Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.544
Filter
1.
Clin Transl Sci ; 17(7): e13876, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38963161

ABSTRACT

Plerixafor is a CXCR4 antagonist approved in 2008 by the FDA for hematopoietic stem cell collection. Subsequently, plerixafor has shown promise as a potential pathogen-agnostic immunomodulator in a variety of preclinical animal models. Additionally, investigator-led studies demonstrated plerixafor prevents viral and bacterial infections in patients with WHIM syndrome, a rare immunodeficiency with aberrant CXCR4 signaling. Here, we investigated whether plerixafor could be repurposed to treat sepsis or severe wound infections, either alone or as an adjunct therapy. In a Pseudomonas aeruginosa lipopolysaccharide (LPS)-induced zebrafish sepsis model, plerixafor reduced sepsis mortality and morbidity assessed by tail edema. There was a U-shaped response curve with the greatest effect seen at 0.1 µM concentration. We used Acinetobacter baumannii infection in a neutropenic murine thigh infection model. Plerixafor did not show reduced bacterial growth at 24 h in the mouse thigh model, nor did it amplify the effects of a rifampin antibiotic therapy, in varying regimens. While plerixafor did not mitigate or treat bacterial wound infections in mice, it did reduce sepsis mortality in zebra fish. The observed mortality reduction in our LPS model of zebrafish was consistent with prior research demonstrating a mortality benefit in a murine model of sepsis. However, based on our results, plerixafor is unlikely to be successful as an adjunct therapy for wound infections. Further research is needed to better define the scope of plerixafor as a pathogen-agnostic therapy. Future directions may include the use of longer acting CXCR4 antagonists, biased CXCR4 signaling, and optimization of animal models.


Subject(s)
Benzylamines , Cyclams , Disease Models, Animal , Heterocyclic Compounds , Receptors, CXCR4 , Sepsis , Zebrafish , Animals , Cyclams/pharmacology , Cyclams/administration & dosage , Benzylamines/pharmacology , Sepsis/drug therapy , Sepsis/microbiology , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/administration & dosage , Mice , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Thigh/microbiology , Acinetobacter Infections/drug therapy , Acinetobacter Infections/microbiology , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/isolation & purification , Pseudomonas Infections/drug therapy , Pseudomonas Infections/microbiology , Female , Lipopolysaccharides , Wound Infection/microbiology , Wound Infection/drug therapy , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use
2.
Front Immunol ; 15: 1383136, 2024.
Article in English | MEDLINE | ID: mdl-38979422

ABSTRACT

Multiple myeloma (MM) is a plasma cell disease with a preferential bone marrow (BM) tropism. Enforced expression of tissue-specific chemokine receptors has been shown to successfully guide adoptively-transferred CAR NK cells towards the malignant milieu in solid cancers, but also to BM-resident AML and MM. For redirection towards BM-associated chemokine CXCL12, we armored BCMA CAR-NK-92 as well as primary NK cells with ectopic expression of either wildtype CXCR4 or a gain-of-function mutant CXCR4R334X. Our data showed that BCMA CAR-NK-92 and -primary NK cells equipped with CXCR4 gained an improved ability to migrate towards CXCL12 in vitro. Beyond its classical role coordinating chemotaxis, CXCR4 has been shown to participate in T cell co-stimulation, which prompted us to examine the functionality of CXCR4-cotransduced BCMA-CAR NK cells. Ectopic CXCR4 expression enhanced the cytotoxic capacity of BCMA CAR-NK cells, as evidenced by the ability to eliminate BCMA-expressing target cell lines and primary MM cells in vitro and through accelerated cytolytic granule release. We show that CXCR4 co-modification prolonged BCMA CAR surface deposition, augmented ZAP-70 recruitment following CAR-engagement, and accelerated distal signal transduction kinetics. BCMA CAR sensitivity towards antigen was enhanced by virtue of an enhanced ZAP-70 recruitment to the immunological synapse, revealing an increased propensity of CARs to become triggered upon CXCR4 overexpression. Unexpectedly, co-stimulation via CXCR4 occurred in the absence of CXCL12 ligand-stimulation. Collectively, our findings imply that co-modification of CAR-NK cells with tissue-relevant chemokine receptors affect adoptive NK cell therapy beyond improved trafficking and retention within tumor sites.


Subject(s)
B-Cell Maturation Antigen , Chemokine CXCL12 , Immunotherapy, Adoptive , Killer Cells, Natural , Multiple Myeloma , Receptors, CXCR4 , Receptors, Chimeric Antigen , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Humans , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , B-Cell Maturation Antigen/immunology , B-Cell Maturation Antigen/metabolism , B-Cell Maturation Antigen/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Immunotherapy, Adoptive/methods , Chemokine CXCL12/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic
3.
Cell Mol Life Sci ; 81(1): 296, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992165

ABSTRACT

Next to its classical role in MHC II-mediated antigen presentation, CD74 was identified as a high-affinity receptor for macrophage migration inhibitory factor (MIF), a pleiotropic cytokine and major determinant of various acute and chronic inflammatory conditions, cardiovascular diseases and cancer. Recent evidence suggests that CD74 is expressed in T cells, but the functional relevance of this observation is poorly understood. Here, we characterized the regulation of CD74 expression and that of the MIF chemokine receptors during activation of human CD4+ T cells and studied links to MIF-induced T-cell migration, function, and COVID-19 disease stage. MIF receptor profiling of resting primary human CD4+ T cells via flow cytometry revealed high surface expression of CXCR4, while CD74, CXCR2 and ACKR3/CXCR7 were not measurably expressed. However, CD4+ T cells constitutively expressed CD74 intracellularly, which upon T-cell activation was significantly upregulated, post-translationally modified by chondroitin sulfate and could be detected on the cell surface, as determined by flow cytometry, Western blot, immunohistochemistry, and re-analysis of available RNA-sequencing and proteomic data sets. Applying 3D-matrix-based live cell-imaging and receptor pathway-specific inhibitors, we determined a causal involvement of CD74 and CXCR4 in MIF-induced CD4+ T-cell migration. Mechanistically, proximity ligation assay visualized CD74/CXCR4 heterocomplexes on activated CD4+ T cells, which were significantly diminished after MIF treatment, pointing towards a MIF-mediated internalization process. Lastly, in a cohort of 30 COVID-19 patients, CD74 surface expression was found to be significantly upregulated on CD4+ and CD8+ T cells in patients with severe compared to patients with only mild disease course. Together, our study characterizes the MIF receptor network in the course of T-cell activation and reveals CD74 as a novel functional MIF receptor and MHC II-independent activation marker of primary human CD4+ T cells.


Subject(s)
Antigens, Differentiation, B-Lymphocyte , CD4-Positive T-Lymphocytes , COVID-19 , Histocompatibility Antigens Class II , Intramolecular Oxidoreductases , Lymphocyte Activation , Macrophage Migration-Inhibitory Factors , SARS-CoV-2 , Humans , Antigens, Differentiation, B-Lymphocyte/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class II/immunology , Macrophage Migration-Inhibitory Factors/metabolism , Macrophage Migration-Inhibitory Factors/genetics , Lymphocyte Activation/immunology , SARS-CoV-2/metabolism , SARS-CoV-2/immunology , COVID-19/immunology , COVID-19/metabolism , COVID-19/pathology , Intramolecular Oxidoreductases/metabolism , Intramolecular Oxidoreductases/genetics , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Cell Movement , Male , Female , Middle Aged , Receptors, Immunologic
4.
Front Cell Infect Microbiol ; 14: 1415123, 2024.
Article in English | MEDLINE | ID: mdl-38994006

ABSTRACT

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


Subject(s)
Disease Progression , Genetic Variation , HIV Infections , HIV-1 , Viral Load , Humans , HIV-1/genetics , Male , Female , Adult , China/epidemiology , HIV Infections/virology , Prospective Studies , CD4 Lymphocyte Count , Middle Aged , Receptors, CCR5/genetics , Receptors, CCR5/metabolism , Receptors, CXCR4/genetics , Young Adult , CD4-Positive T-Lymphocytes/immunology , Risk Factors
5.
Int J Mol Sci ; 25(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39000362

ABSTRACT

Exposure to microgravity during spaceflight induces the alterations in endothelial cell function associated with post-flight cardiovascular deconditioning. PIEZO1 is a major mechanosensitive ion channel that regulates endothelial cell function. In this study, we used a two-dimensional clinostat to investigate the expression of PIEZO1 and its regulatory mechanism on human umbilical vein endothelial cells (HUVECs) under simulated microgravity. Utilizing quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot analysis, we observed that PIEZO1 expression was significantly increased in response to simulated microgravity. Moreover, we found microgravity promoted endothelial cells migration by increasing expression of PIEZO1. Proteomics analysis highlighted the importance of C-X-C chemokine receptor type 4(CXCR4) as a main target molecule of PIEZO1 in HUVECs. CXCR4 protein level was increased with simulated microgravity and decreased with PIEZO1 knock down. The mechanistic study showed that PIEZO1 enhances CXCR4 expression via Ca2+ influx. In addition, CXCR4 could promote endothelial cell migration under simulated microgravity. Taken together, these results suggest that the upregulation of PIEZO1 in response to simulated microgravity regulates endothelial cell migration due to enhancing CXCR4 expression via Ca2+ influx.


Subject(s)
Cell Movement , Human Umbilical Vein Endothelial Cells , Ion Channels , Receptors, CXCR4 , Weightlessness Simulation , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Humans , Ion Channels/metabolism , Ion Channels/genetics , Cell Movement/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Calcium/metabolism , Endothelial Cells/metabolism , Gene Expression Regulation
6.
Front Immunol ; 15: 1411141, 2024.
Article in English | MEDLINE | ID: mdl-39040098

ABSTRACT

Warts, Hypogammaglobulinemia, Infections, Myelokathexis (WHIM) syndrome is a rare, combined immunodeficiency disease predominantly caused by gain-of-function variants in the CXCR4 gene that typically results in truncation of the carboxyl terminus of C-X-C chemokine receptor type 4 (CXCR4) leading to impaired leukocyte egress from bone marrow to peripheral blood. Diagnosis of WHIM syndrome continues to be challenging and is often made through clinical observations and/or genetic testing. Detection of a pathogenic CXCR4 variant in an affected individual supports the diagnosis of WHIM syndrome but relies on an appropriate annotation of disease-causing variants. Understanding the genotypic-phenotypic associations in WHIM syndrome has the potential to improve time to diagnosis and guide appropriate clinical management, resulting in a true example of precision medicine. This article provides an overview of the spectrum of CXCR4 variants in WHIM syndrome and summarizes the various lines of clinical and functional evidence that can support interpretation of newly identified variants.


Subject(s)
Primary Immunodeficiency Diseases , Receptors, CXCR4 , Warts , Receptors, CXCR4/genetics , Humans , Warts/genetics , Warts/diagnosis , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/diagnosis , Mutation , Genetic Association Studies , Genetic Predisposition to Disease , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/diagnosis
7.
Sci Rep ; 14(1): 15764, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982161

ABSTRACT

C-X-C motif chemokine receptor 4 (CXCR4) is a promising therapeutic target of breast cancer because it is overexpressed on cell surface of all molecular subtypes of breast cancer including triplenegative breast cancer (TNBC). Herein, CXCR4 antagonistic peptide-NaGdF4 nanodot conjugates (termed as anti-CXCR4-NaGdF4 NDs) have been constructed for magnetic resonance imaging (MRI)-guided biotherapy of TNBC through conjugation of the C-X-C Motif Chemokine 12 (CXCL12)-derived cyclic peptide with tryptone coated NaGdF4 nanodots (5 ± 0.5 nm in diameter, termed as Try-NaGdF4 NDs). The as-prepared anti-CXCR4-NaGdF4 NDs exhibits high longitudinal relaxivity (r1) value (21.87 mM-1S-1), reasonable biocompatibility and good tumor accumulation ability. The features of anti-CXCR4-NaGdF4 NDs improve the tumor-MRI sensitivity and facilitate tumor biotherapy after injection in mouse-bearing MDA-MB-231 tumor model in vivo. MRI-guided biotherapy using anti-CXCR4-NaGdF4 NDs enables to suppress 46% tumor growth. In addition, about 47% injection dose of anti-CXCR4-NaGdF4 NDs is found in the mouse urine at 24 h post-injection. These findings demonstrate that anti-CXCR4-NaGdF4 NDs enable to be used as renal clearable nanomedicine for biotherapy and MRI of breast cancer.


Subject(s)
Breast Neoplasms , Magnetic Resonance Imaging , Receptors, CXCR4 , Receptors, CXCR4/metabolism , Animals , Female , Magnetic Resonance Imaging/methods , Humans , Mice , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/therapy , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Cell Line, Tumor , Gadolinium/chemistry , Chemokine CXCL12/metabolism , Mice, Nude , Mice, Inbred BALB C , Nanoparticles/chemistry , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Xenograft Model Antitumor Assays , Peptides/chemistry
8.
Front Immunol ; 15: 1406532, 2024.
Article in English | MEDLINE | ID: mdl-39035006

ABSTRACT

Heterozygous autosomal dominant mutations in the CXCR4 gene cause WHIM syndrome, a severe combined immunodeficiency disorder. The mutations primarily affect the C-terminal region of the CXCR4 chemokine receptor, specifically several potential phosphorylation sites critical for agonist (CXCL12)-mediated receptor internalization and desensitization. Mutant receptors have a prolonged residence time on the cell surface, leading to hyperactive signaling that is responsible for some of the symptoms of WHIM syndrome. Recent studies have shown that the situation is more complex than originally thought, as mutant WHIM receptors and CXCR4 exhibit different dynamics at the cell membrane, which also influences their respective cellular functions. This review examines the functional mechanisms of CXCR4 and the impact of WHIM mutations in both physiological and pathological conditions.


Subject(s)
Mutation , Primary Immunodeficiency Diseases , Receptors, CXCR4 , Signal Transduction , Warts , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Humans , Primary Immunodeficiency Diseases/genetics , Warts/genetics , Animals , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/immunology , Thrombocytopenia/genetics , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism
9.
Cancer Med ; 13(14): e7471, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39015025

ABSTRACT

BACKGROUND: ZNF384-fusion (Z-fusion) genes were recently identified in B-cell acute lymphoblastic leukemia (B-ALL) and are frequent in Japanese adult patients. The frequency is about 20% in those with Philadelphia chromosome-negative B-ALL. ZNF384 is a transcription factor and Z-fusion proteins have increased transcriptional activity; however, the detailed mechanisms of leukemogenesis of Z-fusion proteins have yet to be clarified. METHODS: We established three transfectants of cell lines expressing different types of Z-fusion proteins, and analyzed their gene expression profile (GEP) by RNA-seq. We also analyzed the GEP of clinical ALL samples using our previous RNA-seq data of 323 Japanese ALL patients. We selected upregulated genes in both Z-fusion gene-expressing transfectants and Z-fusion gene-positive ALL samples, and investigated the binding of Z-fusion proteins to regulatory regions of the candidate genes by ChIP-qPCR. RESULTS: We selected six commonly upregulated genes. After the investigation by ChIP-qPCR, we finally identified CREB5 and RGS1 as direct and common target genes. RGS1 is an inhibitor of CXCL12-CXCR4 signaling that is required for the homing of hematopoietic progenitor cells to the bone marrow microenvironment and development of B cells. Consistent with this, Z-fusion gene transfectants showed impaired migration toward CXCL12. CONCLUSIONS: We identified CREB5 and RGS1 as direct and common transcriptional targets of Z-fusion proteins. The present results provide novel insight into the aberrant transcriptional regulation by Z-fusion proteins.


Subject(s)
Cyclic AMP Response Element-Binding Protein , Oncogene Proteins, Fusion , RGS Proteins , Humans , Cell Line, Tumor , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Gene Expression Regulation, Leukemic , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , RGS Proteins/genetics , RGS Proteins/metabolism , Trans-Activators
10.
Med Sci Monit ; 30: e945471, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38864115

ABSTRACT

The Editors of Medical Science Monitor wish to inform you that the above manuscript has been retracted from publication due to concerns with the credibility and originality of the study, the manuscript content, and the Figure images. Reference: Rongfeng Zhang, Jianwei Liu, Shengpeng Yu, Dong Sun, Xiaohua Wang, Jingshu Fu, Jie Shen, Zhao Xie. Osteoprotegerin (OPG) Promotes Recruitment of Endothelial Progenitor Cells (EPCs) via CXCR4 Signaling Pathway to Improve Bone Defect Repair. Med Sci Monit, 2019; 25: 5572-5579. DOI: 10.12659/MSM.916838.


Subject(s)
Endothelial Progenitor Cells , Osteoprotegerin , Receptors, CXCR4 , Signal Transduction , Endothelial Progenitor Cells/metabolism , Receptors, CXCR4/metabolism , Osteoprotegerin/metabolism , Animals , Bone Regeneration/drug effects , Humans , Bone and Bones/metabolism , Osteogenesis/drug effects , Male , Mice , Wound Healing/drug effects
11.
Elife ; 122024 Jun 18.
Article in English | MEDLINE | ID: mdl-38896451

ABSTRACT

Durable serological memory following vaccination is critically dependent on the production and survival of long-lived plasma cells (LLPCs). Yet, the factors that control LLPC specification and survival remain poorly resolved. Using intravital two-photon imaging, we find that in contrast to most plasma cells (PCs) in the bone marrow (BM), LLPCs are uniquely sessile and organized into clusters that are dependent on APRIL, an important survival factor. Using deep, bulk RNA sequencing, and surface protein flow-based phenotyping, we find that LLPCs express a unique transcriptome and phenotype compared to bulk PCs, fine-tuning expression of key cell surface molecules, CD93, CD81, CXCR4, CD326, CD44, and CD48, important for adhesion and homing. Conditional deletion of Cxcr4 in PCs following immunization leads to rapid mobilization from the BM, reduced survival of antigen-specific PCs, and ultimately accelerated decay of antibody titer. In naïve mice, the endogenous LLPCs BCR repertoire exhibits reduced diversity, reduced somatic mutations, and increased public clones and IgM isotypes, particularly in young mice, suggesting LLPC specification is non-random. As mice age, the BM PC compartment becomes enriched in LLPCs, which may outcompete and limit entry of new PCs into the LLPC niche and pool.


Subject(s)
Plasma Cells , Animals , Mice , Plasma Cells/immunology , Plasma Cells/metabolism , Mice, Inbred C57BL , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/genetics , Cell Survival , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Spatio-Temporal Analysis
12.
Biochem Biophys Res Commun ; 725: 150249, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-38880081

ABSTRACT

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


Subject(s)
CD4 Antigens , HIV Envelope Protein gp120 , HIV-1 , HIV-1/metabolism , Humans , CD4 Antigens/metabolism , CD4 Antigens/chemistry , HIV Envelope Protein gp120/metabolism , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/genetics , Cryoelectron Microscopy , env Gene Products, Human Immunodeficiency Virus/metabolism , env Gene Products, Human Immunodeficiency Virus/chemistry , env Gene Products, Human Immunodeficiency Virus/genetics , Receptors, CCR5/metabolism , Receptors, CCR5/chemistry , Protein Binding , Models, Molecular , Protein Conformation , HIV Infections/virology , HIV Infections/metabolism , Protein Multimerization , Receptors, CXCR4/metabolism , Receptors, CXCR4/chemistry , Asia
13.
Cells ; 13(12)2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38920657

ABSTRACT

The reciprocal modulation between the CXCL12/CXCR4/ACKR3 axis and the STAT3 signaling pathway plays a crucial role in the progression of various diseases and neoplasms. Activation of the CXCL12/CXCR4/ACKR3 axis triggers the STAT3 pathway through multiple mechanisms, while the STAT3 pathway also regulates the expression of CXCL12. This review offers a thorough and systematic analysis of the reciprocal regulatory mechanisms between the CXCL12/CXCR4/ACKR3 signaling axis and the STAT3 signaling pathway in the context of diseases, particularly tumors. It explores the potential clinical applications in tumor treatment, highlighting possible therapeutic targets and novel strategies for targeted tumor therapy.


Subject(s)
Chemokine CXCL12 , Neoplasms , Receptors, CXCR4 , STAT3 Transcription Factor , Signal Transduction , Humans , STAT3 Transcription Factor/metabolism , Receptors, CXCR4/metabolism , Chemokine CXCL12/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Animals , Receptors, CXCR/metabolism , Receptors, CXCR/genetics
14.
J Transl Med ; 22(1): 598, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937806

ABSTRACT

BACKGROUND: Monocyte-derived alveolar macrophages (Mo_AMs) are increasingly recognised as potential pathogenic factors for idiopathic pulmonary fibrosis (IPF). While scRNAseq analysis has proven valuable in the transcriptome profiling of Mo_AMs, the integration analysis of multi-omics may provide additional dimensions of understanding of these cellular populations. METHODS: We performed multi-omics analysis on 116 scRNAseq, 119 bulkseq and five scATACseq lung tissue samples from IPF. We built a large-scale IPF scRNAseq atlas and conducted the Monocle 2/3 as well as the Cellchat to explore the developmental path and intercellular communication on Mo_AMs. We also reported the difference in metabolisms, tissue repair and phagocytosis between Mo_AMs and tissue-resident alveolar macrophages (TRMs). To determine whether Mo_AMs affected pulmonary function, we projected clinical phenotypes (FVC%pred) from the bulkseq dataset onto the scRNAseq atlas. Finally, we used scATATCseq to uncover the upstream regulatory mechanisms and determine key drivers in Mo_AMs. RESULTS: We identified three Mo_AMs clusters and the trajectory analysis further validated the origin of these clusters. Moreover, via the Cellchat analysis, the CXCL12/CXCR4 axis was found to be involved in the molecular basis of reciprocal interactions between Mo_AMs and fibroblasts through the activation of the ERK pathway in Mo_AMs. SPP1_RecMacs (RecMacs, recruited macrophages) were higher in the low-FVC group than in the high-FVC group. Specifically, compared with TRMs, the functions of lipid and energetic metabolism as well as tissue repair were higher in Mo_AMs than TRMs. But, TRMs may have higher level of phagocytosis than TRMs. SPIB (PU.1), JUNB, JUND, BACH2, FOSL2, and SMARCC1 showed stronger association with open chromatin of Mo_AMs than TRMs. Significant upregulated expression and deep chromatin accessibility of APOE were observed in both SPP1_RecMacs and TRMs. CONCLUSION: Through trajectory analysis, it was confirmed that SPP1_RecMacs derived from Monocytes. Besides, Mo_AMs may influence FVC% pred and aggravate pulmonary fibrosis through the communication with fibroblasts. Furthermore, distinctive transcriptional regulators between Mo_AMs and TRMs implied that they may depend on different upstream regulatory mechanisms. Overall, this work provides a global overview of how Mo_AMs govern IPF and also helps determine better approaches and intervention therapies.


Subject(s)
Idiopathic Pulmonary Fibrosis , Macrophages, Alveolar , Monocytes , Humans , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/genetics , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Monocytes/metabolism , Male , Gene Expression Profiling , Female , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Middle Aged , Phenotype , Lung/pathology , Lung/metabolism , Gene Expression Regulation
16.
Eur J Med Chem ; 275: 116594, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38879970

ABSTRACT

Chemokine receptor 4 (CXCR4) is a subtype receptor protein of the GPCR family with a seven-transmembrane structure widely distributed in human tissues. CXCR4 is involved in diseases (e.g., HIV-1 infection), cancer proliferation and metastasis, inflammation signaling pathways, and leukemia, making it a promising drug target. Clinical trials on CXCR4 antagonists mainly focused on peptides and antibodies, with a few small molecule compounds, such as AMD11070 (2) and MSX-122 (3), showing promise in cancer treatment. This perspective discusses the structure-activity relationship (SAR) of CXCR4 and its role in diseases, mainly focusing on the SAR of CXCR4 antagonists. It also explores the standard structural features and target interactions of CXCR4 binding in different disease categories. Furthermore, it investigates various modification strategies to propose potential improvements in the effectiveness of CXCR4 drugs.


Subject(s)
Receptors, CXCR4 , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Humans , Structure-Activity Relationship , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/metabolism , Molecular Structure , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Drug Development
17.
Eur J Med Chem ; 275: 116605, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38885550

ABSTRACT

C-X-C chemokine receptor type 4 (CXCR4) exerts considerable influence on the pathogenesis of inflammatory disorders and offers a potent avenue for drug intervention. This research utilizes a hybrid virtual screening methodology constructed using computer-aided drug design to discover novel CXCR4 inhibitors for the treatment of inflammation. First, a compound library was screened by Lipinski's five rules and adsorption, distribution, metabolism, excretion and toxicity properties. Second, the HypoGen algorithm was used in constructing a 3D-QSAR pharmacophore model and verify it layer by layer, and the obtained optimal pharmacophore 1 (Hypo 1) was used as a 3D query for compound screening. Then, hit compounds were obtained through molecular docking (Libdock and CDOCKER). The toxicity of the compounds to MDA-MB-231 cells was evaluated in vitro, and their binding affinity to the target was evaluated according to how they compete with 12G5 antibody for CXCR4 on the surfaces of the MDA-MB-231 cells. Compound Hit14 showed the strongest binding affinity among the hit compounds and inhibited cell migration and invasion in Matrigel invasion and wound healing assay at a concentration of 100 nM, demonstrating a better effect than AMD3100. Western Blot experiments further showed that Hit14 blocked the CXCR4/CXCL12-mediated phosphorylation of Akt. Meanwhile, cellular thermal displacement assay analysis showed that CXCR4 protein bound to Hit14 had high thermal stability. Finally, through in vivo experiments, we found that Hit14 inhibited mouse ear inflammation and reduced ear swelling and damage. Therefore, Hit14 is a promising drug for the further development of CXCR4 inhibitors for inflammation treatment.


Subject(s)
Inflammation , Molecular Docking Simulation , Receptors, CXCR4 , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Humans , Animals , Mice , Inflammation/drug therapy , Inflammation/metabolism , Drug Discovery , Cell Movement/drug effects , Molecular Structure , Drug Evaluation, Preclinical , Dose-Response Relationship, Drug , Cell Line, Tumor , Quantitative Structure-Activity Relationship , Male
18.
Biomacromolecules ; 25(7): 4569-4580, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38869359

ABSTRACT

Acute myeloid leukemia (AML) is often associated with poor prognosis and survival. Small molecule inhibitors, though widening the treatment landscape, have limited monotherapy efficacy. The combination therapy, however, shows suboptimal clinical outcomes due to low bioavailability, overlapping systemic toxicity and drug resistance. Here, we report that CXCR4-mediated codelivery of the BCL-2 inhibitor venetoclax (VEN) and the FLT3 inhibitor sorafenib (SOR) via T22 peptide-tagged disulfide cross-linked polymeric micelles (TM) achieves synergistic treatment of FLT3-ITD AML. TM-VS with a VEN/SOR weight ratio of 1/4 and T22 peptide density of 20% exhibited an extraordinary inhibitory effect on CXCR4-overexpressing MV4-11 AML cells. TM-VS at a VEN/SOR dosage of 2.5/10 mg/kg remarkably reduced leukemia burden, prolonged mouse survival, and impeded bone loss in orthotopic MV4-11-bearing mice, outperforming the nontargeted M-VS and oral administration of free VEN/SOR. CXCR4-mediated codelivery of BCL-2 and FLT3 inhibitors has emerged as a prospective clinical treatment for FLT3-ITD AML.


Subject(s)
Leukemia, Myeloid, Acute , Proto-Oncogene Proteins c-bcl-2 , Receptors, CXCR4 , Sorafenib , Sulfonamides , fms-Like Tyrosine Kinase 3 , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/genetics , Animals , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Humans , Mice , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Sulfonamides/pharmacology , Sulfonamides/administration & dosage , Sorafenib/pharmacology , Sorafenib/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Micelles
19.
J Med Chem ; 67(12): 10057-10075, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38863440

ABSTRACT

Artificial intelligence (AI) de novo molecular generation provides leads with novel structures for drug discovery. However, the target affinity and synthesizability of the generated molecules present critical challenges for the successful application of AI technology. Therefore, we developed an advanced reinforcement learning model to bridge the gap between the theory of de novo molecular generation and the practical aspects of drug discovery. This model utilizes chemical reaction templates and commercially available building blocks as a starting point and employs forward reaction prediction to generate molecules, while real-time docking and drug-likeness predictions are conducted to ensure synthesizability and drug-likeness. We applied this model to design active molecules targeting the inflammation-related receptor CXCR4 and successfully prepared them according to the AI-proposed synthetic routes. Several molecules exhibited potent anti-CXCR4 and anti-inflammatory activity in subsequent in vitro and in vivo assays. The top-performing compound XVI alleviated symptoms related to inflammatory bowel disease and showed reasonable pharmacokinetic properties.


Subject(s)
Artificial Intelligence , Drug Design , Receptors, CXCR4 , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Humans , Animals , Molecular Docking Simulation , Inflammatory Bowel Diseases/drug therapy , Mice , Drug Discovery , Structure-Activity Relationship , Male , Molecular Structure
20.
PLoS One ; 19(6): e0302530, 2024.
Article in English | MEDLINE | ID: mdl-38905184

ABSTRACT

At present, the mechanism of fluorosis-induced damage to the hepatic system is unclear. Studies have shown that excess fluoride causes some degree of damage to the liver, including inflammation. The SDF-1/CXCR4 signaling axis has been reported to have an impact on the regulation of inflammation in human cells. In this study, we investigated the role of the SDF-1/CXCR4 signaling axis and related inflammatory factors in fluorosis through in vitro experiments on human hepatic astrocytes (LX-2) cultured with sodium fluoride. CCK-8 assays showed that the median lethal dose at 24 h was 2 mmol/l NaF, and these conditions were used for subsequent enzyme-linked immunosorbent assays (ELISAs) and quantitative real-time polymerase chain reaction (qPCR) analysis. The protein expression levels of SDF-1/CXCR4 and the related inflammatory factors nuclear factor-κB (NF-κB), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α) and interleukin 1ß (IL-1ß) were detected by ELISAs from the experimental and control groups. The mRNA expression levels of these inflammatory indicators were also determined by qPCR in both groups. Moreover, the expression levels of these factors were significantly higher in the experimental group than in the control group at both the protein and mRNA levels (P < 0.05). Excess fluorine may stimulate the SDF-1/CXCR4 signaling axis, activating the inflammatory NF-κB signaling pathway and increasing the expression levels of the related inflammatory factors IL-6, TNF-α and IL-1ß. Identification of this mechanism is important for elucidating the pathogenesis of fluorosis-induced liver injury.


Subject(s)
Chemokine CXCL12 , Hepatocytes , Receptors, CXCR4 , Sodium Fluoride , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Humans , Chemokine CXCL12/metabolism , Chemokine CXCL12/genetics , Sodium Fluoride/toxicity , Sodium Fluoride/pharmacology , Hepatocytes/metabolism , Hepatocytes/drug effects , Signal Transduction/drug effects , NF-kappa B/metabolism , Cell Line , Interleukin-1beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Interleukin-6/metabolism , Interleukin-6/genetics , Inflammation/metabolism , Inflammation/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL