Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 11.893
Filtrer
1.
Gen Physiol Biophys ; 43(4): 301-312, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38953570

RÉSUMÉ

Vascular endothelial growth factor A (VEGFA) is an important regulator for non-small cell lung cancer (NSCLC). Our study aimed to reveal its upstream pathway to provide new ideas for developing the therapeutic targets of NSCLC. The mRNA and protein levels of VEGFA, ubiquitin-specific peptidase 35 (USP35), and FUS were determined by quantitative real-time PCR and Western blot. Cell proliferation, apoptosis, invasion and angiogenesis were detected using CCK8 assay, EdU assay, flow cytometry, transwell assay and tube formation assay. The interaction between USP35 and VEGFA was assessed by Co-IP assay and ubiquitination assay. Animal experiments were performed to assess USP35 and VEGFA roles in vivo. VEGFA had elevated expression in NSCLC tissues and cells. Interferences of VEGFA inhibited NSCLC cell proliferation, invasion, angiogenesis, and increased apoptosis. USP35 could stabilize VEGFA protein level by deubiquitination, and USP35 knockdown suppressed NSCLC cell growth, invasion and angiogenesis via reducing VEGFA expression. FUS interacted with USP35 to promote its mRNA stability, thereby positively regulating VEGFA expression. Also, USP35 silencing could reduce NSCLC tumorigenesis by downregulating VEGFA. FUS-stabilized USP35 facilitated NSCLC cell growth, invasion and angiogenesis through deubiquitinating VEGFA, providing a novel idea for NSCLC treatment.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Prolifération cellulaire , Tumeurs du poumon , Invasion tumorale , Néovascularisation pathologique , Protéine FUS de liaison à l'ARN , Ubiquitination , Facteur de croissance endothéliale vasculaire de type A , Humains , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Protéine FUS de liaison à l'ARN/métabolisme , Protéine FUS de liaison à l'ARN/génétique , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Prolifération cellulaire/génétique , Néovascularisation pathologique/métabolisme , Néovascularisation pathologique/génétique , Invasion tumorale/génétique , Lignée cellulaire tumorale , Souris , Animaux , Ubiquitin-specific proteases/métabolisme , Ubiquitin-specific proteases/génétique , Souris nude ,
2.
BMC Vet Res ; 20(1): 282, 2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-38951783

RÉSUMÉ

BACKGROUND: Wound management is a critical procedure in veterinary practice. A wound is an injury that requires the body's cells' alignment to break down due to external assault, such as trauma, burns, accidents, and diseases. Re-epithelization, extracellular matrix deposition, especially collagen, inflammatory cell infiltration, and development of new blood capillaries are the four features that are used to evaluate the healing process. Using a natural extract for wound management is preferred to avoid the side effects of synthetic drugs. The current study aimed to assess the effect of major pregnane glycoside arabincoside B (AR-B) isolated from Caralluma arabica (C. arabica) for the wound healing process. METHOD: AR-B was loaded on a gel for wound application. Rats were randomly distributed into six groups: normal, positive control (PC), MEBO®, AR-B 0.5%, AR-B 1%, and AR-B 1.5%, to be 6 animals in each group. Wounds were initiated under anesthesia with a 1 cm diameter tissue needle, and treatments were applied daily for 14 days. The collected samples were tested for SOD, NO, and MDA. Gene expression of VEGF and Caspase-3. Histopathological evaluation was performed at two-time intervals (7 and 14 days), and immunohistochemistry was done to evaluate α -SMA, TGF-ß, and TNF-α. RESULT: It was found that AR-B treatment enhanced the wound healing process. AR-B treated groups showed reduced MDA and NO in tissue, and SOD activity was increased. Re-epithelization and extracellular matrix deposition were significantly improved, which was confirmed by the increase in TGF-ß and α -SMA as well as increased collagen deposition. TNF-α was reduced, which indicated the subsiding of inflammation. VEGF and Caspase-3 expression were reduced. CONCLUSION: Our findings confirmed the efficiency of AR-B in enhancing the process of wound healing and its potential use as a topical wound dressing in veterinary practice.


Sujet(s)
Cicatrisation de plaie , Animaux , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Rats , Mâle , Apocynaceae/composition chimique , Bandages , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Hétérosides/pharmacologie , Hétérosides/usage thérapeutique , Prégnanes/pharmacologie , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/génétique , Superoxide dismutase/métabolisme , Caspase-3/métabolisme , Caspase-3/génétique , Rat Sprague-Dawley
3.
Acta Cir Bras ; 39: e392724, 2024.
Article de Anglais | MEDLINE | ID: mdl-38958304

RÉSUMÉ

PURPOSE: Gene expressions of vascular Endothelial Growth Factor Alpha (VEGFa), Nuclear Factor Kappa-Light-Chain-Enhancer of Activated B cells (NFkB) and cytokines could be useful for identifying potential therapeutic targets to alleviate ischemia-reperfusion injury after liver transplantation. Cytokine gene expressions, VEGFa and NFkB were investigated in a preclinical swine model of liver transplantation. METHODS: A total of 12 pigs were used as donors and recipients in liver transplantation without venovenous bypass or aortic clamping. NFkB, IL-6, IL-10, VEGFa and Notch1 gene expression were assessed. These samples were collected in two specific times: group 1 (n= 6) - control, samples were collected before recipient's total hepatectomy and group 2 - liver transplantation group (n=6), where the samples were collected one hour after graft reperfusion. RESULTS: Liver transplantation was successfully performed in all recipients. Liver enzymes were elevated in the transplantation group. NFkB gene expression was significantly decreased in the transplantation group in comparison with the control group (0.62±0.19 versus 0.39±0.08; p= 0.016). No difference was observed between groups Interleucine 6 (IL-6), interleucine 10 (IL-10), VEGFa and Notch homolog 1 (Notch1). CONCLUSIONS: In this survey a decreased NFkB gene expression in a porcine model of liver transplantation was observed.


Sujet(s)
Transplantation hépatique , Facteur de transcription NF-kappa B , Facteur de croissance endothéliale vasculaire de type A , Animaux , Facteur de croissance endothéliale vasculaire de type A/génétique , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/analyse , Suidae , Facteur de transcription NF-kappa B/métabolisme , Interleukine-10/analyse , Interleukine-6/analyse , Interleukine-6/génétique , Lésion d'ischémie-reperfusion , Expression des gènes , Modèles animaux de maladie humaine , Récepteur Notch1/génétique , Cytokines , Foie/métabolisme , Modèles animaux , Mâle
4.
Theranostics ; 14(9): 3509-3525, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948065

RÉSUMÉ

Rationale: Current treatments for ocular angiogenesis primarily focus on blocking the activity of vascular endothelial growth factor (VEGF), but unfavorable side effects and unsatisfactory efficacy remain issues. The identification of novel targets for anti-angiogenic treatment is still needed. Methods: We investigated the role of tsRNA-1599 in ocular angiogenesis using endothelial cells, a streptozotocin (STZ)-induced diabetic model, a laser-induced choroidal neovascularization model, and an oxygen-induced retinopathy model. CCK-8 assays, EdU assays, transwell assays, and matrigel assays were performed to assess the role of tsRNA-1599 in endothelial cells. Retinal digestion assays, Isolectin B4 (IB4) staining, and choroidal sprouting assays were conducted to evaluate the role of tsRNA-1599 in ocular angiogenesis. Transcriptomic analysis, metabolic analysis, RNA pull-down assays, and mass spectrometry were utilized to elucidate the mechanism underlying angiogenic effects mediated by tsRNA-1599. Results: tsRNA-1599 expression was up-regulated in experimental ocular angiogenesis models and endothelial cells in response to angiogenic stress. Silencing of tsRNA-1599 suppressed angiogenic effects in endothelial cells in vitro and inhibited pathological ocular angiogenesis in vivo. Mechanistically, tsRNA-1599 exhibited little effect on VEGF signaling but could cause reduced glycolysis and NAD+/NADH production in endothelial cells by regulating the expression of HK2 gene through interacting with YBX1, thus affecting endothelial effects. Conclusions: Targeting glycolytic reprogramming of endothelial cells by a tRNA-derived small RNA represents an exploitable therapeutic approach for ocular neovascular diseases.


Sujet(s)
Néovascularisation choroïdienne , Cellules endothéliales , Glycolyse , Animaux , Glycolyse/effets des médicaments et des substances chimiques , Souris , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Néovascularisation choroïdienne/traitement médicamenteux , Néovascularisation choroïdienne/métabolisme , Humains , Protéine-1 de liaison à la boîte Y/métabolisme , Protéine-1 de liaison à la boîte Y/génétique , Inhibiteurs de l'angiogenèse/pharmacologie , Hexokinase/métabolisme , Hexokinase/génétique , Diabète expérimental/traitement médicamenteux , Diabète expérimental/métabolisme , Souris de lignée C57BL , Mâle , Modèles animaux de maladie humaine , Néovascularisation pathologique/traitement médicamenteux , Néovascularisation pathologique/métabolisme , Néovascularisation pathologique/génétique , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Rétinopathie diabétique/traitement médicamenteux , Rétinopathie diabétique/métabolisme , Rétinopathie diabétique/génétique , Cellules endothéliales de la veine ombilicale humaine , Petit ARN non traduit/génétique , Petit ARN non traduit/métabolisme
5.
PeerJ ; 12: e17534, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948219

RÉSUMÉ

Ischemic stroke (IS) is a disease with a high mortality and disability rate worldwide, and its incidence is increasing per year. Angiogenesis after IS improves blood supply to ischemic areas, accelerating neurological recovery. ß-asarone has been reported to exhibit a significant protective effect against hypoxia injury. The ability of ß-asarone to improve IS injury by inducing angiogenesis has not been distinctly clarified. The experimental rats were induced with middle cerebral artery occlusion (MCAO), and oxygen-glucose deprivation (OGD) model cells were constructed using human microvascular endothelial cell line (HMEC-1) cells. Cerebral infarction and pathological damage were first determined via triphenyl tetrazolium chloride (TTC) and hematoxylin and eosin (H&E) staining. Then, cell viability, apoptosis, and angiogenesis were assessed by utilizing cell counting kit-8 (CCK-8), flow cytometry, spheroid-based angiogenesis, and tube formation assays in OGD HMEC-1 cells. Besides, angiogenesis and other related proteins were identified with western blot. The study confirms that ß-asarone, like nimodipine, can ameliorate cerebral infarction and pathological damage. ß-asarone can also upregulate vascular endothelial growth factor A (VEGFA) and endothelial nitric oxide synthase (eNOS) and induce phosphorylation of p38. Besides, the study proves that ß-asarone can protect against IS injury by increasing the expression of VEGFA. In vitro experiments affirmed that ß-asarone can induce viability and suppress apoptosis in OGD-mediated HMEC-1 cells and promote angiogenesis of OGD HMEC-1 cells by upregulating VEGFA. This establishes the potential for ß-asarone to be a latent drug for IS therapy.


Sujet(s)
Dérivés de l'allylbenzène , Anisoles , Apoptose , Survie cellulaire , Cellules endothéliales , Accident vasculaire cérébral ischémique , Régulation positive , Facteur de croissance endothéliale vasculaire de type A , Dérivés de l'allylbenzène/pharmacologie , Anisoles/pharmacologie , Anisoles/usage thérapeutique , Apoptose/effets des médicaments et des substances chimiques , Accident vasculaire cérébral ischémique/traitement médicamenteux , Accident vasculaire cérébral ischémique/anatomopathologie , Accident vasculaire cérébral ischémique/métabolisme , Humains , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Survie cellulaire/effets des médicaments et des substances chimiques , Animaux , Régulation positive/effets des médicaments et des substances chimiques , Rats , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Mâle , Lignée cellulaire , Rat Sprague-Dawley , Néovascularisation physiologique/effets des médicaments et des substances chimiques ,
6.
Genes (Basel) ; 15(6)2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38927656

RÉSUMÉ

Gene therapy holds promise as a transformative approach in the treatment landscape of age-related macular degeneration (AMD), diabetic retinopathy (DR), and diabetic macular edema (DME), aiming to address the challenges of frequent intravitreal anti-vascular endothelial growth factor (VEGF) injections. This manuscript reviews ongoing gene therapy clinical trials for these disorders, including ABBV-RGX-314, ixoberogene soroparvovec (ixo-vec), and 4D-150. ABBV-RGX-314 utilizes an adeno-associated virus (AAV) vector to deliver a transgene encoding a ranibizumab-like anti-VEGF antibody fragment, demonstrating promising results in Phase 1/2a and ongoing Phase 2b/3 trials. Ixo-vec employs an AAV2.7m8 capsid for intravitreal delivery of a transgene expressing aflibercept, showing encouraging outcomes in Phase 1 and ongoing Phase 2 trials. 4D-150 utilizes an evolved vector to express both aflibercept and a VEGF-C inhibitory RNAi, exhibiting positive interim results in Phase 1/2 studies. Other therapies reviewed include EXG102-031, FT-003, KH631, OLX10212, JNJ-1887, 4D-175, and OCU410. These therapies offer potential advantages of reduced treatment frequency and enhanced safety profiles, representing a paradigm shift in management towards durable and efficacious cellular-based biofactories. These advancements in gene therapy hold promise for improving outcomes in AMD and addressing the complex challenges of DME and DR, providing new avenues for the treatment of diabetic eye diseases.


Sujet(s)
Rétinopathie diabétique , Thérapie génétique , Dégénérescence maculaire , Humains , Rétinopathie diabétique/thérapie , Rétinopathie diabétique/génétique , Thérapie génétique/méthodes , Dégénérescence maculaire/thérapie , Dégénérescence maculaire/génétique , Vecteurs génétiques/génétique , Dependovirus/génétique , Facteur de croissance endothéliale vasculaire de type A/génétique , Animaux
7.
Reprod Biol Endocrinol ; 22(1): 74, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38918809

RÉSUMÉ

BACKGROUND: Erectile dysfunction (ED) is a common male sexual dysfunction, with an increasing incidence, and the current treatment is often ineffective. METHODS: Vascular endothelial growth factor (VEGFA) was used to treat bone marrow-derived mesenchymal stem cells (BM-MSCs), and their cell migration rates were determined by Transwell assays. The expression of the von Willebrand Factor (vWF)VE-cadherin, and endothelial nitric oxide synthase(eNOS) endothelial markers was determined by qRT‒PCR and Western blot analyses. The MALAT1-induced differentiation of BM-MCs to ECs via the CDC42/PAK1/paxillin pathway was explored by transfecting VEGFA-induced BM-MSC with si-MALAT1 and overexpressing CDC42 and PAK1. The binding capacity between CDC42, PAK1, and paxillin in VEGFA-treated and non-VEGFA-treated BM-MSCs was examined by protein immunoprecipitation. MiR-206 was overexpressed in VEGFA-induced BM-MSC, and the binding sites of MALAT1, miR-206, and CDC42 were identified using a luciferase assay. Sixty male Sprague‒Dawley rats were divided into six groups (n = 10/group). DMED modelling was demonstrated by APO experiments and was assessed by measuring blood glucose levels. Erectile function was assessed by measuring the intracavernosa pressure (ICP) and mean arterial pressure (MAP). Penile erectile tissue was analysed by qRT‒PCR, Western blot analysis, and immunohistochemical staining. RESULTS: MALAT1 under VEGFA treatment conditions regulates the differentiation of BM-MSCs into ECs by modulating the CDC42/PAK1/paxillin axis. In vitro experiments demonstrated that interference with CDC42 and MALAT1 expression inhibited the differentiation of BM-MSCs to ECs. CDC42 binds to PAK1, and PAK1 binds to paxillin. In addition, CDC42 in the VEGFA group had a greater ability to bind to PAK1, whereas PAK1 in the VEGFA group had a greater ability to bind to paxillin. Overexpression of miR-206 in VEGFA-induced BM-MSCs demonstrated that MALAT1 competes with the CDC42 3'-UTR for binding to miR-206, which in turn is involved in the differentiation of BM-MSCs to ECs. Compared to the DMED model group, the ICP/MAP ratio was significantly greater in the three BM-MSCs treatment groups. CONCLUSIONS: MALAT1 facilitates BM-MSC differentiation into ECs by regulating the miR-206/CDC42/PAK1/paxillin axis to improve ED. The present findings revealed the vital role of MALAT1 in the repair of BM-MSCs for erectile function and provided new mechanistic insights into the BM-MSC-mediated repair of DMED.


Sujet(s)
Différenciation cellulaire , Dysfonctionnement érectile , Cellules souches mésenchymateuses , microARN , Paxilline , ARN long non codant , Rat Sprague-Dawley , Transduction du signal , Protéine G cdc42 , p21-Activated Kinases , Mâle , Animaux , ARN long non codant/génétique , ARN long non codant/métabolisme , microARN/génétique , microARN/métabolisme , Différenciation cellulaire/génétique , Protéine G cdc42/métabolisme , Protéine G cdc42/génétique , Rats , p21-Activated Kinases/génétique , p21-Activated Kinases/métabolisme , Cellules souches mésenchymateuses/métabolisme , Dysfonctionnement érectile/thérapie , Dysfonctionnement érectile/génétique , Dysfonctionnement érectile/métabolisme , Paxilline/métabolisme , Paxilline/génétique , Cellules endothéliales/métabolisme , Cellules cultivées , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique
8.
Int J Nanomedicine ; 19: 6035-6055, 2024.
Article de Anglais | MEDLINE | ID: mdl-38911505

RÉSUMÉ

Background: Autism Spectrum Disorder (ASD) is a neurodevelopmental condition that affects social interaction and communication and can cause stereotypic behavior. Fullerenols, a type of carbon nanomaterial known for its neuroprotective properties, have not yet been studied for their potential in treating ASD. We aimed to investigate its role in improving autistic behaviors in BTBR T+Itpr3tf/J (BTBR) mice and its underlying mechanism, which could provide reliable clues for future ASD treatments. Methods: Our research involved treating C57BL/6J (C57) and BTBR mice with either 0.9% NaCl or fullerenols (10 mg/kg) daily for one week at seven weeks of age. We then conducted ASD-related behavioral tests in the eighth week and used RNA-seq to screen for vital pathways in the mouse hippocampus. Additionally, we used real-time quantitative PCR (RT-qPCR) to verify related pathway genes and evaluated the number of stem cells in the hippocampal dentate gyrus (DG) by Immunofluorescence staining. Results: Our findings revealed that fullerenols treatment significantly improved the related ASD-like behaviors of BTBR mice, manifested by enhanced social ability and improved cognitive deficits. Immunofluorescence results showed that fullerenols treatment increased the number of DCX+ and SOX2+/GFAP+ cells in the DG region of BTBR mice, indicating an expanded neural progenitor cell (NPC) pool of BTBR mice. RNA-seq analysis of the mouse hippocampus showed that VEGFA was involved in the rescued hippocampal neurogenesis by fullerenols treatment. Conclusion: In conclusion, our findings suggest that fullerenols treatment improves ASD-like behavior in BTBR mice by upregulating VEGFA, making nanoparticle- fullerenols a promising drug for ASD treatment.


Sujet(s)
Trouble du spectre autistique , Dysfonctionnement cognitif , Modèles animaux de maladie humaine , Protéine doublecortine , Fullerènes , Souris de lignée C57BL , Animaux , Souris , Fullerènes/pharmacologie , Fullerènes/composition chimique , Trouble du spectre autistique/traitement médicamenteux , Dysfonctionnement cognitif/traitement médicamenteux , Mâle , Comportement social , Comportement animal/effets des médicaments et des substances chimiques , Hippocampe/effets des médicaments et des substances chimiques , Facteur de croissance endothéliale vasculaire de type A/génétique , Neuroprotecteurs/pharmacologie , Neurogenèse/effets des médicaments et des substances chimiques , Trouble autistique/traitement médicamenteux
9.
Elife ; 132024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38842166

RÉSUMÉ

Endothelial and skeletal muscle lineages arise from common embryonic progenitors. Despite their shared developmental origin, adult endothelial cells (ECs) and muscle stem cells (MuSCs; satellite cells) have been thought to possess distinct gene signatures and signaling pathways. Here, we shift this paradigm by uncovering how adult MuSC behavior is affected by the expression of a subset of EC transcripts. We used several computational analyses including single-cell RNA-seq (scRNA-seq) to show that MuSCs express low levels of canonical EC markers in mice. We demonstrate that MuSC survival is regulated by one such prototypic endothelial signaling pathway (VEGFA-FLT1). Using pharmacological and genetic gain- and loss-of-function studies, we identify the FLT1-AKT1 axis as the key effector underlying VEGFA-mediated regulation of MuSC survival. All together, our data support that the VEGFA-FLT1-AKT1 pathway promotes MuSC survival during muscle regeneration, and highlights how the minor expression of select transcripts is sufficient for affecting cell behavior.


Sujet(s)
Survie cellulaire , Cellules endothéliales , Protéines proto-oncogènes c-akt , Transduction du signal , Facteur de croissance endothéliale vasculaire de type A , Récepteur-1 au facteur croissance endothéliale vasculaire , Animaux , Récepteur-1 au facteur croissance endothéliale vasculaire/métabolisme , Récepteur-1 au facteur croissance endothéliale vasculaire/génétique , Souris , Protéines proto-oncogènes c-akt/métabolisme , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Cellules endothéliales/métabolisme , Cellules endothéliales/physiologie , Muscles squelettiques/métabolisme , Cellules satellites du muscle squelettique/métabolisme , Cellules satellites du muscle squelettique/physiologie , Souris de lignée C57BL , Mâle
10.
Aging (Albany NY) ; 16(12): 10321-10347, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38874512

RÉSUMÉ

Microangiogenesis is an important prognostic factor in various cancers, including hepatocellular carcinoma (HCC). The Vascular Endothelial Growth Factor (VEGF) has been shown to contribute to tumor angiogenesis. Recently, several studies have investigated the regulation of VEGF production by a single gene, with few researchers exploring all genes that affect VEGF production. In this study, we comprehensively analyzed all genes affecting VEGF production in HCC and developed a risk model and gene-based risk score based on VEGF production. Moreover, the model's predictive capacity on prognosis of HCCs was verified using training and validation datasets. The developed model showed good prediction of the overall survival rate. Patients with a higher risk score experienced poor outcomes compared to those with a lower risk score. Furthermore, we identified the immunological causes of the poor prognosis of patients with high-risk scores comparing with those with low-risk scores.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , Néovascularisation pathologique , Facteur de croissance endothéliale vasculaire de type A , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/mortalité , Humains , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Tumeurs du foie/mortalité , Pronostic , Néovascularisation pathologique/génétique , Facteur de croissance endothéliale vasculaire de type A/génétique , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Régulation de l'expression des gènes tumoraux , Marqueurs biologiques tumoraux/génétique
11.
Aging (Albany NY) ; 16(12): 10546-10562, 2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-38916406

RÉSUMÉ

Metastasis is the primary cause of cancer-related deaths, and colorectal cancer (CRC) liver metastasis is a major poor prognostic factor in CRC. NAT1 (N-acetyltransferase 1) plays a crucial role in the invasive and metastatic processes of colorectal cancer. The role and molecular mechanism of NAT1 on tumor cells were verified by establishing a cell model of overexpression and knockdown of NAT1, and further verified by establishing a liver metastasis model of colorectal cancer for animal experiments. In vivo and in vitro experiments have demonstrated that overexpression of NAT1 reduces the ability of metastasis and invasion of colorectal cancer cells. NAT1 overexpression inhibits the PI3K/AKT/mTOR signaling pathway, thereby suppressing the EMT (epithelial-mesenchymal transition) process and glycolytic ability of tumor cells. Additionally, decreased glycolytic ability results in reduced VEGF (Vascular endothelial growth factor) expression in colorectal cancer cells. The decreased VEGF expression leads to decreased angiogenesis and vascular permeability in liver metastases, ultimately reducing the occurrence of liver metastasis. Our findings highlight that overexpression of NAT1 significantly inhibits the PI3K/AKT/mTOR signaling pathway, thereby suppressing EMT, glycolytic ability, and VEGF expression in colorectal cancer cells, collectively preventing the development of liver metastasis.


Sujet(s)
Arylamine N-acetyltransferase , Tumeurs colorectales , Transition épithélio-mésenchymateuse , Glycolyse , Tumeurs du foie , Transduction du signal , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Transition épithélio-mésenchymateuse/génétique , Humains , Tumeurs du foie/secondaire , Tumeurs du foie/génétique , Tumeurs du foie/métabolisme , Animaux , Arylamine N-acetyltransferase/génétique , Arylamine N-acetyltransferase/métabolisme , Lignée cellulaire tumorale , Souris , Sérine-thréonine kinases TOR/métabolisme , Isoenzymes/métabolisme , Isoenzymes/génétique , Phosphatidylinositol 3-kinases/métabolisme , Phosphatidylinositol 3-kinases/génétique , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Protéines proto-oncogènes c-akt/métabolisme , Régulation de l'expression des gènes tumoraux , Souris nude
12.
Int J Mol Sci ; 25(11)2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38892459

RÉSUMÉ

The aim of this study was to explore how the total flavonoids from Eucommia ulmoides leaves (EULs) regulate ischemia-induced nerve damage, as well as the protective effects mediated by oxidative stress. The cell survival rate was significantly improved compared to the ischemic group (p < 0.05) after treatment with the total flavonoids of EULs. The levels of reactive oxygen species (ROS), lactate dehydrogenase (LDH), and malondialdehyde (MDA) decreased, while catalase (CAT) and glutathione (GSH) increased, indicating that the total flavonoids of EULs can significantly alleviate neurological damage caused by ischemic stroke by inhibiting oxidative stress (p < 0.01). The mRNA expression level of VEGF increased (p < 0.01), which was consistent with the protein expression results. Meanwhile, the protein expression of ERK and CCND1 increased (p < 0.01), suggesting that the total flavonoids of EULs could protect PC12 cells from ischemic injury via VEGF-related pathways. MCAO rat models indicated that the total flavonoids of EULs could reduce brain ischemia-reperfusion injury. In conclusion, this study demonstrates the potential mechanisms of the total flavonoids of EULs in treating ischemic stroke and their potential therapeutic effects in reducing ischemic injury, which provides useful information for ischemic stroke drug discovery.


Sujet(s)
Eucommiaceae , Flavonoïdes , Accident vasculaire cérébral ischémique , Stress oxydatif , Feuilles de plante , Animaux , Rats , Flavonoïdes/pharmacologie , Eucommiaceae/composition chimique , Feuilles de plante/composition chimique , Cellules PC12 , Accident vasculaire cérébral ischémique/traitement médicamenteux , Accident vasculaire cérébral ischémique/métabolisme , Accident vasculaire cérébral ischémique/anatomopathologie , Stress oxydatif/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Mâle , Espèces réactives de l'oxygène/métabolisme , Extraits de plantes/pharmacologie , Extraits de plantes/composition chimique , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Survie cellulaire/effets des médicaments et des substances chimiques , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/métabolisme , Rat Sprague-Dawley , Malonaldéhyde/métabolisme
13.
J Cell Mol Med ; 28(12): e18489, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38899522

RÉSUMÉ

This study explores the impact of senescence on autocrine C-C motif chemokine ligand 5 (CCL5) in human endothelial progenitor cell (EPCs), addressing the poorly understood decline in number and function of EPCs during ageing. We examined the effects of replication-induced senescence on CCL5/CCL5 receptor (CCR5) signalling and angiogenic activity of EPCs in vitro and in vivo. We also explored microRNAs controlling CCL5 secretion in senescent EPCs, its impact on EPC angiogenic activity, and validated our findings in humans. CCL5 secretion and CCR5 levels in senescent EPCs were reduced, leading to attenuated angiogenic activity. CCL5 enhanced EPC proliferation via the CCR5/AKT/P70S6K axis and increased vascular endothelial growth factor (VEGF) secretion. Up-regulation of miR-409 in senescent EPCs resulted in decreased CCL5 secretion, inhibiting the angiogenic activity, though these negative effects were counteracted by the addition of CCL5 and VEGF. In a mouse hind limb ischemia model, CCL5 improved the angiogenic activity of senescent EPCs. Analysis involving 62 healthy donors revealed a negative association between CCL5 levels, age and Framingham Risk Score. These findings propose CCL5 as a potential biomarker for detection of EPC senescence and cardiovascular risk assessment, suggesting its therapeutic potential for age-related cardiovascular disorders.


Sujet(s)
Vieillissement de la cellule , Chimiokine CCL5 , Progéniteurs endothéliaux , microARN , Néovascularisation physiologique , Chimiokine CCL5/métabolisme , Chimiokine CCL5/génétique , Progéniteurs endothéliaux/métabolisme , Progéniteurs endothéliaux/cytologie , Humains , microARN/génétique , microARN/métabolisme , Animaux , Néovascularisation physiologique/génétique , Souris , Prolifération cellulaire , Mâle , Récepteurs CCR5/métabolisme , Récepteurs CCR5/génétique , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Régulation négative/génétique , Ischémie/métabolisme , Ischémie/anatomopathologie , Ischémie/génétique , Transduction du signal ,
14.
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
15.
Ecotoxicol Environ Saf ; 280: 116554, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38878335

RÉSUMÉ

Long non-coding RNA (lncRNA) is a novel emerging type of competitive endogenous RNA (ceRNA) that performs key functions in multiple biological processes. However, little is known about the roles of lncRNA under hypoxia stress in fish. Here, vascular endothelial growth factor-Aa (vegfaa) was cloned in rainbow trout (Oncorhynchus mykiss), with the complete cDNA sequence of 2914 bp, encoding 218 amino acids. The molecular weight of the protein was approximately 25.33 kDa, and contained PDGF and VEGF_C domains. Time-course and spatial expression patterns revealed that LOC110520012 was a key regulator of rainbow trout in response to hypoxia stress, and LOC110520012, miR-206-y and vegfaa exhibited a ceRNA regulatory relationship in liver, gill, muscle and rainbow trout liver cells treated with acute hypoxia. Subsequently, the targeting relationship of LOC110520012 and vegfaa with miR-206-y was confirmed by dual-luciferase reporter analysis, and overexpression of LOC110520012 mediated the inhibition of miR-206-y expression in rainbow trout liver cells, while the opposite results were obtained after LOC110520012 silencing with siRNA. We also proved that vegfaa was a target of miR-206-y in vitro and in vivo, and the vegfaa expression and anti-proliferative effect on rainbow trout liver cells regulated by miR-206-y mimics could be reversed by LOC110520012. These results suggested that LOC110520012 can positively regulate vegfaa expression by sponging miR-206-y under hypoxia stress in rainbow trout, which facilitate in-depth understanding of the molecular mechanisms of fish adaptation and tolerance to hypoxia.


Sujet(s)
Prolifération cellulaire , Foie , microARN , Oncorhynchus mykiss , ARN long non codant , Facteur de croissance endothéliale vasculaire de type A , Animaux , Oncorhynchus mykiss/génétique , microARN/génétique , ARN long non codant/génétique , Facteur de croissance endothéliale vasculaire de type A/génétique , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Foie/effets des médicaments et des substances chimiques , Hypoxie/génétique , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Néovascularisation physiologique/génétique ,
16.
Transl Vis Sci Technol ; 13(6): 13, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38899953

RÉSUMÉ

Purpose: To compare gene expression changes following branch retinal vein occlusion (BRVO) in the pig with and without bevacizumab (BEV) and triamcinolone acetonide (TA). Methods: Photothrombotic BRVOs were created in both eyes of four groups of nine pigs (2, 6, 10, and 20 days). In each group, six pigs received intravitreal injections of BEV in one eye and TA in the fellow eye, with three pigs serving as untreated BRVO controls. Three untreated pigs served as healthy controls. Expression of mRNA of vascular endothelial growth factor (VEGF), glial fibrillary acidic protein (GFAP), dystrophin (DMD), potassium inwardly rectifying channel subfamily J member 10 protein (Kir4.1, KCNJ10), aquaporin-4 (AQP4), stromal cell-derived factor-1α (CXCL12), interleukin-6 (IL6), interleukin-8 (IL8), monocyte chemoattractant protein-1 (CCL2), intercellular adhesion molecule 1 (ICAM1), and heat shock factor 1 (HSF1) were analyzed by quantitative reverse-transcription polymerase chain reaction. Retinal VEGF protein levels were characterized by immunohistochemistry. Results: In untreated eyes, BRVO significantly increased expression of GFAP, IL8, CCL2, ICAM1, HSF1, and AQP4. Expression of VEGF, KCNJ10, and CXCL12 was significantly reduced by 6 days post-BRVO, with expression recovering to healthy control levels by day 20. Treatment with BEV or TA significantly increased VEGF, DMD, and IL6 expression compared with untreated BRVO eyes and suppressed BRVO-induced CCL2 and AQP4 upregulation, as well as recovery of KCNJ10 expression, at 10 to 20 days post-BRVO. Conclusions: Inflammation and cellular osmohomeostasis rather than VEGF suppression appear to play important roles in BRVO-induced retinal neurodegeneration, enhanced in both BEV- and TA-treated retinas. Translational Relevance: Inner retinal neurodegeneration seen in this acute model of BRVO appears to be mediated by inflammation and alterations in osmohomeostasis rather than VEGF inhibition, which may have implications for more specific treatment modalities in the acute phase of BRVO.


Sujet(s)
Inhibiteurs de l'angiogenèse , Bévacizumab , Cytokines , Modèles animaux de maladie humaine , Injections intravitréennes , Occlusion veineuse rétinienne , Triamcinolone acétonide , Animaux , Bévacizumab/pharmacologie , Bévacizumab/usage thérapeutique , Triamcinolone acétonide/pharmacologie , Triamcinolone acétonide/administration et posologie , Triamcinolone acétonide/usage thérapeutique , Occlusion veineuse rétinienne/traitement médicamenteux , Occlusion veineuse rétinienne/métabolisme , Inhibiteurs de l'angiogenèse/pharmacologie , Inhibiteurs de l'angiogenèse/usage thérapeutique , Inhibiteurs de l'angiogenèse/administration et posologie , Cytokines/métabolisme , Cytokines/génétique , Suidae , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , ARN messager/métabolisme , ARN messager/génétique , Glucocorticoïdes/pharmacologie , Glucocorticoïdes/usage thérapeutique , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Protéine gliofibrillaire acide/métabolisme , Protéine gliofibrillaire acide/génétique , Canaux potassiques rectifiants entrants
17.
Sci Rep ; 14(1): 14325, 2024 06 21.
Article de Anglais | MEDLINE | ID: mdl-38906906

RÉSUMÉ

Diabetic retinopathy (DR) is a multifactorial disease displaying vascular-associated pathologies, including vascular leakage and neovascularization, ultimately leading to visual impairment. However, animal models accurately reflecting these pathologies are lacking. Vascular endothelial growth factor A (VEGF-A) is an important factor in the development of micro- and macro-vascular pathology in DR. In this study, we evaluated the feasibility of using a cumate-inducible lentivirus (LV) mediated expression of vegf-a to understand DR pathology in vitro and in vivo. Retinal pigment epithelial cells (ARPE-19) were transduced with cumate-inducible LV expressing vegf-a, with subsequent analysis of vegf-a expression and its impact on cell proliferation, viability, motility, and permeability. Cumate tolerability in adult Wistar rat eyes was assessed as an initial step towards a potential DR animal model development, by administering cumate via intravitreal injections (IVT) and evaluating consequent effects by spectral domain optical coherence tomography (SD-OCT), flash electroretinography (fERG), ophthalmic examination (OE), and immunohistochemistry. Transduction of ARPE-19 cells with cumate-inducible LV resulted in ~ 2.5-fold increase in vegf-a mRNA and ~ threefold increase in VEGF-A protein secretion. Transduced cells displayed enhanced cell proliferation, viability, permeability, and migration in tube-like structures. However, IVT cumate injections led to apparent retinal toxicity, manifesting as retinal layer abnormalities, haemorrhage, vitreous opacities, and significant reductions in a- and b-wave amplitudes, along with increased microglial activation and reactive gliosis. In summary, while cumate-inducible LV-mediated vegf-a expression is valuable for in vitro mechanistic studies in cellular drug discovery, its use is not a feasible approach to model DR in in vivo studies due to cumate-induced retinal toxicity.


Sujet(s)
Rétinopathie diabétique , Lentivirus , Épithélium pigmentaire de la rétine , Facteur de croissance endothéliale vasculaire de type A , Animaux , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Rétinopathie diabétique/anatomopathologie , Rétinopathie diabétique/métabolisme , Lentivirus/génétique , Rats , Épithélium pigmentaire de la rétine/métabolisme , Épithélium pigmentaire de la rétine/anatomopathologie , Humains , Rat Wistar , Prolifération cellulaire , Modèles animaux de maladie humaine , Lignée cellulaire , Injections intravitréennes , Mâle , Mouvement cellulaire , Survie cellulaire , Tomographie par cohérence optique , Vecteurs génétiques/administration et posologie , Vecteurs génétiques/génétique
18.
BMC Ophthalmol ; 24(1): 267, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38907191

RÉSUMÉ

OBJECTIVE: Diabetic retinopathy (DR) is a common complication of diabetes, and recent findings have shown that long noncoding RNAs (lncRNAs) may be involved in its pathogenesis. Through bioinformatics analysis, we found that lncRNA ATP2B2-IT2 may be involved in this process. This study primarily investigated the expression of the lncRNA ATP2B2-IT2 in human retinal microvascular endothelial cells (HRMECs) under high-glucose conditions and its effects on HRMEC proliferation, migration, and neovascularization. METHODS: We used RT‒PCR to assess the expression levels of lncRNA ATP2B2-IT2 and vascular endothelial growth factor (VEGF) in HRMECs under normal glucose (5.5 mmol/L) and high glucose (30 mmol/L) conditions. HRMECs were subsequently divided into four groups: the normal glucose (NG), high glucose (HG), high glucose with lncRNA ATP2B2-IT2 silencing (HG + si-lncRNA ATP2B2-IT2), and high glucose with silencing control (HG + si-NC) groups. The expression levels of the lncRNA ATP2B2-IT2 and VEGF in each group were determined using RT‒PCR. Thereafter, cell proliferation, migration, and neovascularization were assessed using CCK-8, Transwell, and tube formation assays, respectively. RESULTS: RT‒PCR revealed that the expression levels of the lncRNA ATP2B2-IT2 and VEGF were greater in the HG group than in the NG group (P < 0.05). After silencing of the lncRNA ATP2B2-IT2, the expression of VEGF decreased significantly (P < 0.05). Subsequent CCK-8, Transwell, and tube formation assays demonstrated that compared to those in the NG group, the HRMECs in the HG group exhibited significantly increased proliferation, migration, and neovascularization (P < 0.05). However, after silencing of the lncRNA ATP2B2-IT2, the proliferation, migration, and neovascularization of HRMECs were significantly decreased in the HG + si-lncRNA ATP2B2-IT2 group compared to those in the HG group (P < 0.05). CONCLUSION: LncRNA ATP2B2-IT2 may promote the proliferation, migration and neovascularization of HRMECs under high-glucose conditions.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Rétinopathie diabétique , ARN long non codant , Rétinopathie diabétique/génétique , Rétinopathie diabétique/métabolisme , ARN long non codant/génétique , Humains , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Cellules cultivées , Néovascularisation rétinienne/génétique , Néovascularisation rétinienne/métabolisme , Vaisseaux rétiniens/métabolisme , Vaisseaux rétiniens/anatomopathologie , Régulation de l'expression des gènes , Cellules endothéliales/métabolisme , Néovascularisation pathologique/génétique , Néovascularisation pathologique/métabolisme
19.
Sci Rep ; 14(1): 13612, 2024 06 13.
Article de Anglais | MEDLINE | ID: mdl-38871929

RÉSUMÉ

One of the most prevalent disorders of the urinary system is urinary tract infection, which is mostly brought on by uropathogenic Escherichia coli (UPEC). The objective of this study was to evaluate the regenerative therapeutic and antibacterial efficacy of PRP for induced bacterial cystitis in dogs in comparison to conventional antibiotics. 25 healthy male mongrel dogs were divided into 5 groups (n = 5). Control negative group that received neither induced infection nor treatments. 20 dogs were randomized into 4 groups after two weeks of induction of UPEC cystitis into; Group 1 (control positive; G1) received weekly intravesicular instillation of sodium chloride 0.9%. Group 2 (syst/PRP; G2), treated with both systemic intramuscular antibiotic and weekly intravesicular instillation of PRP; Group 3 (PRP; G3), treated with weekly intravesicular instillation of PRP, and Group 4 (syst; G4) treated with an intramuscular systemic antibiotic. Animals were subjected to weekly clinical, ultrasonographic evaluation, urinary microbiological analysis, and redox status biomarkers estimation. Urinary matrix metalloproteinases (MMP-2, MMP-9) and urinary gene expression for platelet-derived growth factor -B (PDGF-B), nerve growth factor (NGF), and vascular endothelial growth factor (VEGF) were measured. At the end of the study, dogs were euthanized, and the bladder tissues were examined macroscopically, histologically, and immunohistochemically for NF-κB P65 and Cox-2. The PRP-treated group showed significant improvement for all the clinical, Doppler parameters, and the urinary redox status (p < 0.05). The urinary MMPs activity was significantly decreased in the PRP-treated group and the expression level of urinary NGF and VEGF were downregulated while PDGFB was significantly upregulated (p < 0.05). Meanwhile, the urinary viable cell count was significantly reduced in all treatments (P < 0.05). Gross examination of bladder tissue showed marked improvement for the PRP-treated group, expressed in the histopathological findings. Immunohistochemical analysis revealed a marked increase in Cox-2 and NF-κB P65 in the PRP-treated group (P < 0.05). autologous CaCl2-activated PRP was able to overcome the bacterial infection, generating an inflammatory environment to overcome the old one and initiate tissue healing. Hence, PRP is a promising alternative therapeutic for UPEC cystitis instead of conventional antibiotics.


Sujet(s)
Cystite , Matrix metalloproteinase 2 , Matrix metalloproteinase 9 , Facteur de croissance nerveuse , Plasma riche en plaquettes , Facteur de croissance endothéliale vasculaire de type A , Animaux , Chiens , Facteur de croissance nerveuse/métabolisme , Plasma riche en plaquettes/métabolisme , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Cystite/métabolisme , Matrix metalloproteinase 9/métabolisme , Matrix metalloproteinase 9/génétique , Mâle , Matrix metalloproteinase 2/métabolisme , Matrix metalloproteinase 2/génétique , Modèles animaux de maladie humaine , Escherichia coli uropathogène/pathogénicité , Infections à Escherichia coli , Régulation négative , Infections urinaires/traitement médicamenteux
20.
Mol Biol Rep ; 51(1): 720, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38824268

RÉSUMÉ

BACKGROUND: Tumor-associated macrophages (TAM) exert a significant influence on the progression and heterogeneity of various subtypes of breast cancer (BRCA). However, the roles of heterogeneous TAM within BRCA subtypes remain unclear. Therefore, this study sought to elucidate the role of TAM across the following three BRCA subtypes: triple-negative breast cancer, luminal, and HER2. MATERIALS AND METHODS: This investigation aimed to delineate the variations in marker genes, drug sensitivity, and cellular communication among TAM across the three BRCA subtypes. We identified specific ligand-receptor (L-R) pairs and downstream mechanisms regulated by VEGFA-VEGFR1, SPP1-CD44, and SPP1-ITGB1 L-R pairs. Experimental verification of these pairs was conducted by co-culturing macrophages with three subtypes of BRCA cells. RESULTS: Our findings reveal the heterogeneity of macrophages within the three BRCA subtypes, evidenced by variations in marker gene expression, composition, and functional characteristics. Notably, heterogeneous TAM were found to promote invasive migration and epithelial-mesenchymal transition (EMT) in MDA-MB-231, MCF-7, and SKBR3 cells, activating NF-κB pathway via P38 MAPK, TGF-ß1, and AKT, respectively, through distinct VEGFA-VEGFR1, SPP1-CD44, and SPP1-ITGB1 L-R pairs. Inhibition of these specific L-R pairs effectively reversed EMT, migration, and invasion of each cancer cells. Furthermore, we observed a correlation between ligand gene expression and TAM sensitivity to anticancer drugs, suggesting a potential strategy for optimizing personalized treatment guidance. CONCLUSION: Our study highlights the capacity of heterogeneous TAM to modulate biological functions via distinct pathways mediated by specific L-R pairs within diverse BRCA subtypes. This study might provide insights into precision immunotherapy of different subtypes of BRCA.


Sujet(s)
Tumeurs du sein , Transition épithélio-mésenchymateuse , Macrophages associés aux tumeurs , Humains , Femelle , Macrophages associés aux tumeurs/métabolisme , Macrophages associés aux tumeurs/immunologie , Transition épithélio-mésenchymateuse/génétique , Lignée cellulaire tumorale , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Régulation de l'expression des gènes tumoraux , Analyse sur cellule unique/méthodes , Cellules MCF-7 , Mouvement cellulaire/génétique , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/métabolisme , Analyse de séquence d'ARN/méthodes , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Transduction du signal/génétique , Microenvironnement tumoral/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...