Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 5.176
Filtrer
1.
Int J Nanomedicine ; 19: 6519-6546, 2024.
Article de Anglais | MEDLINE | ID: mdl-38957181

RÉSUMÉ

Background: Salidroside (SAL) is the most effective component of Rhodiola rosea, a traditional Chinese medicine. Cryptotanshinone (CT) is the main fat-soluble extract of Salvia miltiorrhiza, exhibiting considerable potential for application in osteogenesis. Herein, a polycaprolactone/gelatin nanofiber membrane loaded with CT and SAL (PSGC membrane) was successfully fabricated via coaxial electrospinning and characterized. Methods and Results: This membrane capable of sustained and controlled drug release was employed in this study. Co-culturing the membrane with bone marrow mesenchymal stem cells and human umbilical vein endothelial cells revealed excellent biocompatibility and demonstrated osteogenic and angiogenic capabilities. Furthermore, drug release from the PSGC membrane activated the Wnt/ß-catenin signaling pathway and promoted osteogenic differentiation and vascularization. Evaluation of the membrane's vascularization and osteogenic capacities involved transplantation onto a rat's subcutaneous area and assessing rat cranium defects for bone regeneration, respectively. Microcomputed tomography, histological tests, immunohistochemistry, and immunofluorescence staining confirmed the membrane's outstanding angiogenic capacity two weeks post-operation, with a higher incidence of osteogenesis observed in rat cranial defects eight weeks post-surgery. Conclusion: Overall, the SAL- and CT-loaded coaxial electrospun nanofiber membrane synergistically enhances bone repair and regeneration.


Sujet(s)
Gélatine , Glucosides , Cellules endothéliales de la veine ombilicale humaine , Cellules souches mésenchymateuses , Nanofibres , Néovascularisation physiologique , Ostéogenèse , Phénanthrènes , Phénols , Polyesters , Rat Sprague-Dawley , Ostéogenèse/effets des médicaments et des substances chimiques , Animaux , Nanofibres/composition chimique , Gélatine/composition chimique , Polyesters/composition chimique , Glucosides/composition chimique , Glucosides/pharmacologie , Phénols/composition chimique , Phénols/pharmacologie , Phénanthrènes/composition chimique , Phénanthrènes/pharmacologie , Phénanthrènes/pharmacocinétique , Phénanthrènes/administration et posologie , Humains , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/cytologie , Rats , Mâle , Régénération osseuse/effets des médicaments et des substances chimiques , Membrane artificielle , Techniques de coculture , Libération de médicament , Différenciation cellulaire/effets des médicaments et des substances chimiques
2.
Int J Nanomedicine ; 19: 6449-6462, 2024.
Article de Anglais | MEDLINE | ID: mdl-38946883

RÉSUMÉ

Purpose: Functional inorganic nanomaterials (NMs) are widely exploited as bioactive materials and drug depots. The lack of a stable form of application of NMs at the site of skin injury, may impede the removal of the debridement, elevate pH, induce tissue toxicity, and limit their use in skin repair. This necessitates the advent of innovative wound dressings that overcome the above limitations. The overarching objective of this study was to exploit strontium-doped mesoporous silicon particles (PSiSr) to impart multifunctionality to poly(lactic-co-glycolic acid)/gelatin (PG)-based fibrous dressings (PG@PSiSr) for excisional wound management. Methods: Mesoporous silicon particles (PSi) and PSiSr were synthesized using a chemo-synthetic approach. Both PSi and PSiSr were incorporated into PG fibers using electrospinning. A series of structure, morphology, pore size distribution, and cumulative pH studies on the PG@PSi and PG@PSiSr membranes were performed. Cytocompatibility, hemocompatibility, transwell migration, scratch wound healing, and delineated angiogenic properties of these composite dressings were tested in vitro. The biocompatibility of composite dressings in vivo was assessed by a subcutaneous implantation model of rats, while their potential for wound healing was discerned by implantation in a full-thickness excisional defect model of rats. Results: The PG@PSiSr membranes can afford the sustained release of silicon ions (Si4+) and strontium ions (Sr2+) for up to 192 h as well as remarkably promote human umbilical vein endothelial cells (HUVECs) and NIH-3T3 fibroblasts migration. The PG@PSiSr membranes also showed better cytocompatibility, hemocompatibility, and significant formation of tubule-like networks of HUVECs in vitro. Moreover, PG@PSiSr membranes also facilitated the infiltration of host cells and promoted the deposition of collagen while reducing the accumulation of inflammatory cells in a subcutaneous implantation model in rats as assessed for up to day 14. Further evaluation of membranes transplanted in a full-thickness excisional wound model in rats showed rapid wound closure (PG@SiSr vs control, 96.1% vs 71.7%), re-epithelialization, and less inflammatory response alongside skin appendages formation (eg, blood vessels, glands, hair follicles, etc.). Conclusion: To sum up, we successfully fabricated PSiSr particles and prepared PG@PSiSr dressings using electrospinning. The PSiSr-mediated release of therapeutic ions, such as Si4+ and Sr2+, may improve the functionality of PLGA/Gel dressings for an effective wound repair, which may also have implications for the other soft tissue repair disciplines.


Sujet(s)
Bandages , Gélatine , Copolymère d'acide poly(lactique-co-glycolique) , Silicium , Peau , Strontium , Cicatrisation de plaie , Gélatine/composition chimique , Animaux , Strontium/composition chimique , Strontium/pharmacologie , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Copolymère d'acide poly(lactique-co-glycolique)/composition chimique , Peau/effets des médicaments et des substances chimiques , Porosité , Rats , Humains , Silicium/composition chimique , Rat Sprague-Dawley , Souris , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Mâle , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/pharmacologie
3.
Mar Drugs ; 22(6)2024 Jun 16.
Article de Anglais | MEDLINE | ID: mdl-38921594

RÉSUMÉ

Endothelial hyperpermeability is pivotal in sepsis-associated multi-organ dysfunction. Increased von Willebrand factor (vWF) plasma levels, stemming from activated platelets and endothelium injury during sepsis, can bind to integrin αvß3, exacerbating endothelial permeability. Hence, targeting this pathway presents a potential therapeutic avenue for sepsis. Recently, we identified isaridin E (ISE), a marine-derived fungal cyclohexadepsipeptide, as a promising antiplatelet and antithrombotic agent with a low bleeding risk. ISE's influence on septic mortality and sepsis-induced lung injury in a mouse model of sepsis, induced by caecal ligation and puncture, is investigated in this study. ISE dose-dependently improved survival rates, mitigating lung injury, thrombocytopenia, pulmonary endothelial permeability, and vascular inflammation in the mouse model. ISE markedly curtailed vWF release from activated platelets in septic mice by suppressing vesicle-associated membrane protein 8 and soluble N-ethylmaleide-sensitive factor attachment protein 23 overexpression. Moreover, ISE inhibited healthy human platelet adhesion to cultured lipopolysaccharide (LPS)-stimulated human umbilical vein endothelial cells (HUVECs), thereby significantly decreasing vWF secretion and endothelial hyperpermeability. Using cilengitide, a selective integrin αvß3 inhibitor, it was found that ISE can improve endothelial hyperpermeability by inhibiting vWF binding to αvß3. Activation of the integrin αvß3-FAK/Src pathway likely underlies vWF-induced endothelial dysfunction in sepsis. In conclusion, ISE protects against sepsis by inhibiting endothelial hyperpermeability and platelet-endothelium interactions.


Sujet(s)
Plaquettes , Cellules endothéliales de la veine ombilicale humaine , Sepsie , Facteur de von Willebrand , Animaux , Sepsie/traitement médicamenteux , Facteur de von Willebrand/métabolisme , Humains , Souris , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Mâle , Plaquettes/effets des médicaments et des substances chimiques , Plaquettes/métabolisme , Modèles animaux de maladie humaine , Souris de lignée C57BL , Endothélium vasculaire/effets des médicaments et des substances chimiques , Endothélium vasculaire/métabolisme , Intégrine alphaVbêta3/métabolisme , Intégrine alphaVbêta3/antagonistes et inhibiteurs , Perméabilité capillaire/effets des médicaments et des substances chimiques
4.
J Mater Chem B ; 12(25): 6221-6241, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38835196

RÉSUMÉ

Traumatic injuries, neurodegenerative diseases and oxidative stress serve as the early biomarkers for neuronal damage and impede angiogenesis and subsequently neuronal growth. Considering this, the present work aimed to develop a poly(N-acryloylglycine)-co-(acrylamide)-co-(N-acryloylglutamate) hydrogel [p(NAG-Ac-NAE)] with angiogenesis/neurogenesis properties. As constituents of this polymer modulate their vital role in biological functions, inhibitory neurotransmitter glycine regulates neuronal homeostasis, and glutamatergic signalling regulates angiogenesis. The p(NAG-Ac-NAE) hydrogel is a highly branched, biodegradable and pH-responsive polymer with a very high swelling behavior of 6188%. The mechanical stability (G', 2.3-2.7 kPa) of this polymeric hydrogel is commendable in the differentiation of mature neurons. This hydrogel is biocompatible (as tested in HUVEC cells) and helps to proliferate PC12 cells (152.7 ± 13.7%), whereas it is cytotoxic towards aggressive cancers such as glioblastoma (LN229 cells) and triple negative breast cancer (TNBC; MDA-MB-231 cells) and helps to maintain the healthy cytoskeleton framework structure of primary cortical neurons by facilitating the elongation of the axonal pathway. Furthermore, FACS results revealed that the synthesized hydrogel potentiates neurogenesis by inducing the cell cycle (G0/G1) and arresting the sub-G1 phase by limiting apoptosis. Additionally, RT-PCR results revealed that this hydrogel induced an increased level of HIF-1α expression, providing preconditioning effects towards neuronal cells under oxidative stress by scavenging ROS and initiating neurogenic and angiogenic signalling. This hydrogel further exhibits more pro-angiogenic activities by increasing the expression of VEGF isoforms compared to previously reported hydrogels. In conclusion, the newly synthesized p(NAG-Ac-NAE) hydrogel can be one of the potential neuroregenerative materials for vasculogenesis-assisted neurogenic applications and paramount for the management of neurodegenerative diseases.


Sujet(s)
Hydrogels , Stress oxydatif , Stress oxydatif/effets des médicaments et des substances chimiques , Hydrogels/composition chimique , Hydrogels/pharmacologie , Hydrogels/synthèse chimique , Humains , Animaux , Rats , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/pharmacologie , Matériaux biocompatibles/synthèse chimique , Neurogenèse/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Cellules PC12 , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Polymères/composition chimique , Polymères/pharmacologie , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique
5.
Chem Biol Interact ; 398: 111096, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38844257

RÉSUMÉ

Breast cancer is currently one of the most prevalent cancers worldwide. The mechanisms by which pesticides can increase breast cancer risk are multiple and complex. We have previously observed that two aryl hydrocarbon receptor (AhR) agonists ‒pesticides hexachlorobenzene (HCB) and chlorpyrifos (CPF)‒ act on tumor progression, stimulating cell migration and invasion in vitro and tumor growth in animal models. Elevated levels of hypoxia inducible factor-1α (HIF-1α) are found in malignant breast tumors, and HIF-1α is known to induce proangiogenic factors such as vascular endothelial growth factor (VEGF), nitric oxide synthase-2 (NOS-2) and cyclooxygenase-2 (COX-2), which are fundamental in breast cancer progression. In this work, we studied HCB (0.005, 0.05, 0.5 and 5 µM) and CPF (0.05, 0.5, 5 and 50 µM) action on the expression of these proangiogenic factors in triple negative breast cancer cells MDA-MB-231, as well as the effect of their conditioned medium (CM) on endothelial cells. Exposure to pesticides increased HIF-1α and VEGF protein expression in an AhR-dependent manner. In addition, HCB and CPF boosted NOS-2 and COX-2 content and VEGF secretion in MDA-MB-231 cells. The treatment of endothelial cells with CM from tumor cells exposed to pesticides increased cell proliferation, migration, and tubule formation, enhancing both tubule length and branching points. Of note, these effects were VEGF-dependent, as they were blocked in the presence of a VEGF receptor-2 (VEGFR-2) inhibitor. In sum, our results highlight the harmful impact of HCB and CPF in modulating the interaction between breast cancer and endothelial cells and promoting angiogenesis.


Sujet(s)
Chlorpyriphos , Cyclooxygenase 2 , Hexachloro-benzène , Sous-unité alpha du facteur-1 induit par l'hypoxie , Récepteurs à hydrocarbure aromatique , Tumeurs du sein triple-négatives , Facteur de croissance endothéliale vasculaire de type A , Chlorpyriphos/toxicité , Récepteurs à hydrocarbure aromatique/métabolisme , Humains , Hexachloro-benzène/métabolisme , Hexachloro-benzène/toxicité , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Lignée cellulaire tumorale , Cyclooxygenase 2/métabolisme , Ligands , Nitric oxide synthase type II/métabolisme , Femelle , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques
6.
Clinics (Sao Paulo) ; 79: 100403, 2024.
Article de Anglais | MEDLINE | ID: mdl-38878321

RÉSUMÉ

OBJECTIVES: This study aims to elucidate the role of circUSP9X (Circular RNA Ubiquitin Specific Peptidase 9 X-Linked) in the development of venous thrombosis in the lower extremities. METHODS: An animal model of Deep Vein Thrombosis (DVT) and a hypoxic model of Human Umbilical Vein Endothelial Cells (HUVECs) treated with Cobalt (II) Chloride (CoCl2) were developed. The expression levels of circUSP9X, microRNA-148b-3p (miR-148b-3p), and SRC Kinase Signaling Inhibitor 1 (SRCIN1) were quantified using quantitative reverse transcription Polymerase Chain Reaction and Western blot analysis. Cell cytotoxicity, viability, apoptosis, and inflammation in HUVECs were assessed via Lactate Dehydrogenase (LDH) assay, MTT assay, flow cytometry, Enzyme-Linked Immunosorbent Assay, and Western blot, respectively. Hematoxylin and Eosin staining were employed for histopathological examination of the venous tissues in the animal model. The interaction between circUSP9X, miR-148b-3p, and SRCIN1 was further explored through dual-luciferase reporter assays and RNA Immunoprecipitation experiments. RESULTS: The present findings reveal a significant upregulation of circUSP9X and SRCIN1 and a concurrent downregulation of miR-148b-3p in DVT cases. Knockdown of circUSP9X or overexpression of miR-148b-3p ameliorated CoCl2-induced apoptosis in HUVECs, reduced LDH release, enhanced cellular viability, and mitigated inflammation. Conversely, overexpression of circUSP9X intensified CoCl2's cytotoxic effects. The effects of manipulating circUSP9X expression were counteracted by the corresponding modulation of miR-148b-3p and SRCIN1 levels. Additionally, circUSP9X knockdown effectively inhibited the formation of DVT in the mouse model. A competitive binding mechanism of circUSP9X for miR-148b-3p, modulating SRCIN1 expression, was identified. CONCLUSION: circUSP9X promotes the formation of DVT through the regulation of the miR-148b-3p/SRCIN1 axis.


Sujet(s)
Modèles animaux de maladie humaine , Cellules endothéliales de la veine ombilicale humaine , microARN , Régulation positive , Thrombose veineuse , microARN/métabolisme , Animaux , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Humains , Régulation positive/effets des médicaments et des substances chimiques , ARN circulaire/génétique , Apoptose/effets des médicaments et des substances chimiques , Mâle , Souris
7.
Int J Biol Macromol ; 273(Pt 2): 132762, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38876232

RÉSUMÉ

Wound dressing diligently facilitate healing by fostering hemostasis, immunoregulation, the angiogenesis, and collagen deposition. Our methodology entails fabricating chitosan-taurine nanoparticles (CS-Tau) through an ionic gelation method. The morphology of CS-Tau was observed utilizing Transmission electron microscopy (TEM), scanning electron microscopy (SEM) and Dynamic Light Scattering (DLS). The nanoparticles are subsequently incorporated into carboxymethyl chitosan hydrogels for crosslinking by EDC-NHS, yielding hydrogel dressings (CMCS-CS-Tau) designed to extend the duration of taurine release. In vitro investigations confirmed that these innovative compound dressings displayed superior biodegradation, biocompatibility, cytocompatibility, and non-toxicity, in addition to possessing anti-inflammatory properties, and stimulating the proliferation and mobility of human umbilical vein endothelial cells (HUVECs). Experiments conducted on mice models with full-thickness skin removal demonstrated that CMCS-CS-Tau efficaciously aided in wound healing by spurring angiogenesis, and encouraging collagen deposition. CMCS-CS-Tau can also minimize inflammation and promote collagen deposition in chronic diabetic wound. Hence, CMCS-CS-Tau promotes both acute and chronic diabetic wound healing. Furthermore, the sustained release mechanism of CMCS-CS-Tau on taurine reveals promising potential for extending its clinical utility in relation to various biological effects of taurine.


Sujet(s)
Chitosane , Cellules endothéliales de la veine ombilicale humaine , Hydrogels , Nanoparticules , Taurine , Cicatrisation de plaie , Chitosane/composition chimique , Chitosane/analogues et dérivés , Chitosane/pharmacologie , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Animaux , Nanoparticules/composition chimique , Humains , Souris , Hydrogels/composition chimique , Hydrogels/pharmacologie , Taurine/analogues et dérivés , Taurine/composition chimique , Taurine/pharmacologie , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Diabète expérimental/traitement médicamenteux , Prolifération cellulaire/effets des médicaments et des substances chimiques , Mâle , Réactifs réticulants/composition chimique
8.
Biomed Pharmacother ; 176: 116907, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38865849

RÉSUMÉ

The plant alkaloid homoharringtonine (HHT) is a Food and Drug Administration (FDA)-approved drug for the treatment of hematologic malignancies. In addition to its well-established antitumor activity, accumulating evidence attributes anti-inflammatory effects to HHT, which have mainly been studied in leukocytes to date. However, a potential influence of HHT on inflammatory activation processes in endothelial cells, which are a key feature of inflammation and a prerequisite for the leukocyte-endothelial cell interaction and leukocyte extravasation, remains poorly understood. In this study, the anti-inflammatory potential of HHT and its derivative harringtonine (HT) on the TNF-induced leukocyte-endothelial cell interaction was assessed, and the underlying mechanistic basis of these effects was elucidated. HHT affected inflammation in vivo in a murine peritonitis model by reducing leukocyte infiltration and proinflammatory cytokine expression as well as ameliorating abdominal pain behavior. In vitro, HT and HHT impaired the leukocyte-endothelial cell interaction by decreasing the expression of the endothelial cell adhesion molecules intracellular adhesion molecule -1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1). This effect was mediated by a bipartite mechanism. While HHT did not affect the prominent TNF-induced pro-inflammatory NF-ĸB signaling cascade, the compound downregulated the VCAM1 mRNA expression in an IRF-1-dependent manner and diminished active ICAM1 mRNA translation as determined by polysome profiling. This study highlights HHT as an anti-inflammatory compound that efficiently hampers the leukocyte-endothelial cell interaction by targeting endothelial activation processes.


Sujet(s)
Régulation négative , Homoharringtonine , Inflammation , Facteur-1 de régulation d'interféron , ARN messager , Molécule-1 d'adhérence des cellules vasculaires , Animaux , Régulation négative/effets des médicaments et des substances chimiques , Molécule-1 d'adhérence des cellules vasculaires/métabolisme , Molécule-1 d'adhérence des cellules vasculaires/génétique , Inflammation/traitement médicamenteux , Inflammation/anatomopathologie , Inflammation/métabolisme , ARN messager/métabolisme , ARN messager/génétique , Humains , Facteur-1 de régulation d'interféron/métabolisme , Facteur-1 de régulation d'interféron/génétique , Souris , Homoharringtonine/pharmacologie , Mâle , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Anti-inflammatoires/pharmacologie , Molécule-1 d'adhérence intercellulaire/métabolisme , Molécule-1 d'adhérence intercellulaire/génétique , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Souris de lignée C57BL , Molécules d'adhérence cellulaire/métabolisme , Molécules d'adhérence cellulaire/génétique , Leucocytes/effets des médicaments et des substances chimiques , Leucocytes/métabolisme
9.
Molecules ; 29(11)2024 May 25.
Article de Anglais | MEDLINE | ID: mdl-38893380

RÉSUMÉ

Breast cancer is a major health concern and the leading cause of death among women worldwide. Standard treatment often involves surgery, radiotherapy, and chemotherapy, but these come with side effects and limitations. Researchers are exploring natural compounds like baicalin and baicalein, derived from the Scutellaria baicalensis plant, as potential complementary therapies. This study investigated the effects of baicalin and baicalein on the cytotoxic, proapoptotic, and genotoxic activity of doxorubicin and docetaxel, commonly used chemotherapeutic drugs for breast cancer. The analysis included breast cancer cells (MCF-7) and human endothelial cells (HUVEC-ST), to assess potential effects on healthy tissues. We have found that baicalin and baicalein demonstrated cytotoxicity towards both cell lines, with more potent effects observed in baicalein. Both flavonoids, baicalin (167 µmol/L) and baicalein (95 µmol/L), synergistically enhanced the cytotoxic, proapoptotic, and genotoxic activity of doxorubicin and docetaxel in breast cancer cells. In comparison, their effects on endothelial cells were mixed and depended on concentration and time. The results suggest that baicalin and baicalein might be promising complementary agents to improve the efficacy of doxorubicin and docetaxel anticancer activity. However, further research is needed to validate their safety and efficacy in clinical trials.


Sujet(s)
Apoptose , Tumeurs du sein , Docetaxel , Doxorubicine , Flavanones , Flavonoïdes , Humains , Flavonoïdes/pharmacologie , Flavanones/pharmacologie , Docetaxel/pharmacologie , Doxorubicine/pharmacologie , Cellules MCF-7 , Apoptose/effets des médicaments et des substances chimiques , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Femelle , Altération de l'ADN/effets des médicaments et des substances chimiques , Synergie des médicaments , Antinéoplasiques/pharmacologie , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques
10.
Int J Nanomedicine ; 19: 5157-5172, 2024.
Article de Anglais | MEDLINE | ID: mdl-38855731

RÉSUMÉ

Background: Poly-L-lactic acid (PLLA) stents have broad application prospects in the treatment of cardiovascular diseases due to their excellent mechanical properties and biodegradability. However, foreign body reactions caused by stent implantation remain a bottleneck that limits the clinical application of PLLA stents. To solve this problem, the biocompatibility of PLLA stents must be urgently improved. Albumin, the most abundant inert protein in the blood, possesses the ability to modify the surface of biomaterials, mitigating foreign body reactions-a phenomenon described as the "stealth effect". In recent years, a strategy based on albumin camouflage has become a focal point in nanomedicine delivery and tissue engineering research. Therefore, albumin surface modification is anticipated to enhance the surface biological characteristics required for vascular stents. However, the therapeutic applicability of this modification has not been fully explored. Methods: Herein, a bionic albumin (PDA-BSA) coating was constructed on the surface of PLLA by a mussel-inspired surface modification technique using polydopamine (PDA) to enhance the immobilization of bovine serum albumin (BSA). Results: Surface characterization revealed that the PDA-BSA coating was successfully constructed on the surface of PLLA materials, significantly improving their hydrophilicity. Furthermore, in vivo and in vitro studies demonstrated that this PDA-BSA coating enhanced the anticoagulant properties and pro-endothelialization effects of the PLLA material surface while inhibiting the inflammatory response and neointimal hyperplasia at the implantation site. Conclusion: These findings suggest that the PDA-BSA coating provides a multifunctional biointerface for PLLA stent materials, markedly improving their biocompatibility. Further research into the diverse applications of this coating in vascular implants is warranted.


Sujet(s)
Matériaux revêtus, biocompatibles , Polyesters , Polymères , Sérumalbumine bovine , Endoprothèses , Polyesters/composition chimique , Animaux , Sérumalbumine bovine/composition chimique , Matériaux revêtus, biocompatibles/composition chimique , Matériaux revêtus, biocompatibles/pharmacologie , Polymères/composition chimique , Matériaux biomimétiques/composition chimique , Matériaux biomimétiques/pharmacologie , Indoles/composition chimique , Indoles/pharmacologie , Propriétés de surface , Humains , Test de matériaux , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques
11.
Sci Rep ; 14(1): 11157, 2024 06 04.
Article de Anglais | MEDLINE | ID: mdl-38834598

RÉSUMÉ

Snakebite envenomation is a major public health issue which causes severe morbidity and mortality, affecting millions of people annually. Of a diverse range of clinical manifestations, local and systemic haemorrhage are of particular relevance, as this may result in ischemia, organ failure and even cardiovascular shock. Thus far, in vitro studies have failed to recapitulate the haemorrhagic effects observed in vivo. Here, we present an organ-on-a-chip approach to investigate the effects of four different snake venoms on a perfused microfluidic blood vessel model. We assess the effect of the venoms of four snake species on epithelial barrier function, cell viability, and contraction/delamination. Our findings reveal two different mechanisms by which the microvasculature is being affected, either by disruption of the endothelial cell membrane or by delamination of the endothelial cell monolayer from its matrix. The use of our blood vessel model may shed light on the key mechanisms by which tissue-damaging venoms exert their effects on the capillary vessels, which could be helpful for the development of effective treatments against snakebites.


Sujet(s)
Laboratoires sur puces , Venins de serpent , Animaux , Humains , Cellules endothéliales/effets des médicaments et des substances chimiques , Hémorragie , Survie cellulaire/effets des médicaments et des substances chimiques , Morsures de serpent/traitement médicamenteux , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Systèmes microphysiologiques
12.
Sci Rep ; 14(1): 14109, 2024 06 19.
Article de Anglais | MEDLINE | ID: mdl-38898080

RÉSUMÉ

Developing a reliable method to predict thrombocytopenia is imperative in drug discovery. Here, we establish an assay using a microphysiological system (MPS) to recapitulate the in-vivo mechanisms of platelet aggregation and adhesion. This assay highlights the role of shear stress on platelet aggregation and their interactions with vascular endothelial cells. Platelet aggregation induced by soluble collagen was detected under agitated, but not static, conditions using a plate shaker and gravity-driven flow using MPS. Notably, aggregates adhered on vascular endothelial cells under gravity-driven flow in the MPS, and this incident increased in a concentration-dependent manner. Upon comparing the soluble collagen-induced aggregation activity in platelet-rich plasma (PRP) and whole blood, remarkable platelet aggregate formation was observed at concentrations of 30 µg/mL and 3 µg/mL in PRP and whole blood, respectively. Moreover, ODN2395, an oligonucleotide, induced platelet aggregation and adhesion to vascular endothelial cells. SYK inhibition, which mediated thrombogenic activity via glycoprotein VI on platelets, ameliorated platelet aggregation in the system, demonstrating that the mechanism of platelet aggregation was induced by soluble collagen and oligonucleotide. Our evaluation system partially recapitulated the aggregation mechanisms in blood vessels and can contribute to the discovery of safe drugs to mitigate the risk of thrombocytopenia.


Sujet(s)
Plaquettes , Agrégation plaquettaire , Thrombopénie , Agrégation plaquettaire/effets des médicaments et des substances chimiques , Humains , Thrombopénie/induit chimiquement , Plaquettes/effets des médicaments et des substances chimiques , Plaquettes/métabolisme , Collagène/métabolisme , Collagène/pharmacologie , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Adhésivité plaquettaire/effets des médicaments et des substances chimiques , Syk kinase/métabolisme , Syk kinase/antagonistes et inhibiteurs , Plasma riche en plaquettes/métabolisme , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Systèmes microphysiologiques
13.
PLoS One ; 19(6): e0305906, 2024.
Article de Anglais | MEDLINE | ID: mdl-38905201

RÉSUMÉ

Uric acid induces radical oxygen species formation, endothelial inflammation, and endothelial dysfunction which contributes to the progression of atherosclerosis. Febuxostat inhibits BCRP- and allopurinol stimulates MRP4-mediated uric acid efflux in human embryonic kidney cells. We hypothesized that endothelial cells express uric acid transporters that regulate intracellular uric acid concentration and that modulation of these transporters by febuxostat and allopurinol contributes to their different impact on cardiovascular mortality. The aim of this study was to explore a potential difference between the effect of febuxostat and allopurinol on uric acid uptake by human umbilical vein endothelial cells. Febuxostat increased intracellular uric acid concentrations compared with control. In contrast, allopurinol did not affect intracellular uric acid concentration. In line with this observation, febuxostat increased mRNA expression of GLUT9 and reduced MRP4 expression, while allopurinol did not affect mRNA expression of these uric acid transporters. These findings provide a possible pathophysiological pathway which could explain the higher cardiovascular mortality for febuxostat compared to allopurinol but should be explored further.


Sujet(s)
Allopurinol , Fébuxostat , Transporteurs de glucose par diffusion facilitée , Cellules endothéliales de la veine ombilicale humaine , Protéines associées à la multirésistance aux médicaments , Acide urique , Humains , Allopurinol/pharmacologie , Fébuxostat/pharmacologie , Acide urique/métabolisme , Protéines associées à la multirésistance aux médicaments/métabolisme , Protéines associées à la multirésistance aux médicaments/génétique , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Transporteurs de glucose par diffusion facilitée/métabolisme , Transporteurs de glucose par diffusion facilitée/génétique , Transport biologique/effets des médicaments et des substances chimiques , ARN messager/génétique , ARN messager/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques
14.
Pharmacol Res ; 205: 107259, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38871237

RÉSUMÉ

The osteopontin-derived peptide FOL-005 stimulates hair growth. Using ligand-receptor glyco-capture technology we identified neuropilin-1 (NRP-1), a known co-receptor for vascular endothelial growth factor (VEGF) receptors, as the most probable receptor for FOL-005 and the more stable analogue FOL-026. X-ray diffraction and microscale thermophoresis analysis revealed that FOL-026 shares binding site with VEGF in the NRP-1 b1-subdomain. Stimulation of human umbilical vein endothelial cells with FOL-026 resulted in phosphorylation of VEGFR-2, ERK1/2 and AKT, increased cell growth and migration, stimulation of endothelial tube formation and inhibition of apoptosis in vitro. FOL-026 also promoted angiogenesis in vivo as assessed by subcutaneous Matrigel plug and hind limb ischemia models. NRP-1 knock-down or treatment of NRP-1 antagonist EG00229 blocked the stimulatory effects of FOL-026 on endothelial cells. Exposure of human coronary artery smooth muscle cells to FOL-026 stimulated cell growth, migration, inhibited apoptosis, and induced VEGF gene expression and VEGFR-2/AKT phosphorylation by an NRP-1-dependent mechanism. RNA sequencing showed that FOL-026 activated pathways involved in tissue repair. These findings identify NRP-1 as the receptor for FOL-026 and show that its biological effects mimic that of growth factors binding to the VEGF receptor family. They also suggest that FOL-026 may have therapeutical potential in conditions that require vascular repair and/or enhanced angiogenesis.


Sujet(s)
Cellules endothéliales de la veine ombilicale humaine , Néovascularisation physiologique , Neuropiline 1 , Ostéopontine , Neuropiline 1/métabolisme , Humains , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Animaux , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Ostéopontine/métabolisme , Ostéopontine/génétique , Mouvement cellulaire/effets des médicaments et des substances chimiques , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/métabolisme , Mâle , Peptides/pharmacologie , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Liaison aux protéines , Ischémie/traitement médicamenteux , Ischémie/métabolisme , Souris ,
15.
Biochem Biophys Res Commun ; 723: 150188, 2024 Sep 03.
Article de Anglais | MEDLINE | ID: mdl-38824808

RÉSUMÉ

Steroid (glucocorticoid)-induced necrosis of the femoral head (SONFH) represents a prevalent, progressive, and challenging bone and joint disease characterized by diminished osteogenesis and angiogenesis. Omaveloxolone (OMA), a semi-synthetic oleanocarpane triterpenoid with antioxidant, anti-inflammatory, and osteogenic properties, emerges as a potential therapeutic agent for SONFH. This study investigates the therapeutic impact of OMA on SONFH and elucidates its underlying mechanism. The in vitro environment of SONFH cells was simulated by inducing human bone marrow mesenchymal stem cells (hBMSCs) and human umbilical vein endothelial cells (HUVECs) using dexamethasone (DEX).Various assays, including CCK-8, alizarin red staining, Western blot, qPCR, immunofluorescence, flow cytometry, and TUNNEL, were employed to assess cell viability, STING/NF-κB signaling pathway-related proteins, hBMSCs osteogenesis, HUVECs migration, angiogenesis, and apoptosis. The results demonstrate that OMA promotes DEX-induced osteogenesis, HUVECs migration, angiogenesis, and anti-apoptosis in hBMSCs by inhibiting the STING/NF-κB signaling pathway. This experimental evidence underscores the potential of OMA in regulating DEX-induced osteogenesis, HUVECs migration, angiogenesis, and anti-apoptosis in hBMSCs through the STING/NF-κB pathway, thereby offering a promising avenue for improving the progression of SONFH.


Sujet(s)
Nécrose de la tête fémorale , Glucocorticoïdes , Cellules endothéliales de la veine ombilicale humaine , Néovascularisation physiologique , Ostéogenèse , Humains , Ostéogenèse/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Nécrose de la tête fémorale/induit chimiquement , Nécrose de la tête fémorale/anatomopathologie , Nécrose de la tête fémorale/traitement médicamenteux , Nécrose de la tête fémorale/métabolisme , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Glucocorticoïdes/pharmacologie , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Cellules cultivées , Apoptose/effets des médicaments et des substances chimiques , Dexaméthasone/pharmacologie , Facteur de transcription NF-kappa B/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Tête du fémur/anatomopathologie , Tête du fémur/effets des médicaments et des substances chimiques , Tête du fémur/vascularisation , Tête du fémur/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques , Triterpènes/pharmacologie , Survie cellulaire/effets des médicaments et des substances chimiques ,
16.
Life Sci Alliance ; 7(8)2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38839106

RÉSUMÉ

Targeted therapies against mutant BRAF are effectively used in combination with MEK inhibitors (MEKi) to treat advanced melanoma. However, treatment success is affected by resistance and adverse events (AEs). Approved BRAF inhibitors (BRAFi) show high levels of target promiscuity, which can contribute to these effects. The blood vessel lining is in direct contact with high plasma concentrations of BRAFi, but effects of the inhibitors in this cell type are unknown. Hence, we aimed to characterize responses to approved BRAFi for melanoma in the vascular endothelium. We showed that clinically approved BRAFi induced a paradoxical activation of endothelial MAPK signaling. Moreover, phosphoproteomics revealed distinct sets of off-targets per inhibitor. Endothelial barrier function and junction integrity were impaired upon treatment with vemurafenib and the next-generation dimerization inhibitor PLX8394, but not with dabrafenib or encorafenib. Together, these findings provide insights into the surprisingly distinct side effects of BRAFi on endothelial signaling and functionality. Better understanding of off-target effects could help to identify molecular mechanisms behind AEs and guide the continued development of therapies for BRAF-mutant melanoma.


Sujet(s)
Mélanome , Inhibiteurs de protéines kinases , Protéines proto-oncogènes B-raf , Transduction du signal , Vémurafénib , Protéines proto-oncogènes B-raf/antagonistes et inhibiteurs , Protéines proto-oncogènes B-raf/métabolisme , Humains , Inhibiteurs de protéines kinases/pharmacologie , Mélanome/traitement médicamenteux , Mélanome/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Vémurafénib/pharmacologie , Oximes/pharmacologie , Sulfonamides/pharmacologie , Endothélium vasculaire/effets des médicaments et des substances chimiques , Endothélium vasculaire/métabolisme , Imidazoles/pharmacologie , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Carbamates/pharmacologie , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Lignée cellulaire tumorale , Mutation
17.
Cancer Lett ; 596: 216961, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38823764

RÉSUMÉ

Extracellular vesicles are essential for intercellular communication and are involved in tumor progression. Inhibiting the direct release of extracellular vesicles seems to be an effective strategy in inhibiting tumor progression, but lacks of investigation. Here, we report a natural flavonoid compound, apigenin, could significantly inhibit the growth of hepatocellular carcinoma by preventing microvesicle secretion. Mechanistically, apigenin primarily targets the guanine nucleotide exchange factor ARHGEF1, inhibiting the activity of small G protein Cdc42, which is essential in regulating the release of microvesicles from tumor cells. In turn, this inhibits tumor angiogenesis related to VEGF90K transported on microvesicles, ultimately impeding tumor progression. Collectively, these findings highlight the therapeutic potential of apigenin and shed light on its anticancer mechanisms through inhibiting microvesicle biogenesis, providing a solid foundation for the refinement and practical application of apigenin.


Sujet(s)
Apigénine , Carcinome hépatocellulaire , Microparticules membranaires , Tumeurs du foie , Néovascularisation pathologique , Rho guanine nucleotide exchange factors , Humains , Néovascularisation pathologique/traitement médicamenteux , Néovascularisation pathologique/métabolisme , Animaux , Apigénine/pharmacologie , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Rho guanine nucleotide exchange factors/métabolisme , Rho guanine nucleotide exchange factors/génétique , Microparticules membranaires/métabolisme , Microparticules membranaires/effets des médicaments et des substances chimiques , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/vascularisation , Souris , Lignée cellulaire tumorale , Protéine G cdc42/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Cellules HepG2 , Souris nude ,
18.
Eur J Med Chem ; 274: 116510, 2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-38843585

RÉSUMÉ

Anti-angiogenic therapy has long been used as an adjunct therapy for the resolution of tumor burden. The current findings describe the synthesis of novel marine-based azirine-containing compounds that exhibit anti-angiogenic mediated anti-tumor activity. Azirine-2-carboxylate inhibited HUVEC-mediated tubulogenesis without causing cell death in a dose-dependent manner. Ex-vivo CAM, in-vivo Matrigel implantation, and ear angiogenesis experiments have all shown that azirine-2-carboxylate effectively inhibits angiogenesis. Furthermore, azirine-2-carboxylate inhibits the migration of ECs without disrupting the preformed tubule network. Azirine-2-carboxylate had adequate intramuscular systemic exposure and inhibited tumor growth in a xenograft mouse model. DARTS analysis, competitive binding assay, and gene expression investigations revealed that azirine-2-carboxylate inhibits endothelin-1-mediated angiogenesis. Overall, the discovery of azirine-2-carboxylate demonstrated a potent inhibition of angiogenesis targeting ET1 and a possible application in anti-angiogenic therapy.


Sujet(s)
Inhibiteurs de l'angiogenèse , Azirines , Cellules endothéliales de la veine ombilicale humaine , Humains , Inhibiteurs de l'angiogenèse/pharmacologie , Inhibiteurs de l'angiogenèse/composition chimique , Inhibiteurs de l'angiogenèse/synthèse chimique , Animaux , Souris , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Azirines/composition chimique , Azirines/pharmacologie , Azirines/synthèse chimique , Relation structure-activité , Structure moléculaire , Relation dose-effet des médicaments , Prolifération cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Néovascularisation pathologique/traitement médicamenteux
19.
Biomater Adv ; 162: 213927, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38917649

RÉSUMÉ

Metals are widely utilized as implant materials for bone fixtures as well as stents. Biodegradable versions of these implants are highly desirable since patients do not have to undergo a second surgery for the materials to be removed. Attractive options for such materials are zinc silver alloys since they also offer the benefit of being antibacterial. However, it is important to investigate the effect of the degradation products of such alloys on the surrounding cells, taking into account silver cytotoxicity. Here we investigated zinc alloyed with 1 % of silver (Zn1Ag) and how differently concentrated extracts (1 %-100 %) of this material impact human umbilical vein endothelial cells (HUVECs). More specifically, we focused on free radical generation and oxidative stress as well as the impact on cell viability. To determine free radical production we used diamond-based quantum sensing as well as conventional fluorescent assays. The viability was assessed by observing cell morphology and the metabolic activity via the MTT assay. We found that 1 % and 10 % extracts are well tolerated by the cells. However, at higher extract concentrations we observed severe impact on cell viability and oxidative stress. We were also able to show that quantum sensing was able to detect significant free radical generation even at the lowest tested concentrations.


Sujet(s)
Alliages , Survie cellulaire , Cellules endothéliales de la veine ombilicale humaine , Nanodiamants , Stress oxydatif , Zinc , Humains , Alliages/composition chimique , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Stress oxydatif/effets des médicaments et des substances chimiques , Nanodiamants/composition chimique , Argent/toxicité , Argent/composition chimique , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/pharmacologie , Radicaux libres/métabolisme , Test de matériaux/méthodes , Implant résorbable/effets indésirables
20.
Biol Pharm Bull ; 47(7): 1248-1254, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38866477

RÉSUMÉ

Ethanol (alcohol) is a risk factor that contributes to non-communicable diseases. Chronic abuse of ethanol is toxic to both the heart and overall health, and even results in death. Ethanol and its byproduct acetaldehyde can harm the cardiovascular system by impairing mitochondrial function, causing oxidative damage, and reducing contractile proteins. Endothelial cells are essential components of the cardiovascular system, are highly susceptible to ethanol, either through direct or indirect exposure. Thus, protection against endothelial injury is of great importance for persons who chronic abuse of ethanol. In this study, an in vitro model of endothelial injury was created using ethanol. The findings revealed that a concentration of 20.0 mM of ethanol reduced cell viability and Bcl-2 expression, while increasing cell apoptosis, intracellular reactive oxygen species (ROS) levels, mitochondrial depolarization, and the expression of Bax and cleaved-caspase-3 in endothelial cells. Further study showed that ethanol promoted nuclear translocation of nuclear factor kappa B (NF-κB), increased the secretion of tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, IL-6 in the culture medium, and inhibited nuclear factor-erythroid 2-related factor 2 (Nrf2) signaling pathway. The aforementioned findings suggest that ethanol has a harmful impact on endothelial cells. Nevertheless, the application of epigallocatechin-3-gallate (EGCG) to the cells can effectively mitigate the detrimental effects of ethanol on endothelial cells. In conclusion, EGCG alleviates ethanol-induced endothelial injury partly through alteration of NF-κB translocation and activation of the Nrf2 signaling pathway. Therefore, EGCG holds great potential in safeguarding individuals who chronically abuse ethanol from endothelial dysfunction.


Sujet(s)
Catéchine , Éthanol , Facteur-2 apparenté à NF-E2 , Facteur de transcription NF-kappa B , Transduction du signal , Éthanol/toxicité , Facteur-2 apparenté à NF-E2/métabolisme , Catéchine/analogues et dérivés , Catéchine/pharmacologie , Catéchine/usage thérapeutique , Facteur de transcription NF-kappa B/métabolisme , Humains , Transduction du signal/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Espèces réactives de l'oxygène/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Protéines proto-oncogènes c-bcl-2/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...