Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Rep ; 36(8): 109602, 2021 08 24.
Article in English | MEDLINE | ID: mdl-34433042

ABSTRACT

Inappropriate CD4+ T helper (Th) differentiation can compromise host immunity or promote autoimmune disease. To identify disease-relevant regulators of T cell fate, we examined mutations that modify risk for multiple sclerosis (MS), a canonical organ-specific autoimmune disease. This analysis identified a role for Zinc finger E-box-binding homeobox (ZEB1). Deletion of ZEB1 protects against experimental autoimmune encephalitis (EAE), a mouse model of multiple sclerosis (MS). Mechanistically, ZEB1 in CD4+ T cells is required for pathogenic Th1 and Th17 differentiation. Genomic analyses of paired human and mouse expression data elucidated an unexpected role for ZEB1 in JAK-STAT signaling. ZEB1 inhibits miR-101-3p that represses JAK2 expression, STAT3/STAT4 phosphorylation, and subsequent expression of interleukin-17 (IL-17) and interferon gamma (IFN-γ). Underscoring its clinical relevance, ZEB1 and JAK2 downregulation decreases pathogenic cytokines expression in T cells from MS patients. Moreover, a Food and Drug Administration (FDA)-approved JAK2 inhibitor is effective in EAE. Collectively, these findings identify a conserved, potentially targetable mechanism regulating disease-relevant inflammation.


Subject(s)
Cell Differentiation/physiology , Interleukin-17/metabolism , Multiple Sclerosis/pathology , Th17 Cells/immunology , Zinc Finger E-box-Binding Homeobox 1/metabolism , Animals , Cell Differentiation/immunology , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Interleukin-17/immunology , Mice , Multiple Sclerosis/immunology , Th1 Cells/immunology , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/immunology
2.
J Clin Invest ; 130(3): 1252-1270, 2020 03 02.
Article in English | MEDLINE | ID: mdl-32039918

ABSTRACT

Current antiangiogenic therapy is limited by its cytostatic property, scarce drug delivery to the tumor, and side toxicity. To address these limitations, we unveiled the role of ZEB1, a tumor endothelium-enriched zinc-finger transcription factor, during tumor progression. We discovered that the patients who had lung adenocarcinomas with high ZEB1 expression in tumor endothelium had increased prevalence of metastases and markedly reduced overall survival after the diagnosis of lung cancer. Endothelial ZEB1 deletion in tumor-bearing mice diminished tumor angiogenesis while eliciting persistent tumor vascular normalization by epigenetically repressing TGF-ß signaling. This consequently led to improved blood and oxygen perfusion, enhanced chemotherapy delivery and immune effector cell infiltration, and reduced tumor growth and metastasis. Moreover, targeting vascular ZEB1 remarkably potentiated the anticancer activity of nontoxic low-dose cisplatin. Treatment with low-dose anti-programmed cell death protein 1 (anti-PD-1) antibody elicited tumor regression and markedly extended survival in ZEB1-deleted mice, conferring long-term protective anticancer immunity. Collectively, we demonstrated that inactivation of endothelial ZEB1 may offer alternative opportunities for cancer therapy with minimal side effects. Targeting endothelium-derived ZEB1 in combination with conventional chemotherapy or immune checkpoint blockade therapy may yield a potent and superior anticancer effect.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Zinc Finger E-box-Binding Homeobox 1/deficiency , Animals , Antineoplastic Agents, Immunological/pharmacology , Cisplatin/pharmacology , Endothelium/immunology , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/immunology , Gene Deletion , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Mice , Mice, Transgenic , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Zinc Finger E-box-Binding Homeobox 1/immunology
3.
Trends Immunol ; 40(5): 431-446, 2019 05.
Article in English | MEDLINE | ID: mdl-30956067

ABSTRACT

ZEB1 and ZEB2 are zinc-finger E homeobox-binding transcription factors best known for their role in driving epithelial to mesenchymal transition. However, in recent years our understanding of these two transcription factors has broadened, and it is now clear that they are expressed by a variety of immune cells of both myeloid and lymphoid lineages, including dendritic cells, macrophages, monocytes, B, T, and NK cells. In these cells, ZEBs function to regulate important transcriptional networks necessary for cell differentiation, maintenance, and function. Here, we review the current understanding of ZEB regulation across immune cell lineages, particularly in mice, highlighting present gaps in our knowledge. We also speculate on important questions for the future.


Subject(s)
Zinc Finger E-box Binding Homeobox 2/immunology , Zinc Finger E-box-Binding Homeobox 1/immunology , B-Lymphocytes/immunology , Dendritic Cells/immunology , Epithelial-Mesenchymal Transition/immunology , Humans , Killer Cells, Natural/immunology , Macrophages/immunology , Monocytes/immunology , T-Lymphocytes/immunology
4.
Nat Commun ; 10(1): 1364, 2019 03 25.
Article in English | MEDLINE | ID: mdl-30910999

ABSTRACT

The mechanisms linking muscle injury and regeneration are not fully understood. Here we report an unexpected role for ZEB1 regulating inflammatory and repair responses in dystrophic and acutely injured muscles. ZEB1 is upregulated in the undamaged and regenerating myofibers of injured muscles. Compared to wild-type counterparts, Zeb1-deficient injured muscles exhibit enhanced damage that corresponds with a retarded p38-MAPK-dependent transition of their macrophages towards an anti-inflammatory phenotype. Zeb1-deficient injured muscles also display a delayed and poorer regeneration that is accounted by the retarded anti-inflammatory macrophage transition and their intrinsically deficient muscle satellite cells (MuSCs). Macrophages in Zeb1-deficient injured muscles show lower phosphorylation of p38 and its forced activation reverts the enhanced muscle damage and poorer regeneration. MuSCs require ZEB1 to maintain their quiescence, prevent their premature activation following injury, and drive efficient regeneration in dystrophic muscles. These data indicate that ZEB1 protects muscle from damage and is required for its regeneration.


Subject(s)
Muscle, Skeletal/metabolism , Muscular Dystrophies/genetics , RNA, Messenger/genetics , Regeneration/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics , p38 Mitogen-Activated Protein Kinases/genetics , Animals , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Chromones/pharmacology , Disease Models, Animal , Flavonoids/pharmacology , Gene Expression Regulation , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/immunology , Laminin/genetics , Laminin/immunology , Macrophages/immunology , Macrophages/pathology , Mice , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Morpholines/pharmacology , Muscle, Skeletal/immunology , Muscle, Skeletal/injuries , Muscular Dystrophies/immunology , Muscular Dystrophies/pathology , Phenotype , Phosphorylation , RNA, Messenger/immunology , Regeneration/immunology , Satellite Cells, Skeletal Muscle/immunology , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/pathology , Signal Transduction , Zinc Finger E-box-Binding Homeobox 1/deficiency , Zinc Finger E-box-Binding Homeobox 1/immunology , p38 Mitogen-Activated Protein Kinases/immunology
5.
Cell Death Dis ; 9(12): 1178, 2018 12 05.
Article in English | MEDLINE | ID: mdl-30518749

ABSTRACT

Cutaneous T-cell lymphoma is a group of incurable extranodal non-Hodgkin lymphomas that develop from the skin-homing CD4+ T cell. Mycosis fungoides and Sézary syndrome are the most common histological subtypes. Although next-generation sequencing data provided significant advances in the comprehension of the genetic basis of this lymphoma, there is not uniform consensus on the identity and prevalence of putative driver genes for this heterogeneous group of tumors. Additional studies may increase the knowledge about the complex genetic etiology characterizing this lymphoma. We used SNP6 arrays and GISTIC algorithm to prioritize a list of focal somatic copy-number alterations in a dataset of multiple sequential samples from 21 Sézary syndrome patients. Our results confirmed a prevalence of significant focal deletions over amplifications: single well-known tumor suppressors, such as TP53, PTEN, and RB1, are targeted by these aberrations. In our cohort, ZEB1 (TCF8, ZFHX1A) spans a deletion having the highest level of significance. In a larger group of 43 patients, we found that ZEB1 is affected by deletions and somatic inactivating mutations in 46.5% of cases; also, we found potentially relevant ZEB1 germline variants. The survival analysis shows a worse clinical course for patients with ZEB1 biallelic inactivation. Multiple abnormal expression signatures were found associated with ZEB1 depletion in Sézary patients we verified that ZEB1 exerts a role in oxidative response of Sézary cells. Our data confirm the importance of deletions in the pathogenesis of cutaneous T-cell lymphoma. The characterization of ZEB1 abnormalities in Sézary syndrome fulfils the criteria of a canonical tumor suppressor gene. Although additional confirmations are needed, our findings suggest, for the first time, that ZEB1 germline variants might contribute to the risk of developing this disease. Also, we provide evidence that ZEB1 activity in Sézary cells, influencing the reactive oxygen species production, affects cell viability and apoptosis.


Subject(s)
Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Germ-Line Mutation , Sezary Syndrome/genetics , Skin Neoplasms/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics , Apoptosis , Cell Line, Tumor , Cell Survival , DNA Copy Number Variations , Humans , Oligonucleotide Array Sequence Analysis , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/immunology , Polymorphism, Single Nucleotide , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Retinoblastoma Binding Proteins/genetics , Retinoblastoma Binding Proteins/immunology , Sezary Syndrome/immunology , Sezary Syndrome/mortality , Sezary Syndrome/pathology , Skin/immunology , Skin/pathology , Skin Neoplasms/immunology , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Survival Analysis , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/immunology , Zinc Finger E-box-Binding Homeobox 1/deficiency , Zinc Finger E-box-Binding Homeobox 1/immunology
6.
Front Immunol ; 9: 2604, 2018.
Article in English | MEDLINE | ID: mdl-30483264

ABSTRACT

The role of Epithelial to Mesenchymal Transition (EMT) factor Zeb1 is well defined in metastasis and cancer progression but it's importance in dendritic cells (DCs) is unexplored until now. For the first time we report here that Zeb1 controls immunogenic responses of CD8α+ conventional Type-I (cDC1) DCs. We found that ZEB1 expression increases significantly after TLR9 stimulation and its depletion impairs activation, co-stimulation and secretion of important cytokines like IL-6, IL-10 and IL-12 in cDC1 MutuDC line. We further confirmed our findings in primary cDC1 DCs derived from bone marrow. Co-culture of these Zeb1 knock down (KD) DCs with OT-II CD4+ T helper cells skewed their differentiation toward Th2 subtype. Moreover, adoptive transfer of activated Zeb1 KD DCs cleared intestinal worms in helminth infected mice by increasing Th2 responses in vivo. Integrative genomic analysis showed Zeb1 as an activator of immune response genes in cDC1 MutuDCs as compared to other pathway genes. In addition, differentially regulated genes in Zeb1 KD RNA-seq showed significant enrichment of Th2 activation pathways supporting our in vitro findings. Mechanistically, we showed that decreased IL-12 secreted by Zeb1 KD DCs is the plausible mechanism for increased Th2 differentiation. Collectively our data demonstrate that Zeb1 could be targeted in DCs to modulate T-cell mediated adaptive immune responses.


Subject(s)
Dendritic Cells/immunology , Epithelial-Mesenchymal Transition/immunology , Th1 Cells/immunology , Zinc Finger E-box-Binding Homeobox 1/immunology , Adaptive Immunity/immunology , Adoptive Transfer/methods , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cells, Cultured , Coculture Techniques/methods , Female , HEK293 Cells , Humans , Interleukin-10/immunology , Interleukin-12/immunology , Interleukin-6/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , T-Lymphocytes, Helper-Inducer/immunology , Th2 Cells/immunology , Toll-Like Receptor 9/immunology
7.
Cell Rep ; 22(13): 3574-3586, 2018 03 27.
Article in English | MEDLINE | ID: mdl-29590624

ABSTRACT

Microglia are a key immune-competent cell type that respond to environmental and physiological changes during ischemic stroke. However, the molecular mechanisms controlling post-ischemic microglia activity are unclear. Understanding these mechanisms may ultimately reduce disease burden and allow the manipulation of microglia responses to shape the outcomes of stroke. Here, we report that, after experimentally induced stroke, ZEB1 is highly expressed in ipsilateral cerebral hemisphere, where it is upregulated mainly in microglia. Using a conditional transgenic mouse, we found that ZEB1 upregulation in microglia regulates immune responses in the CNS and alleviates brain injury after ischemic stroke. Our data indicate that ZEB1 overexpression mediates microglia responses and, in turn, inhibits the production of astrocytic CXCL1 through the TGF-ß1-dependent pathway. Reduced CXCL1 leads to a decline in neutrophil infiltration into the brain, thereby reducing CNS inflammation. Our results demonstrate the importance of ZEB1 in microglia-orchestrated neuroinflammation and suggest a potential means for reducing stroke-induced neurological injury.


Subject(s)
Brain Ischemia/metabolism , Microglia/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism , Acute Disease , Animals , Brain Ischemia/genetics , Brain Ischemia/immunology , Brain Ischemia/pathology , Chemokine CXCL1/antagonists & inhibitors , Chemokine CXCL1/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/immunology , Microglia/pathology , Up-Regulation , Zinc Finger E-box-Binding Homeobox 1/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...