Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 387
Filtrer
1.
J Pharmacol Exp Ther ; 390(1): 108-115, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38834354

RÉSUMÉ

Nonsteroidal anti-inflammatory drugs (NSAIDs) possess anti-inflammatory, antipyretic, and analgesic properties and are among the most commonly used drugs. Although the cause of NSAID-induced gastric ulcers is well understood, the mechanism behind small intestinal ulcers remains elusive. In this study, we examined the mechanism through which indomethacin (IM), a prominent NSAID, induces small intestinal ulcers, both in vitro and in vivo. In IEC6 cells, a small intestinal epithelial cell line, IM treatment elevated levels of LC3-II and p62. These expression levels remained unaltered after treatment with chloroquine or bafilomycin, which are vacuolar ATPase (V-ATPase) inhibitors. IM treatment reduced the activity of cathepsin B, a lysosomal protein hydrolytic enzyme, and increased the lysosomal pH. There was a notable increase in subcellular colocalization of LC3 with Lamp2, a lysosome marker, post IM treatment. The increased lysosomal pH and decreased cathepsin B activity were reversed by pretreatment with rapamycin (Rapa) or glucose starvation, both of which stabilize V-ATPase assembly. To validate the in vitro findings in vivo, we established an IM-induced small intestine ulcer mouse model. In this model, we observed multiple ulcerations and heightened inflammation following IM administration. However, pretreatment with Rapa or fasting, which stabilize V-ATPase assembly, mitigated the IM-induced small intestinal ulcers in mice. Coimmunoprecipitation studies demonstrated that IM binds to V-ATPase in vitro and in vivo. These findings suggest that IM induces small intestinal injury through lysosomal dysfunction, likely due to the disassembly of lysosomal V-ATPase caused by direct binding. Moreover, Rapa or starvation can prevent this injury by stabilizing the assembly. SIGNIFICANCE STATEMENT: This study elucidates the largely unknown mechanisms behind small intestinal ulceration induced by indomethacin and reveals the involvement of lysosomal dysfunction via vacuolar ATPase disassembly. The significance lies in identifying potential preventative interventions, such as rapamycin treatment or glucose starvation, offering pivotal insights that extend beyond nonsteroidal anti-inflammatory drugs-induced ulcers to broader gastrointestinal pathologies and treatments, thereby providing a foundation for novel therapeutic strategies aimed at a wide array of gastrointestinal disorders.


Sujet(s)
Indométacine , Lysosomes , Sirolimus , Vacuolar Proton-Translocating ATPases , Animaux , Indométacine/toxicité , Lysosomes/effets des médicaments et des substances chimiques , Lysosomes/métabolisme , Vacuolar Proton-Translocating ATPases/métabolisme , Vacuolar Proton-Translocating ATPases/antagonistes et inhibiteurs , Sirolimus/pharmacologie , Souris , Mâle , Rats , Anti-inflammatoires non stéroïdiens/pharmacologie , Cathepsine B/métabolisme , Souris de lignée C57BL , Lignée cellulaire , Intestin grêle/effets des médicaments et des substances chimiques , Intestin grêle/anatomopathologie , Intestin grêle/métabolisme , Ulcère/induit chimiquement , Ulcère/anatomopathologie , Ulcère/métabolisme
2.
Cell Physiol Biochem ; 58(3): 250-272, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38865588

RÉSUMÉ

BACKGROUND/AIMS: Motivated by the vacuolar proton pump's importance in cancer, we investigate the effects of proton pump inhibition on breast cancer cell migration and proliferation, F-actin polymerization, lamin A/C, heterochromatin, and ETV7 expressions, nuclear size and shape, and AKT/mTOR signaling. METHODS: Lowly metastatic MCF7 and highly metastatic MDA-MB-231 breast cancer cells were treated with 120 nM of proton pump inhibitor Bafilomycin A1 for 24 hours. Cell migration was studied with wound- scratch assays, ATP levels with a chemiluminescent assay; cell proliferation was quantified by a cell area expansion assay. Nuclear size and shape were determined using DAPI nuclear stain and fluorescence microscopy. The levels of F-actin, lamin A/C, heterochromatin, and ETV7 were quantified using both immunocytochemistry and western blots; p-mTORC1, p-mTORC2, mTOR, p-AKT, and AKT were measured by western blots. RESULTS: We reveal that proton pump inhibition reduces F-actin polymerization, cell migration, proliferation, and increases heterochromatin in both lowly and highly metastatic cells. Surprisingly, Bafilomycin decreases lamin A/C in both cell lines. Inhibition has different effects on ETV7 expression in lowly and highly metastatic cells, as well as nuclear area, perimeter, and circularity. Bafilomycin also significantly decreases p-mTORC1, p-MTORC2, and MTOR expression in both cell lines, whereas it significantly decreases p-AKT in lowly metastatic cells and surprisingly significantly increases p-AKT in highly metastatic cells. Our proton pump inhibition protocol reduces V-ATPase levels (~25%) within three hours. V-ATPase levels vary in time for both control and inhibited cells, and inhibition reduces cellular ATP. CONCLUSION: Proton pumps promote F-actin polymerization and decrease heterochromatin, facilitating invasion. These pumps also upregulate both mTORC1 and mTORC2, thus highlighting the relevance of vacuolar proton pumps as metastatic cancer targets.


Sujet(s)
Actines , Tumeurs du sein , Mouvement cellulaire , Prolifération cellulaire , Hétérochromatine , Macrolides , Complexe-2 cible mécanistique de la rapamycine , Protéines proto-oncogènes c-akt , Transduction du signal , Sérine-thréonine kinases TOR , Vacuolar Proton-Translocating ATPases , Humains , Actines/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Femelle , Complexe-2 cible mécanistique de la rapamycine/métabolisme , Complexe-2 cible mécanistique de la rapamycine/antagonistes et inhibiteurs , Macrolides/pharmacologie , Vacuolar Proton-Translocating ATPases/métabolisme , Vacuolar Proton-Translocating ATPases/antagonistes et inhibiteurs , Prolifération cellulaire/effets des médicaments et des substances chimiques , Sérine-thréonine kinases TOR/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Hétérochromatine/métabolisme , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Complexe-1 cible mécanistique de la rapamycine/antagonistes et inhibiteurs , Cellules MCF-7
3.
Int J Nanomedicine ; 19: 3907-3917, 2024.
Article de Anglais | MEDLINE | ID: mdl-38708183

RÉSUMÉ

Background: As highlighted by recent pandemic outbreaks, antiviral drugs are crucial resources in the global battle against viral diseases. Unfortunately, most antiviral drugs are characterized by a plethora of side effects and low efficiency/poor bioavailability owing to their insolubility. This also applies to the arylnaphthalide lignin family member, diphyllin (Diph). Diph acts as a vacuolar ATPase inhibitor and has been previously identified as a promising candidate with broad-spectrum antiviral activity. However, its physicochemical properties preclude its efficient administration in vivo, complicating preclinical testing. Methods: We produced human recombinant H- ferritin (HsaFtH) and used it as a delivery vehicle for Diph encapsulation through pH-mediated reversible reassembly of HsaFtH. Diph nanoformulation was subsequently thoroughly characterized and tested for its non-target cytotoxicity and antiviral efficiency using a panel of pathogenic viral strain. Results: We revealed that loading into HsaFtH decreased the undesired cytotoxicity of Diph in mammalian host cells. We also confirmed that encapsulated Diph exhibited slightly lower antiviral activity than free Diph, which may be due to the differential uptake mechanism and kinetics of free Diph and Diph@HsaFtH. Furthermore, we confirmed that the antiviral effect was mediated solely by Diph with no contribution from HsaFtH. Conclusion: It was confirmed that HsaFtH is a suitable vehicle that allows easy loading of Diph and production of highly homogeneous nanoparticles dispersion with promising broad-spectrum antiviral activity.


Sujet(s)
Antiviraux , Lignanes , Animaux , Humains , Antiviraux/pharmacologie , Antiviraux/composition chimique , Antiviraux/pharmacocinétique , Interactions hydrophobes et hydrophiles , Nanoparticules/composition chimique , Protéines recombinantes/composition chimique , Vacuolar Proton-Translocating ATPases/antagonistes et inhibiteurs , Vacuolar Proton-Translocating ATPases/métabolisme
4.
Biochem Biophys Res Commun ; 718: 149981, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38735134

RÉSUMÉ

In animal cells, vacuoles are absent, but can be induced by diseases and drugs. While phosphoinositides are critical for membrane trafficking, their role in the formation of these vacuoles remains unclear. The immunosuppressive KRP203/Mocravimod, which antagonizes sphingosine-1-phosphate receptors, has been identified as having novel multimodal activity against phosphoinositide kinases. However, the impact of this novel KRP203 activity is unknown. Here, we show that KRP203 disrupts the spatial organization of phosphoinositides and induces extensive vacuolization in tumor cells and immortalized fibroblasts. The KRP203-induced vacuoles are primarily from endosomes, and augmented by inhibition of PIKFYVE and VPS34. Conversely, overexpression of PTEN decreased KRP203-induced vacuole formation. Furthermore, V-ATPase inhibition completely blunted KRP203-induced vacuolization, pointing to a critical requirement of the endosomal maturation process. Importantly, nearly a half of KRP203-induced vacuoles are significantly decorated with PI4P, a phosphoinositide typically enriched at the plasma membrane and Golgi. These results suggest a model that noncanonical spatial reorganization of phosphoinositides by KRP203 alters the endosomal maturation process, leading to vacuolization. Taken together, this study reveals a previously unrecognized bioactivity of KRP203 as a vacuole-inducing agent and its unique mechanism of phosphoinositide modulation, providing a new insight of phosphoinositide regulation into vacuolization-associated diseases and their molecular pathologies.


Sujet(s)
Endosomes , Phosphohydrolase PTEN , Phosphatidyl inositols , Vacuoles , Vacuoles/métabolisme , Vacuoles/effets des médicaments et des substances chimiques , Endosomes/métabolisme , Endosomes/effets des médicaments et des substances chimiques , Humains , Phosphatidyl inositols/métabolisme , Animaux , Phosphohydrolase PTEN/métabolisme , Phosphohydrolase PTEN/génétique , Phosphatidylinositol 3-kinases/métabolisme , Phosphatidylinositol 3-kinases de classe III/métabolisme , Phosphatidylinositol 3-kinases de classe III/génétique , Souris , Morpholines/pharmacologie , Vacuolar Proton-Translocating ATPases/métabolisme , Vacuolar Proton-Translocating ATPases/antagonistes et inhibiteurs , Vacuolar Proton-Translocating ATPases/génétique , Cytoplasme/métabolisme , Cellules HeLa , Aminopyridines , Composés hétérocycliques 3 noyaux
5.
Viruses ; 14(2)2022 02 09.
Article de Anglais | MEDLINE | ID: mdl-35215947

RÉSUMÉ

Diphyllin is a natural arylnaphtalide lignan extracted from tropical plants of particular importance in traditional Chinese medicine. This compound has been described as a potent inhibitor of vacuolar (H+)ATPases and hence of the endosomal acidification process that is required by numerous enveloped viruses to trigger their respective viral infection cascades after entering host cells by receptor-mediated endocytosis. Accordingly, we report here a revised, updated, and improved synthesis of diphyllin, and demonstrate its antiviral activities against a panel of enveloped viruses from Flaviviridae, Phenuiviridae, Rhabdoviridae, and Herpesviridae families. Diphyllin is not cytotoxic for Vero and BHK-21 cells up to 100 µM and exerts a sub-micromolar or low-micromolar antiviral activity against tick-borne encephalitis virus, West Nile virus, Zika virus, Rift Valley fever virus, rabies virus, and herpes-simplex virus type 1. Our study shows that diphyllin is a broad-spectrum host cell-targeting antiviral agent that blocks the replication of multiple phylogenetically unrelated enveloped RNA and DNA viruses. In support of this, we also demonstrate that diphyllin is more than just a vacuolar (H+)ATPase inhibitor but may employ other antiviral mechanisms of action to inhibit the replication cycles of those viruses that do not enter host cells by endocytosis followed by low pH-dependent membrane fusion.


Sujet(s)
Antiviraux/pharmacologie , Lignanes/pharmacologie , Virus/effets des médicaments et des substances chimiques , Animaux , Antigènes viraux/métabolisme , Antiviraux/synthèse chimique , Lignée cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Glucosides/pharmacologie , Lignanes/synthèse chimique , Vacuolar Proton-Translocating ATPases/antagonistes et inhibiteurs , Réplication virale/effets des médicaments et des substances chimiques , Virus/classification , Virus/métabolisme
6.
Molecules ; 27(2)2022 Jan 14.
Article de Anglais | MEDLINE | ID: mdl-35056817

RÉSUMÉ

Melatonin is a hormone secreted by the pineal gland that is involved in the biorhythm of reproductive activities. The present study investigated the inhibitory effects of melatonin on osteoclastogenesis in RAW 264.7 cells according to changes in V-ATPase and the corresponding inhibition of the MAPK and NFATc1 signaling processes. METHODS: the cytotoxic effect of melatonin was investigated by MTT assay. Osteoclast differentiation and gene expression of osteoclast-related factors were confirmed via TRAP staining, pit formation assay, immunofluorescence imaging, western blot, and real-time PCR. RESULTS: melatonin was found to inactivate the p38 and JNK of MAP kinase in RAW264.7 cells treated with RANKL and treated with a combination RANKL and melatonin for 1, 3, and 5 days. The melatonin treatment group showed a reduction in osteoclastogenesis transcription factors and ATP6v0d2 gene expression. CONCLUSIONS: melatonin inhibits osteoclast differentiation and cell fusion by inhibiting the expression of Atp6v0d2 through the inactivation of MAPK and NFATc1 signaling in RANKL-stimulated RAW264.7 macrophages. The findings of the present study suggest that melatonin could be a suitable therapy for bone loss and imply a potential role of melatonin in bone health.


Sujet(s)
Mélatonine/pharmacologie , Protéines membranaires/antagonistes et inhibiteurs , Mitogen-Activated Protein Kinases/antagonistes et inhibiteurs , Facteurs de transcription NFATC/antagonistes et inhibiteurs , Protéines de tissu nerveux/antagonistes et inhibiteurs , Ostéoclastes/cytologie , Ostéogenèse , Vacuolar Proton-Translocating ATPases/antagonistes et inhibiteurs , Animaux , Antioxydants/pharmacologie , Résorption osseuse/traitement médicamenteux , Résorption osseuse/métabolisme , Résorption osseuse/anatomopathologie , Différenciation cellulaire , Cellules cultivées , Régulation négative , Régulation de l'expression des gènes , Macrophages/cytologie , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Souris , Facteur de transcription NF-kappa B/antagonistes et inhibiteurs , Ostéoclastes/effets des médicaments et des substances chimiques , Ostéoclastes/métabolisme , Cellules RAW 264.7
7.
Oxid Med Cell Longev ; 2021: 9230435, 2021.
Article de Anglais | MEDLINE | ID: mdl-34777698

RÉSUMÉ

Hepatocellular carcinoma (HCC) has high morbidity and mortality. MicroRNAs (miRNAs), which could be regulated by cancer-derived exosomes, play critical regulatory roles in the initiation and development of cancer. However, the expressions, effects, and mechanisms of abundant miRNAs regulated by HCC cancer-derived exosomes in HCC remain largely unclear. Exosomes of HepG2 cells under heat shock, TGF-ß1, doxorubicin, acid and hypoxia/reoxygenation (H/R) conditions, and exosomes were successfully identified by transmission electron microscopy and Western blot analysis. The identified exosomes were then applied to evaluate the miRNA expression profiles by RNA sequencing. Mechanically, we discovered that doxorubicin was upregulated, TGF-ß1 downregulated the expressions of Vps4A, Rab27A, Alix, and Hrs in HepG2 cells and exosomes, and Vps4A and Rab27A, as target genes for miR-4454, could also be downregulated by miR-4454. Functionally, we revealed that miR-4454 inhibitor and miR-4454 inhibitor-mediated exosomes could markedly suppress proliferation, migration, invasion, and vascularization and accelerate cycle arrest, apoptosis, and ROS of HepG2 cells. This study provided many potential HCC cancer-derived exosome-mediated miRNAs in HCC under 5 different stimulus conditions. Meanwhile, we certified that miR-4454 in exosomes could provide a novel and effective mechanism for HCC function.


Sujet(s)
ATPases associated with diverse cellular activities/antagonistes et inhibiteurs , Marqueurs biologiques tumoraux/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Complexes de tri endosomique requis pour le transport/antagonistes et inhibiteurs , Régulation de l'expression des gènes tumoraux , Tumeurs du foie/anatomopathologie , microARN/génétique , Vacuolar Proton-Translocating ATPases/antagonistes et inhibiteurs , Protéines rab27 liant le GTP/antagonistes et inhibiteurs , ATPases associated with diverse cellular activities/génétique , ATPases associated with diverse cellular activities/métabolisme , Apoptose , Marqueurs biologiques tumoraux/génétique , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/métabolisme , Mouvement cellulaire , Prolifération cellulaire , Complexes de tri endosomique requis pour le transport/génétique , Complexes de tri endosomique requis pour le transport/métabolisme , Humains , Tumeurs du foie/génétique , Tumeurs du foie/métabolisme , Cellules cancéreuses en culture , Vacuolar Proton-Translocating ATPases/génétique , Vacuolar Proton-Translocating ATPases/métabolisme , Protéines rab27 liant le GTP/génétique , Protéines rab27 liant le GTP/métabolisme
8.
J Immunol Res ; 2021: 6670495, 2021.
Article de Anglais | MEDLINE | ID: mdl-33860063

RÉSUMÉ

At present, liver ischemia-reperfusion (IR) injury is still a great challenge for clinical liver partial resection and liver transplantation. The innate immunity regulated by liver macrophages orchestrates the cascade of IR inflammation and acts as a bridge. As a specific macrophage subunit of vacuolar ATPase, ATP6V0D2 (V-ATPase D2 subunit) has been shown to promote the formation of autophagolysosome in vitro. Our research fills a gap which has existed in the study of inflammatory stress about the V-ATPase subunit ATP6V0D2 in liver macrophages. We first found that the expression of specific ATP6V0D2 in liver macrophages was upregulated with the induction of inflammatory cascade after liver IR surgery, and knockdown of ATP6V0D2 resulted in increased secretion of proinflammatory factors and chemokines, which enhanced activation of NLRP3 and aggravation of liver injury. Further studies found that the exacerbated activation of NLRP3 was related to the autophagic flux regulated by ATP6V0D2. Knocking down ATP6V0D2 impaired the formation of autophagolysosome and aggravated liver IR injury through nonspecific V-ATPase activation independent of V-ATPase-Notchl-Hesl signal axis. In general, we illustrated that the expression of ATP6V0D2 in liver macrophages was upregulated after liver IR, and by gradually promoting the formation of autophagolysosomes to increase autophagy flux to limit the activation of liver inflammation, this regulation is independent of the Notch1-Hes1 signal axis.


Sujet(s)
Autophagie/immunologie , Inflammasomes/immunologie , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Lésion d'ischémie-reperfusion/immunologie , Vacuolar Proton-Translocating ATPases/métabolisme , Animaux , Autophagosomes/immunologie , Autophagosomes/métabolisme , Cellules cultivées , Modèles animaux de maladie humaine , Techniques de knock-down de gènes , Hépatectomie/effets indésirables , Hépatectomie/méthodes , Humains , Inflammasomes/métabolisme , Foie/vascularisation , Foie/immunologie , Foie/anatomopathologie , Foie/chirurgie , Transplantation hépatique/effets indésirables , Lysosomes/immunologie , Lysosomes/métabolisme , Macrophages/immunologie , Macrophages/métabolisme , Mâle , Souris , Récepteur Notch1/métabolisme , Lésion d'ischémie-reperfusion/étiologie , Lésion d'ischémie-reperfusion/anatomopathologie , Lésion d'ischémie-reperfusion/prévention et contrôle , Transduction du signal/immunologie , Facteur de transcription HES-1/métabolisme , Régulation positive , Vacuolar Proton-Translocating ATPases/antagonistes et inhibiteurs , Vacuolar Proton-Translocating ATPases/génétique
9.
Am J Respir Cell Mol Biol ; 65(2): 146-156, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-33789071

RÉSUMÉ

In a newborn pig cystic fibrosis (CF) model, the ability of gland-containing airways to fight infection was affected by at least two major host-defense defects: impaired mucociliary transport and a lower airway surface liquid (ASL) pH. In the gland-containing airways, the ASL pH is balanced by CFTR (CF transmembrane conductance regulator) and ATP12A, which, respectively, control HCO3- transport and proton secretion. We found that, although porcine small airway tissue expressed lower amounts of ATP12A, the ASL of epithelial cultures from CF distal small airways (diameter < 200 µm) were nevertheless more acidic (compared with non-CF airways). Therefore, we hypothesized that gland-containing airways and small airways control acidification using distinct mechanisms. Our microarray data suggested that small airway epithelia mediate proton secretion via ATP6V0D2, an isoform of the V0 d subunit of the H+-translocating plasma membrane V-type ATPase. Immunofluorescence of small airways verified the expression of the V0 d2 subunit isoform at the apical surface of Muc5B+ secretory cells, but not ciliated cells. Inhibiting the V-type ATPase with bafilomycin A1 elevated the ASL pH of small airway cultures, in the presence or absence of HCO3-, and decreased ASL viscosity. These data suggest that, unlike large airways, which are acidified by ATP12A activity, small airways are acidified by V-type ATPase, thus identifying V-type ATPase as a novel therapeutic target for small airway diseases.


Sujet(s)
Hydrogénocarbonates/métabolisme , Mucoviscidose/métabolisme , Cellules épithéliales/métabolisme , Muqueuse respiratoire/métabolisme , Vacuolar Proton-Translocating ATPases/métabolisme , Animaux , Animal génétiquement modifié , Mucoviscidose/génétique , Protéine CFTR/génétique , Protéine CFTR/métabolisme , Femelle , Concentration en ions d'hydrogène , Mâle , Suidae , Vacuolar Proton-Translocating ATPases/antagonistes et inhibiteurs , Vacuolar Proton-Translocating ATPases/génétique
10.
Nat Commun ; 12(1): 1782, 2021 03 19.
Article de Anglais | MEDLINE | ID: mdl-33741963

RÉSUMÉ

Pharmacological inhibition of vacuolar-type H+-ATPase (V-ATPase) by its specific inhibitor can abrogate tumor metastasis, prevent autophagy, and reduce cellular signaling responses. Bafilomycin A1, a member of macrolide antibiotics and an autophagy inhibitor, serves as a specific and potent V-ATPases inhibitor. Although there are many V-ATPase structures reported, the molecular basis of specific inhibitors on V-ATPase remains unknown. Here, we report the cryo-EM structure of bafilomycin A1 bound intact bovine V-ATPase at an overall resolution of 3.6-Å. The structure reveals six bafilomycin A1 molecules bound to the c-ring. One bafilomycin A1 molecule engages with two c subunits and disrupts the interactions between the c-ring and subunit a, thereby preventing proton translocation. Structural and sequence analyses demonstrate that the bafilomycin A1-binding residues are conserved in yeast and mammalian species and the 7'-hydroxyl group of bafilomycin A1 acts as a unique feature recognized by subunit c.


Sujet(s)
Macrolides/pharmacologie , Vacuolar Proton-Translocating ATPases/antagonistes et inhibiteurs , Séquence d'acides aminés , Animaux , Sites de fixation , Biocatalyse/effets des médicaments et des substances chimiques , Bovins , Cryomicroscopie électronique , Antienzymes/composition chimique , Antienzymes/métabolisme , Antienzymes/pharmacologie , Macrolides/composition chimique , Macrolides/métabolisme , Modèles moléculaires , Structure moléculaire , Liaison aux protéines , Domaines protéiques , Similitude de séquences d'acides aminés , Vacuolar Proton-Translocating ATPases/composition chimique , Vacuolar Proton-Translocating ATPases/ultrastructure
11.
Mol Cancer Ther ; 20(4): 739-748, 2021 04.
Article de Anglais | MEDLINE | ID: mdl-33563753

RÉSUMÉ

Prostate cancer is critically dependent on androgen receptor (AR) signaling. Despite initial responsiveness to androgen deprivation, most patients with advanced prostate cancer subsequently progress to a clinically aggressive castrate-resistant prostate cancer (CRPC) phenotype, typically associated with expression of splice-variant or mutant AR forms. Although current evidence suggests that the vacuolar-ATPase (V-ATPase), a multiprotein complex that catalyzes proton transport across intracellular and plasma membranes, influences wild-type AR function, the effect of V-ATPase inhibition on variant AR function is unknown.Inhibition of V-ATPase reduced AR function in wild-type and mutant AR luciferase reporter models. In hormone-sensitive prostate cancer cell lines (LNCaP, DuCaP) and mutant AR CRPC cell lines (22Rv1, LNCaP-F877L/T878A), V-ATPase inhibition using bafilomycin-A1 and concanamycin-A reduced AR expression, and expression of AR target genes, at mRNA and protein levels. Furthermore, combining chemical V-ATPase inhibition with the AR antagonist enzalutamide resulted in a greater reduction in AR downstream target expression than enzalutamide alone in LNCaP cells. To investigate the role of individual subunit isoforms, siRNA and CRISPR-Cas9 were used to target the V1C1 subunit in 22Rv1 cells. Whereas transfection with ATP6V1C1-targeted siRNA significantly reduced AR protein levels and function, CRISPR-Cas9-mediated V1C1 knockout showed no substantial change in AR expression, but a compensatory increase in protein levels of the alternate V1C2 isoform.Overall, these results indicate that V-ATPase dysregulation is directly linked to both hormone-responsive prostate cancer and CRPC via impact on AR function. In particular, V-ATPase inhibition can reduce AR signaling regardless of mutant AR expression.


Sujet(s)
Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Récepteurs aux androgènes/effets des médicaments et des substances chimiques , Vacuolar Proton-Translocating ATPases/antagonistes et inhibiteurs , Humains , Mâle , Transfection
12.
Anal Biochem ; 616: 114088, 2021 03 01.
Article de Anglais | MEDLINE | ID: mdl-33358938

RÉSUMÉ

Endosomal escape is considered a crucial barrier that needs to be overcome by integrin-mediated E. coli for gene delivery into mammalian cells. Bafilomycin, a potent inhibitor of the H+ proton pump commonly employed to lower endosomal pH, was evaluated as part of the E. coli protocol during delivery. We found an increase in green fluorescent protein expression up 6.9, 3.2, 5.0, 2.8, and 4.5 fold in HeLa, HEK-293, A549, HT1080, and MCF-7 respectively, compared to untreated cells. Our result showed for the first time that Inhibition of lysosomal V-ATPase enhances E. coli efficiency.


Sujet(s)
Bacterial Proton-Translocating ATPases/antagonistes et inhibiteurs , Escherichia coli/enzymologie , Lysosomes/enzymologie , Vacuolar Proton-Translocating ATPases/antagonistes et inhibiteurs , Lignée de cellules transformées , Lignée cellulaire tumorale , Régulation négative/effets des médicaments et des substances chimiques , Escherichia coli/génétique , Vecteurs génétiques/métabolisme , Protéines à fluorescence verte/biosynthèse , Homéostasie/effets des médicaments et des substances chimiques , Humains , Concentration en ions d'hydrogène , Lysosomes/effets des médicaments et des substances chimiques , Macrolides/pharmacologie , Transfection/méthodes
13.
Antioxid Redox Signal ; 35(2): 93-112, 2021 07 10.
Article de Anglais | MEDLINE | ID: mdl-32757619

RÉSUMÉ

Aims: A high-salt diet can aggravate oxidative stress, and renal fibrosis via the brain and renal renin-angiotensin system (RAS) axis in chronic kidney disease (CKD) rats. (Pro)renin receptor (PRR) plays a role in regulating RAS and oxidative stress locally. However, whether central PRR regulates salt-induced renal injury in CKD remains undefined. Here, we hypothesized that the reduction of central PRR expression could ameliorate central lesions and thereby ameliorate renal injury in high-salt-load CKD rats. Results: We investigated RAS, sympathetic nerve activity, oxidative stress, inflammation, and tissue injury in subfornical organs and kidneys in high-salt-load 5/6 nephrectomy CKD rats after the silencing of central PRR expression by intracerebroventricular lentivirus-RNAi. We found that the sympathetic nerve activity was reduced, and the levels of inflammation and oxidative stress were decreased in both brain and kidney. Renal injury and fibrosis were ameliorated. To explore the mechanism by which central inhibition of PRR expression ameliorates kidney damage, we blocked central MAPK/ERK1/2 and PI3K/Akt signaling pathways as well as angiotensin converting enzyme 1-angiotensin II-angiotensin type 1 receptors (ACE1-Ang II-AT1R) axis. Salt-induced overexpression of renal RAS, inflammation, oxidative stress, and fibrosis in CKD rats were prevented by central blockade of the pathways. Innovation: This study provides new insights into the mechanisms underlying salt-induced kidney damage. Targeting central PRR or PRR-mediated signaling pathway may be a novel strategy for the treatment of CKD. Conclusions: These results suggested that the silencing of central PRR expression ameliorates salt-induced renal injury in CKD through Ang II-dependent and -independent pathways.


Sujet(s)
Vecteurs génétiques/administration et posologie , Récepteurs de surface cellulaire/génétique , Insuffisance rénale chronique/thérapie , Système rénine-angiotensine/effets des médicaments et des substances chimiques , Sodium alimentaire/effets indésirables , Vacuolar Proton-Translocating ATPases/génétique , Angiotensine-II/métabolisme , Animaux , Modèles animaux de maladie humaine , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Extinction de l'expression des gènes , Vecteurs génétiques/génétique , Perfusions intraventriculaires , Lentivirus/génétique , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Mâle , Néphrectomie/effets indésirables , Interférence par ARN , Rats , Rat Sprague-Dawley , Récepteurs de surface cellulaire/antagonistes et inhibiteurs , Récepteurs de surface cellulaire/métabolisme , Insuffisance rénale chronique/induit chimiquement , Insuffisance rénale chronique/génétique , Insuffisance rénale chronique/métabolisme , Vacuolar Proton-Translocating ATPases/antagonistes et inhibiteurs , Vacuolar Proton-Translocating ATPases/métabolisme
14.
J Nat Prod ; 83(11): 3381-3386, 2020 11 25.
Article de Anglais | MEDLINE | ID: mdl-33151675

RÉSUMÉ

Callyspongiolide is a marine-derived macrolide that kills cells in a caspase-independent manner. NCI COMPARE analysis of human tumor cell line toxicity data for synthetic callyspongiolide indicated that its pattern of cytotoxicity correlated with that seen for concanamycin A, an inhibitor of the vacuolar-type H+-ATPase (V-ATPase). Using yeast as a model system, we report that treatment with synthetic callyspongiolide phenocopied a loss of V-ATPase activity including (1) inability to grow on a nonfermentable carbon source, (2) rescue of cell growth via supplementation with Fe2+, (3) pH-sensitive growth, and (4) a vacuolar acidification defect visualized using the fluorescent dye quinacrine. Crucially, in an in vitro assay, callyspongiolide was found to dose-dependently inhibit yeast V-ATPase (IC50 = 10 nM). Together, these data identify callyspongiolide as a new and highly potent V-ATPase inhibitor. Notably, callyspongiolide is the first V-ATPase inhibitor known to be expelled by Pdr5p.


Sujet(s)
Antienzymes/pharmacologie , Macrolides/pharmacologie , Vacuolar Proton-Translocating ATPases/antagonistes et inhibiteurs , Antienzymes/composition chimique , Colorants fluorescents/composition chimique , Humains , Concentration en ions d'hydrogène , Macrolides/composition chimique , Structure moléculaire , Saccharomyces cerevisiae/croissance et développement , Saccharomyces cerevisiae/métabolisme
15.
Mol Oncol ; 14(10): 2436-2454, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-32797726

RÉSUMÉ

Tumor acidity is the key metabolic feature promoting cancer progression and is modulated by pH regulators on a cancer cell's surface that pump out excess protons/lactic acid for cancer cell survival. Neutralizing tumor acidity improves the therapeutic efficacy of current treatments including immunotherapies. Vacuolar-ATPase (V-ATPase) proton pumps encompass unique plasma membrane-associated subunit isoforms, making this molecule an important target for anticancer therapy. Here, we examined the in vivo therapeutic efficacy of an antibody (a2v-mAB) targeting specific V-ATPase-'V0a2' surface isoform in controlling ovarian tumor growth. In vitro a2v-mAb treatment inhibited the proton pump activity in ovarian cancer (OVCA) cells. In vivo intraperitoneal a2v-mAb treatment drastically delayed ovarian tumor growth with no measurable in vivo toxicity in a transplant tumor model. To explore the possible mechanism causing delayed tumor growth, histochemical analysis of the a2v-mAb-treated tumor tissues displayed high immune cell infiltration (M1-macrophages, neutrophils, CD103+ cells, and NK cells) and an enhanced antitumor response (iNOS, IFN-y, IL-1α) compared to control. There was marked decrease in CA-125-positive cancer cells and an enhanced active caspase-3 expression in a2v-mAb-treated tumors. RNA-seq analysis of a2v-mAb tumor tissues further revealed upregulation of apoptosis-related and toll-like receptor pathway-related genes. Indirect coculture of a2v-mAb-treated OVCA cells with human PBMCs in an unbuffered medium led to an enhanced gene expression of antitumor molecules IFN-y, IL-17, and IL-12-A in PBMCs, further validating the in vivo antitumor responses. In conclusion, V-ATPase inhibition using a monoclonal antibody directed against the V0a2 isoform increases antitumor immune responses and could therefore constitute an effective treatment strategy in OVCA.


Sujet(s)
Anticorps monoclonaux/pharmacologie , Immunité , Tumeurs de l'ovaire/immunologie , Tumeurs de l'ovaire/anatomopathologie , Vacuolar Proton-Translocating ATPases/antagonistes et inhibiteurs , Animaux , Carcinogenèse/effets des médicaments et des substances chimiques , Carcinogenèse/anatomopathologie , Caspase-3/métabolisme , Numération cellulaire , Lignée cellulaire tumorale , Prolifération cellulaire , Milieux de culture conditionnés/pharmacologie , Cytokines/génétique , Cytokines/métabolisme , Femelle , Humains , Médiateurs de l'inflammation/métabolisme , Agranulocytes/effets des médicaments et des substances chimiques , Agranulocytes/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Souris nude , Grading des tumeurs , Monoxyde d'azote/métabolisme , Nitric oxide synthase type II/métabolisme , Récepteurs de type Toll/métabolisme , Vacuolar Proton-Translocating ATPases/métabolisme
16.
Mol Cancer Ther ; 19(9): 1844-1855, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32669314

RÉSUMÉ

We previously reported that silencing of the PRR gene, which encodes the (pro)renin receptor [(P)RR], significantly reduced Wnt/ß-catenin-dependent development of pancreatic ductal adenocarcinoma (PDAC). Here, we examined the effects of a panel of blocking mAbs directed against the (P)RR extracellular domain on proliferation of the human PDAC cell lines PK-1 and PANC-1 in vitro and in vivo We observed that four rat anti-(P)RR mAbs induced accumulation of cells in the G0-G1-phase of the cell cycle and significantly reduced proliferation in vitro concomitant with an attenuation of Wnt/ß-catenin signaling. Systemic administration of the anti-(P)RR mAbs to nude mice bearing subcutaneous PK-1 xenografts significantly decreased tumor expression of active ß-catenin and the proliferation marker Ki-67, and reduced tumor growth. In contrast, treatment with the handle region peptide of (pro)renin did not inhibit tumor growth in vitro or in vivo, indicating that the effects of the anti-(P)RR mAbs were independent of the renin-angiotensin system. These data indicate that mAbs against human (P)RR can suppress PDAC cell proliferation by hindering activation of the Wnt/ß-catenin signaling pathway. Thus, mAb-mediated (P)RR blockade could be an attractive therapeutic strategy for PDAC.


Sujet(s)
Anticorps monoclonaux/administration et posologie , Antinéoplasiques immunologiques/administration et posologie , Carcinome du canal pancréatique/traitement médicamenteux , Tumeurs du pancréas/traitement médicamenteux , Récepteurs de surface cellulaire/composition chimique , Vacuolar Proton-Translocating ATPases/composition chimique , Animaux , Anticorps monoclonaux/pharmacologie , Antinéoplasiques immunologiques/pharmacologie , Carcinome du canal pancréatique/métabolisme , Cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Cellules HEK293 , Humains , Mâle , Souris , Tumeurs du pancréas/métabolisme , Domaines protéiques , Rats , Récepteurs de surface cellulaire/antagonistes et inhibiteurs , Vacuolar Proton-Translocating ATPases/antagonistes et inhibiteurs , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
17.
Cell Chem Biol ; 27(11): 1359-1370.e8, 2020 11 19.
Article de Anglais | MEDLINE | ID: mdl-32649904

RÉSUMÉ

Multidrug resistance (MDR) in cancer remains a major challenge for the success of chemotherapy. Natural products have been a rich source for the discovery of drugs against MDR cancers. Here, we applied high-throughput cytotoxicity screening of an in-house natural product library against MDR SGC7901/VCR cells and identified that the cyclodepsipeptide verucopeptin demonstrated notable antitumor potency. Cytological profiling combined with click chemistry-based proteomics revealed that ATP6V1G directly interacted with verucopeptin. ATP6V1G, a subunit of the vacuolar H+-ATPase (v-ATPase) that has not been previously targeted, was essential for SGC7901/VCR cell growth. Verucopeptin exhibited strong inhibition of both v-ATPase activity and mTORC1 signaling, leading to substantial pharmacological efficacy against SGC7901/VCR cell proliferation and tumor growth in vivo. Our results demonstrate that targeting v-ATPase via its V1G subunit constitutes a unique approach for modulating v-ATPase and mTORC1 signaling with great potential for the development of therapeutics against MDR cancers.


Sujet(s)
Antinéoplasiques/pharmacologie , Produits biologiques/pharmacologie , Depsipeptides/pharmacologie , Multirésistance aux médicaments/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Vacuolar Proton-Translocating ATPases/antagonistes et inhibiteurs , Animaux , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Produits biologiques/synthèse chimique , Produits biologiques/composition chimique , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Depsipeptides/synthèse chimique , Depsipeptides/composition chimique , Tests de criblage d'agents antitumoraux , Femelle , Humains , Mâle , Complexe-1 cible mécanistique de la rapamycine/antagonistes et inhibiteurs , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Souris , Souris de lignée BALB C , Souris nude , Tumeurs expérimentales/traitement médicamenteux , Tumeurs expérimentales/métabolisme , Tumeurs expérimentales/anatomopathologie , Sous-unités de protéines/effets des médicaments et des substances chimiques , Protéomique , Vacuolar Proton-Translocating ATPases/métabolisme
18.
Chem Biol Interact ; 327: 109138, 2020 Aug 25.
Article de Anglais | MEDLINE | ID: mdl-32485151

RÉSUMÉ

Altered intracellular distribution of weak base anticancer drugs owing to lysosomal sequestration is one purported mechanism contributing to chemotherapy resistance. This has often been demonstrated with the example of daunorubicin (DNR), chemotherapy with its characteristic red fluorescence used to trace it in cellular compartments. Here we addressed the question whether image analysis of DNR fluorescence can reflect its real intracellular distribution. We observed that the relationship between the intensity of the DNR fluorescence and its concentration in water solutions with or without proteins is far from linear. In contrast, nucleic acids, RNA and DNA in particular, dramatically diminish the DNR fluorescence, however, the intensity was proportional to the amount. Therefore, image analysis reflects the composition of different cell compartments (i.e., the presence of proteins and nucleic acids) rather than the actual concentration of DNR in these compartments. In line with these results, we observed highly fluorescent lysosomes and low fluorescent nucleus in sensitive cancer cells treated with low DNR concentrations, a fluorescence pattern thought to be found only in resistant cancer cells. Importantly, LC/MS/MS analysis of extracts from sensitive cells treated with DNR or DNR in combination with an inhibitor of vacuolar ATPase, concanamycin A, indicated that lysosomal accumulation of DNR increased with increasing extracellular concentration. However, even the highest lysosomal accumulation of DNR failed to reduce its extralysosomal concentration and thus change the cell sensitivity to the drug. In conclusion, our results strongly suggest that DNR fluorescence within cells does not indicate the real drug distribution. Further they suggested that lysosomal sequestration of DNR can hardly contribute to its resistance in cancer cells in vitro.


Sujet(s)
Daunorubicine/analyse , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Lysosomes/métabolisme , Lignée cellulaire tumorale , Chromatographie en phase liquide , Daunorubicine/pharmacocinétique , Antienzymes/pharmacologie , Humains , Microscopie de fluorescence , Spectrométrie de masse en tandem , Vacuolar Proton-Translocating ATPases/antagonistes et inhibiteurs
19.
Int J Mol Sci ; 21(12)2020 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-32575682

RÉSUMÉ

Resistance to chemotherapeutics and targeted drugs is one of the main problems in successful cancer therapy. Various mechanisms have been identified to contribute to drug resistance. One of those mechanisms is lysosome-mediated drug resistance. Lysosomes have been shown to trap certain hydrophobic weak base chemotherapeutics, as well as some tyrosine kinase inhibitors, thereby being sequestered away from their intracellular target site. Lysosomal sequestration is in most cases followed by the release of their content from the cell by exocytosis. Lysosomal accumulation of anticancer drugs is caused mainly by ion-trapping, but active transport of certain drugs into lysosomes was also described. Lysosomal low pH, which is necessary for ion-trapping is achieved by the activity of the V-ATPase. This sequestration can be successfully inhibited by lysosomotropic agents and V-ATPase inhibitors in experimental conditions. Clinical trials have been performed only with lysosomotropic drug chloroquine and their results were less successful. The aim of this review is to give an overview of lysosomal sequestration and expression of acidifying enzymes as yet not well known mechanism of cancer cell chemoresistance and about possibilities how to overcome this form of resistance.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Lysosomes/enzymologie , Tumeurs/enzymologie , Vacuolar Proton-Translocating ATPases/antagonistes et inhibiteurs , Antinéoplasiques/pharmacologie , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Exocytose , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Concentration en ions d'hydrogène , Lysosomes/effets des médicaments et des substances chimiques , Tumeurs/traitement médicamenteux
20.
Hypertens Res ; 43(10): 1079-1088, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-32382157

RÉSUMÉ

Vacuolar H+-adenosine triphosphatase (V-ATPase) stimulates vesicular acidification that may activate cytoplasmic enzymes, hormone secretion and membrane recycling of transporters. We investigated the effect of blockade of V-ATPase by bafilomycin B1 on renal gluconeogenesis, mitochondrial enzymes, and insulin secretion in type 2 diabetic rats. Spontaneous type 2 diabetic Torii rats were treated with intraperitoneal injection of bafilomycin B1 for 1 week, and the kidneys were examined after 24 h of starvation in metabolic cages. The renal expression and activity of V-ATPase were increased in the brush border membrane of the proximal tubules in diabetic rats. The blockade of V-ATPase by bafilomycin B1 reduced renal V-ATPase activity and urinary ammonium in diabetic rats. Treatment with bafilomycin suppressed the enhanced renal gluconeogenesis enzymes and mitochondrial electron transport enzymes in type 2 diabetic rats and reduced the renal cytoplasmic glucose levels. The insulin index and pancreatic insulin granules were decreased in diabetic rats with increased V-ATPase expression in islet cells, and treatment with bafilomycin B1 reversed these changes and increased the insulin secretion index. Hepatosteatosis in type 2 diabetic rats was ameliorated by bafilomycin treatment. As a consequence, treatment with bafilomycin B1 significantly decreased the plasma glucose level after 24 h of starvation in diabetic rats. In conclusion, a V-ATPase inhibitor improved plasma glucose levels in type 2 diabetes by inhibiting renal mitochondrial gluconeogenesis and improving insulin secretion.


Sujet(s)
Diabète expérimental/traitement médicamenteux , Diabète de type 2/traitement médicamenteux , Néoglucogenèse/effets des médicaments et des substances chimiques , Sécrétion d'insuline/effets des médicaments et des substances chimiques , Macrolides/usage thérapeutique , Animaux , Glycémie/effets des médicaments et des substances chimiques , Évaluation préclinique de médicament , Insulinorésistance , Rein/effets des médicaments et des substances chimiques , Rein/enzymologie , Métabolisme lipidique/effets des médicaments et des substances chimiques , Foie/effets des médicaments et des substances chimiques , Macrolides/pharmacologie , Mâle , Pancréas/effets des médicaments et des substances chimiques , Rats , Vacuolar Proton-Translocating ATPases/antagonistes et inhibiteurs , Vacuolar Proton-Translocating ATPases/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...