Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Biochem Biophys Res Commun ; 715: 150007, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38678783

ABSTRACT

Smad4, a critical mediator of TGF-ß signaling, plays a pivotal role in regulating various cellular functions, including immune responses. In this study, we investigated the impact of Smad4 knockout specifically in macrophages on anti-tumor immunity, focusing on lung metastasis of B16 melanoma cells. Using a mouse model with Smad4 knockout in macrophages established via Lyz2-cre mice and Smad4 flox/flox mice, we demonstrated a significant inhibition of B16 metastasis in the lungs. Interestingly, the inhibition of tumor growth was found to be independent of adaptive immunity, as no significant changes were observed in the numbers or activities of T cells, B cells, or NK cells. Instead, Smad4 knockout led to the emergence of an MCHIIlow CD206high subset of lung interstitial macrophages, characterized by enhanced phagocytosis function. Our findings highlight the crucial role of Smad4 in modulating the innate immune response against tumors and provide insights into potential therapeutic strategies targeting lung interstitial macrophages to enhance anti-tumor immunity.


Subject(s)
Lung Neoplasms , Melanoma, Experimental , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis , Smad4 Protein , Animals , Smad4 Protein/deficiency , Smad4 Protein/genetics , Smad4 Protein/metabolism , Lung Neoplasms/secondary , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Melanoma, Experimental/pathology , Melanoma, Experimental/immunology , Mice , Macrophages/immunology , Macrophages/metabolism , Lung/pathology , Lung/immunology , Lung/metabolism , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Cell Line, Tumor
2.
Nat Commun ; 12(1): 6374, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34737283

ABSTRACT

The transcriptional effector SMAD4 is a core component of the TGF-ß family signaling pathways. However, its role in vertebrate embryo development remains unresolved. To address this, we deleted Smad4 in zebrafish and investigated the consequences of this on signaling by the TGF-ß family morphogens, BMPs and Nodal. We demonstrate that in the absence of Smad4, dorsal/ventral embryo patterning is disrupted due to the loss of BMP signaling. However, unexpectedly, Nodal signaling is maintained, but lacks robustness. This Smad4-independent Nodal signaling is sufficient for mesoderm specification, but not for optimal endoderm specification. Furthermore, using Optical Projection Tomography in combination with 3D embryo morphometry, we have generated a BMP morphospace and demonstrate that Smad4 mutants are morphologically indistinguishable from embryos in which BMP signaling has been genetically/pharmacologically perturbed. Smad4 is thus differentially required for signaling by different TGF-ß family ligands, which has implications for diseases where Smad4 is mutated or deleted.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Nodal Protein/metabolism , Smad4 Protein/metabolism , Transforming Growth Factor beta/metabolism , Zebrafish Proteins/metabolism , Zebrafish/growth & development , Animals , Embryonic Development , Endoderm/metabolism , Gene Knockout Techniques , Mesoderm/metabolism , Morphogenesis , Signal Transduction , Smad4 Protein/deficiency , Smad4 Protein/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
3.
Clin Transl Sci ; 14(5): 1822-1829, 2021 09.
Article in English | MEDLINE | ID: mdl-34002944

ABSTRACT

SMAD4, a tumor suppressor gene, is lost in up to 60%-90% of pancreatic adenocarcinomas (PDAs). Loss of SMAD4 allows tumor progression by upregulating autophagy, a cell survival mechanism that counteracts apoptosis and allows intracellular recycling of macromolecules. Hydroxychloroquine (HCQ) is an autophagy inhibitor. We studied whether HCQ treatment in SMAD4 deficient PDA may prevent therapeutic resistance induced by autophagy upregulation. We retrospectively analyzed the SMAD4 status of patients with PDA enrolled in two prospective clinical trials evaluating pre-operative HCQ. The first dose escalation trial demonstrated the safety of preoperative gemcitabine with HCQ (NCT01128296). More recently, a randomized trial of gemcitabine/nab-paclitaxel +/- HCQ evaluated Evans Grade histopathologic response (NCT01978184). The effect of SMAD4 loss on response to HCQ and chemotherapy was studied for association with clinical outcome. Fisher's exact test and log-rank test were used to assess response and survival. Fifty-two patients receiving HCQ with neoadjuvant chemotherapy were studied. Twenty-five patients had SMAD4 loss (48%). 76% of HCQ-treated patients with SMAD4 loss obtained a histopathologic response greater than or equal to 2A, compared with only 37% with SMAD4 intact (p = 0.006). Although loss of SMAD4 has been associated with worse outcomes, in the current study, loss of SMAD4 was not associated with a detriment in median overall survival in HCQ-treated patients (34.43 months in SMAD4 loss vs. 27.27 months in SMAD4 intact, p = 0.18). The addition of HCQ to neoadjuvant chemotherapy in patients with PDA may improve treatment response in those with SMAD4 loss. Further study of the relationship among SMAD4, autophagy, and treatment outcomes in PDA is warranted.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Pancreatic Ductal/therapy , Hydroxychloroquine/pharmacology , Neoplasm Recurrence, Local/epidemiology , Pancreatic Neoplasms/therapy , Smad4 Protein/genetics , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Autophagy/drug effects , Autophagy/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Disease-Free Survival , Drug Resistance, Neoplasm/genetics , Female , Humans , Hydroxychloroquine/therapeutic use , Male , Middle Aged , Neoadjuvant Therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/prevention & control , Pancreas/pathology , Pancreas/surgery , Pancreatectomy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Retrospective Studies , Smad4 Protein/deficiency , Treatment Outcome
4.
Nat Commun ; 12(1): 2809, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33990575

ABSTRACT

The paradoxical roles of transforming growth factor-ß (TGFß) signaling and nuclear receptor Nur77 in colon cancer development are known but the underlying mechanisms remain obscure. Inhibitor of differentiation 1 (ID1) is a target gene of TGFß and a key promoter for colon cancer progression. Here, we show that Nur77 enhances TGFß/Smad3-induced ID1 mRNA expression through hindering Smurf2-mediated Smad3 mono-ubiquitylation, resulting in ID1 upregulation. In the absence of TGFß, however, Nur77 destabilizes ID1 protein by promoting Smurf2-mediated ID1 poly-ubiquitylation, resulting in ID1 downregulation. Interestingly, TGFß stabilizes ID1 protein by switching Nur77 interaction partners to inhibit ID1 ubiquitylation. This also endows TGFß with an active pro-tumorigenic action in Smad4-deficient colon cancers. Thus, TGFß converts Nur77's role from destabilizing ID1 protein and cancer inhibition to inducing ID1 mRNA expression and cancer promotion, which is highly relevant to colon cancer stemness, metastasis and oxaliplatin resistance. Our data therefore define the integrated duality of Nur77 and TGFß signaling in regulating ID1 expression and provide mechanistic insights into the paradoxical roles of TGFß and Nur77 in colon cancer progression.


Subject(s)
Colonic Neoplasms/etiology , Colonic Neoplasms/metabolism , Inhibitor of Differentiation Protein 1/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Transforming Growth Factor beta/metabolism , Animals , Carcinogenesis , Cell Line, Tumor , Colonic Neoplasms/genetics , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Inhibitor of Differentiation Protein 1/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Models, Biological , Protein Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Smad3 Protein/metabolism , Smad4 Protein/deficiency , Smad4 Protein/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
5.
Cell Mol Gastroenterol Hepatol ; 12(2): 689-713, 2021.
Article in English | MEDLINE | ID: mdl-33774196

ABSTRACT

BACKGROUND & AIMS: Esophageal adenocarcinoma (EAC) develops from its precursor Barrett's esophagus through intermediate stages of low- and high-grade dysplasia. However, knowledge of genetic drivers and molecular mechanisms implicated in disease progression is limited. Herein, we investigated the effect of Mothers against decapentaplegic homolog 4 (SMAD4) loss on transforming growth factor ß (TGF-ß) signaling functionality and in vivo tumorigenicity in high-grade dysplastic Barrett's cells. METHODS: An in vivo xenograft model was used to test tumorigenicity of SMAD4 knockdown or knockout in CP-B high-grade dysplastic Barrett's cells. RT2 polymerase chain reaction arrays were used to analyze TGF-ß signaling functionality, and low-coverage whole-genome sequencing was performed to detect copy number alterations upon SMAD4 loss. RESULTS: We found that SMAD4 knockout significantly alters the TGF-ß pathway target gene expression profile. SMAD4 knockout positively regulates potential oncogenes such as CRYAB, ACTA2, and CDC6, whereas the CDKN2A/B tumor-suppressor locus was regulated negatively. We verified that SMAD4 in combination with CDC6-CDKN2A/B or CRYAB genetic alterations in patient tumors have significant predictive value for poor prognosis. Importantly, we investigated the effect of SMAD4 inactivation in Barrett's tumorigenesis. We found that genetic knockdown or knockout of SMAD4 was sufficient to promote tumorigenesis in dysplastic Barrett's esophagus cells in vivo. Progression to invasive EAC was accompanied by distinctive and consistent copy number alterations in SMAD4 knockdown or knockout xenografts. CONCLUSIONS: Altogether, up-regulation of oncogenes, down-regulation of tumor-suppressor genes, and chromosomal instability within the tumors after SMAD4 loss implicates SMAD4 as a protector of genome integrity in EAC development and progression. Foremost, SMAD4 loss promotes tumorigenesis from dysplastic Barrett's toward EAC.


Subject(s)
Barrett Esophagus/pathology , Carcinogenesis/pathology , Smad4 Protein/metabolism , Xenograft Model Antitumor Assays , Animals , Barrett Esophagus/genetics , Base Sequence , Carcinogenesis/genetics , Cell Line , Down-Regulation , Gene Dosage , Genes, Tumor Suppressor , Humans , Mice , Neoplasm Metastasis , Oncogenes , Principal Component Analysis , Signal Transduction , Smad4 Protein/deficiency , Transforming Growth Factor beta/metabolism
6.
Protein Cell ; 12(2): 128-144, 2021 02.
Article in English | MEDLINE | ID: mdl-32737864

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) has poor prognosis due to limited therapeutic options. This study examines the roles of genome-wide association study identified PDAC-associated genes as therapeutic targets. We have identified HNF4G gene whose silencing most effectively repressed PDAC cell invasiveness. HNF4G overexpression is induced by the deficiency of transcriptional factor and tumor suppressor SMAD4. Increased HNF4G are correlated with SMAD4 deficiency in PDAC tumor samples and associated with metastasis and poor survival time in xenograft animal model and in patients with PDAC (log-rank P = 0.036; HR = 1.60, 95% CI = 1.03-2.47). We have found that Metformin suppresses HNF4G activity via AMPK-mediated phosphorylation-coupled ubiquitination degradation and inhibits in vitro invasion and in vivo metastasis of PDAC cells with SMAD4 deficiency. Furthermore, Metformin treatment significantly improve clinical outcomes and survival in patients with SMAD4-deficient PDAC (log-rank P = 0.022; HR = 0.31, 95% CI = 0.14-0.68) but not in patients with SMAD4-normal PDAC. Pathway analysis shows that HNF4G may act in PDAC through the cell-cell junction pathway. These results indicate that SMAD4 deficiency-induced overexpression of HNF4G plays a critical oncogenic role in PDAC progression and metastasis but may form a druggable target for Metformin treatment.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Hepatocyte Nuclear Factor 4 , Metformin/pharmacology , Pancreatic Neoplasms , Smad4 Protein/deficiency , Up-Regulation/drug effects , Animals , Cell Line , Female , Hepatocyte Nuclear Factor 4/biosynthesis , Hepatocyte Nuclear Factor 4/genetics , Humans , Mice, Inbred BALB C , Mice, Nude , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Smad4 Protein/metabolism , Xenograft Model Antitumor Assays
7.
Front Immunol ; 11: 912, 2020.
Article in English | MEDLINE | ID: mdl-32457763

ABSTRACT

Epidermal Langerhans cells (LCs) are skin-resident dendritic cells that are essential for the induction of skin immunity and tolerance. Transforming growth factor-ß 1 (TGFß1) is a crucial factor for LC maintenance and function. However, the underlying TGFß1 signaling pathways remain unclear. Our previous research has shown that the TGFß1/Smad3 signaling pathway does not impact LC homeostasis and maturation. In this study, we generated mice with conditional deletions of either individual Smad2, Smad4, or both Smad2 and Smad4 in the LC lineage or myeloid lineage, to further explore the impact of TGFß1/Smad signaling pathways on LCs. We found that interruption of Smad2 or Smad4 individually or simultaneously in the LC lineage did not significantly impact the maintenance, maturation, antigen uptake, and migration of LCs in vivo or in vitro during steady state. However, the interruption of both Smad2 and Smad4 pathways in the myeloid lineage led to a dramatic inhibition of bone marrow-derived LCs in the inflammatory state. Overall, our data suggest that canonical TGFß1/Smad2/4 signaling pathways are dispensable for epidermal LC homeostasis and maturation at steady state, but are critical for the long-term LC repopulation directly originating from the bone marrow in the inflammatory state.


Subject(s)
Cell Proliferation , Dermatitis/metabolism , Epidermis/metabolism , Langerhans Cells/metabolism , Smad2 Protein/metabolism , Smad4 Protein/metabolism , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Lineage , Cell Movement , Cells, Cultured , Dermatitis/genetics , Dermatitis/immunology , Dermatitis/pathology , Disease Models, Animal , Epidermis/immunology , Epidermis/pathology , Female , Langerhans Cells/immunology , Langerhans Cells/pathology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction , Smad2 Protein/deficiency , Smad2 Protein/genetics , Smad4 Protein/deficiency , Smad4 Protein/genetics , Transforming Growth Factor beta1/metabolism
8.
Clin Cancer Res ; 26(12): 3058-3070, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32139402

ABSTRACT

PURPOSE: SMAD4 loss causes genomic instability and the initiation/progression of head and neck squamous cell carcinoma (HNSCC). Here, we study whether SMAD4 loss sensitizes HNSCCs to olaparib (PARP inhibitor) in combination with radiotherapy (RT). EXPERIMENTAL DESIGN: We analyzed HNSCC The Cancer Genome Atlas data for SMAD4 expression in association with FANC/BRCA family gene expression. Human HNSCC cell lines were screened for sensitivity to olaparib. Isogenic HNSCC cell lines were generated to restore or reduce SMAD4 expression and treated with olaparib, radiation, or the combination. HNSCC pretreatment specimens from a phase I trial investigating olaparib were analyzed. RESULTS: SMAD4 levels correlated with levels of FANC/BRCA genes in HNSCC. HNSCC cell lines with SMAD4 homozygous deletion were sensitive to olaparib. In vivo, olaparib or RT monotherapy reduced tumor volumes in SMAD4-mutant but not SMAD4-positive tumors. Olaparib with RT dual therapy sustained tumor volume reduction in SMAD4-deficient (mutant or knockdown) xenografts, which exhibited increased DNA damage and cell death compared with vehicle-treated tumors. In vitro, olaparib alone or in combination with radiation caused lower clonogenic survival, more DNA damage-associated cell death, and less proliferation in SMAD4-deficient cells than in SMAD4-positive (endogenous SMAD4 or transduced SMAD4) cells. Applicable to clinic, 5 out of 6 SMAD4-negative HNSCCs and 4 out of 8 SMAD4-positive HNSCCs responded to a standard treatment plus olaparib in a phase I clinical trial, and SMAD4 protein levels inversely correlated with DNA damage. CONCLUSIONS: SMAD4 levels are causal in determining sensitivity to PARP inhibition in combination with RT in HNSCCs.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Disease Models, Animal , Head and Neck Neoplasms/radiotherapy , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Smad4 Protein/deficiency , Squamous Cell Carcinoma of Head and Neck/radiotherapy , Animals , Apoptosis , Cell Proliferation , Cetuximab/administration & dosage , Female , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Mice , Mice, Nude , Phthalazines/administration & dosage , Piperazines/administration & dosage , Prognosis , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Immunity ; 52(2): 328-341.e5, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32049050

ABSTRACT

Fever, an evolutionarily conserved physiological response to infection, is also commonly associated with many autoimmune diseases, but its role in T cell differentiation and autoimmunity remains largely unclear. T helper 17 (Th17) cells are critical in host defense and autoinflammatory diseases, with distinct phenotypes and pathogenicity. Here, we show that febrile temperature selectively regulated Th17 cell differentiation in vitro in enhancing interleukin-17 (IL-17), IL-17F, and IL-22 expression. Th17 cells generated under febrile temperature (38.5°C-39.5°C), compared with those under 37°C, showed enhanced pathogenic gene expression with increased pro-inflammatory activities in vivo. Mechanistically, febrile temperature promoted SUMOylation of SMAD4 transcription factor to facilitate its nuclear localization; SMAD4 deficiency selectively abrogated the effects of febrile temperature on Th17 cell differentiation both in vitro and ameliorated an autoimmune disease model. Our results thus demonstrate a critical role of fever in shaping adaptive immune responses with implications in autoimmune diseases.


Subject(s)
Body Temperature/immunology , Fever/immunology , Th17 Cells/immunology , Adaptive Immunity , Animals , Cell Differentiation/immunology , Cell Nucleus/metabolism , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Fever/genetics , Gene Expression Regulation , Heat-Shock Response/immunology , Mice , Smad4 Protein/deficiency , Smad4 Protein/metabolism , Sumoylation , Th17 Cells/metabolism
10.
Cells ; 8(6)2019 06 19.
Article in English | MEDLINE | ID: mdl-31248101

ABSTRACT

ERBB2 is an oncogenic driver with frequent gene mutations and amplification in human lung tumors and is an attractive target for lung cancer therapy. However, target therapies can be improved by understanding the in vivo mechanisms regulated by ERBB2 during lung tumor development. Here, we generated genetic mouse models to show that Erbb2 loss inhibited lung tumor development induced by deletion of Pten and Smad4. Transcriptome analysis showed that Erbb2 loss suppressed the significant changes of most of the induced genes by ablation of Pten and Smad4. Overlapping with ERBB2-associated human lung cancer genes further identified those ERBB2 downstream players potentially conserved in human and mouse lung tumors. Furthermore, MED24 was identified as a crucial oncogenic target of ERBB2 in lung tumor development. Taken together, ERBB2 is required for the dysregulation of cancer-related genes, such as MED24, during lung tumor development.


Subject(s)
Disease Progression , Epithelium/metabolism , Gene Deletion , Lung Neoplasms/pathology , Mediator Complex/metabolism , PTEN Phosphohydrolase/deficiency , Receptor, ErbB-2/metabolism , Smad4 Protein/deficiency , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Epithelium/pathology , Gene Expression Regulation, Neoplastic , Humans , Lung/pathology , Lung Neoplasms/genetics , Mice , PTEN Phosphohydrolase/metabolism , Smad4 Protein/metabolism
11.
Nat Genet ; 51(5): 777-785, 2019 05.
Article in English | MEDLINE | ID: mdl-30988513

ABSTRACT

BMP/SMAD signaling is a crucial regulator of intestinal differentiation1-4. However, the molecular underpinnings of the BMP pathway in this context are unknown. Here, we characterize the mechanism by which BMP/SMAD signaling drives enterocyte differentiation. We establish that the transcription factor HNF4A acts redundantly with an intestine-restricted HNF4 paralog, HNF4G, to activate enhancer chromatin and upregulate the majority of transcripts enriched in the differentiated epithelium; cells fail to differentiate on double knockout of both HNF4 paralogs. Furthermore, we show that SMAD4 and HNF4 function via a reinforcing feed-forward loop, activating each other's expression and co-binding to regulatory elements of differentiation genes. This feed-forward regulatory module promotes and stabilizes enterocyte cell identity; disruption of the HNF4-SMAD4 module results in loss of enterocyte fate in favor of progenitor and secretory cell lineages. This intersection of signaling and transcriptional control provides a framework to understand regenerative tissue homeostasis, particularly in tissues with inherent cellular plasticity5.


Subject(s)
Enterocytes/cytology , Enterocytes/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Smad4 Protein/metabolism , Animals , Binding Sites/genetics , Caco-2 Cells , Cell Differentiation/genetics , Cell Differentiation/physiology , Enhancer Elements, Genetic , Hepatocyte Nuclear Factor 4/deficiency , Hepatocyte Nuclear Factor 4/genetics , Humans , Mice , Mice, Knockout , Signal Transduction , Smad4 Protein/deficiency , Smad4 Protein/genetics
12.
Circulation ; 139(17): 2049-2063, 2019 04 23.
Article in English | MEDLINE | ID: mdl-30744395

ABSTRACT

BACKGROUND: Hereditary hemorrhagic telangiectasia is an autosomal dominant vascular disorder caused by heterozygous, loss-of-function mutations in 4 transforming growth factor beta (TGFß) pathway members, including the central transcriptional mediator of the TGFß pathway, Smad4. Loss of Smad4 causes the formation of inappropriate, fragile connections between arteries and veins called arteriovenous malformations (AVMs), which can hemorrhage leading to stroke, aneurysm, or death. Unfortunately, the molecular mechanisms underlying AVM pathogenesis remain poorly understood, and the TGFß downstream effectors responsible for hereditary hemorrhagic telangiectasia-associated AVM formation are currently unknown. METHODS: To identify potential biological targets of the TGFß pathway involved in AVM formation, we performed RNA- and chromatin immunoprecipitation-sequencing experiments on BMP9 (bone morphogenetic protein 9)-stimulated endothelial cells (ECs) and isolated ECs from a Smad4-inducible, EC-specific knockout ( Smad4-iECKO) mouse model that develops retinal AVMs. These sequencing studies identified the angiopoietin-Tek signaling pathway as a downstream target of SMAD4. We used monoclonal blocking antibodies to target a specific component in this pathway and assess its effects on AVM development. RESULTS: Sequencing studies uncovered 212 potential biological targets involved in AVM formation, including the EC surface receptor, TEK (TEK receptor tyrosine kinase) and its antagonistic ligand, ANGPT2 (angiopoietin-2). In Smad4-iECKO mice, Angpt2 expression is robustly increased, whereas Tek levels are decreased, resulting in an overall reduction in angiopoietin-Tek signaling. We provide evidence that SMAD4 directly represses Angpt2 transcription in ECs. Inhibition of ANGPT2 function in Smad4-deficient mice, either before or after AVMs form, prevents and alleviates AVM formation and normalizes vessel diameters. These rescue effects are attributed to a reversion in EC morphological changes, such as cell size and shape that are altered in the absence of Smad4. CONCLUSIONS: Our studies provide a novel mechanism whereby the loss of Smad4 causes increased Angpt2 transcription in ECs leading to AVM formation, increased blood vessel calibers, and changes in EC morphology in the retina. Blockade of ANGPT2 function in an in vivo Smad4 model of hereditary hemorrhagic telangiectasia alleviated these vascular phenotypes, further implicating ANGPT2 as an important TGFß downstream mediator of AVM formation. Therefore, alternative approaches that target ANGPT2 function may have therapeutic value for the alleviation of hereditary hemorrhagic telangiectasia symptoms, such as AVMs.


Subject(s)
Angiopoietin-2/antagonists & inhibitors , Arteriovenous Malformations/prevention & control , Smad4 Protein/deficiency , Telangiectasia, Hereditary Hemorrhagic/complications , Angiopoietin-2/biosynthesis , Angiopoietin-2/genetics , Animals , Arteriovenous Malformations/etiology , Arteriovenous Malformations/metabolism , Arteriovenous Malformations/pathology , Cell Size , Disease Models, Animal , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Gene Expression Regulation , Mice , Mice, Knockout , Receptor, TIE-2/physiology , Signal Transduction , Smad4 Protein/genetics , Smad4 Protein/physiology , Telangiectasia, Hereditary Hemorrhagic/genetics , Transcription, Genetic , Transforming Growth Factor beta/metabolism
13.
J Biol Chem ; 294(11): 3974-3986, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30659096

ABSTRACT

Hypoxia-inducible factor 2α (HIF2α) directly regulates a battery of genes essential for intestinal iron absorption. Interestingly, iron deficiency and overload disorders do not result in increased intestinal expression of glycolytic or angiogenic HIF2α target genes. Similarly, inflammatory and tumor foci can induce a distinct subset of HIF2α target genes in vivo These observations indicate that different stimuli activate distinct subsets of HIF2α target genes via mechanisms that remain unclear. Here, we conducted a high-throughput siRNA-based screen to identify genes that regulate HIF2α's transcriptional activity on the promoter of the iron transporter gene divalent metal transporter-1 (DMT1). SMAD family member 3 (SMAD3) and SMAD4 were identified as potential transcriptional repressors. Further analysis revealed that SMAD4 signaling selectively represses iron-absorptive gene promoters but not the inflammatory or glycolytic HIF2α or HIF1α target genes. Moreover, the highly homologous SMAD2 did not alter HIF2α transcriptional activity. During iron deficiency, SMAD3 and SMAD4 expression was significantly decreased via proteasomal degradation, allowing for derepression of iron target genes. Several iron-regulatory genes contain a SMAD-binding element (SBE) in their proximal promoters; however, mutation of the putative SBE on the DMT1 promoter did not alter the repressive function of SMAD3 or SMAD4. Importantly, the transcription factor forkhead box protein A1 (FOXA1) was critical in SMAD4-induced DMT1 repression, and DNA binding of SMAD4 was essential for the repression of HIF2α activity, suggesting an indirect repressive mechanism through DNA binding. These results provide mechanistic clues to how HIF signaling can be regulated by different cellular cues.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Iron-Regulatory Proteins/metabolism , Smad3 Protein/metabolism , Smad4 Protein/metabolism , Animals , Cells, Cultured , Humans , Iron-Regulatory Proteins/genetics , Mice , Mice, Knockout , Smad3 Protein/deficiency , Smad4 Protein/deficiency
14.
Clin Cancer Res ; 25(6): 1948-1956, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30587545

ABSTRACT

PURPOSE: SMAD4 has shown promise in identifying patients with colorectal cancer at high risk of recurrence or death.Experimental Design: A discovery cohort and independent validation cohort were classified by SMAD4 status. SMAD4 status and immune infiltrate measurements were tested for association with recurrence-free survival (RFS). Patient-derived xenografts from SMAD4-deficient and SMAD4-retained tumors were used to examine chemoresistance. RESULTS: The discovery cohort consisted of 364 patients with stage I-IV colorectal cancer. Median age at diagnosis was 53 years. The cohort consisted of 61% left-sided tumors and 62% stage II/III patients. Median follow-up was 5.4 years (interquartile range, 2.3-8.2). SMAD4 loss, noted in 13% of tumors, was associated with higher tumor and nodal stage, adjuvant therapy use, fewer tumor-infiltrating lymphocytes (TIL), and lower peritumoral lymphocyte aggregate (PLA) scores (all P < 0.04). SMAD4 loss was associated with worse RFS (P = 0.02). When stratified by SMAD4 and immune infiltrate status, patients with SMAD4 loss and low TIL or PLA had worse RFS (P = 0.002 and P = 0.006, respectively). Among patients receiving 5-fluorouracil (5-FU)-based systemic chemotherapy, those with SMAD4 loss had a median RFS of 3.8 years compared with 13 years for patients with SMAD4 retained. In xenografted mice, the SMAD4-lost tumors displayed resistance to 5-FU. An independent cohort replicated our findings, in particular, the association of SMAD4 loss with decreased immune infiltrate, as well as worse disease-specific survival. CONCLUSIONS: Our data show SMAD4 loss correlates with worse clinical outcome, resistance to chemotherapy, and decreased immune infiltrate, supporting its use as a prognostic marker in patients with colorectal cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Biomarkers, Tumor/deficiency , Colorectal Neoplasms/pathology , Neoplasm Recurrence, Local/diagnosis , Smad4 Protein/deficiency , Adult , Aged , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/immunology , Chemotherapy, Adjuvant/methods , Colon/pathology , Colon/surgery , Colorectal Neoplasms/immunology , Colorectal Neoplasms/mortality , Colorectal Neoplasms/therapy , Disease-Free Survival , Drug Resistance, Neoplasm/immunology , Female , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Follow-Up Studies , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Middle Aged , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/prevention & control , Prognosis , Prospective Studies , Rectum/pathology , Rectum/surgery , Smad4 Protein/immunology , Xenograft Model Antitumor Assays
15.
J Am Heart Assoc ; 7(21): e009514, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30571376

ABSTRACT

Background Hereditary hemorrhagic telangiectasia ( HHT ) is a rare genetic vascular disorder caused by mutations in endoglin ( ENG ), activin receptor-like kinase 1 ( ACVRL 1; ALK 1), or SMAD 4. Major clinical symptoms of HHT are arteriovenous malformations ( AVM s) found in the brain, lungs, visceral organs, and mucosal surface. Animal models harboring mutations in Eng or Alk1 recapitulate all of these HHT clinical symptoms and have been useful resources for studying mechanisms and testing potential drugs. However, animal models representing SMAD 4 mutations have been lacking. The goal of this study is to evaluate Smad4-inducible knockout ( iKO ) mice as an animal model of HHT and compare the phenotypes with other established HHT animal models. Methods and Results Global Smad4 deletion was induced at neonatal and adult stages, and hemoglobin levels, gastrointestinal hemorrhage, and presence of aberrant arteriovenous connections were examined. Neonatal Smad4- iKO mice exhibited signs of gastrointestinal bleeding and AVM s in the brain, intestine, nose, and retina. The radial expansion was decreased, and AVM s were detected on both distal and proximal retinal vasculature of Smad4- iKO s. Aberrant smooth muscle actin staining was observed in the initial stage AVM s and their connecting veins, indicating abnormal arterial flow to veins. In adult mice, Smad4 deficiency caused gastrointestinal bleeding and AVM s along the gastrointestinal tract and wounded skin. HHT -related phenotypes of Smad4- iKO s appeared to be comparable with those found in Alk1- iKO and Eng- iKO mice. Conclusions These data further confirm that SMAD signaling is crucial for normal arteriovenous network formation, and Smad4- iKO will be an alternative animal model of AVM research associated with HHT .


Subject(s)
Arteriovenous Malformations/genetics , Disease Models, Animal , Mice , Smad4 Protein/deficiency , Smad4 Protein/genetics , Telangiectasia, Hereditary Hemorrhagic/genetics , Age Factors , Animals , Animals, Newborn , Mice, Knockout , Phenotype
16.
Aging (Albany NY) ; 10(12): 3683-3701, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30530919

ABSTRACT

Thoracic aortic aneurysm and dissection (TAAD) is the most fatal macro vascular disease. The mortality of 48h after diagnosis of dissection is up to approximately 50-68%. However, the genetic factors and potential mechanism underlying sporadic TAAD remain largely unknown. Our previous study suggested rs12455792 variant of SMAD4 gene significantly contributed to the increased risk and might participated the pathological progression of TAAD. This investigation aims to test (1) the associations between rs12455792 and MØ recruitment, inflammatory response in aggressiveness of TAAD, and (2) the molecular mechanism accounting for their effects. In TGF-ß signaling molecular detection, rs12455792 C>T variant activated the canonical and non-canonical TGF-ß mediators. It also increased the secretion of chemotactic factors of HASMCs. To confirm the impact of this change, we detected MØ recruitment and infiltration in HASMCs and aortic tissues of TAAD patients. We found that MØ recruitment in cells and tissues with rs12455792 variant genotypes was increased than that in wild type groups. Moreover, rs12455792 variant increased M1 type inflammatory response, which might contribute much to TAAD progression. To mimic the SMAD4 suppression effect of rs12455792 in vivo, we constructed the SMAD4 KD mouse. After induction with Ang II for 4w, the thoracic aorta dilatation and vascular remodeling were more serious than that of wild type group. In conclusion, rs12455792 increased MØ recruitment, M1 type inflammatory response via activated TGF-ß signaling, and further promoted vascular remodeling and pathological progress of TAAD.


Subject(s)
Aortic Aneurysm, Thoracic/metabolism , Aortic Dissection/metabolism , Inflammation Mediators/metabolism , Macrophages/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Smad4 Protein/metabolism , Transforming Growth Factor beta/metabolism , Aortic Dissection/genetics , Aortic Dissection/pathology , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/pathology , Case-Control Studies , Cells, Cultured , Dilatation, Pathologic , Disease Models, Animal , Female , Macrophages/pathology , Male , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Polymorphism, Single Nucleotide , Signal Transduction , Smad4 Protein/deficiency , Smad4 Protein/genetics , Vascular Remodeling
17.
Angiogenesis ; 21(2): 363-380, 2018 05.
Article in English | MEDLINE | ID: mdl-29460088

ABSTRACT

Hereditary hemorrhagic telangiectasia (HHT) is an autosomal dominant vascular disorder that leads to abnormal connections between arteries and veins termed arteriovenous malformations (AVM). Mutations in TGFß pathway members ALK1, ENG and SMAD4 lead to HHT. However, a Smad4 mouse model of HHT does not currently exist. We aimed to create and characterize a Smad4 endothelial cell (EC)-specific, inducible knockout mouse (Smad4f/f;Cdh5-CreERT2) that could be used to study AVM development in HHT. We found that postnatal ablation of Smad4 caused various vascular defects, including the formation of distinct AVMs in the neonate retina. Our analyses demonstrated that increased EC proliferation and size, altered mural cell coverage and distorted artery-vein gene expression are associated with Smad4 deficiency in the vasculature. Furthermore, we show that depletion of Smad4 leads to decreased Vegfr2 expression, and concurrent loss of endothelial Smad4 and Vegfr2 in vivo leads to AVM enlargement. Our work provides a new model in which to study HHT-associated phenotypes and links the TGFß and VEGF signaling pathways in AVM pathogenesis.


Subject(s)
Arteriovenous Malformations , Endothelial Cells , Eye Proteins/metabolism , Retinal Vessels , Smad4 Protein/deficiency , Telangiectasia, Hereditary Hemorrhagic , Animals , Arteriovenous Malformations/genetics , Arteriovenous Malformations/metabolism , Arteriovenous Malformations/pathology , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Eye Proteins/genetics , Mice , Mice, Knockout , Retinal Vessels/abnormalities , Retinal Vessels/metabolism , Telangiectasia, Hereditary Hemorrhagic/genetics , Telangiectasia, Hereditary Hemorrhagic/metabolism , Telangiectasia, Hereditary Hemorrhagic/pathology
18.
Nature ; 551(7678): 105-109, 2017 11 02.
Article in English | MEDLINE | ID: mdl-29072299

ABSTRACT

T helper 17 (TH17) cells are critically involved in host defence, inflammation, and autoimmunity. Transforming growth factor ß (TGFß) is instrumental in TH17 cell differentiation by cooperating with interleukin-6 (refs 6, 7). Yet, the mechanism by which TGFß enables TH17 cell differentiation remains elusive. Here we reveal that TGFß enables TH17 cell differentiation by reversing SKI-SMAD4-mediated suppression of the expression of the retinoic acid receptor (RAR)-related orphan receptor γt (RORγt). We found that, unlike wild-type T cells, SMAD4-deficient T cells differentiate into TH17 cells in the absence of TGFß signalling in a RORγt-dependent manner. Ectopic SMAD4 expression suppresses RORγt expression and TH17 cell differentiation of SMAD4-deficient T cells. However, TGFß neutralizes SMAD4-mediated suppression without affecting SMAD4 binding to the Rorc locus. Proteomic analysis revealed that SMAD4 interacts with SKI, a transcriptional repressor that is degraded upon TGFß stimulation. SKI controls histone acetylation and deacetylation of the Rorc locus and TH17 cell differentiation via SMAD4: ectopic SKI expression inhibits H3K9 acetylation of the Rorc locus, Rorc expression, and TH17 cell differentiation in a SMAD4-dependent manner. Therefore, TGFß-induced disruption of SKI reverses SKI-SMAD4-mediated suppression of RORγt to enable TH17 cell differentiation. This study reveals a critical mechanism by which TGFß controls TH17 cell differentiation and uncovers the SKI-SMAD4 axis as a potential therapeutic target for treating TH17-related diseases.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Smad4 Protein/metabolism , Th17 Cells/cytology , Th17 Cells/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Differentiation/genetics , Female , Gene Deletion , Humans , Interleukin-6/metabolism , Male , Mice , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/deficiency , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Smad4 Protein/deficiency , Smad4 Protein/genetics
19.
Oncotarget ; 8(10): 16704-16711, 2017 Mar 07.
Article in English | MEDLINE | ID: mdl-28053288

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer mortality. Although advances have been made in understanding the pathogenesis of PDAC, the outcome still remains poor. The aim of this study is to conduct a meta-analysis to evaluate the precise association between SMAD4 loss and clinicopathological significance in PDAC. A literature search was made in PubMed, Web of Science, Google scholar, and EMBASE for related publications. The data were extracted and assessed by two reviewers independently. Analysis of pooled data was performed, Odds Ratio or Hazard Ratio with corresponding confidence intervals was calculated and summarized. 12 relevant articles were included for full review in detail and meta-analysis. The frequency of SMAD4 protein loss was significantly increased in PDAC than in nonmalignant pancreatic tissue, Odd Ratio was 0.05 with 95% confidence interval 0.01-0.23, p<0.0001. SMAD4 loss was significantly associated with poor overall survival in patients with PDAC, Hazard Ratio was 0.61 with 95% confidence interval 0.38-0.99, p=0.05. SMAD4 loss was not correlated with the size, grades, and lymph node metastasis of PDAC. In conclusion, SMAD4 is a biomarker for the diagnosis of PDAC. SMAD4 loss is significantly related to poor prognosis in patients with PDAC.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Pancreatic Ductal/genetics , Pancreatic Neoplasms/genetics , Smad4 Protein/deficiency , Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/pathology , Humans , Pancreatic Neoplasms/pathology , Smad4 Protein/genetics
20.
Immunol Cell Biol ; 95(3): 287-296, 2017 03.
Article in English | MEDLINE | ID: mdl-27686408

ABSTRACT

Type 1 diabetes results from autoimmune-mediated pancreatic beta-cell destruction and transforming growth factor-beta (TGF-ß) is known to play a preventive role in type 1 diabetes in non-obese diabetic (NOD) mice. In this study, we investigated the role of Smad4, a key molecule for Smad-dependent TGF-ß signaling, in T cells of NOD mice in the pathogenesis of autoimmune diabetes. We generated T-cell-specific Smad4 knockout (Smad4 tKO) NOD mice and assessed the pathological and immunological changes. Smad4 tKO showed earlier onset and increased incidence of diabetes than wild type (WT) NOD mice. Pathological features such as insulitis, anti-glutamic acid decarboxylase auto-antibody levels and serum IFN-γ levels were significantly increased in Smad4 tKO compared with WT NOD mice. Proportion and number of activated/memory CD4+ T cell were significantly increased in pancreatic lymph nodes of Smad4 tKO compared with WT NOD mice. However, the proportion and function of regulatory T cells was not different. Effector CD4+ T cells from Smad4 tKO were more resistant to suppression by regulatory T cells than effector cells from WT NOD mice. The proliferative potential of effector T cells from Smad4 tKO was significantly elevated compared with WT NOD mice, and activation of sterol regulatory element binding protein-1c (SREBP-1c) in T cells of Smad4 tKO NOD mice was correlated with this proliferative activity. We conclude that Smad4 deletion in T cells of NOD mice accelerated the development of autoimmune diabetes and increased the incidence of the disease by dysregulation of T cell activation at least in part via SREBP-1c activation.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Smad4 Protein/deficiency , T-Lymphocytes/immunology , Animals , Autoantibodies/metabolism , Cell Proliferation , Cytokines/metabolism , Immunologic Memory , Inflammation Mediators/metabolism , Islets of Langerhans/pathology , Lymph Nodes/pathology , Lymphocyte Activation , Male , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Receptors, Antigen, T-Cell/metabolism , Smad4 Protein/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , T-Lymphocytes, Regulatory/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...