, University of Virginia
- Department of Plastic Surgery, University of Virginia
| |
Collapse
|
49
|
Paton MCB, McDonald CA, Allison BJ, Fahey MC, Jenkin G, Miller SL. Perinatal Brain Injury As a Consequence of Preterm Birth and Intrauterine Inflammation: Designing Targeted Stem Cell Therapies. Front Neurosci 2017; 11:200. [PMID: 28442989 PMCID: PMC5385368 DOI: 10.3389/fnins.2017.00200] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 03/24/2017] [Indexed: 12/20/2022] Open
Abstract
Chorioamnionitis is a major cause of preterm birth and brain injury. Bacterial invasion of the chorion and amnion, and/or the placenta, can lead to a fetal inflammatory response, which in turn has significant adverse consequences for the developing fetal brain. Accordingly, there is a strong causal link between chorioamnionitis, preterm brain injury and the pathogenesis of severe postnatal neurological deficits and cerebral palsy. Currently there are no treatments to protect or repair against brain injury in preterm infants born after pregnancy compromised by intrauterine infection. This review describes the injurious cascade of events in the preterm brain in response to a severe fetal inflammatory event. We will highlight specific periods of increased vulnerability, and the potential effects of therapeutic intervention with cell-based therapies. Many clinical trials are underway to investigate the efficacy of stem cells to treat patients with cerebral palsy. Stem cells, obtained from umbilical cord tissue and cord blood, normally discarded after birth, are emerging as a safe and potentially effective therapy. It is not yet known, however, which stem cell type(s) are the most efficacious for administration to preterm infants to treat brain injury-mediated inflammation. Individual stem cell populations found in cord blood and tissue, such as mesenchymal stem cells (MSCs) and endothelial progenitor cells (EPCs), have a number of potential benefits that may specifically target preterm inflammatory-induced brain injury. MSCs have strong immunomodulatory potential, protecting against global and local neuroinflammatory cascades triggered during infection to the fetus. EPCs have angiogenic and vascular reparative qualities that make them ideal for neurovascular repair. A combined therapy using both MSCs and EPCs to target inflammation and promote angiogenesis for re-establishment of vital vessel networks is a treatment concept that warrants further investigation.
Collapse
Affiliation(s)
- Madison C B Paton
- Neurodevelopment and Neuroprotection Research Group, The Ritchie Centre, Hudson Institute of Medical Research, Monash UniversityClayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash Medical Centre, Monash UniversityClayton, VIC, Australia
| | - Courtney A McDonald
- Neurodevelopment and Neuroprotection Research Group, The Ritchie Centre, Hudson Institute of Medical Research, Monash UniversityClayton, VIC, Australia
| | - Beth J Allison
- Neurodevelopment and Neuroprotection Research Group, The Ritchie Centre, Hudson Institute of Medical Research, Monash UniversityClayton, VIC, Australia
| | - Michael C Fahey
- Neurodevelopment and Neuroprotection Research Group, The Ritchie Centre, Hudson Institute of Medical Research, Monash UniversityClayton, VIC, Australia.,Department of Paediatrics, Monash UniversityClayton, VIC, Australia
| | - Graham Jenkin
- Neurodevelopment and Neuroprotection Research Group, The Ritchie Centre, Hudson Institute of Medical Research, Monash UniversityClayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash Medical Centre, Monash UniversityClayton, VIC, Australia
| | - Suzanne L Miller
- Neurodevelopment and Neuroprotection Research Group, The Ritchie Centre, Hudson Institute of Medical Research, Monash UniversityClayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash Medical Centre, Monash UniversityClayton, VIC, Australia
| |
Collapse
|
50
|
Dubský M, Jirkovská A, Bem R, Nemcová A, Fejfarová V, Jude EB. Cell therapy of critical limb ischemia in diabetic patients - State of art. Diabetes Res Clin Pract 2017; 126:263-271. [PMID: 28288436 DOI: 10.1016/j.diabres.2017.02.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/19/2016] [Accepted: 02/22/2017] [Indexed: 01/15/2023]
Abstract
In this review we report on the state of cell therapy of critical limb ischemia (CLI) with respect to differences between diabetic and non-diabetic patients mainly from the clinical point of view. CLI is the most severe form of peripheral arterial disease and its diagnosis and treatment in diabetic patients is very difficult. The therapeutic effect of standard methods of CLI treatment is only partial - more than one third of diabetic patients are not eligible for standard revascularization; therefore, new therapeutic techniques such as cell therapy have been studied in clinical trials. Presence of CLI in patients with diabetic foot disease is associated with worse clinical outcomes such as lack of healing of foot ulcers, major amputations and premature mortality. A revascularization procedure cannot be successful as the only method in contrast to patients without diabetes, but it must always be part of a complex therapy focused not only on ischemia, but also on treatment of infection, off-loading, metabolic control of diabetes and nutrition, local therapy, etc. Therefore, the main criteria for cell therapy may vary in diabetic patients and non-diabetic persons and results of this treatment method should always be assessed in the context of ensuring comprehensive therapy. This review carries out an analysis of the source of precursor cells, route of administration and brings a brief report of published data with respect to diabetic and non-diabetic patients and our experience with autologous cell therapy of diabetic patients with CLI. Analysis of the studies in terms of diabetes is difficult, because in most of them sub-analysis for diabetic patients is not performed separately. The other problem is that it is not clear if diabetic patients received adequate complex treatment for their foot ulcers which can strongly affect the rate of major amputation as an outcome of CLI treatment.
Collapse
Affiliation(s)
- Michal Dubský
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | | | - Robert Bem
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Andrea Nemcová
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | | | - Edward B Jude
- Diabetes Centre, Tameside Hospital NHS Foundation Trust and University of Manchester, Lancashire, UK
| |
Collapse
|