101
|
Atg7 Regulates Brain Angiogenesis via NF-κB-Dependent IL-6 Production. Int J Mol Sci 2017; 18:ijms18050968. [PMID: 28467355 PMCID: PMC5454881 DOI: 10.3390/ijms18050968] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 04/25/2017] [Accepted: 04/28/2017] [Indexed: 12/31/2022] Open
Abstract
The formation of brain vasculature is an essential step during central nervous system development. The molecular mechanism underlying brain angiogenesis remains incompletely understood. The role of Atg7, an autophagy-related protein, in brain angiogenesis was investigated in this study. We found that the microvessel density in mice brains with endothelial-specific knockout of Atg7 (Atg7 EKO) was significantly decreased compared to wild-type control. Consistently, in vitro angiogenesis assays showed that Atg7 knockdown impaired angiogenesis in brain microvascular endothelial cells. Further results indicated that knockdown of Atg7 reduced interleukin-6 (IL-6) expression in brain microvascular endothelial cells, which is mediated by NF-κB-dependent transcriptional control. Interestingly, exogenous IL-6 restored the impaired angiogenesis and reduced cell motility caused by Atg7 knockdown. These results demonstrated that Atg7 has proangiogenic activity in brain angiogenesis which is mediated by IL-6 production in a NF-κB-dependent manner.
Collapse
|
102
|
Dorr A, Thomason LA, Koletar MM, Joo IL, Steinman J, Cahill LS, Sled JG, Stefanovic B. Effects of voluntary exercise on structure and function of cortical microvasculature. J Cereb Blood Flow Metab 2017; 37:1046-1059. [PMID: 27683451 PMCID: PMC5363487 DOI: 10.1177/0271678x16669514] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Aerobic activity has been shown highly beneficial to brain health, yet much uncertainty still surrounds the effects of exercise on the functioning of cerebral microvasculature. This study used two-photon fluorescence microscopy to examine cerebral hemodynamic alterations as well as accompanying geometric changes in the cortical microvascular network following five weeks of voluntary exercise in transgenic mice endogenously expressing tdTomato in vascular endothelial cells to allow visualization of microvessels irrespective of their perfusion levels. We found a diminished microvascular response to a hypercapnic challenge (10% FiCO2) in running mice when compared to that in nonrunning controls despite commensurate increases in transcutaneous CO2 tension. The flow increase to hypercapnia in runners was 70% lower than that in nonrunners (p = 0.0070) and the runners' arteriolar red blood cell speed changed by only half the amount seen in nonrunners (p = 0.0085). No changes were seen in resting hemodynamics or in the systemic physiological parameters measured. Although a few unperfused new vessels were observed on visual inspection, running did not produce significant morphological differences in the microvascular morphometric parameters, quantified following semiautomated tracking of the microvascular networks. We propose that voluntary running led to increased cortical microvascular efficiency and desensitization to CO2 elevation.
Collapse
Affiliation(s)
| | | | | | - Illsung L Joo
- 1 Sunnybrook Research Institute, Toronto, Canada.,2 Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Joe Steinman
- 2 Department of Medical Biophysics, University of Toronto, Toronto, Canada.,3 Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Canada
| | - Lindsay S Cahill
- 3 Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Canada
| | - John G Sled
- 2 Department of Medical Biophysics, University of Toronto, Toronto, Canada.,3 Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Canada
| | - Bojana Stefanovic
- 1 Sunnybrook Research Institute, Toronto, Canada.,2 Department of Medical Biophysics, University of Toronto, Toronto, Canada
| |
Collapse
|
103
|
Watanabe H, Shitara Y, Aoki Y, Inoue T, Tsuchida S, Takahashi N, Taga G. Hemoglobin phase of oxygenation and deoxygenation in early brain development measured using fNIRS. Proc Natl Acad Sci U S A 2017; 114:E1737-E1744. [PMID: 28196885 PMCID: PMC5338505 DOI: 10.1073/pnas.1616866114] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
A crucial issue in neonatal medicine is the impact of preterm birth on the developmental trajectory of the brain. Although a growing number of studies have shown alterations in the structure and function of the brain in preterm-born infants, we propose a method to detect subtle differences in neurovascular and metabolic functions in neonates and infants. Functional near-infrared spectroscopy (fNIRS) was used to obtain time-averaged phase differences between spontaneous low-frequency (less than 0.1 Hz) oscillatory changes in oxygenated hemoglobin (oxy-Hb) and those in deoxygenated hemoglobin (deoxy-Hb). This phase difference was referred to as hemoglobin phase of oxygenation and deoxygenation (hPod) in the cerebral tissue of sleeping neonates and infants. We examined hPod in term, late preterm, and early preterm infants with no evidence of clinical issues and found that all groups of infants showed developmental changes in the values of hPod from an in-phase to an antiphase pattern. Comparison of hPod among the groups revealed that developmental changes in hPod in early preterm infants precede those in late preterm and term infants at term equivalent age but then, progress at a slower pace. This study suggests that hPod measured using fNIRS is sensitive to the developmental stage of the integration of circular, neurovascular, and metabolic functions in the brains of neonates and infants.
Collapse
Affiliation(s)
- Hama Watanabe
- Graduate School of Education, The University of Tokyo, Tokyo 113-0033, Japan;
| | - Yoshihiko Shitara
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Yoshinori Aoki
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Takanobu Inoue
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Shinya Tsuchida
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Naoto Takahashi
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Gentaro Taga
- Graduate School of Education, The University of Tokyo, Tokyo 113-0033, Japan
| |
Collapse
|
104
|
Wälchli T, Ulmann-Schuler A, Hintermüller C, Meyer E, Stampanoni M, Carmeliet P, Emmert MY, Bozinov O, Regli L, Schwab ME, Vogel J, Hoerstrup SP. Nogo-A regulates vascular network architecture in the postnatal brain. J Cereb Blood Flow Metab 2017; 37:614-631. [PMID: 27927704 PMCID: PMC5381465 DOI: 10.1177/0271678x16675182] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Recently, we discovered a new role for the well-known axonal growth inhibitory molecule Nogo-A as a negative regulator of angiogenesis in the developing central nervous system. However, how Nogo-A affected the three-dimensional (3D) central nervous system (CNS) vascular network architecture remained unknown. Here, using vascular corrosion casting, hierarchical, synchrotron radiation μCT-based network imaging and computer-aided network analysis, we found that genetic ablation of Nogo-A significantly increased the three-dimensional vascular volume fraction in the postnatal day 10 (P10) mouse brain. More detailed analysis of the cerebral cortex revealed that this effect was mainly due to an increased number of capillaries and capillary branchpoints. Interestingly, other vascular parameters such as vessel diameter, -length, -tortuosity, and -volume were comparable between both genotypes for non-capillary vessels and capillaries. Taken together, our three-dimensional data showing more vessel segments and branchpoints at unchanged vessel morphology suggest that stimulated angiogenesis upon Nogo-A gene deletion results in the insertion of complete capillary micro-networks and not just single vessels into existing vascular networks. These findings significantly enhance our understanding of how angiogenesis, vascular remodeling, and three-dimensional vessel network architecture are regulated during central nervous system development. Nogo-A may therefore be a potential novel target for angiogenesis-dependent central nervous system pathologies such as brain tumors or stroke.
Collapse
Affiliation(s)
- Thomas Wälchli
- 1 Group of CNS Angiogenesis and Neurovascular Link, and Physician-Scientist Program, Institute for Regenerative Medicine, Neuroscience Center Zurich, and Division of Neurosurgery, University and University Hospital Zurich, Switzerland, and Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.,2 Division of Neurosurgery and Laboratory of Molecular Neuro-Oncology, University Hospital Zurich, Zurich, Switzerland.,3 Brain Research Institute, University of Zurich and Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | | | | | - Eric Meyer
- 3 Brain Research Institute, University of Zurich and Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Marco Stampanoni
- 6 Swiss Light Source, Paul Scherrer Institute, Villigen, Switzerland.,7 Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
| | - Peter Carmeliet
- 8 Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium.,9 Department of Oncology, Laboratory of Angiogenesis and Neurovascular Link, Leuven, Belgium
| | - Maximilian Y Emmert
- 10 Institute for Regenerative Medicine and Clinic for Cardiovascular Surgery, University Hospital Zurich.,11 Wyss Translational Center Zurich, University of Zurich and Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Oliver Bozinov
- 2 Division of Neurosurgery and Laboratory of Molecular Neuro-Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Luca Regli
- 2 Division of Neurosurgery and Laboratory of Molecular Neuro-Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Martin E Schwab
- 3 Brain Research Institute, University of Zurich and Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Johannes Vogel
- 12 Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Simon P Hoerstrup
- 10 Institute for Regenerative Medicine and Clinic for Cardiovascular Surgery, University Hospital Zurich.,11 Wyss Translational Center Zurich, University of Zurich and Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| |
Collapse
|
105
|
Figueiró-Filho EA, Croy BA, Reynolds JN, Dang F, Piro D, Rätsep MT, Forkert ND, Paolozza A, Smith GN, Stroman PW. Diffusion Tensor Imaging of White Matter in Children Born from Preeclamptic Gestations. AJNR Am J Neuroradiol 2017; 38:801-806. [PMID: 28126749 DOI: 10.3174/ajnr.a5064] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 11/06/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND PURPOSE Individuals born from pregnancies complicated by preeclampsia have an elevated risk for cognitive impairment. Deviations in maternal plasma angiokines occur for prolonged intervals before clinical signs of preeclampsia. We hypothesized that fetal brain vascular and nervous tissue development become deviated during maternal progression toward preeclampsia and that such deviations would be detectable by MR imaging. MATERIALS AND METHODS In this pilot study, 10 matched (gestational and current ages) pairs (5 boys/5 girls, 7-10 years of age) from preeclampsia or control pregnancies were examined by using diffusion tensor MR imaging. An unbiased voxel-based analysis was conducted on fractional anisotropy and mean diffusivity parametric maps. Six brain ROIs were identified for subsequent analysis by tractography (middle occipital gyrus, caudate nucleus and precuneus, cerebellum, superior longitudinal fasciculus, and cingulate gyrus). RESULTS Statistical differences were present between groups for fractional anisotropy in the caudate nucleus (offspring from preeclamptic gestation > controls), volume of the tract for the superior longitudinal fasciculus (offspring from preeclamptic gestation > controls) and the caudate nucleus (offspring from preeclamptic gestation > controls), and for parallel diffusivity of the cingulate gyrus (offspring from preeclamptic gestation > controls). CONCLUSIONS These novel preliminary results along with previous results from the same children that identified altered cerebral vessel calibers and increased regional brain volumes justify fully powered MR imaging studies to address the impact of preeclampsia on human fetal brain development.
Collapse
Affiliation(s)
- E A Figueiró-Filho
- From the Departments of Biomedical and Molecular Sciences (E.A.F.-F., B.A.C., J.N.R., F.D., D.P., N.D.F., G.N.S.) .,Centre for Neuroscience Studies (E.A.F.-F., J.N.R., P.W.S.), Queen's University, Kingston, Ontario, Canada.,Faculty of Medicine (E.A.F.-F.), Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - B A Croy
- From the Departments of Biomedical and Molecular Sciences (E.A.F.-F., B.A.C., J.N.R., F.D., D.P., N.D.F., G.N.S.)
| | - J N Reynolds
- From the Departments of Biomedical and Molecular Sciences (E.A.F.-F., B.A.C., J.N.R., F.D., D.P., N.D.F., G.N.S.).,Centre for Neuroscience Studies (E.A.F.-F., J.N.R., P.W.S.), Queen's University, Kingston, Ontario, Canada
| | - F Dang
- From the Departments of Biomedical and Molecular Sciences (E.A.F.-F., B.A.C., J.N.R., F.D., D.P., N.D.F., G.N.S.)
| | - D Piro
- From the Departments of Biomedical and Molecular Sciences (E.A.F.-F., B.A.C., J.N.R., F.D., D.P., N.D.F., G.N.S.)
| | - M T Rätsep
- Obstetrics and Gynecology (M.T.R., G.N.S.)
| | - N D Forkert
- From the Departments of Biomedical and Molecular Sciences (E.A.F.-F., B.A.C., J.N.R., F.D., D.P., N.D.F., G.N.S.).,Department of Radiology and Hotchkiss Brain Institute (N.D.F.), University of Calgary, Calgary, Alberta, Canada
| | - A Paolozza
- Laboratory for Infant Studies (A.P.), University of Toronto Scarborough, Scarborough, Ontario, Canada
| | - G N Smith
- From the Departments of Biomedical and Molecular Sciences (E.A.F.-F., B.A.C., J.N.R., F.D., D.P., N.D.F., G.N.S.).,Obstetrics and Gynecology (M.T.R., G.N.S.)
| | - P W Stroman
- Centre for Neuroscience Studies (E.A.F.-F., J.N.R., P.W.S.), Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
106
|
Brain microvasculature defects and Glut1 deficiency syndrome averted by early repletion of the glucose transporter-1 protein. Nat Commun 2017; 8:14152. [PMID: 28106060 PMCID: PMC5263887 DOI: 10.1038/ncomms14152] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 12/03/2016] [Indexed: 12/14/2022] Open
Abstract
Haploinsufficiency of the SLC2A1 gene and paucity of its translated product, the glucose transporter-1 (Glut1) protein, disrupt brain function and cause the neurodevelopmental disorder, Glut1 deficiency syndrome (Glut1 DS). There is little to suggest how reduced Glut1 causes cognitive dysfunction and no optimal treatment for Glut1 DS. We used model mice to demonstrate that low Glut1 protein arrests cerebral angiogenesis, resulting in a profound diminution of the brain microvasculature without compromising the blood-brain barrier. Studies to define the temporal requirements for Glut1 reveal that pre-symptomatic, AAV9-mediated repletion of the protein averts brain microvasculature defects and prevents disease, whereas augmenting the protein late, during adulthood, is devoid of benefit. Still, treatment following symptom onset can be effective; Glut1 repletion in early-symptomatic mutants that have experienced sustained periods of low brain glucose nevertheless restores the cerebral microvasculature and ameliorates disease. Timely Glut1 repletion may thus constitute an effective treatment for Glut1 DS.
Collapse
|
107
|
Maki T. Novel roles of oligodendrocyte precursor cells in the developing and damaged brain. ACTA ACUST UNITED AC 2017. [DOI: 10.1111/cen3.12358] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Takakuni Maki
- Department of Neurology; Graduate School of Medicine; Kyoto University; Kyoto Japan
| |
Collapse
|
108
|
Arango-Lievano M, Giannoni P, Claeysen S, Marchi N, Jeanneteau F. Longitudinal In Vivo Imaging of the Cerebrovasculature: Relevance to CNS Diseases. J Vis Exp 2016. [PMID: 28060355 DOI: 10.3791/54796] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Remodeling of the brain vasculature is a common trait of brain pathologies. In vivo imaging techniques are fundamental to detect cerebrovascular plasticity or damage occurring overtime and in relation to neuronal activity or blood flow. In vivo two-photon microscopy allows the study of the structural and functional plasticity of large cellular units in the living brain. In particular, the thinned-skull window preparation allows the visualization of cortical regions of interest (ROI) without inducing significant brain inflammation. Repetitive imaging sessions of cortical ROI are feasible, providing the characterization of disease hallmarks over time during the progression of numerous CNS diseases. This technique accessing the pial structures within 250 μm of the brain relies on the detection of fluorescent probes encoded by genetic cellular markers and/or vital dyes. The latter (e.g., fluorescent dextrans) are used to map the luminal compartment of cerebrovascular structures. Germane to the protocol described herein is the use of an in vivo marker of amyloid deposits, Methoxy-O4, to assess Alzheimer's disease (AD) progression. We also describe the post-acquisition image processing used to track vascular changes and amyloid depositions. While focusing presently on a model of AD, the described protocol is relevant to other CNS disorders where pathological cerebrovascular changes occur.
Collapse
Affiliation(s)
| | - Patrizia Giannoni
- Inserm, U1191, Institute of Functional Genomics; CNRS, UMR-5203; Université de Montpellier
| | - Sylvie Claeysen
- Inserm, U1191, Institute of Functional Genomics; CNRS, UMR-5203; Université de Montpellier
| | - Nicola Marchi
- Inserm, U1191, Institute of Functional Genomics; CNRS, UMR-5203; Université de Montpellier;
| | - Freddy Jeanneteau
- Inserm, U1191, Institute of Functional Genomics; CNRS, UMR-5203; Université de Montpellier;
| |
Collapse
|
109
|
Zuo XN, He Y, Betzel RF, Colcombe S, Sporns O, Milham MP. Human Connectomics across the Life Span. Trends Cogn Sci 2016; 21:32-45. [PMID: 27865786 DOI: 10.1016/j.tics.2016.10.005] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 10/10/2016] [Accepted: 10/17/2016] [Indexed: 01/19/2023]
Abstract
Connectomics has enhanced our understanding of neurocognitive development and decline by the integration of network sciences into studies across different stages of the human life span. However, these studies commonly occurred independently, missing the opportunity to test integrated models of the dynamical brain organization across the entire life span. In this review article, we survey empirical findings in life-span connectomics and propose a generative framework for computationally modeling the connectome over the human life span. This framework highlights initial findings that across the life span, the human connectome gradually shifts from an 'anatomically driven' organization to one that is more 'topological'. Finally, we consider recent advances that are promising to provide an integrative and systems perspective of human brain plasticity as well as underscore the pitfalls and challenges.
Collapse
Affiliation(s)
- Xi-Nian Zuo
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Beijing, China; Magnetic Resonance Imaging Research Center, Institute of Psychology, Beijing, China; Lifespan Connectomics and Behavior Team, Institute of Psychology, Beijing, China; Key Laboratory for Brain and Education Sciences, Guangxi Teachers Education University, Nanning, Guangxi, China; Center for Longevity Research, Guangxi Teachers Education University, Nanning, Guangxi, China.
| | - Ye He
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Beijing, China; Magnetic Resonance Imaging Research Center, Institute of Psychology, Beijing, China; Lifespan Connectomics and Behavior Team, Institute of Psychology, Beijing, China; Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Richard F Betzel
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Stan Colcombe
- Nathan S. Kline Institute for Psychiatric Research, Orangeburg, SC, USA
| | - Olaf Sporns
- Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Michael P Milham
- Nathan S. Kline Institute for Psychiatric Research, Orangeburg, SC, USA; Center for the Developing Brain, Child Mind Institute, New York, NY, USA.
| |
Collapse
|
110
|
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: 6.8] [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
|
111
|
Reeson P, Jeffery A, Brown CE. Illuminating the Effects of Stroke on the Diabetic Brain: Insights From Imaging Neural and Vascular Networks in Experimental Animal Models. Diabetes 2016; 65:1779-88. [PMID: 27329953 DOI: 10.2337/db16-0064] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 03/31/2016] [Indexed: 11/13/2022]
Abstract
Type 1 diabetes is known to cause circulatory problems in the eyes, heart, and limbs, and the brain is no exception. Because of the insidious effects of diabetes on brain circulation, patients with diabetes are two to four times more likely to have an ischemic stroke and are less likely to regain functions that are lost. To provide a more mechanistic understanding of this clinically significant problem, imaging studies have focused on how stroke affects neural and vascular networks in experimental models of type 1 diabetes. The emerging picture is that diabetes leads to maladaptive changes in the cerebrovascular system that ultimately limit neuronal rewiring and recovery of functions after stroke. At the cellular and systems level, diabetes is associated with abnormal cerebral blood flow in surviving brain regions and greater disruption of the blood-brain barrier. The abnormal vascular responses to stroke can be partly attributed to aberrant vascular endothelial growth factor (VEGF) signaling because genetic or pharmacological inhibition of VEGF signaling can mitigate vascular dysfunction and improve stroke recovery in diabetic animals. These experimental studies offer new insights and strategies for optimizing stroke recovery in diabetic populations.
Collapse
Affiliation(s)
- Patrick Reeson
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Andrew Jeffery
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Craig E Brown
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada Department of Biology, University of Victoria, Victoria, British Columbia, Canada Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
112
|
Kozberg M, Hillman E. Neurovascular coupling and energy metabolism in the developing brain. PROGRESS IN BRAIN RESEARCH 2016; 225:213-42. [PMID: 27130418 DOI: 10.1016/bs.pbr.2016.02.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In the adult brain, increases in local neural activity are almost always accompanied by increases in local blood flow. However, many functional imaging studies of the newborn and developing human brain have observed patterns of hemodynamic responses that differ from adult responses. Among the proposed mechanisms for the observed variations is that neurovascular coupling itself is still developing in the perinatal brain. Many of the components thought to be involved in actuating and propagating this hemodynamic response are known to still be developing postnatally, including perivascular cells such as astrocytes and pericytes. Both neural and vascular networks expand and are then selectively pruned over the first year of human life. Additionally, the metabolic demands of the newborn brain are still evolving. These changes are highly likely to affect early postnatal neurovascular coupling, and thus may affect functional imaging signals in this age group. This chapter will discuss the literature relating to neurovascular development. Potential effects of normal and aberrant development of neurovascular coupling on the newborn brain will also be explored, as well as ways to effectively utilize imaging techniques that rely on hemodynamic modulation such as fMRI and NIRS in younger populations.
Collapse
Affiliation(s)
- M Kozberg
- Columbia University, New York, NY, United States.
| | - E Hillman
- Columbia University, New York, NY, United States; Kavli Institute for Brain Science, Columbia University, New York, NY, United States; Mortimer B. Zuckerman Institute for Mind Brain and Behavior, Columbia University, New York, NY, United States.
| |
Collapse
|
113
|
COX-2-Derived Prostaglandin E2 Produced by Pyramidal Neurons Contributes to Neurovascular Coupling in the Rodent Cerebral Cortex. J Neurosci 2015; 35:11791-810. [PMID: 26311764 DOI: 10.1523/jneurosci.0651-15.2015] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Vasodilatory prostaglandins play a key role in neurovascular coupling (NVC), the tight link between neuronal activity and local cerebral blood flow, but their precise identity, cellular origin and the receptors involved remain unclear. Here we show in rats that NMDA-induced vasodilation and hemodynamic responses evoked by whisker stimulation involve cyclooxygenase-2 (COX-2) activity and activation of the prostaglandin E2 (PgE2) receptors EP2 and EP4. Using liquid chromatography-electrospray ionization-tandem mass spectrometry, we demonstrate that PgE2 is released by NMDA in cortical slices. The characterization of PgE2 producing cells by immunohistochemistry and single-cell reverse transcriptase-PCR revealed that pyramidal cells and not astrocytes are the main cell type equipped for PgE2 synthesis, one third expressing COX-2 systematically associated with a PgE2 synthase. Consistent with their central role in NVC, in vivo optogenetic stimulation of pyramidal cells evoked COX-2-dependent hyperemic responses in mice. These observations identify PgE2 as the main prostaglandin mediating sensory-evoked NVC, pyramidal cells as their principal source and vasodilatory EP2 and EP4 receptors as their targets. SIGNIFICANCE STATEMENT Brain function critically depends on a permanent spatiotemporal match between neuronal activity and blood supply, known as NVC. In the cerebral cortex, prostaglandins are major contributors to NVC. However, their biochemical identity remains elusive and their cellular origins are still under debate. Although astrocytes can induce vasodilations through the release of prostaglandins, the recruitment of this pathway during sensory stimulation is questioned. Using multidisciplinary approaches from single-cell reverse transcriptase-PCR, mass spectrometry, to ex vivo and in vivo pharmacology and optogenetics, we provide compelling evidence identifying PgE2 as the main prostaglandin in NVC, pyramidal neurons as their main cellular source and the vasodilatory EP2 and EP4 receptors as their main targets. These original findings will certainly change the current view of NVC.
Collapse
|
114
|
Schmidt-Kastner R. Genomic approach to selective vulnerability of the hippocampus in brain ischemia–hypoxia. Neuroscience 2015; 309:259-79. [DOI: 10.1016/j.neuroscience.2015.08.034] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Revised: 08/12/2015] [Accepted: 08/17/2015] [Indexed: 01/06/2023]
|
115
|
Abstract
The vascular and the nervous system are responsible for oxygen, nutrient, and information transfer and thereby constitute highly important communication systems in higher organisms. These functional similarities are reflected at the anatomical, cellular, and molecular levels, where common developmental principles and mutual crosstalks have evolved to coordinate their action. This resemblance of the two systems at different levels of complexity has been termed the "neurovascular link." Most of the evidence demonstrating neurovascular interactions derives from studies outside the CNS and from the CNS tissue of the retina. However, little is known about the specific properties of the neurovascular link in the brain. Here, we focus on regulatory effects of molecules involved in the neurovascular link on angiogenesis in the periphery and in the brain and distinguish between general and CNS-specific cues for angiogenesis. Moreover, we discuss the emerging molecular interactions of these angiogenic cues with the VEGF-VEGFR-Delta-like ligand 4 (Dll4)-Jagged-Notch pathway.
Collapse
|
116
|
Kang HM, Sohn I, Jung J, Jeong JW, Park C. Age-related changes in pial arterial structure and blood flow in mice. Neurobiol Aging 2015; 37:161-170. [PMID: 26460142 DOI: 10.1016/j.neurobiolaging.2015.09.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 09/10/2015] [Accepted: 09/11/2015] [Indexed: 10/23/2022]
Abstract
Age-related cerebral blood flow decreases are thought to deteriorate cognition and cause senescence, although the related mechanism is unclear. To investigate the relationships between aging and changes in cerebral blood flow and vasculature, we obtained fluorescence images of young (2-month-old) and old (12-month-old) mice using indocyanine green (ICG). First, we found that the blood flow in old mice's brains is lower than that in young mice and that old mice had more curved pial arteries and fewer pial artery junctions than young mice. Second, using Western blotting, we determined that the ratio of collagen to elastin (related to cerebral vascular wall distensibility) increased with age. Finally, we found that the peak ICG intensity and blood flow index decreased, whereas the mean transit time increased, with age in the middle cerebral artery and superior sagittal sinus. Age-related changes in pial arterial structure and composition, concurrent with the observed changes in the blood flow parameters, suggest that age-related changes in the cerebral vasculature structure and distensibility may induce altered brain blood flow.
Collapse
Affiliation(s)
- Hye-Min Kang
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Inkyung Sohn
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Junyang Jung
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Joo-Won Jeong
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Chan Park
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Seoul, Korea.
| |
Collapse
|
117
|
Hill J, Cave J. Targeting the vasculature to improve neural progenitor transplant survival. Transl Neurosci 2015; 6:162-167. [PMID: 28123800 PMCID: PMC4936624 DOI: 10.1515/tnsci-2015-0016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 08/05/2015] [Indexed: 12/18/2022] Open
Abstract
Neural progenitor transplantation is a promising therapeutic option for several neurological diseases and injuries. In nearly all human clinical trials and animal models that have tested this strategy, the low survival rate of progenitors after engraftment remains a significant challenge to overcome. Developing methods to improve the survival rate will reduce the number of cells required for transplant and will likely enhance functional improvements produced by the procedure. Here we briefly review the close relationship between the blood vasculature and neural progenitors in both the embryo and adult nervous system. We also discuss previous studies that have explored the role of the vasculature and hypoxic pre-conditioning in neural transplants. From these studies, we suggest that hypoxic pre-conditioning of a progenitor pool containing both neural and endothelial cells will improve engrafted transplanted neuronal survival rates.
Collapse
Affiliation(s)
- Justin Hill
- Burke Medical Research Institute, 785 Mamaroneck Ave, White Plains, NY 10605, USA; Burke Rehabilitation Hospital, 785 Mamaroneck Ave, White Plains, NY 10605, USA; Brain and Mind Research Institute, Weill Cornell Medical College, 1300 York Ave, New York, NY 10605, USA
| | - John Cave
- Burke Medical Research Institute, 785 Mamaroneck Ave, White Plains, NY 10605, USA; Brain and Mind Research Institute, Weill Cornell Medical College, 1300 York Ave, New York, NY 10605, USA
| |
Collapse
|
118
|
COX-2-Derived Prostaglandin E2 Produced by Pyramidal Neurons Contributes to Neurovascular Coupling in the Rodent Cerebral Cortex. J Neurosci 2015. [PMID: 26311764 DOI: 10.1523/jneurosci.0651‐15.2015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Vasodilatory prostaglandins play a key role in neurovascular coupling (NVC), the tight link between neuronal activity and local cerebral blood flow, but their precise identity, cellular origin and the receptors involved remain unclear. Here we show in rats that NMDA-induced vasodilation and hemodynamic responses evoked by whisker stimulation involve cyclooxygenase-2 (COX-2) activity and activation of the prostaglandin E2 (PgE2) receptors EP2 and EP4. Using liquid chromatography-electrospray ionization-tandem mass spectrometry, we demonstrate that PgE2 is released by NMDA in cortical slices. The characterization of PgE2 producing cells by immunohistochemistry and single-cell reverse transcriptase-PCR revealed that pyramidal cells and not astrocytes are the main cell type equipped for PgE2 synthesis, one third expressing COX-2 systematically associated with a PgE2 synthase. Consistent with their central role in NVC, in vivo optogenetic stimulation of pyramidal cells evoked COX-2-dependent hyperemic responses in mice. These observations identify PgE2 as the main prostaglandin mediating sensory-evoked NVC, pyramidal cells as their principal source and vasodilatory EP2 and EP4 receptors as their targets. SIGNIFICANCE STATEMENT Brain function critically depends on a permanent spatiotemporal match between neuronal activity and blood supply, known as NVC. In the cerebral cortex, prostaglandins are major contributors to NVC. However, their biochemical identity remains elusive and their cellular origins are still under debate. Although astrocytes can induce vasodilations through the release of prostaglandins, the recruitment of this pathway during sensory stimulation is questioned. Using multidisciplinary approaches from single-cell reverse transcriptase-PCR, mass spectrometry, to ex vivo and in vivo pharmacology and optogenetics, we provide compelling evidence identifying PgE2 as the main prostaglandin in NVC, pyramidal neurons as their main cellular source and the vasodilatory EP2 and EP4 receptors as their main targets. These original findings will certainly change the current view of NVC.
Collapse
|
119
|
Di Marco LY, Venneri A, Farkas E, Evans PC, Marzo A, Frangi AF. Vascular dysfunction in the pathogenesis of Alzheimer's disease--A review of endothelium-mediated mechanisms and ensuing vicious circles. Neurobiol Dis 2015; 82:593-606. [PMID: 26311408 DOI: 10.1016/j.nbd.2015.08.014] [Citation(s) in RCA: 190] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 07/23/2015] [Accepted: 08/17/2015] [Indexed: 12/17/2022] Open
Abstract
Late-onset dementia is a major health concern in the ageing population. Alzheimer's disease (AD) accounts for the largest proportion (65-70%) of dementia cases in the older population. Despite considerable research effort, the pathogenesis of late-onset AD remains unclear. Substantial evidence suggests that the neurodegenerative process is initiated by chronic cerebral hypoperfusion (CCH) caused by ageing and cardiovascular conditions. CCH causes reduced oxygen, glucose and other nutrient supply to the brain, with direct damage not only to the parenchymal cells, but also to the blood-brain barrier (BBB), a key mediator of cerebral homeostasis. BBB dysfunction mediates the indirect neurotoxic effects of CCH by promoting oxidative stress, inflammation, paracellular permeability, and dysregulation of nitric oxide, a key regulator of regional blood flow. As such, BBB dysfunction mediates a vicious circle in which cerebral perfusion is reduced further and the neurodegenerative process is accelerated. Endothelial interaction with pericytes and astrocytes could also play a role in the process. Reciprocal interactions between vascular dysfunction and neurodegeneration could further contribute to the development of the disease. A comprehensive overview of the complex scenario of interacting endothelium-mediated processes is currently lacking, and could prospectively contribute to the identification of adequate therapeutic interventions. This study reviews the current literature of in vitro and ex vivo studies on endothelium-mediated mechanisms underlying vascular dysfunction in AD pathogenesis, with the aim of presenting a comprehensive overview of the complex network of causative relationships. Particular emphasis is given to vicious circles which can accelerate the process of neurovascular degeneration.
Collapse
Affiliation(s)
- Luigi Yuri Di Marco
- Centre for Computational Imaging and Simulation Technologies in Biomedicine (CISTIB), Department of Electronic and Electrical Engineering, University of Sheffield, Sheffield, UK.
| | - Annalena Venneri
- Department of Neuroscience, Medical School, University of Sheffield, Sheffield, UK; IRCCS San Camillo Foundation Hospital, Venice, Italy
| | - Eszter Farkas
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Paul C Evans
- Department of Cardiovascular Science, Medical School, University of Sheffield, Sheffield, UK
| | - Alberto Marzo
- Centre for Computational Imaging and Simulation Technologies in Biomedicine (CISTIB), Department of Mechanical Engineering, University of Sheffield, Sheffield, UK
| | - Alejandro F Frangi
- Centre for Computational Imaging and Simulation Technologies in Biomedicine (CISTIB), Department of Electronic and Electrical Engineering, University of Sheffield, Sheffield, UK
| |
Collapse
|
120
|
Winkler EA, Sagare AP, Zlokovic BV. The pericyte: a forgotten cell type with important implications for Alzheimer's disease? Brain Pathol 2015; 24:371-86. [PMID: 24946075 DOI: 10.1111/bpa.12152] [Citation(s) in RCA: 192] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 05/13/2014] [Indexed: 12/13/2022] Open
Abstract
Pericytes are cells in the blood-brain barrier (BBB) that degenerate in Alzheimer's disease (AD), a neurodegenerative disorder characterized by early neurovascular dysfunction, elevation of amyloid β-peptide (Aβ), tau pathology and neuronal loss, leading to progressive cognitive decline and dementia. Pericytes are uniquely positioned within the neurovascular unit between endothelial cells of brain capillaries, astrocytes and neurons. Recent studies have shown that pericytes regulate key neurovascular functions including BBB formation and maintenance, vascular stability and angioarchitecture, regulation of capillary blood flow, and clearance of toxic cellular by-products necessary for normal functioning of the central nervous system (CNS). Here, we review the concept of the neurovascular unit and neurovascular functions of CNS pericytes. Next, we discuss vascular contributions to AD and review new roles of pericytes in the pathogenesis of AD such as vascular-mediated Aβ-independent neurodegeneration, regulation of Aβ clearance and contributions to tau pathology, neuronal loss and cognitive decline. We conclude that future studies should focus on molecular mechanisms and pathways underlying aberrant signal transduction between pericytes and its neighboring cells within the neurovascular unit, that is, endothelial cells, astrocytes and neurons, which could represent potential therapeutic targets to control pericyte degeneration in AD and the resulting secondary vascular and neuronal degeneration.
Collapse
Affiliation(s)
- Ethan A Winkler
- Zilkha Neurogenetic Institute, University of Southern California Keck School of Medicine, Los Angeles, CA; Department of Neurosurgery, University of California San Francisco, San Francisco, CA
| | | | | |
Collapse
|
121
|
Bastian TW, Santarriaga S, Nguyen TA, Prohaska JR, Georgieff MK, Anderson GW. Fetal and neonatal iron deficiency but not copper deficiency increases vascular complexity in the developing rat brain. Nutr Neurosci 2015; 18:365-75. [PMID: 26177275 DOI: 10.1179/1476830515y.0000000037] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVES Anemia caused by nutritional deficiencies, such as iron and copper deficiencies, is a global health problem. Iron and copper deficiencies have their most profound effect on the developing fetus/infant, leading to brain development deficits and poor cognitive outcomes. Tissue iron depletion or chronic anemia can induce cellular hypoxic signaling. In mice, chronic hypoxia induces a compensatory increase in brain blood vessel outgrowth. We hypothesized that developmental anemia, due to iron or copper deficiencies, induces angiogenesis/vasculogenesis in the neonatal brain. METHODS To test our hypothesis, three independent experiments were performed where pregnant rats were fed iron- or copper-deficient diets from gestational day 2 through mid-lactation. Effects on the neonatal brain vasculature were determined using quantitative real-time polymerase chain reaction to assess mRNA levels of angiogenesis/vasculogenesis-associated genes and GLUT1 immunohistochemistry to assess brain blood vessel density and complexity. RESULTS Iron deficiency, but not copper deficiency, increased mRNA expression of brain endothelial cell- and angiogenesis/vasculogenesis-associated genes (i.e. Glut1, Vwf, Vegfa, Ang2, Cxcl12, and Flk1) in the neonatal brain, suggesting increased cerebrovascular density. Iron deficiency also increased hippocampal and cerebral cortical blood vessel branching by 62 and 78%, respectively. DISCUSSION This study demonstrates increased blood vessel complexity in the neonatal iron-deficient brain, which is likely due to elevated angiogenic/vasculogenic signaling. At least initially, this is probably an adaptive response to maintain metabolic substrate homeostasis in the developing iron-deficient brain. However, this may also contribute to long-term neurodevelopmental deficits.
Collapse
MESH Headings
- Anemia, Iron-Deficiency/blood
- Anemia, Iron-Deficiency/complications
- Animals
- Animals, Newborn
- Cerebral Cortex/blood supply
- Cerebral Cortex/growth & development
- Ceruloplasmin/metabolism
- Copper/blood
- Copper/deficiency
- Disease Models, Animal
- Female
- Fetus
- Gene Expression Regulation, Developmental
- Hemoglobins/metabolism
- Hippocampus/blood supply
- Hippocampus/growth & development
- Male
- Neovascularization, Pathologic/blood
- Neovascularization, Pathologic/etiology
- Pregnancy
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
Collapse
|
122
|
Janota C, Lemere CA, Brito MA. Dissecting the Contribution of Vascular Alterations and Aging to Alzheimer's Disease. Mol Neurobiol 2015; 53:3793-3811. [PMID: 26143259 DOI: 10.1007/s12035-015-9319-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Accepted: 06/24/2015] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by cognitive decline that afflicts as many as 45 % of individuals who survive past the age of 85. AD has been associated with neurovascular dysfunction and brain accumulation of amyloid-β peptide, as well as tau phosphorylation and neurodegeneration, but the pathogenesis of the disease is still somewhat unclear. According to the amyloid cascade hypothesis of AD, accumulation of amyloid-β peptide (Aβ) aggregates initiates a sequence of events leading to neuronal injury and loss, and dementia. Alternatively, the vascular hypothesis of AD incorporates the vascular contribution to the disease, stating that a primary insult to brain microcirculation (e.g., stroke) not only contributes to amyloidopathy but initiates a non-amyloidogenic pathway of vascular-mediated neuronal dysfunction and injury, which involves blood-brain barrier compromise, with increased permeability of blood vessels, leakage of blood-borne components into the brain, and, consequently, neurotoxicity. Vascular dysfunction also includes a diminished brain capillary flow, causing multiple focal ischemic or hypoxic microinjuries, diminished amyloid-β clearance, and formation of neurotoxic oligomers, which lead to neuronal dysfunction. Here we present and discuss relevant findings on the contribution of vascular alterations during aging to AD, with the hope that a better understanding of the players in the "orchestra" of neurodegeneration will be useful in developing therapies to modulate the "symphony".
Collapse
Affiliation(s)
- Cátia Janota
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal
| | - Cynthia A Lemere
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, 77 Avenue Louis Pasteur (NRB 636F), Boston, MA, 02115, USA
| | - Maria Alexandra Brito
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal. .,Department of Biochemistry and Human Biology, Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal.
| |
Collapse
|
123
|
Hill RA, Tong L, Yuan P, Murikinati S, Gupta S, Grutzendler J. Regional Blood Flow in the Normal and Ischemic Brain Is Controlled by Arteriolar Smooth Muscle Cell Contractility and Not by Capillary Pericytes. Neuron 2015; 87:95-110. [PMID: 26119027 PMCID: PMC4487786 DOI: 10.1016/j.neuron.2015.06.001] [Citation(s) in RCA: 495] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 04/24/2015] [Accepted: 05/22/2015] [Indexed: 01/25/2023]
Abstract
The precise regulation of cerebral blood flow is critical for normal brain function, and its disruption underlies many neuropathologies. The extent to which smooth muscle-covered arterioles or pericyte-covered capillaries control vasomotion during neurovascular coupling remains controversial. We found that capillary pericytes in mice and humans do not express smooth muscle actin and are morphologically and functionally distinct from adjacent precapillary smooth muscle cells (SMCs). Using optical imaging we investigated blood flow regulation at various sites on the vascular tree in living mice. Optogenetic, whisker stimulation, or cortical spreading depolarization caused microvascular diameter or flow changes in SMC but not pericyte-covered microvessels. During early stages of brain ischemia, transient SMC but not pericyte constrictions were a major cause of hypoperfusion leading to thrombosis and distal microvascular occlusions. Thus, capillary pericytes are not contractile, and regulation of cerebral blood flow in physiological and pathological conditions is mediated by arteriolar SMCs.
Collapse
Affiliation(s)
- Robert A Hill
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neurobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Lei Tong
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neurobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Peng Yuan
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neurobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sasidhar Murikinati
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neurobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Shobhana Gupta
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neurobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Jaime Grutzendler
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neurobiology, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
124
|
Wang S, Chen L, Zhang L, Huang C, Xiu Y, Wang F, Zhou C, Luo Y, Xiao Q, Tang Y. Effects of long-term exercise on spatial learning, memory ability, and cortical capillaries in aged rats. Med Sci Monit 2015; 21:945-54. [PMID: 25828032 PMCID: PMC4395020 DOI: 10.12659/msm.893935] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Background This study aimed to determine the effects of long-term running exercise on spatial learning, spatial memory, and cortical capillaries in aged rats. Material/Methods Fourteen-month-old female and male Sprague-Dawley rats were randomly divided into an exercised group (EG) and a non-exercised group (NG). The EG rats were trained on treadmill running for 4 or 14 months. The NG rats were housed under identical conditions without running. Spatial learning and memory were assessed with the Morris water maze. The cortical capillary parameters were quantitatively investigated using immunohistochemical and stereological methods. Results The escaped latencies of the EG were significantly different from those of the NG in 18-month-old females and 28-month-old males (p<0.05). However, 28-month-old females and 18-month-old males showed no differences in escape latency between the EG and NG (p>0.05). In 28-month-old female rats, stereological techniques showed significant differences between the EG and NG in the cortical capillary volume (median, 22.55 vs. 11.42, p<0.05) and the cortical capillary surface area (median, 7474.13 vs. 3935.90, p<0.05). In 28-month-old male rats, the EG had a significantly longer total cortical capillary length (median, 530.35 vs. 156.27, p<0.05), significantly larger cortical capillary volume (median, 16.47 vs. 3.65, p<0.01), and a significantly larger cortical capillary total surface area (median, 7885.79 vs. 1957.16, p<0.01) compared with the NG group. Conclusions These data demonstrate that exercise improved spatial learning, memory capacity and cortical capillaries in aged rats.
Collapse
Affiliation(s)
- Sanrong Wang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, China (mainland)
| | - Lin Chen
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, China (mainland)
| | - Lei Zhang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, China (mainland)
| | - Chunxia Huang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, China (mainland)
| | - Yun Xiu
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, China (mainland)
| | - Feifei Wang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, China (mainland)
| | - Chunni Zhou
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, China (mainland)
| | - Yanmin Luo
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, China (mainland)
| | - Qian Xiao
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, China (mainland)
| | - Yong Tang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, China (mainland)
| |
Collapse
|
125
|
Leger PL, Bonnin P, Renolleau S, Baud O, Charriaut-Marlangue C. Ischemic postconditioning in cerebral ischemia: Differences between the immature and mature brain? Int J Dev Neurosci 2015; 45:39-43. [PMID: 25777940 DOI: 10.1016/j.ijdevneu.2015.03.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 03/12/2015] [Accepted: 03/12/2015] [Indexed: 10/23/2022] Open
Abstract
Ischemic postconditioning (postC), defined as serial mechanical interruptions of blood flow at reperfusion, effectively reduces myocardial infarct size in all species tested so far, including humans. In the brain, ischemic postC leads to controversial results regardless of variations in factors such as onset time of beginning, the duration of ischemia and/or reperfusion, and the number of cycles of occlusion/reperfusion. Thus, many major issues remain to be resolved regarding its protective effects. Future studies should aim to identify the parameters that yield the strongest protection, as well as to understand why the efficacy of ischemic postC differs between models. This review will focus on initial hemodynamic changes and their consequences, and on specific features such as NO-dependent vascular tone and/or prolonged acidosis in cerebral ischemia-reperfusion in order to better understand the dynamics of ischemic postC in the developing brain.
Collapse
Affiliation(s)
- Pierre-Louis Leger
- Univ. Paris Diderot, Sorbonne Paris Cité, INSERM UMR 1141, 75019 Paris, France; PremUp Foundation, 75006 Paris, France; UPMC-Paris6, AP-HP, Hôpital Armand Trousseau, Service de Réanimation Néonatale et Pédiatrique, 75012 Paris, France
| | - Philippe Bonnin
- Univ. Paris Diderot, Sorbonne Paris Cité, AP-HP, Hôpital Lariboisière, Physiologie Clinique, Explorations-Fonctionnelles, 75010 Paris, France; Univ. Paris Diderot, Sorbonne Paris Cité, INSERM, U965, 75010 Paris, France
| | - Sylvain Renolleau
- Univ. Paris Diderot, Sorbonne Paris Cité, INSERM UMR 1141, 75019 Paris, France; Univ. Paris Descartes, AP-HP, CHU Necker-Enfants Malades, Réanimation et USC médico-chirurgicales pédiatriques, 75015 Paris, France
| | - Olivier Baud
- Univ. Paris Diderot, Sorbonne Paris Cité, INSERM UMR 1141, 75019 Paris, France; PremUp Foundation, 75006 Paris, France
| | | |
Collapse
|
126
|
Boehme J, Maltepe E. Spare hypoxia, spoil the child? J Clin Invest 2015; 125:965-7. [PMID: 25689257 DOI: 10.1172/jci80820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
127
|
Pienaar IS, Lee CH, Elson JL, McGuinness L, Gentleman SM, Kalaria RN, Dexter DT. Deep-brain stimulation associates with improved microvascular integrity in the subthalamic nucleus in Parkinson's disease. Neurobiol Dis 2014; 74:392-405. [PMID: 25533682 DOI: 10.1016/j.nbd.2014.12.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 12/01/2014] [Accepted: 12/05/2014] [Indexed: 12/25/2022] Open
Abstract
Deep brain stimulation (DBS) of the subthalamic nucleus (STN) has become an accepted treatment for motor symptoms in a subset of Parkinson's disease (PD) patients. The mechanisms why DBS is effective are incompletely understood, but previous studies show that DBS targeted in brain structures other than the STN may modify the microvasculature. However, this has not been studied in PD subjects who have received STN-DBS. Here we investigated the extent and nature of microvascular changes in post-mortem STN samples from STN-DBS PD patients, compared to aged controls and PD patients who had not been treated with STN-DBS. We used immunohistochemical and immunofluorescent methods to assess serial STN-containing brain sections from PD and STN-DBS PD cases, compared to similar age controls using specific antibodies to detect capillaries, an adherens junction and tight junction-associated proteins as well as activated microglia. Cellular features in stained sections were quantified by confocal fluorescence microscopy and stereological methods in conjunction with in vitro imaging tools. We found significant upregulation of microvessel endothelial cell thickness, length and density but lowered activated microglia density and striking upregulation of all analysed adherens junction and tight junction-associated proteins in STN-DBS PD patients compared to non-DBS PD patients and controls. Moreover, in STN-DBS PD samples, expression of an angiogenic factor, vascular endothelial growth factor (VEGF), was significantly upregulated compared to the other groups. Our findings suggest that overexpressed VEGF and downregulation of inflammatory processes may be critical mechanisms underlying the DBS-induced microvascular changes.
Collapse
Affiliation(s)
- Ilse S Pienaar
- Centre for Neuroinflammation & Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, United Kingdom.
| | - Cecilia Heyne Lee
- The Sir William Dunn School of Pathology, South Parks Road, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - Joanna L Elson
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, United Kingdom; Centre for Human Metabonomics, North-West University, Potchefstroom, South Africa
| | - Louisa McGuinness
- Centre for Neuroinflammation & Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, United Kingdom
| | - Stephen M Gentleman
- Centre for Neuroinflammation & Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, United Kingdom
| | - Raj N Kalaria
- Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, United Kingdom
| | - David T Dexter
- Centre for Neuroinflammation & Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, United Kingdom
| |
Collapse
|
128
|
Letourneur A, Chen V, Waterman G, Drew PJ. A method for longitudinal, transcranial imaging of blood flow and remodeling of the cerebral vasculature in postnatal mice. Physiol Rep 2014; 2:2/12/e12238. [PMID: 25524276 PMCID: PMC4332216 DOI: 10.14814/phy2.12238] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
In the weeks following birth, both the brain and the vascular network that supplies it undergo dramatic alteration. While studies of the postnatal evolution of the pial vasculature and blood flow through its vessels have been previously done histologically or acutely, here we describe a neonatal reinforced thin‐skull preparation for longitudinally imaging the development of the pial vasculature in mice using two‐photon laser scanning microscopy. Starting with mice as young as postnatal day 2 (P2), we are able to chronically image cortical areas >1 mm2, repeatedly for several consecutive days, allowing us to observe the remodeling of the pial arterial and venous networks. We used this method to measure blood velocity in individual vessels over multiple days, and show that blood flow through individual pial venules was correlated with subsequent diameter changes. This preparation allows the longitudinal imaging of the developing mammalian cerebral vascular network and its physiology. We developed a technique to longitudinally image blood vessels in the neonatal mouse cortex transcranially using two‐photon microscopy. The blood vessels on the surface of the brain undergo substantial pruning after birth. Blood flow through a vessel was correlated with the subsequent diameter change of the vessel.
Collapse
Affiliation(s)
- Annelise Letourneur
- Department of Engineering Science and Mechanics, Center for Neural Engineering, Pennsylvania State University, University Park, Pennsylvania CNRS, CEA, Université de Caen Basse-Normandie, UMR 6301 ISTCT, CERVOxy. GIP CYCERON, Caen, France
| | - Victoria Chen
- Department of Engineering Science and Mechanics, Center for Neural Engineering, Pennsylvania State University, University Park, Pennsylvania
| | - Gar Waterman
- Department of Engineering Science and Mechanics, Center for Neural Engineering, Pennsylvania State University, University Park, Pennsylvania
| | - Patrick J Drew
- Department of Engineering Science and Mechanics, Center for Neural Engineering, Pennsylvania State University, University Park, Pennsylvania Department of Neurosurgery, Pennsylvania State University, University Park, Pennsylvania
| |
Collapse
|
129
|
Quantitative assessment of angiogenesis, perfused blood vessels and endothelial tip cells in the postnatal mouse brain. Nat Protoc 2014; 10:53-74. [PMID: 25502884 DOI: 10.1038/nprot.2015.002] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
During development and in various diseases of the CNS, new blood vessel formation starts with endothelial tip cell selection and vascular sprout migration, followed by the establishment of functional, perfused blood vessels. Here we describe a method that allows the assessment of these distinct angiogenic steps together with antibody-based protein detection in the postnatal mouse brain. Intravascular and perivascular markers such as Evans blue (EB), isolectin B4 (IB4) or laminin (LN) are used alongside simultaneous immunofluorescence on the same sections. By using confocal laser-scanning microscopy and stereological methods for analysis, detailed quantification of the 3D postnatal brain vasculature for perfused and nonperfused vessels (e.g., vascular volume fraction, vessel length and number, number of branch points and perfusion status of the newly formed vessels) and characterization of sprouting activity (e.g., endothelial tip cell density, filopodia number) can be obtained. The entire protocol, from mouse perfusion to vessel analysis, takes ∼10 d.
Collapse
|
130
|
Grutzendler J, Murikinati S, Hiner B, Ji L, Lam CK, Yoo T, Gupta S, Hafler BP, Adelman RA, Yuan P, Rodriguez G. Angiophagy prevents early embolus washout but recanalizes microvessels through embolus extravasation. Sci Transl Med 2014; 6:226ra31. [PMID: 24598589 DOI: 10.1126/scitranslmed.3006585] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Occlusion of the microvasculature by blood clots, atheromatous fragments, or circulating debris is a frequent phenomenon in most human organs. Emboli are cleared from the microvasculature by hemodynamic pressure and the fibrinolytic system. An alternative mechanism of clearance is angiophagy, in which emboli are engulfed by the endothelium and translocate through the microvascular wall. We report that endothelial lamellipodia surround emboli within hours of occlusion, markedly reducing hemodynamic washout and tissue plasminogen activator-mediated fibrinolysis in mice. Over the next few days, emboli are completely engulfed by the endothelium and extravasated into the perivascular space, leading to vessel recanalization and blood flow reestablishment. We find that this mechanism is not limited to the brain, as previously thought, but also occurs in the heart, retina, kidney, and lung. In the lung, emboli cross into the alveolar space where they are degraded by macrophages, whereas in the kidney, they enter the renal tubules, constituting potential routes for permanent removal of circulating debris. Retina photography and angiography in patients with embolic occlusions provide indirect evidence suggesting that angiophagy may also occur in humans. Thus, angiophagy appears to be a ubiquitous mechanism that could be a therapeutic target with broad implications in vascular occlusive disorders. Given its biphasic nature-initially causing embolus retention, and subsequently driving embolus extravasation-it is likely that different therapeutic strategies will be required during these distinct post-occlusion time windows.
Collapse
Affiliation(s)
- Jaime Grutzendler
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Sim J, Johnson RS. A whiter shade of gray: HIF and coordination of angiogenesis with postnatal myelination. Dev Cell 2014; 30:116-7. [PMID: 25073152 DOI: 10.1016/j.devcel.2014.07.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Oligodendrocyte precursor cells are the primary source of myelinating oligodendrocytes in the neonatal CNS. In a recent issue of Cell, Yuen and coworkers (2014) show that hypoxia-inducible factors and Wnt7a/7b act in developing white matter, promoting angiogenesis in anticipation of the metabolic demands of oligogenesis, before finally allowing precursor differentiation.
Collapse
Affiliation(s)
- Jingwei Sim
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
| | - Randall S Johnson
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK.
| |
Collapse
|
132
|
Fumagalli S, Ortolano F, De Simoni MG. A close look at brain dynamics: Cells and vessels seen by in vivo two-photon microscopy. Prog Neurobiol 2014; 121:36-54. [DOI: 10.1016/j.pneurobio.2014.06.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 06/17/2014] [Accepted: 06/29/2014] [Indexed: 01/11/2023]
|
133
|
Adams DL, Piserchia V, Economides JR, Horton JC. Vascular Supply of the Cerebral Cortex is Specialized for Cell Layers but Not Columns. Cereb Cortex 2014; 25:3673-81. [PMID: 25246513 DOI: 10.1093/cercor/bhu221] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The vascular supply to layers and columns was compared in macaque primary visual cortex (V1) by labeling red blood cells via their endogenous peroxidase activity. Alternate sections were processed for cytochrome oxidase to reveal "patches" or "blobs," which anchor the interdigitated column systems of striate cortex. More densely populated cell layers received the most profuse blood supply. In the superficial layers the blood supply was organized into microvascular lobules, consisting of a central venule surrounded by arterioles. Each vessel was identified as an arteriole or venule by matching it with the penetration site where it entered the cortex from a parent arteriole or venule in the pial circulation. Although microvascular lobules and cytochrome oxidase patches had a similar periodicity, they bore no mutual relationship. The size and density of penetrating arterioles and venules did not differ between patches and interpatches. The red blood cell labeling in patches and interpatches was equal. Moreover, patches and interpatches were supplied by an anastomotic pial arteriole system, with no segregation of blood supply to the two compartments. Often a focal constriction was present at the origin of pial arterial branches, indicating that local control of cortical perfusion may be accomplished by vascular sphincters.
Collapse
Affiliation(s)
- Daniel L Adams
- Beckman Vision Center, University of California, San Francisco, San Francisco, CA 94143, USA Center for Mind/Brain Sciences, The University of Trento, Trento, Italy
| | | | - John R Economides
- Beckman Vision Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jonathan C Horton
- Beckman Vision Center, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
134
|
Lacoste B, Comin CH, Ben-Zvi A, Kaeser PS, Xu X, Costa LDF, Gu C. Sensory-related neural activity regulates the structure of vascular networks in the cerebral cortex. Neuron 2014; 83:1117-30. [PMID: 25155955 DOI: 10.1016/j.neuron.2014.07.034] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2014] [Indexed: 11/15/2022]
Abstract
Neurovascular interactions are essential for proper brain function. While the effect of neural activity on cerebral blood flow has been extensively studied, whether or not neural activity influences vascular patterning remains elusive. Here, we demonstrate that neural activity promotes the formation of vascular networks in the early postnatal mouse barrel cortex. Using a combination of genetics, imaging, and computational tools to allow simultaneous analysis of neuronal and vascular components, we found that vascular density and branching were decreased in the barrel cortex when sensory input was reduced by either a complete deafferentation, a genetic impairment of neurotransmitter release at thalamocortical synapses, or a selective reduction of sensory-related neural activity by whisker plucking. In contrast, enhancement of neural activity by whisker stimulation led to an increase in vascular density and branching. The finding that neural activity is necessary and sufficient to trigger alterations of vascular networks reveals an important feature of neurovascular interactions.
Collapse
Affiliation(s)
- Baptiste Lacoste
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Cesar H Comin
- IFSC, University of Sao Paulo, Sao Carlos, SP, Brazil
| | - Ayal Ben-Zvi
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Xiaoyin Xu
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Chenghua Gu
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
135
|
Transcranial optical vascular imaging (TOVI) of cortical hemodynamics in mouse brain. Sci Rep 2014; 4:5839. [PMID: 25059112 PMCID: PMC5394480 DOI: 10.1038/srep05839] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 07/09/2014] [Indexed: 12/03/2022] Open
Abstract
In vivo imaging of cerebral vasculature and blood flow provides highly valuable information for clinicians as well as researchers. Nevertheless, currently available methods are complex, time-consuming and expensive. Here, we present a novel, minimally invasive method for vascular imaging through the sufficiently transparent intact skull of young mice. Our method combines laser speckle and fluorescent imaging with dynamic color mapping and image fusion. Quickly generated wide-field images present clear visual information on blood flow and perfusion in the cerebral cortex and meninges. The ability of the method to visualize hemodynamic changes is demonstrated by induced occlusion of the middle cerebral artery. The compact and easily operated system comprises of several pieces of standard and affordable laboratory equipment. This simple, robust and inexpensive method may become an important tool for assessment of brain hemodynamics in preclinical studies.
Collapse
|
136
|
Yuen TJ, Silbereis JC, Griveau A, Chang SM, Daneman R, Fancy SPJ, Zahed H, Maltepe E, Rowitch DH. Oligodendrocyte-encoded HIF function couples postnatal myelination and white matter angiogenesis. Cell 2014; 158:383-396. [PMID: 25018103 DOI: 10.1016/j.cell.2014.04.052] [Citation(s) in RCA: 275] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Revised: 03/13/2014] [Accepted: 04/22/2014] [Indexed: 12/30/2022]
Abstract
Myelin sheaths provide critical functional and trophic support for axons in white matter tracts of the brain. Oligodendrocyte precursor cells (OPCs) have extraordinary metabolic requirements during development as they differentiate to produce multiple myelin segments, implying that they must first secure adequate access to blood supply. However, mechanisms that coordinate myelination and angiogenesis are unclear. Here, we show that oxygen tension, mediated by OPC-encoded hypoxia-inducible factor (HIF) function, is an essential regulator of postnatal myelination. Constitutive HIF1/2α stabilization resulted in OPC maturation arrest through autocrine activation of canonical Wnt7a/7b. Surprisingly, such OPCs also show paracrine activity that induces excessive postnatal white matter angiogenesis in vivo and directly stimulates endothelial cell proliferation in vitro. Conversely, OPC-specific HIF1/2α loss of function leads to insufficient angiogenesis in corpus callosum and catastrophic axon loss. These findings indicate that OPC-intrinsic HIF signaling couples postnatal white matter angiogenesis, axon integrity, and the onset of myelination in mammalian forebrain.
Collapse
Affiliation(s)
- Tracy J Yuen
- Department of Pediatrics, Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine and Howard Hughes Medical Institute, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - John C Silbereis
- Department of Pediatrics, Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine and Howard Hughes Medical Institute, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA; Neuroscience Graduate Program, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Amelie Griveau
- Department of Pediatrics, Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine and Howard Hughes Medical Institute, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Sandra M Chang
- Department of Pediatrics, Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine and Howard Hughes Medical Institute, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Richard Daneman
- Department of Anatomy, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Stephen P J Fancy
- Department of Pediatrics, Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine and Howard Hughes Medical Institute, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Hengameh Zahed
- Department of Pediatrics, Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine and Howard Hughes Medical Institute, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA; Medical Science Training Program, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Emin Maltepe
- Division of Neonatology, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - David H Rowitch
- Department of Pediatrics, Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine and Howard Hughes Medical Institute, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA; Division of Neonatology, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA.
| |
Collapse
|
137
|
Escudero C, Roberts JM, Myatt L, Feoktistov I. Impaired adenosine-mediated angiogenesis in preeclampsia: potential implications for fetal programming. Front Pharmacol 2014; 5:134. [PMID: 24926270 PMCID: PMC4046493 DOI: 10.3389/fphar.2014.00134] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 05/16/2014] [Indexed: 01/24/2023] Open
Abstract
Preeclampsia is a pregnancy-specific syndrome, defined by such clinical hallmarks as the onset of maternal hypertension and proteinuria after 20 weeks of gestation. The syndrome is also characterized by impaired blood flow through the utero-placental circulation and relative placental ischemia, which in turn, may generate feto-placental endothelial dysfunction. Endothelial dysfunction in offspring born from preeclamptic pregnancies has been associated with an increased risk of cardiovascular disease, including hypertension, later in life. Interestingly, diminished endothelial function, manifested by low angiogenic capacity, leads to hypertension in animal studies. Recently, we have shown that the adenosine receptor A2A/nitric oxide/vascular endothelial growth factor axis is reduced in human umbilical vein endothelial cells derived from preeclamptic pregnancies, an effect correlated with gestational age at onset of preeclampsia. We and others suggested that impaired vascular function might be associated with high cardiovascular risk in offspring exposed to pregnancy diseases. However, we are not aware of any studies that examine impaired adenosine-mediated angiogenesis as a possible link to hypertension in offspring born from preeclamptic pregnancies. In this review, we present evidence supporting the hypothesis that reduced adenosine-mediated angiogenesis during preeclamptic pregnancies might be associated with development of hypertension in the offspring.
Collapse
Affiliation(s)
- Carlos Escudero
- Vascular Physiology Laboratory, Group of Investigation in Tumor Angiogenesis, Group of Research and Innovation in Vascular Health, Department of Basic Sciences, Faculty of Sciences, Universidad del Bío-Bío Chillán, Chile
| | - James M Roberts
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology, and Reproductive Sciences, Department of Epidemiology and Clinical and Translational Science Institute, University of Pittsburgh Pittsburgh, PA, USA
| | - Leslie Myatt
- Center for Pregnancy and Newborn Research, University of Texas Health Science Center San Antonio, TX, USA
| | - Igor Feoktistov
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Nashville, TN, USA ; Department of Pharmacology, School of Medicine, Vanderbilt University Nashville, TN, USA
| |
Collapse
|
138
|
Katsimpardi L, Litterman NK, Schein PA, Miller CM, Loffredo FS, Wojtkiewicz GR, Chen JW, Lee RT, Wagers AJ, Rubin LL. Vascular and neurogenic rejuvenation of the aging mouse brain by young systemic factors. Science 2014; 344:630-4. [PMID: 24797482 PMCID: PMC4123747 DOI: 10.1126/science.1251141] [Citation(s) in RCA: 756] [Impact Index Per Article: 75.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In the adult central nervous system, the vasculature of the neurogenic niche regulates neural stem cell behavior by providing circulating and secreted factors. Age-related decline of neurogenesis and cognitive function is associated with reduced blood flow and decreased numbers of neural stem cells. Therefore, restoring the functionality of the niche should counteract some of the negative effects of aging. We show that factors found in young blood induce vascular remodeling, culminating in increased neurogenesis and improved olfactory discrimination in aging mice. Further, we show that GDF11 alone can improve the cerebral vasculature and enhance neurogenesis. The identification of factors that slow the age-dependent deterioration of the neurogenic niche in mice may constitute the basis for new methods of treating age-related neurodegenerative and neurovascular diseases.
Collapse
Affiliation(s)
- Lida Katsimpardi
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA
- Harvard Stem Cell Institute, Cambridge, MA
| | - Nadia K. Litterman
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA
- Harvard Stem Cell Institute, Cambridge, MA
| | - Pamela A. Schein
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA
- Harvard Stem Cell Institute, Cambridge, MA
| | - Christine M. Miller
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Howard Hughes Medical Institute, Joslin Diabetes Center and the Paul F. Glenn Laboratories for the Biological Mechanisms of Aging, Harvard Medical School
| | - Francesco S. Loffredo
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - Gregory R. Wojtkiewicz
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital, Harvard Medical School
| | - John W. Chen
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital, Harvard Medical School
| | - Richard T. Lee
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - Amy J. Wagers
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Howard Hughes Medical Institute, Joslin Diabetes Center and the Paul F. Glenn Laboratories for the Biological Mechanisms of Aging, Harvard Medical School
| | - Lee L. Rubin
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA
- Harvard Stem Cell Institute, Cambridge, MA
| |
Collapse
|
139
|
Schain AJ, Hill RA, Grutzendler J. Label-free in vivo imaging of myelinated axons in health and disease with spectral confocal reflectance microscopy. Nat Med 2014; 20:443-9. [PMID: 24681598 PMCID: PMC3981936 DOI: 10.1038/nm.3495] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 07/02/2013] [Indexed: 12/14/2022]
Abstract
We report a newly developed technique for high-resolution in vivo imaging of myelinated axons in the brain, spinal cord and peripheral nerve that requires no fluorescent labeling. This method, based on spectral confocal reflectance microscopy (SCoRe), uses a conventional laser-scanning confocal system to generate images by merging the simultaneously reflected signals from multiple lasers of different wavelengths. Striking color patterns unique to individual myelinated fibers are generated that facilitate their tracing in dense axonal areas. These patterns highlight nodes of Ranvier and Schmidt-Lanterman incisures and can be used to detect various myelin pathologies. Using SCoRe we carried out chronic brain imaging up to 400 μm deep, capturing de novo myelination of mouse cortical axons in vivo. We also established the feasibility of imaging myelinated axons in the human cerebral cortex. SCoRe adds a powerful component to the evolving toolbox for imaging myelination in living animals and potentially in humans.
Collapse
Affiliation(s)
- Aaron J. Schain
- Yale University School of Medicine, Department of Neurology, 300
George St. Suite 8201, New Haven, CT 06511
| | - Robert A. Hill
- Yale University School of Medicine, Department of Neurology, 300
George St. Suite 8201, New Haven, CT 06511
| | - Jaime Grutzendler
- Yale University School of Medicine, Department of Neurology, 300
George St. Suite 8201, New Haven, CT 06511
- Yale University School of Medicine, Department of Neurobiology, 300
George St. Suite 8201, New Haven, CT 06511
| |
Collapse
|
140
|
Masamoto K, Takuwa H, Seki C, Taniguchi J, Itoh Y, Tomita Y, Toriumi H, Unekawa M, Kawaguchi H, Ito H, Suzuki N, Kanno I. Microvascular sprouting, extension, and creation of new capillary connections with adaptation of the neighboring astrocytes in adult mouse cortex under chronic hypoxia. J Cereb Blood Flow Metab 2014; 34:325-31. [PMID: 24252848 PMCID: PMC3915210 DOI: 10.1038/jcbfm.2013.201] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 10/16/2013] [Accepted: 10/23/2013] [Indexed: 11/09/2022]
Abstract
The present study aimed to determine the spatiotemporal dynamics of microvascular and astrocytic adaptation during hypoxia-induced cerebral angiogenesis. Adult C57BL/6J and Tie2-green fluorescent protein (GFP) mice with vascular endothelial cells expressing GFP were exposed to normobaric hypoxia for 3 weeks, whereas the three-dimensional microvessels and astrocytes were imaged repeatedly using two-photon microscopy. After 7 to 14 days of hypoxia, a vessel sprout appeared from the capillaries with a bump-like head shape (mean diameter 14 μm), and stagnant blood cells were seen inside the sprout. However, no detectable changes in the astrocyte morphology were observed for this early phase of the hypoxia adaptation. More than 50% of the sprouts emerged from capillaries 60 μm away from the center penetrating arteries, which indicates that the capillary distant from the penetrating arteries is a favored site for sprouting. After 14 to 21 days of hypoxia, the sprouting vessels created a new connection with an existing capillary. In this phase, the shape of the new vessel and its blood flow were normalized, and the outside of the vessels were wrapped with numerous processes from the neighboring astrocytes. The findings indicate that hypoxia-induced cerebral angiogenesis provokes the adaptation of neighboring astrocytes, which may stabilize the blood-brain barrier in immature vessels.
Collapse
Affiliation(s)
- Kazuto Masamoto
- 1] Brain Science Inspired Life Support Research Center, University of Electro-Communications, Chofu, Tokyo, Japan [2] Molecular Imaging Center, National Institute of Radiological Sciences, Inage, Chiba, Japan
| | - Hiroyuki Takuwa
- Molecular Imaging Center, National Institute of Radiological Sciences, Inage, Chiba, Japan
| | - Chie Seki
- Molecular Imaging Center, National Institute of Radiological Sciences, Inage, Chiba, Japan
| | - Junko Taniguchi
- Molecular Imaging Center, National Institute of Radiological Sciences, Inage, Chiba, Japan
| | - Yoshiaki Itoh
- Department of Neurology, School of Medicine Keio University, Shinjuku, Tokyo, Japan
| | - Yutaka Tomita
- Department of Neurology, School of Medicine Keio University, Shinjuku, Tokyo, Japan
| | - Haruki Toriumi
- Department of Neurology, School of Medicine Keio University, Shinjuku, Tokyo, Japan
| | - Miyuki Unekawa
- Department of Neurology, School of Medicine Keio University, Shinjuku, Tokyo, Japan
| | - Hiroshi Kawaguchi
- Molecular Imaging Center, National Institute of Radiological Sciences, Inage, Chiba, Japan
| | - Hiroshi Ito
- Molecular Imaging Center, National Institute of Radiological Sciences, Inage, Chiba, Japan
| | - Norihiro Suzuki
- Department of Neurology, School of Medicine Keio University, Shinjuku, Tokyo, Japan
| | - Iwao Kanno
- Molecular Imaging Center, National Institute of Radiological Sciences, Inage, Chiba, Japan
| |
Collapse
|
141
|
Abstract
Stroke usually affects people with underlying medical conditions. In particular, diabetics are significantly more likely to have a stroke and the prognosis for recovery is poor. Because diabetes is associated with degenerative changes in the vasculature of many organs, we sought to determine how hyperglycemia affects blood flow dynamics after an ischemic stroke. Longitudinal in vivo two-photon imaging was used to track microvessels before and after photothrombotic stroke in a diabetic mouse model. Chronic hyperglycemia exacerbated acute (3-7 d) ischemia-induced increases in blood flow velocity, vessel lumen diameter, and red blood cell flux in peri-infarct regions. These changes in blood flow dynamics were most evident in superficial blood vessels within 500 μm from the infarct, rather than deeper or more distant cortical regions. Long-term imaging of diabetic mice not subjected to stroke indicated that these acute stroke-related changes in vascular function could not be attributed to complications from hyperglycemia alone. Treating diabetic mice with insulin immediately after stroke resulted in less severe alterations in blood flow within the first 7 d of recovery, but had more variable results at later time points. Analysis of microvessel branching patterns revealed that stroke led to a pruning of microvessels in peri-infarct cortex, with very few instances of sprouting. These results indicate that chronic hyperglycemia significantly affects the vascular response to ischemic stroke and that insulin only partially mitigates these changes. The combination of these acute and chronic alterations in blood flow dynamics could underlie diabetes-related deficits in cortical plasticity and stroke recovery.
Collapse
|
142
|
Whiteus C, Freitas C, Grutzendler J. Perturbed neural activity disrupts cerebral angiogenesis during a postnatal critical period. Nature 2013; 505:407-11. [PMID: 24305053 PMCID: PMC3947100 DOI: 10.1038/nature12821] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 10/29/2013] [Indexed: 11/09/2022]
Abstract
During the neonatal period, activity-dependent neural-circuit remodelling coincides with growth and refinement of the cerebral microvasculature. Whether neural activity also influences the patterning of the vascular bed is not known. Here we show in neonatal mice, that neither reduction of sensory input through whisker trimming nor moderately increased activity by environmental enrichment affects cortical microvascular development. Unexpectedly, chronic stimulation by repetitive sounds, whisker deflection or motor activity led to a near arrest of angiogenesis in barrel, auditory and motor cortices, respectively. Chemically induced seizures also caused robust reductions in microvascular density. However, altering neural activity in adult mice did not affect the vasculature. Histological analysis and time-lapse in vivo two-photon microscopy revealed that hyperactivity did not lead to cell death or pruning of existing vessels but rather to reduced endothelial proliferation and vessel sprouting. This anti-angiogenic effect was prevented by administration of the nitric oxide synthase (NOS) inhibitor L-NAME and in mice with neuronal and inducible NOS deficiency, suggesting that excessive nitric oxide released from hyperactive interneurons and glia inhibited vessel growth. Vascular deficits persisted long after cessation of hyperstimulation, providing evidence for a critical period after which proper microvascular patterning cannot be re-established. Reduced microvascular density diminished the ability of the brain to compensate for hypoxic challenges, leading to dendritic spine loss in regions distant from capillaries. Therefore, excessive sensorimotor stimulation and repetitive neural activation during early childhood may cause lifelong deficits in microvascular reserve, which could have important consequences for brain development, function and pathology.
Collapse
Affiliation(s)
- Christina Whiteus
- 1] Department of Neurology, Yale University School of Medicine, New Haven, Connecticut 06511, USA [2] Department of Neurobiology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Catarina Freitas
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Jaime Grutzendler
- 1] Department of Neurology, Yale University School of Medicine, New Haven, Connecticut 06511, USA [2] Department of Neurobiology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| |
Collapse
|
143
|
Charriaut-Marlangue C, Bonnin P, Leger P, Renolleau S. Brief update on hemodynamic responses in animal models of neonatal stroke and hypoxia–ischemia. Exp Neurol 2013; 248:316-20. [DOI: 10.1016/j.expneurol.2013.06.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 06/25/2013] [Indexed: 11/30/2022]
|
144
|
Buga AM, Di Napoli M, Popa-Wagner A. Preclinical models of stroke in aged animals with or without comorbidities: role of neuroinflammation. Biogerontology 2013; 14:651-62. [PMID: 24057280 DOI: 10.1007/s10522-013-9465-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 09/10/2013] [Indexed: 12/31/2022]
Abstract
Age is the principal nonmodifiable risk factor for stroke. Over the past 10 years, suitable models for stroke in aged rats have been established. At genetic and cellular level there are significant differences in behavioral, cytological and genomics responses to injury in old animals as compared with the young ones. Behaviorally, the aged rats have the capacity to recover after cortical infarcts albeit to a lower extent than the younger counterparts. Similarly, the increased vulnerability of the aged brain to stroke, together with a decreased interhemisphere synchrony after stroke, assessed by different experimental methods (MRI, fMRI, in vivo microscopy, EEG) leads to unfavorable recovery of physical and cognitive functions in aged people and may have a prognostic value for the recovery of stroke patients. Furthermore, in elderly, comorbidities like diabetes or arterial hypertension are associated with higher risk of stroke, increased mortality and disability, and poorer functional status and quality of life. Aging brain reacts strongly to ischemia-reperfusion injury with an early inflammatory response. The process of cellular senescence can be an important additional contributor to chronic post-stroke by creating a "primed" inflammatory environment in the brain. Overall, these pro-inflammatory reactions promote early scar formation associated with tissue fibrosis and reduce functional recovery. A better understanding of molecular factors and signaling pathways underlying the contribution of comorbidities to stroke-induced pathological sequelae, may be translated into successful treatment or prevention therapies for age-associated diseases which would improve lifespan and quality of life.
Collapse
Affiliation(s)
- A-M Buga
- Department of Functional Sciences, Center of Clinical and Experimental Medicine, University of Medicine and Pharmacy, Craiova, Craiova, Romania
| | | | | |
Collapse
|
145
|
Grutzendler J. Angiophagy: mechanism of microvascular recanalization independent of the fibrinolytic system. Stroke 2013; 44:S84-6. [PMID: 23709741 DOI: 10.1161/strokeaha.112.678730] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Jaime Grutzendler
- Yale University School of Medicine, 300 George St, New Haven, CT 06511, USA.
| |
Collapse
|