Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.720
Filtrer
1.
PLoS One ; 19(8): e0308132, 2024.
Article de Anglais | MEDLINE | ID: mdl-39106252

RÉSUMÉ

To investigate the sex-dependent differentiation of Sox10 cells and their response to pathological conditions such as lipopolysaccharide (LPS) exposure or ischemia, we utilized Sox10 Cre-ERT2, tdTomato mice. Tamoxifen administration induced the expression of red fluorescent protein (RFP) in these cells, facilitating their subsequent tracking and analysis after LPS injection and ischemia via immunofluorescence staining. Propidium iodide (PI) was injected to label necrotic cells following LPS administration. We found that the conversion of Sox10 cells to pericytes in female mice was significantly higher than in male mice, especially in those exposed to LPS. After LPS injection, the number of PI+ necrotic cells were significantly greater in females than in males. Moreover, RFP+ cells did not co-localize with glial fibrillary acidic protein (GFAP) or cluster of differentiation 11b (CD11b). Similarly, after brain ischemia, RFP+ cells did not express cluster of differentiation 13 (CD13), neuronal nuclei (NeuN), GFAP, or ionised calcium binding adaptor molecule 1 (Iba-1). These findings indicate that the conversion of Sox10 cells to pericytes following LPS exposure is sex-dependent, with neither male nor female groups showing differentiation into other cell types after LPS exposure or under ischemic conditions. The differences in LPS-induced necrosis of pericytes between sexes may explain the variations in the conversion of Sox10 cells to pericytes in both sexes.


Sujet(s)
Lipopolysaccharides , Oligodendroglie , Péricytes , Facteurs de transcription SOX-E , Animaux , Femelle , Mâle , Péricytes/métabolisme , Péricytes/effets des médicaments et des substances chimiques , Souris , Facteurs de transcription SOX-E/métabolisme , Oligodendroglie/métabolisme , Oligodendroglie/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Caractères sexuels , Facteurs sexuels , Souris transgéniques
2.
Redox Rep ; 29(1): 2382943, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39092597

RÉSUMÉ

OBJECTIVES: Diabetes is closely linked to hearing loss, yet the exact mechanisms remain unclear. Cochlear stria vascularis and pericytes (PCs) are crucial for hearing. This study investigates whether high glucose induces apoptosis in the cochlear stria vascularis and pericytes via elevated ROS levels due to oxidative stress, impacting hearing loss. METHODS: We established a type II diabetes model in C57BL/6J mice and used auditory brainstem response (ABR), Evans blue staining, HE staining, immunohistochemistry, and immunofluorescence to observe changes in hearing, blood-labyrinth barrier (BLB) permeability, stria vascularis morphology, and apoptosis protein expression. Primary cultured stria vascularis pericytes were subjected to high glucose, and apoptosis levels were assessed using flow cytometry, Annexin V-FITC, Hoechst 33342 staining, Western blot, Mitosox, and JC-1 probes. RESULTS: Diabetic mice showed decreased hearing thresholds, reduced stria vascularis density, increased oxidative stress, cell apoptosis, and decreased antioxidant levels. High glucose exposure increased apoptosis and ROS content in pericytes, while mitochondrial membrane potential decreased, with AIF and cytochrome C (CytC) released from mitochondria to the cytoplasm. Adding oxidative scavengers reduced AIF and CytC release, decreasing pericyte apoptosis. DISCUSSION: Hyperglycemia may induce mitochondrial apoptosis of cochlear stria vascularis pericytes through oxidative stress.


Sujet(s)
Facteur inducteur d'apoptose , Apoptose , Cytochromes c , Hyperglycémie , Souris de lignée C57BL , Mitochondries , Stress oxydatif , Péricytes , Protéines proto-oncogènes c-bcl-2 , Espèces réactives de l'oxygène , Strie vasculaire , Animaux , Péricytes/métabolisme , Péricytes/effets des médicaments et des substances chimiques , Péricytes/anatomopathologie , Strie vasculaire/métabolisme , Strie vasculaire/anatomopathologie , Souris , Espèces réactives de l'oxygène/métabolisme , Mitochondries/métabolisme , Cytochromes c/métabolisme , Facteur inducteur d'apoptose/métabolisme , Hyperglycémie/métabolisme , Protéines proto-oncogènes c-bcl-2/métabolisme , Mâle , Diabète expérimental/métabolisme , Diabète expérimental/anatomopathologie , Cochlée/métabolisme , Cochlée/anatomopathologie
3.
Neuron ; 112(13): 2081-2083, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38964282

RÉSUMÉ

Preterm infants can face lasting neurodevelopmental challenges due to hypoxia-induced injury of the cerebral white matter. In this issue of Neuron, Ren et al.1 identify microvascular pericytes as unexpected targets for growth hormone signaling, which enhances angiogenesis and remyelination after hypoxic injury in the developing mouse brain.


Sujet(s)
Hypoxie cérébrale , Gaine de myéline , Péricytes , Péricytes/métabolisme , Gaine de myéline/métabolisme , Animaux , Hypoxie cérébrale/métabolisme , Souris , Humains , Animaux nouveau-nés , Encéphale/métabolisme
4.
Nat Commun ; 15(1): 6321, 2024 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-39060269

RÉSUMÉ

Spinal cord injury (SCI) leads to fibrotic scar formation at the lesion site, yet the heterogeneity of fibrotic scar remains elusive. Here we show the heterogeneity in distribution, origin, and function of fibroblasts within fibrotic scars after SCI in mice and female monkeys. Utilizing lineage tracing and single-cell RNA sequencing (scRNA-seq), we found that perivascular fibroblasts (PFs), and meningeal fibroblasts (MFs), rather than pericytes/vascular smooth cells (vSMCs), primarily contribute to fibrotic scar in both transection and crush SCI. Crabp2 + /Emb+ fibroblasts (CE-F) derived from meninges primarily localize in the central region of fibrotic scars, demonstrating enhanced cholesterol synthesis and secretion of type I collagen and fibronectin. In contrast, perivascular/pial Lama1 + /Lama2+ fibroblasts (LA-F) are predominantly found at the periphery of the lesion, expressing laminin and type IV collagen and functionally involved in angiogenesis and lipid transport. These findings may provide a comprehensive understanding for remodeling heterogeneous fibrotic scars after SCI.


Sujet(s)
Cicatrice , Fibroblastes , Fibrose , Laminine , Traumatismes de la moelle épinière , Animaux , Traumatismes de la moelle épinière/anatomopathologie , Traumatismes de la moelle épinière/métabolisme , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Cicatrice/anatomopathologie , Cicatrice/métabolisme , Souris , Femelle , Laminine/métabolisme , Méninges/anatomopathologie , Méninges/métabolisme , Fibronectines/métabolisme , Modèles animaux de maladie humaine , Collagène de type I/métabolisme , Souris de lignée C57BL , Péricytes/métabolisme , Péricytes/anatomopathologie , Collagène de type IV/métabolisme , Cholestérol/métabolisme
5.
Int J Mol Sci ; 25(14)2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39063134

RÉSUMÉ

Tick-borne encephalitis virus (TBEV) targets the central nervous system (CNS), leading to potentially severe neurological complications. The neurovascular unit plays a fundamental role in the CNS and in the neuroinvasion of TBEV. However, the role of human brain pericytes, a key component of the neurovascular unit, during TBEV infection has not yet been elucidated. In this study, TBEV infection of the primary human brain perivascular pericytes was investigated with highly virulent Hypr strain and mildly virulent Neudoerfl strain. We used Luminex assay to measure cytokines/chemokines and growth factors. Both viral strains showed comparable replication kinetics, peaking at 3 days post infection (dpi). Intracellular viral RNA copies peaked at 6 dpi for Hypr and 3 dpi for Neudoerfl cultures. According to immunofluorescence staining, only small proportion of pericytes were infected (3% for Hypr and 2% for Neudoerfl), and no cytopathic effect was observed in the infected cells. In cell culture supernatants, IL-6 production was detected at 3 dpi, together with slight increases in IL-15 and IL-4, but IP-10, RANTES and MCP-1 were the main chemokines released after TBEV infection. These chemokines play key roles in both immune defense and immunopathology during TBE. This study suggests that pericytes are an important source of these signaling molecules during TBEV infection in the brain.


Sujet(s)
Encéphale , Chimiokine CCL5 , Chimiokine CXCL10 , Virus de l'encéphalite à tiques (sous-groupe) , Encéphalites à tiques , Péricytes , Péricytes/virologie , Péricytes/métabolisme , Humains , Virus de l'encéphalite à tiques (sous-groupe)/physiologie , Virus de l'encéphalite à tiques (sous-groupe)/pathogénicité , Encéphale/virologie , Encéphale/métabolisme , Encéphale/anatomopathologie , Chimiokine CXCL10/métabolisme , Encéphalites à tiques/virologie , Encéphalites à tiques/métabolisme , Chimiokine CCL5/métabolisme , Cellules cultivées , Réplication virale , Cytokines/métabolisme
6.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-39000224

RÉSUMÉ

Pericytes are a distinct type of cells interacting with endothelial cells in blood vessels and contributing to endothelial barrier integrity. Furthermore, pericytes show mesenchymal stem cell properties. Muscle-derived pericytes can demonstrate both angiogenic and myogenic capabilities. It is well known that regenerative abilities and muscle stem cell potential decline during aging, leading to sarcopenia. Therefore, this study aimed to investigate the potential of pericytes in supporting muscle differentiation and angiogenesis in elderly individuals and in patients affected by Ullrich congenital muscular dystrophy or by Bethlem myopathy, two inherited conditions caused by mutations in collagen VI genes and sharing similarities with the progressive skeletal muscle changes observed during aging. The study characterized pericytes from different age groups and from individuals with collagen VI deficiency by mass spectrometry-based proteomic and bioinformatic analyses. The findings revealed that aged pericytes display metabolic changes comparable to those seen in aging skeletal muscle, as well as a decline in their stem potential, reduced protein synthesis, and alterations in focal adhesion and contractility, pointing to a decrease in their ability to form blood vessels. Strikingly, pericytes from young patients with collagen VI deficiency showed similar characteristics to aged pericytes, but were found to still handle oxidative stress effectively together with an enhanced angiogenic capacity.


Sujet(s)
Collagène de type VI , Péricytes , Protéome , Humains , Péricytes/métabolisme , Collagène de type VI/métabolisme , Collagène de type VI/génétique , Protéome/métabolisme , Cellules cultivées , Adulte , Adulte d'âge moyen , Sujet âgé , Vieillissement/métabolisme , Protéomique/méthodes , Mâle , Femelle , Stress oxydatif , Différenciation cellulaire
7.
J Cell Mol Med ; 28(13): e18505, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39001579

RÉSUMÉ

Hypoxia-ischaemia (HI) can induce the death of cerebrovascular constituent cells through oxidative stress. Hydrogen is a powerful antioxidant which can activate the antioxidant system. A hypoxia-ischaemia brain damage (HIBD) model was established in 7-day-old SD rats. Rats were treated with different doses of hydrogen-rich water (HRW), and brain pericyte oxidative stress damage, cerebrovascular function and brain tissue damage were assessed. Meanwhile, in vitro-cultured pericytes were subjected to oxygen-glucose deprivation and treated with different concentrations of HRW. Oxidative injury was measured and the molecular mechanism of how HRW alleviated oxidative injury of pericytes was also examined. The results showed that HRW significantly attenuated HI-induced oxidative stress in the brain pericytes of neonatal rats, partly through the Nrf2-HO-1 pathway, further improving cerebrovascular function and reducing brain injury and dysfunction. Furthermore, HRW is superior to a single-cell death inhibitor for apoptosis, ferroptosis, parthanatos, necroptosis and autophagy and can better inhibit HI-induced pericyte death. The liver and kidney functions of rats were not affected by present used HRW dose. This study elucidates the role and mechanism of hydrogen in treating HIBD from the perspective of pericytes, providing new theoretical evidence and mechanistic references for the clinical application of hydrogen in neonatal HIE.


Sujet(s)
Animaux nouveau-nés , Encéphale , Hydrogène , Hypoxie-ischémie du cerveau , Stress oxydatif , Péricytes , Rat Sprague-Dawley , Animaux , Péricytes/effets des médicaments et des substances chimiques , Péricytes/métabolisme , Hydrogène/pharmacologie , Hypoxie-ischémie du cerveau/anatomopathologie , Hypoxie-ischémie du cerveau/métabolisme , Hypoxie-ischémie du cerveau/traitement médicamenteux , Rats , Stress oxydatif/effets des médicaments et des substances chimiques , Encéphale/anatomopathologie , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Facteur-2 apparenté à NF-E2/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Antioxydants/pharmacologie
8.
Zhonghua Xin Xue Guan Bing Za Zhi ; 52(7): 814-826, 2024 Jul 24.
Article de Chinois | MEDLINE | ID: mdl-39019831

RÉSUMÉ

Objective: To investigate the role and underlying mechanisms of methyltransferase (Mettl) 3 in the process of angiotensin Ⅱ (Ang Ⅱ)-induced pericyte-to-myofibroblast transdifferentiation and renal fibrosis. Methods: C57BL/6J mice were used, in cell experiments, mouse renal pericytes were isolated and cultured using magnetic bead sorting. These pericytes were then induced to transdifferentiate into myofibroblasts with 1×106 mmol/L Ang Ⅱ, which was the Ang Ⅱ group, while pericytes cultured in normal conditions served as the control group. Successful transdifferentiation was verified by immunofluorescence staining, Western blotting, and real-time reverse transcription PCR (RT-qPCR) for α-smooth muscle actin (α-SMA). The levels of m6A modifications and related enzymes (Mettl3, Mettl14), Wilms tumor 1-associated protein (WTAP), fat mass and obesity protein (FTO), ALKBH5, YTHDF1, YTHDF2, YTHDC1, YTHDC2, YTHDC3 were assessed by Dot blot, RT-qPCR and Western blot. Mettl3 expression was inhibited in cells using lentivirus-mediated Mettl3-shRNA transfection, creating sh-Mettl3 and Ang Ⅱ+sh-Mettl3 groups, while lentivirus empty vector transfection served as the negative control (Ang Ⅱ+sh-NC group). The impact of Ang Ⅱ on pericyte transdifferentiation was observed, and the expression of downstream phosphatidylinositol 3-kinase (PI3K)/AKT signaling pathway proteins, including PI3K, AKT, phosphorylated AKT at serine 473 (p-AKT (S473)), and phosphorylated AKT at threonine 308 (p-AKT (T308)), were examined. PI3K gene transcription was inhibited by co-culturing cells with actinomycin D, and the half-life of PI3K mRNA was calculated by measuring residual PI3K mRNA expression over different co-culture time. The reversibility of Mettl3 inhibition on Ang Ⅱ-induced pericyte-to-myofibroblast transdifferentiation was assessed by adding the AKT activator SC79 to the Ang Ⅱ+sh-Mettl3 group. In animal experiments, mice were divided into these groups: sham group (administered 0.9% sterile saline), Ang Ⅱ group (infused with Ang Ⅱ solution), sh-Mettl3 group (injected with Mettl3 shRNA lentivirus solution), Ang Ⅱ+sh-Mettl3 group (infused with Ang Ⅱ solution and injected with Mettl3 shRNA lentivirus solution), and Ang Ⅱ+sh-Mettl3+SC79 group (administered Ang Ⅱ solution and Mettl3 shRNA lentivirus, with an additional injection of SC79). Each group consisted of six subject mice. Blood pressure was measured using the tail-cuff method before and after surgery, and serum creatinine, urea, and urinary albumin levels were determined 4 weeks post-surgery. Kidney tissues were collected at 28 days and stained using hematoxylin-eosin (HE) and Masson's trichrome to assess the extent of renal fibrosis. Results: Primary renal pericytes were successfully obtained by magnetic bead sorting, and intervened with 1×106 mmol/L Ang Ⅱ for 48 hours to induce pericyte-to-myofibroblast transdifferentiation. Dot blot results indicated higher m6A modification levels in the Ang Ⅱ group compared to the control group (P<0.05). RT-qPCR and Western blot results showed upregulation of Mettl3 mRNA and protein levels in the Ang Ⅱ group compared to the control group (both P<0.05). In the Ang Ⅱ+sh-Mettl3 group, Mettl3 protein expression was lower than that in the Ang Ⅱ group, with reduced expression levels of α-SMA, vimentin, desmin, fibroblast agonist protein (FAPa) and type Ⅰ collagen (all P<0.05). Compared to the control group, PI3K mRNA expression level was elevated in the Ang Ⅱ group, along with increased p-AKT (S473) and p-AKT (T308) expressions. In the Ang Ⅱ+sh-Mettl3 group, PI3K mRNA expression and p-AKT (S473) and p-AKT (T308) levels were decreased (all P<0.05). The half-life of PI3K mRNA was shorter in the Ang Ⅱ+sh-Mettl3 group than that in the Ang Ⅱ+sh-NC group (2.34 h vs. 3.42 h). The ameliorative effect of Mettl3 inhibition on Ang Ⅱ-induced pericyte-to-myofibroblast transdifferentiation was reversible by SC79. Animal experiments showed higher blood pressure, serum creatinine, urea, and 24-hour urinary protein levels, and a larger fibrosis area in the Ang Ⅱ group compared to the sham group (all P<0.05). The fibrosis area was smaller in the Ang Ⅱ+sh-Mettl3 group than that in the Ang Ⅱ group (P<0.05), but increased again upon addition of SC79. Conclusion: Mettl3-mediated RNA m6A epigenetic regulation is involved in Ang Ⅱ-induced pericyte-to-myofibroblast transdifferentiation and renal fibrosis, potentially by affecting PI3K stability and regulating the PI3K/AKT signaling pathway.


Sujet(s)
Angiotensine-II , Transdifférenciation cellulaire , Methyltransferases , Souris de lignée C57BL , Myofibroblastes , Péricytes , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Animaux , Péricytes/métabolisme , Methyltransferases/métabolisme , Souris , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Angiotensine-II/pharmacologie , Myofibroblastes/métabolisme , Rein , Cellules cultivées
9.
J Transl Med ; 22(1): 712, 2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-39085929

RÉSUMÉ

BACKGROUND: Excessive pericyte coverage promotes tumor growth, and a downregulation may solve this dilemma. Due to the double-edged sword role of vascular pericytes in tumor microenvironment (TME), indiscriminately decreasing pericyte coverage by imatinib causes poor treatment outcomes. Here, we optimized the use of imatinib in a colorectal cancer (CRC) model in high pericyte-coverage status, and revealed the value of multiparametric magnetic resonance imaging (mpMRI) at 9.4T in monitoring treatment-related changes in pericyte coverage and the TME. METHODS: CRC xenograft models were evaluated by histological vascular characterizations and mpMRI. Mice with the highest pericyte coverage were treated with imatinib or saline; then, vascular characterizations, tumor apoptosis and HIF-1α level were analyzed histologically, and alterations in the expression of Bcl-2/bax pathway were assessed through qPCR. The effects of imatinib were monitored by dynamic contrast-enhanced (DCE)-, diffusion-weighted imaging (DWI)- and amide proton transfer chemical exchange saturation transfer (APT CEST)-MRI at 9.4T. RESULTS: The DCE- parameters provided a good histologic match the tumor vascular characterizations. In the high pericyte coverage status, imatinib exhibited significant tumor growth inhibition, necrosis increase and pericyte coverage downregulation, and these changes were accompanied by increased vessel permeability, decreased microvessel density (MVD), increased tumor apoptosis and altered gene expression of apoptosis-related Bcl-2/bax pathway. Strategically, a 4-day imatinib effectively decreased pericyte coverage and HIF-1α level, and continuous treatment led to a less marked decrease in pericyte coverage and re-elevated HIF-1α level. Correlation analysis confirmed the feasibility of using mpMRI parameters to monitor imatinib treatment, with DCE-derived Ve and Ktrans being most correlated with pericyte coverage, Ve with vessel permeability, AUC with microvessel density (MVD), DWI-derived ADC with tumor apoptosis, and APT CEST-derived MTRasym at 1 µT with HIF-1α. CONCLUSIONS: These results provided an optimized imatinib regimen to achieve decreasing pericyte coverage and HIF-1α level in the high pericyte-coverage CRC model, and offered an ultrahigh-field multiparametric MRI approach for monitoring pericyte coverage and dynamics response of the TME to treatment.


Sujet(s)
Apoptose , Tumeurs colorectales , Sous-unité alpha du facteur-1 induit par l'hypoxie , Mésilate d'imatinib , Imagerie par résonance magnétique multiparamétrique , Péricytes , Mésilate d'imatinib/pharmacologie , Mésilate d'imatinib/usage thérapeutique , Animaux , Péricytes/métabolisme , Péricytes/effets des médicaments et des substances chimiques , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/imagerie diagnostique , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Lignée cellulaire tumorale , Apoptose/effets des médicaments et des substances chimiques , Humains , Souris nude , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Souris , Souris de lignée BALB C , Tests d'activité antitumorale sur modèle de xénogreffe
10.
J Exp Clin Cancer Res ; 43(1): 210, 2024 Jul 29.
Article de Anglais | MEDLINE | ID: mdl-39075504

RÉSUMÉ

BACKGROUND: It has been proposed that anti-angiogenesis therapy could induce tumor "vascular normalization" and further enhance the efficacy of chemotherapy, radiotherapy, target therapy, and immunotherapy for nearly twenty years. However, the detailed molecular mechanism of this phenomenon is still obscure. METHOD: Overexpression and knockout of CCL28 in human lung adenocarcinoma cell line A549 and murine lung adenocarcinoma cell line LLC, respectively, were utilized to establish mouse models. Single-cell sequencing was performed to analyze the proportion of different cell clusters and metabolic changes in the tumor microenvironment (TME). Immunofluorescence and multiplex immunohistochemistry were conducted in murine tumor tissues and clinical biopsy samples to assess the percentage of pericytes coverage. Primary pericytes were isolated from lung adenocarcinoma tumor tissues using magnetic-activated cell sorting (MACS). These pericytes were then treated with recombinant human CCL28 protein, followed by transwell migration assays and RNA sequencing analysis. Changes in the secretome and metabolome were examined, and verification of retinoic acid metabolism alterations in pericytes was conducted using quantitative real-time PCR, western blotting, and LC-MS technology. Chromatin immunoprecipitation followed by quantitative PCR (ChIP-qPCR) was employed to validate the transcriptional regulatory ability and affinity of RXRα to specific sites at the ANGPT1 promoter. RESULTS: Our study showed that after undergoing anti-angiogenesis treatment, the tumor exhibited a state of ischemia and hypoxia, leading to an upregulation in the expression of CCL28 in hypoxic lung adenocarcinoma cells by the hypoxia-sensitive transcription factor CEBPB. Increased CCL28 could promote tumor vascular normalization through recruiting and metabolic reprogramming pericytes in the tumor microenvironment. Mechanistically, CCL28 modified the retinoic acid (RA) metabolism and increased ANGPT1 expression via RXRα in pericytes, thereby enhancing the stability of endothelial cells. CONCLUSION: We reported the details of the molecular mechanisms of "vascular normalization" after anti-angiogenesis therapy for the first time. Our work might provide a prospective molecular marker for guiding the clinical arrangement of combination therapy between anti-angiogenesis treatment and other therapies.


Sujet(s)
Adénocarcinome pulmonaire , Angiopoïétine-1 , Chimiokines CC , Tumeurs du poumon , Péricytes , Péricytes/métabolisme , Souris , Humains , Animaux , Angiopoïétine-1/métabolisme , Angiopoïétine-1/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Adénocarcinome pulmonaire/métabolisme , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/traitement médicamenteux , Chimiokines CC/métabolisme , Chimiokines CC/génétique , Inhibiteurs de l'angiogenèse/pharmacologie , Inhibiteurs de l'angiogenèse/usage thérapeutique , Microenvironnement tumoral , Néovascularisation pathologique/métabolisme , Lignée cellulaire tumorale
11.
Nat Commun ; 15(1): 4758, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38902234

RÉSUMÉ

To uncover molecular changes underlying blood-brain-barrier dysfunction in Alzheimer's disease, we performed single nucleus RNA sequencing in 24 Alzheimer's disease and control brains and focused on vascular and astrocyte clusters as main cell types of blood-brain-barrier gliovascular-unit. The majority of the vascular transcriptional changes were in pericytes. Of the vascular molecular targets predicted to interact with astrocytic ligands, SMAD3, upregulated in Alzheimer's disease pericytes, has the highest number of ligands including VEGFA, downregulated in Alzheimer's disease astrocytes. We validated these findings with external datasets comprising 4,730 pericyte and 150,664 astrocyte nuclei. Blood SMAD3 levels are associated with Alzheimer's disease-related neuroimaging outcomes. We determined inverse relationships between pericytic SMAD3 and astrocytic VEGFA in human iPSC and zebrafish models. Here, we detect vast transcriptome changes in Alzheimer's disease at the gliovascular-unit, prioritize perturbed pericytic SMAD3-astrocytic VEGFA interactions, and validate these in cross-species models to provide a molecular mechanism of blood-brain-barrier disintegrity in Alzheimer's disease.


Sujet(s)
Maladie d'Alzheimer , Astrocytes , Barrière hémato-encéphalique , Péricytes , Protéine Smad-3 , Facteur de croissance endothéliale vasculaire de type A , Danio zébré , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Humains , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/anatomopathologie , Protéine Smad-3/métabolisme , Protéine Smad-3/génétique , Astrocytes/métabolisme , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Animaux , Péricytes/métabolisme , Péricytes/anatomopathologie , Mâle , Cellules souches pluripotentes induites/métabolisme , Femelle , Sujet âgé , Transcriptome , Encéphale/métabolisme , Encéphale/anatomopathologie , Encéphale/vascularisation , Sujet âgé de 80 ans ou plus , Modèles animaux de maladie humaine
12.
Commun Biol ; 7(1): 693, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38844781

RÉSUMÉ

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.


Sujet(s)
Prolifération cellulaire , GMP cyclique , Protéine O3 à motif en tête de fourche , Peptide natriurétique de type C , Péricytes , Transduction du signal , Humains , Péricytes/métabolisme , Péricytes/anatomopathologie , Peptide natriurétique de type C/métabolisme , GMP cyclique/métabolisme , Protéine O3 à motif en tête de fourche/métabolisme , Protéine O3 à motif en tête de fourche/génétique , Mâle , Femelle , Hypertension artérielle pulmonaire/métabolisme , Hypertension artérielle pulmonaire/anatomopathologie , Adulte d'âge moyen , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/anatomopathologie , Adulte , Récepteur facteur natriurétique auriculaire/métabolisme , Récepteur facteur natriurétique auriculaire/génétique , Cellules cultivées
13.
Int J Mol Sci ; 25(12)2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38928202

RÉSUMÉ

Blood-brain barrier (BBB) dysfunction is a key feature in neuroimmunological and neurodegenerative diseases. In this study, we developed a microfluidic human BBB-on-a-chip to model barrier dysfunction and immune cell migration using immortalized TY10 brain endothelial cells, pericytes, and astrocytes. It was found that immortalized TY10 brain endothelial cells developed a microvascular structure under flow. Pericytes were localized on the basal side surrounding the TY10 microvascular structure, showing an in vivo-like structure. Barrier integrity increased under co-culture with pericytes. In addition, both ethylenediaminetetraacetic acid (EDTA) and anti-Claudin-5 (CLDN5) neutralizing antibody caused a decrease in the transendothelial electrical resistance (TEER). EDTA caused the leakage of 20 kDa dextran, suggesting different effects on the BBB based on the mechanism of action, whereas anti-CLDN5 antibody did not cause leakage. In the tri-culture model, human T cells migrated through endothelial vessels towards basal C-X-C motif chemokine ligand 12 (CXCL12). The live-imaging analysis confirmed the extravasation of fluorescence-labelled T cells in a CXCL12-concentration- and time-dependent manner. Our BBB model had an in vivo-like structure and successfully represented barrier dysfunction and transendothelial T cell migration. In addition, our study suggests that the inhibition of CLDN5 attenuates the BBB in humans. This platform has various potential uses in relation to the BBB in both drug discovery research and in elucidating the mechanisms of central nervous system diseases.


Sujet(s)
Barrière hémato-encéphalique , Mouvement cellulaire , Cellules endothéliales , Laboratoires sur puces , Humains , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Découverte de médicament/méthodes , Techniques de coculture , Péricytes/métabolisme , Péricytes/effets des médicaments et des substances chimiques , Claudine-5/métabolisme , Astrocytes/métabolisme , Astrocytes/effets des médicaments et des substances chimiques , Chimiokine CXCL12/métabolisme , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Lymphocytes T/effets des médicaments et des substances chimiques
14.
Int J Mol Sci ; 25(12)2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38928298

RÉSUMÉ

Pericytes are multipotent cells embedded within the vascular system, primarily surrounding capillaries and microvessels where they closely interact with endothelial cells. These cells are known for their intriguing properties due to their heterogeneity in tissue distribution, origin, and multifunctional capabilities. Specifically, pericytes are essential in regulating blood flow, promoting angiogenesis, and supporting tissue homeostasis and regeneration. These multifaceted roles draw on pericytes' remarkable ability to respond to biochemical cues, interact with neighboring cells, and adapt to changing environmental conditions. This review aims to summarize existing knowledge on pericytes, emphasizing their versatility and involvement in vascular integrity and tissue health. In particular, a comprehensive view of the major signaling pathways, such as PDGFß/ PDGFRß, TGF-ß, FOXO and VEGF, along with their downstream targets, which coordinate the behavior of pericytes in preserving vascular integrity and promoting tissue regeneration, will be discussed. In this light, a deeper understanding of the complex signaling networks defining the phenotype of pericytes in healthy tissues is crucial for the development of targeted therapies in vascular and degenerative diseases.


Sujet(s)
Homéostasie , Péricytes , Transduction du signal , Péricytes/métabolisme , Péricytes/physiologie , Humains , Animaux , Néovascularisation physiologique , Récepteur au PDGF bêta/métabolisme
15.
Exp Neurol ; 379: 114864, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38866101

RÉSUMÉ

Platelet-derived growth factor receptor ß positive (PDGFRß+) pericytes detach from the microvascular wall and migrate into the injury center following spinal cord injury (SCI), which has been widely regarded as the main source of fibrotic scar, but the mechanism of migration and fibroblast transition remains elusive. Here we show the associated spatiotemporal distribution between microglia and pericytes at three and seven days post-injury (dpi). The increased expression of Sphingosine kinase-1 (SPHK1) in microglia significantly raised the concentration of Sphingosine-1-phosphate (S1P) in the spinal cord, which promotes migration and fibroblast transition of pericyte. In vitro experiments, we found the elevated Sphingosine 1-phosphate receptor 3 (S1P3), the S1P/S1PR3 axis inhibited the phosphorylation of YAP and promoted its nuclear translocation, which contributed to the formation of alpha-smooth muscle actin (α-SMA) and collagen type I (COL1) protein, This process can be blocked by an S1P3 specific inhibitor TY52156 in vitro. The S1P/S1P3/YAP pathway might be a potential target for treatment in SCI.


Sujet(s)
Mouvement cellulaire , Fibroblastes , Lysophospholipides , Microglie , Péricytes , Transduction du signal , Récepteurs de la sphingosine-1-phosphate , Sphingosine , Traumatismes de la moelle épinière , Protéines de signalisation YAP , Traumatismes de la moelle épinière/métabolisme , Traumatismes de la moelle épinière/anatomopathologie , Transduction du signal/physiologie , Lysophospholipides/métabolisme , Animaux , Mouvement cellulaire/physiologie , Péricytes/métabolisme , Sphingosine/analogues et dérivés , Sphingosine/métabolisme , Microglie/métabolisme , Protéines de signalisation YAP/métabolisme , Rats , Fibroblastes/métabolisme , Récepteurs de la sphingosine-1-phosphate/métabolisme , Rat Sprague-Dawley , Protéines adaptatrices de la transduction du signal/métabolisme , Récepteurs aux lysosphingolipides/métabolisme , Cellules cultivées
16.
Tissue Cell ; 89: 102431, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38870572

RÉSUMÉ

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.


Sujet(s)
Péricytes , Humains , Péricytes/cytologie , Péricytes/métabolisme , Tubuline/métabolisme , Microtubules/métabolisme , Derme/cytologie , Derme/métabolisme , Communication cellulaire , Mitochondries/métabolisme , Actines/métabolisme , Nanotubes/composition chimique , Microvaisseaux/cytologie , Microvaisseaux/métabolisme , Cellules cultivées , Structures de la membrane cellulaire
17.
Nat Neurosci ; 27(7): 1285-1298, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38849523

RÉSUMÉ

Fibrotic scar tissue formation occurs in humans and mice. The fibrotic scar impairs tissue regeneration and functional recovery. However, the origin of scar-forming fibroblasts is unclear. Here, we show that stromal fibroblasts forming the fibrotic scar derive from two populations of perivascular cells after spinal cord injury (SCI) in adult mice of both sexes. We anatomically and transcriptionally identify the two cell populations as pericytes and perivascular fibroblasts. Fibroblasts and pericytes are enriched in the white and gray matter regions of the spinal cord, respectively. Both cell populations are recruited in response to SCI and inflammation. However, their contribution to fibrotic scar tissue depends on the location of the lesion. Upon injury, pericytes and perivascular fibroblasts become activated and transcriptionally converge on the generation of stromal myofibroblasts. Our results show that pericytes and perivascular fibroblasts contribute to the fibrotic scar in a region-dependent manner.


Sujet(s)
Cicatrice , Fibroblastes , Fibrose , Péricytes , Traumatismes de la moelle épinière , Animaux , Fibroblastes/anatomopathologie , Fibroblastes/métabolisme , Fibrose/anatomopathologie , Traumatismes de la moelle épinière/anatomopathologie , Souris , Péricytes/anatomopathologie , Péricytes/métabolisme , Mâle , Femelle , Cicatrice/anatomopathologie , Souris de lignée C57BL , Cellules stromales/anatomopathologie
18.
Open Biol ; 14(6): 230349, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38862017

RÉSUMÉ

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.


Sujet(s)
COVID-19 , Cellules endothéliales , Péricytes , SARS-CoV-2 , Péricytes/virologie , Péricytes/métabolisme , Péricytes/anatomopathologie , Humains , Animaux , SARS-CoV-2/physiologie , SARS-CoV-2/pathogénicité , COVID-19/virologie , COVID-19/anatomopathologie , Souris , Cellules endothéliales/virologie , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Facteurs de risque , Lipopolysaccharides/pharmacologie , Apolipoprotéine E4/génétique , Apolipoprotéine E4/métabolisme , Apolipoprotéine E3/génétique , Apolipoprotéine E3/métabolisme , Inflammation/virologie , Inflammation/anatomopathologie
19.
Elife ; 122024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38856719

RÉSUMÉ

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.


Sujet(s)
Dysfonctionnement érectile , Pénis , Péricytes , Analyse de l'expression du gène de la cellule unique , Animaux , Humains , Mâle , Souris , Dysfonctionnement érectile/génétique , Dysfonctionnement érectile/métabolisme , Souris de lignée C57BL , Pénis/métabolisme , Péricytes/métabolisme , Transcriptome
20.
J Clin Invest ; 134(14)2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38885342

RÉSUMÉ

While inflammation is beneficial for insulin secretion during homeostasis, its transformation adversely affects ß cells and contributes to diabetes. However, the regulation of islet inflammation for maintaining glucose homeostasis remains largely unknown. Here, we identified pericytes as pivotal regulators of islet immune and ß cell function in health. Islets and pancreatic pericytes express various cytokines in healthy humans and mice. To interfere with the pericytic inflammatory response, we selectively inhibited the TLR/MyD88 pathway in these cells in transgenic mice. The loss of MyD88 impaired pericytic cytokine production. Furthermore, MyD88-deficient mice exhibited skewed islet inflammation with fewer cells, an impaired macrophage phenotype, and reduced IL-1ß production. This aberrant pericyte-orchestrated islet inflammation was associated with ß cell dedifferentiation and impaired glucose response. Additionally, we found that Cxcl1, a pericytic MyD88-dependent cytokine, promoted immune IL-1ß production. Treatment with either Cxcl1 or IL-1ß restored the mature ß cell phenotype and glucose response in transgenic mice, suggesting a potential mechanism through which pericytes and immune cells regulate glucose homeostasis. Our study revealed pericyte-orchestrated islet inflammation as a crucial element in glucose regulation, implicating this process as a potential therapeutic target for diabetes.


Sujet(s)
Inflammation , Interleukine-1 bêta , Facteur de différenciation myéloïde-88 , Péricytes , Transduction du signal , Animaux , Facteur de différenciation myéloïde-88/génétique , Facteur de différenciation myéloïde-88/métabolisme , Souris , Péricytes/métabolisme , Péricytes/anatomopathologie , Péricytes/immunologie , Humains , Inflammation/anatomopathologie , Inflammation/métabolisme , Inflammation/génétique , Inflammation/immunologie , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/génétique , Interleukine-1 bêta/immunologie , Souris transgéniques , Récepteurs de type Toll/métabolisme , Récepteurs de type Toll/génétique , Chimiokine CXCL1/métabolisme , Chimiokine CXCL1/génétique , Ilots pancréatiques/immunologie , Ilots pancréatiques/métabolisme , Ilots pancréatiques/anatomopathologie , Souris knockout , Cellules à insuline/métabolisme , Cellules à insuline/anatomopathologie , Cellules à insuline/immunologie , Mâle , Glucose/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE