Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.452
Filter
1.
J Clin Invest ; 134(17)2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39225100

ABSTRACT

Sarcoidosis is a multiorgan granulomatous disease that lacks diagnostic biomarkers and targeted treatments. Using blood and skin from patients with sarcoid and non-sarcoid skin granulomas, we discovered that skin granulomas from different diseases exhibit unique immune cell recruitment and molecular signatures. Sarcoid skin granulomas were specifically enriched for type 1 innate lymphoid cells (ILC1s) and B cells and exhibited molecular programs associated with formation of mature tertiary lymphoid structures (TLSs), including increased CXCL12/CXCR4 signaling. Lung sarcoidosis granulomas also displayed similar immune cell recruitment. Thus, granuloma formation was not a generic molecular response. In addition to tissue-specific effects, patients with sarcoidosis exhibited an 8-fold increase in circulating ILC1s, which correlated with treatment status. Multiple immune cell types induced CXCL12/CXCR4 signaling in sarcoidosis, including Th1 T cells, macrophages, and ILCs. Mechanistically, CXCR4 inhibition reduced sarcoidosis-activated immune cell migration, and targeting CXCR4 or total ILCs attenuated granuloma formation in a noninfectious mouse model. Taken together, our results show that ILC1s are a tissue and circulating biomarker that distinguishes sarcoidosis from other skin granulomatous diseases. Repurposing existing CXCR4 inhibitors may offer a new targeted treatment for this devastating disease.


Subject(s)
Granuloma , Immunity, Innate , Receptors, CXCR4 , Sarcoidosis , Receptors, CXCR4/immunology , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Animals , Humans , Mice , Sarcoidosis/immunology , Sarcoidosis/pathology , Granuloma/immunology , Granuloma/pathology , Skin Diseases/immunology , Skin Diseases/pathology , Female , Chemokine CXCL12/immunology , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Lymphocytes/immunology , Lymphocytes/pathology , Male , Skin/immunology , Skin/pathology , Signal Transduction/immunology
2.
Elife ; 132024 Sep 09.
Article in English | MEDLINE | ID: mdl-39248648

ABSTRACT

CXCR4 is a ubiquitously expressed chemokine receptor that regulates leukocyte trafficking and arrest in both homeostatic and pathological states. It also participates in organogenesis, HIV-1 infection, and tumor development. Despite the potential therapeutic benefit of CXCR4 antagonists, only one, plerixafor (AMD3100), which blocks the ligand-binding site, has reached the clinic. Recent advances in imaging and biophysical techniques have provided a richer understanding of the membrane organization and dynamics of this receptor. Activation of CXCR4 by CXCL12 reduces the number of CXCR4 monomers/dimers at the cell membrane and increases the formation of large nanoclusters, which are largely immobile and are required for correct cell orientation to chemoattractant gradients. Mechanistically, CXCR4 activation involves a structural motif defined by residues in TMV and TMVI. Using this structural motif as a template, we performed in silico molecular modeling followed by in vitro screening of a small compound library to identify negative allosteric modulators of CXCR4 that do not affect CXCL12 binding. We identified AGR1.137, a small molecule that abolishes CXCL12-mediated receptor nanoclustering and dynamics and blocks the ability of cells to sense CXCL12 gradients both in vitro and in vivo while preserving ligand binding and receptor internalization.


Subject(s)
Chemokine CXCL12 , Receptors, CXCR4 , Receptors, CXCR4/metabolism , Receptors, CXCR4/chemistry , Chemokine CXCL12/metabolism , Allosteric Regulation , Humans , Animals , Protein Binding , Protein Domains , Models, Molecular
3.
J Clin Invest ; 134(17)2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39225095

ABSTRACT

Sarcoidosis is an inflammatory disease characterized by immune cell-rich granulomas that form in multiple organs. In this issue of the JCI, Sati and colleagues used scRNA-seq and spatial transcriptomics of skin samples from patients with sarcoidosis and non-sarcoidosis granulomatous disease to identify upregulation of a stromal-immune CXCL12/CXCR4 axis and accumulation of type 1 innate lymphoid cells (ILC1s) in sarcoidosis. The accumulation of ILC1s in skin and blood was specific to patients with sarcoidosis and not observed in other granulomatous diseases. The authors used a mouse model of lung granuloma to show that ILCs contribute to granuloma formation and that blockade of CXCR4 reduced the formation of granulomas, providing a proof of concept that sarcoidosis may be treated by CXCR4 blockade to prevent the progression of disease in patients. These results suggest ILC1s could serve as a diagnostic biomarker in sarcoidosis and a potential therapeutic target.


Subject(s)
Biomarkers , Immunity, Innate , Lymphocytes , Receptors, CXCR4 , Sarcoidosis , Humans , Animals , Mice , Sarcoidosis/immunology , Sarcoidosis/pathology , Biomarkers/metabolism , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Receptors, CXCR4/immunology , Lymphocytes/immunology , Lymphocytes/metabolism , Chemokine CXCL12/metabolism , Chemokine CXCL12/genetics , Chemokine CXCL12/immunology
4.
Int J Nanomedicine ; 19: 9213-9226, 2024.
Article in English | MEDLINE | ID: mdl-39263631

ABSTRACT

Introduction: Targeting, imaging, and treating tumors represent major clinical challenges. Developing effective theranostic agents to address these issues is an urgent need. Methods: We introduce an "all-in-one" tumor-targeted theranostic platform using CuFeSe2-based composite nanoparticles (CuFeSe2@PA) for magnetic resonance (MR) and computed tomography (CT) dual model imaging-guided hyperthermia tumor ablation. Plerixafor (AMD3100) is bonded to the surface of CuFeSe2 as a targeting unit. Due to the robust interaction between AMD3100 and the overexpressed Chemokine CXC type receptor 4 (CXCR4) on the membrane of 4T1 cancer cells, CuFeSe2@PA specifically recognizes 4T1 cancer cells, enriching the tumor region. Results: CuFeSe2@PA serves as a contrast agent for T2-weighted MR imaging (relaxivity value of 1.61 mM-1 s-1) and CT imaging. Moreover, it effectively suppresses tumor growth through photothermal therapy (PTT) owing to its high photothermal conversion capability and stability, with minimized side effects demonstrated both in vitro and in vivo. Discussion: CuFeSe2@PA nanoparticles show potential as dual-mode imaging contrast agents for MR and CT and provide an effective means of tumor treatment through photothermal therapy. The surface modification with Plerixafor enhances the targeting ability of the nanoparticles, performing more significant efficacy and biocompatibility in the 4T1 cancer cell model. The study demonstrates that CuFeSe2@PA is a promising multifunctional theranostic platform with clinical application potential.


Subject(s)
Copper , Magnetic Resonance Imaging , Photothermal Therapy , Receptors, CXCR4 , Theranostic Nanomedicine , Tomography, X-Ray Computed , Animals , Receptors, CXCR4/metabolism , Theranostic Nanomedicine/methods , Photothermal Therapy/methods , Cell Line, Tumor , Magnetic Resonance Imaging/methods , Mice , Copper/chemistry , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Mice, Inbred BALB C , Female , Humans , Contrast Media/chemistry , Nanoparticles/chemistry , Cyclams/pharmacology , Cyclams/chemistry , Benzylamines/chemistry
5.
Int J Mol Sci ; 25(17)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39273392

ABSTRACT

The identification of new compounds with potential activity against CXC chemokine receptor type 4 (CXCR4) has been broadly studied, implying several chemical families, particularly AMD3100 derivatives. Molecular modeling has played a pivotal role in the identification of new active compounds. But, has its golden age ended? A virtual library of 450,000 tetraamines of general structure 8 was constructed by using five spacers and 300 diamines, which were obtained from the corresponding commercially available cyclic amines. Diversity selection was performed to guide the virtual screening of the former database and to select the most representative set of compounds. Molecular docking on the CXCR4 crystal structure allowed us to rank the selection and identify those candidate molecules with potential antitumor activity against diffuse large B-cell lymphoma (DLBCL). Among them, compound A{17,18} stood out for being a non-symmetrical structure, synthetically feasible, and with promising activity against DLBCL in in vitro experiments. The focused study of symmetrical-related compounds allowed us to identify potential pre-hits (IC50~20 µM), evidencing that molecular design is still relevant in the development of new CXCR4 inhibitor candidates.


Subject(s)
Antineoplastic Agents , Receptors, CXCR4 , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Drug Design , Lymphoma, Large B-Cell, Diffuse/drug therapy , Models, Molecular , Molecular Docking Simulation , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/chemistry , Receptors, CXCR4/metabolism , Structure-Activity Relationship
6.
Brain Behav ; 14(9): e70039, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39295108

ABSTRACT

BACKGROUND: Hypoxia inducible factor-1α (HIF-1α) is a sensitive indicator of oxygen homeostasis, of which the expression elevates following hypoxia/ischemia. This study reveals the specific mechanisms underlying the effects of HIF-1α on ischemic stroke (IS). METHODS: IS model was established using middle cerebral artery occlusion (MCAO)-modeled male rats and oxygen glucose deprivation/reoxygenation (OGD/R)-treated mice hippocampal cells HT22, followed by the silencing of HIF-1α and the overexpression of C-X-C motif chemokine receptor 4 (CXCR4) and nuclear factor-kappa B (NF-κB). Following the surgery, Garcia's grading scale was applied for neurological evaluation. Cerebral infarcts and injuries were visualized using 2,3,5-triphenyltetrazolium chloride and hematoxylin-eosin staining. The levels of tumor necrosis factor-α, Interleukin (IL)-6, IL-1ß, malondialdehyde, and 8-hydroxy-2'-deoxyguanosine, were calculated via ELISA. MTT assay and lactate dehydrogenase (LDH) assay kit were adopted to determine the viability and cytotoxicity of OGD/R-modeled cells. Reactive oxygen species (ROS) generation was evaluated using a 2'-7'dichlorofluorescin diacetate (DCFH-DA) probe. The levels of HIF-1α, CXCR4, and NF-κB p65 were quantified via Western blot and immunofluorescence, respectively. RESULTS: HIF-1α knockdown improved Garcia's score, attenuated the cerebral infarct, inflammation, and ROS generation, and alleviated the levels of inflammatory cytokines and CXCR4/NF-κB p65 in MCAO-modeled rats. Such effects were reversed following the overexpression of CXCR4 and NF-κB. Also, in OGD/R-treated HT22 cells, HIF-1α silencing diminished the cytotoxicity and ROS production and reduced the expressions of CXCR4/NF-κB p65, while promoting viability. However, CXCR4/NF-κB p65 overexpression did the opposite. CONCLUSION: HIF-1α knockdown alleviates inflammation and oxidative stress in IS through the CXCR4/NF-κB pathway.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit , Inflammation , Ischemic Stroke , NF-kappa B , Oxidative Stress , Rats, Sprague-Dawley , Receptors, CXCR4 , Animals , Male , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Rats , Oxidative Stress/physiology , NF-kappa B/metabolism , Ischemic Stroke/metabolism , Ischemic Stroke/genetics , Inflammation/metabolism , Mice , Signal Transduction/physiology , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/genetics , Disease Models, Animal , Reactive Oxygen Species/metabolism , Gene Knockdown Techniques
7.
Sci Adv ; 10(38): eadq1476, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39292787

ABSTRACT

Cross-talk between hematopoietic stem and progenitor cells (HSPCs) and bone marrow (BM) cells is critical for homing and sustained engraftment after transplantation. In particular, molecular and physical adaptation of sinusoidal endothelial cells (ECs) promote HSPC BM occupancy; however, signals that govern these events are not well understood. Extracellular vesicles (EVs) are mediators of cell-cell communication crucial in shaping tissue microenvironments. Here, we demonstrate that integrin α4ß7 on murine HSPC EVs targets uptake into ECs. In BM ECs, HSPC EVs induce up-regulation of C-C motif chemokine receptor 2 (CCR2) ligands that synergize with CXCL12-CXCR4 signaling to promote BM homing. In nonirradiated murine models, marrow preconditioning with HSPC EVs or recombinant CCR2 ligands improves homing and early graft occupancy after transplantation. These findings identify a role for HSPC EVs in remodeling ECs, newly define CCR2-dependent graft homing, and inform novel translational conditioning strategies to improve HSPC transplantation.


Subject(s)
Bone Marrow , Extracellular Vesicles , Hematopoietic Stem Cells , Receptors, CCR2 , Animals , Receptors, CCR2/metabolism , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Mice , Extracellular Vesicles/metabolism , Bone Marrow/metabolism , Hematopoietic Stem Cell Transplantation , Endothelial Cells/metabolism , Endothelial Cells/cytology , Signal Transduction , Cell Movement , Chemokine CXCL12/metabolism , Receptors, CXCR4/metabolism , Bone Marrow Cells/metabolism , Bone Marrow Cells/cytology , Mice, Inbred C57BL
8.
Sci Rep ; 14(1): 21925, 2024 09 20.
Article in English | MEDLINE | ID: mdl-39300240

ABSTRACT

Glioblastoma (GBM) is the most aggressive glial tumor of the adult brain, associated with invariably fatal outcome, and a deeper understanding of the underlying malignant mechanisms is necessary to address the current therapeutic failure. We previously demonstrated the role of the CXCL12/CXCR4 axis in GBM cell migration and resistance to ionizing radiation. The atypical chemokine receptor ACKR3, responsible for CXCL12 scavenging, was previously suggested as additional important player in the context of GBM. Following validation of the detection tools, we observed that ACKR3 is expressed within GBM patient tumor tissue, distributed in diverse cell types. In contrast to CXCR4, ACKR3 expression in patient-derived stem-like cells (GSCs) remains however low, while ACKR3 gene expression by tumor cells appears to be modulated by the in-vivo environment. Using overexpression models, we also showed that in vitro ACKR3 had no significant direct effect on cell proliferation or invasion. Altogether, these results suggest that in vitro ACKR3 plays a minor role in malignant GBM cell biology and that its expression is possibly regulated by in-vivo influences. The subtle and multifaceted functions ACKR3 could exert in GBM should therefore only be tackled within a comprehensive tumor microenvironment considering tumoral but also non-tumoral cells.


Subject(s)
Brain Neoplasms , Glioblastoma , Receptors, CXCR , Glioblastoma/pathology , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Receptors, CXCR/metabolism , Receptors, CXCR/genetics , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Cell Proliferation , Cell Line, Tumor , Cell Movement/genetics , Chemokine CXCL12/metabolism , Chemokine CXCL12/genetics , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Tumor Microenvironment/genetics
9.
Front Immunol ; 15: 1387566, 2024.
Article in English | MEDLINE | ID: mdl-39253088

ABSTRACT

Introduction: G-protein coupled receptors (GPCRs) expressed on neutrophils regulate their mobilization from the bone marrow into the blood, their half-live in the circulation, and their pro- and anti-inflammatory activities during inflammation. Chronic kidney disease (CKD) is associated with systemic inflammatory responses, and neutrophilia is a hallmark of CKD onset and progression. Nonetheless, the role of neutrophils in CKD is currently unclear. Methods: Blood and renal tissue were collected from non-dialysis CKD (grade 3 - 5) patients to evaluate GPCR neutrophil expressions and functions in CKD development. Results: CKD patients presented a higher blood neutrophil-to-lymphocyte ratio (NLR), which was inversely correlated with the glomerular filtration rate (eGFR). A higher frequency of neutrophils expressing the senescent GPCR receptor (CXCR4) and activation markers (CD18+CD11b+CD62L+) was detected in CKD patients. Moreover, CKD neutrophils expressed higher amounts of GPCR formyl peptide receptors (FPR) 1 and 2, known as neutrophil pro- and anti-inflammatory receptors, respectively. Cytoskeletal organization, migration, and production of reactive oxygen species (ROS) by CKD neutrophils were impaired in response to the FPR1 agonist (fMLP), despite the higher expression of FPR1. In addition, CKD neutrophils presented enhanced intracellular, but reduced membrane expression of the protein Annexin A1 (AnxA1), and an impaired ability to secrete it into the extracellular compartment. Secreted and phosphorylated AnxA1 is a recognized ligand of FPR2, pivotal in anti-inflammatory and efferocytosis effects. CKD renal tissue presented a low number of neutrophils, which were AnxA1+. Conclusion: Together, these data highlight that CKD neutrophils overexpress GPCRs, which may contribute to an unbalanced aging process in the circulation, migration into inflamed tissues, and efferocytosis.


Subject(s)
Neutrophils , Receptors, Formyl Peptide , Renal Insufficiency, Chronic , Humans , Neutrophils/immunology , Neutrophils/metabolism , Receptors, Formyl Peptide/metabolism , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/immunology , Male , Female , Middle Aged , Aged , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Receptors, G-Protein-Coupled/metabolism , Reactive Oxygen Species/metabolism , Receptors, Lipoxin/metabolism , Receptors, CXCR4/metabolism
10.
PLoS Pathog ; 20(9): e1012472, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39226327

ABSTRACT

Warts, Hypogammaglobulinemia, Infections, and Myelokathexis (WHIM) syndrome is a rare primary immunodeficiency disease in humans caused by a gain of function in CXCR4, mostly due to inherited heterozygous mutations in CXCR4. One major clinical symptom of WHIM patients is their high susceptibility to human papillomavirus (HPV) induced disease, such as warts. Persistent high risk HPV infections cause 5% of all human cancers, including cervical, anogenital, head and neck and some skin cancers. WHIM mice bearing the same mutation identified in WHIM patients were created to study the underlying causes for the symptoms manifest in patients suffering from the WHIM syndrome. Using murine papillomavirus (MmuPV1) as an infection model in mice for HPV-induced disease, we demonstrate that WHIM mice are more susceptible to MmuPV1-induced warts (papillomas) compared to wild type mice. Namely, the incidence of papillomas is higher in WHIM mice compared to wild type mice when mice are exposed to low doses of MmuPV1. MmuPV1 infection facilitated both myeloid and lymphoid cell mobilization in the blood of wild type mice but not in WHIM mice. Higher incidence and larger size of papillomas in WHIM mice correlated with lower abundance of infiltrating T cells within the papillomas. Finally, we demonstrate that transplantation of bone marrow from wild type mice into WHIM mice normalized the incidence and size of papillomas, consistent with the WHIM mutation in hematopoietic cells contributing to higher susceptibility of WHIM mice to MmuPV1-induced disease. Our results provide evidence that MmuPV1 infection in WHIM mice is a powerful preclinical infectious model to investigate treatment options for alleviating papillomavirus infections in WHIM syndrome.


Subject(s)
Papillomavirus Infections , Primary Immunodeficiency Diseases , Warts , Animals , Mice , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Warts/immunology , Warts/virology , Primary Immunodeficiency Diseases/immunology , Primary Immunodeficiency Diseases/genetics , Disease Models, Animal , Papillomaviridae , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/virology , Immunologic Deficiency Syndromes/genetics , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Mice, Inbred C57BL , Disease Susceptibility , Female
11.
Eur J Radiol ; 180: 111694, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39213763

ABSTRACT

PURPOSE: Gliomas account for 75 % of primary malignant CNS tumors. High-grade glioma (CNS WHO grades 3 and 4) have an unfavorable treatment response and poor outcome. CXCR4 is a G protein-coupled receptor that plays an important part in the signaling pathway between cancer cells and tumor microenvironment. CXCR4 overexpression has been shown in a variety of cancers. In this study, we evaluate the potential value of [68Ga]Ga-Pentixafor as a PET/CT CXCR4-probe for in vivo assessment of CXCR4 expression in patients with high-grade glioma and its correlation with tumor grade. MATERIALS AND METHODS: [68Ga]Ga-CXCR4 PET/CT was performed in the prospective single-center study in treatment-naïve biopsy-proven patients with high-grade glioma. The acquired images were analyzed qualitatively and semi-quantitatively. RESULT: A total of 26 patients (mean age: 53.3±14.4 years, 11 women, 15 men) were enrolled. CNS WHO grade 3 pathology was seen in 19 % (5/26) of the sample. The patient-based sensitivity of 68Ga-CXCR4 was 96.2 %. Overall, 28 pathologic lesions were detected, leading to a lesion-based sensitivity of 96.4 %. The median (IQR) SUVmax of grade 4 lesions was substantially greater than the grade 3(3.03(2.5-3.7) vs. 1.51(1.2-1.8), p = 0.0145).). The highest tracer activity of organs -beside bladder as the main excretion reservoir-was in lymphoid tissue of Waldeyer's ring (mean SUVmax: 7.41), and spleen (mean SUVmax: 6.62). CONCLUSION: In conclusion, this new application for [68Ga]Ga-Pentixafor PET tracer exhibits excellent visual and semi-quantitative diagnostic properties. Further studies are warranted.


Subject(s)
Brain Neoplasms , Glioma , Positron Emission Tomography Computed Tomography , Radiopharmaceuticals , Receptors, CXCR4 , Humans , Receptors, CXCR4/metabolism , Female , Male , Glioma/diagnostic imaging , Glioma/metabolism , Glioma/pathology , Positron Emission Tomography Computed Tomography/methods , Middle Aged , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/metabolism , Radiopharmaceuticals/pharmacokinetics , Prospective Studies , Gallium Radioisotopes , Neoplasm Grading , Sensitivity and Specificity , Peptides, Cyclic , Adult , Aged , Coordination Complexes
12.
Pharmacol Res Perspect ; 12(5): e70003, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39207051

ABSTRACT

The ability to accurately measure drug-target interaction is critical for the discovery of new therapeutics. Classical pharmacological bioassays such as radioligand or fluorescent ligand binding assays can define the affinity or Kd of a ligand for a receptor with the lower the Kd, the stronger the binding and the higher the affinity. However, in many drug discovery laboratories today, the target of interest if often artificially upregulated by means of transfection to modify the host cell's genetic makeup. This then potentially invalidates the assumptions of classical pharmacology affinity calculations as the receptor of interest is no longer at normal physiological densities. The CXCR4 receptor is expressed on many different cancer cell types and is associated with metastasis and poor prognosis. Therefore, the CXCR4 receptor is a desirable target for novel therapeutics. In this study, we explore the applicability of the newly developed fluorescently tagged CXCR4 antagonists, IS4-FAM as an investigative tool to study CXCR4 affinity and competitive antagonism in native, non-transfected cancer cells using confocal microscopy and flow cytometry. IS4-FAM directly labels CXCR4 in several cell lines including high CXCR4 expressing SK-MEL-28 (malignant melanoma) and PC3 (metastatic prostate cancer) and lower CXCR4 expressing THP-1 (acute monocytic leukemia) and was competitive with the established CXCR4 antagonist, AMD3100. This highlights the potential of IS4-FAM as a pharmacological tool for drug discovery in native cells lines and tissues.


Subject(s)
Fluorescent Dyes , Receptors, CXCR4 , Receptors, CXCR4/metabolism , Receptors, CXCR4/antagonists & inhibitors , Humans , Cell Line, Tumor , Fluorescent Dyes/chemistry , Flow Cytometry , Binding, Competitive , Microscopy, Confocal , PC-3 Cells , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/metabolism
13.
Clin Nucl Med ; 49(9): 817-821, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39086048

ABSTRACT

PURPOSE: To investigate the role of CXCR4-targeted 68 Ga-pentixafor PET/CT imaging in inflammatory bowel disease (IBD). METHODS: Five IBD patients and 12 control subjects performing 68 Ga-pentixafor PET/CT examinations were included. 68 Ga-pentixafor PET/CT imaging and endoscopic findings were recorded and compared. The semiquantitative parameters of 68 Ga-pentixafor uptake by the lesion segments in IBD patients and the normal intestines in the control were investigated. RESULTS: Among these 5 IBD patients, endoscopy successfully examined a total of 26 intestinal segments, with 13 segments showing endoscopic lesions. 68 Ga-pentixafor PET/CT was positive in all endoscopy-proven lesions (13/13). Additionally, 68 Ga-pentixafor PET/CT revealed the lesions in small intestines and colons that cannot be reached by endoscopy due to severe stenosis, and mesenteric lymphadenitis accompanied IBD. The SUV max of the lesion segments in IBD patients was significantly higher than that of the normal intestines in the control group (median, 3.15 [range, 1.61-6.26] vs 1.67 [1.18-2.29], P < 0.001). Moreover, the SUV max ratios of the lesion segments/liver or blood pool were higher when compared with the control (2.20 [1.13-3.26] vs 0.85 [0.54-1.20]; 1.66 [0.94-2.95] vs 0.67 [0.52-1.04]; P ≤ 0.001). CONCLUSIONS: 68 Ga-pentixafor PET/CT can be a potentially valuable tool to assess the active intestinal lesions of IBD with high sensitivity. Moreover, this noninvasive approach does not require fasting or bowel preparation, offering good tolerance and safety.


Subject(s)
Inflammatory Bowel Diseases , Positron Emission Tomography Computed Tomography , Receptors, CXCR4 , Humans , Male , Female , Inflammatory Bowel Diseases/diagnostic imaging , Middle Aged , Receptors, CXCR4/metabolism , Adult , Aged , Coordination Complexes , Peptides, Cyclic/pharmacokinetics
14.
Free Radic Biol Med ; 223: 237-249, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39094710

ABSTRACT

Chronic prostatitis-induced excessive inflammation and oxidative stress (OS) damage substantially affect men's quality of life. However, its treatment remains a major clinical challenge. Therefore, the identification of drugs that can decrease chronic prostatitis and oxidative stress targets is urgent and essential. CXCR4 is a classic chemokine receptor that is crucially associated with the occurrence and development of inflammation. This investigation aimed to elucidate how CXCR4 affects prostatitis regression and progression. The effect of CXCR4 on chronic prostatitis was evaluated by HE staining, immunohistochemistry, immunofluorescence, PCR, and TUNEL analyses. Furthermore, CXCR4 influence on metabolism was also evaluated by monitoring body weight, body temperature, food intake, and LC/MS. Additionally, chromatin immunoprecipitation, Western blot, and double luciferase reporter gene assays were carried out to elucidate the mechanism by which CXCR4 modulates Fads2 transcription by PPARγ. Lastly, ROS, DHE, mito-tracker, and ATP were utilized to validate the α-linolenic acid's protective effect against OS in prostate epithelial cells. It was revealed that the inhibition of CXCR4 can effectively alleviate prostatitis in mice. Furthermore, downregulating CXCR4 expression can markedly reduce the inflammatory cell infiltration in mouse prostates, decrease the elevated levels of DNA damage markers,MDA and 4-HNE, and mitigate apoptosis of prostatic epithelial cells. Moreover, treatment of CXCR4 knockdown mice with a PPARγ inhibitor revealed different degrees of changes in the above phenotypes. Mechanistically, the PPARγ protein translocates to the nucleus and serves as a transcription factor to regulate Fads2 expression, thereby altering PUFA metabolism. Additionally, in vitro experiments indicated that α-linolenic acid can effectively alleviate OS damage and RWPE-1 cell apoptosis by protecting mitochondrial function and enhancing the antioxidant capacity of prostatic epithelial cells. In conclusion, reducing the levels of CXCR4 can alleviate inflammation and OS damage in chronic prostatitis.


Subject(s)
Fatty Acid Desaturases , Oxidative Stress , PPAR gamma , Prostatitis , Receptors, CXCR4 , Male , Animals , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Mice , Prostatitis/metabolism , Prostatitis/pathology , Prostatitis/genetics , Prostatitis/drug therapy , PPAR gamma/metabolism , PPAR gamma/genetics , Fatty Acid Desaturases/genetics , Fatty Acid Desaturases/metabolism , Humans , Disease Models, Animal , Apoptosis , Fatty Acids, Unsaturated/metabolism , alpha-Linolenic Acid/pharmacology , alpha-Linolenic Acid/metabolism , Prostate/pathology , Prostate/metabolism , Prostate/drug effects , Mice, Inbred C57BL , Gene Expression Regulation
15.
Int Immunopharmacol ; 140: 112777, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39088923

ABSTRACT

Even while accelerated cardiomyocyte apoptosis is one of the primary causes of cardiac damage, the underlying mechanism is still mostly unknown. In addition to examining potential protective effects of bisoprolol and diosmin against CoCl2-induced cardiac injury, the goal of this study was to identify potential mechanisms regulating the hypoxic cardiac damage caused by cobalt chloride (CoCl2). For a period of 21 days except Cocl2 14 days from the first day of the experiment, rats were split into the following groups: Normal control group, rats received vehicle only (2 ml/kg/day, p.o.), (Cocl2, 150 mg/kg/day, p.o.), bisoprolol (25 mg/kg/day, p.o.); diosmin (100 mg/kg/day, p.o.) and bisoprolol + diosmin + Cocl2 groups. At the end of the experimental period, serum was taken for estimation of cardiac function, lipid profile, and pro/anti-inflammatory cytokines. Moreover, tissue samples were collected for evaluation of oxidative stress, endothelial dysfunction, α-SMA, PKC-α, MiR-143-3P, MAPK, ERK5, MCP-1, CXCR4, Orai-1, and STIM-1. Diosmin and bisoprolol, either alone or in combination, enhance heart function by reducing abnormalities in the electrocardiogram and the hypotension brought on by CoCl2. Additionally, they significantly ameliorate endothelial dysfunction by downregulating the cardiac expressions of α-SMA, PKC-α, MiR-143-3P, MAPK, ERK5, MCP-1, CXCR4, Orai-1, and STIM-1. Bisoprolol and diosmin produced modulatory activity against inflammatory state, redox balance, and atherogenic index concurrently. Together, diosmin and bisoprolol, either alone or in combination, significantly reduced all the cardiac alterations brought on by CoCl2. The capacity to obstruct hypoxia-induced α-SMA, PKC-α, MiR-143-3P/MAPK/MCP-1, MiR-143-3P/ERK5/CXCR4, Orai-1/STIM-1 signaling activation, as well as their anti-inflammatory, antioxidant, and anti-apoptotic properties, may be responsible for these cardio-protective results.


Subject(s)
Bisoprolol , Cardiotoxicity , Cobalt , Diosmin , MicroRNAs , ORAI1 Protein , Receptors, CXCR4 , Signal Transduction , Animals , Cobalt/toxicity , Male , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Cardiotoxicity/drug therapy , Rats , Bisoprolol/pharmacology , Bisoprolol/therapeutic use , Signal Transduction/drug effects , MicroRNAs/metabolism , MicroRNAs/genetics , Diosmin/pharmacology , Diosmin/therapeutic use , ORAI1 Protein/metabolism , ORAI1 Protein/genetics , Rats, Wistar , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Apoptosis/drug effects , Oxidative Stress/drug effects , Chemokine CCL2
16.
Biomolecules ; 14(8)2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39199390

ABSTRACT

The treatment of patients diagnosed with hematologic malignancies typically includes hematopoietic stem cell transplantation (HSCT) as part of a therapeutic standard of care. The primary graft source of hematopoietic stem and progenitor cells (HSPCs) for HSCT is mobilized from the bone marrow into the peripheral blood of allogeneic donors or patients. More recently, these mobilized HSPCs have also been the source for gene editing strategies to treat diseases such as sickle-cell anemia. For a HSCT to be successful, it requires the infusion of a sufficient number of HSPCs that are capable of adequate homing to the bone marrow niche and the subsequent regeneration of stable trilineage hematopoiesis in a timely manner. Granulocyte-colony-stimulating factor (G-CSF) is currently the most frequently used agent for HSPC mobilization. However, it requires five or more daily infusions to produce an adequate number of HSPCs and the use of G-CSF alone often results in suboptimal stem cell yields in a significant number of patients. Furthermore, there are several undesirable side effects associated with G-CSF, and it is contraindicated for use in sickle-cell anemia patients, where it has been linked to serious vaso-occlusive and thrombotic events. The chemokine receptor CXCR4 and the cell surface integrin α4ß1 (very late antigen 4 (VLA4)) are both involved in the homing and retention of HSPCs within the bone marrow microenvironment. Preclinical and/or clinical studies have shown that targeted disruption of the interaction of the CXCR4 or VLA4 receptors with their endogenous ligands within the bone marrow niche results in the rapid and reversible mobilization of HSPCs into the peripheral circulation and is synergistic when combined with G-CSF. In this review, we discuss the roles CXCR4 and VLA4 play in bone marrow homing and retention and will summarize more recent development of small-molecule CXCR4 and VLA4 inhibitors that, when combined, can synergistically improve the magnitude, quality and convenience of HSPC mobilization for stem cell transplantation and ex vivo gene therapy after the administration of just a single dose. This optimized regimen has the potential to afford a superior alternative to G-CSF for HSPC mobilization.


Subject(s)
Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells , Integrin alpha4beta1 , Receptors, CXCR4 , Humans , Receptors, CXCR4/metabolism , Receptors, CXCR4/antagonists & inhibitors , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/cytology , Integrin alpha4beta1/metabolism , Integrin alpha4beta1/antagonists & inhibitors , Animals , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte Colony-Stimulating Factor/metabolism , Hematopoietic Stem Cell Transplantation
17.
Biochem Pharmacol ; 227: 116457, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39098732

ABSTRACT

The chemokine receptor CXCR4 is involved in the development and migration of stem and immune cells but is also implicated in tumor progression and metastasis for a variety of cancers. Antagonizing ligand (CXCL12)-induced CXCR4 signaling is, therefore, of therapeutic interest. Currently, there are two small-molecule CXCR4 antagonists on the market for the mobilization of hematopoietic stem cells. Other molecules with improved potencies and safety profiles are being developed for different indications, including cancer. Moreover, multiple antagonistic nanobodies targeting CXCR4 displayed similar or better potencies as compared to the CXCR4-targeting molecule AMD3100 (Plerixafor), which was further enhanced through avid binding of bivalent derivatives. In this study, we aimed to compare the affinities of various multivalent nanobody formats which might be differently impacted by avidity. By fusion to a flexible GS-linker, Fc-region of human IgG1, different C4bp/CLR multimerization domains, or via site-directed conjugation to a trivalent linker scaffold, we generated different types of multivalent nanobodies with varying valencies ranging from bivalent to decavalent. Of these, C-terminal fusion, especially to human Fc, was most advantageous with a 2-log-fold and 3-log-fold increased potency in inhibiting CXCL12-mediated Gαi- or ß-arrestin recruitment, respectively. Overall, we describe strategies for generating multivalent and high-potency CXCR4 antagonistic nanobodies able to induce receptor clustering and conclude that fusion to an Fc-tail results in the highest avidity effect irrespective of the hinge linker.


Subject(s)
Receptors, CXCR4 , Single-Domain Antibodies , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Receptors, CXCR4/immunology , Humans , Single-Domain Antibodies/pharmacology , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/immunology , Animals , Chemokine CXCL12/metabolism , Chemokine CXCL12/antagonists & inhibitors , Chemokine CXCL12/immunology , HEK293 Cells , Antibody Affinity
18.
J Cancer Res Ther ; 20(4): 1241-1250, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39206986

ABSTRACT

AIMS: Papillary thyroid cancer (PTC) is a serious threat to human health worldwide, while metastasis in the early phase limits therapeutic success and leads to poor survival outcomes. The CXC chemokine receptor type 4 (CXCR4) plays an important role in many cellular movements such as transcriptional modulation, cell skeleton rearrangement, and cell migration, and the change in CXCR4 levels are crucial in various diseases including cancer. In this study, we explored the role of CXCR4 in the migration and invasion of PTC and investigated the potential mechanisms underlying its effects. SUBJECTS AND METHODS: We analyzed the expression levels of CXCR4 in PTC tissues and cell lines. Would healing migration, Transwell invasion assay in vitro, and tail-vein lung metastasis assay In vivo were performed to evaluated the migration and invasion abilities of PTC cells with stable CXCR4 knockdown or overexpression. Signal transducers and activators of transcription (STAT3) signaling pathway-related protein expressions were examined by Western blotting assays. RESULTS: The results showed that CXCR4 was highly expressed in PTC cell lines and PTC tissues. CXCR4 knockdown in PTC cells dampened the migration, invasion, and epithelial-mesenchymal transition (EMT), whereas CXCR4 overexpression enhanced these properties. In vivo, we also found that CXCR4 promoted the metastasis of PTC. Mechanistic studies showed that CXCR4 played these vital roles through the STAT3 signaling pathway. Furthermore, PTC patients with high CXCR4 or p-STAT3 expression correlated with aggressive clinical characteristics such as extrathyroidal extension (ETE), and lymph node metastasis (LNM). CONCLUSIONS: We provided evidence that CXCR4 might activate the STAT3 signaling pathway and further promote PTC development. Thus, CXCR4 might be a novel therapeutic target for PTC.


Subject(s)
Cell Movement , Epithelial-Mesenchymal Transition , Neoplasm Invasiveness , Receptors, CXCR4 , Signal Transduction , Thyroid Cancer, Papillary , Thyroid Neoplasms , Animals , Female , Humans , Male , Mice , Middle Aged , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Thyroid Cancer, Papillary/pathology , Thyroid Cancer, Papillary/metabolism , Thyroid Cancer, Papillary/genetics , Thyroid Neoplasms/pathology , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/genetics , Xenograft Model Antitumor Assays
19.
Cells ; 13(16)2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39195225

ABSTRACT

Colorectal cancer (CRC) is a leading cause of cancer mortality worldwide, and cancer-associated fibroblasts (CAFs) play a major role in the tumor microenvironment (TME), which facilitates the progression of CRC. It is critical to understand how CAFs promote the progression of CRC for the development of novel therapeutic approaches. The purpose of this study was to understand how CAF-derived stromal-derived factor-1 (SDF-1) and its interactions with the corresponding C-X-C motif chemokine receptor 4 (CXCR4) promote CRC progression. Our study focused on their roles in promoting tumor cell migration and invasion and their effects on the characteristics of cancer stem cells (CSCs), which ultimately impact patient outcomes. Here, using in vivo approaches and clinical histological samples, we analyzed the influence of secreted SDF-1 on CRC progression, especially in terms of tumor cell behavior and stemness. We demonstrated that CAF-secreted SDF-1 significantly enhanced CRC cell migration and invasion through paracrine signaling. In addition, the overexpression of SDF-1 in CRC cell lines HT29 and HCT-116 triggered these cells to generate autocrine SDF-1 signaling, which further enhanced their CSC characteristics, including those of migration, invasion, and spheroid formation. An immunohistochemical study showed a close relationship between SDF-1 and CXCR4 expression in CRC tissue, and this significantly affected patient outcomes. The administration of AMD3100, an inhibitor of CXCR4, reversed the entire phenomenon. Our results strongly suggest that targeting this signaling axis in CRC is a feasible approach to attenuating tumor progression, and it may, therefore, serve as an alternative treatment method to improve the prognosis of patients with CRC, especially those with advanced, recurrent, or metastatic CRC following standard therapy.


Subject(s)
Autocrine Communication , Cancer-Associated Fibroblasts , Cell Movement , Chemokine CXCL12 , Colorectal Neoplasms , Neoplastic Stem Cells , Paracrine Communication , Receptors, CXCR4 , Signal Transduction , Humans , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/genetics , Chemokine CXCL12/metabolism , Receptors, CXCR4/metabolism , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Animals , Neoplasm Invasiveness , Mice , Tumor Microenvironment , Cell Line, Tumor , HCT116 Cells , Male , Female , HT29 Cells
20.
Cell Rep ; 43(8): 114578, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39093700

ABSTRACT

CXCR4 binding of its endogenous agonist CXCL12 leads to diverse functions, including bone marrow retention of hematopoietic progenitors and cancer metastasis. However, the structure of the CXCL12-bound CXCR4 remains unresolved despite available structures of CXCR4 in complex with antagonists. Here, we present the cryoelectron microscopy (cryo-EM) structure of the CXCL12-CXCR4-Gi complex at an overall resolution of 2.65 Å. CXCL12 forms a 1:1 stoichiometry complex with CXCR4, following the two-site model. The first 8 amino acids of mature CXCL12 are crucial for CXCR4 activation by forming polar interactions with minor sub-pocket residues in the transmembrane binding pocket. The 3.2-Å distance between V3 of CXCL12 and the "toggle switch" W6.48 marks the deepest insertion among all chemokine-receptor pairs, leading to conformational changes of CXCR4 for G protein activation. These results, combined with functional assays and computational analysis, provide the structural basis for CXCR4 activation by CXCL12.


Subject(s)
Chemokine CXCL12 , Cryoelectron Microscopy , Protein Binding , Receptors, CXCR4 , Receptors, CXCR4/metabolism , Receptors, CXCR4/chemistry , Chemokine CXCL12/metabolism , Chemokine CXCL12/chemistry , Cryoelectron Microscopy/methods , Humans , Models, Molecular , Binding Sites , HEK293 Cells
SELECTION OF CITATIONS
SEARCH DETAIL