Skip to main content
Cell Transplantation logoLink to Cell Transplantation
. 2018 May 30;27(12):1798–1808. doi: 10.1177/0963689718768455

Roles of Pericytes in Stroke Pathogenesis

Jyoti Gautam 1, Yao Yao 1,
PMCID: PMC6300777  PMID: 29845887

Abstract

Stroke is a cerebrovascular disorder that affects many people worldwide. In addition to the well-established functions of astrocytes and microglia in stroke pathogenesis, pericytes also play an important role in stroke progression and recovery. As perivascular multi-potent cells and an important component of the blood–brain barrier (BBB), pericytes have been shown to exert a large variety of functions, including serving as stem/progenitor cells and maintaining BBB integrity. Here in this review, we summarize the roles of pericytes in stroke pathogenesis, with a focus on their effects in cerebral blood flow, BBB integrity, angiogenesis, immune responses, scar formation and fibrosis.

Keywords: stroke, hemorrhage, ischemia, pericytes, blood–brain barrier

Introduction

Stroke is the 5th leading cause of death and is a leading cause of long-term disability in the United States1. Based on the type of injury (occlusion or rupture of blood vessels), it is broadly categorized into ischemic stroke and hemorrhagic stroke. When stroke occurs, a cascade of molecular and cellular events take place, which eventually result in cerebral blood flow interruption, blood–brain barrier (BBB) breakdown, inflammation, glial cell activation, vascular malformation, and neuronal death24. Accumulating evidence suggests that astrocytes, microglia, and inflammatory leukocytes play critical roles in the pathogenesis of stroke57. Recent studies demonstrate that pericytes also affect stroke pathology and contribute to disease progression and recovery.

Pericytes are perivascular multi-potent cells located on the abluminal side of capillaries. In the central nervous system (CNS), pericytes have the highest density with an endothelial-to-pericyte ratio estimated to be 1:1∼3:18,9. This high density/coverage of pericytes has been shown to be crucial for the maintenance of BBB integrity1014. Another unique feature of CNS pericytes is that they are derived from neural crest cells, whereas pericytes from peripheral organs are mainly derived from the mesothelium10,1517. The different embryonic origins of pericytes suggest that CNS and peripheral pericytes may have distinct biological functions. There is evidence showing that CNS pericytes can exert a large variety of functions10, including regulation of cerebral blood flow, maintenance of BBB integrity, modulation of angiogenesis and inflammation, and serving as stem/progenitor cells. These functions are dependent on appropriate interactions and signaling between pericytes and other cells at the BBB, especially endothelial cells and astrocytes.

Although pericytes are embedded in the basement membrane, which separates them and other cells, pericytes and endothelial cells do form numerous direct contacts, including peg-socket contact, adhesion plaques, N-cadherin junctions, and gap junctions10. In addition, a large variety of signaling cascades, including platelet-derived growth factor (PDGF), vascular endothelial growth factor (VEGF), transforming growth factor-beta (TGF-β), and Wnt pathways, are involved in pericyte–endothelium interaction9,11,1824. Defective interactions/signaling between these cells lead to various pathological conditions in humans, including diabetic retinopathy, tumor angiogenesis, ectopic calcification, dementia syndrome, cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), and stroke10. It has been shown that PDGF secreted by endothelial cells attracts pericytes and mediates their attachment and coverage on newly formed blood vessels11,25. The recruitment of pericytes is closely associated with the barrier function of newly formed vessels. In vitro studies demonstrated that pericytes induced the synthesis of tight junction proteins through the release of angiopoetin-126, suggesting that pericyte–endothelium interaction promotes BBB integrity by reducing paracellular leakage. Contrary to this report, reduced pericyte recruitment/density failed to affect the expression of tight junction proteins in vivo12,27. Instead, it induces BBB leakage by up-regulating endothelial expression of genes known to increase vascular permeability, such as plasmalemmal vesicle-associated protein-1 (PLVAP)12,2729. PLVAP, which regulates vesicle trafficking, is highly expressed in permeable/peripheral vessels or in CNS vessels with pathological breakdown of BBB. These results suggest that pericyte–endothelium interaction also contributes to BBB integrity by suppressing the intracellular pathway (transcytosis)27.

Another cell type that actively communicates with pericytes is astrocytes. During vessel formation, astrocytes are also recruited to newly formed blood vessels, although it occurs later than pericyte recruitment30,31. It has been shown that tight junctions form before astrocytic endfeet wrap endothelial cells and pericytes30, suggesting that direct interaction with astrocytes is not required for tight junction formation. Astrocytes, however, can regulate tight junction integrity via Wnt and hedgehog signaling pathways32,33. In addition, astrocytes also contribute to BBB barrier property at the vessel–neuron interface through a group of unique channels and transporters that are exclusively expressed at their endfeet, including aquaporin 4 (Aqp4) and Kir4.134. Pericytes, which are sandwiched between astrocytes and endothelial cells, have been speculated to modulate astrocyte functions. Consistent with this hypothesis, expression of polarized astrocytic endfoot markers was significantly reduced in pericyte-deficient mice11. Together, these results suggest that, by interacting with both endothelial cells and astrocytes, pericytes actively regulate the formation and maintenance of BBB integrity.

Here in this review, we summarize recent findings on the biological roles of pericytes in the pathogenesis of both ischemic and hemorrhagic stroke. Specifically, the functions of pericytes in cerebral blood flow, BBB integrity, angiogenesis, immune responses, scar formation and fibrosis are discussed. The functions and related signaling pathways of pericytes in both types of stroke are summarized in Tables 1 and 2, respectively.

Table 1.

Functions of Pericytes in Stroke.

Functions Stroke types Roles References
Cerebral blood flow control Ischemic stroke Capillary constriction 22,35,36,37
BBB maintenance Intracerebral hemorrhage Regulation of vessel stability and vascular permeability 11,12,25
Regulation of contractile capability 38,39
Intracranial hemorrhage Regulation of endothelial cell permeability 4042
Ischemic stroke Pericyte survival Modulation of BBB integrity and VEGF/Nox4/ROS expression 43,44 4549
Angiogenesis Ischemic stroke Revascularization 23,46,50
Blood vessel stabilization 47,51,52
Disruption of vascular integrity 53,54
Regulation of basement membrane formation 55
Hemorrhagic stroke Vascular development 56
Vessel stabilization 57
Immunological properties Ischemic Stroke Differentiating into microglia-like cells and performing microglia-like functions 58,59,60,61
Reprogramming into NPCs 60,62
Scar formation and fibrosis Ischemic stroke Neuroprotection 21
Promoting fibrosis and regulating fibronectin deposition 63,64

BBB: blood–brain barrier; NPCs: neural precursor cells ; ROS: reactive oxygen species; VEGF: vascular endothelial growth factor.

Table 2.

Signaling Pathways related to Pericyte Functions in Stroke.

Signaling pathways Stroke types Functions References
PDGF-BB/PDGFRβ Ischemic stroke Neuroprotection, angiogenesis and vascular remodeling, pericyte recruitment, scar formation and fibrosis regulation 11,12,21,25,65,66
Hemorrhagic stroke BBB maintenance 11,12,20,25
Notch CADASIL Regulation of pericyte survival and angiogenesis 55,43,44
Neonatal intraventricular hemorrhage Pericyte recruitment, Pericyte–endothelium interaction, BBB maintenance 55,67
Canonical Wnt/β-catenin Ischemic stroke BBB maintenance 18
Hemorrhagic stroke Angiogenesis regulation 18
TGF-β/TGFβR2 Neonatal Intraventricular hemorrhage Maintenance of vascular stability/BBB integrity 67,68
Ischemic stroke Angiogenesis regulation and vessel stabilization 69,70
VEGF-A/VEGFR2 Ischemic stroke and hypoxia Angiogenesis induction and tight junction stabilization 7173
Ang/Tie2 Ischemic stroke ECM protein deposition and BBB protection 10,19
aPKC-CBP Ischemic stroke Vascular remodeling and motor recovery 62

aPKC: atypical protein kinase C; BBB: blood–brain barrier; CADASIL: cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy; CBP: Creb binding protein; ECM: extracellular matrix; PDGF: platelet-derived growth factor; PDGFRβ: beta-type platelet-derived growth factor receptor; TGF: transforming growth factor; VEGF: vascular endothelial growth factor; VEGFR: vascular endothelial growth factor receptor.

Pericytes and Cerebral Blood Flow in Stroke

Whether pericytes regulate cerebral blood flow remains controversial. On one hand, there is evidence supporting that pericytes contribute to blood flow. For example, an early electron microscopy study revealed filamentous structures in the cytoplasm of pericytes, similar to myofilaments in smooth muscle cells (SMCs)74. Later, actin- and myosin-like filaments were identified in rat brain pericytes75. In addition, various contractile proteins, such as α-smooth muscle actin (SMA), were found in brain pericytes in culture22,76,77, suggesting that pericytes are able to contract. Consistent with these studies, rat brain pericytes have been shown to contract in response to intracellular Ca2+ in vitro78. Recently, using an innovative iCelligence electrical impedance system, cultured brain pericytes have been shown to contract and relax in response to endothelin-1 and adenosine, respectively35. Using whole retina and brain slices, Dr. Attwell’s group reported that electrical stimulation and chemicals (adenosine triphosphate and noradrenaline) induced capillary constriction by pericytes, whereas glutamate reversed noradrenaline-induced constriction79. Furthermore, the same group elegantly showed that glutamate-induced pericyte relaxation was mediated by prostaglandin E2 and nitric oxide, and that capillary dilation occurred before arteriole dilation in response to whisker stimulation-induced increase of blood flow36. Together, these results strongly indicate that pericytes initiate blood flow control and actively regulate cerebral blood flow in physiological conditions.

In addition, pericyte constriction is also observed in pathological conditions, such as stroke. It has been shown that pericyte contraction entraps erythrocytes at the capillary constriction sites during stroke, which obstructs microcirculation37. Similarly, Hall and colleagues reported that pericytes constricted capillaries and died quickly in vivo after ischemia36. They further demonstrated that the death of pericytes in rigor produced a long-lasting constriction of the capillaries, leading to prolonged reduction of cerebral blood flow even when arterial flow was restored36. These pathological changes of pericytes after stroke are replicated in the iCelligence electrical impedance system. Specifically, chemical ischemia induced profound and irreversible pericyte contraction before their death in this in vitro system35. These results suggest that pericyte constriction and death contribute to the pathogenesis of stroke via regulating cerebral blood flow. Further mechanistic study revealed that pericyte death was mediated in part by glutamate and was not reduced by free radical scavenging36. In contrast to this report, suppressing oxidative-nitrate stress has been found to alleviate ischemia and reperfusion-induced pericyte contraction and positively affect tissue survival37. Additionally, administration of free radical scavenger edaravone also reduced infarct volume through preventing pericyte contraction and promoting pericyte proliferation in a rat model of focal cerebral ischemia80. The exact molecular mechanism underlying pericyte constriction and death needs further investigation.

On the other hand, there are also studies showing that pericytes cannot constrict and do not regulate cerebral blood flow. Using various transgenic mouse lines and two-photon microscopy, Hill and colleagues failed to detect SMA expression in pericytes in both mice and humans81. They also showed that optogenetic-, whisker stimulation-, and cortical spreading depolarization-induced changes in vessel diameter and blood flow occurred in SMC-covered micro-vessels but not pericyte-covered capillaries81. Furthermore, using a transient middle cerebral artery occlusion (MCAO) model, they found that SMC, rather than pericyte constriction, caused hypo-perfusion, leading to distal microvascular occlusion81. Together, these data suggest that pre-capillary SMCs rather than pericytes are responsible for blood flow regulation in physiological and pathological conditions. This discrepancy may be partially explained by the difficulty in distinguishing pericytes from pre-capillary SMCs due to their similar biochemical, structural, and functional properties.

Compared with ischemic stroke, blood flow is less studied in intracerebral hemorrhage. Although perihematomal tissue constantly undergoes edematous and metabolic changes8284, reduced blood flow with increased oxygen extraction fraction has been reported in perihematomal regions85,86. Whether pericytes play a role in the reduced perihematomal blood flow remains unknown. Addressing this question will enrich our understanding of pericyte biology/function in hemorrhagic stroke.

Pericytes and BBB During Stroke

The BBB is a dynamic structure located at the interface of the CNS and circulation system. By actively regulating material exchange between these two systems, it functions to maintain CNS homeostasis87,88. Not only is BBB breakdown a consequence of stroke, it also exacerbates stroke outcome89,90. Pericytes, as a major component of the BBB10,87, have been hypothesized to contribute to stroke pathogenesis via regulating BBB integrity.

Accumulating evidence shows that reduced pericyte coverage on blood vessels compromises vascular integrity and causes hemorrhagic stroke. During development and/or recovery after injury, pericytes are recruited to newly formed immature blood vessels mainly via the PDGFB–PDGFRβ axis25,9193. Pericyte recruitment and subsequent coverage on endothelial cells reduce vascular leakage and stabilize these vessels1012,25,94,95. Mutations in either PDGFB or PDGFRβ diminish pericyte recruitment11,12,92,93, resulting in severe BBB disruption and massive hemorrhage11,12,20,25. In addition, Notch3 mutation led to substantially reduced pericyte numbers, BBB disruption, and intraventricular hemorrhage in a zebrafish model96. Similarly, reduced pericyte coverage and arteriovenous malformations were observed in Notch1+/−Notch3−/− mice55. These findings strongly indicate that pericyte coverage on blood vessels is important for the maintenance of vascular integrity, and that loss of pericytes causes hemorrhagic stroke.

In addition, abnormal differentiation of pericytes/SMCs has also been linked to BBB disruption and intracerebral hemorrhage. Previous work from our laboratory showed that loss of astrocyte-derived laminin induced aberrant differentiation of brain pericytes and compromised the maturation of vascular SMCs, leading to BBB disruption and intracerebral hemorrhage38,39. Additionally, forkhead transcription factor 2 (FOXF2), a gene specifically expressed in pericytes and SMCs, has been found to associate with a higher risk of stroke40. Inactivation of FOXF2 at both embryonic and adult stages results in BBB breakdown and intracranial hemorrhage in mice due to defects in the differentiation of pericytes and/or SMCs41,42. These data suggest that abnormal pericyte/SMC differentiation plays a causative role in BBB breakdown and intracerebral hemorrhage.

Although there is no direct causal relationship between pericyte dysfunction and ischemic stroke, pericytes may contribute to ischemia pathogenesis indirectly via their effect on vascular integrity. For example, mutations in Notch3 gene, which is crucial for the survival of pericytes43, are linked to CADASIL44, a hereditary stroke disorder. There is also evidence showing that pericytes can modulate BBB integrity and thus ischemic injury through VEGF. Using sodium cyanide (NaCN) treatment as an in vitro ischemic model, it has been shown that NaCN substantially increases VEGF expression in brain pericytes, and that conditioned medium from NaCN-treated pericytes disrupts vascular integrity in an in vitro BBB model45. Consistent with this report, VEGF has been found to induce BBB leakage in ischemic brain46. It should be noted that, however, there is also evidence suggesting that prolonged exposure to VEGF enhances post-ischemic BBB integrity47. This discrepancy could be due to different dosage, treatment strategy, and timing. In addition, pericytes can also affect BBB integrity and thus ischemia progression via reactive oxygen species (ROS). A previous study showed that nicotinamide adenine dinucleotide phosphate (NADPH) oxidase NOX4, an enzymatic source of ROS production, was highly up-regulated in the peri-infarct region in MCAO model48. Recently, it has been reported that pericytes are a main cellular source of NOX4 and its expression in pericytes is greatly enhanced in peri-infarct areas after MCAO49. Further study revealed that overexpression of NOX4 in pericytes induced BBB breakdown by up-regulating metalloproteinase-949, highlighting an important role of ROS in BBB integrity.

Angiogenic Property of Pericytes in Stroke

Angiogenesis, the generation of new blood vessels from existing vasculature, is an important process that occurs in both normal and pathological conditions97. Using a variety of signaling pathways, including PDGFB-PDGFRβ, angiogenic endothelial cells recruit pericytes10,9193,98, which stabilize newly formed blood vessels10,25,94,95. Stroke is a brain vascular disease caused by occlusion and/or rupture of blood vessels. Revascularization in injured brain regions promotes stroke recovery23,46,50,99. Given the critical role of pericytes in angiogenesis24,100102, it has been speculated that pericytes may promote stroke recovery via modulating angiogenesis.

In ischemic stroke, various reports support this hypothesis. First, many important angiogenic factors, such as VEGF and TGF-β, have been found in pericytes24,100,101, and their expression is significantly altered in various types of ischemic stroke in rodents103,104. Next, it has been shown that transplantation of saphenous vein-derived pericyte progenitor cells to a mouse model of myocardial infarct exerts a beneficial role, which is mediated by their pro-angiogenic, pro-survival, and anti-fibrotic activities105. Similarly, bone marrow-derived pericytes also contribute to revascularization after ischemia. It has been reported that transplanted bone marrow-derived cells differentiate into microglia and pericytes in mouse brain after ischemia51. These bone marrow-derived pericytes express high levels of VEGF and TGF-β51, suggesting that they are involved in ischemia-induced angiogenesis and blood vessel stabilization. Furthermore, recombinant human VEGF has been demonstrated to increase capillary density and pericyte coverage, improve cerebral energy state and blood flow, and reduce brain infarction size in MCAO model47. Consistent with this finding, VEGF receptor-inhibition promotes cell death, reduces endothelial cell proliferation, and worsens injury in a neonatal stroke model52. Together, these observations suggest that pericytes play a beneficial role in ischemic stroke via promoting angiogenesis.

It should be noted, however, that there is also evidence showing that angiogenesis may play a detrimental role in ischemic stroke. For instance, elevated VEGF expression in ischemia has been associated with uncoupling of endothelial cell–cell junctions and increased vascular permeability & edema53. Consistent with this report, VEGF antagonism reduced edema formation and tissue damage following MCAO in mouse brain54. In addition, signs of enhanced angiogenesis, including exacerbated endothelial cell activation and retinal hyper-vascularization, were observed in Notch1+/−Notch3−/− mice55, a model of hereditary stroke disorder CADASIL. Furthermore, increased risk of ischemic stroke is associated with both type1 and type 2 diabetes106, a disease characterized by persistent and uncontrolled angiogenesis107. Together, these studies suggest that the angiogenic property of pericytes may exert a dual role in ischemic stroke, depending on animal models, injury types, and timing.

Unlike in ischemic stroke, the angiogenic effect of pericytes is less studied in hemorrhagic stroke. Increased proliferation of pericytes and endothelial cells has been reported in mice with mural cell-specific deletion of Foxc1, which develop late-gestation cerebral micro-hemorrhages56. Additionally, excessive angiogenesis and enhanced sensitivity to angiogenic stimuli were observed in hereditary hemorrhagic telangiectasia57, a genetic disease characterized by arteriovenous malformations and brain hemorrhage. Furthermore, type1 but not type 2 diabetes is associated with elevated risk of hemorrhagic stroke106. These findings demonstrate a positive correlation between angiogenesis and hemorrhagic stroke. The exact role of the angiogenic effect of pericytes in hemorrhagic stroke, however, remains largely unclear.

Immunological Property of Pericytes in Stroke

Pericytes participate in CNS defense by exhibiting both innate and adaptive immune responses. Numerous studies conducted in both rodents and humans have shown that pericytes are able to respond to pro-inflammatory signals and release anti-inflammatory cytokines/chemokines. For instance, mouse brain pericytes constitutively produce chemokines and cytokines, including granulocyte-colony stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin (IL)-1α, IL-6, monocyte chemoattractant protein-1 (MCP-1) and nitric oxide, under normal conditions108,109. In lipopolysaccharide-induced inflammation, not only do the expression levels of these chemokines and cytokines change, many new factors (e.g. IL-5 and RANTES) are also induced108,109. A recent study identified two different pericyte populations in the brain based on CD90 expression110. An in vitro study showed that, when compared with CD90 pericytes, CD90+ pericytes expressed lower levels of pericyte markers (e.g. SMA and PDGFRβ) and extracellular matrix proteins, had higher basal proliferation, and exhibited a reduced pro-inflammatory response110. Consistent with previous reports10,16,111119, these results suggest that pericytes are a heterogeneous population, and that different subpopulations may have distinct biological functions.

There is also evidence showing that pericytes, like classical antigen-presenting cells (e.g. dendritic cells and macrophages), can express cell surface proteins, essential for the acquired immune system to recognize foreign molecules. For example, primary rat CNS pericytes constitutively express low levels of intercellular adhesion molecule-1 and major histocompatibility complex (MHC) class I molecules120. Upon induction by interferon (IFN)-γ, they also express MHC class II molecules, which enable them to present antigens to primed syngeneic T-lymphocytes121. Like CNS pericytes, placenta- and human pluripotent stem cell-derived pericytes constitutively express MHC class I and co-stimulatory molecules under basal culture conditions122. Upon IFN-γ treatment, they start to express MHC class II molecules122. In addition, like microglia/macrophages, pericytes also exhibit phagocytic activity. It has been reported that human brain pericytes phagocytose erythrocytes in brain trauma123. Consistent with this finding, cultured rat pericytes expressed macrophage markers ED-2 and CD11b, and phagocytosed fluorochrome-conjugated polystyrene beads and antibody-coated zymosan120, suggesting both Fc receptor-independent and -dependent phagocytic activity. Together, these studies support that pericytes have immune cell-like properties and are able to regulate immune responses.

Stroke is a neurological condition that involves a local inflammatory reaction and a plethora of immune responses in the brain. It has been speculated that pericytes might transform into microglia/macrophage-like cells in response to CNS injuries, including stroke58. Recently, a few studies provide evidence that brain pericytes acquire a microglia-like phenotype after ischemic stroke. First, it has been shown that RGS5 (regulator of G-protein signaling 5)-expressing cells, which are predominantly pericytes124126, leave blood vessel wall, proliferate and give rise to CD11b+ and galectin-3+ microglia-like cells after ischemic injury in rodents59. Second, under oxygen glucose deprivation, which mimics ischemic injury in vitro, human brain pericytes acquire stemness and differentiate into cells of various lineages, including microglia60. Similarly, stemness is also detected in pericytes isolated from ischemic regions of the mouse brain60,62, suggesting that ischemia induces multi-potency in pericytes. Additionally, Iba1+ microglia also express PDGFRβ in ischemic brains61, suggesting that some microglia may be derived from multi-potent pericytes after ischemia. Consistent with these findings, PDGFRβ+ pericytes isolated from ischemic but not non-ischemic brains differentiated into microglia-like cells and obtained phagocytic activity61. Together, these studies strongly suggest that pericytes can differentiate into microglia-like cells and exert microglia-like functions under ischemic conditions.

Whether pericytes are able to differentiate into microglia-like cells and exert microglia-like functions after hemorrhagic stroke remains unclear. Future studies should focus on investigating pericyte’s stemness in hemorrhagic stroke and other pathological conditions.

Pericytes and Scar Formation and Fibrosis in Stroke

Upon CNS injury, glial cells become activated and contribute to the formation of a glial scar around the injury site by depositing chondroitin sulfate proteoglycans, including neurocan and phosphacan127131. Scar tissue functions to prevent the spreading of toxic substances in the CNS132,133. However, excessive or long-lasting scar formation inhibits axon regeneration and stalls the recovery process, leading to fibrosis133135.

Recent studies show that pericytes also contribute to scar formation and organ fibrosis. In a spinal cord injury model, pericytes are categorized into type A and type B based on glutamate aspartate transporter (GLAST) expression, and GLAST+ type A pericytes are identified as the major cell type that forms a glial scar136. In addition, pericytes are also classified into type I (Nestin-GFP-/NG2-DsRed+) and type II (Nestin-GFP+/NG2-DsRed+), which generate adipocytes/fibroblasts and neural/myogenic cells, respectively137,138. Type I pericytes have been shown to accumulate and participate in scar formation in multiple organs after injury, including the kidney, myocardium, spinal cord, and brain139. In ischemic injury, PDGFRβ+ pericytes have been shown to trigger fibrotic responses in both kidney and CNS63,140. Compared with the controls, PDGFRβ+/− mice demonstrated decreased fibrosis with reduced fibronectin deposition in the ischemic area and enlarged infarct volume, suggesting that PDGFRβ signaling-induced fibronectin production is indispensable for the repair processes after ischemic stroke63. This finding is consistent with the neuroprotective role of fibronectin in CNS injury141,64. Currently, it is unclear whether and how pericytes affect scar formation and fibrosis after hemorrhagic stroke. Answers to these questions may lead to innovative pericyte-based therapies for stroke.

Therapeutic Implications and Future Directions

With the increase of knowledge in pericyte biology/function, therapeutic strategies utilizing/targeting pericytes have been developed and tested. For example, in an acute myocardial infarction model, transplantation of human microvascular pericytes (CD146+CD34CD45CD56 cells) has been shown to attenuate ventricular dilatation, improve cardiac contractility, reduce myocardial fibrosis, and significantly diminish infiltration of host inflammatory cells at the injury site142. More importantly, these pericytes demonstrated a much better therapeutic effect when compared with CD56+ myogenic progenitor cells142. Based on the finding that human brain pericytes can be reprogrammed to neuronal cells by co-expression of SOX2 and MASH1143, it is logical to hypothesize that neuronal reprogramming of pericytes may replace degenerated/damaged neurons and have a therapeutic potential143. In addition, drugs targeting pericytes or modulating their activities also show promising results in treating stroke. In an experimental intracerebral hemorrhage model, administration of recombinant ADAMTS-13 (a disintegrin and metalloprotease with thrombospondin type I motif, member 13) led to enhanced pericyte coverage, attenuated BBB leakage, reduced inflammation markers, ameliorated cerebral edema, diminished hematoma volume, and improved neurological functions144. In an ischemic model, Cilostazol, an antiplatelet drug, showed a neuroprotective effect by preventing pathological detachment of astrocytic endfeet145. The same drug also exerted a beneficial role in a collagenase-induced hemorrhage model by enhancing pericyte coverage in the brain146. Additionally, the free radical scavenger edaravone has been shown to ameliorate brain damage after MCAO via increasing pericyte proliferation and their coverage around endothelial cells80.

Although significant progresses have been made on how pericytes regulate stroke pathogenesis, a few critical questions need further investigation. First, no pericyte-specific markers are available currently, although various markers have been used to identify pericytes10. It should be noted that the ‘pericyte’ populations described in most studies also contain other cells. For example, PDGFRβ+ cells include both pericytes and SMCs10,25,147, whereas RGS5 labels pericytes, SMCs and possibly cardiomyocytes10,125,126. Next, pericytes are a heterogeneous population10. It is speculated that different subtypes of pericytes may exert distinct roles in stroke. The marker expression and function of these subpopulations remain largely unknown at present. Future work should focus on identifying pericyte-specific & subtype-specific markers and characterizing subpopulations of pericytes. Third, compared with ischemic stroke, hemorrhagic stroke is relatively less studied. The biological function and therapeutic potential of pericytes in hemorrhagic stroke remain largely elusive. Understanding this information will enable us to study pericytes in a cell type- and subtype-specific manner, which will promote the development of innovative and effective treatments for stroke.

Footnotes

Declaration of Conflicting Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding: The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This work was supported by the American Heart Association Scientist Development Grant (16SDG29320001 to Y.Y.).

References

  • 1. Writing Group M, Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, Das SR, de Ferranti S, Despres JP, et al. Heart disease and stroke statistics-2016 update: a report From the American Heart Association. Circulation. 2016;133(4):e38–e360. [DOI] [PubMed] [Google Scholar]
  • 2. Terasaki Y, Liu Y, Hayakawa K, Pham LD, Lo EH, Ji X, Arai K. Mechanisms of neurovascular dysfunction in acute ischemic brain. Curr Med Chem. 2014;21(18):2035–2042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Moskowitz MA, Lo EH, Iadecola C. The science of stroke: mechanisms in search of treatments. Neuron. 2010;67(2):181–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Lo EH, Dalkara T, Moskowitz MA. Mechanisms, challenges and opportunities in stroke. Nat Rev Neurosci. 2003;4(5):399–415. [DOI] [PubMed] [Google Scholar]
  • 5. Kim JY, Park J, Chang JY, Kim SH, Lee JE. Inflammation after ischemic stroke: the role of leukocytes and glial cells. Exp Neurobiol. 2016;25(5):241–251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Patel AR, Ritzel R, McCullough LD, Liu F. Microglia and ischemic stroke: a double-edged sword. Int J Physiol Pathophysiol Pharmacol. 2013;5(2):73–90. [PMC free article] [PubMed] [Google Scholar]
  • 7. Becerra-Calixto A, Cardona-Gomez GP. The role of astrocytes in neuroprotection after brain stroke: potential in cell therapy. Front Mol Neurosci. 2017;10:88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Shepro D, Morel NM. Pericyte physiology. FASEB J. 1993;7(11):1031–1038. [DOI] [PubMed] [Google Scholar]
  • 9. Daneman R, Prat A. The blood–brain barrier. Cold Spring Harb Perspect Biol. 2015;7(1):a020412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Armulik A, Genove G, Betsholtz C. Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. Dev Cell. 2011;21(2):193–215. [DOI] [PubMed] [Google Scholar]
  • 11. Armulik A, Genove G, Mae M, Nisancioglu MH, Wallgard E, Niaudet C, He L, Norlin J, Lindblom P, Strittmatter K. Pericytes regulate the blood–brain barrier. Nature. 2010;468(7323):557–561. [DOI] [PubMed] [Google Scholar]
  • 12. Daneman R, Zhou L, Kebede AA, Barres BA. Pericytes are required for blood–brain barrier integrity during embryogenesis. Nature. 2010;468(7323):562–566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Bell RD, Winkler EA, Sagare AP, Singh I, LaRue B, Deane R, Zlokovic BV. Pericytes control key neurovascular functions and neuronal phenotype in the adult brain and during brain aging. Neuron. 2010;68(3):409–427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Sweeney MD, Ayyadurai S, Zlokovic BV. Pericytes of the neurovascular unit: key functions and signaling pathways. Nat Neurosci. 2016;19(6):771–783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Majesky MW. Developmental basis of vascular smooth muscle diversity. Arterioscler Thromb Vasc Biol. 2007;27(6):1248–1258. [DOI] [PubMed] [Google Scholar]
  • 16. Etchevers HC, Vincent C, Le Douarin NM, Couly GF. The cephalic neural crest provides pericytes and smooth muscle cells to all blood vessels of the face and forebrain. Development. 2001;128(7):1059–1068. [DOI] [PubMed] [Google Scholar]
  • 17. Winkler EA, Bell RD, Zlokovic BV. Central nervous system pericytes in health and disease. Nat Neurosci. 2011;14(11):1398–1405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Chang J, Mancuso MR, Maier C, Liang X, Yuki K, Yang L, Kwong JW, Wang J, Rao V, Vallon M, et al. Gpr124 is essential for blood–brain barrier integrity in central nervous system disease. Nat Med. 2017;23(4):450–460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Cui X, Chopp M, Zacharek A, Ye X, Roberts C, Chen J. Angiopoietin/Tie2 pathway mediates type 2 diabetes induced vascular damage after cerebral stroke. Neurobiol Dis. 2011;43(1):285–292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Hellstrom M, Gerhardt H, Kalen M, Li X, Eriksson U, Wolburg H, Betsholtz C. Lack of pericytes leads to endothelial hyperplasia and abnormal vascular morphogenesis. J Cell Biol. 2001;153(3):543–553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Shen J, Ishii Y, Xu G, Dang TC, Hamashima T, Matsushima T, Yamamoto S, Hattori Y, Takatsuru Y, Nabekura J, et al. PDGFR-beta as a positive regulator of tissue repair in a mouse model of focal cerebral ischemia. J Cereb Blood Flow Metab. 2012;32(2):353–367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Sieczkiewicz GJ, Herman IM. TGF-beta 1 signaling controls retinal pericyte contractile protein expression. Microvasc Res. 2003;66(3):190–196. [DOI] [PubMed] [Google Scholar]
  • 23. Sun Y, Jin K, Xie L, Childs J, Mao XO, Logvinova A, Greenberg DA. VEGF-induced neuroprotection, neurogenesis, and angiogenesis after focal cerebral ischemia. J Clin Invest. 2003;111(12):1843–1851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Teichert M, Milde L, Holm A, Stanicek L, Gengenbacher N, Savant S, Ruckdeschel T, Hasanov Z, Srivastava K, Hu J, et al. Pericyte-expressed Tie2 controls angiogenesis and vessel maturation. Nat Commun. 2017;8:16106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Lindahl P, Johansson BR, Leveen P, Betsholtz C. Pericyte loss and microaneurysm formation in PDGF-B-deficient mice. Science. 1997;277(5323):242–245. [DOI] [PubMed] [Google Scholar]
  • 26. Hori S, Ohtsuki S, Hosoya K, Nakashima E, Terasaki T. A pericyte-derived angiopoietin-1 multimeric complex induces occludin gene expression in brain capillary endothelial cells through Tie-2 activation in vitro. J Neurochem. 2004;89(2):503–513. [DOI] [PubMed] [Google Scholar]
  • 27. Shue EH, Carson-Walter EB, Liu Y, Winans BN, Ali ZS, Chen J, Walter KA. Plasmalemmal vesicle associated protein-1 (PV-1) is a marker of blood–brain barrier disruption in rodent models. BMC Neurosci. 2008;9:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Lee SW, Kim WJ, Jun HO, Choi YK, Kim KW. Angiopoietin-1 reduces vascular endothelial growth factor-induced brain endothelial permeability via upregulation of ZO-2. Int J Mol Med. 2009;23(2):279–284. [PubMed] [Google Scholar]
  • 29. Nag S, Papneja T, Venugopalan R, Stewart DJ. Increased angiopoietin2 expression is associated with endothelial apoptosis and blood–brain barrier breakdown. Lab Invest. 2005;85(10):1189–1198. [DOI] [PubMed] [Google Scholar]
  • 30. Bonkowski D, Katyshev V, Balabanov RD, Borisov A, Dore-Duffy P. The CNS microvascular pericyte: pericyte-astrocyte crosstalk in the regulation of tissue survival. Fluids Barriers CNS. 2011;8(1):8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Haseloff RF, Blasig IE, Bauer HC, Bauer H. In search of the astrocytic factor(s) modulating blood–brain barrier functions in brain capillary endothelial cells in vitro. Cell Mol Neurobiol. 2005;25(1):25–39. [DOI] [PubMed] [Google Scholar]
  • 32. Liebner S, Corada M, Bangsow T, Babbage J, Taddei A, Czupalla CJ, Reis M, Felici A, Wolburg H, Fruttiger M, et al. Wnt/beta-catenin signaling controls development of the blood–brain barrier. J Cell Biol. 2008;183(3):409–417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Chien AJ, Conrad WH, Moon RT. A Wnt survival guide: from flies to human disease. J Invest Dermatol. 2009;129(7):1614–1627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Bernacki J, Dobrowolska A, Nierwinska K, Malecki A. Physiology and pharmacological role of the blood–brain barrier. Pharmacol Rep. 2008;60(5):600–622. [PubMed] [Google Scholar]
  • 35. Neuhaus AA, Couch Y, Sutherland BA, Buchan AM. Novel method to study pericyte contractility and responses to ischaemia in vitro using electrical impedance. J Cereb Blood Flow Metab. 2017;37(6):2013–2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Hall CN, Reynell C, Gesslein B, Hamilton NB, Mishra A, Sutherland BA, O’Farrell FM, Buchan AM, Lauritzen M, Attwell D. Capillary pericytes regulate cerebral blood flow in health and disease. Nature. 2014;508(7494):55–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Yemisci M, Gursoy-Ozdemir Y, Vural A, Can A, Topalkara K, Dalkara T. Pericyte contraction induced by oxidative-nitrative stress impairs capillary reflow despite successful opening of an occluded cerebral artery. Nat Med. 2009;15(9):1031–1037. [DOI] [PubMed] [Google Scholar]
  • 38. Yao Y, Chen ZL, Norris EH, Strickland S. Astrocytic laminin regulates pericyte differentiation and maintains blood–brain barrier integrity. Nat Commun. 2014;5:3413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Chen ZL, Yao Y, Norris EH, Kruyer A, Jno-Charles O, Akhmerov A, Strickland S. Ablation of astrocytic laminin impairs vascular smooth muscle cell function and leads to hemorrhagic stroke. J Cell Biol. 2013;202(2):381–395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. French CR, Seshadri S, Destefano AL, Fornage M, Arnold CR, Gage PJ, Skarie JM, Dobyns WB, Millen KJ, Liu T, et al. Mutation of FOXC1 and PITX2 induces cerebral small-vessel disease. J Clin Invest. 2014;124(11):4877–4881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Neurology Working Group of the Cohorts for H, Aging Research in Genomic Epidemiology Consortium tSGN, the International Stroke Genetics C. Identification of additional risk loci for stroke and small vessel disease: a meta-analysis of genome-wide association studies. Lancet Neurol. 2016;15(7):695–707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Reyahi A, Nik AM, Ghiami M, Gritli-Linde A, Ponten F, Johansson BR, Carlsson P. FOXF2 is required for brain pericyte differentiation and development and maintenance of the blood–brain barrier. Dev Cell. 2015;34(1):19–32. [DOI] [PubMed] [Google Scholar]
  • 43. Arboleda-Velasquez JF, Primo V, Graham M, James A, Manent J, D’Amore PA. Notch signaling functions in retinal pericyte survival. Invest Ophthalmol Vis Sci. 2014;55(8):5191–5199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Joutel A, Corpechot C, Ducros A, Vahedi K, Chabriat H, Mouton P, Alamowitch S, Domenga V, Cecillion M, Marechal E, et al. Notch3 mutations in CADASIL, a hereditary adult-onset condition causing stroke and dementia. Nature. 1996;383(6602):707–710. [DOI] [PubMed] [Google Scholar]
  • 45. Bai Y, Zhu X, Chao J, Zhang Y, Qian C, Li P, Liu D, Han B, Zhao L, Zhang J, et al. Pericytes contribute to the disruption of the cerebral endothelial barrier via increasing VEGF expression: implications for stroke. PLoS One. 2015;10(4):e0124362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Zhang ZG, Zhang L, Jiang Q, Zhang R, Davies K, Powers C, Bruggen N, Chopp M. VEGF enhances angiogenesis and promotes blood–brain barrier leakage in the ischemic brain. J Clin Invest. 2000;106(7):829–838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Zechariah A, ElAli A, Doeppner TR, Jin F, Hasan MR, Helfrich I, Mies G, Hermann DM. Vascular endothelial growth factor promotes pericyte coverage of brain capillaries, improves cerebral blood flow during subsequent focal cerebral ischemia, and preserves the metabolic penumbra. Stroke. 2013;44(6):1690–1697. [DOI] [PubMed] [Google Scholar]
  • 48. Vallet P, Charnay Y, Steger K, Ogier-Denis E, Kovari E, Herrmann F, Michel JP, Szanto I. Neuronal expression of the NADPH oxidase NOX4, and its regulation in mouse experimental brain ischemia. Neuroscience. 2005;132(2):233–238. [DOI] [PubMed] [Google Scholar]
  • 49. Nishimura A, Ago T, Kuroda J, Arimura K, Tachibana M, Nakamura K, Wakisaka Y, Sadoshima J, Iihara K, Kitazono T. Detrimental role of pericyte NOX4 in the acute phase of brain ischemia. J Cereb Blood Flow Metab. 2016;36(6):1143–1154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Zaidat OO, Suarez JI, Sunshine JL, Tarr RW, Alexander MJ, Smith TP, Enterline DS, Selman WR, Landis DM. Thrombolytic therapy of acute ischemic stroke: correlation of angiographic recanalization with clinical outcome. AJNR Am J Neuroradiol. 2005;26(4):880–884. [PMC free article] [PubMed] [Google Scholar]
  • 51. Kokovay E, Li L, Cunningham LA. Angiogenic recruitment of pericytes from bone marrow after stroke. J Cereb Blood Flow Metab. 2006;26(4):545–555. [DOI] [PubMed] [Google Scholar]
  • 52. Shimotake J, Derugin N, Wendland M, Vexler ZS, Ferriero DM. Vascular endothelial growth factor receptor-2 inhibition promotes cell death and limits endothelial cell proliferation in a neonatal rodent model of stroke. Stroke. 2010;41(2):343–349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Weis SM, Cheresh DA. Pathophysiological consequences of VEGF-induced vascular permeability. Nature. 2005;437(7058):497–504. [DOI] [PubMed] [Google Scholar]
  • 54. Van Bruggen N, Thibodeaux H, Palmer JT, Lee WP, Fu L, Cairns B, Tumas D, Gerlai R, Williams SP, van Lookeren Campagne M, et al. VEGF antagonism reduces edema formation and tissue damage after ischemia/reperfusion injury in the mouse brain. J Clin Invest. 1999;104(11):1613–1620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Kofler NM, Cuervo H, Uh MK, Murtomaki A, Kitajewski J. Combined deficiency of Notch1 and Notch3 causes pericyte dysfunction, models CADASIL, and results in arteriovenous malformations. Sci Rep. 2015;5:16449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Siegenthaler JA, Choe Y, Patterson KP, Hsieh I, Li D, Jaminet SC, Daneman R, Kume T, Huang EJ, Pleasure SJ. Foxc1 is required by pericytes during fetal brain angiogenesis. Biol Open. 2013;2(7):647–659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Thalgott J, Dos-Santos-Luis D, Lebrin F. Pericytes as targets in hereditary hemorrhagic telangiectasia. Front Genet. 2015;6:37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Boya J. An ultrastructural study of the relationship between pericytes and cerebral macrophages. Acta Anat (Basel). 1976;95(4):598–608. [DOI] [PubMed] [Google Scholar]
  • 59. Ozen I, Deierborg T, Miharada K, Padel T, Englund E, Genove G, Paul G. Brain pericytes acquire a microglial phenotype after stroke. Acta Neuropathol. 2014;128(3):381–396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Nakagomi T, Kubo S, Nakano-Doi A, Sakuma R, Lu S, Narita A, Kawahara M, Taguchi A, Matsuyama T. Brain vascular pericytes following ischemia have multipotential stem cell activity to differentiate into neural and vascular lineage cells. Stem Cells. 2015;33(6):1962–1974. [DOI] [PubMed] [Google Scholar]
  • 61. Sakuma R, Kawahara M, Nakano-Doi A, Takahashi A, Tanaka Y, Narita A, Kuwahara-Otani S, Hayakawa T, Yagi H, Matsuyama T, et al. Brain pericytes serve as microglia-generating multipotent vascular stem cells following ischemic stroke. J Neuroinflammation. 2016;13(1):57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Gouveia A, Seegobin M, Kannangara TS, He L, Wondisford F, Comin CH, Costa LDF, Beique JC, Lagace DC, Lacoste B, et al. The aPKC-CBP pathway regulates post-stroke neurovascular remodeling and functional recovery. Stem Cell Reports. 2017;9(6):1735–1744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Makihara N, Arimura K, Ago T, Tachibana M, Nishimura A, Nakamura K, Matsuo R, Wakisaka Y, Kuroda J, Sugimori H, et al. Involvement of platelet-derived growth factor receptor beta in fibrosis through extracellular matrix protein production after ischemic stroke. Exp Neurol. 2015;264:127–134. [DOI] [PubMed] [Google Scholar]
  • 64. Yanqing Z, Yu-Min L, Jian Q, Bao-Guo X, Chuan-Zhen L. Fibronectin and neuroprotective effect of granulocyte colony-stimulating factor in focal cerebral ischemia. Brain Res. 2006;1098(1):161–169. [DOI] [PubMed] [Google Scholar]
  • 65. Arimura K, Ago T, Kamouchi M, Nakamura K, Ishitsuka K, Kuroda J, Sugimori H, Ooboshi H, Sasaki T, Kitazono T. PDGF receptor beta signaling in pericytes following ischemic brain injury. Curr Neurovasc Res. 2012;9(1):1–9. [DOI] [PubMed] [Google Scholar]
  • 66. Renner O, Tsimpas A, Kostin S, Valable S, Petit E, Schaper W, Marti HH. Time- and cell type-specific induction of platelet-derived growth factor receptor-beta during cerebral ischemia. Brain Res Mol Brain Res. 2003;113(1–2):44–51. [DOI] [PubMed] [Google Scholar]
  • 67. Li F, Lan Y, Wang Y, Wang J, Yang G, Meng F, Han H, Meng A, Wang Y, Yang X. Endothelial Smad4 maintains cerebrovascular integrity by activating N-cadherin through cooperation with Notch. Dev Cell. 2011;20(3):291–302. [DOI] [PubMed] [Google Scholar]
  • 68. Vinukonda G, Dummula K, Malik S, Hu F, Thompson CI, Csiszar A, Ungvari Z, Ballabh P. Effect of prenatal glucocorticoids on cerebral vasculature of the developing brain. Stroke. 2010;41(8):1766–1773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Slevin M, Krupinski J, Slowik A, Kumar P, Szczudlik A, Gaffney J. Serial measurement of vascular endothelial growth factor and transforming growth factor-beta1 in serum of patients with acute ischemic stroke. Stroke. 2000;31(8):1863–1870. [DOI] [PubMed] [Google Scholar]
  • 70. Bottner M, Krieglstein K, Unsicker K. The transforming growth factor-betas: structure, signaling, and roles in nervous system development and functions. J Neurochem. 2000;75(6):2227–2240. [DOI] [PubMed] [Google Scholar]
  • 71. Gonul E, Duz B, Kahraman S, Kayali H, Kubar A, Timurkaynak E. Early pericyte response to brain hypoxia in cats: an ultrastructural study. Microvasc Res. 2002;64(1):116–119. [DOI] [PubMed] [Google Scholar]
  • 72. Dore-Duffy P, Wang X, Mehedi A, Kreipke CW, Rafols JA. Differential expression of capillary VEGF isoforms following traumatic brain injury. Neurol Res. 2007;29(4):395–403. [DOI] [PubMed] [Google Scholar]
  • 73. Al Ahmad A, Gassmann M, Ogunshola OO. Maintaining blood–brain barrier integrity: pericytes perform better than astrocytes during prolonged oxygen deprivation. J Cell Physiol. 2009;218(3):612–622. [DOI] [PubMed] [Google Scholar]
  • 74. Stensaas LJ. Pericytes and perivascular microglial cells in the basal forebrain of the neonatal rabbit. Cell Tissue Res. 1975;158(4):517–541. [DOI] [PubMed] [Google Scholar]
  • 75. Le Beux YJ, Willemot J. Actin- and myosin-like filaments in rat brain pericytes. Anat Rec. 1978;190(4):811–826. [DOI] [PubMed] [Google Scholar]
  • 76. Dore-Duffy P, LaManna JC. Physiologic angiodynamics in the brain. Antioxid Redox Signal. 2007;9(9):1363–1371. [DOI] [PubMed] [Google Scholar]
  • 77. Verbeek MM, Otte-Holler I, Wesseling P, Ruiter DJ, De Waal RM. Induction of alpha-smooth muscle actin expression in cultured human brain pericytes by transforming growth factor-beta 1. Am J Pathol. 1994;144(2):372–382. [PMC free article] [PubMed] [Google Scholar]
  • 78. Kamouchi M, Kitazono T, Ago T, Wakisaka M, Ooboshi H, Ibayashi S, Iida M. Calcium influx pathways in rat CNS pericytes. Brain Res Mol Brain Res. 2004;126(2):114–120. [DOI] [PubMed] [Google Scholar]
  • 79. Peppiatt CM, Howarth C, Mobbs P, Attwell D. Bidirectional control of CNS capillary diameter by pericytes. Nature. 2006;443(7112):700–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Deguchi K, Liu N, Liu W, Omote Y, Kono S, Yunoki T, Deguchi S, Yamashita T, Ikeda Y, Abe K. Pericyte protection by edaravone after tissue plasminogen activator treatment in rat cerebral ischemia. J Neurosci Res. 2014;92(11):1509–1519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. 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(1):95–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Keep RF, Hua Y, Xi G. Intracerebral haemorrhage: mechanisms of injury and therapeutic targets. Lancet Neurol. 2012;11(8):720–731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Xi G, Keep RF, Hoff JT. Mechanisms of brain injury after intracerebral haemorrhage. Lancet Neurol. 2006;5(1):53–63. [DOI] [PubMed] [Google Scholar]
  • 84. Egashira Y, Hua Y, Keep RF, Xi G. Intercellular cross-talk in intracerebral hemorrhage. Brain Res. 2015;1623:97–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Zazulia AR, Diringer MN, Videen TO, Adams RE, Yundt K, Aiyagari V, Grubb RL, Jr, Powers WJ. Hypoperfusion without ischemia surrounding acute intracerebral hemorrhage. J Cereb Blood Flow Metab. 2001;21(7):804–810. [DOI] [PubMed] [Google Scholar]
  • 86. Mayer SA, Lignelli A, Fink ME, Kessler DB, Thomas CE, Swarup R, Van Heertum RL. Perilesional blood flow and edema formation in acute intracerebral hemorrhage: a SPECT study. Stroke. 1998;29(9):1791–1798. [DOI] [PubMed] [Google Scholar]
  • 87. Zlokovic BV. The blood–brain barrier in health and chronic neurodegenerative disorders. Neuron. 2008;57(2):178–201. [DOI] [PubMed] [Google Scholar]
  • 88. Persidsky Y, Ramirez SH, Haorah J, Kanmogne GD. Blood–brain barrier: structural components and function under physiologic and pathologic conditions. J Neuroimmune Pharmacol. 2006;1(3):223–236. [DOI] [PubMed] [Google Scholar]
  • 89. Haley MJ, Lawrence CB. The blood–brain barrier after stroke: Structural studies and the role of transcytotic vesicles. J Cereb Blood Flow Metab. 2017;37(2):456–470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Brouns R, Wauters A, De Surgeloose D, Marien P, De Deyn PP. Biochemical markers for blood–brain barrier dysfunction in acute ischemic stroke correlate with evolution and outcome. Eur Neurol. 2011;65(1):23–31. [DOI] [PubMed] [Google Scholar]
  • 91. Hellstrom M, Kalen M, Lindahl P, Abramsson A, Betsholtz C. Role of PDGF-B and PDGFR-beta in recruitment of vascular smooth muscle cells and pericytes during embryonic blood vessel formation in the mouse. Development. 1999;126(14):3047–3055. [DOI] [PubMed] [Google Scholar]
  • 92. Bjarnegard M, Enge M, Norlin J, Gustafsdottir S, Fredriksson S, Abramsson A, Takemoto M, Gustafsson E, Fassler R, Betsholtz C. Endothelium-specific ablation of PDGFB leads to pericyte loss and glomerular, cardiac and placental abnormalities. Development. 2004;131(8):1847–1857. [DOI] [PubMed] [Google Scholar]
  • 93. Enge M, Bjarnegard M, Gerhardt H, Gustafsson E, Kalen M, Asker N, Hammes HP, Shani M, Fassler R, Betsholtz C. Endothelium-specific platelet-derived growth factor-B ablation mimics diabetic retinopathy. EMBO J. 2002;21(16):4307–4316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Leveen P, Pekny M, Gebre-Medhin S, Swolin B, Larsson E, Betsholtz C. Mice deficient for PDGF B show renal, cardiovascular, and hematological abnormalities. Genes Dev. 1994;8(16):1875–1887. [DOI] [PubMed] [Google Scholar]
  • 95. Soriano P. Abnormal kidney development and hematological disorders in PDGF beta-receptor mutant mice. Genes Dev. 1994;8(16):1888–1896. [DOI] [PubMed] [Google Scholar]
  • 96. Wang Y, Pan L, Moens CB, Appel B. Notch3 establishes brain vascular integrity by regulating pericyte number. Development. 2014;141(2):307–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Ribatti D, Vacca A, Roncali L, Dammacco F. Angiogenesis under normal and pathological conditions. Haematologica. 1991;76(4):311–320. [PubMed] [Google Scholar]
  • 98. Gaengel K, Genove G, Armulik A, Betsholtz C. Endothelial-mural cell signaling in vascular development and angiogenesis. Arterioscler Thromb Vasc Biol. 2009;29(5):630–638. [DOI] [PubMed] [Google Scholar]
  • 99. Ferrara N, Gerber HP. The role of vascular endothelial growth factor in angiogenesis. Acta Haematol. 2001;106(4):148–156. [DOI] [PubMed] [Google Scholar]
  • 100. Ribatti D, Nico B, Crivellato E. The role of pericytes in angiogenesis. Int J Dev Biol. 2011;55(3):261–268. [DOI] [PubMed] [Google Scholar]
  • 101. Raza A, Franklin MJ, Dudek AZ. Pericytes and vessel maturation during tumor angiogenesis and metastasis. Am J Hematol. 2010;85(8):593–598. [DOI] [PubMed] [Google Scholar]
  • 102. Liu AY, Ouyang G. Tumor angiogenesis: a new source of pericytes. Curr Biol. 2013;23(13): R565–R568. [DOI] [PubMed] [Google Scholar]
  • 103. Nakamura K, Arimura K, Nishimura A, Tachibana M, Yoshikawa Y, Makihara N, Wakisaka Y, Kuroda J, Kamouchi M, Ooboshi H, et al. Possible involvement of basic FGF in the upregulation of PDGFRbeta in pericytes after ischemic stroke. Brain Res. 2016;1630:98–108. [DOI] [PubMed] [Google Scholar]
  • 104. Navaratna D, Guo S, Arai K, Lo EH. Mechanisms and targets for angiogenic therapy after stroke. Cell Adh Migr. 2009;3(2):216–223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Katare R, Riu F, Mitchell K, Gubernator M, Campagnolo P, Cui Y, Fortunato O, Avolio E, Cesselli D, Beltrami AP, et al. Transplantation of human pericyte progenitor cells improves the repair of infarcted heart through activation of an angiogenic program involving micro-RNA-132. Circ Res. 2011;109(8):894–906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Janghorbani M, Hu FB, Willett WC, Li TY, Manson JE, Logroscino G, Rexrode KM. Prospective study of type 1 and type 2 diabetes and risk of stroke subtypes: the Nurses’ Health Study. Diabetes Care. 2007;30(7):1730–1735. [DOI] [PubMed] [Google Scholar]
  • 107. Cheng R, Ma JX. Angiogenesis in diabetes and obesity. Rev Endocr Metab Disord. 2015;16(1):67–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Kovac A, Erickson MA, Banks WA. Brain microvascular pericytes are immunoactive in culture: cytokine, chemokine, nitric oxide, and LRP-1 expression in response to lipopolysaccharide. J Neuroinflammation. 2011;8:139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Dohgu S, Banks WA. Brain pericytes increase the lipopolysaccharide-enhanced transcytosis of HIV-1 free virus across the in vitro blood–brain barrier: evidence for cytokine-mediated pericyte-endothelial cell crosstalk. Fluids Barriers CNS. 2013;10(1):23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Park TI, Feisst V, Brooks AE, Rustenhoven J, Monzo HJ, Feng SX, Mee EW, Bergin PS, Oldfield R, Graham ES, et al. Cultured pericytes from human brain show phenotypic and functional differences associated with differential CD90 expression. Sci Rep. 2016;6:26587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Bergwerff M, Verberne ME, DeRuiter MC, Poelmann RE, Gittenberger-de Groot AC. Neural crest cell contribution to the developing circulatory system: implications for vascular morphology? Circ Res. 1998;82(2):221–231. [DOI] [PubMed] [Google Scholar]
  • 112. Korn J, Christ B, Kurz H. Neuroectodermal origin of brain pericytes and vascular smooth muscle cells. J Comp Neurol. 2002;442(1):78–88. [DOI] [PubMed] [Google Scholar]
  • 113. Heglind M, Cederberg A, Aquino J, Lucas G, Ernfors P, Enerback S. Lack of the central nervous system- and neural crest-expressed forkhead gene Foxs1 affects motor function and body weight. Mol Cell Biol. 2005;25(13):5616–5625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Wilm B, Ipenberg A, Hastie ND, Burch JB, Bader DM. The serosal mesothelium is a major source of smooth muscle cells of the gut vasculature. Development. 2005;132(23):5317–5328. [DOI] [PubMed] [Google Scholar]
  • 115. Que J, Wilm B, Hasegawa H, Wang F, Bader D, Hogan BL. Mesothelium contributes to vascular smooth muscle and mesenchyme during lung development. Proc Natl Acad Sci USA. 2008;105(43):16626–16630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Asahina K, Zhou B, Pu WT, Tsukamoto H. Septum transversum-derived mesothelium gives rise to hepatic stellate cells and perivascular mesenchymal cells in developing mouse liver. Hepatology. 2011;53(3):983–995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Cai CL, Martin JC, Sun Y, Cui L, Wang L, Ouyang K, Yang L, Bu L, Liang X, Zhang X, et al. A myocardial lineage derives from Tbx18 epicardial cells. Nature. 2008;454(7200):104–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Wessels A, Perez-Pomares JM. The epicardium and epicardially derived cells (EPDCs) as cardiac stem cells. Anat Rec A Discov Mol Cell Evol Biol. 2004;276(1):43–57. [DOI] [PubMed] [Google Scholar]
  • 119. Zhou B, Ma Q, Rajagopal S, Wu SM, Domian I, Rivera-Feliciano J, Jiang D, von Gise A, Ikeda S, Chien KR, et al. Epicardial progenitors contribute to the cardiomyocyte lineage in the developing heart. Nature. 2008;454(7200):109–113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Balabanov R, Washington R, Wagnerova J, Dore-Duffy P. CNS microvascular pericytes express macrophage-like function, cell surface integrin alpha M, and macrophage marker ED-2. Microvasc Res. 1996;52(2):127–142. [DOI] [PubMed] [Google Scholar]
  • 121. Balabanov R, Beaumont T, Dore-Duffy P. Role of central nervous system microvascular pericytes in activation of antigen-primed splenic T-lymphocytes. J Neurosci Res. 1999;55(5):578–587. [DOI] [PubMed] [Google Scholar]
  • 122. Domev H, Milkov I, Itskovitz-Eldor J, Dar A. Immunoevasive pericytes from human pluripotent stem cells preferentially modulate induction of allogeneic regulatory T cells. Stem Cells Transl Med. 2014;3(10):1169–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Castejon OJ. Submicroscopic changes of cortical capillary pericytes in human perifocal brain edema. J Submicrosc Cytol. 1984;16(3):601–18. [PubMed] [Google Scholar]
  • 124. Berger M, Bergers G, Arnold B, Hammerling GJ, Ganss R. Regulator of G-protein signaling-5 induction in pericytes coincides with active vessel remodeling during neovascularization. Blood. 2005;105(3):1094–1101. [DOI] [PubMed] [Google Scholar]
  • 125. Bondjers C, Kalen M, Hellstrom M, Scheidl SJ, Abramsson A, Renner O, Lindahl P, Cho H, Kehrl J, Betsholtz C. Transcription profiling of platelet-derived growth factor-B-deficient mouse embryos identifies RGS5 as a novel marker for pericytes and vascular smooth muscle cells. Am J Pathol. 2003;162(3):721–729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Cho H, Kozasa T, Bondjers C, Betsholtz C, Kehrl JH. Pericyte-specific expression of Rgs5: implications for PDGF and EDG receptor signaling during vascular maturation. FASEB J. 2003;17(3):440–442. [DOI] [PubMed] [Google Scholar]
  • 127. Li HP, Komuta Y, Kimura-Kuroda J, van Kuppevelt TH, Kawano H. Roles of chondroitin sulfate and dermatan sulfate in the formation of a lesion scar and axonal regeneration after traumatic injury of the mouse brain. J Neurotrauma. 2013;30(5):413–425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Silver J, Miller JH. Regeneration beyond the glial scar. Nat Rev Neurosci. 2004;5(2):146–156. [DOI] [PubMed] [Google Scholar]
  • 129. Cregg JM, DePaul MA, Filous AR, Lang BT, Tran A, Silver J. Functional regeneration beyond the glial scar. Exp Neurol. 2014;253:197–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. McKeon RJ, Jurynec MJ, Buck CR. The chondroitin sulfate proteoglycans neurocan and phosphacan are expressed by reactive astrocytes in the chronic CNS glial scar. J Neurosci. 1999;19(24):10778–10788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Fawcett JW, Asher RA. The glial scar and central nervous system repair. Brain Res Bull. 1999;49(6):377–391. [DOI] [PubMed] [Google Scholar]
  • 132. Kawano H, Kimura-Kuroda J, Komuta Y, Yoshioka N, Li HP, Kawamura K, Li Y, Raisman G. Role of the lesion scar in the response to damage and repair of the central nervous system. Cell Tissue Res. 2012;349(1):169–180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Fitch MT, Silver J. CNS injury, glial scars, and inflammation: inhibitory extracellular matrices and regeneration failure. Exp Neurol. 2008;209(2):294–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Asher RA, Morgenstern DA, Moon LD, Fawcett JW. Chondroitin sulphate proteoglycans: inhibitory components of the glial scar. Prog Brain Res. 2001;132:611–619. [DOI] [PubMed] [Google Scholar]
  • 135. Hoke A, Silver J. Proteoglycans and other repulsive molecules in glial boundaries during development and regeneration of the nervous system. Prog Brain Res. 1996;108:149–63. [DOI] [PubMed] [Google Scholar]
  • 136. Goritz C, Dias DO, Tomilin N, Barbacid M, Shupliakov O, Frisen J. A pericyte origin of spinal cord scar tissue. Science. 2011;333(6039):238–242. [DOI] [PubMed] [Google Scholar]
  • 137. Birbrair A, Zhang T, Wang ZM, Messi ML, Enikolopov GN, Mintz A, Delbono O. Skeletal muscle pericyte subtypes differ in their differentiation potential. Stem Cell Res. 2013;10(1):67–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Birbrair A, Zhang T, Wang ZM, Messi ML, Enikolopov GN, Mintz A, Delbono O. Role of pericytes in skeletal muscle regeneration and fat accumulation. Stem Cells Dev. 2013;22(16):2298–2314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Birbrair A, Zhang T, Files DC, Mannava S, Smith T, Wang ZM, Messi ML, Mintz A, Delbono O. Type-1 pericytes accumulate after tissue injury and produce collagen in an organ-dependent manner. Stem Cell Res Ther. 2014;5(6):122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Chen YT, Chang FC, Wu CF, Chou YH, Hsu HL, Chiang WC, Shen J, Chen YM, Wu KD, Tsai TJ, et al. Platelet-derived growth factor receptor signaling activates pericyte-myofibroblast transition in obstructive and post-ischemic kidney fibrosis. Kidney Int. 2011;80(11):1170–1181. [DOI] [PubMed] [Google Scholar]
  • 141. Tom VJ, Doller CM, Malouf AT, Silver J. Astrocyte-associated fibronectin is critical for axonal regeneration in adult white matter. J Neurosci 2004;24(42):9282–9290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Chen CW, Okada M, Proto JD, Gao X, Sekiya N, Beckman SA, Corselli M, Crisan M, Saparov A, Tobita K, et al. Human pericytes for ischemic heart repair. Stem Cells. 2013;31(2):305–316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Karow M, Sanchez R, Schichor C, Masserdotti G, Ortega F, Heinrich C, Gascon S, Khan MA, Lie DC, Dellavalle A, et al. Reprogramming of pericyte-derived cells of the adult human brain into induced neuronal cells. Cell Stem Cell. 2012;11(4):471–476. [DOI] [PubMed] [Google Scholar]
  • 144. Cai P, Luo H, Xu H, Zhu X, Xu W, Dai Y, Xiao J, Cao Y, Zhao Y, Zhao BQ, et al. Recombinant ADAMTS 13 attenuates brain injury after intracerebral hemorrhage. Stroke. 2015;46(9):2647–2653. [DOI] [PubMed] [Google Scholar]
  • 145. Omote Y, Deguchi K, Kono S, Liu N, Liu W, Kurata T, Yamashita T, Ikeda Y, Abe K. Neurovascular protection of cilostazol in stroke-prone spontaneous hypertensive rats associated with angiogenesis and pericyte proliferation. J Neurosci Res. 2014;92(3):369–374. [DOI] [PubMed] [Google Scholar]
  • 146. Takagi T, Hara H. Protective effects of cilostazol against hemorrhagic stroke: current and future perspectives. J Pharmacol Sci. 2016;131(3):155–161. [DOI] [PubMed] [Google Scholar]
  • 147. Winkler EA, Bell RD, Zlokovic BV. Pericyte-specific expression of PDGF beta receptor in mouse models with normal and deficient PDGF beta receptor signaling. Mol Neurodegener. 2010;5:32. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cell Transplantation are provided here courtesy of SAGE Publications

RESOURCES