Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Cell Commun Signal ; 22(1): 191, 2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38528533

ABSTRACT

BACKGROUND: The incidence of diabetic kidney disease (DKD) continues to rapidly increase, with limited available treatment options. One of the hallmarks of DKD is persistent inflammation, but the underlying molecular mechanisms of early diabetic kidney injury remain poorly understood. C-X-C chemokine receptor 2 (CXCR2), plays an important role in the progression of inflammation-related vascular diseases and may bridge between glomerular endothelium and persistent inflammation in DKD. METHODS: Multiple methods were employed to assess the expression levels of CXCR2 and its ligands, as well as renal inflammatory response and endothelial glycocalyx shedding in patients with DKD. The effects of CXCR2 on glycocalyx shedding, and persistent renal inflammation was examined in a type 2 diabetic mouse model with Cxcr2 knockout specifically in endothelial cells (DKD-Cxcr2 eCKO mice), as well as in glomerular endothelial cells (GECs), cultured in high glucose conditions. RESULTS: CXCR2 was associated with early renal decline in DKD patients, and endothelial-specific knockout of CXCR2 significantly improved renal function in DKD mice, reduced inflammatory cell infiltration, and simultaneously decreased the expression of proinflammatory factors and chemokines in renal tissue. In DKD conditions, glycocalyx shedding was suppressed in endothelial Cxcr2 knockout mice compared to Cxcr2 L/L mice. Modulating CXCR2 expression also affected high glucose-induced inflammation and glycocalyx shedding in GECs. Mechanistically, CXCR2 deficiency inhibited the activation of NF-κB signaling, thereby regulating inflammation, restoring the endothelial glycocalyx, and alleviating DKD. CONCLUSIONS: Taken together, under DKD conditions, activation of CXCR2 exacerbates inflammation through regulation of the NF-κB pathway, leading to endothelial glycocalyx shedding and deteriorating renal function. Endothelial CXCR2 deficiency has a protective role in inflammation and glycocalyx dysfunction, suggesting its potential as a promising therapeutic target for DKD treatment.


Subject(s)
Diabetic Nephropathies , NF-kappa B , Receptors, Interleukin-8B , Animals , Humans , Mice , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Endothelial Cells/metabolism , Endothelium/metabolism , Glucose , Glycocalyx/metabolism , Inflammation/metabolism , Mice, Knockout , NF-kappa B/metabolism , Receptors, Chemokine/therapeutic use , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism , Diabetes Complications/genetics , Diabetes Complications/metabolism
2.
Arab J Gastroenterol ; 25(1): 28-36, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38220479

ABSTRACT

BACKGROUND AND STUDY AIM: Hepatocellular carcinoma (HCC) is the fifth leading cause of cancer-related mortality worldwide, and, more than half of these cases are diagnosed in China. However, effective treatment for HCC is still limited. MATERIAL AND METHODS: C-X-C motif chemokine receptor 4 (CXCR4) was first activated and inhibited in HepG2 cells using a pharmacological method. HepG2 cell proliferation was detected using the CCK-8 method. Metastasis and apoptosis of HepG2 cells were detected using wound healing and flow cytometry. The expression of each target molecule related to metastasis and invasion, such as MMPs, E-cadherin and the PI3K/AKT/Mcl-1/PARP signaling pathway was detected by western blotting. The secretion of molecular metastases was detected using competitive ELISA. RESULTS: This study constructed a CXCR4 activation and inhibition model in HepG2 cells. CXCR4 inhibition promoted the inhibitory effect of plantamajoside on the proliferation and metastasis of cells, which led to apoptosis. Furthermore, we found that the expression of apoptosis-related proteins was increased after treatment with plantamajoside combined with CXCR4 inhibition. In addition, the expression and secretion of pro-metastatic proteins, including MMPs and E-cadherin were decreased. We also noticed that this effect might be mediated by the PI3K/AKT/Mcl-1/PARP signaling pathway. CONCLUSION: CXCR4 inhibition may contribute to the treatment of HCC. Inhibition of CXCR4 expression contributes to the therapeutic effect of plantamajoside; the effect of plantamajoside might be mediated by the PI3K/AKT/Mcl-1/PARP signaling pathway; and CXCR4 might be a therapeutic target of HCC.


Subject(s)
Carcinoma, Hepatocellular , Catechols , Glucosides , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/pharmacology , Proto-Oncogene Proteins c-akt/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/pharmacology , Phosphatidylinositol 3-Kinases/therapeutic use , Myeloid Cell Leukemia Sequence 1 Protein/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Cell Movement , Apoptosis , Cadherins , Receptors, Chemokine/therapeutic use
3.
Phytomedicine ; 123: 155238, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38128394

ABSTRACT

BACKGROUND: Ischemic stroke, a severe and life-threatening neurodegenerative condition, currently relies on thrombolytic therapy with limited therapeutic window and potential risks of hemorrhagic transformation. Thus, there is a crucial need to explore novel therapeutic agents for ischemic stroke. Ginsenoside Rg1 (Rg1), a potential neuroprotective agent, exhibits anti-ischemic effects attributed to its anti-inflammatory, anti-oxidant, and anti-apoptotic properties. Nevertheless, the precise underlying mechanism of action remains to be fully elucidated. PURPOSE: This study aimed to explore whether Rg1 exerts anti-ischemic stroke effects by inhibiting pyroptotic neuronal cell death through modulation of the chemokine like factor 1 (CKLF1)/ C-C chemokine receptor type 5 (CCR5) axis. METHODS: In this study, the MCAO model was used as an ischemic stroke model, and experimental tests were performed after 6 hours of ischemia. The anti-ischemic effect of Rg1 was examined by TTC staining, nissl-staining and neurobehavioral tests. In the in vitro experiments, PC12 cells were subjected to stimulation with CKLF1's mimetic peptide C27 to assess the potential of CKLF1 to induce focal neuronal cell death. Additionally, the impact of CKLF1 mimetic peptide C27, antagonistic peptide C19, and CCR5 inhibitor MVC on PC12 cells subjected to oxygen-glucose deprivation (OGD) and subsequently treated with Rg1 was investigated. In vivo, Rg1 treatment was examined by quantitative real-time PCR (qPCR), ELISA, immunohistochemistry (IHC), immunofluorescence (IF), western blot (WB), and co-immunoprecipitate (Co-IP) assays to perspective whether Rg1 treatment reduces CKLF1/CCR5 axis-induced pyroptotic neuronal cell death. In addition, to further explore the biological significance of CKLF1 in ischemic stroke, CKLF1-/- rats were used as the observation subjects in this study. RESULTS: The in vitro results suggested that CKLF1 was able to induce neuronal cells to undergo pyroptosis. In vivo pharmacodynamic results showed that Rg1 treatment was able to significantly improve symptoms in ischemic stroke rats. In addition, Rg1 treatment was able to inhibit the interaction between CKLF1 and CCR5 after ischemic stroke and inhibited CKLF1/CCR5 axis-induced pyroptosis. The results of related experiments in CKLF1-/- rats showed that Rg1 lost its therapeutic effect after CKLF1 knockdown. CONCLUSION: Our findings indicate that the activation of the NLRP3 inflammasome is initiated by the CKLF1/CCR5 axis, facilitated through the activation of the NF-κB pathway, ultimately resulting in the pyroptosis of neuronal cells. Conversely, Rg1 demonstrates the capability to mitigate neuronal cell damage following CKLF1-induced effects by suppressing the expression of CKLF1. Thus, CKLF1 represents a crucial target for Rg1 in the context of cerebral ischemia treatment, and it also holds promise as a potential target for drug screening in the management of ischemic stroke.


Subject(s)
Brain Ischemia , Ginsenosides , Ischemic Stroke , Reperfusion Injury , Humans , Rats , Animals , Ischemic Stroke/drug therapy , Pyroptosis , Receptors, Chemokine/therapeutic use , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Reperfusion Injury/drug therapy , Receptors, CCR5/therapeutic use
4.
Clin Transl Gastroenterol ; 14(12): e00635, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37655708

ABSTRACT

INTRODUCTION: Ustekinumab, a monoclonal antibody to the p40 subunit of interleukin (IL)-12 and IL-23, is used for Crohn's disease (CD), and the documented clinical remission rate after 1 year was observed in approximately 50% of patients. We aimed to identify predictors for a clinical response using peripheral blood obtained from patients with CD just before ustekinumab treatment initiation. METHODS: RNA extraction from peripheral blood mononuclear cells was followed by mRNA paired-end sequencing. Differential gene expression was performed using DESeq2. RESULTS: We processed samples from 36 adults with CD (13 men, 36%) obtained at baseline before starting ustekinumab treatment. Twenty-two of 36 (61%) were defined as responders and 14/36 (39%) as nonresponders after 1 year based on Physician Global Assessment. Differential gene expression between responders (n = 22) and nonresponders (n = 14) did not show a gene expression signature that passed false discovery rate (FDR) correction. However, the analyses identified 68 genes, including CXCL1/2/3, which were induced in nonresponders vs responders with P < 0.05 and fold change above 1.5. Functional annotation enrichments of these 68 genes using ToppGene indicated enrichment for cytokine activity (FDR = 1.98E-05), CXCR chemokine receptor binding (FDR = 2.11E-05), IL-10 signaling (FDR = 5.03E-07), genes encoding secreted soluble factors (FDR = 1.73E-05), and myeloid dendritic cells (FDR = 1.80E-08). DISCUSSION: No substantial differences were found in peripheral blood mononuclear cell transcriptomics between responders and nonresponders. However, among the nonresponders, we noted an increased inflammatory response enriched for pathways linked with cytokine activity and chemokine receptor binding and innate myeloid signature. A larger cohort is required to validate and further explore these findings.


Subject(s)
Crohn Disease , Ustekinumab , Male , Adult , Humans , Ustekinumab/therapeutic use , Ustekinumab/pharmacology , Crohn Disease/diagnosis , Crohn Disease/drug therapy , Crohn Disease/genetics , Leukocytes, Mononuclear , Interleukin-12/therapeutic use , Gene Expression Profiling , Receptors, Chemokine/therapeutic use
5.
J Pak Med Assoc ; 73(Suppl 4)(4): S52-S55, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37482830

ABSTRACT

Objectives: To examine the C-X-C Motif Chemokine Receptor 1 expression in breast cancer tissues prior to neo-adjuvant chemotherapy, and its relationship to neo-adjuvant chemotherapy effectiveness and other prognostic variables. Method: The prospective study was conducted at Kafrelsheikh University Hospital, Egypt, from November 2018 to March 2021, and comprised patients with recent histopathologically proven breast cancer cases eligible for chemotherapy. Paraffin blocks of tumourspecimens were stained by immunohistochemicalstain using concentrating rabbit anti-human C-X-C Motif Chemokine Receptor 1 polyclonal antibody kits. C-X-C Motif Chemokine Receptor 1 expression was classified into low and high categories. Patients were followed for 2 years for treatment response, disease recurrence and mortality. Data was analysed using SPSS 25. RESULTS: Of the 100 females with mean age 50.2±12.1 years, 52(52%) had their left side affected, while 48(48%) had their rightside affected. There were 52(52%) cases with mean age 49.2±12.9 years having high C-X-C Motif Chemokine Receptor 1 expresssion, while 48(48%) with mean age 51.4±11.2 years had low expression. There was a significant association between high expression and advanced tumour grade, advanced tumourstage, higher frequency of triple negative breast cancer and higher frequency of Ki-67-positive cancers (p<0.05). Patients with high C-X-C Motif Chemokine Receptor 1 expression had significantly lower frequency of complete pathological response when compared with patients with low expression (p<0.001). Patients with high expression had higher frequency of recurrence, shorter disease-free survival, higher mortality and shorter overall survival, but the difference was not significant (p>0.05). Multivariate logistic regression analysis identified triple negative hormonal status (p=0.031) and high baseline C-X-C Motif Chemokine Receptor 1 expression (p<0.001) as significant predictors of complete pathological response. CONCLUSIONS: There was found to be a link between baseline C-X-C Motif Chemokine Receptor 1 expression in breast cancer tissues and pathological response to neoadjuvant therapy in breast cancer patients.


Subject(s)
Breast Neoplasms , Triple Negative Breast Neoplasms , Female , Humans , Adult , Middle Aged , Breast Neoplasms/pathology , Neoadjuvant Therapy , Prospective Studies , Retrospective Studies , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasm Recurrence, Local , Prognosis , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Chemotherapy, Adjuvant , Receptors, Chemokine/therapeutic use , Receptor, ErbB-2/metabolism
6.
Kidney Blood Press Res ; 48(1): 405-413, 2023.
Article in English | MEDLINE | ID: mdl-37231814

ABSTRACT

INTRODUCTION: Diabetic nephropathy (DN) is a common complication in diabetic patients. Chemerin, a novel adipokine, has been associated with renal damage in DN. The chemerin chemokine-like receptor 1 (CMKLR1) has been reported to participate in DN. In this study, we aimed to investigate the effect of a CMKLR1 antagonist, 2-(anaphthoyl)ethyltrimethylammonium iodide (α-NETA), on DN. METHODS: To induce diabetes, 8-week-old male C57BL/6J mice were given a single intraperitoneal injection of 65 mg/kg streptozotocin (STZ). Diabetic mice were randomly assigned to receive daily doses of 0, 5, or 10 mg/kg α-NETA for 4 weeks. RESULTS: α-NETA dose-dependently induced body weight and reduced fasting blood glucose levels in STZ-induced diabetic mice. Furthermore, α-NETA significantly reduced the expressions of renal injury markers, including serum creatinine, kidney weight/body weight, urine volume, total proteins, and albumin in the urine, and increased creatinine clearance. Periodic acid-Schiff staining also indicated that α-NETA could effectively ameliorate renal injuries in DN mice. In addition, α-NETA inhibited renal inflammation and the expressions of chemerin and CMKLR1 in mice with DN. CONCLUSION: In summary, our findings suggested that α-NETA has beneficial effects on the management of DN. Specifically, α-NETA effectively ameliorated renal damage and inflammation in a dose-dependent manner in mice with DN. Thus, targeting the chemerin and CMKLR1 axis with α-NETA may be a promising therapeutic strategy for the treatment of DN.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , Humans , Mice , Male , Animals , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Mice, Inbred C57BL , Kidney/metabolism , Inflammation/metabolism , Body Weight , Receptors, Chemokine/metabolism , Receptors, Chemokine/therapeutic use , Chemokines/metabolism , Intercellular Signaling Peptides and Proteins
7.
Cancer Commun (Lond) ; 43(5): 525-561, 2023 05.
Article in English | MEDLINE | ID: mdl-37005490

ABSTRACT

Tumor development and metastasis are facilitated by the complex interactions between cancer cells and their microenvironment, which comprises stromal cells and extracellular matrix (ECM) components, among other factors. Stromal cells can adopt new phenotypes to promote tumor cell invasion. A deep understanding of the signaling pathways involved in cell-to-cell and cell-to-ECM interactions is needed to design effective intervention strategies that might interrupt these interactions. In this review, we describe the tumor microenvironment (TME) components and associated therapeutics. We discuss the clinical advances in the prevalent and newly discovered signaling pathways in the TME, the immune checkpoints and immunosuppressive chemokines, and currently used inhibitors targeting these pathways. These include both intrinsic and non-autonomous tumor cell signaling pathways in the TME: protein kinase C (PKC) signaling, Notch, and transforming growth factor (TGF-ß) signaling, Endoplasmic Reticulum (ER) stress response, lactate signaling, Metabolic reprogramming, cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) and Siglec signaling pathways. We also discuss the recent advances in Programmed Cell Death Protein 1 (PD-1), Cytotoxic T-Lymphocyte Associated Protein 4 (CTLA4), T-cell immunoglobulin mucin-3 (TIM-3) and Lymphocyte Activating Gene 3 (LAG3) immune checkpoint inhibitors along with the C-C chemokine receptor 4 (CCR4)- C-C class chemokines 22 (CCL22)/ and 17 (CCL17), C-C chemokine receptor type 2 (CCR2)- chemokine (C-C motif) ligand 2 (CCL2), C-C chemokine receptor type 5 (CCR5)- chemokine (C-C motif) ligand 3 (CCL3) chemokine signaling axis in the TME. In addition, this review provides a holistic understanding of the TME as we discuss the three-dimensional and microfluidic models of the TME, which are believed to recapitulate the original characteristics of the patient tumor and hence may be used as a platform to study new mechanisms and screen for various anti-cancer therapies. We further discuss the systemic influences of gut microbiota in TME reprogramming and treatment response. Overall, this review provides a comprehensive analysis of the diverse and most critical signaling pathways in the TME, highlighting the associated newest and critical preclinical and clinical studies along with their underlying biology. We highlight the importance of the most recent technologies of microfluidics and lab-on-chip models for TME research and also present an overview of extrinsic factors, such as the inhabitant human microbiome, which have the potential to modulate TME biology and drug responses.


Subject(s)
Neoplasms , Tumor Microenvironment , Humans , Neoplasms/metabolism , Signal Transduction , Neoplastic Processes , Receptors, Chemokine/therapeutic use , Chemokines/pharmacology , Chemokines/therapeutic use
8.
Gastroenterology ; 164(7): 1261-1278, 2023 06.
Article in English | MEDLINE | ID: mdl-36863689

ABSTRACT

BACKGROUND & AIMS: The therapeutic effect of immune checkpoint inhibitors (ICIs) is poor in hepatocellular carcinoma (HCC) and varies greatly among individuals. Schlafen (SLFN) family members have important functions in immunity and oncology, but their roles in cancer immunobiology remain unclear. We aimed to investigate the role of the SLFN family in immune responses against HCC. METHODS: Transcriptome analysis was performed in human HCC tissues with or without response to ICIs. A humanized orthotopic HCC mouse model and a co-culture system were constructed, and cytometry by time-of-flight technology was used to explore the function and mechanism of SLFN11 in the immune context of HCC. RESULTS: SLFN11 was significantly up-regulated in tumors that responded to ICIs. Tumor-specific SLFN11 deficiency increased the infiltration of immunosuppressive macrophages and aggravated HCC progression. HCC cells with SLFN11 knockdown promoted macrophage migration and M2-like polarization in a C-C motif chemokine ligand 2-dependent manner, which in turn elevated their own PD-L1 expression by activating the nuclear factor-κB pathway. Mechanistically, SLFN11 suppressed the Notch pathway and C-C motif chemokine ligand 2 transcription by binding competitively with tripartite motif containing 21 to the RNA recognition motif 2 domain of RBM10, thereby inhibiting tripartite motif containing 21-mediated RBM10 degradation to stabilize RBM10 and promote NUMB exon 9 skipping. Pharmacologic antagonism of C-C motif chemokine receptor 2 potentiated the antitumor effect of anti-PD-1 in humanized mice bearing SLFN11 knockdown tumors. ICIs were more effective in patients with HCC with high serum SLFN11 levels. CONCLUSIONS: SLFN11 serves as a critical regulator of microenvironmental immune properties and an effective predictive biomarker of ICIs response in HCC. Blockade of C-C motif chemokine ligand 2/C-C motif chemokine receptor 2 signaling sensitized SLFN11low HCC patients to ICI treatment.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Animals , Mice , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Ligands , Macrophages/metabolism , Receptors, Chemokine/metabolism , Receptors, Chemokine/therapeutic use , Cell Line, Tumor , Tumor Microenvironment , Chemokine CCL2 , RNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism
9.
Drug Res (Stuttg) ; 73(4): 213-223, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36754055

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSCs) modulate immune responses, and their immunomodulatory potential can be enhanced using inflammatory cytokines. Here, the modulatory effects of IFN-γ-licensed MSCs on expression of T cell-related chemokines and chemokine receptors were evaluated using an experimental autoimmune encephalomyelitis (EAE) model. MATERIAL AND METHODS: EAE was induced in 3 groups of C57bl/6 mice and then treated with PBS, MSCs and IFN-γ-treated MSCs. The EAE manifestations were registered daily and finally, the brain and spinal cords were isolated for histopathological and gene expression studies. RESULTS: The clinical scores were lowered in MSCs and IFN-γ-licensed MSCs groups, however, mice treated with IFN-γ-licensed MSCs exhibited lower clinical scores than MSCs-treated mice. Leukocyte infiltration into the brain was reduced after treatment with MSCs or IFN-γ-licensed MSCs compared to untreated group (P<0.05 and P<0.01, respectively). In comparison with untreated EAE mice, treatment with MSCs reduced CCL20 expression (P<0.001) and decreased CXCR3 and CCR6 expression (P<0.02 and P<0.04, respectively). In comparison with untreated EAE mice, treatment with IFN-γ-licensed MSCs reduced CXCL10, CCL17 and CCL20 expression (P<0.05, P<0.05, and P<0.001, respectively) as well as decreased CXCR3 and CCR6 expression (P<0.002 and P<0.02, respectively), whilst promoting expression of CCL22 and its receptor CCR4 (P<0.0001 and P<0.02, respectively). In comparison with MSC-treated group, mice treated with IFN-γ-licensed MSCs exhibited lower CXCL10 and CCR6 expression (P<0.002 and P<0.01, respectively), whereas greater expression of CCL22 and CCR4 (P<0.0001 and P<0.01, respectively). CONCLUSION: Priming the MSC with IFN-γ can be an efficient approach to enhance the immunomodulatory potential of MSCs.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Mesenchymal Stem Cells , Animals , Mice , Encephalomyelitis, Autoimmune, Experimental/therapy , Interferon-gamma , Receptors, Chemokine/metabolism , Receptors, Chemokine/therapeutic use , Chemokines/metabolism , Chemokines/pharmacology , Chemokines/therapeutic use , T-Lymphocytes , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL
10.
EBioMedicine ; 84: 104254, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36150362

ABSTRACT

BACKGROUND: Immune checkpoint blockade (ICB) partially reverses the dysfunctional state of antigen-specific T cell in chronic infections. However, its impact on the diverse subsets of CD4+ T cells in humans is largely unknown. METHODS: We examined immune checkpoint (IC) expression and function in HIV-specific CD4+ T cells of viremic individuals (≥5000 vRNA cp/ml, n = 17) prior to ART and persons with spontaneous (n = 11) or therapy-induced (n = 16) viral suppression (<40 cp/ml). We investigated IC patterns associated with exhaustion-related transcription factors and chemokine receptors using activation-induced marker assays. We determined effector functions representative of TFH, TH1, and TH17/TH22 using RNA flow cytometric fluorescence in situ hybridization (FISH). We compared increase in cytokine expression upon ICB across functions and patient status. FINDINGS: Expression of dysfunction-related molecules, such as transcription factors and ICs PD-1, TIGIT, and CD200, followed a hierarchy associated with infection status and effector profile. In vitro responsiveness to PD-L1 blockade varied with defined functions rather than IC levels: frequencies of cells with TH1- and TH17/TH22-, but not TFH-related functions, increased. Cells co-expressing TH1 and TFH functions showed response to ICB, suggesting that the cell's state rather than function dictates responsiveness to PD-L1 blockade. Response to PD-L1 blockade was strongest in viremic participants and reduced after ART initiation. INTERPRETATION: Our data highlight a polarization-specific regulation of IC expression and differing sensitivities of antigen-specific T helper subsets to PD-1-mediated inhibition. This heterogeneity may direct and constrain ICB efficacy in restoring CD4+ T cell function in HIV infection and other diseases. FUNDING: NIH, CIHR, CFI, FRQS.


Subject(s)
B7-H1 Antigen , HIV Infections , B7-H1 Antigen/metabolism , CD4-Positive T-Lymphocytes , Cytokines/metabolism , Humans , Immune Checkpoint Inhibitors , In Situ Hybridization, Fluorescence , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , RNA/therapeutic use , Receptors, Chemokine/metabolism , Receptors, Chemokine/therapeutic use , Receptors, Immunologic/metabolism , Transcription Factors/metabolism
11.
Curr HIV/AIDS Rep ; 19(5): 344-357, 2022 10.
Article in English | MEDLINE | ID: mdl-35867211

ABSTRACT

PURPOSE OF REVIEW: Reducing the risk of HIV-associated neurocognitive disorders (HAND) is an elusive treatment goal for people living with HIV. Combination antiretroviral therapy (cART) has reduced the prevalence of HIV-associated dementia, but milder, disabling HAND is an unmet challenge. As newer cART regimens that more consistently suppress central nervous system (CNS) HIV replication are developed, the testing of adjunctive neuroprotective therapies must accelerate. RECENT FINDINGS: Successes in modifying cART regimens for CNS efficacy (penetrance, chemokine receptor targeting) and delivery (nanoformulations) in pilot studies suggest that improving cART neuroprotection and reducing HAND risk is achievable. Additionally, drugs currently used in neuroinflammatory, neuropsychiatric, and metabolic disorders show promise as adjuncts to cART, likely by broadly targeting neuroinflammation, oxidative stress, aerobic metabolism, and/or neurotransmitter metabolism. Adjunctive cognitive brain therapy and aerobic exercise may provide additional efficacy. Adjunctive neuroprotective therapies, including available FDA-approved drugs, cognitive therapy, and aerobic exercise combined with improved cART offer plausible strategies for optimizing the prevention and treatment of HAND.


Subject(s)
AIDS Dementia Complex , HIV Infections , AIDS Dementia Complex/drug therapy , AIDS Dementia Complex/prevention & control , HIV Infections/complications , HIV Infections/drug therapy , Humans , Neurocognitive Disorders/etiology , Neurocognitive Disorders/prevention & control , Neuroprotection , Neurotransmitter Agents/therapeutic use , Receptors, Chemokine/therapeutic use
12.
Eur Arch Otorhinolaryngol ; 279(11): 5089-5095, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35732904

ABSTRACT

Allergic rhinitis (AR) is an immune-mediated inflammatory condition characterized by immune cell infiltration of the nasal mucosa, with symptoms of rhinorrhea, sneezing, nasal obstruction, and itchiness. Currently, common medication for AR is anti-inflammatory treatment including intranasal steroids, oral, or intranasal anti-histamines, and immunotherapy. These strategies are effective to the majority of patients with AR, but some patients under medication cannot achieve symptom relieve and suffer from bothersome side effects, indicating a demand for novel anti-inflammatory treatment as alternatives. Chemokines, a complex superfamily of small, secreted proteins, were initially recognized for their chemotactic effects on various immune cells. Chemokines constitute both physiological and inflammatory cell positioning systems and mediate cell localization to certain sites via interaction with their receptors, which are expressed on responding cells. Chemokines and their receptors participate in the sensitization, early phase response, and late phase response of AR by promoting inflammatory cell recruitment, differentiation, and allergic mediator release. In this review, we first systemically summarize chemokines and chemokine receptors that are important in AR pathophysiology and then discuss potential strategies targeting chemokines and their receptors for AR therapy.


Subject(s)
Receptors, Chemokine , Rhinitis, Allergic , Anti-Inflammatory Agents/therapeutic use , Chemokines/metabolism , Chemokines/therapeutic use , Humans , Nasal Mucosa/metabolism , Receptors, Chemokine/metabolism , Receptors, Chemokine/therapeutic use , Steroids/therapeutic use
13.
J Biomol Struct Dyn ; 40(23): 13115-13126, 2022.
Article in English | MEDLINE | ID: mdl-34569417

ABSTRACT

C-C chemokine receptor 5 (CCR5), which is part of the chemokine receptor family, is a member of the G protein-coupled receptor superfamily. The interactions of CCR5 with HIV-1 during viral entry position it as an effective therapeutic target for designing potent antiviral therapies. The small-molecule Maraviroc was approved by the FDA as a CCR5 drug in 2007, while clinical trials failure has characterised many of the other CCR5 inhibitors. Thus, the continual identification of potential CCR5 inhibitors is, therefore, warranted. In this study, a structure-based discovery approach has been utilised to screen and retrieved novel potential CCR5 inhibitors from the Asinex antiviral compound (∼ 8,722) database. Explicit lipid-bilayer molecular dynamics simulation, in silico physicochemical and pharmacokinetic analyses, were further performed for the top compounds. A total of 23 structurally diverse compounds with binding scores higher than Maraviroc were selected. Subsequent molecular dynamics (MD) simulations analysis of the top four compounds LAS 51495192, BDB 26405401, BDB 26419079, and LAS 34154543, maintained stability at the CCR5 binding site. Furthermore, these compounds made pertinent interactions with CCR5 residues critical for the HIV-1 gp120-V3 loop binding such as Trp86, Tyr89, Phe109, Tyr108, Glu283 and Tyr251. Additionally, the predicted in silico physicochemical and pharmacokinetic descriptors of the selected compounds were within the acceptable range for drug-likeness. The results suggest positive indications that the identified molecules may represent promising CCR5 entry inhibitors. Further structural optimisations and biochemical testing of the proposed compounds may assist in the discovery of effective HIV-1 therapy.Communicated by Ramaswamy H. Sarma.


Subject(s)
HIV Fusion Inhibitors , HIV Infections , HIV-1 , Humans , Maraviroc/pharmacology , Maraviroc/metabolism , Maraviroc/therapeutic use , CCR5 Receptor Antagonists/pharmacology , CCR5 Receptor Antagonists/chemistry , CCR5 Receptor Antagonists/therapeutic use , Receptors, Chemokine/metabolism , Receptors, Chemokine/therapeutic use , Cyclohexanes/pharmacology , Cyclohexanes/chemistry , Triazoles/pharmacology , Triazoles/chemistry , HIV Fusion Inhibitors/pharmacology , HIV Fusion Inhibitors/chemistry , HIV Fusion Inhibitors/therapeutic use , Receptors, CCR5/chemistry , Receptors, CCR5/metabolism , Receptors, CCR5/therapeutic use , HIV Envelope Protein gp120/metabolism , HIV Infections/drug therapy
14.
Oncol Rep ; 45(3): 809-823, 2021 03.
Article in English | MEDLINE | ID: mdl-33650640

ABSTRACT

Hepatocellular carcinoma (HCC) is a prevalent malignant tumor worldwide, with an unsatisfactory prognosis, although treatments are improving. One of the main challenges for the treatment of HCC is the prevention or management of recurrence and metastasis of HCC. It has been found that chemokines and their receptors serve a pivotal role in HCC progression. In the present review, the literature on the multifactorial roles of exosomes in HCC from PubMed, Cochrane library and Embase were obtained, with a specific focus on the functions and mechanisms of chemokines in HCC. To date, >50 chemokines have been found, which can be divided into four families: CXC, CX3C, CC and XC, according to the different positions of the conserved N­terminal cysteine residues. Chemokines are involved in the inflammatory response, tumor immune response, proliferation, invasion and metastasis via modulation of various signaling pathways. Thus, chemokines and their receptors directly or indirectly shape the tumor cell microenvironment, and regulate the biological behavior of the tumor. In addition, the potential application of chemokines in chemotaxis of exosomes as drug vehicles is discussed. Exosomes containing chemokines or expressing receptors for chemokines may improve chemotaxis to HCC and may thus be exploited for targeted drug delivery.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Chemokines/metabolism , Liver Neoplasms/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Chemokines/therapeutic use , Disease Progression , Exosomes/metabolism , Exosomes/transplantation , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Molecular Targeted Therapy , Receptors, Chemokine/metabolism , Receptors, Chemokine/therapeutic use , Signal Transduction , Tumor Microenvironment
15.
Mediators Inflamm ; 2020: 8198963, 2020.
Article in English | MEDLINE | ID: mdl-33029105

ABSTRACT

The novel coronavirus is not only causing respiratory problems, but it may also damage the heart, kidneys, liver, and other organs; in Wuhan, 14 to 30% of COVID-19 patients have lost their kidney function and now require either dialysis or kidney transplants. The novel coronavirus gains entry into humans by targeting the ACE2 receptor that found on lung cells, which destroy human lungs through cytokine storms, and this leads to hyperinflammation, forcing the immune cells to destroy healthy cells. This is why some COVID-19 patients need intensive care. The inflammatory chemicals released during COVID-19 infection cause the liver to produce proteins that defend the body from infections. However, these proteins can cause blood clotting, which can clog blood vessels in the heart and other organs; as a result, the organs are deprived of oxygen and nutrients which could ultimately lead to multiorgan failure and consequent progression to acute lung injury, acute respiratory distress syndrome, and often death. However, there are novel protein modification tools called the QTY code, which are similar in their structure to antibodies, which could provide a solution to excess cytokines. These synthetic proteins can be injected into the body to bind the excess cytokines created by the cytokine storm; this will eventually remove the excessive cytokines and inhibit the severe symptoms caused by the COVID-19 infection. In this review, we will focus on cytokine storm in COVID-19 patients, their impact on the body organs, and the potential treatment by QTY code-designed detergent-free chemokine receptors.


Subject(s)
Coronavirus Infections/complications , Coronavirus Infections/immunology , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/therapy , Pneumonia, Viral/complications , Pneumonia, Viral/immunology , Receptors, Chemokine/therapeutic use , Betacoronavirus , COVID-19 , Coronavirus Infections/drug therapy , Coronavirus Infections/therapy , Cytokine Release Syndrome/immunology , Cytokines/antagonists & inhibitors , Drug Design , Humans , Inflammation Mediators/blood , Inflammation Mediators/immunology , Models, Molecular , Multiple Organ Failure/etiology , Multiple Organ Failure/immunology , Multiple Organ Failure/therapy , Pandemics , Pneumonia, Viral/therapy , Protein Engineering , Protein Modification, Translational , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , SARS-CoV-2 , COVID-19 Drug Treatment
16.
Viruses ; 10(8)2018 08 20.
Article in English | MEDLINE | ID: mdl-30127279

ABSTRACT

US28 is one of four G protein coupled receptors (GPCRs) encoded by human cytomegalovirus (HCMV). The US28 protein (pUS28) is a potent signaling molecule that alters a variety of cellular pathways that ultimately alter the host cell environment. This viral GPCR is expressed not only in the context of lytic replication but also during viral latency, highlighting its multifunctional properties. pUS28 is a functional GPCR, and its manipulation of multiple signaling pathways likely impacts HCMV pathogenesis. Herein, we will discuss the impact of pUS28 on both lytic and latent infection, pUS28-mediated signaling and its downstream consequences, and the influence this viral GPCR may have on disease states, including cardiovascular disease and cancer. We will also discuss the potential for and progress towards exploiting pUS28 as a novel therapeutic to combat HCMV.


Subject(s)
Cardiovascular Diseases/virology , Cytomegalovirus Infections/virology , Cytomegalovirus/pathogenicity , Gene Expression Regulation, Viral , Host-Pathogen Interactions , Neoplasms/virology , Receptors, Chemokine/genetics , Viral Proteins/genetics , Cardiovascular Diseases/pathology , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/pathology , Humans , Models, Molecular , Neoplasms/pathology , Protein Structure, Secondary , Receptors, Chemokine/therapeutic use , Signal Transduction , Viral Proteins/therapeutic use , Virus Latency/genetics , Virus Replication/genetics
17.
Article in Japanese | MEDLINE | ID: mdl-27320932

ABSTRACT

Chemokines induce migration of inflammatory cells. In the synovial tissue of rheumatoid arthritis (RA), abundant chemokines are expressed, which contribute migration of lymphocytes and monocytes/macrophages, stimulation of synovial cells, and angiogenesis. Blockade of CCL2, CCL3, CCL5, CCR1, CCR9, CXCL2, CXCL5, CXCL13, CXCL16, CXCR3, CXCR4, CXCR7, and CX3CL1 showed improvement of arthritis of animal models. Moreover, CCR1 antagonist and anti-CXCL10 antibody reduced arthritis of patients with RA. Chemokine is a promising target for RA therapy.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/etiology , Chemokines/antagonists & inhibitors , Inflammation Mediators/antagonists & inhibitors , Receptors, Chemokine/therapeutic use , Animals , Antibodies, Monoclonal/therapeutic use , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cell Movement , Chemokine CXCL10/immunology , Chemokines/metabolism , Disease Models, Animal , Humans , Inflammation Mediators/metabolism , Lymphocytes/pathology , Macrophages/pathology , Molecular Targeted Therapy , Monocytes/pathology , Neovascularization, Pathologic , Receptors, CCR1/antagonists & inhibitors , Synovial Membrane/blood supply , Synovial Membrane/cytology , Synovial Membrane/metabolism
18.
J. investig. allergol. clin. immunol ; 25(1): 26-33, 2015. ilus, tab
Article in English | IBECS | ID: ibc-134344

ABSTRACT

Background: Findings regarding the associations between the CC motif chemokine ligand 5 (CCL5) -403G/A and -28C/G polymorphisms and asthma risk are controversial. We performed a meta-analysis to determine whether CCL5 polymorphisms are associated with asthma risk. Methods: We searched the Pubmed, Embase, Chinese National Knowledge Infrastructure (CNKI), and Wanfang databases for studies published before June 2013. The strength of associations was calculated using ORs with 95% CIs. Results: Twenty case-control studies were included in this meta-analysis. We did not observe a significant association between the CCL5 -403G/A polymorphism and asthma risk (OR, 1.10; 95% CI, 0.93-1.30; P=.25). The CCL5 -28C/G polymorphism, however, was associated with a significantly elevated asthma risk (OR, 1.17; 95% CI, 1.02-1.33; P=.02). Subgroup analyses found that the CCL5 -28C/G polymorphism was significantly associated with asthma risk in Asians (OR, 1.16; 95% CI, 1.01-1.33; P=.04) and children (OR, 1.29; 95% CI, 1.03-1.63; P=.03). Conclusions: This meta-analysis suggests that the CCL5 -28C/G polymorphism is a risk factor for asthma (AU)


Introducción: Existen discrepancias entre la asociación del riesgo de padecer asma y diferentes polimorfismos del ligando de la quimiocina CC5 (CCL5). En este trabajo se ha realizado un meta-análisis para determinar si los polimorfismos CCL5-403G / A y CCL5-28C / G se asocian con el riesgo de asma bronquial. Métodos: Se utilizaron diversas bases de datos para realizar las búsquedas de estudios publicados antes de junio de 2013, incluyendo: PubMed, EMBASE, CNKI (Infraestructura del Conocimiento Nacional Chino) y Wanfang Se calcularon los odd ratios combinados (OR) con intervalos de confianza del 95% (IC). Resultados: Se incluyeron un total de 20 estudios de casos y controles. No se encontró una asociación significativa entre el polimorfismo CCL5-403G / A y el riesgo de asma (OR = 1,10, IC del 95%: 0,93 a 1,30, p = 0,25). Por el contrario, el polimorfismo CCL5-28C / G, se asoció con un riesgo significativamente elevado de asma (OR = 1,17, IC del 95%: 1,02 a 1,33, p = 0,02). En los análisis de subgrupos, el riesgo de asma fue significativamente mayor en los asiáticos con el polimorfismo CCL5-28C / G (OR = 1.16, 95% IC 1,01-1,33, P = 0,04) y los niños (OR = 1.29, 95% IC 1,03-1,63, P = 0,03). Conclusiones: Este meta-análisis sugiere que el polimorfismo CCL5-28C / G es un factor de riesgo significativo para padecer asma bronquial (AU)


Subject(s)
Humans , Child , Adult , Amplified Fragment Length Polymorphism Analysis/classification , Amplified Fragment Length Polymorphism Analysis/methods , Asthma/diagnosis , Asthma/metabolism , Receptors, Chemokine/biosynthesis , Lymphocytes/cytology , Luciferases/administration & dosage , Amplified Fragment Length Polymorphism Analysis/standards , Amplified Fragment Length Polymorphism Analysis , Asthma/genetics , Asthma/prevention & control , Receptors, Chemokine/therapeutic use , Lymphocytes/pathology , Luciferases/supply & distribution
19.
Trends Mol Med ; 18(6): 304-10, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22554906

ABSTRACT

Inflammatory responses now have a defined central role in cancer cell growth, invasion, and metastases. Anti-inflammatory proteins from viruses target key stages in immune response pathways and have potential as novel therapeutics for cancer, including highly potent virus-derived inhibitors of protease, chemokine, cytokine, and apoptotic cascades that have been identified. Serine proteases, in addition to their conventional roles in thrombosis, thrombolysis, and apoptotic pathways, are essential regulators of inflammation and are associated with developing cancers. Chemokines drive other inflammatory response pathways with central roles in cell invasion and activation as well as establishing the microenvironment of tumors, modulating immune cell infiltration, cancer cell proliferation, metastasis, and angiogenesis. This review focuses on the mechanisms of action and potential for application of viral immunomodulatory proteins as anticancer therapeutics.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Neoplasms/drug therapy , Viral Proteins/therapeutic use , Animals , Anti-Inflammatory Agents/immunology , Anti-Inflammatory Agents/metabolism , Chemokines/metabolism , Humans , Immunomodulation , Mice , Neoplasms/immunology , Neoplasms/metabolism , Protein Binding , Receptors, Chemokine/immunology , Receptors, Chemokine/metabolism , Receptors, Chemokine/therapeutic use , Serpins/immunology , Serpins/metabolism , Serpins/therapeutic use , Viral Proteins/immunology , Viral Proteins/metabolism , Viruses/immunology , Viruses/metabolism
20.
Am J Respir Cell Mol Biol ; 45(1): 72-80, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20833968

ABSTRACT

CC chemokines play an important role in the pathogenesis of idiopathic pulmonary fibrosis. Few studies have evaluated the efficacy of therapeutically targeting CC chemokines and their receptors during interstitial lung diseases. In the present study, the therapeutic effects of Evasin-1, a tick-derived chemokine-binding protein that has high affinity for CCL3/microphage inflammatory protein (MIP)-1α, was investigated in a murine model of bleomycin-induced lung fibrosis. CCL3/MIP-1α concentrations in lung homogenates increased significantly with time after bleomycin challenge, and this was accompanied by increased number of leukocytes and elevated levels of CCL2/monocyte chemoattractant protein (MCP)-1, CCL5/regulated upon activation, normal T cell expressed and secreted, TNF-α and transforming growth factor-ß(1), and pulmonary fibrosis. Administration of evasin-1 on a preventive (from the day of bleomycin administration) or therapeutic (from Day 8 after bleomycin) schedule decreased number of leukocytes in the lung, reduced levels of TNF-α and transforming growth factor-ß(1), and attenuated lung fibrosis. These protective effects were similar to those observed in CCL3/MIP-1α-deficient mice. In conclusion, targeting CCL3/MIP-1α by treatment with evasin-1 is beneficial in the context of bleomycin-induced lung injury, even when treatment is started after the fibrogenic insult. Mechanistically, evasin-1 treatment was associated with decreased recruitment of leukocytes and production of fibrogenic cytokines. Modulation of CCL3/MIP-1α function by evasin-1 could be useful for the treatment of idiopathic pulmonary fibrosis.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Bleomycin/adverse effects , Chemokine CCL3/antagonists & inhibitors , Chemokine CCL3/immunology , Pulmonary Fibrosis/drug therapy , Receptors, Chemokine/therapeutic use , Animals , Antibiotics, Antineoplastic/administration & dosage , Bleomycin/pharmacology , Chemokine CCL3/genetics , Chemokine CCL3/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Leukocytes/immunology , Leukocytes/metabolism , Leukocytes/pathology , Male , Mice , Mice, Knockout , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Receptors, Chemokine/chemistry , Rhipicephalus sanguineus/chemistry , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...