Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
1.
J Phys Chem B ; 128(29): 7086-7101, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39016126

ABSTRACT

BACKGROUND: Human immunodeficiency virus (HIV) infection continues to pose a major global health challenge. HIV entry into host cells via membrane fusion mediated by the viral envelope glycoprotein gp120/gp41 is a key step in the HIV life cycle. CCR5, expressed on CD4+ T cells and macrophages, acts as a coreceptor facilitating HIV-1 entry. The CCR5 antagonist maraviroc is used to treat HIV infection. However, it can cause adverse effects and has limitations such as only inhibiting CCR5-tropic viruses. There remains a need to develop alternative CCR5 inhibitors with improved safety profiles. PROBLEM STATEMENT: Natural products may offer advantages over synthetic inhibitors including higher bioavailability, binding affinity, effectiveness, lower toxicity, and molecular diversity. However, screening the vast chemical space of natural compounds to identify novel CCR5 inhibitors presents challenges. This study aimed to address this gap through a hybrid ligand-based pharmacophore modeling and molecular docking approach to virtually screen large natural product databases. METHODS: A reliable pharmacophore model was developed based on 311 known CCR5 antagonists and validated against an external data set. Five natural product databases containing over 306,000 compounds were filtered based on drug-likeness rules. The validated pharmacophore model screened the databases to identify 611 hits. Key residues of the CCR5 receptor crystal structure were identified for docking. The top hits were docked, and interactions were analyzed. Molecular dynamics simulations were conducted to examine complex stability. Computational prediction evaluated pharmacokinetic properties. RESULTS: Three compounds exhibited similar interactions and binding energies to maraviroc. MD simulations demonstrated complex stability comparable to maraviroc. One compound showed optimal predicted absorption, minimal metabolism, and a lower likelihood of interactions than maraviroc. CONCLUSION: This computational screening workflow identified three natural compounds with promising CCR5 inhibition and favorable pharmacokinetic profiles. One compound emerged as a lead based on bioavailability potential and minimal interaction risk. These findings present opportunities for developing alternative CCR5 antagonists and warrant further experimental investigation. Overall, the hybrid virtual screening approach proved effective for mining large natural product spaces to discover novel molecular entities with drug-like properties.


Subject(s)
Biological Products , CCR5 Receptor Antagonists , HIV-1 , Molecular Docking Simulation , Receptors, CCR5 , Receptors, CCR5/metabolism , Receptors, CCR5/chemistry , Biological Products/chemistry , Biological Products/pharmacology , CCR5 Receptor Antagonists/chemistry , CCR5 Receptor Antagonists/pharmacology , HIV-1/drug effects , Humans , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Drug Evaluation, Preclinical , Maraviroc/chemistry , Maraviroc/pharmacology
2.
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
3.
Sci Rep ; 14(1): 13980, 2024 06 17.
Article in English | MEDLINE | ID: mdl-38886484

ABSTRACT

Maraviroc (MVC) is an antiretroviral drug capable of binding to CCR5 receptors and block HIV entry into target cells. Moreover, MVC can activate NF-kB pathway and induce viral transcription in HIV-infected cells, being proposed as a latency reversal agent (LRA) in HIV cure strategies. However, the evaluation of immunological and metabolic parameters induced by MVC concentrations capable of inducing HIV transcription have not been explored in depth. We cultured isolated CD4 T cells in the absence or presence of MVC, and evaluated the frequency of CD4 T cell subpopulations and activation markers levels by flow cytometry, and the oxidative and glycolytic metabolic rates of CD4 T cells using a Seahorse Analyzer. Our results indicate that a high concentration of MVC did not increase the levels of activation markers, as well as glycolytic or oxidative metabolic rates in CD4 T cells. Furthermore, MVC did not induce significant changes in the frequency and activation levels of memory cell subpopulations. Our data support a safety profile of MVC as a promising LRA candidate since it does not induce alterations of the immunological and metabolic parameters that could affect the functionality of these immune cells.


Subject(s)
CD4-Positive T-Lymphocytes , Maraviroc , Maraviroc/pharmacology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Humans , Glycolysis/drug effects , HIV Infections/drug therapy , HIV Infections/metabolism , HIV Infections/immunology , Cells, Cultured , Triazoles/pharmacology , HIV-1/drug effects , Lymphocyte Activation/drug effects , Male , CCR5 Receptor Antagonists/pharmacology , Cyclohexanes/pharmacology , Adult
5.
Endocrinology ; 165(7)2024 May 27.
Article in English | MEDLINE | ID: mdl-38862137

ABSTRACT

The inhibition of hepatic macrophage and Kupfer cell recruitment and activation is a potential strategy for treating insulin resistance and nonalcoholic steatohepatitis (NASH). Cenicriviroc (CVC), a dual C-C chemokine receptor 2 (CCR2) and CCR5 antagonist, has shown antifibrotic activity in murine models of NASH and has been evaluated in clinical trials on patients with NASH. This study investigated the effects of CVC on macrophage infiltration and polarization in a lipotoxic model of NASH. C57BL/6 mice were fed a high-cholesterol, high-fat (CL) diet or a CL diet containing 0.015% CVC (CL + CVC) for 12 weeks. Macrophage recruitment and activation were assayed by immunohistochemistry and flow cytometry. CVC supplementation attenuated excessive hepatic lipid accumulation and peroxidation and alleviated glucose intolerance and hyperinsulinemia in the mice that were fed the CL diet. Flow cytometry analysis revealed that compared with the CL group, mice fed the CL + CVC diet had fewer M1-like macrophages, more M2-like macrophages, and fewer T cell counts, indicating that CVC caused an M2-dominant shift of macrophages in the liver. Similarly, CVC decreased lipopolysaccharide-stimulated M1-like macrophage activation, whereas it increased interleukin-4-induced M2-type macrophage polarization in vitro. In addition, CVC attenuated hepatic fibrosis by repressing hepatic stellate cell activation. Lastly, CVC reversed insulin resistance as well as steatosis, inflammation, and fibrosis of the liver in mice with pre-existing NASH. In conclusion, CVC prevented and reversed hepatic steatosis, insulin resistance, inflammation, and fibrogenesis in the liver of NASH mice via M2 macrophage polarization.


Subject(s)
Liver , Macrophages , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease , Animals , Macrophages/drug effects , Macrophages/metabolism , Mice , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/pathology , Male , Liver/drug effects , Liver/pathology , Liver/metabolism , Diet, High-Fat/adverse effects , Receptors, CCR2/metabolism , Sulfoxides/pharmacology , Macrophage Activation/drug effects , CCR5 Receptor Antagonists/pharmacology , CCR5 Receptor Antagonists/therapeutic use , Insulin Resistance , Imidazoles
6.
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
7.
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
8.
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
9.
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
10.
Eur J Immunol ; 54(7): e2350847, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38643381

ABSTRACT

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.


Subject(s)
CCR5 Receptor Antagonists , Cell Differentiation , Colitis , Receptors, CCR2 , Receptors, CCR5 , T-Lymphocytes, Regulatory , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Animals , Mice , Receptors, CCR2/metabolism , Receptors, CCR2/antagonists & inhibitors , Colitis/immunology , Colitis/drug therapy , Colitis/chemically induced , Cell Differentiation/drug effects , Cell Differentiation/immunology , CCR5 Receptor Antagonists/pharmacology , CCR5 Receptor Antagonists/therapeutic use , Receptors, CCR5/metabolism , Humans , Th17 Cells/immunology , Th17 Cells/drug effects , Sulfoxides/pharmacology , Mice, Inbred C57BL , Th1 Cells/immunology , Th1 Cells/drug effects , Interleukin-10/metabolism , Th2 Cells/immunology , Imidazoles
11.
Mucosal Immunol ; 17(1): 41-53, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37866719

ABSTRACT

Despite their importance for immunity against sexually transmitted infections, the composition of female reproductive tract (FRT) memory T-cell populations in response to changes within the local tissue environment under the regulation of the menstrual cycle remains poorly defined. Here, we show that in humans and pig-tailed macaques, the cycle determines distinct clusters of differentiation 4 T-cell surveillance behaviors by subsets corresponding to migratory memory (TMM) and resident memory T cells. TMM displays tissue-itinerant trafficking characteristics, restricted distribution within the FRT microenvironment, and distinct effector responses to infection. Gene pathway analysis by RNA sequencing identified TMM-specific enrichment of genes involved in hormonal regulation and inflammatory responses. FRT T-cell subset fluctuations were discovered that synchronized to cycle-driven CCR5 signaling. Notably, oral administration of a CCR5 antagonist drug blocked TMM trafficking. Taken together, this study provides novel insights into the dynamic nature of FRT memory CD4 T cells and identifies the menstrual cycle as a key regulator of immune surveillance at the site of STI pathogen exposure.


Subject(s)
CD4-Positive T-Lymphocytes , Genitalia, Female , Menstrual Cycle , Receptors, CCR5 , Signal Transduction , Female , Humans , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Genitalia, Female/immunology , Genitalia, Female/metabolism , Menstrual Cycle/immunology , Menstrual Cycle/physiology , Receptors, CCR5/genetics , Receptors, CCR5/metabolism , T-Lymphocyte Subsets/immunology , Macaca nemestrina/immunology , Immunologic Memory , Cellular Microenvironment/immunology , Cellular Microenvironment/physiology , CCR5 Receptor Antagonists/pharmacology
12.
Acta Pharmacol Sin ; 44(10): 1935-1947, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37198412

ABSTRACT

Chemokine receptor 5 (CCR5) is one of the main co-receptors of HIV-1, and has been found to be a potential therapeutic target for stroke. Maraviroc is a classic CCR5 antagonist, which is undergoing clinical trials against stroke. As maraviroc shows poor blood-brain barrier (BBB) permeability, it is of interest to find novel CCR5 antagonists suitable for neurological medication. In this study we characterized the therapeutic potential of a novel CCR5 antagonist A14 in treating ischemic stroke mice. A14 was discovered in screening millions compounds in the Chemdiv library based on the molecular docking diagram of CCR5 and maraviroc. We found that A14 dose-dependently inhibited the CCR5 activity with an IC50 value of 4.29 µM. Pharmacodynamic studies showed that A14 treatment exerted protective effects against neuronal ischemic injury both in vitro and vivo. In a SH-SY5Y cell line overexpressing CCR5, A14 (0.1, 1 µM) significantly alleviated OGD/R-induced cell injury. We found that the expression of CCR5 and its ligand CKLF1 was significantly upregulated during both acute and recovery period in focal cortical stroke mice; oral administration of A14 (20 mg·kg-1·d-1, for 1 week) produced sustained protective effect against motor impairment. A14 treatment had earlier onset time, lower onset dosage and much better BBB permeability compared to maraviroc. MRI analysis also showed that A14 treatment significantly reduced the infarction volume after 1 week of treatment. We further revealed that A14 treatment blocked the protein-protein interaction between CCR5 and CKLF1, increasing the activity of CREB signaling pathway in neurons, thereby improving axonal sprouting and synaptic density after stroke. In addition, A14 treatment remarkably inhibited the reactive proliferation of glial cells after stroke and reduced the infiltration of peripheral immune cells. These results demonstrate that A14 is a promising novel CCR5 antagonist for promoting neuronal repair after ischemic stroke. A14 blocked the protein-protein interaction between CKLF1 and CCR5 after stroke by binding with CCR5 stably, improved the infarct area and promoted motor recovery through reversing the CREB/pCREB signaling which was inhibited by activated CCR5 Gαi pathway, and benefited to the dendritic spines and axons sprouting.


Subject(s)
CCR5 Receptor Antagonists , Ischemic Stroke , Neuroblastoma , Stroke , Animals , Humans , Mice , Ischemic Stroke/drug therapy , Maraviroc/therapeutic use , Maraviroc/pharmacology , Molecular Docking Simulation , Receptors, CCR5/metabolism , Stroke/drug therapy , CCR5 Receptor Antagonists/chemistry , CCR5 Receptor Antagonists/pharmacology
13.
J Med Chem ; 65(24): 16526-16540, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36472561

ABSTRACT

Blocking the entry of an HIV-1 targeting CCR5 coreceptor has emerged as an attractive strategy to develop HIV therapeutics. Maraviroc is the only CCR5 antagonist approved by FDA; however, serious side effects limited its clinical use. Herein, 21 novel tropane derivatives (6-26) were designed and synthesized based on the CCR5-maraviroc complex structure. Among them, compounds 25 and 26 had comparable activity to maraviroc and presented more potent inhibitory activity against a series of HIV-1 strains. In addition, compound 26 exhibited synergistic or additive antiviral effects in combination with other antiretroviral agents. Compared to maraviroc, both 25 and 26 displayed higher Cmax and AUC0-∞ and improved oral bioavailability in SD rats. In addition, compounds 25 and 26 showed no significant CYP450 inhibition and showed a novel binding mode with CCR5 different from that of maraviroc-CCR5. In summary, compounds 25 and 26 are promising drug candidates for the treatment of HIV-1 infection.


Subject(s)
Anti-HIV Agents , HIV Infections , HIV-1 , Rats , Animals , Maraviroc/pharmacology , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , CCR5 Receptor Antagonists/pharmacology , Cyclohexanes/pharmacology , Triazoles/pharmacology , Triazoles/therapeutic use , Biological Availability , Rats, Sprague-Dawley , HIV Infections/drug therapy , Tropanes/pharmacology , Receptors, CCR5/metabolism
14.
Viruses ; 14(11)2022 10 31.
Article in English | MEDLINE | ID: mdl-36366513

ABSTRACT

With the aim of rationally devising a refined and potent HIV-1 blocker, the cDNA of CCL5 5p12 5m, an extremely potent CCR5 antagonist, was fused to that of C37, a gp41-targeted fusion inhibitor. The resulting CCL5 5p12 5m-C37 fusion protein was expressed in E. coli and proved to be capable of inhibiting R5 HIV-1 strains with low to sub-picomolar IC50, maintaining its antagonism toward CCR5. In addition, CCL5 5p12 5m-C37 inhibits R5/X4 and X4 HIV-1 strains in the picomolar concentration range. The combination of CCL5 5p12 5m-C37 with tenofovir (TDF) exhibited a synergic effect, promoting this antiviral cocktail. Interestingly, a CCR5-targeted combination of maraviroc (MVC) with CCL5 5p12 5m-C37 led to a synergic effect that could be explained by an extensive engagement of different CCR5 conformational populations. Within the mechanism of HIV-1 entry, the CCL5 5p12 5m-C37 chimera may fit as a powerful blocker in several instances. In its possible consideration for systemic therapy or pre-exposure prophylaxis, this protein design represents an interesting lead in the combat of HIV-1 infection.


Subject(s)
HIV Infections , HIV Seropositivity , HIV-1 , Humans , Receptors, CCR5/genetics , Receptors, CCR5/metabolism , Escherichia coli/metabolism , Maraviroc/pharmacology , Maraviroc/therapeutic use , HIV Infections/metabolism , CCR5 Receptor Antagonists/pharmacology , CCR5 Receptor Antagonists/therapeutic use
15.
PLoS One ; 17(10): e0275269, 2022.
Article in English | MEDLINE | ID: mdl-36251708

ABSTRACT

CC Chemokine receptor 5 (CCR5), a member of the Superfamily of G Protein-Coupled Receptors (GPCRs), is an important effector in multiple physiopathological processes such as inflammatory and infectious entities, including central nervous system neuroinflammatory diseases such as Alzheimer's disease, recovery from nervous injuries, and in the HIV-AIDS infective processes. Thus, CCR5 is an attractive target for pharmacological modulation. Since maraviroc was described as a CCR5 ligand that modifies the HIV-AIDS progression, multiple efforts have been developed to describe the functionality of the receptor. In this work, we characterized key structural features of the CCR5 receptor employing extensive atomistic molecular dynamics (MD) in its apo form and in complex with an endogenous agonist, the chemokine CCL5/RANTES, an HIV entry inhibitor, the partial inverse agonist maraviroc, and the experimental antagonists Compound 21 and 34, aiming to elucidate the structural features and mechanistic processes that constitute its functional states, contributing with structural details and a general understanding of this relevant system.


Subject(s)
HIV Fusion Inhibitors , HIV Infections , CCR5 Receptor Antagonists/pharmacology , CCR5 Receptor Antagonists/therapeutic use , Chemokine CCL5/pharmacology , HIV Infections/drug therapy , Humans , Imidazoles , Ligands , Maraviroc/therapeutic use , Receptors, CCR5 , Sulfonamides , Thiophenes
16.
PLoS Pathog ; 18(6): e1010547, 2022 06.
Article in English | MEDLINE | ID: mdl-35749425

ABSTRACT

Coronavirus Disease 2019 (COVID-19), caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), has created a global pandemic infecting over 230 million people and costing millions of lives. Therapies to attenuate severe disease are desperately needed. Cenicriviroc (CVC), a C-C chemokine receptor type 5 (CCR5) and C-C chemokine receptor type 2 (CCR2) antagonist, an agent previously studied in advanced clinical trials for patients with HIV or nonalcoholic steatohepatitis (NASH), may have the potential to reduce respiratory and cardiovascular organ failures related to COVID-19. Inhibiting the CCR2 and CCR5 pathways could attenuate or prevent inflammation or fibrosis in both early and late stages of the disease and improve outcomes of COVID-19. Clinical trials using CVC either in addition to standard of care (SoC; e.g., dexamethasone) or in combination with other investigational agents in patients with COVID-19 are currently ongoing. These trials intend to leverage the anti-inflammatory actions of CVC for ameliorating the clinical course of COVID-19 and prevent complications. This article reviews the literature surrounding the CCR2 and CCR5 pathways, their proposed role in COVID-19, and the potential role of CVC to improve outcomes.


Subject(s)
CCR5 Receptor Antagonists , COVID-19 Drug Treatment , CCR5 Receptor Antagonists/pharmacology , CCR5 Receptor Antagonists/therapeutic use , Humans , Imidazoles , Receptors, CCR2 , Receptors, CCR5 , SARS-CoV-2 , Sulfoxides
17.
J Exp Med ; 219(5)2022 05 02.
Article in English | MEDLINE | ID: mdl-35404390

ABSTRACT

The resistance of pancreatic ductal adenocarcinoma (PDAC) to immune checkpoint inhibitors (ICIs) is attributed to the immune-quiescent and -suppressive tumor microenvironment (TME). We recently found that CCR2 and CCR5 were induced in PDAC following treatment with anti-PD-1 antibody (αPD-1); thus, we examined PDAC vaccine or radiation therapy (RT) as T cell priming mechanisms together with BMS-687681, a dual antagonist of CCR2 and CCR5 (CCR2/5i), in combination with αPD-1 as new treatment strategies. Using PDAC mouse models, we demonstrated that RT followed by αPD-1 and prolonged treatment with CCR2/5i conferred better antitumor efficacy than other combination treatments tested. The combination of RT + αPD-1 + CCR2/5i enhanced intratumoral effector and memory T cell infiltration but suppressed regulatory T cell, M2-like tumor-associated macrophage, and myeloid-derived suppressive cell infiltration. RNA sequencing showed that CCR2/5i partially inhibited RT-induced TLR2/4 and RAGE signaling, leading to decreased expression of immunosuppressive cytokines including CCL2/CCL5, but increased expression of effector T cell chemokines such as CCL17/CCL22. This study thus supports the clinical development of CCR2/5i in combination with RT and ICIs for PDAC treatment.


Subject(s)
Adenocarcinoma , CCR5 Receptor Antagonists , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Receptors, CCR2 , Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Animals , CCR5 Receptor Antagonists/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/radiotherapy , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/radiotherapy , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR5 , Tumor Microenvironment , Pancreatic Neoplasms
18.
Front Immunol ; 13: 826418, 2022.
Article in English | MEDLINE | ID: mdl-35126399

ABSTRACT

The large number of pathologies that position CCR5 as a central molecular determinant substantiates the studies aimed at understanding receptor-ligand interactions, as well as the development of compounds that efficiently block this receptor. This perspective focuses on CCR5 antagonism as the preferred landscape for therapeutic intervention, thus the receptor active site occupancy by known antagonists of different origins is overviewed. CCL5 is a natural agonist ligand for CCR5 and an extensively studied scaffold for CCR5 antagonists production through chemokine N-terminus modification. A retrospective 3D modeling analysis on recently developed CCL5 mutants and their contribution to enhanced anti-HIV-1 activity is reported here. These results allow us to prospect the development of conceptually novel amino acid substitutions outside the CCL5 N-terminus hotspot. CCR5 interaction improvement in regions distal to the chemokine N-terminus, as well as the stabilization of the chemokine hydrophobic core are strategies that influence binding affinity and stability beyond the agonist/antagonist dualism. Furthermore, the development of allosteric antagonists topologically remote from the orthosteric site (e.g., intracellular or membrane-embedded) is an intriguing new avenue in GPCR druggability and thus a conceivable novel direction for CCR5 blockade. Ultimately, the three-dimensional structure elucidation of the interaction between various ligands and CCR5 helps illuminate the active site occupancy and mechanism of action.


Subject(s)
CCR5 Receptor Antagonists/pharmacology , Chemokine CCL5/chemistry , HIV-1/physiology , Models, Molecular , Receptors, CCR5/chemistry , Animals , CCR5 Receptor Antagonists/chemistry , Chemokine CCL5/metabolism , HIV Infections/drug therapy , HIV Infections/immunology , Humans , Ligands , Protein Binding , Receptors, CCR5/metabolism
19.
J Immunol ; 208(5): 1170-1179, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35140134

ABSTRACT

Mucosa-associated invariant T (MAIT) cells recognize bacterial riboflavin metabolite Ags presented by MHC class Ib-related protein (MR1) and play important roles in immune control of microbes that synthesize riboflavin. This includes the pathobiont Staphylococcus aureus, which can also express a range of virulence factors, including the secreted toxin leukocidin ED (LukED). In this study, we found that human MAIT cells are hypersensitive to LukED-mediated lysis and lost on exposure to the toxin, leaving a T cell population devoid of MAIT cells. The cytolytic effect of LukED on MAIT cells was rapid and occurred at toxin concentrations lower than those required for toxicity against conventional T cells. Furthermore, this coincided with high MAIT cell expression of CCR5, and loss of these cells was efficiently inhibited by the CCR5 inhibitor maraviroc. Interestingly, exposure and preactivation of MAIT cells with IL-12 and IL-18, or activation via TCR triggering, partially protected from LukED toxicity. Furthermore, analysis of NK cells indicated that LukED targeted the mature cytotoxic CD57+ NK cell subset in a CCR5-independent manner. Overall, these results indicate that LukED efficiently eliminates immune cells that can respond rapidly to S. aureus in an innate fashion without the need for clonal expansion, and that MAIT cells are exceptionally vulnerable to this toxin. Thus, the findings support a model where LukED secretion may allow S. aureus to avoid recognition by the rapid cell-mediated responses mediated by MAIT cells and NK cells.


Subject(s)
Immune Evasion/immunology , Killer Cells, Natural/immunology , Leukocidins/metabolism , Mucosal-Associated Invariant T Cells/pathology , Receptors, CCR5/metabolism , Staphylococcus aureus/pathogenicity , CCR5 Receptor Antagonists/pharmacology , Cell Line , Humans , Interleukin-12 Subunit p35/metabolism , Interleukin-18/metabolism , Lymphocyte Activation/immunology , Maraviroc/pharmacology , Mucosal-Associated Invariant T Cells/immunology , Staphylococcal Infections/pathology , Staphylococcus aureus/immunology , THP-1 Cells , Virulence Factors/metabolism
20.
Fluids Barriers CNS ; 19(1): 7, 2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35062973

ABSTRACT

BACKGROUND: C-C chemokine receptor type 1 (CCR1) and its endogenous ligand, CCL5, participate in the pathogenesis of neuroinflammatory diseases. However, much remains unknown regarding CCL5/CCR1 signaling in blood-brain barrier (BBB) permeability after intracerebral hemorrhage (ICH). METHODS: A total of 250 CD1 male mice were used and ICH was induced via autologous whole blood injection. Either Met-RANTES, a selective CCR1 antagonist, or Met-RANTES combined with a Rac1 CRISPR activator was administered to the mice 1 h after ICH. Post-ICH assessments included neurobehavioral tests, brain water content, BBB integrity, hematoma volume, Western blot, and immunofluorescence staining. The CCR1 ligand, rCCL5, and SRC CRISPR knockout in naïve mice were used to further elucidate detrimental CCL5/CCR1/SRC signaling. RESULTS: Brain endogenous CCR1 and CCL5 were upregulated after ICH in mice with a peak at 24 h, and CCR1 was expressed in endothelial cells, astrocytes, and neurons. Met-R treatment reduced brain edema and neurobehavioral impairment, as well as preserved BBB integrity and tight junction protein expression in ICH mice. Met-R treatment decreased expression of p-SRC, Rac1, albumin, and MMP9, but increased claudin-5, occludin, and ZO-1 tight junction proteins after ICH. These effects were regressed using the Rac1 CRISPR activator. Administration of rCCL5 in naïve mice increased expression of p-SRC, Rac1, albumin, and MMP9, but decreased levels of claudin-5, occludin, and ZO-1 tight junction proteins. These effects in naïve mice were reversed with SRC CRISPR (KO). CONCLUSIONS: Our findings demonstrate that CCR5 inhibition by Met-R improves neurological deficits after ICH by preserving BBB integrity through inhibiting CCR1/SRC/Rac1 signaling pathway in mice. Thus, Met-R has therapeutic potential in the management of ICH patients.


Subject(s)
Blood-Brain Barrier/metabolism , CCR5 Receptor Antagonists/pharmacology , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/metabolism , Chemokine CCL5/pharmacology , Neuropeptides/metabolism , Receptors, CCR1/metabolism , rac1 GTP-Binding Protein/metabolism , src-Family Kinases/metabolism , Animals , Blood-Brain Barrier/drug effects , Chemokine CCL5/administration & dosage , Male , Mice , Neuropeptides/drug effects , Receptors, CCR1/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , rac1 GTP-Binding Protein/drug effects , src-Family Kinases/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL