Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 866
Filter
1.
Stem Cell Reports ; 19(7): 946-956, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38876110

ABSTRACT

Functionality of the blood-brain barrier (BBB) relies on the interaction between endothelial cells (ECs), pericytes, and astrocytes to regulate molecule transport within the central nervous system. Most experimental models for the BBB rely on freshly isolated primary brain cells. Here, we explored human induced pluripotent stem cells (hiPSCs) as a cellular source for astrocytes in a 3D vessel-on-chip (VoC) model. Self-organized microvascular networks were formed by combining hiPSC-derived ECs, human brain vascular pericytes, and hiPSC-derived astrocytes within a fibrin hydrogel. The hiPSC-ECs and pericytes showed close interactions, but, somewhat unexpectedly, addition of astrocytes disrupted microvascular network formation. However, continuous fluid perfusion or activation of cyclic AMP (cAMP) signaling rescued the vascular organization and decreased vascular permeability. Nevertheless, astrocytes did not affect the expression of proteins related to junction formation, transport, or extracellular matrix, indicating that, despite other claims, hiPSC-derived ECs do not entirely acquire a BBB-like identity in the 3D VoC model.


Subject(s)
Astrocytes , Blood-Brain Barrier , Endothelial Cells , Induced Pluripotent Stem Cells , Astrocytes/metabolism , Astrocytes/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/cytology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Pericytes/cytology , Pericytes/metabolism , Cell Differentiation , Lab-On-A-Chip Devices , Cells, Cultured , Hydrogels , Cyclic AMP/metabolism , Models, Biological
2.
ACS Biomater Sci Eng ; 10(7): 4388-4399, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38856968

ABSTRACT

In this study, fibrous polyurethane (PU) materials with average fiber diameter of 200, 500, and 1000 nm were produced using a solution blow spinning (SBS) process. The effects of the rotation speed of the collector (in the range of 200-25 000 rpm) on the fiber alignment and diameter were investigated. The results showed that fiber alignment was influenced by the rotation speed of the collector, and such alignment was possible when the fiber diameter was within a specific range. Homogeneously oriented fibers were obtained only for a fiber diameter ≥500 nm. Moreover, the changes in fiber orientation and fiber diameter (resulting from changes in the rotation speed of the collector) were more noticeable for materials with an average fiber diameter of 1000 nm in comparison to 500 nm, which suggests that the larger the fiber diameter, the better the controlled architectures that can be obtained. The porosity of the produced scaffolds was about 65-70%, except for materials with a fiber diameter of 1000 nm and aligned fibers, which had a higher porosity (76%). Thus, the scaffold pore size increased with increasing fiber diameter but decreased with increasing fiber alignment. The mechanical properties of fibrous materials strongly depend on the direction of stretching, whereby the fiber orientation influences the mechanical strength only for materials with a fiber diameter of 1000 nm. Furthermore, the fiber diameter and alignment affected the pericyte growth. Significant differences in cell growth were observed after 7 days of cell culture between materials with a fiber diameter of 1000 nm (cell coverage 96-99%) and those with a fiber diameter of 500 nm (cell coverage 70-90%). By appropriately setting the SBS process parameters, scaffolds can be easily adapted to the cell requirements, which is of great importance in producing complex 3D structures for guided tissue regeneration.


Subject(s)
Pericytes , Polyurethanes , Tissue Scaffolds , Polyurethanes/chemistry , Tissue Scaffolds/chemistry , Pericytes/cytology , Pericytes/physiology , Porosity , Animals , Cell Proliferation , Tissue Engineering/methods , Materials Testing
3.
Tissue Cell ; 89: 102431, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38870572

ABSTRACT

Tunneling nanotubes (TNTs) represent an innovative way for cells to communicate with one another, as they act as long conduits between cells. However, their roles in human dermal microvascular pericytes (HDMPCs) interaction remain elusive in vitro. In this work, we identified and characterized the TNT-like structures that connected two or more pericytes in two-dimensional cultures and formed a functional network in the human dermis. Immunofluorescence assay indicated that the F-actin was an essential element to form inter-pericyte TNT-like structures, as it decreased in actin polymer inhibitor-cytochalasin B treated groups, and microtubules were present in almost half of the TNT-like structures. Most importantly, we only found the presence of mitochondrial in TNT-like structures containing α-tubulin, and the application of microtubule assembly inhibitor-Nocodazole significantly reduced the percentage of TNT-like structures that contain α-tubulin, resulting in a sudden decrease in the positive rate of cytochrome c oxidase subunit 4 isoform 1 (COX IV, a marker of mitochondria) in TNT-like structures. In summary, we described a novel intercellular communication-TNT-like structures-between HDMPCs in vitro, and this work allows us to properly understand the cellular mechanisms of spreading materials between HDMPCs, shedding light on the role of HDMPCs.


Subject(s)
Pericytes , Humans , Pericytes/cytology , Pericytes/metabolism , Tubulin/metabolism , Microtubules/metabolism , Dermis/cytology , Dermis/metabolism , Cell Communication , Mitochondria/metabolism , Actins/metabolism , Nanotubes/chemistry , Microvessels/cytology , Microvessels/metabolism , Cells, Cultured , Cell Membrane Structures
4.
PLoS Biol ; 22(4): e3002590, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38683849

ABSTRACT

Brain pericytes are one of the critical cell types that regulate endothelial barrier function and activity, thus ensuring adequate blood flow to the brain. The genetic pathways guiding undifferentiated cells into mature pericytes are not well understood. We show here that pericyte precursor populations from both neural crest and head mesoderm of zebrafish express the transcription factor nkx3.1 develop into brain pericytes. We identify the gene signature of these precursors and show that an nkx3.1-, foxf2a-, and cxcl12b-expressing pericyte precursor population is present around the basilar artery prior to artery formation and pericyte recruitment. The precursors later spread throughout the brain and differentiate to express canonical pericyte markers. Cxcl12b-Cxcr4 signaling is required for pericyte attachment and differentiation. Further, both nkx3.1 and cxcl12b are necessary and sufficient in regulating pericyte number as loss inhibits and gain increases pericyte number. Through genetic experiments, we have defined a precursor population for brain pericytes and identified genes critical for their differentiation.


Subject(s)
Brain , Pericytes , Transcription Factors , Zebrafish Proteins , Animals , Brain/metabolism , Brain/embryology , Cell Differentiation , Chemokine CXCL12/metabolism , Chemokine CXCL12/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Mesoderm/metabolism , Mesoderm/cytology , Neural Crest/metabolism , Neural Crest/cytology , Pericytes/metabolism , Pericytes/cytology , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Signal Transduction , Transcription Factors/metabolism , Transcription Factors/genetics , Zebrafish/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics
5.
Dev Cell ; 59(10): 1233-1251.e5, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38569546

ABSTRACT

De novo brown adipogenesis holds potential in combating the epidemics of obesity and diabetes. However, the identity of brown adipocyte progenitor cells (APCs) and their regulation have not been extensively explored. Here, through in vivo lineage tracing and mouse modeling, we observed that platelet-derived growth factor receptor beta (PDGFRß)+ pericytes give rise to developmental brown adipocytes but not to those in adult homeostasis. By contrast, T-box 18 (TBX18)+ pericytes contribute to brown adipogenesis throughout both developmental and adult stages, though in a depot-specific manner. Mechanistically, Notch inhibition in PDGFRß+ pericytes promotes brown adipogenesis by downregulating PDGFRß. Furthermore, inhibition of Notch signaling in PDGFRß+ pericytes mitigates high-fat, high-sucrose (HFHS)-induced glucose and metabolic impairment in mice during their development and juvenile phases. Collectively, these findings show that the Notch/PDGFRß axis negatively regulates developmental brown adipogenesis, and its repression promotes brown adipose tissue expansion and improves metabolic health.


Subject(s)
Adipocytes, Brown , Adipogenesis , Cell Differentiation , Receptor, Platelet-Derived Growth Factor beta , Receptors, Notch , Stem Cells , Animals , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptors, Notch/metabolism , Mice , Adipocytes, Brown/metabolism , Adipocytes, Brown/cytology , Stem Cells/metabolism , Stem Cells/cytology , Signal Transduction , Pericytes/metabolism , Pericytes/cytology , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/cytology , Mice, Inbred C57BL , Male
7.
Adv Healthc Mater ; 13(18): e2400388, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38465502

ABSTRACT

Hydrogel-based 3D cell cultures can recapitulate (patho)physiological phenomena ex vivo. However, due to their complex multifactorial regulation, adapting these tissue and disease models for high-throughput screening workflows remains challenging. In this study, a new precision culture scaling (PCS-X) methodology combines statistical techniques (design of experiment and multiple linear regression) with automated, parallelized experiments and analyses to customize hydrogel-based vasculogenesis cultures using human umbilical vein endothelial cells and retinal microvascular endothelial cells. Variations of cell density, growth factor supplementation, and media composition are systematically explored to induce vasculogenesis in endothelial mono- and cocultures with mesenchymal stromal cells or retinal microvascular pericytes in 384-well plate formats. The developed cultures are shown to respond to vasculogenesis inhibitors in a compound- and dose-dependent manner, demonstrating the scope and power of PCS-X in creating parallelized tissue and disease models for drug discovery and individualized therapies.


Subject(s)
Human Umbilical Vein Endothelial Cells , Neovascularization, Physiologic , Humans , Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Physiologic/drug effects , Hydrogels/chemistry , Coculture Techniques/methods , High-Throughput Screening Assays/methods , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Pericytes/cytology , Pericytes/metabolism , Pericytes/drug effects , Cell Culture Techniques/methods , Cell Culture Techniques/instrumentation , Endothelial Cells/cytology , Endothelial Cells/metabolism
8.
Biophys J ; 123(14): 2110-2121, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38444160

ABSTRACT

Capillaries, composed of electrically coupled endothelial cells and overlying pericytes, constitute the vast majority of blood vessels in the brain. The most arteriole-proximate three to four branches of the capillary bed are covered by α-actin-expressing, contractile pericytes. These mural cells have a distinctive morphology and express different markers compared with their smooth muscle cell (SMC) cousins but share similar excitation-coupling contraction machinery. Despite this similarity, pericytes are considerably more depolarized than SMCs at low intravascular pressures. We have recently shown that pericytes, such as SMCs, possess functional voltage-dependent Ca2+ channels and ATP-sensitive K+ channels. Here, we further investigate the complement of pericyte ion channels, focusing on members of the K+ channel superfamily. Using NG2-DsRed-transgenic mice and diverse configurations of the patch-clamp technique, we demonstrate that pericytes display robust inward-rectifier K+ currents that are primarily mediated by the Kir2 family, based on their unique biophysical characteristics and sensitivity to micromolar concentrations of Ba2+. Moreover, multiple lines of evidence, including characteristic kinetics, sensitivity to specific blockers, biophysical attributes, and distinctive single-channel properties, established the functional expression of two voltage-dependent K+ channels: KV1 and BKCa. Although these three types of channels are also present in SMCs, they exhibit distinctive current density and kinetics profiles in pericytes. Collectively, these findings underscore differences in the operation of shared molecular features between pericytes and SMCs and highlight the potential contribution of these three K+ ion channels in setting pericyte membrane potential, modulating capillary hemodynamics, and regulating cerebral blood flow.


Subject(s)
Brain , Capillaries , Pericytes , Pericytes/metabolism , Pericytes/cytology , Animals , Capillaries/metabolism , Capillaries/cytology , Mice , Brain/blood supply , Brain/cytology , Brain/metabolism , Potassium Channels/metabolism , Mice, Transgenic , Potassium Channels, Inwardly Rectifying/metabolism , Mice, Inbred C57BL
9.
Dev Dyn ; 253(5): 519-541, 2024 May.
Article in English | MEDLINE | ID: mdl-38112237

ABSTRACT

BACKGROUND: Mural cells are an essential perivascular cell population that associate with blood vessels and contribute to vascular stabilization and tone. In the embryonic zebrafish vasculature, pdgfrb and tagln are commonly used as markers for identifying pericytes and vascular smooth muscle cells. However, the overlapping and distinct expression patterns of these markers in tandem have not been fully described. RESULTS: Here, we used the Tg(pdgfrb:Gal4FF; UAS:RFP) and Tg(tagln:NLS-EGFP) transgenic lines to identify single- and double-positive perivascular cell populations on the cranial, axial, and intersegmental vessels between 1 and 5 days postfertilization. From this comparative analysis, we discovered two novel regions of tagln-positive cell populations that have the potential to function as mural cell precursors. Specifically, we found that the hypochord-a reportedly transient structure-contributes to tagln-positive cells along the dorsal aorta. We also identified a unique mural cell progenitor population that resides along the midline between the neural tube and notochord and contributes to intersegmental vessel mural cell coverage. CONCLUSION: Together, our findings highlight the variability and versatility of tracking both pdgfrb and tagln expression in mural cells of the developing zebrafish embryo and reveal unexpected embryonic cell populations that express pdgfrb and tagln.


Subject(s)
Animals, Genetically Modified , Pericytes , Zebrafish Proteins , Zebrafish , Animals , Blood Vessels/embryology , Blood Vessels/cytology , Blood Vessels/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Embryonic Development/physiology , Pericytes/cytology , Pericytes/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptor, Platelet-Derived Growth Factor beta/genetics , Zebrafish/embryology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
10.
Cells ; 12(8)2023 04 15.
Article in English | MEDLINE | ID: mdl-37190075

ABSTRACT

Cardiac diseases are the foremost cause of morbidity and mortality worldwide. The heart has limited regenerative potential; therefore, lost cardiac tissue cannot be replenished after cardiac injury. Conventional therapies are unable to restore functional cardiac tissue. In recent decades, much attention has been paid to regenerative medicine to overcome this issue. Direct reprogramming is a promising therapeutic approach in regenerative cardiac medicine that has the potential to provide in situ cardiac regeneration. It consists of direct cell fate conversion of one cell type into another, avoiding transition through an intermediary pluripotent state. In injured cardiac tissue, this strategy directs transdifferentiation of resident non-myocyte cells (NMCs) into mature functional cardiac cells that help to restore the native tissue. Over the years, developments in reprogramming methods have suggested that regulation of several intrinsic factors in NMCs can help to achieve in situ direct cardiac reprogramming. Among NMCs, endogenous cardiac fibroblasts have been studied for their potential to be directly reprogrammed into both induced cardiomyocytes and induced cardiac progenitor cells, while pericytes can transdifferentiate towards endothelial cells and smooth muscle cells. This strategy has been indicated to improve heart function and reduce fibrosis after cardiac injury in preclinical models. This review summarizes the recent updates and progress in direct cardiac reprogramming of resident NMCs for in situ cardiac regeneration.


Subject(s)
Cell Transdifferentiation , Cellular Reprogramming Techniques , Cellular Reprogramming , Fibroblasts , Heart Diseases , Heart , Pericytes , Regeneration , Heart/physiology , Heart Diseases/therapy , Fibroblasts/cytology , Fibroblasts/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Pericytes/cytology , Pericytes/physiology , Endothelial Cells/cytology , Endothelial Cells/physiology , Humans , Animals
11.
Cell Biochem Funct ; 40(5): 439-450, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35707856

ABSTRACT

Regular soft tissue healing relies on the well-organized interaction of different stromal cell types with endothelial cells. However, spatiotemporal conditions might provoke high densities of one special stromal cell type, potentially leading to impaired healing. Detailed knowledge of the functions of rivaling stromal cell types aiming for tissue contraction and stabilization as well as vascular support is mandatory. By the application of an in vitro approach comprising the evaluation of cell proliferation, cell morphology, myofibroblastoid differentiation, and cytokine release, we verified a density-dependent modulation of these functions among juvenile and adult fibroblasts, pericytes, and adipose-derived stem cells during their interaction with microvascular endothelial cells in cocultures. Results indicate that juvenile fibroblasts rather support angiogenesis via paracrine regulation at the early stage of healing, a role potentially compromised in adult fibroblasts. In contrast, pericytes showed a more versatile character aiming at angiogenesis, vessel stabilization, and tissue contraction. Such a universal character was even more pronounced among adipose-derived stem cells. The explicit knowledge of the characteristic functions of stromal cell types is a prerequisite for the development of new analytical and therapeutic approaches for impaired soft tissue healing. The present study delivers new considerations concerning the roles of rivaling stromal cell types within a granulation tissue, pointing to extraordinary properties of pericytes and adipose-derived stem cells.


Subject(s)
Endothelial Cells , Stromal Cells , Wound Healing , Adipose Tissue/cytology , Cell Count , Endothelial Cells/cytology , Fibroblasts/cytology , Humans , Neovascularization, Pathologic , Pericytes/cytology , Stem Cells/cytology , Stromal Cells/cytology
12.
Nat Protoc ; 17(1): 95-128, 2022 01.
Article in English | MEDLINE | ID: mdl-34997242

ABSTRACT

The blood-brain barrier (BBB) greatly restricts the entry of biological and engineered therapeutic molecules into the brain. Due to challenges in translating results from animal models to the clinic, relevant in vitro human BBB models are needed to assess pathophysiological molecular transport mechanisms and enable the design of targeted therapies for neurological disorders. This protocol describes an in vitro model of the human BBB self-assembled within microfluidic devices from stem-cell-derived or primary brain endothelial cells, and primary brain pericytes and astrocytes. This protocol requires 1.5 d for device fabrication, 7 d for device culture and up to 5 d for downstream imaging, protein and gene expression analyses. Methodologies to measure the permeability of any molecule in the BBB model, which take 30 min per device, are also included. Compared with standard 2D assays, the BBB model features relevant cellular organization and morphological characteristics, as well as values of molecular permeability within the range expected in vivo. These properties, coupled with a functional brain endothelial expression profile and the capability to easily test several repeats with low reagent consumption, make this BBB model highly suitable for widespread use in academic and industrial laboratories.


Subject(s)
Blood-Brain Barrier , Capillary Permeability/physiology , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/instrumentation , Models, Cardiovascular , Astrocytes/cytology , Blood-Brain Barrier/cytology , Blood-Brain Barrier/physiology , Brain/cytology , Cells, Cultured , Endothelial Cells/cytology , Humans , Pericytes/cytology
13.
Science ; 375(6584): eabi7377, 2022 03 04.
Article in English | MEDLINE | ID: mdl-35084939

ABSTRACT

Cerebrovascular diseases are a leading cause of death and neurologic disability. Further understanding of disease mechanisms and therapeutic strategies requires a deeper knowledge of cerebrovascular cells in humans. We profiled transcriptomes of 181,388 cells to define a cell atlas of the adult human cerebrovasculature, including endothelial cell molecular signatures with arteriovenous segmentation and expanded perivascular cell diversity. By leveraging this reference, we investigated cellular and molecular perturbations in brain arteriovenous malformations, which are a leading cause of stroke in young people, and identified pathologic endothelial transformations with abnormal vascular patterning and the ontology of vascularly derived inflammation. We illustrate the interplay between vascular and immune cells that contributes to brain hemorrhage and catalog opportunities for targeting angiogenic and inflammatory programs in vascular malformations.


Subject(s)
Blood Vessels/cytology , Brain/blood supply , Intracranial Arteriovenous Malformations/pathology , Transcriptome , Adult , Blood Vessels/pathology , Blood Vessels/physiology , Blood Vessels/physiopathology , Cells, Cultured , Cerebral Cortex/blood supply , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/physiopathology , Cerebrovascular Circulation , Endothelial Cells/cytology , Endothelial Cells/pathology , Endothelial Cells/physiology , Fibroblasts/cytology , Fibroblasts/physiology , Humans , Inflammation , Intracranial Arteriovenous Malformations/metabolism , Monocytes/cytology , Monocytes/physiology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiology , Pericytes/cytology , Pericytes/physiology , RNA-Seq , Single-Cell Analysis
14.
Arterioscler Thromb Vasc Biol ; 42(2): 205-222, 2022 02.
Article in English | MEDLINE | ID: mdl-34879709

ABSTRACT

OBJECTIVE: We sought to determine how endothelial cell (EC) expression of the activating k-Ras (kirsten rat sarcoma 2 viral oncogene homolog) mutation, k-RasV12, affects their ability to form lumens and tubes and interact with pericytes during capillary assembly Approach and Results: Using defined bioassays where human ECs undergo observable tubulogenesis, sprouting behavior, pericyte recruitment to EC-lined tubes, and pericyte-induced EC basement membrane deposition, we assessed the impact of EC k-RasV12 expression on these critical processes that are necessary for proper capillary network formation. This mutation, which is frequently seen in human ECs within brain arteriovenous malformations, was found to markedly accentuate EC lumen formation mechanisms, with strongly accelerated intracellular vacuole formation, vacuole fusion, and lumen expansion and with reduced sprouting behavior, leading to excessively widened tube networks compared with control ECs. These abnormal tubes demonstrate strong reductions in pericyte recruitment and pericyte-induced EC basement membranes compared with controls, with deficiencies in fibronectin, collagen type IV, and perlecan deposition. Analyses of signaling during tube formation from these k-RasV12 ECs reveals strong enhancement of Src (Src proto-oncogene, non-receptor tyrosine kinase), Pak2 (P21 [RAC1 (Rac family small GTPase 1)] activated kinase 2), b-Raf (v-raf murine sarcoma viral oncogene homolog B1), Erk (extracellular signal-related kinase), and Akt (AK strain transforming) activation and increased expression of PKCε (protein kinase C epsilon), MT1-MMP (membrane-type 1 matrix metalloproteinase), acetylated tubulin and CDCP1 (CUB domain-containing protein 1; most are known EC lumen regulators). Pharmacological blockade of MT1-MMP, Src, Pak, Raf, Mek (mitogen-activated protein kinase) kinases, Cdc42 (cell division cycle 42)/Rac1, and Notch markedly interferes with lumen and tube formation from these ECs. CONCLUSIONS: Overall, this novel work demonstrates that EC expression of k-RasV12 disrupts capillary assembly due to markedly excessive lumen formation coupled with strongly reduced pericyte recruitment and basement membrane deposition, which are critical pathogenic features predisposing the vasculature to develop arteriovenous malformations.


Subject(s)
Basement Membrane/cytology , Capillaries/physiology , Endothelial Cells/cytology , Neovascularization, Physiologic , Pericytes/cytology , Proto-Oncogene Proteins p21(ras)/genetics , Basement Membrane/metabolism , Cell Line , Endothelial Cells/metabolism , Gene Expression , Human Umbilical Vein Endothelial Cells , Humans , Mutation , Pericytes/metabolism
15.
J Neurosci ; 42(3): 362-376, 2022 01 19.
Article in English | MEDLINE | ID: mdl-34819341

ABSTRACT

Multifaceted microglial functions in the developing brain, such as promoting the differentiation of neural progenitors and contributing to the positioning and survival of neurons, have been progressively revealed. Although previous studies have noted the relationship between vascular endothelial cells and microglia in the developing brain, little attention has been given to the importance of pericytes, the mural cells surrounding endothelial cells. In this study, we attempted to dissect the role of pericytes in microglial distribution and function in developing mouse brains. Our immunohistochemical analysis showed that approximately half of the microglia attached to capillaries in the cerebral walls. Notably, a magnified observation of the position of microglia, vascular endothelial cells and pericytes demonstrated that microglia were preferentially associated with pericytes that covered 79.8% of the total capillary surface area. Through in vivo pericyte depletion induced by the intraventricular administration of a neutralizing antibody against platelet-derived growth factor receptor (PDGFR)ß (clone APB5), we found that microglial density was markedly decreased compared with that in control antibody-treated brains because of their low proliferative capacity. Moreover, in vitro coculture of isolated CD11b+ microglia and NG2+PDGFRα- cells, which are mostly composed of pericytes, from parenchymal cells indicated that pericytes promote microglial proliferation via the production of soluble factors. Furthermore, pericyte depletion by APB5 treatment resulted in a failure of microglia to promote the differentiation of neural stem cells into intermediate progenitors. Taken together, our findings suggest that pericytes facilitate microglial homeostasis in the developing brains, thereby indirectly supporting microglial effects on neural progenitors.SIGNIFICANCE STATEMENT This study highlights the novel effect of pericytes on microglia in the developing mouse brain. Through multiple analyses using an in vivo pericyte depletion mouse model and an in vitro coculture study of isolated pericytes and microglia from parenchymal cells, we demonstrated that pericytes contribute to microglial proliferation and support microglia in efficiently promoting the differentiation of neural stem cells into intermediate progenitors. Our present data provide evidence that pericytes function not only in the maintenance of cerebral microcirculation and blood brain barrier (BBB) integrity but also in microglial homeostasis in the developing cerebral walls. These findings will expand our knowledge and help elucidate the mechanism of brain development both in healthy and disease conditions.


Subject(s)
Cerebral Cortex/cytology , Homeostasis/physiology , Microglia/cytology , Neural Stem Cells/cytology , Pericytes/cytology , Animals , Antibodies, Neutralizing , Blood-Brain Barrier/cytology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/embryology , Capillary Permeability/drug effects , Cell Line , Cell Proliferation/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/embryology , Clodronic Acid/pharmacology , Homeostasis/drug effects , Liposomes , Mice , Microglia/drug effects , Neural Stem Cells/drug effects , Pericytes/drug effects , Receptor, Platelet-Derived Growth Factor beta
16.
Exp Cell Res ; 411(1): 112983, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34921827

ABSTRACT

After the severe initial insults of acute kidney injury, progressive kidney tubulointerstitial fibrosis may occur, the peritubular capillary (PTC) rarefaction plays a key role in the disease progression. However, the mechanisms of PTC damage were not fully understood and potential therapeutic interventions were not explored. Previous studies of our research team and others in this field suggested that bone marrow-derived mesenchymal stem cells (BMSCs) transplanted into the AKI rat model may preserve the kidney function and pathological changes. In the current study, with the ischemia/reperfusion AKI rat model, we revealed that BMSCs transplantation attenuated the renal function decrease in the AKI model through preserving the peritubular capillaries (PTCs) function. The density of PTCs is maintained by BMSCs transplantation in the AKI model, detachment and relocation of pericytes in the PTCs diminished. Then we established that BMSCs transplantation may attenuate the renal fibrosis and preserve the kidney function after AKI by repairing the PTCs. Improving the vitality of pericytes, suppressing the detachment and trans-differentiation of pericytes, directly differentiation of BMSCs into pericytes by BMSCs transplantation all participate in the PTC repair. Through these processes, BMSCs rescued the microvascular damage and improved the density of PTCs. As a result, a preliminary conclusion can be reached that BMSCs transplantation can be an effective therapy for delaying renal fibrosis after AKI.


Subject(s)
Acute Kidney Injury/complications , Endothelium, Vascular/cytology , Fibrosis/therapy , Kidney Diseases/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Pericytes/cytology , Animals , Fibrosis/etiology , Fibrosis/metabolism , Fibrosis/pathology , Kidney Diseases/etiology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Male , Rats , Rats, Sprague-Dawley
17.
Int J Mol Sci ; 22(24)2021 Dec 14.
Article in English | MEDLINE | ID: mdl-34948214

ABSTRACT

The term "stromal cells" refers to a highly heterogeneous class of connective tissue cells that build the infrastructure of any organ and fulfill a variety of fundamental roles in health and disease [...].


Subject(s)
Stromal Cells/cytology , Connective Tissue Cells/cytology , Fibroblasts/cytology , Humans , Mesenchymal Stem Cells/cytology , Pericytes/cytology , Telocytes/cytology
18.
Neurobiol Dis ; 161: 105561, 2021 12.
Article in English | MEDLINE | ID: mdl-34780863

ABSTRACT

Coronavirus disease 19 (COVID-19) is a respiratory illness caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). COVID-19 pathogenesis causes vascular-mediated neurological disorders via elusive mechanisms. SARS-CoV-2 infects host cells via the binding of viral Spike (S) protein to transmembrane receptor, angiotensin-converting enzyme 2 (ACE2). Although brain pericytes were recently shown to abundantly express ACE2 at the neurovascular interface, their response to SARS-CoV-2 S protein is still to be elucidated. Using cell-based assays, we found that ACE2 expression in human brain vascular pericytes was increased upon S protein exposure. Pericytes exposed to S protein underwent profound phenotypic changes associated with an elongated and contracted morphology accompanied with an enhanced expression of contractile and myofibrogenic proteins, such as α-smooth muscle actin (α-SMA), fibronectin, collagen I, and neurogenic locus notch homolog protein-3 (NOTCH3). On the functional level, S protein exposure promoted the acquisition of calcium (Ca2+) signature of contractile ensheathing pericytes characterized by highly regular oscillatory Ca2+ fluctuations. Furthermore, S protein induced lipid peroxidation, oxidative and nitrosative stress in pericytes as well as triggered an immune reaction translated by activation of nuclear factor-kappa-B (NF-κB) signaling pathway, which was potentiated by hypoxia, a condition associated with vascular comorbidities that exacerbate COVID-19 pathogenesis. S protein exposure combined to hypoxia enhanced the production of pro-inflammatory cytokines involved in immune cell activation and trafficking, namely macrophage migration inhibitory factor (MIF). Using transgenic mice expressing the human ACE2 that recognizes S protein, we observed that the intranasal infection with SARS-CoV-2 rapidly induced hypoxic/ischemic-like pericyte reactivity in the brain of transgenic mice, accompanied with an increased vascular expression of ACE2. Moreover, we found that SARS-CoV-2 S protein accumulated in the intranasal cavity reached the brain of mice in which the nasal mucosa is deregulated. Collectively, these findings suggest that SARS-CoV-2 S protein impairs the vascular and immune regulatory functions of brain pericytes, which may account for vascular-mediated brain damage. Our study provides a better understanding for the mechanisms underlying cerebrovascular disorders in COVID-19, paving the way to develop new therapeutic interventions.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , Brain/metabolism , COVID-19/metabolism , Hypoxia-Ischemia, Brain/metabolism , Hypoxia/metabolism , Inflammation/metabolism , Pericytes/metabolism , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Actins/metabolism , Angiotensin-Converting Enzyme 2/drug effects , Angiotensin-Converting Enzyme 2/genetics , Animals , Brain/blood supply , COVID-19/physiopathology , Calcium Signaling , Collagen Type I/metabolism , Fibronectins/metabolism , Humans , Hypoxia-Ischemia, Brain/physiopathology , Lipid Peroxidation/drug effects , Lipid Peroxidation/genetics , Macrophage Migration-Inhibitory Factors/drug effects , Macrophage Migration-Inhibitory Factors/metabolism , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Myofibroblasts , NF-kappa B/drug effects , NF-kappa B/metabolism , Nasal Mucosa , Nitrosative Stress , Oxidative Stress , Pericytes/cytology , Pericytes/drug effects , Phenotype , Receptor, Notch3/metabolism , Receptors, Coronavirus/drug effects , Receptors, Coronavirus/genetics , Receptors, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/pharmacology
19.
Exp Eye Res ; 212: 108771, 2021 11.
Article in English | MEDLINE | ID: mdl-34624336

ABSTRACT

The choriocapillaris (CC), the capillary bed in the choroid, essentially nourishes the photoreceptor cells. Its damage in aging and age-related diseases significantly influences the survival of the photoreceptor cells. Earlier reports implicated endothelial loss in aged and diseased CC; however, age-related pericyte changes and their contribution in CC death remain unknown. We examined human donor eyes (age: 56-94 years; N = 24), and found that CC pericyte damage preceded endothelial changes. With aging (>70 years), the sub-macular choroid accumulated debris in Bruch's membrane (BM). Of the debris content, the long-spaced collagens had a tendency to settle over the capillary basal lamina (BL), and this often resulted in endothelial projection into capillary lumen. Between 75 and 83 years, pericytes contained dark mitochondria, and their processes facing the BM debris showed partial loss of BL and intermediate filaments (IFs), when the endothelium remained unaltered. The endothelial changes appeared beyond 83 years, the abundance of IFs and autophagy reinforced their survival until late aging. TUNEL+ pericytes, and immunoreactivity to carboxymethyl lysine and 4-hydroxy 2-nonenal, but no nitro-tyrosine, was detected in aged CC walls. Iba-1+ dystrophic microglia were present in the vicinity of the CC. Our data indicate that (1) BM debris exerts pressure on the CC, leading to the damage of the capillary BL and pericyte processes (2) loss of IFs results in early pericyte destabilization (3) capillary wall undergoes lipid peroxidative and glycative damage, and (4) pericyte damage leads to late endothelial changes and ultimately CC loss. Future research should explore the normal ways of pericyte maintenance in the aging nervous system.


Subject(s)
Aging/physiology , Choroid/cytology , Endothelium, Vascular/cytology , Oxidative Stress/physiology , Pericytes/cytology , Aged , Aged, 80 and over , Female , Humans , In Situ Nick-End Labeling , Male , Middle Aged
20.
Dis Markers ; 2021: 7124835, 2021.
Article in English | MEDLINE | ID: mdl-34630739

ABSTRACT

Inner blood-retina barrier (iBRB) is primarily formed of retinal microvascular endothelial cells (ECs) with tight junctions, which are surrounded and supported by retinal microvascular pericytes (RMPs) and basement membrane. Pericytes are believed to be critically involved in the physiology and pathology of iBRB. However, the underlying mechanism remains to be fully elucidated. We developed a novel in vitro iBRB model which was composed of primary cultures of rat retinal ECs and RMPs based on Transwell system. We tested the involvement of pericytes in the migration and invasion of ECs, examined the expression and activity of matrix metalloproteinase- (MMP-) 2/MMP-9 in the culture, evaluated the TEER and permeability of iBRB, and assessed the expression of ZO-1, occludin, claudin-5, and VE-cadherin of endothelial junctions. We found that RMPs with indirect contact of ECs can increase the expression of MMP-2 and upgrade the activity of MMP-2/9 in the coculture, which subsequently decreased TJ protein abundance of ZO-1 and occludin in ECs, promoted the migration of ECs, and finally reduced the integrity of iBRB. Taken together, our data show that RMP relative location with ECs is involved in the integrity of iBRB via MMP-2/9 and has important implications for treating diabetic retinopathy and other retinal disorders involving iBRB dysfunction.


Subject(s)
Blood-Retinal Barrier/cytology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Pericytes/cytology , Retina/cytology , Animals , Blood-Retinal Barrier/metabolism , Cell Movement , Cells, Cultured , Coculture Techniques , Endothelial Cells/cytology , Endothelial Cells/metabolism , Male , Models, Biological , Pericytes/metabolism , Primary Cell Culture , Rats , Retina/metabolism , Tight Junctions/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL