Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.575
Filter
1.
Elife ; 122024 Jun 10.
Article in English | MEDLINE | ID: mdl-38856719

ABSTRACT

Erectile dysfunction (ED) affects a significant proportion of men aged 40-70 and is caused by cavernous tissue dysfunction. Presently, the most common treatment for ED is phosphodiesterase 5 inhibitors; however, this is less effective in patients with severe vascular disease such as diabetic ED. Therefore, there is a need for development of new treatment, which requires a better understanding of the cavernous microenvironment and cell-cell communications under diabetic condition. Pericytes are vital in penile erection; however, their dysfunction due to diabetes remains unclear. In this study, we performed single-cell RNA sequencing to understand the cellular landscape of cavernous tissues and cell type-specific transcriptional changes in diabetic ED. We found a decreased expression of genes associated with collagen or extracellular matrix organization and angiogenesis in diabetic fibroblasts, chondrocytes, myofibroblasts, valve-related lymphatic endothelial cells, and pericytes. Moreover, the newly identified pericyte-specific marker, Limb Bud-Heart (Lbh), in mouse and human cavernous tissues, clearly distinguishing pericytes from smooth muscle cells. Cell-cell interaction analysis revealed that pericytes are involved in angiogenesis, adhesion, and migration by communicating with other cell types in the corpus cavernosum; however, these interactions were highly reduced under diabetic conditions. Lbh expression is low in diabetic pericytes, and overexpression of LBH prevents erectile function by regulating neurovascular regeneration. Furthermore, the LBH-interacting proteins (Crystallin Alpha B and Vimentin) were identified in mouse cavernous pericytes through LC-MS/MS analysis, indicating that their interactions were critical for maintaining pericyte function. Thus, our study reveals novel targets and insights into the pathogenesis of ED in patients with diabetes.


Subject(s)
Erectile Dysfunction , Penis , Pericytes , Single-Cell Analysis , Male , Pericytes/metabolism , Erectile Dysfunction/genetics , Erectile Dysfunction/metabolism , Animals , Mice , Humans , Penis/metabolism , Gene Expression Profiling , Transcriptome , Mice, Inbred C57BL , Single-Cell Gene Expression Analysis
2.
Open Biol ; 14(6): 230349, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38862017

ABSTRACT

Coronavirus disease 2019 (COVID-19) was initially considered a primarily respiratory disease but is now known to affect other organs including the heart and brain. A major route by which COVID-19 impacts different organs is via the vascular system. We studied the impact of apolipoprotein E (APOE) genotype and inflammation on vascular infectivity by pseudo-typed severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viruses in mouse and human cultured endothelial cells and pericytes. Possessing the APOE4 allele or having existing systemic inflammation is known to enhance the severity of COVID-19. Using targeted replacement human APOE3 and APOE4 mice and inflammation induced by bacterial lipopolysaccharide (LPS), we investigated infection by SARS-CoV-2. Here, we show that infectivity was higher in murine cerebrovascular pericytes compared to endothelial cells and higher in cultures expressing APOE4. Furthermore, increasing the inflammatory state of the cells by prior incubation with LPS increased infectivity into human and mouse pericytes and human endothelial cells. Our findings provide insights into the mechanisms underlying severe COVID-19 infection, highlighting how risk factors such as APOE4 genotype and prior inflammation may exacerbate disease severity by augmenting the virus's ability to infect vascular cells.


Subject(s)
COVID-19 , Endothelial Cells , Pericytes , SARS-CoV-2 , Pericytes/virology , Pericytes/metabolism , Pericytes/pathology , Humans , Animals , SARS-CoV-2/physiology , SARS-CoV-2/pathogenicity , COVID-19/virology , COVID-19/pathology , Mice , Endothelial Cells/virology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Risk Factors , Lipopolysaccharides/pharmacology , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Apolipoprotein E3/genetics , Apolipoprotein E3/metabolism , Inflammation/virology , Inflammation/pathology
3.
Commun Biol ; 7(1): 693, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38844781

ABSTRACT

Pericyte dysfunction, with excessive migration, hyperproliferation, and differentiation into smooth muscle-like cells contributes to vascular remodeling in Pulmonary Arterial Hypertension (PAH). Augmented expression and action of growth factors trigger these pathological changes. Endogenous factors opposing such alterations are barely known. Here, we examine whether and how the endothelial hormone C-type natriuretic peptide (CNP), signaling through the cyclic guanosine monophosphate (cGMP) -producing guanylyl cyclase B (GC-B) receptor, attenuates the pericyte dysfunction observed in PAH. The results demonstrate that CNP/GC-B/cGMP signaling is preserved in lung pericytes from patients with PAH and prevents their growth factor-induced proliferation, migration, and transdifferentiation. The anti-proliferative effect of CNP is mediated by cGMP-dependent protein kinase I and inhibition of the Phosphoinositide 3-kinase (PI3K)/AKT pathway, ultimately leading to the nuclear stabilization and activation of the Forkhead Box O 3 (FoxO3) transcription factor. Augmentation of the CNP/GC-B/cGMP/FoxO3 signaling pathway might be a target for novel therapeutics in the field of PAH.


Subject(s)
Cell Proliferation , Cyclic GMP , Forkhead Box Protein O3 , Natriuretic Peptide, C-Type , Pericytes , Signal Transduction , Humans , Pericytes/metabolism , Pericytes/pathology , Natriuretic Peptide, C-Type/metabolism , Cyclic GMP/metabolism , Forkhead Box Protein O3/metabolism , Forkhead Box Protein O3/genetics , Male , Female , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/pathology , Middle Aged , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Adult , Receptors, Atrial Natriuretic Factor/metabolism , Receptors, Atrial Natriuretic Factor/genetics , Cells, Cultured
4.
Sci Prog ; 107(2): 368504241257126, 2024.
Article in English | MEDLINE | ID: mdl-38863331

ABSTRACT

Pericytes (PCs) are versatile cells integral to the microcirculation wall, exhibiting specific stem cell traits. They are essential in modulating blood flow, ensuring vascular permeability, maintaining homeostasis, and aiding tissue repair process. Given their involvement in numerous disease-related pathological and physiological processes, the regulation of PCs has emerged as a focal point of research. Adenomyosis is characterized by the presence of active endometrial glands and stroma encased by an enlarged and proliferative myometrial layer, further accompanied by fibrosis and new blood vessel formation. This distinct pathological condition might be intricately linked with PCs. This article comprehensively reviews the markers associated with PCs, their contributions to angiogenesis, blood flow modulation, and fibrotic processes. Moreover, it provides a comprehensive overview of the current research on adenomyosis pathophysiology, emphasizing the potential correlation and future implications regarding PCs and the development of adenomyosis.


Subject(s)
Adenomyosis , Pericytes , Adenomyosis/pathology , Adenomyosis/physiopathology , Pericytes/pathology , Humans , Female , Neovascularization, Pathologic/pathology , Animals , Fibrosis/pathology , Endometrium/pathology , Endometrium/blood supply , Myometrium/pathology , Biomarkers/metabolism
5.
ACS Nano ; 18(22): 14348-14366, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38768086

ABSTRACT

Pericyte dysfunction severely undermines cerebrovascular integrity and exacerbates neurodegeneration in Alzheimer's disease (AD). However, pericyte-targeted therapy is a yet-untapped frontier for AD. Inspired by the elevation of vascular cell adhesion molecule-1 (VCAM-1) and reactive oxygen species (ROS) levels in pericyte lesions, we fabricated a multifunctional nanoprodrug by conjugating the hybrid peptide VLC, a fusion of the VCAM-1 high-affinity peptide VHS and the neuroprotective apolipoprotein mimetic peptide COG1410, to curcumin (Cur) through phenylboronic ester bond (VLC@Cur-NPs) to alleviate complex pericyte-related pathological changes. Importantly, VLC@Cur-NPs effectively homed to pericyte lesions via VLC and released their contents upon ROS stimulation to maximize their regulatory effects. Consequently, VLC@Cur-NPs markedly increased pericyte regeneration to form a positive feedback loop and thus improved neurovascular function and ultimately alleviated memory defects in APP/PS1 transgenic mice. We present a promising therapeutic strategy for AD that can precisely modulate pericytes and has the potential to treat other cerebrovascular diseases.


Subject(s)
Alzheimer Disease , Mice, Transgenic , Pericytes , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Pericytes/drug effects , Pericytes/metabolism , Pericytes/pathology , Mice , Reactive Oxygen Species/metabolism , Curcumin/pharmacology , Curcumin/chemistry , Prodrugs/pharmacology , Prodrugs/chemistry , Nanoparticles/chemistry , Vascular Cell Adhesion Molecule-1/metabolism , Humans , Peptides/chemistry , Peptides/pharmacology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry
7.
Alzheimer Dis Assoc Disord ; 38(2): 107-111, 2024.
Article in English | MEDLINE | ID: mdl-38752577

ABSTRACT

BACKGROUND: Blood-brain barrier (BBB) dysfunction is emerging as an important pathophysiologic factor in Alzheimer disease (AD). Cerebrospinal fluid (CSF) platelet-derived growth factor receptor-ß (PDGFRß) is a biomarker of BBB pericyte injury and has been implicated in cognitive impairment and AD. METHODS: We aimed to study CSF PDGFRß protein levels, along with CSF biomarkers of brain amyloidosis and tau pathology in a well-characterized population of cognitively unimpaired individuals and correlated CSF findings with amyloid-PET positivity. We performed an institutional review board (IRB)-approved cross-sectional analysis of a prospectively enrolled cohort of 36 cognitively normal volunteers with available CSF, Pittsburgh compound B PET/CT, Mini-Mental State Exam score, Global Deterioration Scale, and known apolipoprotein E ( APOE ) ε4 status. RESULTS: Thirty-six subjects were included. Mean age was 63.3 years; 31 of 36 were female, 6 of 36 were amyloid-PET-positive and 12 of 36 were APOE ε4 carriers. We found a moderate positive correlation between CSF PDGFRß and both total Tau (r=0.45, P =0.006) and phosphorylated Tau 181 (r=0.51, P =0.002). CSF PDGFRß levels were not associated with either the CSF Aß42 or the amyloid-PET. CONCLUSIONS: We demonstrated a moderate positive correlation between PDGFRß and both total Tau and phosphorylated Tau 181 in cognitively normal individuals. Our data support the hypothesis that BBB dysfunction represents an important early pathophysiologic step in AD, warranting larger prospective studies. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT00094939.


Subject(s)
Alzheimer Disease , Biomarkers , Pericytes , tau Proteins , Humans , Female , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/diagnostic imaging , Male , Biomarkers/cerebrospinal fluid , Middle Aged , Cross-Sectional Studies , Aged , tau Proteins/cerebrospinal fluid , Pericytes/pathology , Positron-Emission Tomography , Amyloid beta-Peptides/cerebrospinal fluid , Blood-Brain Barrier , Receptor, Platelet-Derived Growth Factor beta/cerebrospinal fluid , Prospective Studies , Cohort Studies
8.
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
9.
Microbiol Spectr ; 12(6): e0069024, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38752731

ABSTRACT

Enterovirus A71 (EV-A71) is associated with neurological conditions such as acute meningitis and encephalitis. The virus is detected in the bloodstream, and high blood viral loads are associated with central nervous system (CNS) manifestations. We used an in vitro blood-brain barrier (BBB) model made up of human brain-like endothelial cells (hBLECs) and brain pericytes grown in transwell systems to investigate whether three genetically distinct EV-A71 strains (subgenogroups C1, C1-like, and C4) can cross the human BBB. EV-A71 poorly replicated in hBLECs, which released moderate amounts of infectious viruses from their luminal side and trace amounts of infectious viruses from their basolateral side. The barrier properties of hBLECs were not impaired by EV-A71 infection. We investigated the passage through hBLECs of EV-A71-infected white blood cells. EV-A71 strains efficiently replicated in immune cells, including monocytes, neutrophils, and NK/T cells. Attachment to hBLECs of immune cells infected with the C1-like virus was higher than attachment of cells infected with C1-06. EV-A71 infection did not impair the transmigration of immune cells through hBLECs. Overall, EV-A71 targets different white blood cell populations that have the potential to be used as a Trojan horse to cross hBLECs more efficiently than cell-free EV-A71 particles.IMPORTANCEEnterovirus A71 (EV-A71) was first reported in the USA, and numerous outbreaks have since occurred in Asia and Europe. EV-A71 re-emerged as a new multirecombinant strain in 2015 in Europe and is now widespread. The virus causes hand-foot-and-mouth disease in young children and is involved in nervous system infections. How the virus spreads to the nervous system is unclear. We investigated whether white blood cells could be infected by EV-A71 and transmit it across human endothelial cells mimicking the blood-brain barrier protecting the brain from adverse effects. We found that endothelial cells provide a strong roadblock to prevent the passage of free virus particles but allow the migration of infected immune cells, including monocytes, neutrophils, and NK/T cells. Our data are consistent with the potential role of immune cells in the pathogenesis of EV-A71 infections by spreading the virus in the blood and across the human blood-brain barrier.


Subject(s)
Blood-Brain Barrier , Endothelial Cells , Enterovirus A, Human , Enterovirus Infections , Blood-Brain Barrier/virology , Humans , Enterovirus A, Human/genetics , Enterovirus A, Human/physiology , Enterovirus Infections/virology , Enterovirus Infections/immunology , Endothelial Cells/virology , Virus Replication , Monocytes/virology , Monocytes/immunology , Pericytes/virology , Leukocytes/virology , Leukocytes/immunology , Brain/virology , Killer Cells, Natural/immunology , Neutrophils/immunology , Neutrophils/virology
10.
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
11.
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
12.
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
13.
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
14.
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
15.
J Med Virol ; 96(5): e29671, 2024 May.
Article in English | MEDLINE | ID: mdl-38747003

ABSTRACT

The coronavirus disease of 2019 (COVID-19) pandemic has led to more than 700 million confirmed cases and nearly 7 million deaths. Although severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) virus mainly infects the respiratory system, neurological complications are widely reported in both acute infection and long-COVID cases. Despite the success of vaccines and antiviral treatments, neuroinvasiveness of SARS-CoV-2 remains an important question, which is also centered on the mystery of whether the virus is capable of breaching the barriers into the central nervous system. By studying the K18-hACE2 infection model, we observed clear evidence of microvascular damage and breakdown of the blood-brain barrier (BBB). Mechanistically, SARS-CoV-2 infection caused pericyte damage, tight junction loss, endothelial activation and vascular inflammation, which together drive microvascular injury and BBB impairment. In addition, the blood-cerebrospinal fluid barrier at the choroid plexus was also impaired after infection. Therefore, cerebrovascular and choroid plexus dysfunctions are important aspects of COVID-19 and may contribute to neurological complications both acutely and in long COVID.


Subject(s)
Blood-Brain Barrier , COVID-19 , Choroid Plexus , SARS-CoV-2 , Blood-Brain Barrier/virology , Animals , Choroid Plexus/virology , Choroid Plexus/pathology , COVID-19/virology , COVID-19/pathology , COVID-19/complications , COVID-19/physiopathology , Mice , Tight Junctions/virology , Disease Models, Animal , Angiotensin-Converting Enzyme 2/metabolism , Inflammation/virology , Humans , Pericytes/virology , Pericytes/pathology
16.
NanoImpact ; 34: 100508, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38663501

ABSTRACT

The objective of this investigation was to evaluate the influence of micro- and nanoplastic particles composed of polyethylene terephthalate (PET), a significant contributor to plastic pollution, on human brain vascular pericytes. Specifically, we delved into their impact on mitochondrial functionality, oxidative stress, and the expression of genes associated with oxidative stress, ferroptosis and mitochondrial functions. Our findings demonstrate that the exposure of a monoculture of human brain vascular pericytes to PET particles in vitro at a concentration of 50 µg/ml for a duration of 3, 6 and 10 days did not elicit oxidative stress. Notably, we observed a reduction in various aspects of mitochondrial respiration, including maximal respiration, spare respiratory capacity, and ATP production in pericytes subjected to PET particles for 3 days, with a mitochondrial function recovery at 6 and 10 days. Furthermore, there were no statistically significant alterations in mitochondrial DNA copy number, or in the expression of genes linked to oxidative stress and ferroptosis, but an increase of the expression of the gene mitochondrial transcription factor A (TFAM) was noted at 3 days exposure. These outcomes suggest that, at a concentration of 50 µg/ml, PET particles do not induce oxidative stress in human brain vascular pericytes. Instead, at 3 days exposure, PET exposure impairs mitochondrial functions, but this is recovered at 6-day exposure. This seems to indicate a potential mitochondrial hormesis response (mitohormesis) is incited, involving the gene TFAM. Further investigations are warranted to explore the stages of mitohormesis and the potential consequences of plastics on the integrity of the blood-brain barrier and intercellular interactions. This research contributes to our comprehension of the potential repercussions of nanoplastic pollution on human health and underscores the imperative need for ongoing examinations into the exposure to plastic particles.


Subject(s)
Brain , Mitochondria , Oxidative Stress , Pericytes , Polyethylene Terephthalates , Humans , Oxidative Stress/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Pericytes/drug effects , Pericytes/metabolism , Brain/metabolism , Brain/blood supply , Brain/drug effects , Nanoparticles , Microplastics/toxicity , Cells, Cultured
17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...