Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.524
Filter
1.
Front Cell Infect Microbiol ; 14: 1415123, 2024.
Article in English | MEDLINE | ID: mdl-38994006

ABSTRACT

The high proportion of AIDS cases and mortality rates in Guangxi underscores the urgency to investigate the influence of HIV-1 genetic diversity on disease progression in this region. Newly diagnosed HIV-1 patients were enrolled from January 2016 to December 2021, and the follow-up work and detection of CD4+T lymphocytes were carried out every six months until December 2022. Multivariate logistic regression was used to analyze the factors affecting pre-treatment CD4+T lymphocyte counts, while local weighted regression models (LOESS) and generalized estimating equation models (GEE) were conducted to assess factors influencing CD4+T Lymphocyte Recovery. Cox regression analysis was utilized to examine the impact of subtypes on survival risk. Additionally, HIV-1 env sequences were utilized for predicting CXCR4 and CCR5 receptors. The study encompassed 1867 individuals with pol sequences and 281 with env sequences. Our findings indicate that age over 30, divorced/widowed, peasant, heterosexual infection, CRF01_AE, long-term infection, and Pre-treatment Viral load >10000 copies/ml were factors associated with higher risk for pre-treatment CD4+T lymphocyte decline. Specifically, male gender, age over 30, heterosexual infection (HETs), long-term infection, CRF01_AE, and Pre-treatment CD4 T cell counts below 350/µL were identified as risk factors impeding CD4+T lymphocyte recovery. Pre-treatment CD4+T lymphocyte counts and recovery in individuals infected with CRF01_AE were lower compared to CRF07_BC and CRF55_01B. Additionally, CRF01_AE and CRF08_BC subtypes exhibited higher mortality rates than CRF07_BC, CRF55_01B, and other subtypes. Notably, CRF01_AE demonstrated the highest percentage of CXCR4 affinity ratios. This research unveils the intricate influence of HIV-1 gene diversity on CD4+T lymphocyte dynamics and clinical outcomes. It highlights the multifaceted nature of HIV infection in Guangxi, providing novel insights into subtype-specific disease progression among HIV-infected individuals in this region.


Subject(s)
Disease Progression , Genetic Variation , HIV Infections , HIV-1 , Viral Load , Humans , HIV-1/genetics , Male , Female , Adult , China/epidemiology , HIV Infections/virology , Prospective Studies , CD4 Lymphocyte Count , Middle Aged , Receptors, CCR5/genetics , Receptors, CCR5/metabolism , Receptors, CXCR4/genetics , Young Adult , CD4-Positive T-Lymphocytes/immunology , Risk Factors
2.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(6): 1098-1108, 2024 Jun 20.
Article in Chinese | MEDLINE | ID: mdl-38977339

ABSTRACT

OBJECTIVE: To identify the biomarkers for early rheumatoid arthritis (RA) diagnosis and explore the possible immune regulatory mechanisms. METHODS: The differentially expressed genesin RA were screened and functionally annotated using the limma, RRA, batch correction, and clusterProfiler. The protein-protein interaction network was retrieved from the STRING database, and Cytoscape 3.8.0 and GeneMANIA were used to select the key genes and predicting their interaction mechanisms. ROC curves was used to validate the accuracy of diagnostic models based on the key genes. The disease-specific immune cells were selected via machine learning, and their correlation with the key genes were analyzed using Corrplot package. Biological functions of the key genes were explored using GSEA method. The expression of STAT1 was investigated in the synovial tissue of rats with collagen-induced arthritis (CIA). RESULTS: We identified 9 core key genes in RA (CD3G, CD8A, SYK, LCK, IL2RG, STAT1, CCR5, ITGB2, and ITGAL), which regulate synovial inflammation primarily through cytokines-related pathways. ROC curve analysis showed a high predictive accuracy of the 9 core genes, among which STAT1 had the highest AUC (0.909). Correlation analysis revealed strong correlations of CD3G, ITGAL, LCK, CD8A, and STAT1 with disease-specific immune cells, and STAT1 showed the strongest correlation with M1-type macrophages (R=0.68, P=2.9e-08). The synovial tissues of the ankle joints of CIA rats showed high expressions of STAT1 and p-STAT1 with significant differential expression of STAT1 between the nucleus and the cytoplasm of the synovial fibroblasts. The protein expressions of p-STAT1 and STAT1 in the cell nuclei were significantly reduced after treatment. CONCLUSION: CD3G, CD8A, SYK, LCK, IL2RG, STAT1, CCR5, ITGB2, and ITGAL may serve as biomarkers for early diagnosis of RA. Gene-immune cell pathways such as CD3G/CD8A/LCK-γδ T cells, ITGAL-Tfh cells, and STAT1-M1-type macrophages may be closely related with the development of RA.


Subject(s)
Arthritis, Rheumatoid , Biomarkers , Protein Interaction Maps , STAT1 Transcription Factor , Synovial Membrane , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Animals , Rats , STAT1 Transcription Factor/metabolism , Biomarkers/metabolism , Synovial Membrane/metabolism , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Gene Expression Profiling , Databases, Genetic , Humans , CD8 Antigens/metabolism , Receptors, CCR5/metabolism , Receptors, CCR5/genetics , Syk Kinase/metabolism , Syk Kinase/genetics , ROC Curve
3.
Int J Mol Sci ; 25(13)2024 Jul 05.
Article in English | MEDLINE | ID: mdl-39000516

ABSTRACT

The ligands of chemokine receptors 2 and 5 (CCR2 and CCR5, respectively) are associated with the pathomechanism of neuropathic pain development, but their role in painful diabetic neuropathy remains unclear. Therefore, the aim of our study was to examine the function of these factors in the hypersensitivity accompanying diabetes. Additionally, we analyzed the analgesic effect of cenicriviroc (CVC), a dual CCR2/CCR5 antagonist, and its influence on the effectiveness of morphine. An increasing number of experimental studies have shown that targeting more than one molecular target is advantageous compared with the coadministration of individual pharmacophores in terms of their analgesic effect. The advantage of using bifunctional compounds is that they gain simultaneous access to two receptors at the same dose, positively affecting their pharmacokinetics and pharmacodynamics and consequently leading to improved analgesia. Experiments were performed on male and female Swiss albino mice with a streptozotocin (STZ, 200 mg/kg, i.p.) model of diabetic neuropathy. We found that the blood glucose level increased, and the mechanical and thermal hypersensitivity developed on the 7th day after STZ administration. In male mice, we observed increased mRNA levels of Ccl2, Ccl5, and Ccl7, while in female mice, we observed additional increases in Ccl8 and Ccl12 levels. We have demonstrated for the first time that a single administration of cenicriviroc relieves pain to a similar extent in male and female mice. Moreover, repeated coadministration of cenicriviroc with morphine delays the development of opioid tolerance, while the best and longest-lasting analgesic effect is achieved by repeated administration of cenicriviroc alone, which reduces pain hypersensitivity in STZ-exposed mice, and unlike morphine, no tolerance to the analgesic effects of CVC is observed until Day 15 of treatment. Based on these results, we suggest that targeting CCR2 and CCR5 with CVC is a potent therapeutic option for novel pain treatments in diabetic neuropathy patients.


Subject(s)
CCR5 Receptor Antagonists , Diabetic Neuropathies , Disease Models, Animal , Receptors, CCR2 , Receptors, CCR5 , Animals , Mice , Diabetic Neuropathies/drug therapy , Male , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR2/metabolism , Female , Receptors, CCR5/metabolism , Receptors, CCR5/genetics , CCR5 Receptor Antagonists/pharmacology , CCR5 Receptor Antagonists/therapeutic use , Morphine/pharmacology , Morphine/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/complications , Analgesics/pharmacology , Analgesics/therapeutic use , Hyperalgesia/drug therapy , Imidazoles , Sulfoxides
4.
Front Cell Infect Microbiol ; 14: 1408245, 2024.
Article in English | MEDLINE | ID: mdl-39006742

ABSTRACT

While simian immunodeficiency virus (SIV) infection is non-pathogenic in naturally infected African nonhuman primate hosts, experimental or accidental infection in rhesus macaques often leads to AIDS. Baboons, widely distributed throughout Africa, do not naturally harbor SIV, and experimental infection of baboons with SIVmac results in transient low-level viral replication. Elucidation of mechanisms of natural immunity in baboons could uncover new targets of antiviral intervention. We tested the hypothesis that an SIVmac adapted to replicate in baboon primary cells will gain the capacity to establish chronic infections in vivo. Here, we generated SIVmac variants in baboon cells through serial passage in PBMC from different donors (SIVbn-PBMC s1), in PBMC from the same donors (SIVbn-PBMC s2), or in isolated CD4 cells from the same donors used for series 2 (SIVbn-CD4). While SIVbn-PBMC s1 and SIVbn-CD4 demonstrated increased replication capacity, SIVbn-PBMC s2 did not. Pharmacological blockade of CCR5 revealed SIVbn-PBMC s1 could more efficiently use available CCR5 than SIVmac, a trait we hypothesize arose to circumvent receptor occupation by chemokines. Sequencing analysis showed that all three viruses accumulated different types of mutations, and that more non-synonymous mutations became fixed in SIVbn-PBMC s1 than SIVbn-PBMC s2 and SIVbn-CD4, supporting the notion of stronger fitness pressure in PBMC from different genetic backgrounds. Testing the individual contribution of several newly fixed SIV mutations suggested that is the additive effect of these mutations in SIVbn-PBMC s1 that contributed to its enhanced fitness, as recombinant single mutant viruses showed no difference in replication capacity over the parental SIVmac239 strain. The replicative capacity of SIVbn-PBMC passage 4 (P4) s1 was tested in vivo by infecting baboons intravenously with SIVbn-PBMC P4 s1 or SIVmac251. While animals infected with SIVmac251 showed the known pattern of transient low-level viremia, animals infected with SIVbn-PBMC P4 s1 had undetectable viremia or viral DNA in lymphoid tissue. These studies suggest that adaptation of SIV to grow in baboon primary cells results in mutations that confer increased replicative capacity in the artificial environment of cell culture but make the virus unable to avoid the restrictive factors generated by a complex multicellular organism.


Subject(s)
Papio , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Virus Replication , Animals , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/physiology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Acquired Immunodeficiency Syndrome/immunology , Leukocytes, Mononuclear/virology , Leukocytes, Mononuclear/immunology , Receptors, CCR5/metabolism , Receptors, CCR5/genetics , CD4-Positive T-Lymphocytes/virology , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Serial Passage
5.
Int J Immunopathol Pharmacol ; 38: 3946320241265265, 2024.
Article in English | MEDLINE | ID: mdl-38889772

ABSTRACT

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.


Subject(s)
COVID-19 , Chemokine CCL5 , Humans , COVID-19/immunology , COVID-19/epidemiology , Comorbidity , Male , Female , Middle Aged , Receptors, CCR5/metabolism , Aged , Jaw Diseases/epidemiology , Jaw Diseases/immunology , SARS-CoV-2 , Cytokine Release Syndrome , Antibodies, Monoclonal, Humanized/therapeutic use , Adult
7.
Biochem Biophys Res Commun ; 725: 150249, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-38880081

ABSTRACT

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.


Subject(s)
CD4 Antigens , HIV Envelope Protein gp120 , HIV-1 , HIV-1/metabolism , Humans , CD4 Antigens/metabolism , CD4 Antigens/chemistry , HIV Envelope Protein gp120/metabolism , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/genetics , Cryoelectron Microscopy , env Gene Products, Human Immunodeficiency Virus/metabolism , env Gene Products, Human Immunodeficiency Virus/chemistry , env Gene Products, Human Immunodeficiency Virus/genetics , Receptors, CCR5/metabolism , Receptors, CCR5/chemistry , Protein Binding , Models, Molecular , Protein Conformation , HIV Infections/virology , HIV Infections/metabolism , Protein Multimerization , Receptors, CXCR4/metabolism , Receptors, CXCR4/chemistry , Asia
8.
J Cell Mol Med ; 28(12): e18489, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38899522

ABSTRACT

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.


Subject(s)
Cellular Senescence , Chemokine CCL5 , Endothelial Progenitor Cells , MicroRNAs , Neovascularization, Physiologic , Animals , Humans , Male , Mice , Angiogenesis , Cell Proliferation , Chemokine CCL5/metabolism , Chemokine CCL5/genetics , Down-Regulation/genetics , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/cytology , Ischemia/metabolism , Ischemia/pathology , Ischemia/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Neovascularization, Physiologic/genetics , Receptors, CCR5/metabolism , Receptors, CCR5/genetics , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics
9.
BMC Neurol ; 24(1): 190, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38844862

ABSTRACT

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.


Subject(s)
CCR5 Receptor Antagonists , Maraviroc , Proof of Concept Study , Stroke , Humans , Maraviroc/administration & dosage , Maraviroc/therapeutic use , Male , Female , Middle Aged , CCR5 Receptor Antagonists/therapeutic use , CCR5 Receptor Antagonists/administration & dosage , Stroke/complications , Stroke/psychology , Stroke/drug therapy , Aged , Depression/drug therapy , Depression/etiology , Treatment Outcome , Triazoles/therapeutic use , Triazoles/administration & dosage , Adult , Receptors, CCR5/metabolism
10.
Cell Rep ; 43(6): 114317, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38848213

ABSTRACT

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/.


Subject(s)
CD4-Positive T-Lymphocytes , Cell Differentiation , Malaria , Phenotype , Animals , Malaria/immunology , Malaria/parasitology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Th1 Cells/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, CCR5/metabolism , Receptors, CCR5/genetics , Spleen/immunology
11.
Int Immunopharmacol ; 136: 112264, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38810308

ABSTRACT

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.


Subject(s)
Artesunate , Arthritis, Experimental , Arthritis, Rheumatoid , Chemotaxis , Dendritic Cells , Receptors, CCR5 , Animals , Dendritic Cells/drug effects , Dendritic Cells/immunology , Receptors, CCR5/metabolism , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Chemotaxis/drug effects , Artesunate/pharmacology , Artesunate/therapeutic use , Mice , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Mice, Inbred DBA , Male , Cells, Cultured , Humans
12.
Cancer Cell ; 42(6): 1032-1050.e10, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38759656

ABSTRACT

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.


Subject(s)
CD8-Positive T-Lymphocytes , Immunotherapy , Macrophages , Animals , CD8-Positive T-Lymphocytes/immunology , Immunotherapy/methods , Mice , Humans , Macrophages/immunology , Receptors, CCR5/metabolism , Receptors, CCR5/genetics , Mice, Inbred C57BL , Macrophage Activation/immunology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Lymphocyte Activation/immunology , Female , Tumor Microenvironment/immunology
13.
Neuropharmacology ; 254: 109981, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38704022

ABSTRACT

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.


Subject(s)
CCR5 Receptor Antagonists , Doxorubicin , Maraviroc , Mice, Inbred C57BL , NF-kappa B , NLR Family, Pyrin Domain-Containing 3 Protein , Signal Transduction , Animals , Maraviroc/pharmacology , CCR5 Receptor Antagonists/pharmacology , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Female , NF-kappa B/metabolism , Signal Transduction/drug effects , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/chemically induced , Breast Neoplasms/drug therapy , Hippocampus/drug effects , Hippocampus/metabolism , Antibiotics, Antineoplastic/toxicity , Chemotherapy-Related Cognitive Impairment/drug therapy , Receptors, CCR5/metabolism , Neuroprotective Agents/pharmacology , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/metabolism
14.
Sci Rep ; 14(1): 10852, 2024 05 13.
Article in English | MEDLINE | ID: mdl-38741006

ABSTRACT

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.


Subject(s)
CRISPR-Cas Systems , Gene Editing , HIV Fusion Inhibitors , HIV Infections , HIV-1 , Receptors, CCR5 , Receptors, CCR5/genetics , Receptors, CCR5/metabolism , Gene Editing/methods , Humans , HIV-1/genetics , HIV-1/drug effects , HIV Infections/genetics , HIV Infections/virology , HIV Infections/therapy , HIV Fusion Inhibitors/pharmacology , Cell Line , Virus Replication/drug effects , Recombinant Fusion Proteins
15.
Microbiol Spectr ; 12(7): e0389523, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38809042

ABSTRACT

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.


Subject(s)
CCR5 Receptor Antagonists , HIV Infections , HIV-1 , Maraviroc , Viral Tropism , HIV-1/drug effects , HIV-1/genetics , HIV-1/physiology , Maraviroc/pharmacology , Humans , CCR5 Receptor Antagonists/pharmacology , HIV Infections/virology , HIV Infections/drug therapy , Inhibitory Concentration 50 , Triazoles/pharmacology , Phenotype , Microbial Sensitivity Tests , Receptors, CCR5/metabolism , Receptors, CCR5/genetics , Anti-HIV Agents/pharmacology , Cyclohexanes/pharmacology , Drug Resistance, Viral/genetics , HIV Fusion Inhibitors/pharmacology
16.
AIDS ; 38(10): 1449-1459, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38770825

ABSTRACT

OBJECTIVE: CCR5, a G protein-coupled receptor (GPCR), is used by most HIV strains as a coreceptor. In this study, we looked for other GPCR able to modify HIV-1 infection. DESIGN: We analyzed the effects of one GPCR coexpressed with CCR5, EBI2, on HIV-1 replicative cycle. METHODS: We identified GPCR expressed in primary CD4 + CCR5 + T cells by multi-RT-qPCR. We studied GPCR dimerization by FRET technology. Cell lines expressing EBI2 were established by transduction with HIV vectors. HIV-1 entry was quantified with virions harboring ß-lactamase fused to the viral protein vpr, early and late HIV-1 transcriptions by qPCR, NFkB nuclear activation by immunofluorescence and transfection, and viral production by measuring p24 concentration in culture supernatant by ELISA. RESULTS: We showed that EBI2 is naturally expressed in primary CD4 + CCR5 + T cells, and that CCR5 and EBI2 heterodimerize. We observed that this coexpression reduced viral entry by 50%. The amount of HIV reverse transcripts was similar in cells expressing or not EBI2. Finally, the presence of EBI2 induced the translocation of NFkB and activated HIV-1 genome expression. Globally, the result was a drastic HIV-1 R5, but not X4, overproduction in EBI2 -transduced cells. CONCLUSION: EBI2 expression in CD4 + CCR5 + cells boosts HIV-1 R5 productive infection. As the natural ligand for EBI2 is present in blood and lymphoid tissues, the constant EBI2 activation might increase HIV replication in CD4 + T cells. It might be of interest to test the effect of EBI2 antagonists on the residual viral production persisting in patients aviremic under treatment.


Subject(s)
CD4-Positive T-Lymphocytes , HIV-1 , Receptors, CCR5 , Receptors, G-Protein-Coupled , Virus Replication , Humans , Receptors, CCR5/metabolism , CD4-Positive T-Lymphocytes/virology , CD4-Positive T-Lymphocytes/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Virus Internalization , Cells, Cultured , HIV Infections/virology , HIV Infections/metabolism , Protein Multimerization , Gene Expression
17.
PLoS Pathog ; 20(5): e1012211, 2024 May.
Article in English | MEDLINE | ID: mdl-38709823

ABSTRACT

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.


Subject(s)
CD8-Positive T-Lymphocytes , Cell Movement , Leishmaniasis, Cutaneous , Receptors, CCR5 , Animals , Receptors, CCR5/metabolism , Receptors, CCR5/immunology , CD8-Positive T-Lymphocytes/immunology , Mice , Humans , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/parasitology , Leishmaniasis, Cutaneous/pathology , Mice, Knockout , Mice, Inbred C57BL , CCR5 Receptor Antagonists/pharmacology , Maraviroc/pharmacology , Female
18.
Int Immunopharmacol ; 135: 112331, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38795597

ABSTRACT

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.


Subject(s)
Asthma , CCR5 Receptor Antagonists , Macrophages , Maraviroc , Pulmonary Fibrosis , Receptors, CCR5 , Transforming Growth Factor beta , Animals , Asthma/immunology , Asthma/drug therapy , Asthma/pathology , Asthma/metabolism , Receptors, CCR5/metabolism , Receptors, CCR5/genetics , Maraviroc/pharmacology , Maraviroc/therapeutic use , CCR5 Receptor Antagonists/pharmacology , CCR5 Receptor Antagonists/therapeutic use , Macrophages/immunology , Macrophages/drug effects , Transforming Growth Factor beta/metabolism , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/drug therapy , Mice , Lung/pathology , Lung/immunology , Lung/drug effects , Mice, Inbred BALB C , Disease Models, Animal , Humans , Female
20.
J Neuroinflammation ; 21(1): 136, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802924

ABSTRACT

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.


Subject(s)
Mesenchymal Stem Cells , Mice, Inbred C57BL , Receptors, CCR5 , Single-Cell Analysis , Uveitis , Animals , Mice , Mesenchymal Stem Cells/metabolism , Uveitis/immunology , Receptors, CCR5/metabolism , Receptors, CCR5/genetics , Autoimmune Diseases/therapy , Gene Expression Profiling , Disease Models, Animal , Female , Single-Cell Gene Expression Analysis
SELECTION OF CITATIONS
SEARCH DETAIL