Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Acta Neuropathol Commun ; 12(1): 45, 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38509621

ABSTRACT

Interactions between extracellular matrix (ECM) proteins and ß1 integrins play an essential role maintaining vascular integrity in the brain, particularly under vascular remodeling conditions. As blood vessels in the spinal cord are reported to have distinct properties from those in the brain, here we examined the impact of ß1 integrin inhibition on spinal cord vascular integrity, both under normoxic conditions, when blood vessels are stable, and during exposure to chronic mild hypoxia (CMH), when extensive vascular remodeling occurs. We found that a function-blocking ß1 integrin antibody triggered a small degree of vascular disruption in the spinal cord under normoxic conditions, but under hypoxic conditions, it greatly enhanced (20-fold) vascular disruption, preferentially in spinal cord white matter (WM). This resulted in elevated microglial activation as well as marked loss of myelin integrity and reduced density of oligodendroglial cells. To understand why vascular breakdown is localized to WM, we compared expression levels of major BBB components of WM and grey matter (GM) blood vessels, but this revealed no obvious differences. Interestingly however, hypoxyprobe staining demonstrated that the most severe levels of spinal cord hypoxia induced by CMH occurred in the WM. Analysis of brain tissue revealed a similar preferential vulnerability of WM tracts to show vascular disruption under these conditions. Taken together, these findings demonstrate an essential role for ß1 integrins in maintaining vascular integrity in the spinal cord, and unexpectedly, reveal a novel and fundamental difference between WM and GM blood vessels in their dependence on ß1 integrin function during hypoxic exposure. Our data support the concept that the preferential WM vulnerability described may be less a result of intrinsic differences in vascular barrier properties between WM and GM, and more a consequence of differences in vascular density and architecture.


Subject(s)
White Matter , Humans , White Matter/metabolism , Integrin beta1/metabolism , Vascular Remodeling/physiology , Spinal Cord/metabolism , Gray Matter/metabolism , Hypoxia/metabolism
2.
Microvasc Res ; 152: 104625, 2024 03.
Article in English | MEDLINE | ID: mdl-37979909

ABSTRACT

Previous studies have shown that expression of the endothelial laminin receptor α6ß4 integrin in the brain is uniquely restricted to arterioles. As exposure to chronic mild hypoxia (CMH, 8 % O2) stimulates robust angiogenic and arteriogenic remodeling responses in the brain, the goal of this study was to determine how CMH influences cerebrovascular expression of the ß4 integrin as well as its potential ligands, laminin 411 and 511, containing the α4 and α5 laminin subunits respectively, and then define how aging impacts this expression. We observed the following: (i) CMH launched a robust arteriogenic remodeling response both in the young (10 weeks) and aged (20 months) brain, correlating with an increased number of ß4 integrin+ vessels, (ii) while the laminin α4 subunit is expressed evenly across all cerebral blood vessels, laminin α5 was highly expressed preferentially on ß4 integrin+ arterioles, (iii) CMH-induced arteriolar remodeling was associated with strong downregulation of the laminin α4 subunit but no change in the laminin α5 subunit, (iv) in addition to its expression on arterioles, ß4 integrin was also expressed at lower levels on capillaries specifically in white matter (WM) tracts but not in the grey matter (GM), and (v), these observations were consistent in both the brain and spinal cord, and age had no obvious impact. Taken together, our findings suggest that laminin 511 may be a specific ligand for α6ß4 integrin and that dynamic switching of the laminin subunits α4 and α5 might play an instructive role in arteriogenic remodeling. Furthermore, ß4 integrin expression differentiates WM from GM capillaries, highlighting a novel and important difference.


Subject(s)
Integrin alpha6beta4 , Integrin beta4 , Humans , Arterioles/metabolism , Integrin alpha6beta4/metabolism , Laminin/metabolism , Hypoxia
3.
Fluids Barriers CNS ; 20(1): 52, 2023 Jul 03.
Article in English | MEDLINE | ID: mdl-37400852

ABSTRACT

BACKGROUND: Maintaining a tight blood-brain barrier (BBB) is an important prerequisite for the preservation of neurological health, though current evidence suggests it declines with age. While extracellular matrix-integrin interactions play critical roles in regulating the balance between vascular stability and remodeling, it remains to be established whether manipulation of integrin function weakens or strengthens vascular integrity. Indeed, recent reports have generated conflicting outcomes in this regard. METHODS: Here, in young (8-10 weeks) and aged (20 months) mice, we examined the impact of intraperitoneal injection of a function-blocking ß1 integrin antibody, both under normoxic conditions, when the BBB is stable, and during chronic mild hypoxic (CMH; 8% O2) conditions, when a vigorous vascular remodeling response is ongoing. Brain tissue was examined by immunofluorescence (IF) for markers of vascular remodeling and BBB disruption, and microglial activation and proliferation. Data were analyzed using one-way analysis of variance (ANOVA) followed by Tukey's multiple comparison post-hoc test. RESULTS: In both young and aged mice, ß1 integrin block greatly amplified hypoxia-induced vascular disruption, though it was much less under normoxic conditions. Interestingly, under both normoxic and hypoxic conditions, ß1 integrin antibody-induced BBB disruption was greater in young mice. Enhanced BBB breakdown was associated with increased levels of the leaky BBB marker MECA-32 and with greater loss of endothelial tight junction proteins and the adherens protein VE-cadherin. Surprisingly, ß1 integrin blockade did not reduce hypoxia-induced endothelial proliferation, nor did it prevent the hypoxia-associated increase in vascularity. Commensurate with the increased vascular disruption, ß1 integrin blockade enhanced microglial activation both in young and aged brain, though the impact was much greater in young brain. In vitro studies revealed that ß1 integrin blockade also reduced the integrity of a brain endothelial monolayer and triggered disruptions in tight junction proteins. CONCLUSIONS: These data demonstrate that ß1 integrin plays an essential role in maintaining BBB integrity, both under stable normoxic conditions and during hypoxia-induced vascular remodeling. As ß1 integrin blockade had a greater disruptive effect in young brain, effectively shifting the BBB phenotype of young brain towards that of the aged, we speculate that enhancing ß1 integrin function at the aged BBB may hold therapeutic potential by reverting the deteriorating BBB phenotype back towards that of the young.


Subject(s)
Blood-Brain Barrier , Integrin beta1 , Mice , Animals , Blood-Brain Barrier/metabolism , Integrin beta1/metabolism , Vascular Remodeling , Hypoxia/metabolism , Tight Junction Proteins/metabolism
4.
Int J Mol Sci ; 24(14)2023 Jul 08.
Article in English | MEDLINE | ID: mdl-37510999

ABSTRACT

In response to chronic mild hypoxia (CMH, 8% O2), spinal cord blood vessels launch a robust angiogenic response that is associated with transient disruption of the blood-spinal cord barrier (BSCB) which, in turn, triggers a microglial vasculo-protective response. Because hypoxia occurs in many age-related conditions, the goal of this study was to define how aging influences these responses by comparing events in young (8-10 weeks) and aged (20 months) mice. This revealed that aged mice had much greater (3-4-fold) levels of hypoxic-induced BSCB disruption than young mice and that, while the early stage of the angiogenic response in aged mice was no different to young mice, the maturation of newly formed vessels was significantly delayed. Interestingly, microglia in the spinal cords of aged mice were much more activated than young mice, even under normoxic conditions, and this was further enhanced by CMH, though, surprisingly, this resulted in reduced microglial clustering around leaky blood vessels and diminished vasculo-protection. Vascular disruption was associated with loss of myelin in spinal cord white matter (WM) in both young and aged mice. Furthermore, it was notable that the spinal cord of aged mice contained a lower density of Olig2+ oligodendroglial cells even under normoxic conditions and that CMH significantly reduced the density of Olig2+ cells in spinal cord WM of the aged, but not the young, mice. These results demonstrate that spinal cord blood vessels of aged mice are much more vulnerable to the damaging effects of hypoxia than young mice, in part due to the reduced vasculo-protection conferred by chronically activated microglial cells. These observations may have implications for the pathogenesis and/or treatment of spinal cord diseases such as amyotrophic lateral sclerosis (ALS) and suggest that an improvement in microglial function could offer therapeutic potential for treating these age-related conditions.


Subject(s)
Amyotrophic Lateral Sclerosis , White Matter , Mice , Animals , Microglia/pathology , Spinal Cord/pathology , Amyotrophic Lateral Sclerosis/pathology , White Matter/pathology , Hypoxia
5.
Microvasc Res ; 148: 104517, 2023 07.
Article in English | MEDLINE | ID: mdl-36894025

ABSTRACT

Exposure to chronic mild hypoxia (CMH; 8-10% O2) promotes a robust vascular remodeling response in the brain resulting in 50% increased vessel density over a period of two weeks. It is currently unknown whether blood vessels in other organs show similar responses. To address this question, mice were exposed to CMH for 4 days and various markers of vascular remodeling were examined in the brain along with heart, skeletal muscle, kidney, and liver. In contrast to brain, where CMH strongly promoted endothelial proliferation, none of the peripheral organs showed this response and in heart and liver, CMH notably reduced endothelial proliferation. While the MECA-32 endothelial activation marker was strongly induced by CMH in brain, in peripheral organs it was constitutively expressed either on a sub-population of vessels (heart and skeletal muscle) or on all vessels (kidney and liver), and notably, CMH did not affect expression. Endothelial expression of the tight junction proteins claudin-5 and ZO-1 were markedly increased on cerebral vessels, but in the peripheral organs examined, CMH either had no effect or reduced ZO-1 expression (liver). Finally, while CMH had no impact on the number of Mac-1 positive macrophages in the brain, heart, or skeletal muscle, this number was markedly decreased in the kidney but increased in the liver. Our findings show that the vascular remodeling responses to CMH are organ-specific, with the brain showing a strong angiogenic response and enhanced tight junction protein expression, but heart, skeletal muscle, kidney, and liver failing to show these responses.


Subject(s)
Brain , Vascular Remodeling , Mice , Animals , Brain/metabolism , Hypoxia , Endothelium/metabolism
7.
Aging Cell ; 21(11): e13720, 2022 11.
Article in English | MEDLINE | ID: mdl-36130175

ABSTRACT

In a recent study of young mice, we showed that chronic mild hypoxia (CMH, 8% O2 ) triggers transient blood-brain barrier (BBB) disruption, and that microglia play an important vasculo-protective function in maintaining BBB integrity. As hypoxia is a common component of many age-related diseases, here we extended these studies to aged mice and found that hypoxia-induced vascular leak was greatly amplified (5-fold to 10-fold) in aged mice, being particularly high in the olfactory bulb and midbrain. While aged mice showed no obvious difference in the early stages of hypoxic angiogenic remodeling, the compensatory increase in vascularity and vessel maturation was significantly delayed. Compared with young brain, microglia in the normoxic aged brain were markedly activated, and this was further increased under hypoxic conditions, but paradoxically, this correlated with reduced vasculo-protection. Microglial depletion studies showed that microglial still play an important vasculo-protective role in aged brain, but interestingly, partial attenuation of microglial activation with minocycline resulted in fewer vascular leaks and reduced loss of endothelial tight junction proteins. Taken together, these findings suggest that increased BBB disruption in hypoxic aged mice can be explained both by a delayed vascular remodeling response and reduced microglial vasculo-protection. Importantly, they show that overly activated microglia in the aged brain are less effective at maintaining vascular integrity, though this can be improved by reducing microglial activation with minocycline, suggesting therapeutic potential for enhancing BBB integrity in the hypoxia-predisposed elderly population.


Subject(s)
Microglia , Minocycline , Aged , Humans , Animals , Mice , Microglia/metabolism , Minocycline/pharmacology , Brain/metabolism , Blood-Brain Barrier/metabolism , Hypoxia/metabolism
8.
Fluids Barriers CNS ; 18(1): 50, 2021 Nov 17.
Article in English | MEDLINE | ID: mdl-34789271

ABSTRACT

BACKGROUND: Chronic mild hypoxia (CMH, 8% O2) stimulates robust vascular remodelling in the brain, but it also triggers transient vascular disruption. This raises the fundamental question: is the vascular leak an unwanted side-effect of angiogenic remodelling or is it a pathological response, unrelated to endothelial proliferation, in which declining oxygen levels trigger endothelial dysfunction? METHODS: To answer this question, mice were exposed to CMH (8% O2) for periods up to 14 days, after which, brain tissue was examined by immunofluorescence (IF) to determine which type of blood vessel (arteriole, capillary or venule) was most commonly associated with endothelial proliferation and vascular leak and how this correlated with tight junction protein expression. Vascular perfusion was examined using DiI. Data were analysed using one-way analysis of variance (ANOVA) followed by Tukey's multiple comparison post-hoc test. RESULTS: The following was observed: (1) most endothelial proliferation and extravascular fibrinogen leak occurred in capillaries and to a lesser degree in venules, (2) much to our surprise, endothelial proliferation and extravascular fibrinogen leak never colocalized, (3) interestingly however, endothelial proliferation was strongly associated with an intravascular fibrinogen staining pattern not seen in stable blood vessels, (4) DiI perfusion studies revealed that angiogenic vessels were adequately perfused, suggesting that fibrinogen retention in angiogenic vessels is not due to temporary closure of the vessel, but more likely because fibrinogen is retained within the vessel wall, (5) bromodeoxyuridine (BrdU) labelling as a means to more permanently label proliferating endothelial cells, confirmed lack of any connection between endothelial proliferation and extravascular fibrinogen leak, while (6) in contrast, proliferating microglia were detected within extravascular leaks. CONCLUSIONS: Taken together, our findings support the concept that in the short-term, hypoxia-induced endothelial proliferation triggers transient fibrinogen deposition within the walls of angiogenic blood vessels, but no overt vascular leak occurs in these vessels. Importantly, endothelial proliferation and extravascular fibrinogen leaks never co-localize, demonstrating that extravascular leak is not an unwanted side-effect of angiogenic endothelial proliferation, but rather a dysfunctional vascular response to hypoxia that occurs in a distinct group of non-angiogenic blood vessels.


Subject(s)
Blood-Brain Barrier/physiopathology , Cerebrovascular Disorders/physiopathology , Endothelium, Vascular/physiopathology , Hypoxia/physiopathology , Neovascularization, Pathologic/physiopathology , Vascular Remodeling/physiology , Animals , Cerebrovascular Disorders/etiology , Disease Models, Animal , Female , Hypoxia/complications , Male , Mice , Mice, Inbred C57BL
9.
Acta Neuropathol Commun ; 8(1): 175, 2020 10 28.
Article in English | MEDLINE | ID: mdl-33115539

ABSTRACT

We recently demonstrated that when mice are exposed to chronic mild hypoxia (CMH, 8% O2), blood vessels in the spinal cord show transient vascular leak that is associated with clustering and activation of microglia around disrupted vessels. Importantly, microglial depletion profoundly increased hypoxia-induced vascular leak, implying that microglia play a critical role maintaining vascular integrity in the hypoxic spinal cord. The goal of the current study was to examine if microglia play a similar vasculo-protective function in the brain. Employing extravascular fibrinogen leak as an index of blood-brain barrier (BBB) disruption, we found that CMH provoked transient vascular leak in cerebral blood vessels that was associated with activation and aggregation of Mac-1-positive microglia around leaky vessels. Interestingly, CMH-induced vascular leak showed regional selectivity, being much more prevalent in the brainstem and olfactory bulb than the cerebral cortex and cerebellum. Pharmacological depletion of microglia with the colony stimulating factor-1 receptor inhibitor PLX5622, had no effect under normoxic conditions, but markedly increased hypoxia-induced cerebrovascular leak in all regions examined. As in the spinal cord, this was associated with endothelial induction of MECA-32, a marker of leaky CNS endothelium, and greater loss of endothelial tight junction proteins. Brain regions displaying the highest levels of hypoxic-induced vascular leak also showed the greatest levels of angiogenic remodeling, suggesting that transient BBB disruption may be an unwanted side-effect of hypoxic-induced angiogenic remodeling. As hypoxia is common to a multitude of human diseases including obstructive sleep apnea, lung disease, and age-related pulmonary, cardiac and cerebrovascular dysfunction, our findings have important translational implications. First, they point to a potential pathogenic role of chronic hypoxia in triggering BBB disruption and subsequent neurological dysfunction, and second, they demonstrate an important protective role for microglia in maintaining vascular integrity in the hypoxic brain.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/metabolism , Capillary Permeability/physiology , Fibrinogen/metabolism , Hypoxia/metabolism , Microglia/physiology , Animals , Antigens, Surface/metabolism , Blood-Brain Barrier/drug effects , Brain/blood supply , Brain/drug effects , Brain Stem/blood supply , Brain Stem/drug effects , Brain Stem/metabolism , Capillary Permeability/drug effects , Cerebellum/blood supply , Cerebellum/drug effects , Cerebellum/metabolism , Cerebral Cortex/blood supply , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebrovascular Circulation , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Hypoxia/physiopathology , Macrophage-1 Antigen/metabolism , Mice , Microglia/drug effects , Olfactory Bulb/blood supply , Olfactory Bulb/drug effects , Olfactory Bulb/metabolism , Organic Chemicals/pharmacology , Tight Junction Proteins/drug effects , Tight Junction Proteins/metabolism
10.
Proc Natl Acad Sci U S A ; 117(20): 11126-11135, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32371484

ABSTRACT

While several studies have shown that hypoxic preconditioning suppresses development of the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis (MS), no one has yet examined the important clinically relevant question of whether mild hypoxia can impact the progression of preexisting disease. Using a relapsing-remitting model of EAE, here we demonstrate that when applied to preexisting disease, chronic mild hypoxia (CMH, 10% O2) markedly accelerates clinical recovery, leading to long-term stable reductions in clinical score. At the histological level, CMH led to significant reductions in vascular disruption, leukocyte accumulation, and demyelination. Spinal cord blood vessels of CMH-treated mice showed reduced expression of the endothelial activation molecule VCAM-1 but increased expression of the endothelial tight junction proteins ZO-1 and occludin, key mechanisms underlying vascular integrity. Interestingly, while equal numbers of inflammatory leukocytes were present in the spinal cord at peak disease (day 14 postimmunization; i.e., 3 d after CMH started), apoptotic removal of infiltrated leukocytes during the remission phase was markedly accelerated in CMH-treated mice, as determined by increased numbers of monocytes positive for TUNEL and cleaved caspase-3. The enhanced monocyte apoptosis in CMH-treated mice was paralleled by increased numbers of HIF-1α+ monocytes, suggesting that CMH enhances monocyte removal by amplifying the hypoxic stress manifest within monocytes in acute inflammatory lesions. These data demonstrate that mild hypoxia promotes recovery from preexisting inflammatory demyelinating disease and suggest that this protection is primarily the result of enhanced vascular integrity and accelerated apoptosis of infiltrated monocytes.


Subject(s)
Apoptosis/physiology , Encephalomyelitis, Autoimmune, Experimental , Hypoxia/metabolism , Monocytes/metabolism , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Caspase 3 , Disease Models, Animal , Endothelium/metabolism , Female , Mice , Multiple Sclerosis , Occludin/metabolism , Spinal Cord/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Zonula Occludens-1 Protein/metabolism
11.
Exp Neurol ; 283(Pt A): 396-403, 2016 09.
Article in English | MEDLINE | ID: mdl-27412766

ABSTRACT

Activated protein C (APC) is a serine protease that promotes favorable changes in vascular barrier integrity and post-ischemic angiogenic remodeling in animal models of ischemic stroke, and its efficacy is currently being investigated in clinical ischemic stroke trials. Interestingly, application of sub-clinical chronic mild hypoxia (CMH) (8% O2) also promotes angiogenic remodeling and increased tight junction protein expression, suggestive of enhanced blood-brain barrier (BBB) integrity, though the role of APC in mediating the influence of CMH has not been investigated. To examine this potential link, we studied CMH-induced cerebrovascular remodeling after treating mice with two different reagents: (i) a function-blocking antibody that neutralizes APC activity, and (ii) exogenous recombinant murine APC. While CMH promoted endothelial proliferation, increased vascular density, and upregulated the angiogenic endothelial integrins α5ß1 and αvß3, these events were almost completely abolished by functional blockade of APC. Consistent with these findings, addition of exogenous recombinant APC enhanced CMH-induced endothelial proliferation, expansion of total vascular area and further enhanced the CMH-induced right-shift in vessel size distribution. Taken together, our findings support a key role for APC in mediating physiological remodeling of cerebral blood vessels in response to CMH.


Subject(s)
Brain Ischemia/pathology , Brain Ischemia/physiopathology , Brain/pathology , Gene Expression Regulation/physiology , Neovascularization, Physiologic/physiology , Protein C/metabolism , Animals , Antibodies/pharmacology , Disease Models, Animal , Endothelium, Vascular/metabolism , Integrin alpha5/metabolism , Ki-67 Antigen/metabolism , Male , Mice , Mice, Inbred C57BL , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Protein C/immunology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL