Littérature scientifique sur le sujet « Tumor-associated macrophages, DAB2, metastases, extracellular matrix »

Créez une référence correcte selon les styles APA, MLA, Chicago, Harvard et plusieurs autres

Choisissez une source :

Consultez les listes thématiques d’articles de revues, de livres, de thèses, de rapports de conférences et d’autres sources académiques sur le sujet « Tumor-associated macrophages, DAB2, metastases, extracellular matrix ».

À côté de chaque source dans la liste de références il y a un bouton « Ajouter à la bibliographie ». Cliquez sur ce bouton, et nous générerons automatiquement la référence bibliographique pour la source choisie selon votre style de citation préféré : APA, MLA, Harvard, Vancouver, Chicago, etc.

Vous pouvez aussi télécharger le texte intégral de la publication scolaire au format pdf et consulter son résumé en ligne lorsque ces informations sont inclues dans les métadonnées.

Articles de revues sur le sujet "Tumor-associated macrophages, DAB2, metastases, extracellular matrix"

1

Paolillo et Schinelli. « Extracellular Matrix Alterations in Metastatic Processes ». International Journal of Molecular Sciences 20, no 19 (7 octobre 2019) : 4947. http://dx.doi.org/10.3390/ijms20194947.

Texte intégral
Résumé :
The extracellular matrix (ECM) is a complex network of extracellular-secreted macromolecules, such as collagen, enzymes and glycoproteins, whose main functions deal with structural scaffolding and biochemical support of cells and tissues. ECM homeostasis is essential for organ development and functioning under physiological conditions, while its sustained modification or dysregulation can result in pathological conditions. During cancer progression, epithelial tumor cells may undergo epithelial-to-mesenchymal transition (EMT), a morphological and functional remodeling, that deeply alters tumor cell features, leading to loss of epithelial markers (i.e., E-cadherin), changes in cell polarity and intercellular junctions and increase of mesenchymal markers (i.e., N-cadherin, fibronectin and vimentin). This process enhances cancer cell detachment from the original tumor mass and invasiveness, which are necessary for metastasis onset, thus allowing cancer cells to enter the bloodstream or lymphatic flow and colonize distant sites. The mechanisms that lead to development of metastases in specific sites are still largely obscure but modifications occurring in target tissue ECM are being intensively studied. Matrix metalloproteases and several adhesion receptors, among which integrins play a key role, are involved in metastasis-linked ECM modifications. In addition, cells involved in the metastatic niche formation, like cancer associated fibroblasts (CAF) and tumor associated macrophages (TAM), have been found to play crucial roles in ECM alterations aimed at promoting cancer cells adhesion and growth. In this review we focus on molecular mechanisms of ECM modifications occurring during cancer progression and metastatic dissemination to distant sites, with special attention to lung, liver and bone. Moreover, the functional role of cells forming the tumor niche will also be reviewed in light of the most recent findings.
Styles APA, Harvard, Vancouver, ISO, etc.
2

Mizuno, Rei, Kenji Kawada, Yoshiro Itatani, Ryotaro Ogawa, Yoshiyuki Kiyasu et Yoshiharu Sakai. « The Role of Tumor-Associated Neutrophils in Colorectal Cancer ». International Journal of Molecular Sciences 20, no 3 (27 janvier 2019) : 529. http://dx.doi.org/10.3390/ijms20030529.

Texte intégral
Résumé :
Colorectal cancer (CRC) is one of the most common causes of cancer deaths worldwide and the number of CRC patients is increasing progressively. Despite the improvement of the surgical techniques and chemotherapy, we have not completely overcome this disease yet due to the metastases. Therefore, understanding the mechanisms through which metastasis occurs is important for overcoming CRC. Normal host cells in the tumor microenvironment, such as macrophages and fibroblasts, have been reported to promote the growth of CRCs. Although neutrophils were originally considered to have defensive functions against tumor cells, it has been revealed that some populations of neutrophils, called as tumor-associated neutrophils (TANs), have tumor-supportive functions. The plasticity between tumor-suppressive and -supportive neutrophils are regulated by transforming growth factor (TGF)-β and Interferon-β signaling. Some studies have demonstrated that TANs promote the spread of cancer cells to distant organs. TANs contribute to the tumor invasion and angiogenesis through the production of matrix metalloproteinase-9 (MMP9), vascular endothelial growth factor (VEGF), and hepatocyte growth factor (HGF) in the primary and metastatic sites. Neutrophils also promotes tumor cell dissemination by capturing circulating tumor cells using neutrophil extracellular traps and promote their migration to distant sites. The neutrophil-to-lymphocyte ratio is a well-defined predictive marker for CRC patients. In this review, we highlight the molecular signaling between TANs and CRC cells and the possibility of TANs as a potential target for cancer therapy.
Styles APA, Harvard, Vancouver, ISO, etc.
3

Vacca, Angelo, Domenico Ribatti, Roberto Ria, Antonio Pellegrino, Michele Bruno, Francesca Merchionne et Franco Dammacco. « Proteolytic Activity of Human Lymphoid Tumor Cells. Correlation with Tumor Progression ». Developmental Immunology 7, no 2-4 (2000) : 77–88. http://dx.doi.org/10.1155/2000/74372.

Texte intégral
Résumé :
Matrix metalloproteinase (MMP) expression and production are associated with advanced-stage tumor and contribute to tumor progression, invasion and metastases. The current study was designed to determine the expression and production of MMP-2 (gelatinase A) and MMP-9 (gelatinase B) by human lymphoid tumor cells. Changes in expression and production were also investigated during tumor progression of multiple myeloma and mycosis fungoides.In situhybridization analysis revealed that lymphoblastic leukemia B cells (SB cell line), multiple myeloma (MM) cells (U266 cell line) and lymphoblastic leukemia T cells (CEM and Jurkat cell lines) express constitutively the mRNA for MMP-2 and/or MMP-9. We demonstrated by gelatin-zymography of cell culture medium that both enzymes were secreted in their cleaved (activated) form.In situhybridization of bone marrow plasma cells and gelatin- zymography of the medium showed that patients with active MM (diagnosis, relapse, leukemic progression) express higher levels of MMP-2 mRNA and protein than patients with non-active MM (complete/objective response, plateau) and with monoclonal gammopathies of undetermined significance (MGUS). MMP-9 expression and secretion was similar in all patient groups. In patients with mycosis fungoides (MF), the expression of MMP-2 and MMP-9 mRNAs was significantly upregulated with advancing stage, in terms of lesions both positive for one of two mRNAs and with the greatest intensity of expression. Besides MF cells, the MMP-2 and/or MMP-9 mRNAs were expressed by some stromal cell populations (microvascular endothelial cells, fibroblasts, macrophages), suggesting that these cells cooperate in the process of tumor invasion. Our studies identify MMPs as an important class of proteinases involved in the extracellular matrix (ECM) degradation by human lymphoid tumors, and suggest that MMPs inhibitors may lead to important new treatment for their control.
Styles APA, Harvard, Vancouver, ISO, etc.
4

Browne, Alice, Miranda Clements, Wei Ju, Daniel Brock et Rosandra Kaplan. « Abstract B007 : Remodeling the hyaluronan-rich tumor extracellular matrix using hyaluronidase-expressing genetically engineered MSCs for tumor regression and metastasis prevention in sarcomas ». Cancer Research 83, no 2_Supplement_2 (15 janvier 2023) : B007. http://dx.doi.org/10.1158/1538-7445.metastasis22-b007.

Texte intégral
Résumé :
Abstract Sarcomas are a diverse group of rare malignant tumors the develop in the bone and connective tissue and are often aggressive with ~50% of patients developing metastases. Due to their rarity and diversity, there is a great lack of standard therapies resulting in a 65% and 15% 5-year survival rate for regional and metastatic disease, respectively. Tumorigenesis is largely influenced by communication between the tumor microenvironment (TME), cancer cells, and the extracellular matrix (ECM). The ECM promotes cancer initiation and dissemination from the primary tumor and fosters a favorable microenvironment for metastatic colonization of tumor cells. Due to their mesenchymal origin, sarcomas are thought to produce and deposit large quantities of ECM. One component of the tumor-ECM known to promote an aggressive cancer phenotype is hyaluronan (HA). Abundant HA in the TME arises from overactive HA synthases that produce HA or downregulation of hyaluronidases (Hyals) that catalyze HA degradation. HA-rich tumor ECM promote cancer progression by regulating chemokine and cytokine trafficking, increasing hypoxia and interstitial pressure, and limiting delivery of antitumor therapies. In-silico profiling of sarcomas revealed an increase in HA synthase mRNA expression in 50.78% and correlated with 35% decrease in overall patient survival (P = 0.0027), compared to sarcomas with low HA synthase expression. To target this HA-rich tumor ECM, we sought to develop a cell-based approach to locally deliver HA-remodeling enzymes to minimize the drawbacks of systemic therapy and enhance anti-tumor responses. We developed genetically engineered mesenchymal stromal cells (GEMesys) that express Hyal, using mesenchymal stem cells derived from mouse lungs. We hypothesized that the Hyal-GEMesys would home to and degrade the HA-rich ECM preventing cancer progression. The therapeutic efficacy of Hyal-GEMesys was tested in osteosarcoma (F42010) and rhabdomyosarcoma (M-3-9M) syngeneic murine models. Preliminary data demonstrated encouraging efficacy of Hyal-GEMesys in reducing tumor volume in the osteosarcoma (up to 1.4-fold, p=0.0035-0.0172) and rhabdomyosarcoma M-3-9M model (up to 2-fold, P= 0.0428). Further, analysis of osteosarcoma tumors revealed reduced HA (up to 2.3-fold, P=0.0078) and collagen-I content (up to 3-fold, P= 0.0025) indicating our Hyal-GEMesys functionally remodel the ECM in vivo. Changes in ECM in GEMesy treated group were associated with changes in immune infiltration with an increase in CD4+ (P=0.0221) and CD8+ (P=0.0087) T cells and a decrease in IBA1+ (P=0.0144) macrophages. Analysis of rhabdomyosarcoma tumors revealed reduced HA content (up to 2-fold, P-0.00206). Future studies will more deeply explore the mechanisms of ECM-mediated immune changes and impact on metastatic progression. Our cell-based approach to deliver hyaluronidases holds promise of effectively degrading and remodel the HA-rich tumor ECM to enhance anti-tumor responses, prevent metastases and improve clinical outcomes for patients with high-risk sarcoma malignancies. Citation Format: Alice Browne, Miranda Clements, Wei Ju, Daniel Brock, Rosandra Kaplan. Remodeling the hyaluronan-rich tumor extracellular matrix using hyaluronidase-expressing genetically engineered MSCs for tumor regression and metastasis prevention in sarcomas [abstract]. In: Proceedings of the AACR Special Conference: Cancer Metastasis; 2022 Nov 14-17; Portland, OR. Philadelphia (PA): AACR; Cancer Res 2022;83(2 Suppl_2):Abstract nr B007.
Styles APA, Harvard, Vancouver, ISO, etc.
5

Horn, Lucas, Haiyan Qin, Kristen Fousek, Masafumi Iida, Dallas Flies, Ronald Copeland, Zachary Cusumano et al. « 570 Blockade of the inhibitory collagen receptor LAIR-1, PD-L1, and TGF-β promotes anti-tumor activity through T cell activation and myeloid cell polarization ». Journal for ImmunoTherapy of Cancer 9, Suppl 2 (novembre 2021) : A599. http://dx.doi.org/10.1136/jitc-2021-sitc2021.570.

Texte intégral
Résumé :
BackgroundLeukocyte-associated immunoglobulin-like receptor 1 (LAIR-1) is an immune inhibitory receptor that binds collagen-like domains commonly found in extracellular matrix (ECM) collagens and complement component C1q. LAIR-1 is expressed on several immune cell types including activated T cells, B cells, NK cells, dendritic cells, and macrophages. Numerous cancer types including gastric, colon, ovarian, bladder, and others, upregulate collagens which enhances tumor growth, metastases, and invasion while actively suppressing antitumor immunity. While a soluble decoy, LAIR-2, is expressed in humans and competes with LAIR-1 for binding of collagen domains, excess LAIR ligands in the tumor often result in an immune suppressive environment.MethodsHere, we report on a novel immunotherapy approach combining NC410, a novel fusion protein consisting of two LAIR-2 molecules grafted on to an IgG1 antibody backbone, capable of targeting the tumor ECM and blocking LAIR-1 signaling; and bintrafusp alfa, a first-in-class bifunctional fusion protein composed of the extracellular domain of the human transforming growth factor β receptor II (TGF-βRII or TGF-β ”trap”) fused via a flexible linker to the C-terminus of each heavy chain of an IgG1 antibody blocking programmed death ligand 1 (anti–PD-L1).ResultsWe have demonstrated that the combination of NC410 and bintrafusp alfa more effectively controls in vivo tumor growth of the collagen rich MC38 colon and EMT6 mammary carcinomas compared to either monotherapy. We demonstrate that this potent anti-tumor immune response is propagated through the synergy of activated tumor infiltrating lymphocytes and a repolarization of myeloid cells in the tumor microenvironment. MC38 tumors treated with the combination of NC410 plus bintrafusp alfa contained higher numbers of infiltrating T cells, NK cells, and M1 polarized macrophages.ConclusionsThis study highlights the synergy of reshaping the large suppressive myeloid cell populations often present in tumors with activation of adaptive T-cell immune responses dampened by checkpoint inhibition. The results also provide the rationale for the future evaluation of this combination therapy in the clinic.
Styles APA, Harvard, Vancouver, ISO, etc.
6

Schmidt, H., A. Roslind, J. S. Johansen, D. L. Nielsen, P. A. Price et E. Balslev. « YKL-40 expression in naevocellular naevi, carcinomas and melanomas of the skin ». Journal of Clinical Oncology 24, no 18_suppl (20 juin 2006) : 18019. http://dx.doi.org/10.1200/jco.2006.24.18_suppl.18019.

Texte intégral
Résumé :
18019 Background: Elevated serum levels of the protein YKL-40 are associated with a poor prognosis in several solid tumors including melanoma (H. Schmidt, JCO 2006, Cancer 2006 in press). YKL-40 is secreted by cancer cells, macrophages and neutrophils. The function of YKL-40 is only poorly understood. It may be a growth or differentiation factor, play a role in angiogenesis or protect against apoptosis. The aim was to describe the tissue expression of YKL-40 in naevocellular naevi, carcinomas and melanomas of the skin by immunohistochemistry. Methods: Paraffin-embedded tissue sections from 40 patients with different naevocellular naevi, carcinomas and melanomas of the skin were developed with EnVision+ System-HRP K4007 (DakoCytomation) using a monoclonal antibody against YKL-40. The intensity of YKL-40 staining was scored using a semi-quantitative method. Results: YKL-40 protein expression appeared as a cytoplasmic, granular staining with no staining of membranes, nuclei or extracellular matrix. In normal skin the basal epithelial layer stained weakly positive. Melanocytes and melanophages stained negative. Macrophages and leucocytes stained positive. Hair follicles, sebaceous glands and sweat glands stained positive. Three basal cell carcinomas all stained positive. In 10 squamous cell carcinomas, the epidermal components stained positive and the invasive tumor islands were negative. In 3 keratoacanthomas, the epithelial components as well as the infiltrative tumor islands stained positive. In naevi (2 junctional, 5 compound and 1 intradermal) the epithelial components stained positive, the intradermal components stained negative. In melanomas the tumor cells stained positive in 9 of 10 specimens. Both the epidermal and the invasive components of melanomas stained positive. Three melanoma metastases all stained positive. Conclusions: YKL-40 is expressed in normal skin and in benign and malignant lesions of the skin. Generally, YKL-40 expression is increased in cells of high activity, although the invasive tumor islands of squamous cell carcinomas stain negative. In melanomas, the tumor cells may be a source of the elevated YKL-40 serum levels observed in these patients. No significant financial relationships to disclose.
Styles APA, Harvard, Vancouver, ISO, etc.
7

Sathe, Anuja, Kaishu Mason, Susan M. Grimes, Zilu Zhou, Billy T. Lau, Xiangqi Bai, Andrew Su et al. « Colorectal cancer metastases in the liver establish immunosuppressive spatial networking between tumor associated SPP1+ macrophages and fibroblasts ». Clinical Cancer Research, 14 octobre 2022. http://dx.doi.org/10.1158/1078-0432.ccr-22-2041.

Texte intégral
Résumé :
Abstract Purpose:The liver is the most frequent metastatic site for colorectal cancer (CRC). Its microenvironment is modified to provide a niche that is conducive for CRC cell growth.This study focused on characterizing the cellular changes in the metastatic CRC (mCRC) liver tumor microenvironment (TME). Experimental Design: We analyzed a series of microsatellite stable (MSS) mCRCs to the liver, paired normal liver tissue and peripheral blood mononuclear cells using single cell RNA-seq (scRNA-seq). We validated our findings using multiplexed spatial imaging and bulk gene expression with cell deconvolution. Results: We identified TME-specific SPP1-expressing macrophages with altered metabolism features, foam cell characteristics and increased activity in extracellular matrix (ECM) organization. SPP1+ macrophages and fibroblasts expressed complementary ligand receptor pairs with the potential to mutually influence their gene expression programs. TME lacked dysfunctional CD8 T cells and contained regulatory T cells, indicative of immunosuppression. Spatial imaging validated these cell states in the TME. Moreover, TME macrophages and fibroblasts had close spatial proximity, which is a requirement for intercellular communication and networking.In an independent cohort of mCRCs in the liver, we confirmed the presence of SPP1+ macrophages and fibroblasts using gene expression data. An increased proportion of TME fibroblasts was associated with a worst prognosis in these patients. Conclusions: We demonstrated that mCRC in the liver is characterized by transcriptional alterations of macrophages in the TME. Intercellular networking between macrophages and fibroblasts supports CRC growth in the immunosuppressed metastatic niche in the liver. These features can be used to target immune checkpoint resistant MSS tumors.
Styles APA, Harvard, Vancouver, ISO, etc.
8

Zhao, Qing, Xuexin He, Xiyi Qin, Yu Liu, Han Jiang, Jing Wang, Shuang Wu et al. « Enhanced Therapeutic Efficacy of Combining Losartan and Chemo-Immunotherapy for Triple Negative Breast Cancer ». Frontiers in Immunology 13 (23 juin 2022). http://dx.doi.org/10.3389/fimmu.2022.938439.

Texte intégral
Résumé :
Triple-negative breast cancer (TNBC) is a particularly aggressive subtype of breast cancer, which is relatively resistant to anti-programmed cell death-1 (α-PD1) therapy, characterized as non-immunogenic, dense stroma and accumulation of M2 tumor-associated macrophages (TAMs). Despite progress in strategies to deplete extracellular matrix (ECM) and enhance tumor-cell immunogenicity, the combinatorial anti-cancer effects with α-PD1 need to be explored. Here, we applied doxorubicin hydrochloride liposome (Dox-L) as immunogenic cell death (ICD)-inducing nano-chemotherapy and used losartan as stroma-depleting agent to improve α-PD1 efficacy (Losartan + Dox-L + α-PD1). The results showed that losartan could cause ECM reduction, facilitating enhanced delivery of Dox-L and further dendritic cell (DC) maturation. Additionally, losartan could also alleviate hypoxia for TNBC, thus reprogramming pro-cancer M2 TAMs to anti-cancer M1 TAMs, successfully overcoming immune-suppressive microenvironment. These modifications led to a significant increase in T cells’ infiltration and augmented anti-tumor immunity as exemplified by the notable reduction in tumor size and lung metastases. In summary, our findings support that combined treatment of losartan with Dox-L normalizes immunological-cold microenvironment, improves immuno-stimulation and optimizes the efficacy of TNBC immunotherapy. A novel combinational strategy with FDA-approved compounds proposed by the study may potentially be useful in TNBC clinical treatment.
Styles APA, Harvard, Vancouver, ISO, etc.
Nous offrons des réductions sur tous les plans premium pour les auteurs dont les œuvres sont incluses dans des sélections littéraires thématiques. Contactez-nous pour obtenir un code promo unique!

Vers la bibliographie