receptor ccr5
Recently Published Documents


TOTAL DOCUMENTS

228
(FIVE YEARS 15)

H-INDEX

44
(FIVE YEARS 2)

2021 ◽  
Vol 12 ◽  
Author(s):  
Raquel Blanco ◽  
Marta Gómez de Cedrón ◽  
Laura Gámez-Reche ◽  
Ana Martín-Leal ◽  
Alicia González-Martín ◽  
...  

The inhibition of anabolic pathways, such as aerobic glycolysis, is a metabolic cornerstone of memory T cell differentiation and function. However, the signals that hamper these anabolic pathways are not completely known. Recent evidence pinpoints the chemokine receptor CCR5 as an important player in CD4+ T cell memory responses by regulating T cell antigen receptor (TCR) nanoclustering in an antigen-independent manner. This paper reports that CCR5 specifically restrains aerobic glycolysis in memory-like CD4+ T cells, but not in effector CD4+ T cells. CCR5-deficient memory CD4+ T cells thus show an abnormally high glycolytic/oxidative metabolism ratio. No CCR5-dependent change in glucose uptake nor in the expression of the main glucose transporters was detected in any of the examined cell types, although CCR5-deficient memory cells did show increased expression of the hexokinase 2 and pyruvate kinase M2 isoforms, plus the concomitant downregulation of Bcl-6, a transcriptional repressor of these key glycolytic enzymes. Further, the TCR nanoclustering defects observed in CCR5-deficient antigen-experienced CD4+ T cells were partially reversed by incubation with 2-deoxyglucose (2-DG), suggesting a link between inhibition of the glycolytic pathway and TCR nanoscopic organization. Indeed, the treatment of CCR5-deficient lymphoblasts with 2-DG enhanced IL-2 production after antigen re-stimulation. These results identify CCR5 as an important regulator of the metabolic fitness of memory CD4+ T cells, and reveal an unexpected link between T cell metabolism and TCR organization with potential influence on the response of memory T cells upon antigen re-encounter.


Life ◽  
2021 ◽  
Vol 11 (12) ◽  
pp. 1283
Author(s):  
Larry D. Sanford ◽  
Laurie L. Wellman ◽  
Richard P. Ciavarra ◽  
Edward C. Oldfield ◽  
Rouzbeh Shams ◽  
...  

Bi-directional interactions amongst the gut microbiota, immune system, and brain function are thought to be critical mediators of health and disease. The role sleep plays in mediating these interactions is not known. We assessed the effects of sleep fragmentation (SF) on the microbiota–gut–brain axis. Male C57BL/6NCrl mice (4 to 5 per cage, fed standard lab chow) experienced SF via mechanical stimulation at 2 min intervals during the light (SF) and dark (DD, dark disturbances) periods. Home cage (HC) controls were undisturbed. After 10 days, fecal samples were collected at light onset, midday, light offset, and midnight. Samples were also collected after 10 days without SF. Subsequently, the mice were randomized across groups and allowed 20 additional days of recovery followed by 10 days of SF or DD. To assess effects on the microbiota, 16S rRNA sequencing was used, and mesenteric lymph nodes (MLNs) and cortex and medial prefrontal cortex were analyzed using cytokine arrays. SF and DD produced significant alterations in the microbiota compared to HC, and DD had greater impact than SF on some organisms. SF produced marked suppression in MLNs of chemokines that regulate inflammation (CCL3, CCL4 and their receptor CCR5) and maintain the immune mucosal barrier (Cxcl13) at the same time that cortical cytokines (IL-33) indicated neuroinflammation. DD effects on immune responses were similar to HC. These data suggest that SF alters the microbiome and suppresses mucosal immunity at the same time that mediators of brain inflammation are upregulated. The translational implications for potential application to clinical care are compelling.


2021 ◽  
Vol 17 ◽  
pp. 2399-2416
Author(s):  
Yudhi Dwi Kurniawan ◽  
A'liyatur Rosyidah

In recent years fifteen 5,6-dihydro-α-pyrone derivatives, bearing either a distinctive cyclopropane or furan ring and named brevipolides A–O (1–15), have been isolated from the invasive plant Hyptis brevipes Poit. Their fascinating structural features, and the potent biological activities, including cytotoxicity against an array of human cancer cell lines and inhibition of the chemokine receptor CCR5, make them attractive synthetic targets. This review article highlights the recent synthetic methodologies and briefly summarizes their biological activities.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Hui Zhang ◽  
Kun Chen ◽  
Qiuxiang Tan ◽  
Qiang Shao ◽  
Shuo Han ◽  
...  

AbstractThe chemokine receptor CCR5 plays a vital role in immune surveillance and inflammation. However, molecular details that govern its endogenous chemokine recognition and receptor activation remain elusive. Here we report three cryo-electron microscopy structures of Gi1 protein-coupled CCR5 in a ligand-free state and in complex with the chemokine MIP-1α or RANTES, as well as the crystal structure of MIP-1α-bound CCR5. These structures reveal distinct binding modes of the two chemokines and a specific accommodate pattern of the chemokine for the distal N terminus of CCR5. Together with functional data, the structures demonstrate that chemokine-induced rearrangement of toggle switch and plasticity of the receptor extracellular region are critical for receptor activation, while a conserved tryptophan residue in helix II acts as a trigger of receptor constitutive activation.


2021 ◽  
Vol 75 (6) ◽  
pp. 489-494
Author(s):  
Kalyana Bharati Akondi ◽  
Marianne Paolini-Bertrand ◽  
Oliver Hartley

Numerous members of the human G protein-coupled receptor (GPCR) superfamily are receptors of therapeutic interest. GPCRs are considered to be highly tractable for drug discovery, representing the targets of approximately one-third of currently licensed drugs. These successful drug discovery outcomes cover only a relatively small subset of the superfamily, however, and many other attractive receptors have proven to present significant challenges. Among these difficult GPCRs are those whose natural ligands are peptides and proteins. In this review we explain the obstacles faced by GPCR drug discovery campaigns, with particular focus on those related to peptide and protein GPCRs. We describe a novel and promising approach for these targets based on engineering of their natural ligands and describe an integrated discovery platform that allows potent ligand analogs to be discovered rapidly and efficiently. Finally, we present a case study involving the chemokine receptor CCR5 to show that this approach can be used to generate new drugs for peptide and protein GPCR targets combining best-in-class potency with tunable signaling activity.


Gene Therapy ◽  
2021 ◽  
Author(s):  
Lea Isabell Schwarze ◽  
Dawid Głów ◽  
Tanja Sonntag ◽  
Almut Uhde ◽  
Boris Fehse

AbstractDisruption of the C-C-Chemokine-receptor-5 (CCR5) gene induces resistance towards CCR5-tropic HIV. Here we optimised our previously described CCR5-Uco-TALEN and its delivery by mRNA electroporation. The novel variant, CCR5-Uco-hetTALEN features an obligatory heterodimeric Fok1-cleavage domain, which resulted in complete abrogation of off-target activity at previously found homodimeric as well as 7/8 in silico predicted, potential heterodimeric off-target sites, the only exception being highly homologous CCR2. Prevailing 18- and 10-bp deletions at the on-target site revealed microhomology-mediated end-joining as a major repair pathway. Notably, the CCR5Δ55–60 protein resulting from the 18-bp deletion was almost completely retained in the cytosol. Simultaneous cutting at CCR5 and CCR2 induced rearrangements, mainly 15-kb deletions between the cut sites, in up to 2% of T cells underlining the necessity to restrict TALEN expression. We optimised in vitro mRNA production and showed that CCR5-on- and CCR2 off-target activities of CCR5-Uco-hetTALEN were limited to the first 72 and 24–48 h post-mRNA electroporation, respectively. Using single-cell HRMCA, we discovered high rates of TALEN-induced biallelic gene editing of CCR5, which translated in large numbers of CCR5-negative cells resistant to HIVenv-pseudotyped lentiviral vectors. We conclude that CCR5-Uco-hetTALEN transfected by mRNA electroporation facilitates specific, high-efficiency CCR5 gene-editing (30%–56%) and it is highly suited for clinical translation subject to further characterisation of off-target effects.


2021 ◽  
Vol 22 (9) ◽  
pp. 4464
Author(s):  
Tamara Lah Turnšek ◽  
Xuanmao Jiao ◽  
Metka Novak ◽  
Sriharsha Jammula ◽  
Gina Cicero ◽  
...  

The mechanisms governing therapeutic resistance of the most aggressive and lethal primary brain tumor in adults, glioblastoma, have increasingly focused on tumor stem cells. These cells, protected by the periarteriolar hypoxic GSC niche, contribute to the poor efficacy of standard of care treatment of glioblastoma. Integrated proteogenomic and metabolomic analyses of glioblastoma tissues and single cells have revealed insights into the complex heterogeneity of glioblastoma and stromal cells, comprising its tumor microenvironment (TME). An additional factor, which isdriving poor therapy response is the distinct genetic drivers in each patient’s tumor, providing the rationale for a more individualized or personalized approach to treatment. We recently reported that the G protein-coupled receptor CCR5, which contributes to stem cell expansion in other cancers, is overexpressed in glioblastoma cells. Overexpression of the CCR5 ligand CCL5 (RANTES) in glioblastoma completes a potential autocrine activation loop to promote tumor proliferation and invasion. CCL5 was not expressed in glioblastoma stem cells, suggesting a need for paracrine activation of CCR5 signaling by the stromal cells. TME-associated immune cells, such as resident microglia, infiltrating macrophages, T cells, and mesenchymal stem cells, possibly release CCR5 ligands, providing heterologous signaling between stromal and glioblastoma stem cells. Herein, we review current therapies for glioblastoma, the role of CCR5 in other cancers, and the potential role for CCR5 inhibitors in the treatment of glioblastoma.


Author(s):  
Girum Tefera Belachew ◽  
Paramesh Hanumanthaiah ◽  
Bitaniya Abera Tekelemariam

The improvement of particularly versatile genome-modifying advancements has outfitted experts with the ability to rapidly and monetarily bring sequence-specific changes into the genomes of a wide scope of cell types and organisms. The CRISPR framework was first found as a protection system in Escherichia coli against infections. Short portions of unfamiliar DNA are coordinated inside the CRISPR locus and translated into CRISPR RNA (crRNA), which at that point toughen to trans-activating crRNA (tracrRNA) to coordinate sequence specific debasement of pathogenic DNA by the Cas9 protein. Many studies have now revealed insight into the primary premise of DNA recognition by Cas9, showing that the heteroduplex shaped by the gRNA and its complementary strand of DNA is housed in a positively charged groove between the two nuclease areas (RuvC and HNH) inside the Cas9 protein, and that PAM recognition is intervened by an arginine-rich motif present in Cas9. Genome altering biological tools likewise bring healing chances. For instance, ZFN-interceded gene interruption has been taken to the clinic, particularly for the treatment of glioblastoma and HIV by Sangamo biosciences. ZFNs focused to the HIV co-receptor CCR5 for the medication of HIV/AIDS are in stage I clinical trials have been finished currently and are in advancement). In these clinical investigations, the security and possibility of autologous infusion of ex vivo extended CD4+ T cells treated with CCR5- specific ZFNs are assessed in patients with HIV/AIDS. Genome altering itself likewise holds huge potential for treating the fundamental hereditary causes for specific infections. Thusly, the point of this survey is to sum up the vital standards of genome altering, focusing a considerable lot of the designing advances that have laid the foundation for the creation, refinement, and usage of the current set-up of genome-changing biological tools.


Proteomes ◽  
2020 ◽  
Vol 8 (3) ◽  
pp. 24
Author(s):  
Mahlet Lemma ◽  
Stefan Petkov ◽  
Yonas Bekele ◽  
Beyene Petros ◽  
Rawleigh Howe ◽  
...  

Treatment of HIV-1-infected patients results in improved clinical and immunological conditions, but severe non-AIDS-related conditions still persist. Novel proteomic platforms have identified inflammatory proteins where abundance is dysregulated in adult treated patients, whereas limited data are available in treated HIV-1 infection of children. Using a proteomic plasma profiling approach comprising 92 inflammation-related molecules, we analyzed specimens from 43 vertically HIV-1-infected children receiving antiretroviral treatment (ART) and matched controls in Ethiopia. The infected children were analyzed as a group and separately, according to age of treatment initiation. Proteins displaying a significantly different abundance between groups were hierarchically clustered and presented in heat maps. Random forest analysis was performed to pin-point proteins discriminating between groups; five proteins (STAMBP, CD5, TFG-α, TRANCE, AXIN1) were the strongest prediction factors for treated HIV-1 infection. TRANCE was previously linked to reduced bone mass levels in HIV-1-infected children. CCL4 chemokine, ligand to HIV-1 co-receptor CCR5, was the most critical protein for successful classification between children who initiated ART at different time points. Our data provide evidence that a dysregulated expression of proteins linked to immunological abnormalities and bone metabolism can be found in HIV-1-infected children with prolonged exposure to ART.


Sign in / Sign up

Export Citation Format

Share Document