Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 14: 1237715, 2023.
Article in English | MEDLINE | ID: mdl-37771579

ABSTRACT

CX3CL1 secreted in the tumor microenvironment serves as a chemoattractant playing a critical role in metastasis of CX3CR1 expressing cancer cells. CX3CR1 can be expressed in both cancer and immune-inhibitory myeloid cells to facilitate their migration. We generated a novel monoclonal antibody against mouse CX3CR1 that binds to CX3CR1 and blocks the CX3CL1-CX3CR1 interaction. We next explored the immune evasion strategies implemented by the CX3CL1-CX3CR1 axis and find that it initiates a resistance program in cancer cells that results in 1) facilitation of tumor cell migration, 2) secretion of soluble mediators to generate a pro-metastatic niche, 3) secretion of soluble mediators to attract myeloid populations, and 4) generation of tumor-inflammasome. The CX3CR1 monoclonal antibody reduces migration of tumor cells and decreases secretion of immune suppressive soluble mediators by tumor cells. In combination with anti-PD-1 immunotherapy, this CX3CR1 monoclonal antibody enhances survival in an immunocompetent mouse colon carcinoma model through a decrease in tumor-promoting myeloid populations. Thus, this axis is involved in the mechanisms of resistance to anti-PD-1 immunotherapy and the combination therapy can overcome a portion of the resistance mechanisms to anti-PD-1.


Subject(s)
Antibodies, Monoclonal , Tumor Escape , Mice , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Chemokine CX3CL1/metabolism
2.
Cancer Res ; 80(22): 5035-5050, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32978170

ABSTRACT

Although epithelial cell adhesion molecule (EpCAM) has previously been shown to promote tumor progression, the underlying mechanisms remain largely unknown. Here, we report that the EGF-like domain I within the extracellular domain of EpCAM (EpEX) binds EGFR, activating both AKT and MAPK signaling to inhibit forkhead transcription factor O3a (FOXO3a) function and stabilize PD-L1 protein, respectively. Treatment with the EpCAM neutralizing antibody, EpAb2-6, inhibited AKT and FOXO3a phosphorylation, increased FOXO3a nuclear translocation, and upregulated high temperature requirement A2 (HtrA2) expression to promote apoptosis while decreasing PD-L1 protein levels to enhance the cytotoxic activity of CD8+ T cells. In vivo, EpAb2-6 markedly extended survival in mouse metastasis and orthotopic models of human colorectal cancer. The combination of EpAb2-6 with atezolizumab, an anti-PD-L1 antibody, almost completely eliminated tumors. Moreover, the number of CD8+ T cells in combination-treated tumors was increased compared with atezolizumab alone. Our findings suggest a new combination strategy for cancer immunotherapy in patients with EpCAM-expressing tumors. SIGNIFICANCE: This study shows that treatment with an EpCAM neutralizing antibody promotes apoptosis while decreasing PD-L1 protein to enhance cytotoxic activity of CD8+ T cells.


Subject(s)
B7-H1 Antigen/chemistry , CD8-Positive T-Lymphocytes/immunology , Disease Progression , Epithelial Cell Adhesion Molecule/metabolism , ErbB Receptors/metabolism , Forkhead Box Protein O3/metabolism , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Neutralizing/pharmacology , Antigens, Neoplasm/metabolism , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Line, Tumor , Cell Nucleus/metabolism , Colorectal Neoplasms/mortality , Colorectal Neoplasms/therapy , Cycloheximide/pharmacology , Enzyme Activation , Heterografts , High-Temperature Requirement A Serine Peptidase 2/metabolism , Humans , Mice , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Transplantation , Phosphorylation/drug effects , Programmed Cell Death 1 Receptor/metabolism , Protein Domains , Protein Stability/drug effects , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Up-Regulation
3.
Biomolecules ; 10(7)2020 07 10.
Article in English | MEDLINE | ID: mdl-32664346

ABSTRACT

Intractable cardiovascular diseases are leading causes of mortality around the world. Adult mammalian hearts have poor regenerative capacity and are not capable of self-repair after injury. Recent studies of cell-free therapeutics such as those designed to stimulate endogenous cardiac regeneration have uncovered new feasible therapeutic avenues for cardiac repair. The Hippo pathway, a fundamental pathway with pivotal roles in cell proliferation, survival and differentiation, has tremendous potential for therapeutic manipulation in cardiac regeneration. In this review, we summarize the most recent studies that have revealed the function of the Hippo pathway in heart regeneration and homeostasis. In particular, we discuss the molecular mechanisms of how the Hippo pathway maintains cardiac homeostasis by directing cardiomyocyte chromatin remodeling and regulating the cell-cell communication between cardiomyocytes and non-cardiomyocytes in the heart.


Subject(s)
Heart Injuries/therapy , Myocytes, Cardiac/physiology , Signal Transduction , Animals , Cell Communication , Cell Differentiation , Cell Proliferation , Cell-Free System , Heart Injuries/metabolism , Hippo Signaling Pathway , Homeostasis , Humans , Protein Serine-Threonine Kinases/metabolism , Regeneration
4.
Cancer Lett ; 433: 165-175, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29981429

ABSTRACT

Epithelial cell adhesion molecule (EpCAM) is highly expressed in colon cancers, but its role in cancer progression remains to be elucidated. In this work, we found that the extracellular domain of EpCAM (EpEX) activated EGFR and downstream ERK1/2 signaling to promote colon cancer cell migration and proliferation, as well as tumor growth. Mechanistically, we discovered that EpEX-EGFR-ERK1/2 signaling positively regulated intramembrane proteolysis (RIP) of EpCAM and shedding of the intracellular domain (EpICD). Treatment with an EGFR inhibitor ablated the EpEX-induced phosphorylation of ERK1/2 and AKT. Additionally, treatment with inhibitors of either EGFR or MEK decreased EpEX-induced EpICD shedding and further revealed that EpICD is necessary for nuclear accumulation of ß-catenin and the induction of HIF1α target gene expression in vitro and in vivo. Moreover, an anti-EpCAM neutralizing monoclonal antibody, EpAb2-6, inhibited the nuclear translocation of EpICD and ß-catenin and induced apoptosis in colon cancer cells. Importantly, analysis of colorectal cancer tissues showed that nuclear accumulation of EpICD was highly correlated with metastasis and poor prognosis, suggesting that it may play an important functional role in cancer progression. Thus, we provide novel insights into the mechanisms and functions of EpEX-mediated signaling, which may be considered as a promising target for the treatment of colon cancer.


Subject(s)
Cell Nucleus/metabolism , Colonic Neoplasms/pathology , Epithelial Cell Adhesion Molecule/chemistry , Epithelial Cell Adhesion Molecule/metabolism , MAP Kinase Signaling System , Animals , Cell Line, Tumor , Cell Movement , Colonic Neoplasms/metabolism , Disease Progression , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Mice , Neoplasm Transplantation , Phosphorylation , Prognosis , Protein Domains , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...