Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
EMBO Mol Med ; 12(8): e11222, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32638512

ABSTRACT

Pancreatitis is the inflammation of the pancreas. However, little is known about the genes associated with pancreatitis severity. Our microarray analysis of pancreatic tissues from mild and severe acute pancreatitis mice models identified angiopoietin-like 4 (ANGPTL4) as one of the most significantly upregulated genes. Clinically, ANGPTL4 expression was also increased in the serum and pancreatic tissues of pancreatitis patients. The deficiency in ANGPTL4 in mice, either by gene deletion or neutralizing antibody, mitigated pancreatitis-associated pathological outcomes. Conversely, exogenous ANGPTL4 exacerbated pancreatic injury with elevated cytokine levels and apoptotic cell death. High ANGPTL4 enhanced macrophage activation and infiltration into the pancreas, which increased complement component 5a (C5a) level through PI3K/AKT signaling. The activation of the C5a receptor led to hypercytokinemia that accelerated acinar cell damage and furthered pancreatitis. Indeed, C5a neutralizing antibody decreased inflammatory response in LPS-activated macrophages and alleviated pancreatitis severity. In agreement, there was a significant positive correlation between C5a and ANGPTL4 levels in pancreatitis patients. Taken together, our study suggests that targeting ANGPTL4 is a potential strategy for the treatment of pancreatitis.


Subject(s)
Pancreatitis , Acinar Cells , Acute Disease , Angiopoietin-Like Protein 4/genetics , Animals , Humans , Mice , Mice, Inbred C57BL , Pancreas , Phosphatidylinositol 3-Kinases , Up-Regulation
2.
Mol Cancer ; 17(1): 152, 2018 10 20.
Article in English | MEDLINE | ID: mdl-30342537

ABSTRACT

Overcoming multidrug resistance has always been a major challenge in cancer treatment. Recent evidence suggested epithelial-mesenchymal transition plays a role in MDR, but the mechanism behind this link remains unclear. We found that the expression of multiple ABC transporters was elevated in concordance with an increased drug efflux in cancer cells during EMT. The metastasis-related angiopoietin-like 4 (ANGPTL4) elevates cellular ATP to transcriptionally upregulate ABC transporters expression via the Myc and NF-κB signaling pathways. ANGPTL4 deficiency reduced IC50 of anti-tumor drugs and enhanced apoptosis of cancer cells. In vivo suppression of ANGPTL4 led to higher accumulation of cisplatin-DNA adducts in primary and metastasized tumors, and a reduced metastatic tumor load. ANGPTL4 empowered cancer cells metabolic flexibility during EMT, securing ample cellular energy that fuels multiple ABC transporters to confer EMT-mediated chemoresistance. It suggests that metabolic strategies aimed at suppressing ABC transporters along with energy deprivation of EMT cancer cells may overcome drug resistance.


Subject(s)
Angiopoietin-Like Protein 4/antagonists & inhibitors , Angiopoietin-Like Protein 4/metabolism , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Energy Metabolism/drug effects , Neoplasms/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphate/metabolism , Angiopoietin-Like Protein 4/genetics , Animals , Cell Line, Tumor , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Humans , Mice , Neoplasms/drug therapy , Neoplasms/genetics
3.
Cell Discov ; 4: 15, 2018.
Article in English | MEDLINE | ID: mdl-29619245

ABSTRACT

Connective tissue diseases of the skin are characterized by excessive collagen deposition in the skin and internal organs. Fibroblasts play a pivotal role in the clinical presentation of these conditions. Nuclear receptor peroxisome-proliferator activated receptors (PPARs) are therapeutic targets for dermal fibrosis, but the contribution of the different PPAR subtypes are poorly understood. Particularly, the role of fibroblast PPARß/δ in dermal fibrosis has not been elucidated. Thus, we generated a mouse strain with selective deletion of PPARß/δ in the fibroblast (FSPCre-Pparb/d-/-) and interrogated its epidermal and dermal transcriptome profiles. We uncovered a downregulated gene, leucine-rich alpha-2-glycoprotein-1 (Lrg1), of previously unknown function in skin development and architecture. Our findings suggest that the regulation of Lrg1 by PPARß/δ in fibroblasts is an important signaling conduit integrating PPARß/δ and TGFß1-signaling networks in skin health and disease. Thus, the FSPCre-Pparb/d-/- mouse model could serve as a novel tool in the current gunnery of animal models to better understand dermal fibrosis.

4.
Sci Rep ; 7(1): 6303, 2017 07 24.
Article in English | MEDLINE | ID: mdl-28740178

ABSTRACT

In adult skin wounds, collagen expression rapidly re-establishes the skin barrier, although the resultant scar is aesthetically and functionally inferior to unwounded tissue. Although TGFß signaling and fibroblasts are known to be responsible for scar-associated collagen production, there are currently no prophylactic treatments for scar management. Fibroblasts in crosstalk with wound keratinocytes orchestrate collagen expression, although the precise paracrine pathways involved remain poorly understood. Herein, we showed that the matricellular protein, angiopoietin-like 4 (ANGPTL4), accelerated wound closure and reduced collagen expression in diabetic and ANGPTL4-knockout mice. Similar observations were made in wild-type rat wounds. Using human fibroblasts as a preclinical model for mechanistic studies, we systematically elucidated that ANGPTL4 binds to cadherin-11, releasing membrane-bound ß-catenin which translocate to the nucleus and transcriptionally upregulate the expression of Inhibitor of DNA-binding/differentiation protein 3 (ID3). ID3 interacts with scleraxis, a basic helix-loop-helix transcription factor, to inhibit scar-associated collagen types 1α2 and 3α1 production by fibroblasts. We also showed ANGPTL4 interaction with cadherin-11 in human scar tissue. Our findings highlight a central role for matricellular proteins such as ANGPTL4 in the attenuation of collagen expression and may have a broader implication for other fibrotic pathologies.


Subject(s)
Angiopoietin-Like Protein 4/genetics , Cicatrix/drug therapy , Diabetes Complications/drug therapy , Fibroblasts/cytology , Inhibitor of Differentiation Proteins/genetics , Neoplasm Proteins/genetics , Skin Physiological Phenomena , beta Catenin/metabolism , Angiopoietin-Like Protein 4/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cadherins/metabolism , Cell Proliferation , Cells, Cultured , Cicatrix/genetics , Cicatrix/metabolism , Collagen/metabolism , Diabetes Complications/genetics , Diabetes Complications/metabolism , Fibroblasts/metabolism , Gene Knockout Techniques , Humans , Mice , Rats , Skin/cytology , Up-Regulation , Wound Healing
5.
Biochim Biophys Acta Mol Cell Res ; 1864(10): 1525-1536, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28641978

ABSTRACT

Angiopoietin-like 4 (ANGPTL4) is a secretory protein that can be cleaved to form an N-terminal and a C-terminal protein. Studies performed thus far have linked ANGPTL4 to several cancer-related and metabolic processes. Notably, several point mutations in the C-terminal ANGPTL4 (cANGPTL4) have been reported, although no studies have been performed that ascribed these mutations to cancer-related and metabolic processes. In this study, we compared the characteristics of tumors with and without wild-type (wt) cANGPTL4 and tumors with cANGPTL4 bearing the T266M mutation (T266M cANGPTL4). We found that T266M cANGPTL4 bound to integrin α5ß1 with a reduced affinity compared to wt, leading to weaker activation of downstream signaling molecules. The mutant tumors exhibited impaired proliferation, anoikis resistance, and migratory capability and had reduced adenylate energy charge. Further investigations also revealed that cANGPTL4 regulated the expression of Glut2. These findings may explain the differences in the tumor characteristics and energy metabolism observed with the cANGPTL4 T266M mutation compared to tumors without the mutation.


Subject(s)
Angiopoietin-Like Protein 4/genetics , Glucose Transporter Type 2/genetics , Integrin alpha5beta1/genetics , Liver Neoplasms/genetics , Stomach Neoplasms/genetics , Angiopoietin-Like Protein 4/metabolism , Animals , Anoikis/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Circular Dichroism , Energy Metabolism/genetics , Gene Expression Regulation, Neoplastic , Glucose/metabolism , Glucose Transporter Type 2/metabolism , Hep G2 Cells , Humans , Integrin alpha5beta1/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mutagenesis, Site-Directed , Mutation , Neoplasm Invasiveness/genetics , Polymorphism, Single Nucleotide , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Xenograft Model Antitumor Assays
6.
Sci Rep ; 7: 44351, 2017 03 13.
Article in English | MEDLINE | ID: mdl-28287161

ABSTRACT

Many gastrointestinal diseases exhibit a protracted and aggravated inflammatory response that can lead to hypercytokinaemia, culminating in extensive tissue damage. Recently, angiopoietin-like 4 (ANGPTL4) has been implicated in many inflammation-associated diseases. However, how ANGPTL4 regulates colonic inflammation remains unclear. Herein, we show that ANGPTL4 deficiency in mice (ANGPTL4-/-) exacerbated colonic inflammation induced by dextran sulfate sodium (DSS) or stearic acid. Microbiota was similar between the two genotypes prior DSS challenge. A microarray gene expression profile of the colon from DSS-treated ANGPTL4-/- mice was enriched for genes involved in leukocyte migration and infiltration, and showed a close association to inflamed ulcerative colitis (UC), whereas the profile from ANGPTL4+/+ littermates resembled that of non-inflamed UC biopsies. Bone marrow transplantation demonstrates the intrinsic role of colonic ANGPTL4 in regulating leukocyte infiltration during DSS-induced inflammation. Using immortalized human colon epithelial cells, we revealed that the ANGPTL4-mediated upregulation of tristetraprolin expression operates through CREB and NF-κB transcription factors, which in turn, regulates the stability of chemokines. Together, our findings suggest that ANGPTL4 protects against acute colonic inflammation and that its absence exacerbates the severity of inflammation. Our findings emphasize the importance of ANGPTL4 as a novel target for therapy in regulating and attenuating inflammation.


Subject(s)
Angiopoietin-Like Protein 4/genetics , Chemokines/genetics , Colon/metabolism , Gene Expression Profiling , Inflammation/genetics , Tristetraprolin/genetics , Angiopoietin-Like Protein 4/metabolism , Animals , Cell Line , Chemokines/metabolism , Colitis, Ulcerative/genetics , Colitis, Ulcerative/metabolism , Colon/pathology , Dextran Sulfate , Humans , Inflammation/chemically induced , Inflammation/metabolism , Mice, Inbred C57BL , Mice, Knockout , RNA Stability , Stearic Acids , THP-1 Cells , Tristetraprolin/metabolism
7.
Cell Death Dis ; 8(1): e2562, 2017 01 19.
Article in English | MEDLINE | ID: mdl-28102840

ABSTRACT

Histological inspection of visually normal tissue adjacent to neoplastic lesions often reveals multiple foci of cellular abnormalities. This suggests the presence of a regional carcinogenic signal that spreads oncogenic transformation and field cancerization. We observed an abundance of mutagenic reactive oxygen species in the stroma of cryosectioned patient tumor biopsies, indicative of extratumoral oxidative stress. Diffusible hydrogen peroxide (H2O2) was elevated in the conditioned medium of cultured skin epithelia at various stages of oncogenic transformation, and H2O2 production increased with greater tumor-forming and metastatic capacity of the studied cell lines. Explanted cancer-associated fibroblasts (CAFs) also had higher levels of H2O2 secretion compared with normal fibroblasts (FIBs). These results suggest that extracellular H2O2 acts as a field effect carcinogen. Indeed, H2O2-treated keratinocytes displayed decreased phosphatase and tensin homolog (PTEN) and increased Src activities because of oxidative modification. Furthermore, treating FIBs with CAF-conditioned medium or exogenous H2O2 resulted in the acquisition of an oxidative, CAF-like state. In vivo, the proliferative potential and invasiveness of composite tumor xenografts comprising cancerous or non-tumor-forming epithelia with CAFs and FIBs could be attenuated by the presence of catalase. Importantly, we showed that oxidatively transformed FIBs isolated from composite tumor xenografts retained their ability to promote tumor growth and aggressiveness when adoptively transferred into new xenografts. Higher H2O2 production by CAFs was contingent on impaired TGFß signaling leading to the suppression of the antioxidant enzyme glutathione peroxidase 1 (GPX1). Finally, we detected a reduction in Smad3, TAK1 and TGFßRII expression in a cohort of 197 clinical squamous cell carcinoma (SCC) CAFs, suggesting that impaired stromal TGFß signaling may be a clinical feature of SCC. Our study indicated that CAFs and cancer cells engage redox signaling circuitries and mitogenic signaling to reinforce their reciprocal relationship, suggesting that future anticancer approaches should simultaneously target ligand receptor and redox-mediated pathways.


Subject(s)
Breast Neoplasms/genetics , Cancer-Associated Fibroblasts/metabolism , Carcinoma, Squamous Cell/genetics , Oxidative Stress/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cancer-Associated Fibroblasts/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Hydrogen Peroxide/metabolism , PTEN Phosphohydrolase/biosynthesis , PTEN Phosphohydrolase/genetics , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics , Xenograft Model Antitumor Assays
8.
Cell Rep ; 10(5): 654-663, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25660016

ABSTRACT

Excessive host inflammatory responses negatively impact disease outcomes in respiratory infection. Host-pathogen interactions during the infective phase of influenza are well studied, but little is known about the host's response during the repair stage. Here, we show that influenza infection stimulated the expression of angiopoietin-like 4 (ANGPTL4) via a direct IL6-STAT3-mediated mechanism. ANGPTL4 enhanced pulmonary tissue leakiness and exacerbated inflammation-induced lung damage. Treatment of infected mice with neutralizing anti-ANGPTL4 antibodies significantly accelerated lung recovery and improved lung tissue integrity. ANGPTL4-deficient mice also showed reduced lung damage and recovered faster from influenza infection when compared to their wild-type counterparts. Retrospective examination of human lung biopsy specimens from infection-induced pneumonia with tissue damage showed elevated expression of ANGPTL4 when compared to normal lung samples. These observations underscore the important role that ANGPTL4 plays in lung infection and damage and may facilitate future therapeutic strategies for the treatment of influenza pneumonia.

9.
Mol Cancer Res ; 10(6): 677-88, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22661548

ABSTRACT

Angiopoietin-like 4 (ANGPTL4) is best known for its role as an adipokine involved in the regulation of lipid and glucose metabolism. The characterization of ANGPTL4 as an adipokine is largely due to our limited understanding of the interaction partners of ANGPTL4 and how ANGPTL4 initiates intracellular signaling. Recent findings have revealed a critical role for ANGPTL4 in cancer growth and progression, anoikis resistance, altered redox regulation, angiogenesis, and metastasis. Emerging evidence suggests that ANGPTL4 function may be drastically altered depending on the proteolytic processing and posttranslational modifications of ANGPTL4, which may clarify several conflicting roles of ANGPTL4 in different cancers. Although the N-terminal coiled-coil region of ANGPTL4 has been largely responsible for the endocrine regulatory role in lipid metabolism, insulin sensitivity, and glucose homeostasis, it has now emerged that the COOH-terminal fibrinogen-like domain of ANGPTL4 may be a key regulator in the multifaceted signaling during cancer development. New insights into the mechanistic action of this functional domain have opened a new chapter into the possible clinical application of ANGPTL4 as a promising candidate for clinical intervention in the fight against cancer. This review summarizes our current understanding of ANGPTL4 in cancer and highlights areas that warrant further investigation. A better understanding of the underlying cellular and molecular mechanisms of ANGPTL4 will reveal novel insights into other aspects of tumorigenesis and the potential therapeutic value of ANGPTL4.


Subject(s)
Angiopoietins/physiology , Neoplasms/metabolism , Neovascularization, Pathologic/physiopathology , Signal Transduction/physiology , Angiopoietin-Like Protein 4 , Angiopoietins/genetics , Angiopoietins/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Lipid Metabolism , Neoplasms/genetics , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Protein Processing, Post-Translational , Signal Transduction/genetics
10.
Blood ; 118(14): 3990-4002, 2011 Oct 06.
Article in English | MEDLINE | ID: mdl-21841165

ABSTRACT

Vascular disruption induced by interactions between tumor-secreted permeability factors and adhesive proteins on endothelial cells facilitates metastasis. The role of tumor-secreted C-terminal fibrinogen-like domain of angiopoietin-like 4 (cANGPTL4) in vascular leakiness and metastasis is controversial because of the lack of understanding of how cANGPTL4 modulates vascular integrity. Here, we show that cANGPTL4 instigated the disruption of endothelial continuity by directly interacting with 3 novel binding partners, integrin α5ß1, VE-cadherin, and claudin-5, in a temporally sequential manner, thus facilitating metastasis. We showed that cANGPTL4 binds and activates integrin α5ß1-mediated Rac1/PAK signaling to weaken cell-cell contacts. cANGPTL4 subsequently associated with and declustered VE-cadherin and claudin-5, leading to endothelial disruption. Interfering with the formation of these cANGPTL4 complexes delayed vascular disruption. In vivo vascular permeability and metastatic assays performed using ANGPTL4-knockout and wild-type mice injected with either control or ANGPTL4-knockdown tumors confirmed that cANGPTL4 induced vascular leakiness and facilitated lung metastasis in mice. Thus, our findings elucidate how cANGPTL4 induces endothelial disruption. Our findings have direct implications for targeting cANGPTL4 to treat cancer and other vascular pathologies.


Subject(s)
Angiopoietins/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Claudins/metabolism , Integrin alpha5beta1/metabolism , Angiopoietin-Like Protein 4 , Angiopoietins/genetics , Animals , Capillary Permeability , Cells, Cultured , Claudin-5 , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Gene Expression Regulation, Neoplastic , Humans , Lung/pathology , Mice , Mice, Nude , Neoplasm Metastasis/pathology , Neoplasms/blood supply , Neoplasms/genetics , Neoplasms/metabolism , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...