Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.682
Filter
2.
FASEB J ; 38(10): e23679, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38780117

ABSTRACT

Retinal vascular diseases (RVDs), in particular diabetic retinopathy, retinal vein occlusion, and retinopathy of prematurity, are leading contributors to blindness. The pathogenesis of RVD involves vessel dilatation, leakage, and occlusion; however, the specific underlying mechanisms remain unclear. Recent findings have indicated that pericytes (PCs), as critical members of the vascular mural cells, significantly contribute to the progression of RVDs, including detachment from microvessels, alteration of contractile and secretory properties, and excessive production of the extracellular matrix. Moreover, PCs are believed to have mesenchymal stem properties and, therefore, might contribute to regenerative therapy. Here, we review novel ideas concerning PC characteristics and functions in RVDs and discuss potential therapeutic strategies based on PCs, including the targeting of pathological signals and cell-based regenerative treatments.


Subject(s)
Pericytes , Pericytes/metabolism , Humans , Animals , Retinal Vessels/metabolism , Retinal Vessels/pathology , Retinal Diseases/therapy , Retinal Diseases/metabolism , Retinal Diseases/pathology , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/therapy , Diabetic Retinopathy/pathology
3.
Nat Commun ; 15(1): 4097, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755144

ABSTRACT

Angiogenesis, the growth of new blood vessels from pre-existing vasculature, is essential for the development of new organ systems, but transcriptional control of angiogenesis remains incompletely understood. Here we show that FOXC1 is essential for retinal angiogenesis. Endothelial cell (EC)-specific loss of Foxc1 impairs retinal vascular growth and expression of Slc3a2 and Slc7a5, which encode the heterodimeric CD98 (LAT1/4F2hc) amino acid transporter and regulate the intracellular transport of essential amino acids and activation of the mammalian target of rapamycin (mTOR). EC-Foxc1 deficiency diminishes mTOR activity, while administration of the mTOR agonist MHY-1485 rescues perturbed retinal angiogenesis. EC-Foxc1 expression is required for retinal revascularization and resolution of neovascular tufts in a model of oxygen-induced retinopathy. Foxc1 is also indispensable for pericytes, a critical component of the blood-retina barrier during retinal angiogenesis. Our findings establish FOXC1 as a crucial regulator of retinal vessels and identify therapeutic targets for treating retinal vascular disease.


Subject(s)
Blood-Retinal Barrier , Endothelial Cells , Forkhead Transcription Factors , Retinal Neovascularization , Animals , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Retinal Neovascularization/metabolism , Retinal Neovascularization/genetics , Retinal Neovascularization/pathology , Mice , Endothelial Cells/metabolism , Blood-Retinal Barrier/metabolism , TOR Serine-Threonine Kinases/metabolism , Pericytes/metabolism , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Fusion Regulatory Protein 1, Heavy Chain/genetics , Retinal Vessels/metabolism , Humans , Large Neutral Amino Acid-Transporter 1/metabolism , Large Neutral Amino Acid-Transporter 1/genetics , Mice, Knockout , Mice, Inbred C57BL , Retina/metabolism , Male , Angiogenesis
4.
Int J Mol Sci ; 25(10)2024 May 07.
Article in English | MEDLINE | ID: mdl-38791110

ABSTRACT

Vascular co-option is a consequence of the direct interaction between perivascular cells, known as pericytes (PCs), and glioblastoma multiforme (GBM) cells (GBMcs). This process is essential for inducing changes in the pericytes' anti-tumoral and immunoreactive phenotypes. Starting from the initial stages of carcinogenesis in GBM, PCs conditioned by GBMcs undergo proliferation, acquire a pro-tumoral and immunosuppressive phenotype by expressing and secreting immunosuppressive molecules, and significantly hinder the activation of T cells, thereby facilitating tumor growth. Inhibiting the pericyte (PC) conditioning mechanisms in the GBM tumor microenvironment (TME) results in immunological activation and tumor disappearance. This underscores the pivotal role of PCs as a key cell in the TME, responsible for tumor-induced immunosuppression and enabling GBM cells to evade the immune system. Other cells within the TME, such as tumor-associated macrophages (TAMs) and microglia, have also been identified as contributors to this immunomodulation. In this paper, we will review the role of these three cell types in the immunosuppressive properties of the TME. Our conclusion is that the cellular heterogeneity of immunocompetent cells within the TME may lead to the misinterpretation of cellular lineage identification due to different reactive stages and the identification of PCs as TAMs. Consequently, novel therapies could be developed to disrupt GBM-PC interactions and/or PC conditioning through vascular co-option, thereby exposing GBMcs to the immune system.


Subject(s)
Brain Neoplasms , Pericytes , Tumor Microenvironment , Pericytes/immunology , Pericytes/pathology , Pericytes/metabolism , Humans , Tumor Microenvironment/immunology , Animals , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Glioma/immunology , Glioma/pathology , Glioma/metabolism , Glioblastoma/immunology , Glioblastoma/pathology , Glioblastoma/metabolism , Disease Progression , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology
5.
Viruses ; 16(5)2024 05 15.
Article in English | MEDLINE | ID: mdl-38793666

ABSTRACT

SARS-CoV-2 primarily infects the lungs via the ACE2 receptor but also other organs including the kidneys, the gastrointestinal tract, the heart, and the skin. SARS-CoV-2 also infects the brain, but the hematogenous route of viral entry to the brain is still not fully characterized. Understanding how SARS-CoV-2 traverses the blood-brain barrier (BBB) as well as how it affects the molecular functions of the BBB are unclear. In this study, we investigated the roles of the receptors ACE2 and DPP4 in the SARS-CoV-2 infection of the discrete cellular components of a transwell BBB model comprising HUVECs, astrocytes, and pericytes. Our results demonstrate that direct infection on the BBB model does not modulate paracellular permeability. Also, our results show that SARS-CoV-2 utilizes clathrin and caveolin-mediated endocytosis to traverse the BBB, resulting in the direct infection of the brain side of the BBB model with a minimal endothelial infection. In conclusion, the BBB is susceptible to SARS-CoV-2 infection in multiple ways, including the direct infection of endothelium, astrocytes, and pericytes involving ACE2 and/or DPP4 and the blood-to-brain transcytosis, which is an event that does not require the presence of host receptors.


Subject(s)
Angiotensin-Converting Enzyme 2 , Astrocytes , Blood-Brain Barrier , COVID-19 , Dipeptidyl Peptidase 4 , Pericytes , SARS-CoV-2 , Transcytosis , Virus Internalization , Blood-Brain Barrier/virology , Blood-Brain Barrier/metabolism , Humans , SARS-CoV-2/physiology , Angiotensin-Converting Enzyme 2/metabolism , Pericytes/virology , Pericytes/metabolism , COVID-19/virology , COVID-19/metabolism , Astrocytes/virology , Astrocytes/metabolism , Dipeptidyl Peptidase 4/metabolism , Brain/virology , Brain/metabolism , Endocytosis , Human Umbilical Vein Endothelial Cells/virology , Permeability
6.
Cell Mol Life Sci ; 81(1): 225, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38769116

ABSTRACT

Ischemic stroke induces neovascularization of the injured tissue as an attempt to promote structural repair and neurological recovery. Angiogenesis is regulated by pericytes that potently react to ischemic stroke stressors, ranging from death to dysfunction. Platelet-derived growth factor (PDGF) receptor (PDGFR)ß controls pericyte survival, migration, and interaction with brain endothelial cells. PDGF-D a specific ligand of PDGFRß is expressed in the brain, yet its regulation and role in ischemic stroke pathobiology remains unexplored. Using experimental ischemic stroke mouse model, we found that PDGF-D is transiently induced in brain endothelial cells at the injury site in the subacute phase. To investigate the biological significance of PDGF-D post-ischemic stroke regulation, its subacute expression was either downregulated using siRNA or upregulated using an active recombinant form. Attenuation of PDGF-D subacute induction exacerbates neuronal loss, impairs microvascular density, alters vascular permeability, and increases microvascular stalling. Increasing PDGF-D subacute bioavailability rescues neuronal survival and improves neurological recovery. PDGF-D subacute enhanced bioavailability promotes stable neovascularization of the injured tissue and improves brain perfusion. Notably, PDGF-D enhanced bioavailability improves pericyte association with brain endothelial cells. Cell-based assays using human brain pericyte and brain endothelial cells exposed to ischemia-like conditions were applied to investigate the underlying mechanisms. PDGF-D stimulation attenuates pericyte loss and fibrotic transition, while increasing the secretion of pro-angiogenic and vascular protective factors. Moreover, PDGF-D stimulates pericyte migration required for optimal endothelial coverage and promotes angiogenesis. Our study unravels new insights into PDGF-D contribution to neurovascular protection after ischemic stroke by rescuing the functions of pericytes.


Subject(s)
Endothelial Cells , Ischemic Stroke , Lymphokines , Pericytes , Platelet-Derived Growth Factor , Pericytes/metabolism , Pericytes/pathology , Animals , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Mice , Lymphokines/metabolism , Lymphokines/genetics , Platelet-Derived Growth Factor/metabolism , Humans , Endothelial Cells/metabolism , Male , Mice, Inbred C57BL , Brain/metabolism , Brain/pathology , Disease Models, Animal , Neovascularization, Physiologic , Cell Movement
7.
Methods Mol Biol ; 2807: 271-283, 2024.
Article in English | MEDLINE | ID: mdl-38743235

ABSTRACT

The blood-brain barrier (BBB) is one of several barriers between the brain and the peripheral blood system to maintain homeostasis. Understanding the interactions between infectious agents such as human immunodeficiency virus type 1 (HIV-1), which are capable of traversing the BBB and causing neuroinflammation requires modeling an authentic BBB in vitro. Such an in vitro BBB model also helps develop means of targeting viruses that reside in the brain via natural immune effectors such as antibodies. The BBB consists of human brain microvascular endothelial cells (HBMECs), astrocytes, and pericytes. Here we report in vitro methods to establish a dual-cell BBB model consisting of primary HBMECs and primary astrocytes to measure the integrity of the BBB and antibody penetration of the BBB, as well as a method to establish a single cell BBB model to study the impact of HIV-1 infected medium on the integrity of such a BBB.


Subject(s)
Astrocytes , Blood-Brain Barrier , Endothelial Cells , HIV Infections , HIV-1 , Blood-Brain Barrier/virology , Blood-Brain Barrier/metabolism , Humans , Astrocytes/virology , Astrocytes/metabolism , Astrocytes/immunology , Endothelial Cells/virology , Endothelial Cells/metabolism , Endothelial Cells/immunology , HIV-1/immunology , HIV-1/physiology , HIV Infections/virology , HIV Infections/immunology , Pericytes/virology , Pericytes/metabolism , Pericytes/immunology , Neuroinflammatory Diseases/virology , Neuroinflammatory Diseases/immunology , Coculture Techniques/methods , Cells, Cultured , Brain/virology , Brain/immunology , Brain/metabolism
8.
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 , Cell Differentiation , Pericytes , Transcription Factors , Zebrafish Proteins , Zebrafish , Pericytes/metabolism , Pericytes/cytology , Animals , Zebrafish/metabolism , Zebrafish/embryology , Zebrafish/genetics , Brain/metabolism , Brain/embryology , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Cell Differentiation/genetics , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Gene Expression Regulation, Developmental , Neural Crest/metabolism , Neural Crest/cytology , Mesoderm/metabolism , Mesoderm/cytology , Signal Transduction , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Chemokine CXCL12/metabolism , Chemokine CXCL12/genetics
9.
Circ Res ; 134(10): 1240-1255, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38563133

ABSTRACT

BACKGROUND: Pericytes are capillary-associated mural cells involved in the maintenance and stability of the vascular network. Although aging is one of the main risk factors for cardiovascular disease, the consequences of aging on cardiac pericytes are unknown. METHODS: In this study, we have combined single-nucleus RNA sequencing and histological analysis to determine the effects of aging on cardiac pericytes. Furthermore, we have conducted in vivo and in vitro analysis of RGS5 (regulator of G-protein signaling 5) loss of function and finally have performed pericytes-fibroblasts coculture studies to understand the effect of RGS5 deletion in pericytes on the neighboring fibroblasts. RESULTS: Aging reduced the pericyte area and capillary coverage in the murine heart. Single-nucleus RNA sequencing analysis further revealed that the expression of Rgs5 was reduced in cardiac pericytes from aged mice. In vivo and in vitro studies showed that the deletion of RGS5 impaired cardiac function, induced fibrosis, and morphological changes in pericytes characterized by a profibrotic gene expression signature and the expression of different ECM (extracellular matrix) components and growth factors, for example, TGFB2 and PDGFB. Indeed, culturing fibroblasts with the supernatant of RGS5-deficient pericytes induced their activation as evidenced by the increased expression of αSMA (alpha smooth muscle actin) in a TGFß (transforming growth factor beta)2-dependent mechanism. CONCLUSIONS: Our results have identified RGS5 as a crucial regulator of pericyte function during cardiac aging. The deletion of RGS5 causes cardiac dysfunction and induces myocardial fibrosis, one of the hallmarks of cardiac aging.


Subject(s)
Fibroblasts , Fibrosis , Pericytes , RGS Proteins , Pericytes/metabolism , Pericytes/pathology , Animals , RGS Proteins/genetics , RGS Proteins/metabolism , RGS Proteins/deficiency , Fibroblasts/metabolism , Fibroblasts/pathology , Mice , Cells, Cultured , Aging/metabolism , Aging/pathology , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Male , Coculture Techniques
10.
J Control Release ; 369: 458-474, 2024 May.
Article in English | MEDLINE | ID: mdl-38575077

ABSTRACT

The blood-brain barrier (BBB)/blood-tumor barrier (BTB) impedes brain entry of most brain-targeted drugs, whether they are water-soluble or hydrophobic. Endothelial WNT signaling and neoplastic pericytes maintain BTB low permeability by regulating tight junctions. Here, we proposed nitazoxanide (NTZ) and ibrutinib (IBR) co-loaded ICAM-1-targeting nanoparticles (NI@I-NPs) to disrupt the BTB in a time-dependent, reversible, and size-selective manner by targeting specific ICAM-1, inactivating WNT signaling and depleting pericytes in tumor-associated blood vessels in breast cancer brain metastases. At the optimal NTZ/IBR mass ratio (1:2), BTB opening reached the optimum effect at 48-72 h without any sign of intracranial edema and cognitive impairment. The combination of NI@I-NPs and chemotherapeutic drugs (doxorubicin and etoposide) extended the median survival of mice with breast cancer brain metastases. Targeting BTB endothelial WNT signaling and tumor pericytes via NI@I-NPs could open the BTB to improve chemotherapeutic efficiency against brain metastases.


Subject(s)
Blood-Brain Barrier , Brain Neoplasms , Nanoparticles , Pericytes , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Brain Neoplasms/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Pericytes/metabolism , Pericytes/drug effects , Female , Humans , Nanoparticles/administration & dosage , Piperidines/administration & dosage , Piperidines/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Thiazoles/administration & dosage , Thiazoles/pharmacology , Cell Line, Tumor , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Doxorubicin/administration & dosage , Doxorubicin/therapeutic use , Mice, Inbred BALB C , Wnt Signaling Pathway/drug effects , Mice , Drug Delivery Systems , Adenine/analogs & derivatives
11.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167169, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38631408

ABSTRACT

Mitochondrial dysregulation is pivotal in Alzheimer's disease (AD) pathogenesis. Calcium governs vital mitochondrial processes impacting energy conversion, oxidative stress, and cell death signaling. Disruptions in mitochondrial calcium (mCa2+) handling induce calcium overload and trigger the opening of mitochondrial permeability transition pore, ensuing energy deprivation and resulting in AD-related neuronal cell death. However, the role of mCa2+ in non-neuronal cells (microglia, astrocytes, oligodendrocytes, endothelial cells, and pericytes) remains elusive. This review provides a comprehensive exploration of mitochondrial heterogeneity and calcium signaling, offering insights into specific differences among various brain cell types in AD.


Subject(s)
Alzheimer Disease , Calcium Signaling , Calcium , Mitochondria , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Humans , Mitochondria/metabolism , Mitochondria/pathology , Calcium Signaling/physiology , Animals , Calcium/metabolism , Astrocytes/metabolism , Astrocytes/pathology , Pericytes/metabolism , Pericytes/pathology , Microglia/metabolism , Microglia/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Oxidative Stress , Oligodendroglia/metabolism , Oligodendroglia/pathology , Mitochondrial Permeability Transition Pore/metabolism , Neurons/metabolism , Neurons/pathology
12.
Front Biosci (Landmark Ed) ; 29(4): 141, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38682199

ABSTRACT

Pericytes, a specific type of mesenchymal cell that surround the basement membrane of pulmonary venules and capillaries. They are crucial pathological features observed in individuals with the severe lung disease of pulmonary fibrosis (PF). The presence of pericytes leads to inflammation and fibrosis in the lung interstitium and alveolar space due to the release of various cytokines and chemokines. Pericytes also stimulate the proliferation and activation of fibroblasts, thereby promoting the progression of PF. Previous studies examining the mechanism of action of pericytes have primarily focused on cell signal transduction pathways, cell growth and death processes, and the synthesis and breakdown of extracellular matrix (ECM). Notably, the transforming growth factor-ß (TGF-ß) and Wnt signaling pathways have been associated with the action of pericytes in driving the progression of PF. It is therefore clear that pericytes play an essential role in the development of PF, while also offering possible avenues for targeted therapeutic intervention against this condition. The current article provides a comprehensive review on how pericytes contribute to inflammatory responses, as well as their importance for understanding the mechanism of PF. In addition, this review discusses the potential use of pericyte-targeted approaches for the treatment of patients affected by this debilitating lung disease.


Subject(s)
Pericytes , Pulmonary Fibrosis , Pericytes/pathology , Pericytes/metabolism , Humans , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/metabolism , Animals , Transforming Growth Factor beta/metabolism , Signal Transduction , Extracellular Matrix/metabolism , Wnt Signaling Pathway
13.
Front Biosci (Landmark Ed) ; 29(4): 136, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38682184

ABSTRACT

Alzheimer's disease (AD) is an age-related progressive neurodegenerative disorder characterized by aberrant amyloid precursor protein (APP) cleavage, pathological aggregations of beta-amyloid (Aß) that make up Aß plaques and hyperphosphorylation of Tau that makes up neurofibrillary tangles (NFTs). Although progress has been made in research on AD, the fundamental causes of this disease have not been fully elucidated. Recent studies have shown that vascular dysfunction especially the loss of pericytes plays a significant role in the onset of AD. Pericytes play a variety of important roles in the nervous system including the regulation of the cerebral blood flow (CBF), the formation and maintenance of the blood-brain barrier (BBB), angiogenesis, and the clearance of toxic substances from the brain. Pericytes participate in the transport of Aß through various receptors, and Aß acts on pericytes to cause them to constrict, detach, and die. The loss of pericytes elevates the levels of Aß1-40 and Aß1-42 by disrupting the integrity of the BBB and reducing the clearance of soluble Aß from the brain interstitial fluid. The aggravated deposition of Aß further exacerbates pericyte dysfunction, forming a vicious cycle. The combined influence of these factors eventually results in the loss of neurons and cognitive decline. Further exploration of the interactions between pericytes and Aß is beneficial for understanding AD and could lead to the identification of new therapeutic targets for the prevention and treatment of AD. In this review, we explore the characterization of pericytes, interactions between pericytes and other cells in the neurovascular unit (NVU), and the physiological functions of pericytes and dysfunctions in AD. This review discusses the interactions between pericytes and Aß, as well as current and further strategies for preventing or treating AD targeting pericytes.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Blood-Brain Barrier , Pericytes , Pericytes/metabolism , Alzheimer Disease/metabolism , Humans , Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/metabolism , Animals , Brain/metabolism
14.
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
15.
J Clin Invest ; 134(6)2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38487999

ABSTRACT

Allergic asthma generally starts during early life and is linked to substantial tissue remodeling and lung dysfunction. Although angiogenesis is a feature of the disrupted airway, the impact of allergic asthma on the pulmonary microcirculation during early life is unknown. Here, using quantitative imaging in precision-cut lung slices (PCLSs), we report that exposure of neonatal mice to house dust mite (HDM) extract disrupts endothelial cell/pericyte interactions in adventitial areas. Central to the blood vessel structure, the loss of pericyte coverage was driven by mast cell (MC) proteases, such as tryptase, that can induce pericyte retraction and loss of the critical adhesion molecule N-cadherin. Furthermore, spatial transcriptomics of pediatric asthmatic endobronchial biopsies suggests intense vascular stress and remodeling linked with increased expression of MC activation pathways in regions enriched in blood vessels. These data provide previously unappreciated insights into the pathophysiology of allergic asthma with potential long-term vascular defects.


Subject(s)
Asthma , Mast Cells , Humans , Child , Animals , Mice , Mast Cells/pathology , Pericytes/metabolism , Endothelial Cells/metabolism , Asthma/pathology , Lung/pathology , Allergens , Pyroglyphidae , Disease Models, Animal
16.
Methods Mol Biol ; 2783: 25-33, 2024.
Article in English | MEDLINE | ID: mdl-38478224

ABSTRACT

Perivascular cells represent an in vivo counterpart of mesenchymal stromal/stem cells that populate the outer layer of blood vessels. Pericytes in capillaries and microvessels and adventitial cells of large arteries and veins give rise to stem/progenitor cells when isolated and cultured in vitro. These cells have been considered candidate cell types for cell therapy. Adipose tissue, being highly vascularized, dispensable, and easily accessed, is a viable option to obtain perivascular cells for use in research and in clinical trials. Here, we describe our established protocol to extract perivascular cells from human fat through fluorescence-activated cell sorting, which allows for the isolation of defined populations of progenitor cells with high reproducibility.


Subject(s)
Mesenchymal Stem Cells , Humans , Flow Cytometry , Reproducibility of Results , Mesenchymal Stem Cells/metabolism , Pericytes/metabolism , Adipose Tissue , Cell Differentiation
17.
Biomed Pharmacother ; 174: 116436, 2024 May.
Article in English | MEDLINE | ID: mdl-38508081

ABSTRACT

In cancer immunotherapy, chimeric antigen receptors (CARs) targeting specific antigens have become a powerful tool for cell-based therapy. CAR-natural killer (NK) cells offer selective anticancer lysis with reduced off-tumor toxicity compared to CAR-T cells, which is beneficial in the heterogeneous milieu of solid tumors. In the tumor microenvironment (TME) of glioblastoma (GBM), pericytes not only support tumor growth but also contribute to immune evasion, underscoring their potential as therapeutic targets in GBM treatment. Given this context, our study aimed to target the GBM TME, with a special focus on pericytes expressing CD19, to evaluate the potential effectiveness of CD19 CAR-iNK cells against GBM. We performed CD19 CAR transduction in induced pluripotent stem cell-derived NK (iNK) cells. To determine whether CD19 CAR targets the TME pericytes in GBM, we developed GBM-blood vessel assembloids (GBVA) by fusing GBM spheroids with blood vessel organoids. When co-cultured with GBVA, CD19 CAR-iNK cells migrated towards the pericytes surrounding the GBM. Using a microfluidic chip, we demonstrated CD19 CAR-iNK cells' targeted action and cytotoxic effects in a perfusion-like environment. GBVA xenografts recapitulated the TME including human CD19-positive pericytes, thereby enabling the application of an in vivo model for validating the efficacy of CD19 CAR-iNK cells against GBM. Compared to GBM spheroids, the presence of pericytes significantly enhanced CD19 CAR-iNK cell migration towards GBM and reduced proliferation. These results underline the efficacy of CD19 CAR-iNK cells in targeting pericytes within the GBM TME, suggesting their potential therapeutic value for GBM treatment.


Subject(s)
Antigens, CD19 , Cell Movement , Glioblastoma , Induced Pluripotent Stem Cells , Killer Cells, Natural , Pericytes , Receptors, Chimeric Antigen , Tumor Microenvironment , Pericytes/metabolism , Pericytes/pathology , Humans , Glioblastoma/pathology , Glioblastoma/immunology , Glioblastoma/therapy , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Antigens, CD19/metabolism , Antigens, CD19/immunology , Animals , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/immunology , Induced Pluripotent Stem Cells/metabolism , Cell Line, Tumor , Immunotherapy, Adoptive/methods , Brain Neoplasms/pathology , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Mice , Xenograft Model Antitumor Assays
18.
Brain Res ; 1832: 148849, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38452844

ABSTRACT

The present study focused on whether hypoxia-inducible factor-1alpha (HIF-1α) and platelet-derived factor-beta (PDGF-ß) are involved in the crosstalk between brain microvascular endothelial cells (BMECs) and brain vascular pericytes (BVPs) under ischaemic-hypoxic conditions. Mono-cultures or co-cultures of BVPs and BMECs were made for the construction of the blood-brain barrier (BBB) model in vitro and then exposed to control and oxygen-glucose deprivation (OGD) conditions. BBB injury was determined by assessing the ability, apoptosis, and migration of BVPs and the transendothelial electrical resistance and horseradish peroxidase permeation of BMECs. Relative mRNA and protein levels of HIF-1α and PDGF-ß, as well as tight junction proteins ZO-1 and claudin-5 were analyzed by western blotting, reverse transcription quantitative PCR, and/or immunofluorescence staining. Dual-luciferase reporter assays assessed the relationship between PDGF-ß and HIF-1α. Co-culturing with BMECs alleviated OGD-induced reduction in BVP viability, elevation in BVP apoptosis, and repression in BVP migration. Co-culturing with BVPs protected against OGD-induced impairment on BMEC permeability. OGD-induced HIF-1α upregulation enhanced PDGF-ß expression in mono-cultured BMECs and co-cultured BMECs with BVPs. Knockdown of HIF-1α impaired the effect of BMECs on BVPs under OGD conditions, and PDGFR-ß silencing in BVPs blocked the crosstalk between BMECs and BVPs under OGD conditions. The crosstalk between BMECs and BVPs was implicated in OGD-induced BBB injury through the HIF-1α/PDGF-ß signaling.


Subject(s)
Endothelial Cells , Oxygen , Brain/metabolism , Endothelial Cells/metabolism , Glucose/metabolism , Hypoxia/metabolism , Oxygen/metabolism , Pericytes/metabolism , Proteins/metabolism
19.
J Cereb Blood Flow Metab ; 44(6): 881-895, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38513138

ABSTRACT

The blood-brain barrier (BBB) is a complex and dynamic interface that regulates the exchange of molecules and cells between the blood and the central nervous system. It undergoes structural and functional changes during aging, which may compromise its integrity and contribute to the pathogenesis of neurodegenerative diseases. In recent years, advances in microscopy and high-throughput bioinformatics have allowed a more in-depth investigation of the aging mechanisms of BBB. This review summarizes age-related alterations of the BBB structure and function from six perspectives: endothelial cells, astrocytes, pericytes, basement membrane, microglia and perivascular macrophages, and fibroblasts, ranging from the molecular level to the human multi-system level. These basic components are essential for the proper functioning of the BBB. Recent imaging methods of BBB were also reviewed. Elucidation of age-associated BBB changes may offer insights into BBB homeostasis and may provide effective therapeutic strategies to protect it during aging.


Subject(s)
Aging , Blood-Brain Barrier , Blood-Brain Barrier/metabolism , Humans , Aging/physiology , Animals , Endothelial Cells/metabolism , Pericytes/metabolism , Pericytes/physiology , Astrocytes/metabolism , Astrocytes/physiology
20.
J Exp Clin Cancer Res ; 43(1): 83, 2024 Mar 16.
Article in English | MEDLINE | ID: mdl-38493151

ABSTRACT

BACKGROUND: Tumor angiogenesis inhibitors have been applied for non-small cell lung cancer (NSCLC) therapy. However, the drug resistance hinders their further development. Intercellular crosstalk between lung cancer cells and vascular cells was crucial for anti-angiogenenic resistance (AAD). However, the understanding of this crosstalk is still rudimentary. Our previous study showed that Glioma-associated oncogene 1 (Gli1) is a driver of NSCLC metastasis, but its role in lung cancer cell-vascular cell crosstalk remains unclear. METHODS: Conditioned medium (CM) from Gli1-overexpressing or Gli1-knockdown NSCLC cells was used to educate endothelia cells and pericytes, and the effects of these media on angiogenesis and the maturation of new blood vessels were evaluated via wound healing assays, Transwell migration and invasion assays, tube formation assays and 3D coculture assays. The xenograft model was conducted to establish the effect of Gli1 on tumor angiogenesis and growth. Angiogenic antibody microarray analysis, ELISA, luciferase reporte, chromatin immunoprecipitation (ChIP), bFGF protein stability and ubiquitination assay were performed to explore how Gli1 regulate bFGF expression. RESULTS: Gli1 overexpression in NSCLC cells enhanced the endothelial cell and pericyte motility required for angiogenesis required for angiogenesis. However, Gli1 knockout in NSCLC cells had opposite effect on this process. bFGF was critical for the enhancement effect on tumor angiogenesis. bFGF treatment reversed the Gli1 knockdown-mediated inhibition of angiogenesis. Mechanistically, Gli1 increased the bFGF protein level by promoting bFGF transcriptional activity and protein stability. Importantly, suppressing Gli1 with GANT-61 obviously inhibited angiogenesis. CONCLUSION: The Gli1-bFGF axis is crucial for the crosstalk between lung cancer cells and vascular cells. Targeting Gli1 is a potential therapeutic approach for NSCLC angiogenesis.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Pericytes/metabolism , Pericytes/pathology , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism , Angiogenesis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Cell Movement , Cell Line, Tumor , Cell Proliferation
SELECTION OF CITATIONS
SEARCH DETAIL
...