Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.548
Filtrer
1.
FASEB J ; 38(15): e23857, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39114953

RÉSUMÉ

Atherosclerotic plaque formation is largely attributed to the impaired efferocytosis, which is known to be associated with the pathologic upregulation of cluster of differentiation 47 (CD47), a key antiphagocytic molecule. By gene expression omnibus (GEO) datasets analysis, we identified that four miRNAs are aberrantly downregulated in atherosclerosis, coronary artery disease, and obesity. Of them, hsa-miR-299-3p (miR-299-3p) was predicted to target the 3'UTR of human CD47 mRNA by bioinformatics analysis. Further, we demonstrated that miR-299-3p negatively regulates CD47 expression by binding to the target sequence "CCCACAU" in the 3'UTR of CD47 mRNA through luciferase reporter assay and site-directed mutagenesis. Additionally, we found that miR-299-3p was downregulated by ~32% in foam cells in response to oxidized low-density lipoprotein (ox-LDL) stimulation, thus upregulating CD47 and contributing to the impaired efferocytosis. Whereas, restoration of miR-299-3p reversed the ox-LDL-induced upregulation of CD47, thereby facilitating efferocytosis. In high-fat diet (HFD) fed ApoE-/- mice, we discovered that miR-299-3p was downregulated thus leading to upregulation of CD47 in abdominal aorta. Conversely, miR-299-3p restoration potently suppressed HFD-induced upregulation of CD47 and promoted phagocytosis of foam cells by macrophages in atherosclerotic plaques, thereby reducing necrotic core, increasing plaque stability, and mitigating atherosclerosis. Conclusively, we identify miR-299-3p as a negative regulator of CD47, and reveal a molecular mechanism whereby the ox-LDL-induced downregulation of miR-299-3p leads to the upregulation of CD47 in foam cells thus contributing to the impaired efferocytosis in atherosclerosis, and propose miR-299-3p can potentially serve as an inhibitor of CD47 to promote efferocytosis and ameliorate atherosclerosis.


Sujet(s)
Athérosclérose , Antigènes CD47 , , microARN , Animaux , Humains , Souris , Régions 3' non traduites , Athérosclérose/métabolisme , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Antigènes CD47/métabolisme , Antigènes CD47/génétique , Alimentation riche en graisse/effets indésirables , Cellules spumeuses/métabolisme , Cellules spumeuses/anatomopathologie , Lipoprotéines LDL/métabolisme , Souris de lignée C57BL , microARN/génétique , microARN/métabolisme
2.
Int J Nanomedicine ; 19: 8253-8270, 2024.
Article de Anglais | MEDLINE | ID: mdl-39157734

RÉSUMÉ

Background: Myocardial infarction (MI) is characterized by irreversible cardiomyocyte death resulting from an inadequate supply of oxygenated blood to the myocardium. Recent studies have indicated that ferroptosis, a form of regulated cell death, exacerbates myocardial injury during MI. Concurrently, the upregulation of CD47 on the surface of damaged myocardium following MI impairs the clearance of dead cells by macrophages, thereby hindering efferocytosis. In this context, simultaneously inhibiting ferroptosis and enhancing efferocytosis may represent a promising strategy to mitigate myocardial damage post-MI. Methods: In this study, we engineered platelet membrane-coated hollow mesoporous silicon nanoparticles (HMSN) to serve as a drug delivery system, encapsulating ferroptosis inhibitor, Ferrostatin-1, along with an anti-CD47 antibody. We aimed to assess the potential of these nanoparticles (designated as Fer-aCD47@PHMSN) to specifically target the site of MI and evaluate their efficacy in reducing cardiomyocyte death and inflammation. Results: The platelet membrane coating on the nanoparticles significantly enhanced their ability to successfully target the site of myocardial infarction (MI). Our findings demonstrate that treatment with Fer-aCD47@PHMSN resulted in a 38.5% reduction in cardiomyocyte ferroptosis under hypoxia, indicated by decreased lipid peroxidation and increased in vitro. Additionally, Fer-aCD47@PHMSN improved cardiomyocyte efferocytosis by approximately 15% in vitro. In MI mice treated with Fer-aCD47@PHMSN, we observed a substantial reduction in cardiomyocyte death (nearly 30%), decreased inflammation, and significant improvement in cardiac function. Conclusion: Our results demonstrated that the cooperation between the two agents induced anti-ferroptosis effects and enhanced dead cardiomyocyte clearance by macrophage as well as anti-inflammation effects. Thus, our nanoparticle Fer-aCD47@PHMSN provides a new therapeutic strategy for targeted therapy of MI.


Sujet(s)
Antigènes CD47 , Ferroptose , Infarctus du myocarde , Myocytes cardiaques , Nanoparticules , Infarctus du myocarde/traitement médicamenteux , Infarctus du myocarde/métabolisme , Ferroptose/effets des médicaments et des substances chimiques , Animaux , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Nanoparticules/composition chimique , Souris , Antigènes CD47/métabolisme , Phagocytose/effets des médicaments et des substances chimiques , Cyclohexylamines/pharmacologie , Cyclohexylamines/composition chimique , Mâle , Phénylènediamines/pharmacologie , Phénylènediamines/composition chimique , Macrophages/effets des médicaments et des substances chimiques , Plaquettes/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Matériaux biomimétiques/composition chimique , Matériaux biomimétiques/pharmacologie , Vecteurs de médicaments/composition chimique , Humains ,
3.
ACS Nano ; 18(33): 22298-22315, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39117621

RÉSUMÉ

A series of successes in RNA interference (RNAi) therapies for liver diseases using lipid nanoparticles and N-acetylgalactosamine have heralded a current era of RNA therapeutics. However, alternative delivery strategies are required to take RNAi out of the comfort zone of hepatocytes. Here we report SIRPα IgV/anti-CD47 siRNA (vS-siCD47) conjugates that selectively and persistently disrupt the antiphagocytic CD47/SIRPα axis in solid tumors. Conjugation of the SIRPα IgV domain protein to siRNAs enables tumor dash through CD47-mediated erythrocyte piggyback, primarily blocking the physical interaction between CD47 on cancer cells and SIRPα on phagocytes. After internalization of the vS-siCD47 conjugates within cancer cells, the detached free-standing anti-CD47 siRNAs subsequently attack CD47 through the RNAi mechanism. The dual-action approach of the vS-siCD47 conjugate effectively overcomes the "don't eat me" barrier and stimulates phagocyte-mediated tumor destruction, demonstrating a highly selective and potent CD47-blocking immunotherapy. This delivery strategy, employing IgV domain protein-siRNA conjugates with a dual mode of target suppression, holds promise for expanding RNAi applications beyond hepatocytes and advancing RNAi-based cancer immunotherapies for solid tumors.


Sujet(s)
Antigènes CD47 , Petit ARN interférent , Récepteurs immunologiques , Antigènes CD47/métabolisme , Antigènes CD47/composition chimique , Humains , Petit ARN interférent/composition chimique , Animaux , Souris , Récepteurs immunologiques/métabolisme , Tumeurs/thérapie , Tumeurs/génétique , Tumeurs/anatomopathologie , Antigènes de différenciation , Lignée cellulaire tumorale
4.
J Exp Clin Cancer Res ; 43(1): 225, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39135122

RÉSUMÉ

BACKGROUND: Long non-coding RNAs (LncRNAs) have been implicated as critical regulators of cancer tumorigenesis and progression. However, their functions and molecular mechanisms in colorectal cancer (CRC) still remain to be further elucidated. METHODS: LINC00460 was identified by differential analysis between human CRC and normal tissues and verified by in situ hybridization (ISH) and qRT-PCR. We investigated the biological functions of LINC00460 in CRC by in vitro and in vivo experiments. We predicted the mechanism and downstream functional molecules of LINC00460 by bioinformatics analysis, and confirmed them by dual luciferase reporter gene assay, RNA immunoprecipitation (RIP), RNA pull-down, etc. RESULTS: LINC00460 was found to be significantly overexpressed in CRC and associated with poor prognosis. Overexpression of LINC00460 promoted CRC cell immune escape and remodeled a suppressive tumor immune microenvironment, thereby promoting CRC proliferation and metastasis. Mechanistic studies showed that LINC00460 served as a molecular sponge for miR-186-3p, and then promoted the expressions of MYC, CD47 and PD-L1 to facilitate CRC cell immune escape. We also demonstrated that MYC upregulated LINC00460 expression at the transcriptional level and formed a positive feedback loop. CONCLUSIONS: The LINC00460/miR-186-3p/MYC feedback loop promotes CRC cell immune escape and subsequently facilitates CRC proliferation and metastasis. Our findings provide novel insight into LINC00460 as a CRC immune regulator, and provide a potential therapeutic target for CRC patients.


Sujet(s)
Antigène CD274 , Antigènes CD47 , Tumeurs colorectales , microARN , ARN long non codant , Humains , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Tumeurs colorectales/immunologie , microARN/génétique , Antigènes CD47/métabolisme , Antigènes CD47/génétique , ARN long non codant/génétique , ARN long non codant/métabolisme , Souris , Antigène CD274/métabolisme , Antigène CD274/génétique , Animaux , Régulation de l'expression des gènes tumoraux , Lignée cellulaire tumorale , Échappement de la tumeur à la surveillance immunitaire/génétique , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Mâle , Femelle , Prolifération cellulaire , Rétrocontrôle physiologique , Pronostic , Souris nude
5.
Cell Rep ; 43(8): 114613, 2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-39116206

RÉSUMÉ

Leptomeningeal metastases (LMs) remain a devastating complication of non-small cell lung cancer (NSCLC), particularly following osimertinib resistance. We conducted single-cell RNA sequencing on cerebrospinal fluid (CSF) from EGFR-mutant NSCLC with central nervous system metastases. We found that macrophages of LMs displayed functional and phenotypic heterogeneity and enhanced immunosuppressive properties. A population of lipid-associated macrophages, namely RNASE1_M, were linked to osimertinib resistance and LM development, which was regulated by Midkine (MDK) from malignant epithelial cells. MDK exhibited significant elevation in both CSF and plasma among patients with LMs, with higher MDK levels correlating to poorer outcomes in an independent cohort. Moreover, MDK could promote macrophage M2 polarization with lipid metabolism and phagocytic function. Furthermore, malignant epithelial cells in CSF, particularly after resistance to osimertinib, potentially achieved immune evasion through CD47-SIRPA interactions with RNASE1_M. In conclusion, we revealed a specific subtype of macrophages linked to osimertinib resistance and LM development, providing a potential target to overcome LMs.


Sujet(s)
Acrylamides , Dérivés de l'aniline , Carcinome pulmonaire non à petites cellules , Résistance aux médicaments antinéoplasiques , Tumeurs du poumon , Macrophages , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Humains , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/traitement médicamenteux , Dérivés de l'aniline/pharmacologie , Dérivés de l'aniline/usage thérapeutique , Acrylamides/pharmacologie , Acrylamides/usage thérapeutique , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Animaux , Souris , Lignée cellulaire tumorale , Femelle , Méningite carcinomateuse/traitement médicamenteux , Méningite carcinomateuse/anatomopathologie , Méningite carcinomateuse/secondaire , Métabolisme lipidique/effets des médicaments et des substances chimiques , Antigènes CD47/métabolisme , Antigènes CD47/génétique , Mâle , Phagocytose/effets des médicaments et des substances chimiques , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Indoles , Pyrimidines
6.
J Pharm Biomed Anal ; 251: 116431, 2024 Dec 15.
Article de Anglais | MEDLINE | ID: mdl-39197208

RÉSUMÉ

The assessment of bioactivity for therapeutic antibody release assay poses challenges, particularly when targeting immune checkpoints. An in vitro bioassay platform was developed using the chimeric antigen receptor on Jurkat cells (Jurkat-CAR) to analyze antibodies targeting immune checkpoints, such as CD47/SIRPα, VEGF/VEGFR1, PD-1/PD-L1, and CD70/CD27. For CD47/SIRPα, the platform involved a Jurkat-CAR cell line expressing the chimeric SIRPα receptor (CarSIRPα). CarSIRPα was created by sequentially fusing the SIRPα extracellular region with the CD8α hinge region, the transmembrane (TM) and intracellular (IC) domains of CD28, and the intracellular signaling domain of CD3ζ. The resulting Jurkat-CarSIRPα cells can undergo "activation-induced cell death (AICD)" upon incubation with purified or cellular CD47, as evidenced by the upregulation of CD69, IL-2, and IFN-γ. Similar results also appeared in Jurkat CarVEGFR1, Jurkat CarPD1 and Jurkat CARCD27 cells. These cells are perfectly utilized for the bioactivity analysis of therapeutic antibody. Our study indicates that the established in vitro assay platform based on Jurkat-CAR has been confirmed repeatedly and has shown robust reproducibility; thus, this platform can be used for screening or for release assays of given antibody drugs targeting immune checkpoints.


Sujet(s)
Dosage biologique , Récepteurs chimériques pour l'antigène , Humains , Cellules Jurkat , Dosage biologique/méthodes , Récepteurs immunologiques/métabolisme , Antigènes CD47/métabolisme , Antigènes CD/immunologie , Interleukine-2 , Interféron gamma , Mort cellulaire/effets des médicaments et des substances chimiques , Antigènes de différenciation
7.
Int J Mol Sci ; 25(16)2024 Aug 17.
Article de Anglais | MEDLINE | ID: mdl-39201643

RÉSUMÉ

An association between high CD47 expression and poor cancer survival has been attributed to its function on malignant cells to inhibit phagocytic clearance. However, CD47 mRNA expression in some cancers lacks correlation or correlates with improved survival. IFT57 encodes an essential primary cilium component and is colinear with CD47 across amniote genomes, suggesting coregulation of these genes. Analysis of The Cancer Genome Atlas datasets identified IFT57 as a top coexpressed gene with CD47 among 1156 human cancer cell lines and in most tumor types. The primary cilium also regulates cancer pathogenesis, and correlations between IFT57 mRNA and survival paralleled those for CD47 in thyroid and lung carcinomas, melanoma, and glioma. CD47 ranked first for coexpression with IFT57 mRNA in papillary thyroid carcinomas, and higher expression of both genes correlated with significantly improved overall survival. CD47 and IFT57 mRNAs were coordinately regulated in thyroid carcinoma cell lines. Transcriptome analysis following knockdown of CD47 or IFT57 in thyroid carcinoma cells identified the cytoskeletal regulator CRACD as a specific target of IFT57. CRACD mRNA expression inversely correlated with IFT57 mRNA and with survival in low-grade gliomas, lung adenocarcinomas, and papillary thyroid carcinomas, suggesting that IFT57 rather than CD47 regulates survival in these cancers.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Antigènes CD47 , Régulation de l'expression des gènes tumoraux , Humains , Antigènes CD47/génétique , Antigènes CD47/métabolisme , Lignée cellulaire tumorale , Analyse de profil d'expression de gènes , Tumeurs/génétique , Tumeurs/mortalité , Tumeurs/anatomopathologie , Tumeurs/métabolisme , ARN messager/génétique , ARN messager/métabolisme , Tumeurs de la thyroïde/génétique , Tumeurs de la thyroïde/anatomopathologie , Tumeurs de la thyroïde/mortalité , Tumeurs de la thyroïde/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/métabolisme
8.
J Adv Res ; 63: 129-158, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39167629

RÉSUMÉ

BACKGROUND: Immunotherapy has emerged as a novel strategy for cancer treatment following surgery, radiotherapy, and chemotherapy. Immune checkpoint blockade and Chimeric antigen receptor (CAR)-T cell therapies have been successful in clinical trials. Cancer cells evade immune surveillance by hijacking inhibitory pathways via overexpression of checkpoint genes. The Cluster of Differentiation 47 (CD47) has emerged as a crucial checkpoint for cancer immunotherapy by working as a "don't eat me" signal and suppressing innate immune signaling. Furthermore, CD47 is highly expressed in many cancer types to protect cancer cells from phagocytosis via binding to SIRPα on phagocytes. Targeting CD47 by either interrupting the CD47-SIRPα axis or combing with other therapies has been demonstrated as an encouraging therapeutic strategy in cancer immunotherapy. Antibodies and small molecules that target CD47 have been explored in pre- and clinical trials. However, formidable challenges such as the anemia and palate aggregation cannot be avoided because of the wide presentation of CD47 on erythrocytes. AIM OF VIEW: This review summarizes the current knowledge on the regulation and function of CD47, and provides a new perspective for immunotherapy targeting CD47. It also highlights the clinical progress of targeting CD47 and discusses challenges and potential strategies. KEY SCIENTIFIC CONCEPTS OF REVIEW: This review provides a comprehensive understanding of targeting CD47 in cancer immunotherapy, it also augments the concept of combination immunotherapy strategies by employing both innate and adaptive immune responses.


Sujet(s)
Antigènes CD47 , Immunothérapie , Tumeurs , Antigènes CD47/métabolisme , Antigènes CD47/immunologie , Humains , Tumeurs/thérapie , Tumeurs/immunologie , Immunothérapie/méthodes , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/immunologie , Animaux , Transduction du signal , Antigènes de différenciation/immunologie , Antigènes de différenciation/métabolisme , Immunité innée , Phagocytose
9.
Sci Immunol ; 9(98): eadk9872, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39121194

RÉSUMÉ

The Aedes aegypti mosquito is a vector of many infectious agents, including flaviviruses such as Zika virus. Components of mosquito saliva have pleomorphic effects on the vertebrate host to enhance blood feeding, and these changes also create a favorable niche for pathogen replication and dissemination. Here, we demonstrate that human CD47, which is known to be involved in various immune processes, interacts with a 34-kilodalton mosquito salivary protein named Nest1. Nest1 is up-regulated in blood-fed female A. aegypti and facilitates Zika virus dissemination in human skin explants. Nest1 has a stronger affinity for CD47 than its natural ligand, signal regulatory protein α, competing for binding at the same interface. The interaction between Nest1 with CD47 suppresses phagocytosis by human macrophages and inhibits proinflammatory responses by white blood cells, thereby suppressing antiviral responses in the skin. This interaction elucidates how an arthropod protein alters the human response to promote arbovirus infectivity.


Sujet(s)
Aedes , Peau , Virus Zika , Aedes/immunologie , Aedes/virologie , Animaux , Humains , Peau/immunologie , Peau/virologie , Virus Zika/immunologie , Virus Zika/physiologie , Femelle , Protéines d'insecte/immunologie , Infection par le virus Zika/immunologie , Protéines et peptides salivaires/immunologie , Vecteurs moustiques/immunologie , Vecteurs moustiques/virologie , Antigènes CD47
10.
Mol Cancer ; 23(1): 166, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39138571

RÉSUMÉ

BACKGROUND: Ovarian cancer (OC) remains one of the most challenging and deadly malignancies facing women today. While PARP inhibitors (PARPis) have transformed the treatment landscape for women with advanced OC, many patients will relapse and the PARPi-resistant setting is an area of unmet medical need. Traditional immunotherapies targeting PD-1/PD-L1 have failed to show any benefit in OC. The CD47/TSP-1 axis may be relevant in OC. We aimed to describe changes in CD47 expression with platinum therapy and their relationship with immune features and prognosis. METHODS: Tumor and blood samples collected from OC patients in the CHIVA trial were assessed for CD47 and TSP-1 before and after neoadjuvant chemotherapy (NACT) and multiplex analysis was used to investigate immune markers. Considering the therapeutic relevance of targeting the CD47/TSP-1 axis, we used the CD47-derived TAX2 peptide to selectively antagonize it in a preclinical model of aggressive ovarian carcinoma. RESULTS: Significant reductions in CD47 expression were observed post NACT. Tumor patients having the highest CD47 expression profile at baseline showed the greatest CD4+ and CD8+ T-cell influx post NACT and displayed a better prognosis. In addition, TSP-1 plasma levels decreased significantly under NACT, and high TSP-1 was associated with a worse prognosis. We demonstrated that TAX2 exhibited a selective and favorable biodistribution profile in mice, localizing at the tumor sites. Using a relevant peritoneal carcinomatosis model displaying PARPi resistance, we demonstrated that post-olaparib (post-PARPi) administration of TAX2 significantly reduced tumor burden and prolonged survival. Remarkably, TAX2 used sequentially was also able to increase animal survival even under treatment conditions allowing olaparib efficacy. CONCLUSIONS: Our study thus (1) proposes a CD47-based stratification of patients who may be most likely to benefit from postoperative immunotherapy, and (2) suggests that TAX2 is a potential alternative therapy for patients relapsing on PARP inhibitors.


Sujet(s)
Marqueurs biologiques tumoraux , Antigènes CD47 , Tumeurs de l'ovaire , Thrombospondine-1 , Antigènes CD47/métabolisme , Femelle , Humains , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/métabolisme , Tumeurs de l'ovaire/anatomopathologie , Marqueurs biologiques tumoraux/métabolisme , Animaux , Souris , Thrombospondine-1/métabolisme , Pronostic , Lignée cellulaire tumorale , Traitement néoadjuvant , Tests d'activité antitumorale sur modèle de xénogreffe , Adulte d'âge moyen , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
11.
Int J Mol Sci ; 25(16)2024 Aug 22.
Article de Anglais | MEDLINE | ID: mdl-39201801

RÉSUMÉ

Despite improvements in vaccination, screening, and treatment, cervical cancer (CC) remains a major healthcare problem on a global scale. The tumor microenvironment (TME) plays an important and controversial role in cancer development, and the mechanism of the tumor's escape from immunological surveillance is still not clearly defined. We aim to investigate the expression of CD68 and CD47 in patients with different histological variants of CC, tumor characteristics, and burden. This is a retrospective cohort study performed on paraffin-embedded tumor tissues from 191 patients diagnosed with CC between 2014 and 2021 at the Medical University Pleven, Bulgaria. Slides for immunohistochemical (IHC) evaluation were obtained, and the expression of CD68 was scored in intratumoral (IT) and stromal (ST) macrophages (CD68+cells) using a three-point scoring scale. The CD47 expression was reported as an H-score. All statistical analyses were performed using R v. 4.3.1 for Windows. Infiltration by CD68-IT cells in the tumor depended on histological type and the expression of CD47. Higher levels of the CD47 H-score were significantly more frequent among patients in the early stage. Higher levels of infiltration by CD68-ST cells were associated with worse prognosis, and the infiltration of CD68-IT cells was associated with reduced risk of death from neoplastic disease. TME is a complex ecosystem that has a major role in the growth and development of tumors. Macrophages are a major component of innate immunity and, when associated with a tumor process, are defined as TAM. Tumor cells try to escape immunological surveillance in three ways, and one of them is reducing immunogenicity by the overexpression of negative coreceptors by T-lymphocytes and their ligands on the surface of tumor cells. One such mechanism is the expression of CD47 in tumor cells, which sends a "don't eat me" signal to the macrophages and, thus, prevents phagocytosis. To our knowledge, this is the first study that has tried to establish the relationship between the CD47 and CD68 expression levels and some clinicopathologic features in CC. We found that the only clinicopathological feature implicating the level of CD68 infiltration was the histological variant of the tumor, and only for CD68-IT-high levels were these observed in SCC. High levels of CD47 expression were seen more frequently in pT1B than pT2A and pT2B in the FIGO I stage than in the FIGO II and III stages. Infiltration by large numbers of CD68-IT cells was much more common among patients with a high expression of CD47 in tumor cells. A high level of infiltration by CD68-ST cells was associated with a worse prognosis, and a high level of infiltration by CD68-ST cells was associated with a lower risk of death from cancer.


Sujet(s)
Antigènes CD , Antigènes de différenciation des myélomonocytes , Marqueurs biologiques tumoraux , Antigènes CD47 , Microenvironnement tumoral , Tumeurs du col de l'utérus , Humains , Tumeurs du col de l'utérus/immunologie , Tumeurs du col de l'utérus/anatomopathologie , Tumeurs du col de l'utérus/métabolisme , Tumeurs du col de l'utérus/mortalité , Femelle , Antigènes CD47/métabolisme , Pronostic , Microenvironnement tumoral/immunologie , Antigènes de différenciation des myélomonocytes/métabolisme , Adulte d'âge moyen , Marqueurs biologiques tumoraux/métabolisme , Antigènes CD/métabolisme , Études rétrospectives , Adulte , Sujet âgé , Macrophages/métabolisme , Macrophages/immunologie , Phagocytose , Macrophages associés aux tumeurs/métabolisme , Macrophages associés aux tumeurs/immunologie ,
12.
Nat Commun ; 15(1): 6043, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39025845

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) is a devastating cancer with dismal prognosis due to distant metastasis, even in the early stage. Using RNA sequencing and multiplex immunofluorescence, here we find elevated expression of mixed lineage kinase domain-like pseudo-kinase (MLKL) and enhanced necroptosis pathway in PDAC from early liver metastasis T-stage (T1M1) patients comparing with non-metastatic (T1M0) patients. Mechanistically, MLKL-driven necroptosis recruits macrophages, enhances the tumor CD47 'don't eat me' signal, and induces macrophage extracellular traps (MET) formation for CXCL8 activation. CXCL8 further initiates epithelial-mesenchymal transition (EMT) and upregulates ICAM-1 expression to promote endothelial adhesion. METs also degrades extracellular matrix, that eventually supports PDAC liver metastasis. Meanwhile, targeting necroptosis and CD47 reduces liver metastasis in vivo. Our study thus reveals that necroptosis facilitates PDAC metastasis by evading immune surveillance, and also suggest that CD47 blockade, combined with MLKL inhibitor GW806742X, may be a promising neoadjuvant immunotherapy for overcoming the T1M1 dilemma and reviving the opportunity for radical surgery.


Sujet(s)
Antigènes CD47 , Carcinome du canal pancréatique , Transition épithélio-mésenchymateuse , Pièges extracellulaires , Tumeurs du foie , Macrophages , Nécroptose , Tumeurs du pancréas , Protein kinases , Humains , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/génétique , Tumeurs du pancréas/immunologie , Tumeurs du foie/secondaire , Tumeurs du foie/métabolisme , Animaux , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/génétique , Souris , Macrophages/métabolisme , Macrophages/immunologie , Lignée cellulaire tumorale , Antigènes CD47/métabolisme , Antigènes CD47/génétique , Protein kinases/métabolisme , Pièges extracellulaires/métabolisme , Molécule-1 d'adhérence intercellulaire/métabolisme , Molécule-1 d'adhérence intercellulaire/génétique , Mâle , Transduction du signal , Femelle , Acrylamides , Sulfonamides
13.
Mol Pharm ; 21(8): 3897-3908, 2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-38959154

RÉSUMÉ

The antitumor strategies based on innate immunity activation have become favored by researchers in recent years. In particular, strategies targeting antiphagocytic signaling blockade to enhance phagocytosis have been widely reported. For example, the addition of prophagocytic signals such as calreticulin could make the strategy significantly more effective. In this study, an antitumor strategy that combines photodynamic therapy (PDT) with CD47 blockade has been reported. This approach promotes the maturation of dendritic cells and the presentation of tumor antigens by PDT-mediated tumor immunogenic cell death, as well as the enhancement of cytotoxic T lymphocyte infiltration in tumor areas and the phagocytic activity of phagocytes. Furthermore, the downregulation and blockage of CD47 protein could further promote phagocytic activity, strengthen the innate immune system, and ultimately elevate the antitumor efficacy and inhibit tumor metastasis.


Sujet(s)
Antigènes CD47 , Cellules dendritiques , Phagocytose , Photothérapie dynamique , Antigènes CD47/antagonistes et inhibiteurs , Antigènes CD47/métabolisme , Photothérapie dynamique/méthodes , Animaux , Souris , Phagocytose/effets des médicaments et des substances chimiques , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/immunologie , Lignée cellulaire tumorale , Tumeurs/traitement médicamenteux , Tumeurs/immunologie , Tumeurs/anatomopathologie , Souris de lignée C57BL , Photosensibilisants/pharmacologie , Photosensibilisants/usage thérapeutique , Humains , Lymphocytes T cytotoxiques/immunologie , Lymphocytes T cytotoxiques/effets des médicaments et des substances chimiques , Immunité innée/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Femelle
14.
Nat Commun ; 15(1): 5759, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38982116

RÉSUMÉ

Type I interferons have been well recognized for their roles in various types of immune cells during tumor immunotherapy. However, their direct effects on tumor cells are less understood. Oxidative phosphorylation is typically latent in tumor cells. Whether oxidative phosphorylation can be targeted for immunotherapy remains unclear. Here, we find that tumor cell responsiveness to type I, but not type II interferons, is essential for CD47-SIRPα blockade immunotherapy in female mice. Mechanistically, type I interferons directly reprogram tumor cell metabolism by activating oxidative phosphorylation for ATP production in an ISG15-dependent manner. ATP extracellular release is also promoted by type I interferons due to enhanced secretory autophagy. Functionally, tumor cells with genetic deficiency in oxidative phosphorylation or autophagy are resistant to CD47-SIRPα blockade. ATP released upon CD47-SIRPα blockade is required for antitumor T cell response induction via P2X7 receptor-mediated dendritic cell activation. Based on this mechanism, combinations with inhibitors of ATP-degrading ectoenzymes, CD39 and CD73, are designed and show synergistic antitumor effects with CD47-SIRPα blockade. Together, these data reveal an important role of type I interferons on tumor cell metabolic reprograming for tumor immunotherapy and provide rational strategies harnessing this mechanism for enhanced efficacy of CD47-SIRPα blockade.


Sujet(s)
Adénosine triphosphate , Antigènes CD47 , Interféron de type I , Phosphorylation oxydative , Récepteurs immunologiques , Transduction du signal , Animaux , Antigènes CD47/métabolisme , Antigènes CD47/génétique , Interféron de type I/métabolisme , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/génétique , Femelle , Souris , Adénosine triphosphate/métabolisme , Phosphorylation oxydative/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Souris de lignée C57BL , Immunothérapie/méthodes , Humains , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Récepteurs purinergiques P2X7/métabolisme , Récepteurs purinergiques P2X7/génétique , Autophagie/effets des médicaments et des substances chimiques , Apyrase/métabolisme , Souris knockout , Tumeurs/immunologie , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Cytokines/métabolisme
16.
ACS Appl Mater Interfaces ; 16(32): 41788-41799, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39079025

RÉSUMÉ

Glycinamide ribonucleotide formyltransferase (GARFT) is an important enzyme in the folate metabolism pathway, and chemical drugs targeting GARFT have been used in tumor treatments over the past few decades. The development of novel antimetabolism drugs that target GARFT with improved performance and superior activity remains an attractive strategy. Herein, we proposed a targeted double-template molecularly imprinted polymer (MIP) for enhancing macrophage phagocytosis and synergistic antimetabolic therapy. The double-template MIP was prepared by imprinting the exposed peptide segment of the extracellular domain of CD47 and the active center of GARFT. Owing to the imprinted cavities on the surface of MIP, it can actively target cancer cells and mask the "do not eat me" signal upon binding to CD47 thereby blocking the CD47-SIRPα pathway and ultimately enhancing phagocytosis by macrophages. In addition, MIP can specifically bind to the active center of GARFT upon entry into the cells, thereby inhibiting its catalytic activity and ultimately interfering with the normal expression of DNA. A series of cell experiments demonstrated that MIP can effectively target CD47 overexpressed 4T1 cancer cells and inhibit the growth of 4T1 cells. The enhanced phagocytosis ability of macrophages-RAW264.7 cells was also clearly observed by confocal imaging experiments. In vivo experiments also showed that the MIP exhibited a satisfactory tumor inhibition effect. Therefore, this study provides a new idea for the application of molecular imprinting technology to antimetabolic therapy in conjunction with macrophage-mediated immunotherapy.


Sujet(s)
Antigènes CD47 , Macrophages , Polymères à empreintes moléculaires , Phagocytose , Antigènes CD47/métabolisme , Antigènes CD47/composition chimique , Phagocytose/effets des médicaments et des substances chimiques , Animaux , Souris , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Cellules RAW 264.7 , Polymères à empreintes moléculaires/composition chimique , Lignée cellulaire tumorale , Femelle , Souris de lignée BALB C , Humains , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie
17.
J Control Release ; 373: 447-462, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39038546

RÉSUMÉ

Immunosuppressive tumor-associated macrophages (TAMs) account for a high proportion of the tumor tissue and significantly impede immunoefficacy. Furthermore, the signal regulatory protein α (SIRPα) expressed in TAMs adversely correlates with macrophage activation and phagocytosis, resulting in immunosurveillance escape. To address these difficulties, a mannose-modified, pH-responsive nanoplatform with resiquimod (R848) and 2', 3'-cyclic GMP-AMP (cGAMP) co-encapsulation (named M-PNP@R@C) is designed to polarize TAMs and lower SIRPα expression. The co-delivery of R848 and cGAMP synergistically facilitates the polarization of TAMs from the anti-inflammatory M2 phenotype into the pro-inflammatory M1 phenotype, thereby enhancing antitumor immunotherapy. Remarkably, activation of the cGAMP-mediated stimulator of interferon genes (STING) in TAMs significantly downregulates the expression of SIRPα, which synergizes with the cluster of differentiation 47 (CD47) antibody for the dual blockade of the CD47-SIRPα axis. Further analysis of single-cell RNA sequencing indicates that STING activation downregulates SIRPα by regulating intracellular fatty acid oxidation metabolism. In vivo studies indicate that M-PNP@R@C significantly inhibits tumor growth with a potent antitumor immune response in melanoma graft tumor models. After synergy with anti-CD47, the double blockade strategies of the SIRPα/CD47 axis result in a notable inhibition of lung metastasis. A prolonged survival rate is observed after combination treatment with CD47 and programmed death ligand-1 antibodies for the triple immune checkpoint blockade. In summary, our study provides original insights into the potential role of the STING pathway in macrophage-based immunotherapy, thus offering a potential combinatorial strategy for cancer therapy.


Sujet(s)
Immunothérapie , Protéines membranaires , Souris de lignée C57BL , Nucleotidyltransferases , Phagocytose , Animaux , Immunothérapie/méthodes , Protéines membranaires/métabolisme , Nucleotidyltransferases/métabolisme , Phagocytose/effets des médicaments et des substances chimiques , Souris , Macrophages/immunologie , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Nanoparticules/administration et posologie , Polymères/administration et posologie , Polymères/composition chimique , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/immunologie , Nucléotides cycliques/administration et posologie , Transduction du signal/effets des médicaments et des substances chimiques , Antigènes CD47/immunologie , Macrophages associés aux tumeurs/immunologie , Macrophages associés aux tumeurs/effets des médicaments et des substances chimiques , Macrophages associés aux tumeurs/métabolisme , Mélanome expérimental/immunologie , Mélanome expérimental/thérapie , Mélanome expérimental/métabolisme , Femelle , Humains , Lignée cellulaire tumorale , Cellules RAW 264.7 , Tumeurs/thérapie , Tumeurs/immunologie , Tumeurs/traitement médicamenteux
18.
JCI Insight ; 9(17)2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39078701

RÉSUMÉ

Thrombospondin-1 (TSP1) is a matricellular protein associated with the regulation of cell migration through direct binding interactions with integrin proteins and by associating with other receptors known to regulate integrin function, including CD47 and CD36. We previously demonstrated that deletion of an epithelial TSP1 receptor, CD47, attenuates epithelial wound repair following intestinal mucosal injury. However, the mechanisms by which TSP1 contributes to intestinal mucosal repair remain poorly understood. Our results show upregulated TSP1 expression in colonic mucosal wounds and impaired intestinal mucosal wound healing in vivo upon intestinal epithelium-specific loss of TSP1 (VillinCre/+ Thbs1fl/fl or Thbs1ΔIEC mice). We report that exposure to exogenous TSP1 enhanced migration of intestinal epithelial cells in a CD47- and TGF-ß1-dependent manner and that deficiency of TSP1 in primary murine colonic epithelial cells resulted in impaired wound healing. Mechanistically, TSP1 modulated epithelial actin cytoskeletal dynamics through suppression of RhoA activity, activation of Rho family small GTPase (Rac1), and changes in filamentous-actin bundling. Overall, TSP1 was found to regulate intestinal mucosal wound healing via CD47 and TGF-ß1, coordinate integrin-containing cell-matrix adhesion dynamics, and remodel the actin cytoskeleton in migrating epithelial cells to enhance cell motility and promote wound repair.


Sujet(s)
Antigènes CD47 , Mouvement cellulaire , Muqueuse intestinale , Thrombospondine-1 , Facteur de croissance transformant bêta-1 , Cicatrisation de plaie , Animaux , Thrombospondine-1/métabolisme , Thrombospondine-1/génétique , Cicatrisation de plaie/physiologie , Souris , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Antigènes CD47/métabolisme , Antigènes CD47/génétique , Facteur de croissance transformant bêta-1/métabolisme , Protéine G RhoA/métabolisme , Souris knockout , Protéine G rac1/métabolisme , Cellules épithéliales/métabolisme , Humains , Côlon/métabolisme , Côlon/anatomopathologie , Mâle , Neuropeptides
20.
Front Immunol ; 15: 1398508, 2024.
Article de Anglais | MEDLINE | ID: mdl-38983860

RÉSUMÉ

Background: CD38 and CD47 are expressed in many hematologic malignancies, including multiple myeloma (MM), B-cell non-Hodgkin lymphoma (NHL), B-cell acute lymphoblastic leukemia (ALL), and B-cell chronic lymphocytic leukemia (CLL). Here, we evaluated the antitumor activities of CD38/CD47 bispecific antibodies (BsAbs). Methods: Five suitable anti-CD38 antibodies for co-targeting CD47 and CD38 BsAb were developed using a 2 + 2 "mAb-trap" platform. The activity characteristics of the CD38/CD47 BsAbs were evaluated using in vitro and in vivo systems. Results: Using hybridoma screening technology, we obtained nine suitable anti-CD38 antibodies. All anti-CD38 antibodies bind to CD38+ tumor cells and kill tumor cells via antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP). Five anti-CD38 antibodies (4A8, 12C10, 26B4, 35G5, and 65A7) were selected for designing CD38/CD47 BsAbs (IMM5605) using a "mAb-trap" platform. BsAbs had higher affinity and binding activity to the CD38 target than those to the CD47 target, decreasing the potential on-target potential and off-tumor effects. The CD38/CD47 BsAbs did not bind to RBCs and did not induce RBC agglutination; thus, BsAbs had much lower blood toxicity. The CD38/CD47 BsAbs had a greater ability to block the CD47/SIRPα signal in CD38+/CD47+ tumor cells than IMM01 (SIRPα Fc fusion protein). Through Fc domain engineering, CD38/CD47 BsAbs were shown to kill tumors more effectively by inducing ADCC and ADCP. IMM5605-26B4 had the strongest inhibitory effect on cellular CD38 enzymatic activity. IMM5605-12C10 had the strongest ability to directly induce the apoptosis of tumor cells. The anti-CD38 antibody 26B4 combined with the SIRPα-Fc fusion proteins showed strong antitumor effects, which were better than any of the mono-therapeutic agents used alone in the NCI-H929 cell xenograft model. The CD38/CD47 BsAbs exhibited strong antitumor effects; specifically, IMM5605-12C10 efficiently eradicated all established tumors in all mice. Conclusion: A panel of BsAbs targeting CD38 and CD47 developed based on the "mAb-tarp" platform showed potent tumor-killing ability in vitro and in vivo. As BsAbs had lower affinity for binding to CD47, higher affinity for binding to CD38, no affinity for binding to RBCs, and did not induce RBC agglutination, we concluded that CD38/CD47 BsAbs are safe and have a satisfactory tolerability profile.


Sujet(s)
Antigènes CD38 , Antigènes CD47 , Tumeurs hématologiques , Antigènes CD47/immunologie , Antigènes CD47/antagonistes et inhibiteurs , Antigènes CD47/métabolisme , Antigènes CD38/antagonistes et inhibiteurs , Antigènes CD38/immunologie , Antigènes CD38/métabolisme , Humains , Animaux , Souris , Tumeurs hématologiques/thérapie , Tumeurs hématologiques/immunologie , Lignée cellulaire tumorale , Anticorps bispécifiques/pharmacologie , Anticorps bispécifiques/immunologie , Tests d'activité antitumorale sur modèle de xénogreffe , Glycoprotéines membranaires/immunologie , Glycoprotéines membranaires/antagonistes et inhibiteurs , Cytotoxicité à médiation cellulaire dépendante des anticorps , Femelle , Antinéoplasiques immunologiques/pharmacologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE