Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 5.076
Filtrer
1.
Nat Commun ; 15(1): 5689, 2024 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-38971796

RÉSUMÉ

Leukemia is a kind of hematological malignancy originating from bone marrow, which provides essential signals for initiation, progression, and recurrence of leukemia. However, how to specifically deliver drugs to the bone marrow remains elusive. Here, we develop biomimetic vesicles by infusing hematopoietic stem and progenitor cell (HSPC) membrane with liposomes (HSPC liposomes), which migrate to the bone marrow of leukemic mice via hyaluronic acid-CD44 axis. Moreover, the biomimetic vesicles exhibit superior binding affinity to leukemia cells through intercellular cell adhesion molecule-1 (ICAM-1)/integrin ß2 (ITGB2) interaction. Further experiments validate that the vesicles carrying chemotherapy drug cytarabine (Ara-C@HSPC-Lipo) markedly inhibit proliferation, induce apoptosis and differentiation of leukemia cells, and decrease number of leukemia stem cells. Mechanically, RNA-seq reveals that Ara-C@HSPC-Lipo treatment induces apoptosis and differentiation and inhibits the oncogenic pathways. Finally, we verify that HSPC liposomes are safe in mice. This study provides a method for targeting bone marrow and treating leukemia.


Sujet(s)
Apoptose , Moelle osseuse , Cytarabine , Systèmes de délivrance de médicaments , Cellules souches hématopoïétiques , Leucémies , Liposomes , Animaux , Cellules souches hématopoïétiques/effets des médicaments et des substances chimiques , Cellules souches hématopoïétiques/métabolisme , Souris , Cytarabine/pharmacologie , Moelle osseuse/effets des médicaments et des substances chimiques , Moelle osseuse/anatomopathologie , Moelle osseuse/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Leucémies/traitement médicamenteux , Leucémies/anatomopathologie , Humains , Différenciation cellulaire/effets des médicaments et des substances chimiques , Membrane cellulaire/métabolisme , Membrane cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Antigènes CD18/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Antigènes CD44/métabolisme , Acide hyaluronique/composition chimique , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/anatomopathologie , Cellules souches tumorales/métabolisme
2.
BMC Oral Health ; 24(1): 729, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38918827

RÉSUMÉ

BACKGROUND: Despite the better prognosis associated with human papillomavirus (HPV)-positive oropharyngeal squamous cell carcinoma (OPSCC), some patients experience relapse and succumb to the disease; thus, there is a need for biomarkers identifying these patients for intensified treatment. Leucine-rich repeats and immunoglobulin-like domain (LRIG) protein 1 is a negative regulator of receptor tyrosine kinase signaling and a positive prognostic factor in OPSCC. Studies indicate that LRIG1 interacts with the LIM domain 7 protein (LMO7), a stabilizer of adherence junctions. Its role in OPSCC has not been studied before. METHODS: A total of 145 patients diagnosed with OPSCC were enrolled. Immunohistochemical LMO7 expression and staining intensity were evaluated in the tumors and correlated with known clinical and pathological prognostic factors, such as HPV status and LRIG1, CD44, Ki67, and p53 expression. RESULTS: Our results show that high LMO7 expression is associated with significantly longer overall survival (OS) (p = 0.044). LMO7 was a positive prognostic factor for OS in univariate analysis (HR 0.515, 95% CI: 0.267-0.994, p = 0.048) but not in multivariate analysis. The LMO7 expression correlated with LRIG1 expression (p = 0.048), consistent with previous findings. Interestingly, strong LRIG1 staining intensity was an independent negative prognostic factor in the HPV-driven group of tumors (HR 2.847, 95% Cl: 1.036-7.825, p = 0.043). CONCLUSIONS: We show for the first time that high LMO7 expression is a positive prognostic factor in OPSCC, and we propose that LMO7 should be further explored as a biomarker. In contrast to previous reports, LRIG1 expression was shown to be an independent negative prognostic factor in HPV-driven OPSCC.


Sujet(s)
Marqueurs biologiques tumoraux , Carcinome épidermoïde , Protéines à domaine LIM , Tumeurs de l'oropharynx , Humains , Tumeurs de l'oropharynx/virologie , Tumeurs de l'oropharynx/métabolisme , Tumeurs de l'oropharynx/anatomopathologie , Tumeurs de l'oropharynx/mortalité , Mâle , Femelle , Adulte d'âge moyen , Pronostic , Protéines à domaine LIM/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/analyse , Carcinome épidermoïde/métabolisme , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/virologie , Sujet âgé , Facteurs de transcription/métabolisme , Glycoprotéines membranaires/métabolisme , Adulte , Antigène KI-67/métabolisme , Antigènes CD44/métabolisme , Antigènes CD44/analyse , Protéine p53 suppresseur de tumeur/métabolisme , Infections à papillomavirus/complications , Immunohistochimie , Sujet âgé de 80 ans ou plus , Taux de survie
3.
Front Immunol ; 15: 1418061, 2024.
Article de Anglais | MEDLINE | ID: mdl-38903499

RÉSUMÉ

Extracellular vesicles (EVs), characterized by low immunogenicity, high biocompatibility and targeting specificity along with excellent blood-brain barrier permeability, are increasingly recognized as promising drug delivery vehicles for treating a variety of diseases, such as cancer, inflammation and viral infection. However, recent findings demonstrate that the intracellular delivery efficiency of EVs fall short of expectations due to phagocytic clearance mediated by the host mononuclear phagocyte system through Fcγ receptors, complement receptors as well as non-opsonic phagocytic receptors. In this text, we investigate a range of bacterial virulence proteins that antagonize host phagocytic machinery, aiming to explore their potential in engineering EVs to counteract phagocytosis. Special emphasis is placed on IdeS secreted by Group A Streptococcus and ImpA secreted by Pseudomonas aeruginosa, as they not only counteract phagocytosis but also bind to highly upregulated surface biomarkers αVß3 on cancer cells or cleave the tumor growth and metastasis-promoting factor CD44, respectively. This suggests that bacterial anti-phagocytic proteins, after decorated onto EVs using pre-loading or post-loading strategies, can not only improve EV-based drug delivery efficiency by evading host phagocytosis and thus achieve better therapeutic outcomes but also further enable an innovative synergistic EV-based cancer therapy approach by integrating both phagocytosis antagonism and cancer targeting or deactivation.


Sujet(s)
Vésicules extracellulaires , Phagocytose , Vésicules extracellulaires/immunologie , Vésicules extracellulaires/métabolisme , Phagocytose/immunologie , Humains , Animaux , Protéines bactériennes/métabolisme , Protéines bactériennes/immunologie , Tumeurs/immunologie , Tumeurs/thérapie , Tumeurs/métabolisme , Intégrine alphaVbêta3/métabolisme , Intégrine alphaVbêta3/immunologie , Antigènes CD44/métabolisme , Antigènes CD44/immunologie , Pseudomonas aeruginosa/immunologie
4.
Sci Rep ; 14(1): 13749, 2024 06 14.
Article de Anglais | MEDLINE | ID: mdl-38877012

RÉSUMÉ

Prion diseases are fatal, infectious, neurodegenerative disorders resulting from accumulation of misfolded cellular prion protein in the brain. Early pathological changes during CNS prion disease also include reactive astrocyte activation with increased CD44 expression, microgliosis, as well as loss of dendritic spines and synapses. CD44 is a multifunctional cell surface adhesion and signalling molecule which is considered to play roles in astrocyte morphology and the maintenance of dendritic spine integrity and synaptic plasticity. However, the role of CD44 in prion disease was unknown. Here we used mice deficient in CD44 to determine the role of CD44 during prion disease. We show that CD44-deficient mice displayed no difference in their response to CNS prion infection when compared to wild type mice. Furthermore, the reactive astrocyte activation and microgliosis that accompanies CNS prion infection was unimpaired in the absence of CD44. Together, our data show that although CD44 expression is upregulated in reactive astrocytes during CNS prion disease, it is dispensable for astrocyte and microglial activation and the development of prion neuropathogenesis.


Sujet(s)
Astrocytes , Antigènes CD44 , Maladies à prions , Animaux , Astrocytes/métabolisme , Astrocytes/anatomopathologie , Antigènes CD44/métabolisme , Antigènes CD44/génétique , Maladies à prions/métabolisme , Maladies à prions/anatomopathologie , Maladies à prions/génétique , Souris , Souris knockout , Microglie/métabolisme , Microglie/anatomopathologie , Encéphale/métabolisme , Encéphale/anatomopathologie , Souris de lignée C57BL
5.
Int J Biol Macromol ; 273(Pt 1): 132671, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38823747

RÉSUMÉ

The delivery of chemotherapeutical drugs via nanomaterials has become a focus of pharmaceutical research over several decades due to improved drug delivery to cancer cells, decreased side effects on normal tissues, and increased therapeutic efficacy. Herein, a novel hyaluronic acid-conjugated methotrexate and 5-fluorouracil nanodrug system has been developed to address the critical limitations associated with the high toxicity and side effects of methotrexate and 5-fluorouracil. Furthermore, this nanodrug system enhances the targeting capacity of drug molecules and facilitates the potential integration of multimodal drug therapies. Concomitantly, the synergistic effects of MTX with 5-fluorouracil have been shown to improve the therapeutic index of MTX while attenuating the associated toxicities of MTX. The structure and micromorphology of the novel nanodrug can be confirmed by 1HNMR, FT-IR, UV-Vis, DLS, TEM, and AFM. Due to the ability of HA to bind to CD44 receptors activated on the surface of cancer cells and its enhanced permeability and retention (EPR) effect, the novel nanodrug we designed and synthesized can effectively target cancer cells. Cell counting Kit-8 (CCK8), flow cytometry, and live-dead staining assays in vitro showed that this nanodrug system had high targeting and antitumor activity against CD44 receptors. By using drugs to act on patient-derived colorectal, liver, and breast cancer organoids, the anticancer effect of the nanodrug was identified and verified. These results showed that the nanodrug system developed in this study may have great potential as a targeted therapy for cancer.


Sujet(s)
Fluorouracil , Acide hyaluronique , Méthotrexate , Méthotrexate/pharmacologie , Méthotrexate/composition chimique , Acide hyaluronique/composition chimique , Acide hyaluronique/pharmacologie , Fluorouracil/pharmacologie , Fluorouracil/composition chimique , Humains , Systèmes de délivrance de médicaments , Lignée cellulaire tumorale , Antigènes CD44/métabolisme , Vecteurs de médicaments/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique
6.
Nat Immunol ; 25(7): 1270-1282, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38877178

RÉSUMÉ

The relative and synergistic contributions of genetics and environment to interindividual immune response variation remain unclear, despite implications in evolutionary biology and medicine. Here we quantify interactive effects of genotype and environment on immune traits by investigating C57BL/6, 129S1 and PWK/PhJ inbred mice, rewilded in an outdoor enclosure and infected with the parasite Trichuris muris. Whereas cellular composition was shaped by interactions between genotype and environment, cytokine response heterogeneity including IFNγ concentrations was primarily driven by genotype with consequence on worm burden. In addition, we show that other traits, such as expression of CD44, were explained mostly by genetics on T cells, whereas expression of CD44 on B cells was explained more by environment across all strains. Notably, genetic differences under laboratory conditions were decreased following rewilding. These results indicate that nonheritable influences interact with genetic factors to shape immune variation and parasite burden.


Sujet(s)
Interaction entre gènes et environnement , Souris de lignée C57BL , Trichocéphalose , Trichuris , Animaux , Trichuris/immunologie , Trichocéphalose/immunologie , Trichocéphalose/parasitologie , Souris , Antigènes CD44/génétique , Antigènes CD44/métabolisme , Lymphocytes B/immunologie , Génotype , Interféron gamma/métabolisme , Lymphocytes T/immunologie , Femelle , Mâle
7.
Int J Biol Macromol ; 273(Pt 2): 133063, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38880443

RÉSUMÉ

The oral delivery of doxorubicin (DOX), an anti-cancer drug, encounters multiple hurdles such as limited gastrointestinal permeability, P-glycoprotein-mediated efflux, brief intestinal residence, and rapid degradation. This study introduced a novel approach utilizing hyaluronic acid (HA)-grafted fatty acid monoglycerides (HGD) to encapsulate DOX, forming HGD-DOX nanoparticles, aimed at enhancing its oral bioavailability. Drug encapsulated by HGD provided several advantages, including extended drug retention in the gastrointestinal tract, controlled release kinetics, and promotion of lymphatic absorption in the intestine. Additionally, HGD-DOX nanoparticles could specifically target CD44 receptors, potentially increasing therapeutic efficacy. The uptake mechanism of HGD-DOX nanoparticles primarily involved clathrin-mediated, caveolin-mediated and macropinocytosis endocytosis. Pharmacokinetic analysis further revealed that HGD significantly prolonged the in vivo residence time of DOX. In vivo imaging and pharmacodynamic studies indicated that HGD possessed tumor-targeting capabilities and exhibited a significant inhibitory effect on tumor growth, while maintaining an acceptable safety profile. Collectively, these findings position HGD-DOX nanoparticles as a promising strategy to boost the oral bioavailability of DOX, offering a potential avenue for improved cancer treatment.


Sujet(s)
Doxorubicine , Antigènes CD44 , Acide hyaluronique , Nanoparticules , Doxorubicine/administration et posologie , Doxorubicine/composition chimique , Doxorubicine/pharmacocinétique , Doxorubicine/pharmacologie , Acide hyaluronique/composition chimique , Animaux , Nanoparticules/composition chimique , Antigènes CD44/métabolisme , Humains , Administration par voie orale , Souris , Vecteurs de médicaments/composition chimique , Lignée cellulaire tumorale , Systèmes de délivrance de médicaments , Tests d'activité antitumorale sur modèle de xénogreffe
8.
Differentiation ; 138: 100789, 2024.
Article de Anglais | MEDLINE | ID: mdl-38896972

RÉSUMÉ

Osteoclast (OC) differentiation, vital for bone resorption, depends on osteoclast and precursor fusion. Osteoprotegerin (OPG) inhibits osteoclast differentiation. OPG's influence on fusion and mechanisms is unclear. Osteoclasts and precursors were treated with OPG alone or with ATP. OPG significantly reduced OC number, area and motility and ATP mitigated OPG's inhibition. However, OPG hardly affected the motility of precusors. OPG downregulated fusion-related molecules (CD44, CD47, DC-STAMP, ATP6V0D2) in osteoclasts, reducing only CD47 in precursors. OPG reduced Connexin43 phosphorylated forms (P1 and P2) in osteoclasts, affecting only P2 in precursors. OPG disrupted subcellular localization of CD44, CD47, DC-STAMP, ATP6V0D2, and Connexin43 in both cell types. Findings underscore OPG's multifaceted impact, inhibiting multinucleated osteoclast and mononuclear precursor fusion through distinct molecular mechanisms. Notably, ATP mitigates OPG's inhibitory effect, suggesting a potential regulatory role for the ATP signaling pathway. This study enhances understanding of intricate processes in osteoclast differentiation and fusion, offering insights into potential therapeutic targets for abnormal bone metabolism.


Sujet(s)
Adénosine triphosphate , Différenciation cellulaire , Ostéoclastes , Ostéoprotégérine , Ostéoprotégérine/métabolisme , Ostéoprotégérine/génétique , Ostéoclastes/métabolisme , Ostéoclastes/cytologie , Animaux , Adénosine triphosphate/métabolisme , Souris , Connexine 43/métabolisme , Connexine 43/génétique , Fusion cellulaire , Antigènes CD47/métabolisme , Antigènes CD47/génétique , Antigènes CD44/métabolisme , Antigènes CD44/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Résorption osseuse/métabolisme , Résorption osseuse/génétique , Résorption osseuse/anatomopathologie , Transduction du signal , Vacuolar Proton-Translocating ATPases/métabolisme , Vacuolar Proton-Translocating ATPases/génétique , Protéines de tissu nerveux
9.
Nanoscale ; 16(24): 11610-11622, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38855987

RÉSUMÉ

Nanomedicine aims to develop smart approaches for treating cancer and other diseases to improve patient survival and quality of life. Novel nanoparticles as nanodiamonds (NDs) represent promising candidates to overcome current limitations. In this study, NDs were functionalized with a 200 kDa hyaluronic acid-phospholipid conjugate (HA/DMPE), enhancing the stability of the nanoparticles in water-based solutions and selectivity for cancer cells overexpressing specific HA cluster determinant 44 (CD44) receptors. These nanoparticles were characterized by diffuse reflectance Fourier-transform infrared spectroscopy, Raman spectroscopy, and photoluminescence spectroscopy, confirming the efficacy of the functionalization process. Scanning electron microscopy was employed to evaluate the size distribution of the dry particles, while dynamic light scattering and zeta potential measurements were utilized to evaluate ND behavior in a water-based medium. Furthermore, the ND biocompatibility and uptake mediated by CD44 receptors in three different models of human adenocarcinoma cells were assessed by performing cytofluorimetric assay and confocal microscopy. HA-functionalized nanodiamonds demonstrated the advantage of active targeting in the presence of cancer cells expressing CD44 on the surface, suggesting higher drug delivery to tumors over non-tumor tissues. Even CD44-poorly expressing cancers could be targeted by the NDs, thanks to their good passive diffusion within cancer cells.


Sujet(s)
Antigènes CD44 , Acide hyaluronique , Nanodiamants , Humains , Nanodiamants/composition chimique , Acide hyaluronique/composition chimique , Antigènes CD44/métabolisme , Lignée cellulaire tumorale , Phospholipides/composition chimique , Imagerie optique , Tumeurs/imagerie diagnostique , Tumeurs/anatomopathologie , Tumeurs/métabolisme
10.
Nanoscale ; 16(24): 11762-11773, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38869001

RÉSUMÉ

Nanohydroxyapatite (nHAp) has attracted significant attention for its tumor suppression and tumor microenvironment modulation capabilities. However, a strong tendency to aggregate greatly affects its anti-tumor efficiency. To address this issue, a hydrogel platform consisting of thiolated hyaluronic acid (HA-SH) modified nanohydroxyapatite (nHAp-HA) and HA-SH was developed for sustained delivery of nHAp for melanoma therapy. The hydrophilic and negatively charged HA-SH significantly improved the size dispersion and stability of nHAp in aqueous media while conferring nHAp targeting effects. Covalent sulfhydryl self-cross-linking between HA-SH and nHAp-HA groups ensured homogeneous dispersion of nHAp in the matrix material. Meanwhile, the modification of HA-SH conferred the targeting properties of nHAp and enhanced cellular uptake through the HA/CD44 receptor. The hydrogel platform could effectively reduce the aggregation of nHAp and release nHAp in a sustained and orderly manner. Antitumor experiments showed that the modified nHAp-HA retained the tumor cytotoxicity of nHAp in vitro and inhibited the growth of highly malignant melanomas up to 78.6% while being able to induce the differentiation of macrophages to the M1 pro-inflammatory and antitumor phenotype. This study will broaden the application of nanohydroxyapatite in tumor therapy.


Sujet(s)
Durapatite , Acide hyaluronique , Hydrogels , Mélanome , Durapatite/composition chimique , Durapatite/pharmacologie , Acide hyaluronique/composition chimique , Acide hyaluronique/pharmacologie , Hydrogels/composition chimique , Hydrogels/pharmacologie , Animaux , Souris , Mélanome/traitement médicamenteux , Mélanome/anatomopathologie , Mélanome/métabolisme , Lignée cellulaire tumorale , Humains , Antigènes CD44/métabolisme , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Nanoparticules/composition chimique , Cellules RAW 264.7
11.
Int J Nanomedicine ; 19: 4893-4906, 2024.
Article de Anglais | MEDLINE | ID: mdl-38828202

RÉSUMÉ

Introduction: The tumor microenvironment (TME) has attracted considerable attention as a potential therapeutic target for cancer. High levels of reactive oxygen species (ROS) in the TME may act as a stimulus for drug release. In this study, we have developed ROS-responsive hyaluronic acid-bilirubin nanoparticles (HABN) loaded with doxorubicin (DOX@HABN) for the specific delivery and release of DOX in tumor tissue. The hyaluronic acid shell of the nanoparticles acts as an active targeting ligand that can specifically bind to CD44-overexpressing tumors. The bilirubin core has intrinsic anti-cancer activity and ROS-responsive solubility change properties. Methods & Results: DOX@HABN showed the HA shell-mediated targeting ability, ROS-responsive disruption leading to ROS-mediated drug release, and synergistic anti-cancer activity against ROS-overproducing CD44-overexpressing HeLa cells. Additionally, intravenously administered HABN-Cy5.5 showed remarkable tumor-targeting ability in HeLa tumor-bearing mice with limited distribution in major organs. Finally, intravenous injection of DOX@HABN into HeLa tumor-bearing mice showed synergistic anti-tumor efficacy without noticeable side effects. Conclusion: These findings suggest that DOX@HABN has significant potential as a cancer-targeting and TME ROS-responsive nanomedicine for targeted cancer treatment.


Sujet(s)
Bilirubine , Doxorubicine , Antigènes CD44 , Acide hyaluronique , Nanomédecine , Nanoparticules , Espèces réactives de l'oxygène , Microenvironnement tumoral , Acide hyaluronique/composition chimique , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Animaux , Espèces réactives de l'oxygène/métabolisme , Humains , Doxorubicine/pharmacologie , Doxorubicine/composition chimique , Doxorubicine/pharmacocinétique , Doxorubicine/administration et posologie , Nanoparticules/composition chimique , Souris , Cellules HeLa , Antigènes CD44/métabolisme , Bilirubine/composition chimique , Bilirubine/pharmacologie , Bilirubine/pharmacocinétique , Libération de médicament , Souris de lignée BALB C , Souris nude , Tests d'activité antitumorale sur modèle de xénogreffe , Vecteurs de médicaments/composition chimique , Vecteurs de médicaments/pharmacocinétique , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacocinétique , Antinéoplasiques/administration et posologie , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme
12.
J Vet Sci ; 25(3): e35, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38834505

RÉSUMÉ

IMPORTANCE: Experimental autoimmune encephalomyelitis (EAE) is an animal model of multiple sclerosis characterized by inflammation within the central nervous system. However, inflammation in non-neuronal tissues, including the lungs, has not been fully evaluated. OBJECTIVE: This study evaluated the inflammatory response in lungs of EAE mice by immunohistochemistry and histochemistry. METHODS: Eight adult C57BL/6 mice were injected with myelin oligodendrocyte glycoprotein35-55 to induce the EAE. Lungs and spinal cords were sampled from the experimental mice at the time of sacrifice and used for the western blotting, histochemistry, and immunohistochemistry. RESULTS: Histopathological examination revealed inflammatory lesions in the lungs of EAE mice, characterized by infiltration of myeloperoxidase (MPO)- and galectin-3-positive cells, as determined by immunohistochemistry. Increased numbers of collagen fibers in the lungs of EAE mice were confirmed by histopathological analysis. Western blotting revealed significantly elevated level of osteopontin (OPN), cluster of differentiation 44 (CD44), MPO and galectin-3 in the lungs of EAE mice compared with normal controls (p < 0.05). Immunohistochemical analysis revealed both OPN and CD44 in ionized calcium-binding adapter molecule 1-positive macrophages within the lungs of EAE mice. CONCLUSIONS AND RELEVANCE: Taken together, these findings suggest that the increased OPN level in lungs of EAE mice led to inflammation; concurrent increases in proinflammatory factors (OPN and galectin-3) caused pulmonary impairment.


Sujet(s)
Encéphalomyélite auto-immune expérimentale , Poumon , Souris de lignée C57BL , Animaux , Encéphalomyélite auto-immune expérimentale/anatomopathologie , Souris , Poumon/anatomopathologie , Femelle , Immunohistochimie , Ostéopontine/métabolisme , Galectine -3/métabolisme , Myeloperoxidase/métabolisme , Antigènes CD44/métabolisme , Moelle spinale/anatomopathologie , Inflammation/anatomopathologie , Technique de Western
13.
Sci Adv ; 10(23): eadl6083, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38838151

RÉSUMÉ

Hepatocellular carcinoma (HCC) acquires an immunosuppressive microenvironment, leading to unbeneficial therapeutic outcomes. Hyaluronan-mediated motility receptor (HMMR) plays a crucial role in tumor progression. Here, we found that aberrant expression of HMMR could be a predictive biomarker for the immune suppressive microenvironment of HCC, but the mechanism remains unclear. We established an HMMR-/- liver cancer mouse model to elucidate the HMMR-mediated mechanism of the dysregulated "don't eat me" signal. HMMR knockout inhibited liver cancer growth and induced phagocytosis. HMMRhigh liver cancer cells escaped from phagocytosis via sustaining CD47 signaling. Patients with HMMRhighCD47high expression showed a worse prognosis than those with HMMRlowCD47low expression. HMMR formed a complex with FAK/SRC in the cytoplasm to activate NF-κB signaling, which could be independent of membrane interaction with CD44. Notably, targeting HMMR could enhance anti-PD-1 treatment efficiency by recruiting CD8+ T cells. Overall, our data revealed a regulatory mechanism of the "don't eat me" signal and knockdown of HMMR for enhancing anti-PD-1 treatment.


Sujet(s)
Antigènes CD47 , Carcinome hépatocellulaire , Antigènes CD44 , Tumeurs du foie , Phagocytes , Phagocytose , Animaux , Humains , Souris , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/immunologie , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/génétique , Antigènes CD47/métabolisme , Antigènes CD47/génétique , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Lignée cellulaire tumorale , Focal adhesion kinase 1/métabolisme , Focal adhesion kinase 1/génétique , Antigènes CD44/métabolisme , Antigènes CD44/génétique , Échappement immunitaire , Tumeurs du foie/anatomopathologie , Tumeurs du foie/immunologie , Tumeurs du foie/métabolisme , Tumeurs du foie/génétique , Souris knockout , Facteur de transcription NF-kappa B/métabolisme , Phagocytes/métabolisme , Phagocytes/immunologie , Transduction du signal , Échappement de la tumeur à la surveillance immunitaire , Microenvironnement tumoral/immunologie
14.
Neoplasia ; 54: 101008, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38823209

RÉSUMÉ

Successful treatment of glioblastoma multiforme (GBM), an aggressive form of primary brain neoplasm, mandates the need to develop new therapeutic strategies. In this study, we investigated the potential of PBI-05204 in targeting GBM stem cells (GSCs) and the underlying mechanisms. Treatment with PBI-05204 significantly reduced both the number and size of tumor spheres derived from patient-derived GSCs (GBM9, GSC28 and TS543), and suppressed the tumorigenesis of GBM9 xenografts. Moreover, PBI-05204 treatment led to a significant decrease in the expression of CD44 and NANOG, crucial markers of progenitor stem cells, in GBM9 and GSC28 GSCs. This treatment also down-regulated GRP78 expression in both GSC types. Knocking down GRP78 expression through GRP78 siRNA transfection in GBM9 and GSC28 GSCs also resulted in reduced spheroid size and CD44 expression. Combining PBI-05204 with GRP78 siRNA further decreased spheroid numbers compared to GRP78 siRNA treatment alone. PBI-05204 treatment led to increased expression of pRIP1K and pRIP3K, along with enhanced binding of RIPK1/RIPK3 in GBM9 and GSC28 cells, resembling the effects observed in GRP78-silenced GSCs, suggesting that PBI-05204 induced necroptosis in these cells. Furthermore, oleandrin, a principle active cardiac glycoside component of PBI-05204, showed the ability to inhibit the self-renewal capacity in GSCs. These findings highlight the potential of PBI-05204 as a promising candidate for the development of novel therapies that target GBM stem cells.


Sujet(s)
Chaperonne BiP du réticulum endoplasmique , Glioblastome , Protéines du choc thermique , Cellules souches tumorales , Tests d'activité antitumorale sur modèle de xénogreffe , Humains , Glioblastome/anatomopathologie , Glioblastome/traitement médicamenteux , Glioblastome/métabolisme , Glioblastome/génétique , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Animaux , Souris , Protéines du choc thermique/métabolisme , Protéines du choc thermique/génétique , Lignée cellulaire tumorale , Extraits de plantes/pharmacologie , Nécroptose/effets des médicaments et des substances chimiques , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Antigènes CD44/métabolisme , Antigènes CD44/génétique
15.
Drug Deliv Transl Res ; 14(8): 2100-2111, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38709442

RÉSUMÉ

Biodegradable nanocarriers possess enormous potential for use as drug delivery systems that can accomplish controlled and targeted drug release, and a wide range of nanosystems have been reported for the treatment and/or diagnosis of various diseases and disorders. Of the various nanocarriers currently available, liposomes and polymer nanoparticles have been extensively studied and some formulations have already reached the market. However, a combination of properties to create a single hybrid system can give these carriers significant advantages, such as improvement in encapsulation efficacy, higher stability, and active targeting towards specific cells or tissues, over lipid or polymer-based platforms. To this aim, this work presents the formulation of poly(lactic-co-glycolic) acid (PLGA) nanoparticles in the presence of a hyaluronic acid (HA)-phospholipid conjugate (HA-DPPE), which was used to anchor HA onto the nanoparticle surface and therefore create an actively targeted hybrid nanosystem. Furthermore, ionic interactions have been proposed for drug encapsulation, leading us to select the free base form of pentamidine (PTM-B) as the model drug. We herein report the preparation of hybrid nanocarriers that were loaded via ion-pairing between the negatively charged PLGA and HA and the positively charged PTM-B, demonstrating an improved loading capacity compared to PLGA-based nanoparticles. The nanocarriers displayed a size of below 150 nm, a negative zeta potential of -35 mV, a core-shell internal arrangement and high encapsulation efficiency (90%). Finally, the ability to be taken up and exert preferential and receptor-mediated cytotoxicity on cancer cells that overexpress the HA specific receptor (CD44) has been evaluated. Competition assays supported the hypothesis that PLGA/HA-DPPE nanoparticles deliver their cargo within cells in a CD44-dependent manner.


Sujet(s)
Antigènes CD44 , Acide hyaluronique , Nanoparticules , Pentamidine , Copolymère d'acide poly(lactique-co-glycolique) , Humains , Acide hyaluronique/composition chimique , Copolymère d'acide poly(lactique-co-glycolique)/composition chimique , Antigènes CD44/métabolisme , Nanoparticules/composition chimique , Nanoparticules/administration et posologie , Pentamidine/composition chimique , Pentamidine/administration et posologie , Vecteurs de médicaments/composition chimique , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Libération de médicament , Lipides/composition chimique , Systèmes de délivrance de médicaments
16.
Genes (Basel) ; 15(5)2024 04 24.
Article de Anglais | MEDLINE | ID: mdl-38790166

RÉSUMÉ

Clear cell RCC (ccRCC) represents the most common type of kidney cancer, with surgery being the only potential curative treatment. Almost one-third of ccRCC patients relapse either locally or as cases of distant metastases. Several biomarkers have been employed in order to separate ccRCC patients with better prognosis or to predict treatment outcomes, with limited results. CD44 is a membrane glycoprotein with multiple roles in normal development but also cancer. Recently, the CD44 standard isoform has been implicated in tumor progression and the metastasis cascade through microenvironment interactions. Here, through CD44 immunohistochemical staining of ccRCC patient samples and TCGA data analysis, we sought to elucidate the expression patterns (mRNA and protein) of CD44 in clear cell RCC and correlate its expression with clinicopathological parameters. We were able to show that CD44 expression presents a positive association with tumor grade and overall survival, predicting a worse patient outcome in ccRCC. In addition, our data indicate that the CD44 mRNA upregulation can be attributed to reduced gene methylation, implicating epigenetic gene regulation in ccRCC development and progression.


Sujet(s)
Néphrocarcinome , Méthylation de l'ADN , Régulation de l'expression des gènes tumoraux , Antigènes CD44 , Tumeurs du rein , Humains , Néphrocarcinome/génétique , Néphrocarcinome/anatomopathologie , Néphrocarcinome/mortalité , Néphrocarcinome/métabolisme , Antigènes CD44/génétique , Antigènes CD44/métabolisme , Tumeurs du rein/génétique , Tumeurs du rein/anatomopathologie , Tumeurs du rein/mortalité , Tumeurs du rein/métabolisme , Femelle , Mâle , Adulte d'âge moyen , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Grading des tumeurs , Sujet âgé , Pronostic , Adulte
17.
Curr Med Sci ; 44(3): 503-511, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38748366

RÉSUMÉ

OBJECTIVE: This study aimed to examine the role of long non-coding RNA PCED1B antisense RNA 1 (PCED1B-AS1) in the development of hepatocellular carcinoma (HCC). METHODS: A total of 62 pairs of HCC tissues and adjacent non-tumor tissues were obtained from 62 HCC patients. The interactions of PCED1B-AS1 and microRNA-34a (miR-34a) were detected by dual luciferase activity assay and RNA pull-down assay. The RNA expression levels of PCED1B-AS1, miR-34a and CD44 were detected by RT-qPCR, and the protein expression level of CD44 was determined by Western blotting. The cell proliferation was detected by cell proliferation assay, and the cell invasion and migration by transwell invasion assay. The HCC tumor growth after PCED1B-AS1 was downregulated was determined by in vivo animal study. RESULTS: PCED1B-AS1 was highly expressed in HCC tissues, which was associated with poor survival of HCC patients. Furthermore, PCED1B-AS1 interacted with miR-34a in HCC cells, but they did not regulate the expression of each other. Additionally, PCED1B-AS1 increased the expression level of CD44, which was targeted by miR-34a. The cell proliferation and invasion assay revealed that miR-34a inhibited the proliferation and invasion of HCC in vitro, while CD44 exhibited the opposite effects. Furthermore, PCED1B-AS1 suppressed the role of miR-34a. Moreover, the knockdown of PCED1B-AS1 repressed the HCC tumor growth in nude mice in vivo. CONCLUSION: PCED1B-AS1 may play an oncogenic role by regulating the miR-34a/CD44 axis in HCC.


Sujet(s)
Carcinome hépatocellulaire , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Antigènes CD44 , Tumeurs du foie , microARN , Invasion tumorale , ARN long non codant , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , microARN/génétique , microARN/métabolisme , Humains , Antigènes CD44/génétique , Antigènes CD44/métabolisme , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Prolifération cellulaire/génétique , ARN long non codant/génétique , ARN long non codant/métabolisme , Animaux , Souris , Invasion tumorale/génétique , Mâle , Lignée cellulaire tumorale , Femelle , Mouvement cellulaire/génétique , Adulte d'âge moyen , Souris nude , ARN antisens/génétique
18.
J Endocrinol ; 262(1)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38692289

RÉSUMÉ

CD44, a cell surface adhesion receptor and stem cell biomarker, is recently implicated in chronic metabolic diseases. Ablation of CD44 ameliorates adipose tissue inflammation and insulin resistance in obesity. Here, we investigated cell type-specific CD44 expression in human and mouse adipose tissue and further studied how CD44 in preadipocytes regulates adipocyte function. Using Crispr Cas9-mdediated gene deletion and lentivirus-mediated gene re-expression, we discovered that deletion of CD44 promotes adipocyte differentiation and adipogenesis, whereas re-expression of CD44 abolishes this effect and decreases insulin responsiveness and adiponectin secretion in 3T3-L1 cells. Mechanistically, CD44 does so via suppressing Pparg expression. Using quantitative proteomics analysis, we further discovered that cell cycle-regulated pathways were mostly decreased by deletion of CD44. Indeed, re-expression of CD44 moderately restored expression of proteins involved in all phases of the cell cycle. These data were further supported by increased preadipocyte proliferation rates in CD44-deficient cells and re-expression of CD44 diminished this effect. Our data suggest that CD44 plays a crucial role in regulating adipogenesis and adipocyte function possibly through regulating PPARγ and cell cycle-related pathways. This study provides evidence for the first time that CD44 expressed in preadipocytes plays key roles in regulating adipocyte function outside immune cells where CD44 is primarily expressed. Therefore, targeting CD44 in (pre)adipocytes may provide therapeutic potential to treat obesity-associated metabolic complications.


Sujet(s)
Cellules 3T3-L1 , Adipocytes , Adipogenèse , Cycle cellulaire , Antigènes CD44 , Récepteur PPAR gamma , Adipogenèse/génétique , Adipogenèse/physiologie , Antigènes CD44/métabolisme , Antigènes CD44/génétique , Animaux , Récepteur PPAR gamma/métabolisme , Récepteur PPAR gamma/génétique , Souris , Cycle cellulaire/génétique , Cycle cellulaire/physiologie , Humains , Adipocytes/métabolisme , Délétion de gène , Différenciation cellulaire/génétique , Mâle , Tissu adipeux/métabolisme , Tissu adipeux/cytologie , Transduction du signal/physiologie
19.
Nat Commun ; 15(1): 3904, 2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38724502

RÉSUMÉ

Chronic wounds are a major complication in patients with diabetes. Here, we identify a therapeutic circRNA and load it into small extracellular vesicles (sEVs) to treat diabetic wounds in preclinical models. We show that circCDK13 can stimulate the proliferation and migration of human dermal fibroblasts and human epidermal keratinocytes by interacting with insulin-like growth factor 2 mRNA binding protein 3 in an N6-Methyladenosine-dependent manner to enhance CD44 and c-MYC expression. We engineered sEVs that overexpress circCDK13 and show that local subcutaneous injection into male db/db diabetic mouse wounds and wounds of streptozotocin-induced type I male diabetic rats could accelerate wound healing and skin appendage regeneration. Our study demonstrates that the delivery of circCDK13 in sEVs may present an option for diabetic wound treatment.


Sujet(s)
Diabète expérimental , Vésicules extracellulaires , Fibroblastes , Kératinocytes , ARN circulaire , Cicatrisation de plaie , Animaux , Humains , Mâle , Souris , Rats , Mouvement cellulaire , Prolifération cellulaire , Modèles animaux de maladie humaine , Vésicules extracellulaires/composition chimique , Fibroblastes/effets des médicaments et des substances chimiques , Antigènes CD44/métabolisme , Antigènes CD44/génétique , Kératinocytes/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , ARN circulaire/pharmacologie , ARN circulaire/usage thérapeutique , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Peau/effets des médicaments et des substances chimiques , Cicatrisation de plaie/effets des médicaments et des substances chimiques
20.
J Biomed Sci ; 31(1): 54, 2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38790021

RÉSUMÉ

BACKGROUND: Alcohol-related liver disease (ALD) is a major health concern worldwide, but effective therapeutics for ALD are still lacking. Tumor necrosis factor-inducible gene 6 protein (TSG-6), a cytokine released from mesenchymal stem cells, was shown to reduce liver fibrosis and promote successful liver repair in mice with chronically damaged livers. However, the effect of TSG-6 and the mechanism underlying its activity in ALD remain poorly understood. METHODS: To investigate its function in ALD mice with fibrosis, male mice chronically fed an ethanol (EtOH)-containing diet for 9 weeks were treated with TSG-6 (EtOH + TSG-6) or PBS (EtOH + Veh) for an additional 3 weeks. RESULTS: Severe hepatic injury in EtOH-treated mice was markedly decreased in TSG-6-treated mice fed EtOH. The EtOH + TSG-6 group had less fibrosis than the EtOH + Veh group. Activation of cluster of differentiation 44 (CD44) was reported to promote HSC activation. CD44 and nuclear CD44 intracellular domain (ICD), a CD44 activator which were upregulated in activated HSCs and ALD mice were significantly downregulated in TSG-6-exposed mice fed EtOH. TSG-6 interacted directly with the catalytic site of MMP14, a proteolytic enzyme that cleaves CD44, inhibited CD44 cleavage to CD44ICD, and reduced HSC activation and liver fibrosis in ALD mice. In addition, a novel peptide designed to include a region that binds to the catalytic site of MMP14 suppressed CD44 activation and attenuated alcohol-induced liver injury, including fibrosis, in mice. CONCLUSIONS: These results demonstrate that TSG-6 attenuates alcohol-induced liver damage and fibrosis by blocking CD44 cleavage to CD44ICD and suggest that TSG-6 and TSG-6-mimicking peptide could be used as therapeutics for ALD with fibrosis.


Sujet(s)
Molécules d'adhérence cellulaire , Antigènes CD44 , Cirrhose du foie , Maladies alcooliques du foie , Animaux , Mâle , Souris , Molécules d'adhérence cellulaire/administration et posologie , Éthanol , Antigènes CD44/métabolisme , Antigènes CD44/génétique , Cirrhose du foie/métabolisme , Cirrhose du foie/traitement médicamenteux , Cirrhose du foie/induit chimiquement , Maladies alcooliques du foie/métabolisme , Maladies alcooliques du foie/traitement médicamenteux , Souris de lignée C57BL , Peptides/pharmacologie , Peptides/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...