1
|
Arsava EM, Yilmaz E, Demirel E, Aykac O, Uysal Kocabas Z, Dogan B, Polat M, Ozdemir AO, Gungor L, Topcuoglu MA. Pre-Stroke Frailty Negatively Affects Leptomeningeal Collateral Flow in Proximal Middle Cerebral Artery Occlusion. Cerebrovasc Dis 2024:1-7. [PMID: 39561741 DOI: 10.1159/000542627] [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: 09/06/2024] [Accepted: 11/12/2024] [Indexed: 11/21/2024] Open
Abstract
INTRODUCTION The adequacy of blood flow from the leptomeningeal collaterals is considered one of the most important factors determining the rate of infarct progression and response to acute stroke treatments in the setting of large vessel occlusions. Several patient-related variables, including age, vascular risk factors, and laboratory parameters, have been proposed to explain the interindividual variability of collateral flow among stroke patients. This study aimed to assess how pre-stroke frailty, an aging-related syndrome characterized by a loss in the physiologic reserve of numerous body functions, affected the degree of leptomeningeal collateral flow in the setting of acute ischemic stroke. METHODS A consecutive series of patients presenting with proximal middle cerebral artery occlusion were enrolled in this prospective, multicenter observational study. Collateral flow was determined by the regional leptomeningeal collateral (rLMC) score on admission computed tomography angiography images. Pre-stroke frailty was assessed by the Edmonton Frailty Scale (EFS), based on the information obtained from patients or their next of kin. The relationship between collateral flow and frailty was evaluated by bivariate and multivariate analyses taking into consideration the demographic, clinical, and imaging characteristics of the patients. RESULTS The study population was comprised of 116 patients (median [interquartile range] age 78 [71-84] years; 60% female). The EFS scores were negatively correlated with the rLMC score (r = -0.264; p = 0.004). A vulnerable or frail (EFS ≥6) status before stroke, higher blood pressure levels at admission, having imaging studies performed at an earlier phase after contrast injection, and presenting with thrombi extending to the proximal half of the M1 portion of the middle cerebral artery were significantly related to poor collateral circulation (rLMC score ≤10). After adjustment for potential confounders in multivariable analyses, a vulnerable/frail status was independently associated with poor leptomeningeal collateral flow (OR: 2.97 [95% CI: 1.15-7.69]; p = 0.025). CONCLUSION Our findings highlight that the leptomeningeal collateral flow is also compromised as part of the diminished physiologic reserve characterizing the frailty status in patients with acute ischemic stroke. Future studies are needed to understand how this interplay contributes to the unfavorable clinical outcomes observed in frail patients after stroke.
Collapse
Affiliation(s)
- Ethem Murat Arsava
- Department of Neurology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Ezgi Yilmaz
- Department of Neurology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Ezgi Demirel
- Department of Neurology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Ozlem Aykac
- Department of Neurology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Zehra Uysal Kocabas
- Department of Neurology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Baki Dogan
- Department of Neurology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Murat Polat
- Department of Neurology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Atilla Ozcan Ozdemir
- Department of Neurology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Levent Gungor
- Department of Neurology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | | |
Collapse
|
2
|
Yan J, Tie G, Tutto A, Messina LM. Hypercholesterolemia impairs collateral artery enlargement by ten-eleven translocation 1-dependent hematopoietic stem cell autonomous mechanism in a murine model of limb ischemia. JVS Vasc Sci 2024; 5:100203. [PMID: 38774713 PMCID: PMC11106542 DOI: 10.1016/j.jvssci.2024.100203] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 03/22/2024] [Indexed: 05/24/2024] Open
Abstract
Objective The extent of collateral artery enlargement determines the risk of limb loss due to peripheral arterial disease. Hypercholesterolemia impairs collateral artery enlargement, but the underlying mechanism remains poorly characterized. This study tests the hypothesis that hypercholesterolemia impairs collateral artery enlargement through a ten-eleven translocation 1 (Tet1)-dependent hematopoietic stem cell (HSC)-autonomous mechanism that increases their differentiation into proinflammatory Ly6Chi monocytes and restricts their conversion into proangiogenic Ly6Clow monocytes. Methods To test our hypothesis, we induced limb ischemia and generated chimeric mouse models by transplanting HSCs from either wild-type (WT) mice or hypercholesterolemic mice into lethally irradiated WT recipient mice. Results We found that the lethally irradiated WT recipient mice reconstituted with HSCs from hypercholesterolemic mice displayed lower blood flow recovery and collateral artery enlargement that was nearly identical to that observed in hypercholesterolemic mice, despite the absence of hypercholesterolemia and consistent with an HSC-autonomous mechanism. We showed that hypercholesterolemia impairs collateral artery enlargement by a Tet1-dependent mechanism that increases HSC differentiation toward proinflammatory Ly6Chi monocytes and restricts the conversion of Ly6Chi monocytes into proangiogenic Ly6Clow monocytes. Moreover, Tet1 epigenetically reprograms monocyte gene expression within the HSCs. Restoration of Tet1 expression in HSCs of hypercholesterolemic mice restores WT collateral artery enlargement and blood flow recovery after induction of hindlimb ischemia. Conclusions These results show that hypercholesterolemia impairs collateral artery enlargement by a novel Tet1-dependent HSC-autonomous mechanism that epigenetically reprograms monocyte gene expression within the HSCs.
Collapse
Affiliation(s)
- Jinglian Yan
- Division of Vascular and Endovascular Surgery, University of Massachusetts Medical School, Worcester, MA
| | - Guodong Tie
- Division of Vascular and Endovascular Surgery, University of Massachusetts Medical School, Worcester, MA
| | - Amanda Tutto
- Division of Vascular and Endovascular Surgery, University of Massachusetts Medical School, Worcester, MA
| | - Louis M. Messina
- Division of Vascular and Endovascular Surgery, University of Massachusetts Medical School, Worcester, MA
| |
Collapse
|
3
|
Hakim AH, Brewster L. Dyslipidemia impairs collateral artery formation after hindlimb ischemia: Adding insult to injury. JVS Vasc Sci 2024; 5:100204. [PMID: 38774714 PMCID: PMC11106532 DOI: 10.1016/j.jvssci.2024.100204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2024] Open
Affiliation(s)
- Ali H Hakim
- Division of Vascular Surgery, Department of Surgery, University of Nebraska Medical Center, Omaha, NE
| | - Luke Brewster
- Division of Vascular Surgery, Department of Surgery, Emory University, Atlanta, GA
- Research and Surgical Services, Atlanta VA Medical Center, Decatur, GA
| |
Collapse
|
4
|
Palzkill VR, Tan J, Tice AL, Ferriera LF, Ryan TE. A 6-minute Limb Function Assessment for Therapeutic Testing in Experimental Peripheral Artery Disease Models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.21.586197. [PMID: 38585832 PMCID: PMC10996543 DOI: 10.1101/2024.03.21.586197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Background The translation of promising therapies from pre-clinical models of hindlimb ischemia (HLI) to patients with peripheral artery disease (PAD) has been inadequate. While this failure is multifactorial, primary outcome measures in preclinical HLI models and clinical trials involving patients with PAD are not aligned well. For example, laser Doppler perfusion recovery measured under resting conditions is the most used outcome in HLI studies, whereas clinical trials involving patients with PAD primarily assess walking performance. Here, we sought to develop a 6-min limb function test for preclinical HLI models that assess muscular performance and hemodynamics congruently. Methods We developed an in situ 6-min limb function test that involves repeated isotonic (shortening) contractions performed against a submaximal load. Continuous measurement of muscle blood flow was performed using laser Doppler flowmetry. Quantification of muscle power, work, and perfusion are obtained across the test. To assess the efficacy of this test, we performed HLI via femoral artery ligation on several mouse strains: C57BL6J, BALBc/J, and MCK-PGC1α (muscle-specific overexpression of PGC1α). Additional experiments were performed using an exercise intervention (voluntary wheel running) following HLI. Results The 6-min limb function test was successful at detecting differences in limb function of C57BL6/J and BALBc/J mice subjected to HLI with effect sizes superior to laser Doppler perfusion recovery. C57BL6/J mice randomized to exercise therapy following HLI had smaller decline in muscle power, greater hyperemia, and performed more work across the 6-min limb function test compared to non-exercise controls with HLI. Mice with muscle-specific overexpression of PGC1α had no differences in perfusion recovery in resting conditions, but exhibited greater capillary density, increased muscle mass and absolute force levels, and performed more work across the 6-min limb function test compared to their wildtype littermates without the transgene. Conclusion These results demonstrate the efficacy of the 6-min limb function test to detect differences in the response to HLI across several interventions including where traditional perfusion recovery, capillary density, and muscle strength measures were unable to detect therapeutic differences.
Collapse
Affiliation(s)
- Victoria R. Palzkill
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Jianna Tan
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | | | - Leonardo F. Ferriera
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
- Center for Exercise Science, The University of Florida, Gainesville, FL, USA
- The Myology Institute, The University of Florida, Gainesville, FL, USA
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Terence E. Ryan
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
- Center for Exercise Science, The University of Florida, Gainesville, FL, USA
- The Myology Institute, The University of Florida, Gainesville, FL, USA
| |
Collapse
|
5
|
Biose IJ, Oremosu J, Bhatnagar S, Bix GJ. Promising Cerebral Blood Flow Enhancers in Acute Ischemic Stroke. Transl Stroke Res 2023; 14:863-889. [PMID: 36394792 PMCID: PMC10640530 DOI: 10.1007/s12975-022-01100-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 11/18/2022]
Abstract
Ischemic stroke presents a major global economic and public health burden. Although recent advances in available endovascular therapies show improved functional outcome, a good number of stroke patients are either ineligible or do not have access to these treatments. Also, robust collateral flow during acute ischemic stroke independently predicts the success of endovascular therapies and the outcome of stroke. Hence, adjunctive therapies for cerebral blood flow (CBF) enhancement are urgently needed. A very clear overview of the pial collaterals and the role of genetics are presented in this review. We review available evidence and advancement for potential therapies aimed at improving CBF during acute ischemic stroke. We identified heme-free soluble guanylate cyclase activators; Sanguinate, remote ischemic perconditioning; Fasudil, S1P agonists; and stimulation of the sphenopalatine ganglion as promising potential CBF-enhancing therapeutics requiring further investigation. Additionally, we outline and discuss the critical steps required to advance research strategies for clinically translatable CBF-enhancing agents in the context of acute ischemic stroke models.
Collapse
Affiliation(s)
- Ifechukwude Joachim Biose
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, 131 S. Robertson, Ste 1300, Room 1349, New Orleans, LA, 70112, USA
| | - Jadesola Oremosu
- School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Somya Bhatnagar
- School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Gregory Jaye Bix
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, 131 S. Robertson, Ste 1300, Room 1349, New Orleans, LA, 70112, USA.
- Tulane Brain Institute, Tulane University, New Orleans, LA, 70112, USA.
- Department of Neurology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, 70122, USA.
| |
Collapse
|
6
|
Dönmez-Demir B, Yemisci M, Uruk G, Söylemezoğlu F, Bolbos R, Kazmi S, Dalkara T. Cortical spreading depolarization-induced constriction of penetrating arteries can cause watershed ischemia: A potential mechanism for white matter lesions. J Cereb Blood Flow Metab 2023; 43:1951-1966. [PMID: 37435741 PMCID: PMC10676143 DOI: 10.1177/0271678x231186959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 06/05/2023] [Accepted: 06/05/2023] [Indexed: 07/13/2023]
Abstract
Periventricular white matter lesions (WMLs) are common MRI findings in migraine with aura (MA). Although hemodynamic disadvantages of vascular supply to this region create vulnerability, the pathophysiological mechanisms causing WMLs are unclear. We hypothesize that prolonged oligemia, a consequence of cortical spreading depolarization (CSD) underlying migraine aura, may lead to ischemia/hypoxia at hemodynamically vulnerable watershed zones fed by long penetrating arteries (PAs). For this, we subjected mice to KCl-triggered single or multiple CSDs. We found that post-CSD oligemia was significantly deeper at medial compared to lateral cortical areas, which induced ischemic/hypoxic changes at watershed areas between the MCA/ACA, PCA/anterior choroidal and at the tip of superficial and deep PAs, as detected by histological and MRI examination of brains 2-4 weeks after CSD. BALB-C mice, in which MCA occlusion causes large infarcts due to deficient collaterals, exhibited more profound CSD-induced oligemia and were more vulnerable compared to Swiss mice such that a single CSD was sufficient to induce ischemic lesions at the tip of PAs. In conclusion, CSD-induced prolonged oligemia has potential to cause ischemic/hypoxic injury at hemodynamically vulnerable brain areas, which may be one of the mechanisms underlying WMLs located at the tip of medullary arteries seen in MA patients.
Collapse
Affiliation(s)
- Buket Dönmez-Demir
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| | - Muge Yemisci
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
- Department of Neurology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Gökhan Uruk
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| | - Figen Söylemezoğlu
- Department of Pathology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Radu Bolbos
- CERMEP – imagerie du vivant, Groupement Hospitalier Est, Bron, France
| | - Shams Kazmi
- Biomedical Engineering Department, The University of Texas at Austin, Austin, Texas, USA
| | - Turgay Dalkara
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| |
Collapse
|
7
|
Faber JE, Zhang H, Xenakis JG, Bell TA, Hock P, Pardo-Manuel de Villena F, Ferris MT, Rzechorzek W. Large differences in collateral blood vessel abundance among individuals arise from multiple genetic variants. J Cereb Blood Flow Metab 2023; 43:1983-2004. [PMID: 37572089 PMCID: PMC10676139 DOI: 10.1177/0271678x231194956] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/14/2023]
Abstract
Collateral blood flow varies greatly among humans for reasons that remain unclear, resulting in significant differences in ischemic tissue damage. A similarly large variation has also been found in mice that is caused by genetic background-dependent differences in the extent of collateral formation, termed collaterogenesis-a unique angiogenic process that occurs during development and determines collateral number and diameter in the adult. Previous studies have identified several quantitative trait loci (QTL) linked to this variation. However, understanding has been hampered by the use of closely related inbred strains that do not model the wide genetic variation present in the "outbred" human population. The Collaborative Cross (CC) multiparent mouse genetic reference panel was developed to address this limitation. Herein we measured the number and average diameter of cerebral collaterals in 60 CC strains, their 8 founder strains, 8 F1 crosses of CC strains selected for abundant versus sparse collaterals, and 2 intercross populations created from the latter. Collateral number evidenced 47-fold variation among the 60 CC strains, with 14% having poor, 25% poor-to-intermediate, 47% intermediate-to-good, and 13% good collateral abundance, that was associated with large differences in post-stroke infarct volume. Collateral number in skeletal muscle and intestine of selected high- and low-collateral strains evidenced the same relative abundance as in brain. Genome-wide mapping demonstrated that collateral abundance is a highly polymorphic trait. Subsequent analysis identified: 6 novel QTL circumscribing 28 high-priority candidate genes harboring putative loss-of-function polymorphisms (SNPs) associated with low collateral number; 335 predicted-deleterious SNPs present in their human orthologs; and 32 genes associated with vascular development but lacking protein coding variants. Six additional suggestive QTL (LOD > 4.5) were also identified in CC-wide QTL mapping. This study provides a comprehensive set of candidate genes for future investigations aimed at identifying signaling proteins within the collaterogenesis pathway whose variants potentially underlie genetic-dependent collateral insufficiency in brain and other tissues.
Collapse
Affiliation(s)
- James E Faber
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
- Curriculum in Neuroscience, University of North Carolina, Chapel Hill, NC, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Hua Zhang
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | - James G Xenakis
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Timothy A Bell
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Pablo Hock
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Fernando Pardo-Manuel de Villena
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Martin T Ferris
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Wojciech Rzechorzek
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
8
|
Faber JE, Zhang H, Xenakis JG, Bell TA, Hock P, de Villena FPM, Ferris MT, Rzechorzek W. Large differences in collateral blood vessel abundance among individuals arise from multiple genetic variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.28.542633. [PMID: 37398475 PMCID: PMC10312463 DOI: 10.1101/2023.05.28.542633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Collateral blood flow varies greatly among humans for reasons that remain unclear, resulting in significant differences in ischemic tissue damage. A similarly large variation has also been found in mice that is caused by genetic background-dependent differences in the extent of collateral formation, termed collaterogenesis-a unique angiogenic process that occurs during development and determines collateral number and diameter in the adult. Previous studies have identified several quantitative trait loci (QTL) linked to this variation. However, understanding has been hampered by the use of closely related inbred strains that do not model the wide genetic variation present in the "outbred" human population. The Collaborative Cross (CC) multiparent mouse genetic reference panel was developed to address this limitation. Herein we measured the number and average diameter of cerebral collaterals in 60 CC strains, their 8 founder strains, 8 F1 crosses of CC strains selected for abundant versus sparse collaterals, and 2 intercross populations created from the latter. Collateral number evidenced 47-fold variation among the 60 CC strains, with 14% having poor, 25% poor-to-intermediate, 47% intermediate-to-good, and 13% good collateral abundance, that was associated with large differences in post-stroke infarct volume. Genome-wide mapping demonstrated that collateral abundance is a highly polymorphic trait. Subsequent analysis identified: 6 novel QTL circumscribing 28 high-priority candidate genes harboring putative loss-of-function polymorphisms (SNPs) associated with low collateral number; 335 predicted-deleterious SNPs present in their human orthologs; and 32 genes associated with vascular development but lacking protein coding variants. This study provides a comprehensive set of candidate genes for future investigations aimed at identifying signaling proteins within the collaterogenesis pathway whose variants potentially underlie genetic-dependent collateral insufficiency in brain and other tissues.
Collapse
|
9
|
Seifert K, Heit JJ. Collateral Blood Flow and Ischemic Core Growth. Transl Stroke Res 2023; 14:13-21. [PMID: 35699917 DOI: 10.1007/s12975-022-01051-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 01/31/2023]
Abstract
Treatment of a large vessel occlusion in the acute ischemic stroke setting focuses on vessel recanalization, and endovascular thrombectomy results in favorable outcomes in appropriate candidates. Expeditious treatment is imperative, but patients often present to institutions that do not have neurointerventional surgeons and need to be transferred to a comprehensive stroke center. These treatment delays are common, and it is important to identify factors that mitigate the progression of the ischemic core in order to maximize the preservation of salvageable brain tissue. Collateral blood flow is the strongest factor known to influence ischemic core growth, which includes the input arterial vessels, tissue-level vessels, and venous outflow. Collateral blood flow at these different levels may be imaged by specific imaging techniques that may also predict ischemic core growth during treatment delays and help identify patients who would benefit from transfer and endovascular therapy, as well as identify those patients in whom transfer may be futile. Here we review collateral blood flow and its relationship to ischemic core growth in stroke patients.
Collapse
Affiliation(s)
- Kimberly Seifert
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jeremy J Heit
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA. .,Radiology and Neurosurgery, Stanford University School of Medicine, 453 Quarry Road, Palo Alto, CA, 94304, USA.
| |
Collapse
|
10
|
Chigogidze M, Mantskava M, Sanikidze T, Pagava Z, Urdulashvili T, Tsimakuridze M, Momtselidze N, Sharashidze N. Study of blood rheological parameters and NO in coronary artery disease patients with and without collaterals. Clin Hemorheol Microcirc 2023; 84:193-203. [PMID: 37066905 DOI: 10.3233/ch-231745] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
BACKGROUND In coronary artery disease (CAD), an alternative way of improvement of blood circulation in the ischemic area of the myocardium is coronary collateral circulation. Our study aimed to investigate the rheological parameters of blood and nitric oxide (NO) content in patients with various degrees of collateral development and the likelihood of the influence of blood fluidity on collateral angiogenesis. METHODS We studied patients with stable CAD who underwent elective coronary angiography and a control group with the same mean age. We investigated patients with different degrees of developing collaterals and those without them. In studied patients, the blood plasma viscosity, aggregability, and deformability of erythrocytes, as the main indicators of blood rheology. We recorded content of stable metabolic end products of nitric oxide (NOx). RESULTS Results of the studies showed that in the blood of studied patients with CAD erythrocyte aggregation was increased and NO content decreased compared to the control level; NO content was as lower, as less was the number of developed collaterals was recorded. CONCLUSION In this work, the role of the aggregation ability of erythrocytes and the endothelial origin of NO in the direct and feedback regulatory mechanism of angiogenesis in patients with CAD are discussed.
Collapse
Affiliation(s)
- Maia Chigogidze
- Faculty of Medicine, Ivane Javakhishvili Tbilisi State University. Tbilisi, Georgia
| | - Maia Mantskava
- Laboratory of Rheology and Diagnosti Analytical Services, Ivane Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
- Department of Clinical Research, Multidisciplinary Science High School, Tbilisi, Georgia
| | - Tamar Sanikidze
- Department of Physics, Biophysics, Biomechanics and IT Technologies, Tbilisi State Medical University, Tbilisi, Georgia
| | - Zurab Pagava
- Department of Cardiopulmonary, Bokhua Memorial Cardiovascular Clinic, Tbilisi, Georgia
| | - Tamar Urdulashvili
- Laboratory of Rheology and Diagnosti Analytical Services, Ivane Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
- Department of Clinical Research, Multidisciplinary Science High School, Tbilisi, Georgia
| | - Marina Tsimakuridze
- Department of Nutrition, Aging Medicine, Environmental and Occupational Health, Tbilisi State Medical University, Tbilisi, Georgia
| | - Nana Momtselidze
- Laboratory of Rheology and Diagnosti Analytical Services, Ivane Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
- Department of Medicine, UNIK-Kutaisi University, Kutaisi, Georgia
| | - Nino Sharashidze
- Faculty of Medicine, Ivane Javakhishvili Tbilisi State University. Tbilisi, Georgia
| |
Collapse
|
11
|
Kulkarni R, Andraska E, McEnaney R. Structural Remodeling of the Extracellular Matrix in Arteriogenesis: A Review. Front Cardiovasc Med 2021; 8:761007. [PMID: 34805316 PMCID: PMC8602576 DOI: 10.3389/fcvm.2021.761007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/11/2021] [Indexed: 01/10/2023] Open
Abstract
Lower extremity arterial occlusive disease (AOD) results in significant morbidity and mortality for the population, with up to 10% of patients ultimately requiring amputation. An alternative method for non-surgical revascularization which is yet to be fully understood is the optimization of the body's own natural collateral arterial network in a process known as arteriogenesis. Under conditions of conductance vessel stenosis or occlusion resulting in increased flow, shear forces, and pressure gradients within collaterals, positive remodeling occurs to increase the diameter and capacity of these vessels. The creation of a distal arteriovenous fistula (AVF) will drive increased arteriogenesis as compared to collateral formation with the occlusion of a conductance vessel alone by further increasing flow through these arterioles, demonstrating the capacity for arteriogenesis to form larger, more efficient collaterals beyond what is spontaneously achieved after arterial occlusion. Arteries rely on an extracellular matrix (ECM) composed of elastic fibers and collagens that provide stability under hemodynamic stress, and ECM remodeling is necessary to allow for increased diameter and flow conductance in mature arterial structures. When positive remodeling occurs, digestion of lamella and the internal elastic lamina (IEL) by matrix metalloproteinases (MMPs) and other elastases results in the rearrangement and thinning of elastic structures and may be replaced with disordered elastin synthesis without recovery of elastic function. This results in transmission of wall strain to collagen and potential for aneurysmal degeneration along collateral networks, as is seen in the pancreaticoduodenal artery (PDA) after celiac occlusion and inferior mesenteric artery (IMA) with concurrent celiac and superior mesenteric artery (SMA) occlusions. Further understanding into the development of collaterals is required to both better understand aneurysmal degeneration and optimize collateral formation in AOD.
Collapse
Affiliation(s)
- Rohan Kulkarni
- Division of Vascular Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Elizabeth Andraska
- Division of Vascular Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Ryan McEnaney
- Division of Vascular Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Veterans Affairs Hospitals Pittsburgh Healthcare System, Pittsburgh, PA, United States
| |
Collapse
|
12
|
Determinants of Leptomeningeal Collateral Status Variability in Ischemic Stroke Patients. Can J Neurol Sci 2021; 49:767-773. [PMID: 34585652 DOI: 10.1017/cjn.2021.226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Collateral status is an indicator of a favorable outcome in stroke. Leptomeningeal collaterals provide alternative routes for brain perfusion following an arterial occlusion or flow-limiting stenosis. Using a large cohort of ischemic stroke patients, we examined the relative contribution of various demographic, laboratory, and clinical variables in explaining variability in collateral status. METHODS Patients with acute ischemic stroke in the anterior circulation were enrolled in a multi-center hospital-based observational study. Intracranial occlusions and collateral status were identified and graded using multiphase computed tomography angiography. Based on the percentage of affected territory filled by collateral supply, collaterals were graded as either poor (0-49%), good (50-99%), or optimal (100%). Between-group differences in demographic, laboratory, and clinical factors were explored using ordinal regression models. Further, we explored the contribution of measured variables in explaining variance in collateral status. RESULTS 386 patients with collateral status classified as poor (n = 64), good (n = 125), and optimal (n = 197) were included. Median time from symptom onset to CT was 120 (IQR: 78-246) minutes. In final multivariable model, male sex (OR 1.9, 95% CIs [1.2, 2.9], p = 0.005) and leukocytosis (OR 1.1, 95% CIs [1.1, 1.2], p = 0.001) were associated with poor collaterals. Measured variables only explained 44.8-53.0% of the observed between-patient variance in collaterals. CONCLUSION Male sex and leukocytosis are associated with poorer collaterals. Nearly half of the variance in collateral flow remains unexplained and could be in part due to genetic differences.
Collapse
|
13
|
PHACTR1 genetic variability is not critical in small vessel ischemic disease patients and PcomA recruitment in C57BL/6J mice. Sci Rep 2021; 11:6072. [PMID: 33727568 PMCID: PMC7966789 DOI: 10.1038/s41598-021-84919-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 02/04/2021] [Indexed: 11/29/2022] Open
Abstract
Recently, several genome-wide association studies identified PHACTR1 as key locus for five diverse vascular disorders: coronary artery disease, migraine, fibromuscular dysplasia, cervical artery dissection and hypertension. Although these represent significant risk factors or comorbidities for ischemic stroke, PHACTR1 role in brain small vessel ischemic disease and ischemic stroke most important survival mechanism, such as the recruitment of brain collateral arteries like posterior communicating arteries (PcomAs), remains unknown. Therefore, we applied exome and genome sequencing in a multi-ethnic cohort of 180 early-onset independent familial and apparently sporadic brain small vessel ischemic disease and CADASIL-like Caucasian patients from US, Portugal, Finland, Serbia and Turkey and in 2 C57BL/6J stroke mouse models (bilateral common carotid artery stenosis [BCCAS] and middle cerebral artery occlusion [MCAO]), characterized by different degrees of PcomAs patency. We report 3 very rare coding variants in the small vessel ischemic disease-CADASIL-like cohort (p.Glu198Gln, p.Arg204Gly, p.Val251Leu) and a stop-gain mutation (p.Gln273*) in one MCAO mouse. These coding variants do not cluster in PHACTR1 known pathogenic domains and are not likely to play a critical role in small vessel ischemic disease or brain collateral circulation. We also exclude the possibility that copy number variants (CNVs) or a variant enrichment in Phactr1 may be associated with PcomA recruitment in BCCAS mice or linked to diverse vascular traits (cerebral blood flow pre-surgery, PcomA size, leptomeningeal microcollateral length and junction density during brain hypoperfusion) in C57BL/6J mice, respectively. Genetic variability in PHACTR1 is not likely to be a common susceptibility factor influencing small vessel ischemic disease in patients and PcomA recruitment in C57BL/6J mice. Nonetheless, rare variants in PHACTR1 RPEL domains may influence the stroke outcome and are worth investigating in a larger cohort of small vessel ischemic disease patients, different ischemic stroke subtypes and with functional studies.
Collapse
|
14
|
Hemodynamics in acute stroke: Cerebral and cardiac complications. HANDBOOK OF CLINICAL NEUROLOGY 2021; 177:295-317. [PMID: 33632449 DOI: 10.1016/b978-0-12-819814-8.00015-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Hemodynamics is the study of blood flow, where parameters have been defined to quantify blood flow and the relationship with systemic circulatory changes. Understanding these perfusion parameters, the relationship between different blood flow variables and the implications for ischemic injury are outlined in the ensuing discussion. This chapter focuses on the hemodynamic changes that occur in ischemic stroke, and their contribution to ischemic stroke pathophysiology. We discuss the interaction between cardiovascular response and hemodynamic changes in stroke. Studying hemodynamic changes has a key role in stroke prevention, therapeutic implications and prognostic importance in acute ischemic stroke: preexisting hemodynamic and autoregulatory impairments predict the occurrence of stroke. Hemodynamic failure predisposes to the formation of thromboemboli and accelerates infarction due to impairing compensatory mechanisms. In ischemic stroke involving occlusion of a large vessel, persistent collateral circulation leads to preservation of ischemic penumbra and therefore justifying endovascular thrombectomy. Following thrombectomy, impaired autoregulation may lead to reperfusion injury and hemorrhage.
Collapse
|
15
|
Diabetes Mellitus/Poststroke Hyperglycemia: a Detrimental Factor for tPA Thrombolytic Stroke Therapy. Transl Stroke Res 2020; 12:416-427. [PMID: 33140258 DOI: 10.1007/s12975-020-00872-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/17/2022]
Abstract
Intravenous administration of tissue-type plasminogen activator (IV tPA) therapy has long been considered a mainstay in ischemic stroke management. However, patients respond to IV tPA therapy unequally with some subsets of patients having worsened outcomes after treatment. In particular, diabetes mellitus (DM) is recognized as a clinically important vascular comorbidity that leads to lower recanalization rates and increased risks of hemorrhagic transformation (HT). In this short-review, we summarize the recent advances in understanding of the underlying mechanisms involved in post-IV tPA worsening of outcome in diabetic stroke. Potential pathologic factors that are related to the suboptimal tPA recanalization in diabetic stroke include higher plasma plasminogen activator inhibitor (PAI)-1 level, diabetic atherogenic vascular damage, glycation of the tPA receptor annexin A2, and alterations in fibrin clot density. While factors contributing to the exacerbation of HT in diabetic stroke include hyperglycemia, vascular oxidative stress, and inflammation, tPA neurovascular toxicity and imbalance in extracellular proteolysis are discussed. Besides, impaired collaterals in DM also compromise the efficacy of IV tPA therapy. Additionally, several tPA combination approaches developed from experimental studies that may help to optimize IV tPA therapy are also briefly summarized. In summary, more research efforts are needed to improve the safety and efficacy of IV tPA therapy in ischemic stroke patients with DM/poststroke hyperglycemia.
Collapse
|
16
|
Zhang H, Rzechorzek W, Aghajanian A, Faber JE. Hypoxia induces de novo formation of cerebral collaterals and lessens the severity of ischemic stroke. J Cereb Blood Flow Metab 2020; 40:1806-1822. [PMID: 32423327 PMCID: PMC7430105 DOI: 10.1177/0271678x20924107] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Pial collaterals provide protection in stroke. Evidence suggests their formation late during gestation (collaterogenesis) is driven by reduced oxygen levels in the cerebral watersheds. The purpose of this study was to determine if collaterogenesis can be re-activated in the adult to induce formation of additional collaterals ("neo-collateral formation", NCF). Mice were gradually acclimated to reduced inspired oxygen (FIO2) and maintained at 12, 10, 8.5 or 7% for two-to-eight weeks. Hypoxemia induced "dose"-dependent NCF and remodeling of native collaterals, and decreased infarct volume after permanent MCA occlusion. In contrast, no formation occurred of addition collateral-like intra-tree anastomoses, PComs, or branches within the MCA tree. Hypoxic NCF, remodeling and infarct protection were durable, i.e. retained for at least six weeks after return to normoxia. Hypoxia increased expression of Hif2α, Vegfa, Rabep2, Angpt2, Tie2 and Cxcr4. Neo-collateral formation was abolished in mice lacking Rabep2, a novel gene involved in VEGFA→Flk1 signaling and required for formation of collaterals during development, and inhibited by knockdown of Vegfa, Flk1 and Cxcr4. Rabep2-dependent NCF was also induced by permanent MCA occlusion. This is the first report that hypoxia induces new pial collaterals to form. Hypoxia- and occlusion-induced neo-collateral formation provide models to study collaterogenesis in the adult.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Cell Biology and Physiology, McAllister Heart Institute, Curriculum in Neurobiology, University of North Carolina at Chapel Hill, NC, USA
| | - Wojciech Rzechorzek
- Department of Cell Biology and Physiology, McAllister Heart Institute, Curriculum in Neurobiology, University of North Carolina at Chapel Hill, NC, USA
| | - Amir Aghajanian
- Department of Cell Biology and Physiology, McAllister Heart Institute, Curriculum in Neurobiology, University of North Carolina at Chapel Hill, NC, USA
| | - James E Faber
- Department of Cell Biology and Physiology, McAllister Heart Institute, Curriculum in Neurobiology, University of North Carolina at Chapel Hill, NC, USA
| |
Collapse
|
17
|
Ma J, Ma Y, Shuaib A, Winship IR. Impaired Collateral Flow in Pial Arterioles of Aged Rats During Ischemic Stroke. Transl Stroke Res 2020; 11:243-253. [PMID: 31203565 PMCID: PMC7067739 DOI: 10.1007/s12975-019-00710-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/02/2019] [Accepted: 06/05/2019] [Indexed: 02/05/2023]
Abstract
Cerebral collateral circulation and age are critical factors in determining outcome from acute ischemic stroke. Aging may lead to rarefaction of cerebral collaterals, and thereby accelerate ischemic injury by reducing penumbral blood flow. Dynamic changes in pial collaterals after onset of cerebral ischemia may vary with age but have not been extensively studied. Here, laser speckle contrast imaging (LSCI) and two-photon laser scanning microscopy (TPLSM) were combined to monitor cerebral pial collaterals between the anterior cerebral artery (ACA) and the middle cerebral artery (MCA) in young adult and aged male Sprague Dawley rats during distal middle cerebral artery occlusion (dMCAo). Histological analysis showed that aged rats had significantly greater volumes of ischemic damage than young rats. LSCI showed that cerebral collateral perfusion declined over time after stroke in aged and young rats, and that this decline was significantly greater in aged rats. TPLSM demonstrated that pial arterioles narrowed faster after dMCAo in aged rats compared to young adult rats. Notably, while arteriole vessel narrowing was comparable 4.5 h after ischemic onset in aged and young adult rats, red blood cell velocity was stable in young adults but declined over time in aged rats. Overall, red blood cell flux through pial arterioles was significantly reduced at all time-points after 90 min post-dMCAo in aged rats relative to young adult rats. Thus, collateral failure is more severe in aged rats with significantly impaired pial collateral dynamics (reduced diameter, red blood cell velocity, and red blood cell flux) relative to young adult rats.
Collapse
Affiliation(s)
- Junqiang Ma
- Neurochemical Research Unit, Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, 12-127 Clinical Sciences Building, Edmonton, AB, T6G 2R3, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Yonglie Ma
- Neurochemical Research Unit, Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, 12-127 Clinical Sciences Building, Edmonton, AB, T6G 2R3, Canada
| | - Ashfaq Shuaib
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Division of Neurology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Ian R Winship
- Neurochemical Research Unit, Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, 12-127 Clinical Sciences Building, Edmonton, AB, T6G 2R3, Canada.
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
18
|
Foddis M, Winek K, Bentele K, Mueller S, Blumenau S, Reichhart N N, Crespo-Garcia S, Harnett D, Ivanov A, Meisel A, Joussen A, Strauss O, Beule D, Dirnagl U, Sassi C. An exploratory investigation of brain collateral circulation plasticity after cerebral ischemia in two experimental C57BL/6 mouse models. J Cereb Blood Flow Metab 2020; 40:276-287. [PMID: 31549895 PMCID: PMC7370619 DOI: 10.1177/0271678x19827251] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Brain collateral circulation is an essential compensatory mechanism in response to acute brain ischemia. To study the temporal evolution of brain macro and microcollateral recruitment and their reciprocal interactions in response to different ischemic conditions, we applied a combination of complementary techniques (T2-weighted magnetic resonance imaging [MRI], time of flight [TOF] angiography [MRA], cerebral blood flow [CBF] imaging and histology) in two different mouse models. Hypoperfusion was either induced by permanent bilateral common carotid artery stenosis (BCCAS) or 60-min transient unilateral middle cerebral artery occlusion (MCAO). In both models, collateralization is a very dynamic phenomenon with a global effect affecting both hemispheres. Patency of ipsilateral posterior communicating artery (PcomA) represents the main variable survival mechanism and the main determinant of stroke lesion volume and recovery in MCAO, whereas the promptness of external carotid artery retrograde flow recruitment together with PcomA patency, critically influence survival, brain ischemic lesion volume and retinopathy in BCCAS mice. Finally, different ischemic gradients shape microcollateral density and size.
Collapse
Affiliation(s)
- Marco Foddis
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Katarzyna Winek
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Kajetan Bentele
- Berlin Institute of Health, BIH, Unit Bioinformatics, Berlin, Germany
| | - Susanne Mueller
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Charité - Universitätsmedizin Berlin, NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Berlin, Germany
| | - Sonja Blumenau
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Nadine Reichhart N
- Department of Ophthalmology, Experimental Ophthalmology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Sergio Crespo-Garcia
- Department of Ophthalmology, Experimental Ophthalmology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Dermot Harnett
- Berlin Institute of Health, BIH, Unit Bioinformatics, Berlin, Germany
| | - Andranik Ivanov
- Berlin Institute of Health, BIH, Unit Bioinformatics, Berlin, Germany
| | - Andreas Meisel
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Antonia Joussen
- Department of Ophthalmology, Experimental Ophthalmology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Olaf Strauss
- Department of Ophthalmology, Experimental Ophthalmology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Dieter Beule
- Berlin Institute of Health, BIH, Unit Bioinformatics, Berlin, Germany
| | - Ulrich Dirnagl
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,QUEST Center for Transforming Biomedical Research, Berlin Institute of Health (BIH), Berlin, Germany
| | - Celeste Sassi
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
19
|
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
|
20
|
Aref Z, de Vries MR, Quax PHA. Variations in Surgical Procedures for Inducing Hind Limb Ischemia in Mice and the Impact of These Variations on Neovascularization Assessment. Int J Mol Sci 2019; 20:ijms20153704. [PMID: 31362356 PMCID: PMC6696155 DOI: 10.3390/ijms20153704] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/22/2019] [Accepted: 07/25/2019] [Indexed: 12/16/2022] Open
Abstract
Mouse hind limb ischemia is the most common used preclinical model for peripheral arterial disease and critical limb ischemia. This model is used to investigate the mechanisms of neovascularization and to develop new therapeutic agents. The literature shows many variations in the model, including the method of occlusion, the number of occlusions, and the position at which the occlusions are made to induce hind limb ischemia. Furthermore, predefined end points and the histopathological and radiological analysis vary. These differences hamper the correlation of results between different studies. In this review, variations in surgical methods of inducing hind limb ischemia in mice are described, and the consequences of these variations on perfusion restoration and vascular remodeling are discussed. This study aims at providing the reader with a comprehensive overview of the methods so far described, and proposing uniformity in research of hind limb ischemia in a mouse model.
Collapse
Affiliation(s)
- Zeen Aref
- Department of Surgery, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Margreet R de Vries
- Department of Surgery, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Paul H A Quax
- Department of Surgery, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.
| |
Collapse
|
21
|
Piedade GS, Schirmer CM, Goren O, Zhang H, Aghajanian A, Faber JE, Griessenauer CJ. Cerebral Collateral Circulation: A Review in the Context of Ischemic Stroke and Mechanical Thrombectomy. World Neurosurg 2019; 122:33-42. [PMID: 30342266 DOI: 10.1016/j.wneu.2018.10.066] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/06/2018] [Accepted: 10/08/2018] [Indexed: 01/28/2023]
Abstract
The pial (leptomenigeal) collateral circulation is a key determinant of functional outcome after mechanical thrombectomy after large-vessel ischemic stroke. Patients with good collateral blood flow benefit up to 24 hours after stroke onset, whereas those with poor collateral flow evidence less or no benefit. However, clues to why collateral flow varies so widely among patients have remained elusive. Recent findings in animal studies, which are currently being tested for confirmation in humans, have found that naturally occurring variants of a novel "collateral gene," Rabep2, result in large differences in the extent of anatomic collaterals and thus blood flow and infarct size in mice after stroke. The comprehension of collagerogenesis in humans and the evaluation of collateral status could aid in identifying patients who will benefit not only from mechanical thrombectomy in the extended time window but also from any reperfusion strategy. We performed a literature review focused on radiographic, clinical, and genetic aspects of the collateral circulation.
Collapse
Affiliation(s)
- Guilherme Santos Piedade
- Department of Neurosurgery, Geisinger, Pennsylvania, USA; Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany
| | | | - Oded Goren
- Department of Neurosurgery, Geisinger, Pennsylvania, USA
| | - Hua Zhang
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Amir Aghajanian
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - James E Faber
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Christoph J Griessenauer
- Department of Neurosurgery, Geisinger, Pennsylvania, USA; Research Institute of Neurointervention, Paracelsus Medical University, Salzburg, Austria; Department of Neurosurgery, Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
22
|
The pial vasculature of the mouse develops according to a sensory-independent program. Sci Rep 2018; 8:9860. [PMID: 29959346 PMCID: PMC6026131 DOI: 10.1038/s41598-018-27910-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 06/12/2018] [Indexed: 12/15/2022] Open
Abstract
The cerebral vasculature is organized to supply the brain’s metabolic needs. Sensory deprivation during the early postnatal period causes altered neural activity and lower metabolic demand. Neural activity is instructional for some aspects of vascular development, and deprivation causes changes in capillary density in the deprived brain region. However, it is not known if the pial arteriole network, which contains many leptomeningeal anastomoses (LMAs) that endow the network with redundancy against occlusions, is also affected by sensory deprivation. We quantified the effects of early-life sensory deprivation via whisker plucking on the densities of LMAs and penetrating arterioles (PAs) in anatomically-identified primary sensory regions (vibrissae cortex, forelimb/hindlimb cortex, visual cortex and auditory cortex) in mice. We found that the densities of penetrating arterioles were the same across cortical regions, though the hindlimb representation had a higher density of LMAs than other sensory regions. We found that the densities of PAs and LMAs, as well as quantitative measures of network topology, were not affected by sensory deprivation. Our results show that the postnatal development of the pial arterial network is robust to sensory deprivation.
Collapse
|
23
|
赵 庆, 王 刚, 肖 浩, 冯 文, 张 国, 李 明, 廖 永, 温 运, 漆 松. [Characteristics of collateral circulation in adult moyamoya disease based on modified Suzuki staging]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:483-489. [PMID: 29735452 PMCID: PMC6765649 DOI: 10.3969/j.issn.1673-4254.2018.04.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Indexed: 06/08/2023]
Abstract
OBJECTIVE To investigate the characteristics of collateral circulation in adult moyamoya disease (MMD). METHODS The clinical data were collected from all adult patients with MMD undergoing digital subtractive angiography (DSA) in our department from 2006 to 2016. Based on the imaging findings, the patients were divided into ischemia group and bleeding group. A double-blind analysis was conducted of the CT or magnetic resonance imaging findings and the severity of the disease was graded using the modified Suzuki score (mSS). We classified the anastomotic networks in MMD into the superficial meningeal type and deep parenchymal type. The superficial meningeal type was further classified into the leptomeningeal and the durocortical networks, and the deep parenchymal networks into subependymal networks and the inner striatal and inner thalamic networks. RESULTS No significant difference was found in the distribution of mSS scores between the hemorrhage group and the ischemic group (Χ2=5.812, v=5, P=0.325), but the posterior communicating artery and internal carotid artery diameter ratio (Pcom/ICA ratio) was significantly greater in the hemorrhage group (t=2.119, v=108, P=0.036). The Pcom/ICA ratio differed significantly among the groups with different mSS scores (f=8.924, P=0.00), higher in groups with mSS scores of 3, 4 and 5. The incidence of anterior choroidal artery dilation differed significantly between hemorrhage and ischemic groups (Χ2=11.79, P=0.001). The incidences of durocortical networks (Χ2=0.327, P=0.567) and subependymal networks (Χ2=0.011, P=0.917) were comparable between hemorrhage group and ischemic groups, but the incidence of leptomeningeal networks (P=0.018) and inner striatal and inner thalamic networks (Χ2=7.551, P=0.006) differed significantly between the two groups. CONCLUSION The collateral circulation vascular system is an important component of cerebral blood flow in MMD patients and varies from patient to patient. Patients with MMD exhibit increased Pcom/ICA ratio with abnormal expansion of the anterior choroidal artery, and the leptomeningeal networks and the inner striatal and inner thalamic networks are independent risk factors for cerebral hemorrhage.
Collapse
Affiliation(s)
- 庆顺 赵
- />南方医科大学南方医院神经外科,广东 广州 510515Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 刚 王
- />南方医科大学南方医院神经外科,广东 广州 510515Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 浩江 肖
- />南方医科大学南方医院神经外科,广东 广州 510515Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 文峰 冯
- />南方医科大学南方医院神经外科,广东 广州 510515Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 国忠 张
- />南方医科大学南方医院神经外科,广东 广州 510515Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 明洲 李
- />南方医科大学南方医院神经外科,广东 广州 510515Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 永鸿 廖
- />南方医科大学南方医院神经外科,广东 广州 510515Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 运宇 温
- />南方医科大学南方医院神经外科,广东 广州 510515Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 松涛 漆
- />南方医科大学南方医院神经外科,广东 广州 510515Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
24
|
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
|
25
|
Coucha M, Abdelsaid M, Ward R, Abdul Y, Ergul A. Impact of Metabolic Diseases on Cerebral Circulation: Structural and Functional Consequences. Compr Physiol 2018; 8:773-799. [PMID: 29687902 DOI: 10.1002/cphy.c170019] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Metabolic diseases including obesity, insulin resistance, and diabetes have profound effects on cerebral circulation. These diseases not only affect the architecture of cerebral blood arteries causing adverse remodeling, pathological neovascularization, and vasoregression but also alter the physiology of blood vessels resulting in compromised myogenic reactivity, neurovascular uncoupling, and endothelial dysfunction. Coupled with the disruption of blood brain barrier (BBB) integrity, changes in blood flow and microbleeds into the brain rapidly occur. This overview is organized into sections describing cerebrovascular architecture, physiology, and BBB in these diseases. In each section, we review these properties starting with larger arteries moving into smaller vessels. Where information is available, we review in the order of obesity, insulin resistance, and diabetes. We also tried to include information on biological variables such as the sex of the animal models noted since most of the information summarized was obtained using male animals. © 2018 American Physiological Society. Compr Physiol 8:773-799, 2018.
Collapse
Affiliation(s)
- Maha Coucha
- South University, School of Pharmacy, Savannah, Georgia, USA
| | | | - Rebecca Ward
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Yasir Abdul
- Charlie Norwood VA Medical Center, Augusta, Georgia, USA.,Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Adviye Ergul
- Charlie Norwood VA Medical Center, Augusta, Georgia, USA.,Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
26
|
Schmidt CA, Amorese AJ, Ryan TE, Goldberg EJ, Tarpey MD, Green TD, Karnekar RR, Yamaguchi DJ, Spangenburg EE, McClung JM. Strain-Dependent Variation in Acute Ischemic Muscle Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1246-1262. [PMID: 29454751 DOI: 10.1016/j.ajpath.2018.01.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 12/15/2017] [Accepted: 01/11/2018] [Indexed: 12/19/2022]
Abstract
Limited efficacy of clinical interventions for peripheral arterial disease necessitates a better understanding of the environmental and genetic determinants of tissue pathology. Existing research has largely ignored the early skeletal muscle injury response during hind limb ischemia (HLI). We compared the hind limb muscle response, after 6 hours of ischemia, in two mouse strains that differ dramatically in their postischemic extended recovery: C57BL/6J and BALB/cJ. Perfusion, measured by laser Doppler and normalized to the control limb, differed only slightly between strains after HLI (<12% across all measures). Similar (<10%) effect sizes in lectin-perfused vessel area and no differences in tissue oxygen saturation measured by reflectance spectroscopy were also found. Muscles from both strains were functionally impaired after HLI, but greater muscle necrosis and loss of dystrophin-positive immunostaining were observed in BALB/cJ muscle compared with C57BL/6J. Muscle cell-specific dystrophin loss and reduced viability were also detected in additional models of ischemia that were independent of residual perfusion differences. Our results indicate that factors other than the completeness of ischemia alone (ie, background genetics) influence the magnitude of acute ischemic muscle injury. These findings may have implications for future development of therapeutic interventions for limb ischemia and for understanding the phasic etiology of chronic and acute ischemic muscle pathophysiology.
Collapse
Affiliation(s)
- Cameron A Schmidt
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina
| | - Adam J Amorese
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina
| | - Terence E Ryan
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina
| | - Emma J Goldberg
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina
| | - Michael D Tarpey
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina
| | - Thomas D Green
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina
| | - Reema R Karnekar
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina
| | - Dean J Yamaguchi
- Department of Cardiovascular Sciences, East Carolina University, Greenville, North Carolina; Division of Vascular Surgery, East Carolina University, Greenville, North Carolina
| | - Espen E Spangenburg
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina
| | - Joseph M McClung
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina; Department of Cardiovascular Sciences, East Carolina University, Greenville, North Carolina.
| |
Collapse
|
27
|
Tong LS, Guo ZN, Ou YB, Yu YN, Zhang XC, Tang J, Zhang JH, Lou M. Cerebral venous collaterals: A new fort for fighting ischemic stroke? Prog Neurobiol 2017; 163-164:172-193. [PMID: 29199136 DOI: 10.1016/j.pneurobio.2017.11.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/03/2017] [Accepted: 11/28/2017] [Indexed: 12/13/2022]
Abstract
Stroke therapy has entered a new era highlighted by the use of endovascular therapy in addition to intravenous thrombolysis. However, the efficacy of current therapeutic regimens might be reduced by their associated adverse events. For example, over-reperfusion and futile recanalization may lead to large infarct, brain swelling, hemorrhagic complication and neurological deterioration. The traditional pathophysiological understanding on ischemic stroke can hardly address these occurrences. Accumulating evidence suggests that a functional cerebral venous drainage, the major blood reservoir and drainage system in brain, may be as critical as arterial infusion for stroke evolution and clinical sequelae. Further exploration of the multi-faceted function of cerebral venous system may add new implications for stroke outcome prediction and future therapeutic decision-making. In this review, we emphasize the anatomical and functional characteristics of the cerebral venous system and illustrate its necessity in facilitating the arterial infusion and maintaining the cerebral perfusion in the pathological stroke content. We then summarize the recent critical clinical studies that underscore the associations between cerebral venous collateral and outcome of ischemic stroke with advanced imaging techniques. A novel three-level venous system classification is proposed to demonstrate the distinct characteristics of venous collaterals in the setting of ischemic stroke. Finally, we discuss the current directions for assessment of cerebral venous collaterals and provide future challenges and opportunities for therapeutic strategies in the light of these new concepts.
Collapse
Affiliation(s)
- Lu-Sha Tong
- Department of Neurology, The 2nd Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China; Departments of Physiology, Loma Linda University, School of Medicine, CA, USA
| | - Zhen-Ni Guo
- Department of Neurology, The First Affiliated Hospital of Jilin University, Changchun, China; Departments of Physiology, Loma Linda University, School of Medicine, CA, USA
| | - Yi-Bo Ou
- Department of Neurosurgery, Tong-ji Hospital, Wuhan, China; Departments of Physiology, Loma Linda University, School of Medicine, CA, USA
| | - Yan-Nan Yu
- Department of Neurology, The 2nd Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Xiao-Cheng Zhang
- Department of Neurology, The 2nd Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Jiping Tang
- Department of Anesthesiology, Loma Linda University, School of Medicine, CA, USA
| | - John H Zhang
- Departments of Physiology, Loma Linda University, School of Medicine, CA, USA.
| | - Min Lou
- Department of Neurology, The 2nd Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China.
| |
Collapse
|
28
|
Hancock AM, Frostig RD. Testing the effects of sensory stimulation as a collateral-based therapeutic for ischemic stroke in C57BL/6J and CD1 mouse strains. PLoS One 2017; 12:e0183909. [PMID: 28902897 PMCID: PMC5597132 DOI: 10.1371/journal.pone.0183909] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 08/14/2017] [Indexed: 11/19/2022] Open
Abstract
Utilizing a rat model of ischemic stroke, we have previously shown that sensory stimulation can completely protect rats from impending ischemic damage of cortex if this treatment is delivered within the first two hours post-permanent middle cerebral artery occlusion (pMCAo). The current study sought to extend our findings in rats to mice, which would allow new avenues of research not available in rats. Thus, young adult C57BL/6J and CD1 mice were tested for protection from ischemic stroke with the same protective sensory stimulation-based treatment. Cortical activity and blood flow were assessed with intrinsic signal optical imaging (ISOI) and laser speckle imaging (LSI), respectively, and histological analysis (TTC) was performed at the completion of the experiments. Standing in stark contrast to the positive results observed in rats, in both strains we found that there were no differences between treated and untreated mice at 24 hours post-pMCAo in terms of infarct volume, negative functional imaging results, and major reduction in retrograde collateral blood flow as compared to pre-pMCAo baseline and surgical controls. Also, no differences were found between the strains in terms of theses variables. Potential reasons for the differences between rats and mice are discussed.
Collapse
Affiliation(s)
- Aneeka M. Hancock
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, California, United States of America
| | - Ron D. Frostig
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, California, United States of America
- Center for the Neurobiology of Learning and Memory, University of California Irvine, Irvine, California, United States of America
- Department of Biomedical Engineering, University of California Irvine, Irvine, California, United States of America
| |
Collapse
|
29
|
Rocha M, Jovin TG. Fast Versus Slow Progressors of Infarct Growth in Large Vessel Occlusion Stroke. Stroke 2017; 48:2621-2627. [DOI: 10.1161/strokeaha.117.017673] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 07/13/2017] [Accepted: 07/20/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Marcelo Rocha
- From the Department of Neurology (M.R., T.G.J.) and Department of Neurosurgery (T.G.J.), Stroke Institute, University of Pittsburgh Medical Center, PA
| | - Tudor G. Jovin
- From the Department of Neurology (M.R., T.G.J.) and Department of Neurosurgery (T.G.J.), Stroke Institute, University of Pittsburgh Medical Center, PA
| |
Collapse
|
30
|
Ginsberg MD. The cerebral collateral circulation: Relevance to pathophysiology and treatment of stroke. Neuropharmacology 2017; 134:280-292. [PMID: 28801174 DOI: 10.1016/j.neuropharm.2017.08.003] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 07/28/2017] [Accepted: 08/06/2017] [Indexed: 12/29/2022]
Abstract
The brain's collateral circulation consists of arterial anastomotic channels capable of providing nutrient perfusion to brain regions whose normal sources of flow have become compromised, as occurs in acute ischemic stroke. Modern CT-based neuroimaging is capable of providing detailed information as to collateral extent and sufficiency and is complemented by magnetic resonance-based methods. In the present era of standard-of-care IV thrombolysis for acute ischemic stroke, and following the recent therapeutic successes of randomized clinical trials of acute endovascular intervention, the sufficiency of the collateral circulation has been convincingly established as a key factor influencing the likelihood of successful reperfusion and favorable clinical outcome. This article reviews the features of the brain's collateral circulation; methods for its evaluation in the acute clinical setting; the relevance of collateral circulation to prognosis in acute ischemic stroke; the specific insights into the collateral circulation learned from recent trials of endovascular intervention; and the major influence of genetic factors. Finally, we emphasize the need to develop therapeutic approaches to augment collateral perfusion as an adjunctive strategy to be employed along with, or prior to, thrombolysis and endovascular interventions, and we highlight the possible potential of inhaled nitric oxide, albumin, and other approaches. This article is part of the Special Issue entitled 'Cerebral Ischemia'.
Collapse
Affiliation(s)
- Myron D Ginsberg
- Department of Neurology, University of Miami Miller School of Medicine, Clinical Research Center, Room 1331, 1120 NW 14th Street, Miami, FL 33136, USA.
| |
Collapse
|
31
|
McClung JM, McCord TJ, Ryan TE, Schmidt CA, Green TD, Southerland KW, Reinardy JL, Mueller SB, Venkatraman TN, Lascola CD, Keum S, Marchuk DA, Spangenburg EE, Dokun A, Annex BH, Kontos CD. BAG3 (Bcl-2-Associated Athanogene-3) Coding Variant in Mice Determines Susceptibility to Ischemic Limb Muscle Myopathy by Directing Autophagy. Circulation 2017; 136:281-296. [PMID: 28442482 PMCID: PMC5537727 DOI: 10.1161/circulationaha.116.024873] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 04/14/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND Critical limb ischemia is a manifestation of peripheral artery disease that carries significant mortality and morbidity risk in humans, although its genetic determinants remain largely unknown. We previously discovered 2 overlapping quantitative trait loci in mice, Lsq-1 and Civq-1, that affected limb muscle survival and stroke volume after femoral artery or middle cerebral artery ligation, respectively. Here, we report that a Bag3 variant (Ile81Met) segregates with tissue protection from hind-limb ischemia. METHODS We treated mice with either adeno-associated viruses encoding a control (green fluorescent protein) or 2 BAG3 (Bcl-2-associated athanogene-3) variants, namely Met81 or Ile81, and subjected the mice to hind-limb ischemia. RESULTS We found that the BAG3 Ile81Met variant in the C57BL/6 (BL6) mouse background segregates with protection from tissue necrosis in a shorter congenic fragment of Lsq-1 (C.B6-Lsq1-3). BALB/c mice treated with adeno-associated virus encoding the BL6 BAG3 variant (Ile81; n=25) displayed reduced limb-tissue necrosis and increased limb tissue perfusion compared with Met81- (n=25) or green fluorescent protein- (n=29) expressing animals. BAG3Ile81, but not BAG3Met81, improved ischemic muscle myopathy and muscle precursor cell differentiation and improved muscle regeneration in a separate, toxin-induced model of injury. Systemic injection of adeno-associated virus-BAG3Ile81 (n=9), but not BAG3Met81 (n=10) or green fluorescent protein (n=5), improved ischemic limb blood flow and limb muscle histology and restored muscle function (force production). Compared with BAG3Met81, BAG3Ile81 displayed improved binding to the small heat shock protein (HspB8) in ischemic skeletal muscle cells and enhanced ischemic muscle autophagic flux. CONCLUSIONS Taken together, our data demonstrate that genetic variation in BAG3 plays an important role in the prevention of ischemic tissue necrosis. These results highlight a pathway that preserves tissue survival and muscle function in the setting of ischemia.
Collapse
Affiliation(s)
- Joseph M McClung
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville.
| | - Timothy J McCord
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Terence E Ryan
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Cameron A Schmidt
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Tom D Green
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Kevin W Southerland
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Jessica L Reinardy
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Sarah B Mueller
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Talaignair N Venkatraman
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Christopher D Lascola
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Sehoon Keum
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Douglas A Marchuk
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Espen E Spangenburg
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Ayotunde Dokun
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Brian H Annex
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Christopher D Kontos
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| |
Collapse
|
32
|
de Vries MR, Peters EAB, Quax PHA, Nossent AY. von Willebrand factor deficiency leads to impaired blood flow recovery after ischaemia in mice. Thromb Haemost 2017; 117:1412-1419. [PMID: 28382367 DOI: 10.1160/th16-12-0957] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/25/2017] [Indexed: 02/01/2023]
Abstract
Neovascularisation, i. e. arteriogenesis and angiogenesis, is an inflammatory process. Therefore attraction and extravasation of leukocytes is essential for effective blood flow recovery after ischaemia. Previous studies have shown that von Willebrand factor (VWF) is a negative regulator of angiogenesis. However, it has also been shown that VWF facilitates leukocyte attraction and extravasation. We aimed to investigate the role of VWF in arteriogenesis and angiogenesis during post-ischaemic neovascularisation. Wild-type (WT) and VWF deficient (VWF-/-) C57BL/6 mice were subjected to hindlimb ischaemia via double ligation of the left femoral artery, and blood flow recovery was followed over time, using Laser Doppler Perfusion Imaging. Blood flow recovery was impaired in VWF-/- mice. After 10 days, VWF-/- mice showed a 43 ± 5 % recovery versus 68 ± 5 % in WT. Immunohistochemistry revealed that both arteriogenesis in the adductor muscles and angiogenesis in the gastrocnemius muscles were reduced in VWF-/- mice. Furthermore, leukocyte infiltration in the affected adductor muscles was reduced in VWF-/- mice. Residual paw perfusion directly after artery ligation was also reduced in VWF-/- mice, indicating a decrease in pre-existing collateral arteriole density. When we quantified collateral arterioles, we observed a 31 % decrease in the average number of collateral arterioles in the pia mater compared to WT mice (57 ± 3 in WT vs 40 ± 4 pial collaterals in VWF-/-). We conclude that VWF facilitates blood flow recovery in mice. VWF deficiency hampers both arteriogenesis and angiogenesis in a hindlimb ischaemia model. This is associated with impaired leukocytes recruitment and decreased pre-existing collateral density in the absence of VWF.
Collapse
Affiliation(s)
| | | | | | - A Yaël Nossent
- A. Y. Nossent, PhD, Department of Surgery, D6-28, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, the Netherlands, Tel.: +31 71 52 65147, E-mail:
| |
Collapse
|
33
|
Nossent AY, Bastiaansen AJNM, Peters EAB, de Vries MR, Aref Z, Welten SMJ, de Jager SCA, van der Pouw Kraan TCTM, Quax PHA. CCR7-CCL19/CCL21 Axis is Essential for Effective Arteriogenesis in a Murine Model of Hindlimb Ischemia. J Am Heart Assoc 2017; 6:JAHA.116.005281. [PMID: 28275068 PMCID: PMC5524034 DOI: 10.1161/jaha.116.005281] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background In order to identify factors that stimulate arteriogenesis after ischemia, we followed gene expression profiles in two extreme models for collateral artery formation over 28 days after hindlimb ischemia, namely “good‐responding” C57BL/6 mice and “poor‐responding” BALB/c mice. Methods and Results Although BALB/c mice show very poor blood flow recovery after ischemia, most known proarteriogenic genes were upregulated more excessively and for a longer period than in C57BL/6 mice. In clear contrast, chemokine genes Ccl19, Ccl21a, and Ccl21c and the chemokine receptor CCR7 were upregulated in C57BL/6 mice 1 day after hindlimb ischemia, but not in BALB/C mice. CCL19 and CCL21 regulate migration and homing of T lymphocytes via CCR7. When subjecting CCR7−/−/LDLR−/− mice to hindlimb ischemia, we observed a 20% reduction in blood flow recovery compared with that in LDLR−/− mice. Equal numbers of α‐smooth muscle actin–positive collateral arteries were found in the adductor muscles of both mouse strains, but collateral diameters were smaller in the CCR7−/−/LDLR−/−. Fluorescence‐activated cell sorter analyses showed that numbers of CCR7+ T lymphocytes (both CD4+ and CD8+) were decreased in the spleen and increased in the blood at day 1 after hindlimb ischemia in LDLR−/− mice. At day 1 after hindlimb ischemia, however, numbers of activated CD4+ T lymphocytes were decreased in the draining lymph nodes of LDLR−/− mice compared with CCR7−/−/LDLR−/− mice. Conclusions These data show that CCR7‐CCL19/CCL21 axis facilitates retention CD4+ T lymphocytes at the site of collateral artery remodeling, which is essential for effective arteriogenesis.
Collapse
Affiliation(s)
- A Yaël Nossent
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands .,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Antonius J N M Bastiaansen
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Erna A B Peters
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Margreet R de Vries
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Zeen Aref
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Sabine M J Welten
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Saskia C A de Jager
- Division of Biopharmaceutics, LACDR, Leiden University, Leiden, the Netherlands.,Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Paul H A Quax
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
34
|
Sommer CJ. Ischemic stroke: experimental models and reality. Acta Neuropathol 2017; 133:245-261. [PMID: 28064357 PMCID: PMC5250659 DOI: 10.1007/s00401-017-1667-0] [Citation(s) in RCA: 375] [Impact Index Per Article: 46.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 12/31/2016] [Accepted: 01/01/2017] [Indexed: 12/11/2022]
Abstract
The vast majority of cerebral stroke cases are caused by transient or permanent occlusion of a cerebral blood vessel (“ischemic stroke”) eventually leading to brain infarction. The final infarct size and the neurological outcome depend on a multitude of factors such as the duration and severity of ischemia, the existence of collateral systems and an adequate systemic blood pressure, etiology and localization of the infarct, but also on age, sex, comorbidities with the respective multimedication and genetic background. Thus, ischemic stroke is a highly complex and heterogeneous disorder. It is immediately obvious that experimental models of stroke can cover only individual specific aspects of this multifaceted disease. A basic understanding of the principal molecular pathways induced by ischemia-like conditions comes already from in vitro studies. One of the most frequently used in vivo models in stroke research is the endovascular suture or filament model in rodents with occlusion of the middle cerebral artery (MCA), which causes reproducible infarcts in the MCA territory. It does not require craniectomy and allows reperfusion by withdrawal of the occluding filament. Although promptly restored blood flow is far from the pathophysiology of spontaneous human stroke, it more closely mimics the therapeutic situation of mechanical thrombectomy which is expected to be increasingly applied to stroke patients. Direct transient or permanent occlusion of cerebral arteries represents an alternative approach but requires craniectomy. Application of endothelin-1, a potent vasoconstrictor, allows induction of transient focal ischemia in nearly any brain region and is frequently used to model lacunar stroke. Circumscribed and highly reproducible cortical lesions are characteristic of photothrombotic stroke where infarcts are induced by photoactivation of a systemically given dye through the intact skull. The major shortcoming of this model is near complete lack of a penumbra. The two models mimicking human stroke most closely are various embolic stroke models and spontaneous stroke models. Closeness to reality has its price and goes along with higher variability of infarct size and location as well as unpredictable stroke onset in spontaneous models versus unpredictable reperfusion in embolic clot models.
Collapse
Affiliation(s)
- Clemens J Sommer
- Institute of Neuropathology, University Medical Center of the Johannes Gutenberg-University Mainz; Focus Program Translational Neuroscience (FTN) and Rhine Main Neuroscience Network (rmn2), Langenbeckstrasse 1, 55131, Mainz, Germany.
| |
Collapse
|
35
|
Schmidt CA, Ryan TE, Lin CT, Inigo MMR, Green TD, Brault JJ, Spangenburg EE, McClung JM. Diminished force production and mitochondrial respiratory deficits are strain-dependent myopathies of subacute limb ischemia. J Vasc Surg 2016; 65:1504-1514.e11. [PMID: 28024849 DOI: 10.1016/j.jvs.2016.04.041] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 04/17/2016] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Reduced skeletal muscle mitochondrial function might be a contributing mechanism to the myopathy and activity based limitations that typically plague patients with peripheral arterial disease (PAD). We hypothesized that mitochondrial dysfunction, myofiber atrophy, and muscle contractile deficits are inherently determined by the genetic background of regenerating ischemic mouse skeletal muscle, similar to how patient genetics affect the distribution of disease severity with clinical PAD. METHODS Genetically ischemia protected (C57BL/6) and susceptible (BALB/c) mice underwent either unilateral subacute hind limb ischemia (SLI) or myotoxic injury (cardiotoxin) for 28 days. Limbs were monitored for blood flow and tissue oxygen saturation and tissue was collected for the assessment of histology, muscle contractile force, gene expression, mitochondrial content, and respiratory function. RESULTS Despite similar tissue O2 saturation and mitochondrial content between strains, BALB/c mice suffered persistent ischemic myofiber atrophy (55.3% of C57BL/6) and muscle contractile deficits (approximately 25% of C57BL/6 across multiple stimulation frequencies). SLI also reduced BALB/c mitochondrial respiratory capacity, assessed in either isolated mitochondria (58.3% of C57BL/6 at SLI on day (d)7, 59.1% of C57BL/6 at SLI d28 across multiple conditions) or permeabilized myofibers (38.9% of C57BL/6 at SLI d7; 76.2% of C57BL/6 at SLI d28 across multiple conditions). SLI also resulted in decreased calcium retention capacity (56.0% of C57BL/6) in BALB/c mitochondria. Nonischemic cardiotoxin injury revealed similar recovery of myofiber area, contractile force, mitochondrial respiratory capacity, and calcium retention between strains. CONCLUSIONS Ischemia-susceptible BALB/c mice suffered persistent muscle atrophy, impaired muscle function, and mitochondrial respiratory deficits during SLI. Interestingly, parental strain susceptibility to myopathy appears specific to regenerative insults including an ischemic component. Our findings indicate that the functional deficits that plague PAD patients could include mitochondrial respiratory deficits genetically inherent to the regenerating muscle myofibers.
Collapse
Affiliation(s)
- Cameron A Schmidt
- Department of Physiology, East Carolina University, Greenville, NC; Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Terence E Ryan
- Department of Physiology, East Carolina University, Greenville, NC; Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Chien-Te Lin
- Department of Physiology, East Carolina University, Greenville, NC; Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Melissa M R Inigo
- Department of Physiology, East Carolina University, Greenville, NC; Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Tom D Green
- Department of Physiology, East Carolina University, Greenville, NC; Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Jeffrey J Brault
- Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, NC; Department of Kinesiology, East Carolina University, Greenville, NC
| | - Espen E Spangenburg
- Department of Physiology, East Carolina University, Greenville, NC; Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Joseph M McClung
- Department of Physiology, East Carolina University, Greenville, NC; Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, NC.
| |
Collapse
|
36
|
Faber JE, Moore SM, Lucitti JL, Aghajanian A, Zhang H. Sex Differences in the Cerebral Collateral Circulation. Transl Stroke Res 2016; 8:273-283. [PMID: 27844273 DOI: 10.1007/s12975-016-0508-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 10/26/2016] [Accepted: 11/01/2016] [Indexed: 12/15/2022]
Abstract
Premenopausal women and intact female rodents sustain smaller cerebral infarctions than males. Several sex-dependent differences have been identified as potential contributors, but many questions remain unanswered. Mice exhibit wide variation in native collateral number and diameter (collateral extent) that is dependent on differences in genetic background, aging, and other comorbidities and that contributes to their also-wide differences in infarct volume. Likewise, variation in infarct volume correlates with differences in collateral-dependent blood flow in patients with acute ischemic stroke. We examined whether extent of pial collateral arterioles and posterior communicating collateral arteries (PComAs) differ depending on sex in young, aged, obese, hypertensive, and genetically different mice. We combined new data with meta-analysis of our previously published data. Females of C57BL/6J (B6) and BALB/cByJ (BC) strains sustained smaller infarctions than males after permanent MCA occlusion. This protection was unchanged in BC mice after introgression of the B6 allele of Dce1, the major genetic determinant of variation in pial collaterals among mouse strains. Consistent with this, collateral extent in these and other strains did not differ with sex. Extent of PComAs and primary cerebral arteries also did not vary with sex. No dimorphism was evident for loss of pial collateral number and/or diameter (collateral rarefaction) caused by aging, obesity, and hypertension, nor for collateral remodeling after pMCAO. However, rarefaction was greater in females with long-standing hypertension. We conclude that smaller infarct volume in female mice is not due to greater collateral extent, greater remodeling, or less rarefaction caused by aging, obesity, or hypertension.
Collapse
Affiliation(s)
- James E Faber
- Department of Cell Biology and Physiology, The McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA.
| | - Scott M Moore
- Department of Surgery, University of Colorado, Denver, CO, USA
| | - Jennifer L Lucitti
- Department of Cell Biology and Physiology, The McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Amir Aghajanian
- Department of Cell Biology and Physiology, The McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Hua Zhang
- Department of Cell Biology and Physiology, The McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA
| |
Collapse
|
37
|
Bosetti F, Galis ZS, Bynoe MS, Charette M, Cipolla MJ, Del Zoppo GJ, Gould D, Hatsukami TS, Jones TLZ, Koenig JI, Lutty GA, Maric-Bilkan C, Stevens T, Tolunay HE, Koroshetz W. "Small Blood Vessels: Big Health Problems?": Scientific Recommendations of the National Institutes of Health Workshop. J Am Heart Assoc 2016; 5:JAHA.116.004389. [PMID: 27815267 PMCID: PMC5210346 DOI: 10.1161/jaha.116.004389] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Francesca Bosetti
- National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD
| | - Zorina S Galis
- National Heart, Lung and Blood Institute, National Institutes of Health (NIH), Bethesda, MD
| | | | - Marc Charette
- National Heart, Lung and Blood Institute, National Institutes of Health (NIH), Bethesda, MD
| | | | | | | | | | - Teresa L Z Jones
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health (NIH), Bethesda, MD
| | - James I Koenig
- National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD
| | | | - Christine Maric-Bilkan
- National Heart, Lung and Blood Institute, National Institutes of Health (NIH), Bethesda, MD
| | | | - H Eser Tolunay
- National Heart, Lung and Blood Institute, National Institutes of Health (NIH), Bethesda, MD
| | - Walter Koroshetz
- National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD
| | | |
Collapse
|
38
|
Lucitti JL, Sealock R, Buckley BK, Zhang H, Xiao L, Dudley AC, Faber JE. Variants of Rab GTPase-Effector Binding Protein-2 Cause Variation in the Collateral Circulation and Severity of Stroke. Stroke 2016; 47:3022-3031. [PMID: 27811335 DOI: 10.1161/strokeaha.116.014160] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 08/23/2016] [Accepted: 09/21/2016] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND PURPOSE The extent (number and diameter) of collateral vessels varies widely and is a major determinant, along with arteriogenesis (collateral remodeling), of variation in severity of tissue injury after large artery occlusion. Differences in genetic background underlie the majority of the variation in collateral extent in mice, through alterations in collaterogenesis (embryonic collateral formation). In brain and other tissues, ≈80% of the variation in collateral extent among different mouse strains has been linked to a region on chromosome 7. We recently used congenic (CNG) fine mapping of C57BL/6 (B6, high extent) and BALB/cByJ (BC, low extent) mice to narrow the region to a 737 Kb locus, Dce1. Herein, we report the causal gene. METHODS We used additional CNG mapping and knockout mice to narrow the number of candidate genes. Subsequent inspection identified a nonsynonymous single nucleotide polymorphism between B6 and BC within Rabep2 (rs33080487). We then created B6 mice with the BC single nucleotide polymorphism at this locus plus 3 other lines for predicted alteration or knockout of Rabep2 using gene editing. RESULTS The single amino acid change caused by rs33080487 accounted for the difference in collateral extent and infarct volume between B6 and BC mice attributable to Dce1. Mechanistically, variants of Rabep2 altered collaterogenesis during embryogenesis but had no effect on angiogenesis examined in vivo and in vitro. Rabep2 deficiency altered endosome trafficking known to be involved in VEGF-A→VEGFR2 signaling required for collaterogenesis. CONCLUSIONS Naturally occurring variants of Rabep2 are major determinants of variation in collateral extent and stroke severity in mice.
Collapse
Affiliation(s)
- Jennifer L Lucitti
- From the Department of Cell Biology and Physiology, The McAllister Heart Institute, University of North Carolina, Chapel Hill
| | - Robert Sealock
- From the Department of Cell Biology and Physiology, The McAllister Heart Institute, University of North Carolina, Chapel Hill
| | - Brian K Buckley
- From the Department of Cell Biology and Physiology, The McAllister Heart Institute, University of North Carolina, Chapel Hill
| | - Hua Zhang
- From the Department of Cell Biology and Physiology, The McAllister Heart Institute, University of North Carolina, Chapel Hill
| | - Lin Xiao
- From the Department of Cell Biology and Physiology, The McAllister Heart Institute, University of North Carolina, Chapel Hill
| | - Andrew C Dudley
- From the Department of Cell Biology and Physiology, The McAllister Heart Institute, University of North Carolina, Chapel Hill
| | - James E Faber
- From the Department of Cell Biology and Physiology, The McAllister Heart Institute, University of North Carolina, Chapel Hill.
| |
Collapse
|
39
|
Ginsberg MD. Expanding the concept of neuroprotection for acute ischemic stroke: The pivotal roles of reperfusion and the collateral circulation. Prog Neurobiol 2016; 145-146:46-77. [PMID: 27637159 DOI: 10.1016/j.pneurobio.2016.09.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/22/2016] [Accepted: 09/10/2016] [Indexed: 12/27/2022]
Abstract
This review surveys the efforts taken to achieve clinically efficacious protection of the ischemic brain and underscores the necessity of expanding our purview to include the essential role of cerebral perfusion and the collateral circulation. We consider the development of quantitative strategies to measure cerebral perfusion at the regional and local levels and the application of these methods to elucidate flow-related thresholds of ischemic viability and to characterize the ischemic penumbra. We stress that the modern concept of neuroprotection must consider perfusion, the necessary substrate upon which ischemic brain survival depends. We survey the major mechanistic approaches to neuroprotection and review clinical neuroprotection trials, focusing on those phase 3 multicenter clinical trials for acute ischemic stroke that have been completed or terminated. We review the evolution of thrombolytic therapies; consider the lessons learned from the initial, negative multicenter trials of endovascular therapy; and emphasize the highly successful positive trials that have finally established a clinical role for endovascular clot removal. As these studies point to the brain's collateral circulation as key to successful reperfusion, we next review the anatomy and pathophysiology of collateral perfusion as it relates to ischemic infarction, as well as the molecular and genetic influences on collateral development. We discuss the current MR and CT-based diagnostic methods for assessing the collateral circulation and the prognostic significance of collaterals in ischemic stroke, and we consider past and possible future therapeutic directions.
Collapse
Affiliation(s)
- Myron D Ginsberg
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States.
| |
Collapse
|
40
|
The Role of Vascular Imaging in the Initial Assessment of Patients with Acute Ischemic Stroke. Curr Neurol Neurosci Rep 2016; 16:32. [PMID: 26898684 DOI: 10.1007/s11910-016-0632-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Over the last few years, improvement in radiological imaging and treatment has changed the management of acute ischemic stroke. We have made significant advances in not only the imaging modalities themselves but also in identifying imaging parameters that can help us predict patient outcomes with both intravascular thrombolysis and endovascular thrombectomy. In this review, we describe the added utility of baseline vascular imaging including computed tomography angiography and magnetic resonance angiography in the diagnosis and management of patients with acute ischemic stroke. We focus on information these imaging modalities provide on clot characteristics, tissue state, collateral status, and endovascular planning. We also highlight the benefits of newer imaging modalities like dynamic computed tomography angiography (CTA) and multi-phase CTA. Lastly, we also describe some of the disadvantages of vascular imaging in ischemic stroke.
Collapse
|
41
|
Wang S, Reeves B, Sparkenbaugh EM, Russell J, Soltys Z, Zhang H, Faber JE, Key NS, Kirchhofer D, Granger DN, Mackman N, Pawlinski R. Protective and detrimental effects of neuroectodermal cell-derived tissue factor in mouse models of stroke. JCI Insight 2016; 1. [PMID: 27489885 DOI: 10.1172/jci.insight.86663] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Within the CNS, a dysregulated hemostatic response contributes to both hemorrhagic and ischemic strokes. Tissue factor (TF), the primary initiator of the extrinsic coagulation cascade, plays an essential role in hemostasis and also contributes to thrombosis. Using both genetic and pharmacologic approaches, we characterized the contribution of neuroectodermal (NE) cell TF to the pathophysiology of stroke. We used mice with various levels of TF expression and found that astrocyte TF activity reduced to ~5% of WT levels was still sufficient to maintain hemostasis after hemorrhagic stroke but was also low enough to attenuate inflammation, reduce damage to the blood-brain barrier, and improve outcomes following ischemic stroke. Pharmacologic inhibition of TF during the reperfusion phase of ischemic stroke attenuated neuronal damage, improved behavioral deficit, and prevented mortality of mice. Our data demonstrate that NE cell TF limits bleeding complications associated with the transition from ischemic to hemorrhagic stroke and also contributes to the reperfusion injury after ischemic stroke. The high level of TF expression in the CNS is likely the result of selective pressure to limit intracerebral hemorrhage (ICH) after traumatic brain injury but, in the modern era, poses the additional risk of increased ischemia-reperfusion injury after ischemic stroke.
Collapse
Affiliation(s)
- Shaobin Wang
- McAllister Heart Institute, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Brandi Reeves
- Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Erica M Sparkenbaugh
- McAllister Heart Institute, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Janice Russell
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA
| | - Zbigniew Soltys
- Department of Neuroanatomy, Institute of Zoology, Jagiellonian University, Krakow, Poland
| | - Hua Zhang
- McAllister Heart Institute, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, North Carolina, USA
| | - James E Faber
- McAllister Heart Institute, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Nigel S Key
- McAllister Heart Institute, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Daniel Kirchhofer
- Department of Early Discovery Biochemistry, Genentech Inc., South San Francisco, California, USA
| | - D Neil Granger
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA
| | - Nigel Mackman
- McAllister Heart Institute, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Rafal Pawlinski
- McAllister Heart Institute, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
42
|
Lee HK, Keum S, Sheng H, Warner DS, Lo DC, Marchuk DA. Natural allelic variation of the IL-21 receptor modulates ischemic stroke infarct volume. J Clin Invest 2016; 126:2827-38. [PMID: 27400126 DOI: 10.1172/jci84491] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 05/12/2016] [Indexed: 02/06/2023] Open
Abstract
Risk for ischemic stroke has a strong genetic basis, but heritable factors also contribute to the extent of damage after a stroke has occurred. We previously identified a locus on distal mouse chromosome 7 that contributes over 50% of the variation in postischemic cerebral infarct volume observed between inbred strains. Here, we used ancestral haplotype analysis to fine-map this locus to 12 candidate genes. The gene encoding the IL-21 receptor (Il21r) showed a marked difference in strain-specific transcription levels and coding variants in neonatal and adult cortical tissue. Collateral vessel connections were moderately reduced in Il21r-deficient mice, and cerebral infarct volume increased 2.3-fold, suggesting that Il21r modulates both collateral vessel anatomy and innate neuroprotection. In brain slice explants, oxygen deprivation (OD) activated apoptotic pathways and increased neuronal cell death in IL-21 receptor-deficient (IL-21R-deficient) mice compared with control animals. We determined that the neuroprotective effects of IL-21R arose from signaling through JAK/STAT pathways and upregulation of caspase 3. Thus, natural genetic variation in murine Il21r influences neuronal cell viability after ischemia by modulating receptor function and downstream signal transduction. The identification of neuroprotective genes based on naturally occurring allelic variations has the potential to inform the development of drug targets for ischemic stroke treatment.
Collapse
|
43
|
Ryan TE, Schmidt CA, Green TD, Brown DA, Neufer PD, McClung JM. Mitochondrial Regulation of the Muscle Microenvironment in Critical Limb Ischemia. Front Physiol 2015; 6:336. [PMID: 26635622 PMCID: PMC4649016 DOI: 10.3389/fphys.2015.00336] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 11/02/2015] [Indexed: 01/11/2023] Open
Abstract
Critical limb ischemia (CLI) is the most severe clinical presentation of peripheral arterial disease and manifests as chronic limb pain at rest and/or tissue necrosis. Current clinical interventions are largely ineffective and therapeutic angiogenesis based trials have shown little efficacy, highlighting the dire need for new ideas and novel therapeutic approaches. Despite a decade of research related to skeletal muscle as a determinant of morbidity and mortality outcomes in CLI, very little progress has been made toward an effective therapy aimed directly at the muscle myopathies of this disease. Within the muscle cell, mitochondria are well positioned to modulate the ischemic cellular response, as they are the principal sites of cellular energy production and the major regulators of cellular redox charge and cell death. In this mini review, we update the crucial importance of skeletal muscle to CLI pathology and examine the evolving influence of muscle and endothelial cell mitochondria in the complex ischemic microenvironment. Finally, we discuss the novelty of muscle mitochondria as a therapeutic target for ischemic pathology in the context of the complex co-morbidities often associated with CLI.
Collapse
Affiliation(s)
- Terence E Ryan
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - Cameron A Schmidt
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - Tom D Green
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - David A Brown
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - P Darrell Neufer
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - Joseph M McClung
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| |
Collapse
|
44
|
Du R, Zhou J, Lorenzano S, Liu W, Charoenvimolphan N, Qian B, Xu J, Wang J, Zhang X, Wang X, Berndt A, Devan WJ, Valant VJ, Wang J, Furie KL, Rosand J, Rost N, Friedlander RM, Paigen B, Weiss ST. Integrative Mouse and Human Studies Implicate ANGPT1 and ZBTB7C as Susceptibility Genes to Ischemic Injury. Stroke 2015; 46:3514-22. [PMID: 26542693 DOI: 10.1161/strokeaha.115.010767] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 10/01/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND PURPOSE The extent of ischemic injury in response to cerebral ischemia is known to be affected by native vasculature. However, the nonvascular and dynamic vascular responses and their genetic basis are not well understood. METHODS We performed a genome-wide association study in 235 mice from 33 inbred strains using the middle cerebral artery occlusion model. Population structure and genetic relatedness were accounted for using the efficient mixed-model association method. Human orthologs to the genes associated with the significant and suggestive single-nucleotide polymorphisms from the mouse strain survey were examined in patients with M1 occlusions admitted with signs and symptoms of acute ischemic stroke. RESULTS We identified 4 genome-wide significant and suggestive single-nucleotide polymorphisms to be associated with infarct volume in mice (rs3694965, P=2.17×10(-7); rs31924033, P=5.61×10(-6); rs32249495, P=2.08×10(-7); and rs3677406, P=9.56×10(-6)). rs32249495, which corresponds to angiopoietin-1 (ANGPT1), was also significant in the recessive model in humans, whereas rs1944577, which corresponds to ZBTB7C, was nominally significant in both the additive and dominant genetic models in humans. ZBTB7C was shown to be upregulated in endothelial cells using both in vitro and in vivo models of ischemia. CONCLUSIONS Genetic variations of ANGPT1 and ZBTB7C are associated with increased infarct size in both mice and humans. ZBTB7C may modulate the ischemic response via neuronal apoptosis and dynamic collateralization and, in addition to ANGPT1, may serve as potential novel targets for treatments of cerebral ischemia.
Collapse
Affiliation(s)
- Rose Du
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.).
| | - Jing Zhou
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Svetlana Lorenzano
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Wenming Liu
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Nareerat Charoenvimolphan
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Baogang Qian
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Jun Xu
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Jian Wang
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Xinmu Zhang
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Xin Wang
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Annerose Berndt
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - William J Devan
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Valerie J Valant
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Jinyi Wang
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Karen L Furie
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Jonathan Rosand
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Natalia Rost
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Robert M Friedlander
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Beverly Paigen
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Scott T Weiss
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| |
Collapse
|
45
|
Tang Z, Li S, Han P, Yin J, Gan Y, Liu Q, Wang J, Wang C, Li Y, Shi J. Pertussis toxin reduces calcium influx to protect ischemic stroke in a middle cerebral artery occlusion model. J Neurochem 2015; 135:998-1006. [PMID: 26365274 DOI: 10.1111/jnc.13359] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 08/30/2015] [Accepted: 09/10/2015] [Indexed: 11/28/2022]
Affiliation(s)
- Zhiwei Tang
- Department of Neurology; Barrow Neurological Institute; St. Joseph's Hospital and Medical Center; Phoenix Arizona USA
- Department of Neurosurgery; the First Affiliated Hospital of Kunming Medical University; Kunming China
| | - Shiping Li
- Department of Neurology; Barrow Neurological Institute; St. Joseph's Hospital and Medical Center; Phoenix Arizona USA
- Department of Neurology; No.2 Hospital of Hebei Medical University; Shijiazhuang China
| | - Pengcheng Han
- Department of Neurology; Barrow Neurological Institute; St. Joseph's Hospital and Medical Center; Phoenix Arizona USA
| | - Junxiang Yin
- Department of Neurology; Barrow Neurological Institute; St. Joseph's Hospital and Medical Center; Phoenix Arizona USA
| | - Yan Gan
- Department of Neurology; Barrow Neurological Institute; St. Joseph's Hospital and Medical Center; Phoenix Arizona USA
| | - Qingwei Liu
- Department of Radiology; Barrow Neurological Institute; St. Joseph's Hospital and Medical Center; Phoenix Arizona USA
| | - Jinkun Wang
- Department of Neurosurgery; the First Affiliated Hospital of Kunming Medical University; Kunming China
| | - Chongqian Wang
- Department of Neurosurgery; the First Affiliated Hospital of Kunming Medical University; Kunming China
| | - Yu Li
- Department of Neurosurgery; the First Affiliated Hospital of Kunming Medical University; Kunming China
| | - Jiong Shi
- Department of Neurology; Barrow Neurological Institute; St. Joseph's Hospital and Medical Center; Phoenix Arizona USA
| |
Collapse
|
46
|
McClung JM, McCord TJ, Southerland K, Schmidt CA, Padgett ME, Ryan TE, Kontos CD. Subacute limb ischemia induces skeletal muscle injury in genetically susceptible mice independent of vascular density. J Vasc Surg 2015; 64:1101-1111.e2. [PMID: 26254821 DOI: 10.1016/j.jvs.2015.06.139] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 06/07/2015] [Indexed: 01/01/2023]
Abstract
OBJECTIVE The primary preclinical model of peripheral artery disease, which involves acute limb ischemia (ALI), can result in appreciable muscle injury that is attributed to the acuity of the ischemic injury. A less acute model of murine limb ischemia using ameroid constrictors (ACs) has been developed in an attempt to mimic the chronic nature of human disease. However, there is currently little understanding of how genetics influence muscle injury following subacute arterial occlusion in the mouse. METHODS We investigated the influence of mouse genetics on skeletal muscle tissue survival, blood flow, and vascular density by subjecting two different mouse strains, C57BL/6 (BL6) and BALB/c, to ALI or subacute limb ischemia using single (1AC) or double (2AC) AC placement on the femoral artery. RESULTS Similar to ALI, the 2AC model resulted in significant tissue necrosis and limb perfusion deficits in genetically susceptible BALB/c but not BL6 mice. In the 1AC model, no outward evidence of tissue necrosis was observed, and there were no differences in limb blood flow between BL6 and BALB/c. However, BALB/c mice displayed significantly greater muscle injury, as evidenced by increased inflammation and myofiber atrophy, despite having no differences in CD31(+) and SMA(+) vascular density and area. BALB/c mice also displayed significantly greater centralized myonuclei, indicating increased muscle regeneration. CONCLUSIONS The susceptibility of skeletal muscle to ischemia-induced injury is at least partly independent of muscle blood flow and vascular density, consistent with a muscle cell autonomous response that is genetically determined. Further development of preclinical models of peripheral artery disease that more accurately reflect the nature of the human disease may allow more accurate identification of genetic targets for therapeutic intervention.
Collapse
Affiliation(s)
- Joseph M McClung
- Department of Physiology, East Carolina University, Brody Medical Center, Greenville, NC; Diabetes and Obesity Institute, East Carolina Heart Institute, Brody Medical Center, Greenville, NC.
| | - Timothy J McCord
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Kevin Southerland
- Division of General Surgery, Department of Surgery, Duke University Medical Center, Durham, NC
| | - Cameron A Schmidt
- Department of Physiology, East Carolina University, Brody Medical Center, Greenville, NC; Diabetes and Obesity Institute, East Carolina Heart Institute, Brody Medical Center, Greenville, NC
| | - Michael E Padgett
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Terence E Ryan
- Department of Physiology, East Carolina University, Brody Medical Center, Greenville, NC; Diabetes and Obesity Institute, East Carolina Heart Institute, Brody Medical Center, Greenville, NC
| | - Christopher D Kontos
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC; Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC
| |
Collapse
|
47
|
Al-Ali F, Elias JJ, Filipkowski DE, Faber JE. Acute ischemic stroke treatment, part 1: patient selection "the 50% barrier and the capillary index score". Front Neurol 2015; 6:83. [PMID: 25954243 PMCID: PMC4406085 DOI: 10.3389/fneur.2015.00083] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/31/2015] [Indexed: 11/13/2022] Open
Abstract
The current strategy for intra-arterial treatment (IAT) of acute ischemic stroke focuses on minimizing time from ictus to revascularization and maximizing revascularization. Employing this strategy has yet to lead to improved rates of successful outcomes. However, the collateral blood supply likely plays a significant role in maintaining viable brain tissue during ischemia. Based on our prior work, we believe that only approximately 50% of patients are genetically predisposed to have sufficient collaterals for a good outcome following treatment, a concept we call the 50% barrier. The Capillary Index Score (CIS) has been developed as a tool to identify patients with a sufficient collateral blood supply to maintain tissue viability prior to treatment. Patients with a favorable CIS (f CIS) may be able to achieve a good outcome with IAT beyond an arbitrary time window. The CIS is incorporated into a proposed patient treatment algorithm. For patients suffering from a large stroke without aphasia, a non-enhanced head CT should be followed by CT angiography (CTA). For patients without signs of stroke mimics or visible signs of structural changes due to large irreversible ischemia, CTA can help confirm the vascular occlusion and location. The CIS can be obtained from a diagnostic cerebral angiogram, with IAT offered to patients categorized as f CIS.
Collapse
Affiliation(s)
- Firas Al-Ali
- Summit Neurovascular Specialists, Akron, OH, USA
| | - John J. Elias
- Department of Research, Akron General Medical Center, Akron, OH, USA
| | | | - James E. Faber
- Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| |
Collapse
|
48
|
Bigler ED, Stern Y. Traumatic brain injury and reserve. HANDBOOK OF CLINICAL NEUROLOGY 2015; 128:691-710. [DOI: 10.1016/b978-0-444-63521-1.00043-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
49
|
Prabhakar P, Zhang H, Chen D, Faber JE. Genetic variation in retinal vascular patterning predicts variation in pial collateral extent and stroke severity. Angiogenesis 2015; 18:97-114. [PMID: 25369734 PMCID: PMC4422395 DOI: 10.1007/s10456-014-9449-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 10/27/2014] [Indexed: 01/21/2023]
Abstract
The presence of a native collateral circulation in tissues lessens injury in occlusive vascular diseases. However, differences in genetic background cause wide variation in collateral number and diameter in mice, resulting in large variation in protection. Indirect estimates of collateral perfusion suggest that wide variation also exists in humans. Unfortunately, methods used to obtain these estimates are invasive and not widely available. We sought to determine whether differences in genetic background in mice result in variation in branch patterning of the retinal arterial circulation, and whether these differences predict strain-dependent differences in pial collateral extent and severity of ischemic stroke. Retinal patterning metrics, collateral extent, and infarct volume were obtained for 10 strains known to differ widely in collateral extent. Multivariate regression was conducted, and model performance was assessed using K-fold cross-validation. Twenty-one metrics varied with strain (p<0.01). Ten metrics (e.g., bifurcation angle, lacunarity, optimality) predicted collateral number and diameter across seven regression models, with the best model closely predicting (p<0.0001) number (±1.2-3.4 collaterals, K-fold R2=0.83-0.98), diameter (±1.2-1.9 μm, R2=0.73-0.88), and infarct volume (±5.1 mm3, R2=0.85-0.87). An analogous set of the most predictive metrics, obtained for the middle cerebral artery (MCA) tree in a subset of the above strains, also predicted (p<0.0001) collateral number (±3.3 collaterals, K-fold R2=0.78) and diameter (±1.6 μm, R2=0.86). Thus, differences in arterial branch patterning in the retina and the MCA trees are specified by genetic background and predict variation in collateral extent and stroke severity. If also true in human, and since genetic variation in cerebral collaterals extends to other tissues at least in mice, a similar "retinal predictor index" could serve as a non- or minimally invasive biomarker for collateral extent in brain and other tissues. This could aid prediction of severity of tissue injury in the event of an occlusive event or development of obstructive disease and in patient stratification for treatment options and clinical studies.
Collapse
Affiliation(s)
- Pranay Prabhakar
- Department of Cell Biology and Physiology and the McAllister Heart Institute, University of North Carolina
| | - Hua Zhang
- Department of Cell Biology and Physiology and the McAllister Heart Institute, University of North Carolina
| | - De Chen
- Optical Microscopy and Analysis Laboratory Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - James E. Faber
- Department of Cell Biology and Physiology and the McAllister Heart Institute, University of North Carolina
| |
Collapse
|
50
|
Michelis KC, Boehm M, Kovacic JC. New vessel formation in the context of cardiomyocyte regeneration--the role and importance of an adequate perfusing vasculature. Stem Cell Res 2014; 13:666-82. [PMID: 24841067 PMCID: PMC4213356 DOI: 10.1016/j.scr.2014.04.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 03/16/2014] [Accepted: 04/18/2014] [Indexed: 02/08/2023] Open
Abstract
The history of revascularization for cardiac ischemia dates back to the early 1960's when the first coronary artery bypass graft procedures were performed in humans. With this 50 year history of providing a new vasculature to ischemic and hibernating myocardium, a profound depth of experience has been amassed in clinical cardiovascular medicine as to what does, and does not work in the context of cardiac revascularization, alleviating ischemia and adequacy of myocardial perfusion. These issues are of central relevance to contemporary cell-based cardiac regenerative approaches. While the cardiovascular cell therapy field is surging forward on many exciting fronts, several well accepted clinical axioms related to the cardiac arterial supply appear to be almost overlooked by some of our current basic conceptual and experimental cell therapy paradigms. We present here information drawn from five decades of the clinical revascularization experience, review relevant new data on vascular formation via cell therapy, and put forward the case that for optimal cell-based cardiac regeneration due attention must be paid to providing an adequate vascular supply.
Collapse
Affiliation(s)
- Katherine C Michelis
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manfred Boehm
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jason C Kovacic
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|