Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.519
Filtrer
2.
Biochem Biophys Res Commun ; 725: 150249, 2024 Sep 17.
Article de Anglais | MEDLINE | ID: mdl-38880081

RÉSUMÉ

The HIV-1 envelope glycoprotein (Env) plays crucial role in viral infection by facilitating viral attachment to host cells and inducing fusion of the virus with the host cell membrane. This fusion allows the HIV-1 viral genome to enter the target cell then triggering various stages of the viral life cycle. The native Env directly interacts with the main receptor CD4 and the co-receptor (CCR5 or CXCR4) in human cell membrane then induces membrane fusion. The elucidation of the structure of Env with CD4 and co-receptors in different HIV-1 subtypes is essential for the understanding of the mechanism of virus entry. Here we report the Cryo-EM structure of the CD4-bound HIV-1 heterotrimeric Env from Asia prevalent CRF07_BC CH119 strain. In this structure, the binding of three CD4 molecules with Env induced extensively conformational changes in gp120, resulting in the transformation of the Env from close state to intermediate open state. Additionally, the conformational shift of V1/V2 loops of the heterotrimeric Env allosterically expose the V3 loop and promoting the further interactions with co-receptor CCR5 or CXCR4. These findings not only illustrate the structural complexity and plasticity of HIV-1 Env but also give new insights how the biological trimeric Env initialize the immune recognition and membrane fusion.


Sujet(s)
Antigènes CD4 , Protéine d'enveloppe gp120 du VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/métabolisme , Humains , Antigènes CD4/métabolisme , Antigènes CD4/composition chimique , Protéine d'enveloppe gp120 du VIH/métabolisme , Protéine d'enveloppe gp120 du VIH/composition chimique , Protéine d'enveloppe gp120 du VIH/génétique , Cryomicroscopie électronique , Produits du gène env du virus de l'immunodéficience humaine/métabolisme , Produits du gène env du virus de l'immunodéficience humaine/composition chimique , Produits du gène env du virus de l'immunodéficience humaine/génétique , Récepteurs CCR5/métabolisme , Récepteurs CCR5/composition chimique , Liaison aux protéines , Modèles moléculaires , Conformation des protéines , Infections à VIH/virologie , Infections à VIH/métabolisme , Multimérisation de protéines , Récepteurs CXCR4/métabolisme , Récepteurs CXCR4/composition chimique , Asie
3.
Int J Immunopathol Pharmacol ; 38: 3946320241265265, 2024.
Article de Anglais | MEDLINE | ID: mdl-38889772

RÉSUMÉ

Introduction: Exceedingly high levels of the chemokine CCL5/RANTES have been found in fatty degenerated osteonecrotic alveolar bone cavities (FDOJ) and aseptic ischemic osteolysis of the jaw (AIOJ) from toothless regions. Because CCL5/RANTES seems to have a prominent role in creating the COVID-19 "cytokine storm", some researchers have used the monoclonal antibody Leronlimab to block the CCR5 on inflammatory cells.Objective: Is preexisting FDOJ/AIOJ jaw marrow pathology a "hidden" co-morbidity affecting some COVID-19 infections? To what extent does the chronic CCL5/RANTES expression from preexisting FDOJ/AIOJ areas contribute to the progression of the acute cytokine storm in COVID-19 patients?Methods: Authors report on reducing the COVID-19 "cytokine storm" by treating infected patients through targeting the chemokine receptor 5 (CCR5) with Leronlimab and interrupting the activation of CCR5 by high CCL5/RANTES signaling, thus dysregulating the inflammatory phase of the viremia. Surgical removal of FDOJ/AIOJ lesions with high CCL5/RANTES from patients with inflammatory diseases may be classified as a co-morbid disease.Results: Both multiplex analysis of 249 FDOJ/AIOJ bone tissue samples as well as serum levels of CCL5/RANTES displayed exceedingly high levels in both specimens.Discussion: By the results the authors hypothesize that chronic CCL5/RANTES induction from FDOJ/AIOJ areas may sensitize CCR5 throughout the immune system, thus, enabling it to amplify its response when confronted with the virus. As conventional intraoral radiography does little to assess the quality of the alveolar bone, ultrasonography units are available to help dentists locate the FDOJ/AIOJ lesions in an office setting.Conclusion: The authors propose a new approach to containment of the COVID-19 cytokine storm by a prophylactic focus for future viral-related pandemics, which may be early surgical clean-up of CCL5/RANTES expression sources in the FDOJ/AIOJ areas, thus diminishing a possible pre-sensitization of CCR5. A more complete dental examination includes trans-alveolar ultrasono-graphy (TAU) for hidden FDOJ/AIOJ lesions.


Sujet(s)
COVID-19 , Chimiokine CCL5 , Humains , COVID-19/immunologie , COVID-19/épidémiologie , Comorbidité , Mâle , Femelle , Adulte d'âge moyen , Récepteurs CCR5/métabolisme , Sujet âgé , Maladies de la mâchoire/épidémiologie , Maladies de la mâchoire/immunologie , SARS-CoV-2 , Syndrome de libération de cytokines , Anticorps monoclonaux humanisés/usage thérapeutique , Adulte
4.
BMC Neurol ; 24(1): 190, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38844862

RÉSUMÉ

BACKGROUND: Post-stroke depression (PSD) is a significant impediment to successful rehabilitation and recovery after a stroke. Current therapeutic options are limited, leaving an unmet demand for specific and effective therapeutic options. Our objective was to investigate the safety of Maraviroc, a CCR5 antagonist, as a possible mechanism-based add-on therapeutic option for PSD in an open-label proof-of-concept clinical trial. METHODS: We conducted a 10-week clinical trial in which ten patients with subcortical and cortical stroke, suffering from PSD. were administered a daily oral dose of 300 mg Maraviroc. Participants were then monitored for an additional eight weeks. The primary outcome measure was serious treatment-emergent adverse events (TEAEs) and TEAEs leading to discontinuation. The secondary outcome measure was a change in the Montgomery-Asberg Depression Rating Scale (MADRS). RESULTS: Maraviroc was well tolerated, with no reports of serious adverse events or discontinuations due to intolerance. The MADRS scores substantially reduced from baseline to week 10 (mean change: -16.4 ± 9.3; p < 0.001). By the conclusion of the treatment phase, a favorable response was observed in five patients, with four achieving remission. The time to response was relatively short, approximately three weeks. After the cessation of treatment, MADRS scores increased at week 18 by 6.1 ± 9.6 points (p = 0.014). CONCLUSIONS: Our proof-of-concept study suggests that a daily dosage of 300 mg of Maraviroc may represent a well-tolerated and potentially effective pharmacological approach to treating PSD. Further comprehensive placebo-controlled studies are needed to assess the impact of Maraviroc augmentation on PSD. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT05932550, Retrospectively registered: 28/06/2023.


Sujet(s)
Antagonistes des récepteurs CCR5 , Maraviroc , Étude de validation de principe , Accident vasculaire cérébral , Humains , Maraviroc/administration et posologie , Maraviroc/usage thérapeutique , Mâle , Femelle , Adulte d'âge moyen , Antagonistes des récepteurs CCR5/usage thérapeutique , Antagonistes des récepteurs CCR5/administration et posologie , Accident vasculaire cérébral/complications , Accident vasculaire cérébral/psychologie , Accident vasculaire cérébral/traitement médicamenteux , Sujet âgé , Dépression/traitement médicamenteux , Dépression/étiologie , Résultat thérapeutique , Triazoles/usage thérapeutique , Triazoles/administration et posologie , Adulte , Récepteurs CCR5/métabolisme
5.
J Cell Mol Med ; 28(12): e18489, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38899522

RÉSUMÉ

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


Sujet(s)
Vieillissement de la cellule , Chimiokine CCL5 , Progéniteurs endothéliaux , microARN , Néovascularisation physiologique , Chimiokine CCL5/métabolisme , Chimiokine CCL5/génétique , Progéniteurs endothéliaux/métabolisme , Progéniteurs endothéliaux/cytologie , Humains , microARN/génétique , microARN/métabolisme , Animaux , Néovascularisation physiologique/génétique , Souris , Prolifération cellulaire , Mâle , Récepteurs CCR5/métabolisme , Récepteurs CCR5/génétique , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Régulation négative/génétique , Ischémie/métabolisme , Ischémie/anatomopathologie , Ischémie/génétique , Transduction du signal ,
6.
Cell Rep ; 43(6): 114317, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38848213

RÉSUMÉ

Naive CD4+ T cells must differentiate in order to orchestrate immunity to Plasmodium, yet understanding of their emerging phenotypes, clonality, spatial distributions, and cellular interactions remains incomplete. Here, we observe that splenic polyclonal CD4+ T cells differentiate toward T helper 1 (Th1) and T follicular helper (Tfh)-like states and exhibit rarer phenotypes not elicited among T cell receptor (TCR) transgenic counterparts. TCR clones present at higher frequencies exhibit Th1 skewing, suggesting that variation in major histocompatibility complex class II (MHC-II) interaction influences proliferation and Th1 differentiation. To characterize CD4+ T cell interactions, we map splenic microarchitecture, cellular locations, and molecular interactions using spatial transcriptomics at near single-cell resolution. Tfh-like cells co-locate with stromal cells in B cell follicles, while Th1 cells in red pulp co-locate with activated monocytes expressing multiple chemokines and MHC-II. Spatial mapping of individual transcriptomes suggests that proximity to chemokine-expressing monocytes correlates with stronger effector phenotypes in Th1 cells. Finally, CRISPR-Cas9 gene disruption reveals a role for CCR5 in promoting clonal expansion and Th1 differentiation. A database of cellular locations and interactions is presented: https://haquelab.mdhs.unimelb.edu.au/spatial_gui/.


Sujet(s)
Lymphocytes T CD4+ , Différenciation cellulaire , Paludisme , Phénotype , Animaux , Paludisme/immunologie , Paludisme/parasitologie , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Souris , Souris de lignée C57BL , Lymphocytes auxiliaires Th1/immunologie , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs CCR5/métabolisme , Récepteurs CCR5/génétique , Rate/immunologie
7.
Int Immunopharmacol ; 136: 112264, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-38810308

RÉSUMÉ

BACKGROUND: Chemotaxis and trafficking of dendritic cells (DCs) induced by cytokine receptors are crucial steps in rheumatoid arthritis (RA) pathogenesis. C-C chemokine receptor type 5 (CCR5) plays a key role in DC movement and has been implicated in multitudinous inflammatory and immunology diseases. Thus, targeting CCR5 to suppress DC chemotaxis is considered as a potential strategy for the management of RA. METHODS: Herein, we first synthesized a new hybrid named CT3-1 which based on artesunate and isatin. Besides, we studied the regulating effectiveness of CT3-1 on bone marrow-derived DCs (BMDCs) and on collagen-induced arthritis (CIA) through RNA-seq analysis, cell function experiments in vitro and mice model in vivo. RESULTS: The results shown that CT3-1 mainly reduced CCR5 expression of immature BMDCs and importantly inhibited immature BMDC migration induced by CCR5 in vitro, with no or minor influence on other functions of DCs, such as phagocytosis and maturation. In the mouse model, CT3-1 relieved arthritis severity and inhibited CIA development. Furthermore, CT3-1 intervention decreased the expression of CCR5 in DCs and reduced the proportion of DCs in the peripheral blood of CIA mice. CONCLUSIONS: Our findings suggest that CCR5-induced chemotaxis and trafficking of immature DCs are important in RA. Targeting CCR5 and inhibiting immature DC chemotaxis may provide a novel choice for the treatment of RA and other similar autoimmune diseases. Moreover, we synthesized a new hybrid compound CT3-1 that could inhibit immature DC trafficking and effectively relieve RA by directly reducing the CCR5 expression of immature DCs.


Sujet(s)
Artésunate , Arthrite expérimentale , Polyarthrite rhumatoïde , Chimiotaxie , Cellules dendritiques , Récepteurs CCR5 , Animaux , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/immunologie , Récepteurs CCR5/métabolisme , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/immunologie , Chimiotaxie/effets des médicaments et des substances chimiques , Artésunate/pharmacologie , Artésunate/usage thérapeutique , Souris , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/immunologie , Souris de lignée DBA , Mâle , Cellules cultivées , Humains
8.
Microbiol Spectr ; 12(7): e0389523, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38809042

RÉSUMÉ

The susceptibility of genetically divergent HIV-1 strains (HIV-1 non-M) from groups O, N, and P to the CCR5 co-receptor antagonist, maraviroc (MVC), was investigated among a large panel of 45 clinical strains, representative of the viral genetic diversity. The results were compared to the reference strains of HIV-1 group M (HIV-1/M) with known tropism. Among the non-M strains, a wide range of phenotypic susceptibilities to MVC were observed. The large majority of HIV-1/O strains (40/42) displayed a high susceptibility to MVC, with median and mean IC50 values of 1.23 and 1.33 nM, respectively, similar to the HIV-1/M R5 strain (1.89 nM). However, the two remaining HIV-1/O strains exhibited a lower susceptibility (IC50 at 482 and 496 nM), in accordance with their dual/mixed (DM) tropism. Interestingly, the two HIV-1/N strains demonstrated varying susceptibility patterns, despite always having relatively low IC50 values (2.87 and 47.5 nM). This emphasized the complexity of determining susceptibility solely based on IC50 values. Our study examined the susceptibility of all HIV-1 non-M groups to MVC and correlated these findings with virus tropism (X4, R5, or DM). The results confirm the critical significance of tropism determination before initiating MVC treatment in patients infected with HIV-1 non-M. Furthermore, we advocate for the consideration of additional parameters, such as the slope of inhibition curves, to provide a more thorough characterization of phenotypic susceptibility profiles. IMPORTANCE: Unlike HIV-1 group M, the scarcity of studies on HIV-1 non-M groups (O, N, and P) presents challenges in understanding their susceptibility to antiretroviral treatments, particularly due to their natural resistance to non-nucleoside reverse transcriptase inhibitors. The TROPI-CO study logically complements our prior investigations into integrase inhibitors and anti-gp120 efficacy. The largest panel of 45 non-M strains existing so far yielded valuable results on maraviroc (MVC) susceptibility. The significant variations in MVC IC50 reveal a spectrum of susceptibilities, with most strains displaying R5 tropism. Notably, the absence of MVC-resistant strains suggests a potential therapeutic avenue. The study also employs a robust novel cell-based phenotropism assay and identifies distinct groups of susceptibilities based on inhibition curve slopes. Our findings emphasize the importance of determining tropism before initiating MVC and provide crucial insights for selecting effective therapeutic strategies in the delicate context of HIV-1 non-M infections.


Sujet(s)
Antagonistes des récepteurs CCR5 , Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Maraviroc , Tropisme viral , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Maraviroc/pharmacologie , Humains , Antagonistes des récepteurs CCR5/pharmacologie , Infections à VIH/virologie , Infections à VIH/traitement médicamenteux , Concentration inhibitrice 50 , Triazoles/pharmacologie , Phénotype , Tests de sensibilité microbienne , Récepteurs CCR5/métabolisme , Récepteurs CCR5/génétique , Agents antiVIH/pharmacologie , Cyclohexanes/pharmacologie , Résistance virale aux médicaments/génétique , Inhibiteurs de fusion du VIH/pharmacologie
9.
Neuropharmacology ; 254: 109981, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38704022

RÉSUMÉ

The chemotherapeutic agent Doxorubicin (DOX) is known to cause chemotherapy-induced cognitive impairment (CICI). Maraviroc, a potent C-C chemokine receptor 5 (CCR5) antagonist, shows neuroprotective properties, while its role in CICI remains unclear. This study determined the therapeutic potential of maraviroc on CICI. Adult C57BL/6J mice with implanted breast cancer cells received four weekly intraperitoneal injections of saline (Control group), 5 mg/kg DOX (DOX group), 10 mg/kg maraviroc (MVC group), or 5 mg/kg DOX with 10 mg/kg maraviroc (DOX + MVC group). The Morris Water Maze (MWM) was used for neurobehavioural test. Western blot analysis and immunofluorescence were used to evaluate the expressions of inflammatory markers, apoptosis-related proteins, and synaptic-related proteins. The volume and weight of tumor were also evaluated after treatments. DOX treatment significantly increased chemokines (CCL3, CCL4) and inflammatory cytokines (IL-1ß, TNF-α) in tumor-bearing mice hippocampus. While maraviroc administration reduced hippocampal proinflammatory factors compared to the DOX group. Furthermore, it also lowered apoptosis markers, restored synaptic proteins levels, and inhibited the NF-κB/NLRP3 pathway. Accordingly, maraviroc treatment significantly improved DOX-induced neurobehavioural impairments as evidenced by an increased number of platform crossings and percentage of target quadrant time in the MWM test. Additionally, when combined with DOX, maraviroc had additional inhibitory effects on tumor growth. These findings suggest that maraviroc can mitigate DOX-induced CICI by suppressing elevated proinflammatory chemokines and cytokines through the NF-κB/NLRP3 pathway, potentially offering an anti-tumor benefit. This research presents a promising therapeutic approach for DOX-induced CICI, enhancing the safety and efficacy of cancer treatments.


Sujet(s)
Antagonistes des récepteurs CCR5 , Doxorubicine , Maraviroc , Souris de lignée C57BL , Facteur de transcription NF-kappa B , Protéine-3 de la famille des NLR contenant un domaine pyrine , Transduction du signal , Animaux , Maraviroc/pharmacologie , Antagonistes des récepteurs CCR5/pharmacologie , Souris , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/antagonistes et inhibiteurs , Femelle , Facteur de transcription NF-kappa B/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Maladies neuro-inflammatoires/traitement médicamenteux , Maladies neuro-inflammatoires/induit chimiquement , Tumeurs du sein/traitement médicamenteux , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Antibiotiques antinéoplasiques/toxicité , Altération cognitive liée à la chimiothérapie/traitement médicamenteux , Récepteurs CCR5/métabolisme , Neuroprotecteurs/pharmacologie , Dysfonctionnement cognitif/traitement médicamenteux , Dysfonctionnement cognitif/induit chimiquement , Dysfonctionnement cognitif/métabolisme
10.
Cancer Cell ; 42(6): 1032-1050.e10, 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38759656

RÉSUMÉ

Total tumor clearance through immunotherapy is associated with a fully coordinated innate and adaptive immune response, but knowledge on the exact contribution of each immune cell subset is limited. We show that therapy-induced intratumoral CD8+ T cells recruited and skewed late-stage activated M1-like macrophages, which were critical for effective tumor control in two different murine models of cancer immunotherapy. The activated CD8+ T cells summon these macrophages into the tumor and their close vicinity via CCR5 signaling. Exposure of non-polarized macrophages to activated T cell supernatant and tumor lysate recapitulates the late-stage activated and tumoricidal phenotype in vitro. The transcriptomic signature of these macrophages is also detected in a similar macrophage population present in human tumors and coincides with clinical response to immune checkpoint inhibitors. The requirement of a functional co-operation between CD8+ T cells and effector macrophages for effective immunotherapy gives warning to combinations with broad macrophage-targeting strategies.


Sujet(s)
Lymphocytes T CD8+ , Immunothérapie , Macrophages , Animaux , Lymphocytes T CD8+/immunologie , Immunothérapie/méthodes , Souris , Humains , Macrophages/immunologie , Récepteurs CCR5/métabolisme , Récepteurs CCR5/génétique , Souris de lignée C57BL , Activation des macrophages/immunologie , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Activation des lymphocytes/immunologie , Femelle , Microenvironnement tumoral/immunologie
11.
PLoS Pathog ; 20(5): e1012211, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38709823

RÉSUMÉ

Cytolytic CD8+ T cells mediate immunopathology in cutaneous leishmaniasis without controlling parasites. Here, we identify factors involved in CD8+ T cell migration to the lesion that could be targeted to ameliorate disease severity. CCR5 was the most highly expressed chemokine receptor in patient lesions, and the high expression of CCL3 and CCL4, CCR5 ligands, was associated with delayed healing of lesions. To test the requirement for CCR5, Leishmania-infected Rag1-/- mice were reconstituted with CCR5-/- CD8+ T cells. We found that these mice developed smaller lesions accompanied by a reduction in CD8+ T cell numbers compared to controls. We confirmed these findings by showing that the inhibition of CCR5 with maraviroc, a selective inhibitor of CCR5, reduced lesion development without affecting the parasite burden. Together, these results reveal that CD8+ T cells migrate to leishmanial lesions in a CCR5-dependent manner and that blocking CCR5 prevents CD8+ T cell-mediated pathology.


Sujet(s)
Lymphocytes T CD8+ , Mouvement cellulaire , Leishmaniose cutanée , Récepteurs CCR5 , Animaux , Récepteurs CCR5/métabolisme , Récepteurs CCR5/immunologie , Lymphocytes T CD8+/immunologie , Souris , Humains , Leishmaniose cutanée/immunologie , Leishmaniose cutanée/parasitologie , Leishmaniose cutanée/anatomopathologie , Souris knockout , Souris de lignée C57BL , Antagonistes des récepteurs CCR5/pharmacologie , Maraviroc/pharmacologie , Femelle
12.
Int Immunopharmacol ; 135: 112331, 2024 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-38795597

RÉSUMÉ

CCR5 may be involved in the pathogenesis of asthma; however, the underlying mechanisms remain unclear. In comparison with a mild asthma model, subepithelial fibrosis was more severe and CCR5 gene expression in the lungs was significantly higher in our recently developed murine model of steroid-resistant severe asthma. Treatment with the CCR5 antagonist, maraviroc, significantly suppressed the development of subepithelial fibrosis in bronchi, whereas dexamethasone did not. On the other hand, increases in leukocytes related to type 2 inflammation, eosinophils, Th2 cells, and group 2 innate lymphoid cells in the lungs were not affected by the treatment with maraviroc. Increases in neutrophils and total macrophages were also not affected by the CCR5 antagonist. However, increases in transforming growth factor (TGF)-ß-producing interstitial macrophages (IMs) were significantly reduced by maraviroc. The present results confirmed increases in CCR5-expressing IMs in the lungs of the severe asthma model. In conclusion, CCR5 on IMs plays significant roles in the development of subepithelial fibrosis in severe asthma through TGF-ß production in the lungs.


Sujet(s)
Asthme , Antagonistes des récepteurs CCR5 , Macrophages , Maraviroc , Fibrose pulmonaire , Récepteurs CCR5 , Facteur de croissance transformant bêta , Animaux , Asthme/immunologie , Asthme/traitement médicamenteux , Asthme/anatomopathologie , Asthme/métabolisme , Récepteurs CCR5/métabolisme , Récepteurs CCR5/génétique , Maraviroc/pharmacologie , Maraviroc/usage thérapeutique , Antagonistes des récepteurs CCR5/pharmacologie , Antagonistes des récepteurs CCR5/usage thérapeutique , Macrophages/immunologie , Macrophages/effets des médicaments et des substances chimiques , Facteur de croissance transformant bêta/métabolisme , Fibrose pulmonaire/immunologie , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/traitement médicamenteux , Souris , Poumon/anatomopathologie , Poumon/immunologie , Poumon/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Modèles animaux de maladie humaine , Humains , Femelle
13.
J Immunother Cancer ; 12(5)2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38719543

RÉSUMÉ

The CCR/L5 axis is known for its role in immune regulation in a variety of settings and has been shown to have dichotomous functions in cancer, influencing both tumor progression and immune responses. Battaglin et al investigated its role using genomic and transcriptomic data from several datasets of patients with advanced colorectal cancer (CRC), including patients treated on CALGB/SWOG 80405, a trial of chemotherapy plus cetuximab versus bevacizumab, as well as a larger population of patients whose CRCs underwent commercially available Caris NGS and CODEai assays. These authors showed that CCR/L5 expression was both prognostic and predictive. They reported that low expression of the CCR/L5 axis was correlated with improved survival broadly, with particular benefit in patients treated with chemotherapy plus cetuximab. They demonstrated that high expression of CCR/L5 was associated with infiltration by negatively prognostic Tregs, M1 and M2 macrophages, myeloid-derived suppressor cells, and cancer-associated fibroblasts. They also showed that increased expression was correlated a wide variety of immune suppressive proteins, including PD-1, PD-L1, PD-L2, CTLA4, CD80, CD86, TIM3, IDO1, LAG3, and IFN-γ. This suggests mechanisms by which CRC resists anti-cancer immune responses. This study enhances our understanding of the role of the CCR/L5 axis in advanced CRC.


Sujet(s)
Chimiokine CCL5 , Tumeurs colorectales , Récepteurs CCR5 , Humains , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Tumeurs colorectales/génétique , Récepteurs CCR5/métabolisme , Récepteurs CCR5/génétique , Chimiokine CCL5/métabolisme , Chimiokine CCL5/génétique , Métastase tumorale
14.
Sci Rep ; 14(1): 10852, 2024 05 13.
Article de Anglais | MEDLINE | ID: mdl-38741006

RÉSUMÉ

Hematopoietic stem-cell (HSC) transplantation using a donor with a homozygous mutation in the HIV co-receptor CCR5 (CCR5Δ32/Δ32) holds great promise as a cure for HIV-1. Previously, there were three patients that had been reported to be completely cured from HIV infection by this approach. However, finding a naturally suitable Human Leukocyte Antigen (HLA)-matched homozygous CCR5Δ32 donor is very difficult. The prevalence of this allele is only 1% in the Caucasian population. Therefore, additional sources of CCR5Δ32/Δ32 HSCs are required. The Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated (Cas) system is one method to mediate CCR5 knockout in HSCs that has been successfully employed as a gene editing tool in clinical trials. Additional anti-HIV-1 strategies are still required for broad-spectrum inhibition of HIV-1 replication. Here in this study, we combined an additional anti-HIV-1 therapy, which is C46, a cell membrane-anchored HIV-1 fusion inhibitor with the CRISPR/Cas9 mediated knockout CCR5. The combined HIV-1 therapeutic genes were investigated for the potential prevention of both CCR5 (R5)- and CXCR4 (X4)-tropic HIV-1 infections in the MT4CCR5 cell line. The combinatorial CRISPR/Cas9 therapies were superior compared to single method therapy for achieving the HIV-1 cure strategy and shows potential for future applications.


Sujet(s)
Systèmes CRISPR-Cas , Édition de gène , Inhibiteurs de fusion du VIH , Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Récepteurs CCR5 , Récepteurs CCR5/génétique , Récepteurs CCR5/métabolisme , Édition de gène/méthodes , Humains , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Infections à VIH/génétique , Infections à VIH/virologie , Infections à VIH/thérapie , Inhibiteurs de fusion du VIH/pharmacologie , Lignée cellulaire , Réplication virale/effets des médicaments et des substances chimiques , Protéines de fusion recombinantes
15.
J Neuroinflammation ; 21(1): 136, 2024 May 27.
Article de Anglais | MEDLINE | ID: mdl-38802924

RÉSUMÉ

Autoimmune uveitis is a leading cause of severe vision loss, and animal models provide unique opportunities for studying its pathogenesis and therapeutic strategies. Here we employ scRNA-seq, RNA-seq and various molecular and cellular approaches to characterize mouse models of classical experimental autoimmune uveitis (EAU), revealing that EAU causes broad retinal neuron degeneration and marker downregulation, and that Müller glia may act as antigen-presenting cells. Moreover, EAU immune response is primarily driven by Th1 cells, and results in dramatic upregulation of CC chemokines, especially CCL5, in the EAU retina. Accordingly, overexpression of CCR5, a CCL5 receptor, in mesenchymal stem cells (MSCs) enhances their homing capacity and improves their immunomodulatory outcomes in preventing EAU, by reducing infiltrating T cells and activated microglia and suppressing Nlrp3 inflammasome activation. Taken together, our data not only provide valuable insights into the molecular characteristics of EAU but also open an avenue for innovative MSC-based therapy.


Sujet(s)
Cellules souches mésenchymateuses , Souris de lignée C57BL , Récepteurs CCR5 , Analyse sur cellule unique , Uvéite , Animaux , Souris , Cellules souches mésenchymateuses/métabolisme , Uvéite/immunologie , Récepteurs CCR5/métabolisme , Récepteurs CCR5/génétique , Maladies auto-immunes/thérapie , Analyse de profil d'expression de gènes , Modèles animaux de maladie humaine , Femelle , Analyse de l'expression du gène de la cellule unique
17.
Ageing Res Rev ; 96: 102286, 2024 04.
Article de Anglais | MEDLINE | ID: mdl-38561044

RÉSUMÉ

Chemokines and their corresponding receptors play crucial roles in orchestrating inflammatory and immune responses, particularly in the context of pathological conditions disrupting the internal environment. Among these receptors, CCR5 has garnered considerable attention due to its significant involvement in the inflammatory cascade, serving as a pivotal mediator of neuroinflammation and other inflammatory pathways associated with various diseases. However, a notable gap persists in comprehending the intricate mechanisms governing the interplay between CCR5 and its ligands across diverse and intricate inflammatory pathologies. Further exploration is warranted, especially concerning the inflammatory cascade instigated by immune cell infiltration and the precise binding sites within signaling pathways. This study aims to illuminate the regulatory axes modulating signaling pathways in inflammatory cells by providing a comprehensive overview of the pathogenic processes associated with CCR5 and its ligands across various disorders. The primary focus lies on investigating the pathomechanisms associated with CCR5 in disorders related to neuroinflammation, alongside the potential impact of aging on these processes and therapeutic interventions. The discourse culminates in addressing current challenges and envisaging potential future applications, advocating for innovative research endeavors to advance our comprehension of this realm.


Sujet(s)
Maladies neuro-inflammatoires , Récepteurs CCR5 , Humains , Récepteurs CCR5/métabolisme , Transduction du signal
18.
Eur J Immunol ; 54(7): e2350847, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38643381

RÉSUMÉ

Cenicriviroc, a dual CCR2/CCR5 antagonist, initially developed as an anti-HIV drug, has shown promising results in nonalcoholic steatohepatitis phase 2 clinical trials. It inhibits the infiltration and activation of CCR2+/CCR5+ monocytes and macrophages to the site of liver injury, preventing liver fibrosis. However, the role of Cenicriviroc in the modulation of helper T cell differentiation and functions remains to be explored. In inflamed colons of Crohn's disease patients, CCR2+ and CCR5+ CD4+ T cells are enriched. Considering the role of CCR2+ and CCR5+ T cells in IBD pathogenesis, we investigated the potential role of Cenicriviroc in colitis. Our in vitro studies revealed that Cenicriviroc inhibits Th1-, Th2-, and Th17-cell differentiation while promoting the generation of type 1 regulatory T cells (Tr1), known for preventing inflammation through induction of IL-10. This study is the first to report that Cenicriviroc promotes Tr1 cell generation by up-regulating the signature of Tr1 cell transcription factors such as c-Maf, Prdm1, Irf-1, Batf, and EGR-2. Cenicriviroc displayed a protective effect in experimental colitis models by preventing body weight loss and intestinal inflammation and preserving epithelial barrier integrity. We show that Cenicriviroc induced IL-10 and inhibited the generation of pro-inflammatory cytokines IFN-γ, IL-17, IL-6, and IL-1ß during colitis. Based on our data, we propose Cenicriviroc as a potential therapeutic in controlling tissue inflammation by inhibiting the generation and functions of effector T cells and promoting the induction of anti-inflammatory Tr1 cells.


Sujet(s)
Antagonistes des récepteurs CCR5 , Différenciation cellulaire , Colite , Récepteurs CCR2 , Récepteurs CCR5 , Lymphocytes T régulateurs , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Animaux , Souris , Récepteurs CCR2/métabolisme , Récepteurs CCR2/antagonistes et inhibiteurs , Colite/immunologie , Colite/traitement médicamenteux , Colite/induit chimiquement , Différenciation cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/immunologie , Antagonistes des récepteurs CCR5/pharmacologie , Antagonistes des récepteurs CCR5/usage thérapeutique , Récepteurs CCR5/métabolisme , Humains , Cellules Th17/immunologie , Cellules Th17/effets des médicaments et des substances chimiques , Sulfoxydes/pharmacologie , Souris de lignée C57BL , Lymphocytes auxiliaires Th1/immunologie , Lymphocytes auxiliaires Th1/effets des médicaments et des substances chimiques , Interleukine-10/métabolisme , Lymphocytes auxiliaires Th2/immunologie , Imidazoles
19.
Eur Rev Med Pharmacol Sci ; 28(6): 2430-2463, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38567606

RÉSUMÉ

Human Immunodeficiency Virus (HIV) has continuously been the greatest epidemic for humanity over a period spanning almost five decades. With no specific cure or treatment available to date despite extensive research, the C-C Chemokine Receptor 5, Delta 32 (CCR5 Δ32) allele genetic point mutation plays an imperative role in the prevention of acquired immunodeficiency syndrome (AIDS). This comprehensive study aims to review the induction of the homozygous recessive deletion genotype using the Clustered Regularly Interspaced Short Palindromic Repeats, Cas 9 Enzyme (CRISPR-Cas9), and hematopoietic stem cell transplantation under positive selection pressure for active immunity in seropositive patients' populations as the phenotype. A methodology is proposed to trigger a significant increase in the expression of Delta 32 beneficial mutant alleles within controlled modern healthcare facilities utilizing totipotent stem cells through somatic gene therapy. It acts upon two dysfunctional CCR5 genes, translating mutant G protein-coupled co-receptors, whose primary function is similar to that of C-X-C Motif Chemokine receptor 4 (CXCR4), by blocking the entry of viral RNA into the CD4+ T helper lymphocytes, halting infection and seizing viral life cycle. This modification is endemic in Northern Europe, where it naturally pertains to the Caucasian descent population samples in the form of polymorphism, p (X=0.01), where X is the probability of frequency of complete immunity against HIV-1 in population samples. The epigenetics of the single nucleotide polymorphism (SNP) are analyzed as they play a significant role in immunity distribution. Furthermore, a comparative analysis within the ethical boundaries of CRISPR-Cas9 is conducted to discuss the practical aspects and challenges of the presented methodologies and treatment alternatives. Additionally, the study assembles all available data and summarizes preexisting research while providing a promising solution to this ethical dilemma. Finally, a methodology is devised to answer the question of whether the variant-specific epidemic of AIDS caused by HIV-1 can be cured via artificially inducing immunity by CRISPR-Cas9.


Sujet(s)
Syndrome d'immunodéficience acquise , Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Humains , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Syndrome d'immunodéficience acquise/génétique , Syndrome d'immunodéficience acquise/thérapie , Infections à VIH/génétique , Infections à VIH/thérapie , Systèmes CRISPR-Cas/génétique , Récepteurs CCR5/génétique , Récepteurs CCR5/métabolisme , Mutation , Thérapie génétique , Polymorphisme de nucléotide simple , Fréquence d'allèle
20.
Viruses ; 16(4)2024 03 27.
Article de Anglais | MEDLINE | ID: mdl-38675853

RÉSUMÉ

HIV-1 typically infects cells via the CD4 receptor and CCR5 or CXCR4 co-receptors. Maraviroc is a CCR5-specific viral entry inhibitor; knowledge of viral co-receptor specificity is important prior to usage. We developed and validated an economical V3-env Illumina-based assay to detect and quantify the frequency of viruses utilizing each co-receptor. Plasma from 54 HIV+ participants (subtype B) was tested. The viral template cDNA was generated from plasma RNA with unique molecular identifiers (UMIs). The sequences were aligned and collapsed by the UMIs with a custom bioinformatics pipeline. Co-receptor usage, determined by codon analysis and online phenotype predictors PSSM and Geno2pheno, were compared to existing Trofile® data. The cost of V3-UMI was tallied. The sequences interpreted by Geno2pheno using the most conservative cut-off, a 2% false-positive-rate (FPR), predicted CXCR4 usage with the greatest sensitivity (76%) and specificity (100%); PSSM and codon analysis had similar sensitivity and lower specificity. Discordant Trofile® and genotypic results were more common when participants had specimens from different dates analyzed by either assay. V3-UMI reagents cost USD$62/specimen. A batch of ≤20 specimens required 5 h of technical time across 1.5 days. V3-UMI predicts HIV tropism at a sensitivity and specificity similar to those of Trofile®, is relatively inexpensive, and could be performed by most central laboratories. The adoption of V3-UMI could expand HIV drug therapeutic options in lower-resource settings that currently do not have access to phenotypic HIV tropism testing.


Sujet(s)
Techniques de génotypage , Récepteurs CCR5 , Récepteurs CXCR4 , Humains , Mâle , Génotype , Techniques de génotypage/méthodes , Infections à VIH/virologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Récepteurs CCR5/métabolisme , Récepteurs CCR5/génétique , Récepteurs CXCR4/génétique , Récepteurs CXCR4/métabolisme , ARN viral/génétique , Sensibilité et spécificité , Tropisme viral
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...