1
|
Gu X, Li T, Yin X, Zhai P, Jiang D, Sun D, Yan H, Wang B. Exosomes Derived from Metformin-Pretreated BMSCs Accelerate Diabetic Wound Repair by Promoting Angiogenesis Via the LINC-PINT/miR-139-3p/FOXC2 Axis. Stem Cell Rev Rep 2025; 21:1096-1112. [PMID: 40111729 DOI: 10.1007/s12015-025-10860-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2025] [Indexed: 03/22/2025]
Abstract
Chronic trauma is a prevalent and significant complication of diabetes. Mesenchymal stem cell(MSC)-derived exosomes (Exos) have been reported to accelerate the healing of chronic diabetic wounds. MSCs pretreated with chemical or biological factors were reported to enhance the biological activity of MSC-derived exosomes. Hence, this study investigated the role of exosomes derived from bone marrow mesenchymal stem cells (BMSCs) pretreated with metformin (MET) on diabetic wound healing. The results showed that MET-Exos promoted endothelial cell migration, tube formation, and angiogenesis, leading to accelerated wound healing in diabetic mice. Mechanistically, MET-Exos upregulated LINC-PINT, which, through competitive binding to miR-139-3p, activated FOXC2, a key regulator of angiogenesis. These data reveal that MET-Exos might promote revascularization and wound healing through the LINC-PINT/miR-139-3p/FOXC2 axis, showing its potential as a therapeutic modality for diabetic wounds.
Collapse
Affiliation(s)
- Xiaobao Gu
- Department of Vascular Surgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Teng Li
- Department of Vascular Surgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiangyang Yin
- Department of Vascular Surgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pengbo Zhai
- Department of Vascular Surgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Deyu Jiang
- Department of Vascular Surgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ding Sun
- Department of Vascular Surgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongxu Yan
- Department of Vascular Surgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bing Wang
- Department of Vascular Surgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
2
|
Wang Y, Liu J. Modulation of the Epithelial-mesenchymal transition process by Forkhead Box C2 in the repair of airway epithelium after injury. Respir Res 2025; 26:96. [PMID: 40065336 PMCID: PMC11895206 DOI: 10.1186/s12931-025-03150-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/11/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Epithelial-mesenchymal transition (EMT) is regarded as a key process in repair of airway epithelium after injury. Forkhead Box C2 (FOXC2) is a transcription factor involved in EMT process, whether it is involved in repair of bronchial epithelium remains unknown. METHODS C57BL/6 mice were subjected to intraperitoneal injection with naphthalene (NAPH; 200 mg/kg) to induce airway injury model. qPCR, immunoblot and FOXC2 immunohistochemistry assays were conducted to detect the expression of FOXC2 in bronchial epithelium. To explore the function of FOXC2 in NAPH-induced airway injury, the mice were given intratracheal administration of shFOXC2- or shNC-lentivirus particles, followed by NAPH treatment. Hematoxylin-and-eosin staining was used to assess the histopathology of the bronchial epithelium. Immunofluorescence analysis of CCSP, a club cell marker confirmed the CCSP expression in bronchial epithelium. Immunoblot and immunofluorescence assays determined the expression of E-cadherin, vimentin, and N-cadherin. In mouse primary bronchial epithelial cells (PBECs), we overexpressed and silenced FOXC2 by lentivirus particles, respectively. Cell migration was analyzed using wound healing assay. Immunoblot assays determined the E-cadherin, vimentin, FN-EDA expression in TGF-β1-induced PBECs. mRNA sequencing (mRNA-seq) and FOXC2 ChIP sequencing (ChIP-seq) to reveal the downstream genes of FOXC2 in TGF-β1-induced PBECs. Luciferase assay, ChIP-PCR and functional rescue experiments were performed to confirm the interaction of FOXC2/formin binding protein 1 (FNBP1) in TGF-β1-induced PBECs. RESULTS FOXC2 expression was up-regulated in the lung tissues of mice at 2, 3 and 6 days post-NAPH. FOXC2 knockdown in bronchial epithelium of mice delayed CCSP+ club cell regeneration and normal repair of the airway epithelium within 14 days after injury. Knockdown of FOXC2 increased E-cadherin but decreased vimentin and N-cadherin, EMT markers during early phase after injury. In vitro, knockdown of endogenous FOXC2 repressed the migration of cells and increased TGF-β1-induced E-cadherin but decreased vimentin, N-cadherin and FN-EDA. Exogenous FOXC2 addition exerted opposite effects. Furthermore, mRNA-seq and FOXC2 ChIP-seq revealed that FNBP1 might be a downstream target of FOXC2. Overexpression of FNBP1 reversed the inhibitory role of FOXC2 knockdown in EMT. CONCLUSIONS These data highlight the important function of FOXC2 as a regulator in repair of bronchial epithelium after injury.
Collapse
Affiliation(s)
- Yudong Wang
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Shenyang, People's Republic of China
| | - Jun Liu
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Shenyang, People's Republic of China.
| |
Collapse
|
3
|
Pei S, Zhang D, Li Z, Liu J, Li Z, Chen J, Xie Z. The Role of the Fox Gene in Breast Cancer Progression. Int J Mol Sci 2025; 26:1415. [PMID: 40003882 PMCID: PMC11855465 DOI: 10.3390/ijms26041415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/25/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Forkhead box (FOX) genes are a family of transcription factors that participate in many biological activities, from early embryogenesis to the formation of organs, and from regulation of glucose metabolism to regulation of longevity. Given the extensive influence in the multicellular process, FOX family proteins are responsible for the progression of many types of cancers, especially lung cancer, breast cancer, prostate cancer, and other cancers. Breast cancer is the most common cancer among women, and 2.3 million women were diagnosed in 2020. So, various drugs targeting the FOX signaling pathway have been developed to inhibit breast cancer progression. While the role of the FOX family gene in cancer development has not received enough attention, discovering more potential drugs targeting the FOX signaling pathway is urgently demanded. Here, we review the main members in the FOX gene family and summarize their signaling pathway, including the regulation of the FOX genes and their effects on breast cancer progression. We hope this review will emphasize the understanding of the role of the FOX gene in breast cancer and inspire the discovery of effective anti-breast cancer medicines targeting the FOX gene in the future.
Collapse
Affiliation(s)
- Shaoxuan Pei
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Dechun Zhang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Zhuohan Li
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Jinkai Liu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Ziyi Li
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Jianrui Chen
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Zhenzhen Xie
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
| |
Collapse
|
4
|
Peng Y, Jia L, Hu X, Shi X, Fang X, Qiu Y, Gan Z, Wang Y. Cellular Feimin enhances exercise performance by suppressing muscle thermogenesis. Nat Metab 2025; 7:84-101. [PMID: 39747484 DOI: 10.1038/s42255-024-01176-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 11/05/2024] [Indexed: 01/04/2025]
Abstract
Exercise can rapidly increase core body temperature, and research has indicated that elevated internal body temperature can independently contribute to fatigue during physical activity. However, the precise mechanisms responsible for regulating thermogenesis in muscles during exercise have remained unclear. Here, we demonstrate that cellular Feimin (cFeimin) enhances exercise performance by inhibiting muscle thermogenesis during physical activity. Mechanistically, we found that AMP-activated protein kinase (AMPK) phosphorylates cFeimin and facilitates its translocation into the cell nucleus during exercise. Within the nucleus, cFeimin binds to the forkhead transcription factor FOXC2, leading to the suppressed expression of sarcolipin (Sln), which is a key regulator of muscle thermogenesis. In addition, our results further reveal that short-term AMPK agonist treatments can enhance exercise performance through the activation of the AMPK-cFeimin signalling pathway. In summary, these results underscore the crucial role of cFeimin in enhancing exercise performance by modulating SLN-mediated thermogenesis.
Collapse
Affiliation(s)
- Ying Peng
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Liangjie Jia
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xiao Hu
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xiaoliu Shi
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xinlei Fang
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yifu Qiu
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Zhenji Gan
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical School of Nanjing University, Nanjing, China
| | - Yiguo Wang
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
5
|
Vakili S, Cao K. Angiopoietin-2: A Therapeutic Target for Vascular Protection in Hutchinson-Gilford Progeria Syndrome. Int J Mol Sci 2024; 25:13537. [PMID: 39769300 PMCID: PMC11676795 DOI: 10.3390/ijms252413537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a pediatric condition characterized by clinical features that resemble accelerated aging. The abnormal accumulation of a toxic form of the lamin A protein known as progerin disrupts cellular functions, leading to various complications, including growth retardation, loss of subcutaneous fat, abnormal skin, alopecia, osteoporosis, and progressive joint contractures. Death primarily occurs as the result of complications from progressive atherosclerosis, especially from cardiac disease, such as myocardial infarction or heart failure, or cerebrovascular disease like stroke. Despite the availability of lonafarnib, the only US Food and Drug Administration-approved treatment for HGPS, cardiovascular complications remain the leading cause of morbidity and mortality in affected patients. Defective angiogenesis-the process of forming new blood vessels from existing ones-plays a crucial role in the development of cardiovascular disease. A recent study suggests that Angiopoietin-2 (Ang2), a pro-angiogenic growth factor that regulates angiogenesis and vascular stability, may offer therapeutic potential for the treatment of HGPS. In this review, we describe the clinical features and key cellular processes impacted by progerin and discuss the therapeutic potential of Ang2 in addressing these challenges.
Collapse
Affiliation(s)
| | - Kan Cao
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA;
| |
Collapse
|
6
|
Warmke N, Bridge KI, Ozber CH, Smith J, Platt F, Haywood NJ, Skromna A, Makava N, Yuldasheva NY, Wheatcroft S, Kearney MT, Cubbon RM, Griffin KJ. Insulin receptor signalling in PDGFRβ-expressing cells influences systemic metabolism and negatively impacts lipid storage. Biochem Biophys Res Commun 2024; 735:150799. [PMID: 39406023 DOI: 10.1016/j.bbrc.2024.150799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/19/2024] [Accepted: 10/07/2024] [Indexed: 11/05/2024]
Abstract
Pericytes are vascular mural cells that support the microvasculature; their dysfunction contributes to diabetic retinopathy and has been linked to obesity in humans. To explore the role of pericyte insulin signalling on systemic metabolism we utilised male mice from our previously described PIR-/- (PIRKO) mouse line which has insulin receptor (Insr) knockout in PDGFRβ-expressing cells. These animals exhibit systemic insulin resistance from as early as 8-weeks of age, despite no change in body weight or activity level, and show altered body composition and hepatosteatosis. When challenged with high fat diet, PIR-/- remain insulin resistant but are protected from weight gain with reduced adipose tissue expansion across all depots and altered adipose morphology. Exhibiting parallels with the metabolically-obese-normal-weight (MONW) human phenotype, the PIR-/- line underlines the importance of pericyte biology in the development of both diabetes and obesity and establishes the angiopoietin (Ang)/Tie signalling pathway as a focus for future research.
Collapse
Affiliation(s)
- Nele Warmke
- Leeds Institute of Cardiovascular & Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Clarendon Way, Leeds, LS2 9JT, UK
| | - Katherine I Bridge
- Leeds Institute of Cardiovascular & Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Clarendon Way, Leeds, LS2 9JT, UK
| | - Claire H Ozber
- Leeds Institute of Cardiovascular & Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Clarendon Way, Leeds, LS2 9JT, UK; Leeds Institute of Medical Research at St James' Hospital, Faculty of Medicine and Health, University of Leeds, Beckett Street, LS9 7TF, UK
| | - Jessica Smith
- Leeds Institute of Cardiovascular & Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Clarendon Way, Leeds, LS2 9JT, UK
| | - Fiona Platt
- Leeds Institute of Cardiovascular & Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Clarendon Way, Leeds, LS2 9JT, UK
| | - Natalie J Haywood
- Leeds Institute of Cardiovascular & Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Clarendon Way, Leeds, LS2 9JT, UK
| | - Anna Skromna
- Leeds Institute of Cardiovascular & Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Clarendon Way, Leeds, LS2 9JT, UK
| | - Natallia Makava
- Leeds Institute of Cardiovascular & Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Clarendon Way, Leeds, LS2 9JT, UK
| | - Nadira Y Yuldasheva
- Leeds Institute of Cardiovascular & Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Clarendon Way, Leeds, LS2 9JT, UK
| | - Stephen Wheatcroft
- Leeds Institute of Cardiovascular & Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Clarendon Way, Leeds, LS2 9JT, UK
| | - Mark T Kearney
- Leeds Institute of Cardiovascular & Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Clarendon Way, Leeds, LS2 9JT, UK
| | - Richard M Cubbon
- Leeds Institute of Cardiovascular & Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Clarendon Way, Leeds, LS2 9JT, UK
| | - Kathryn J Griffin
- Leeds Institute of Cardiovascular & Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Clarendon Way, Leeds, LS2 9JT, UK.
| |
Collapse
|
7
|
Hosaka K, Andersson P, Wu J, He X, Du Q, Jing X, Seki T, Gao J, Zhang Y, Sun X, Huang P, Yang Y, Ge M, Cao Y. KRAS mutation-driven angiopoietin 2 bestows anti-VEGF resistance in epithelial carcinomas. Proc Natl Acad Sci U S A 2023; 120:e2303740120. [PMID: 37428914 PMCID: PMC10629547 DOI: 10.1073/pnas.2303740120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/17/2023] [Indexed: 07/12/2023] Open
Abstract
Defining reliable surrogate markers and overcoming drug resistance are the most challenging issues for improving therapeutic outcomes of antiangiogenic drugs (AADs) in cancer patients. At the time of this writing, no biomarkers are clinically available to predict AAD therapeutic benefits and drug resistance. Here, we uncovered a unique mechanism of AAD resistance in epithelial carcinomas with KRAS mutations that targeted angiopoietin 2 (ANG2) to circumvent antivascular endothelial growth factor (anti-VEGF) responses. Mechanistically, KRAS mutations up-regulated the FOXC2 transcription factor that directly elevated ANG2 expression at the transcriptional level. ANG2 bestowed anti-VEGF resistance as an alternative pathway to augment VEGF-independent tumor angiogenesis. Most colorectal and pancreatic cancers with KRAS mutations were intrinsically resistant to monotherapies of anti-VEGF or anti-ANG2 drugs. However, combination therapy with anti-VEGF and anti-ANG2 drugs produced synergistic and potent anticancer effects in KRAS-mutated cancers. Together, these data demonstrate that KRAS mutations in tumors serve as a predictive marker for anti-VEGF resistance and are susceptible to combination therapy with anti-VEGF and anti-ANG2 drugs.
Collapse
Affiliation(s)
- Kayoko Hosaka
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm171 65, Sweden
| | - Patrik Andersson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm171 65, Sweden
| | - Jieyu Wu
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm171 65, Sweden
| | - Xingkang He
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm171 65, Sweden
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310016, China
| | - Qiqiao Du
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm171 65, Sweden
| | - Xu Jing
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm171 65, Sweden
| | - Takahiro Seki
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm171 65, Sweden
| | - Juan Gao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm171 65, Sweden
| | - Yin Zhang
- School of Pharmacology, Binzhou Medical University, Yantai, Shandong264003, China
| | - Xiaoting Sun
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm171 65, Sweden
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vison and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou325024, China
| | - Ping Huang
- Department of Pharmacy, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou310053, China
| | - Yunlong Yang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai200032, China
| | - Minghua Ge
- Department of Head, Neck and Thyroid Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou31003, China
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm171 65, Sweden
| |
Collapse
|
8
|
Vliora M, Ravelli C, Grillo E, Corsini M, Flouris AD, Mitola S. The impact of adipokines on vascular networks in adipose tissue. Cytokine Growth Factor Rev 2023; 69:61-72. [PMID: 35953434 DOI: 10.1016/j.cytogfr.2022.07.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 02/07/2023]
Abstract
Adipose tissue (AT) is a highly active and plastic endocrine organ. It secretes numerous soluble molecules known as adipokines, which act locally to AT control the remodel and homeostasis or exert pleiotropic functions in different peripheral organs. Aberrant production or loss of certain adipokines contributes to AT dysfunction associated with metabolic disorders, including obesity. The AT plasticity is strictly related to tissue vascularization. Angiogenesis supports the AT expansion, while regression of blood vessels is associated with AT hypoxia, which in turn mediates tissue inflammation, fibrosis and metabolic dysfunction. Several adipokines can regulate endothelial cell functions and are endowed with either pro- or anti-angiogenic properties. Here we address the role of adipokines in the regulation of angiogenesis. A better understanding of the link between adipokines and angiogenesis will open the way for novel therapeutic approaches to treat obesity and metabolic diseases.
Collapse
Affiliation(s)
- Maria Vliora
- FAME Laboratory, Department of Exercise Science, University of Thessaly, Trikala, Greece; Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy
| | - Cosetta Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy
| | - Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy
| | - Michela Corsini
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy
| | - Andreas D Flouris
- FAME Laboratory, Department of Exercise Science, University of Thessaly, Trikala, Greece
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy.
| |
Collapse
|
9
|
Yimingjiang M, Aini A, Tuergan T, Zhang W. Differential Gene Expression Profiling in Alveolar Echinococcosis Identifies Potential Biomarkers Associated With Angiogenesis. Open Forum Infect Dis 2023; 10:ofad031. [PMID: 36817746 PMCID: PMC9927572 DOI: 10.1093/ofid/ofad031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
Background Alveolar echinococcosis (AE) is a worldwide zoonosis caused by Echinococcus multilocularis. Alveolar echinococcosis is a severe chronic parasitic disease that exhibits a tumor-like growth, with the potential for invasion and distant metastasis; however, the molecular mechanism underlying this condition remains unclear. Methods Transcriptome analyses were performed to detect differentially expressed genes (DEGs) in samples from patients with AE with invasion and distant metastasis. The results were further verified by immunohistochemistry. Results A total of 1796 DEGs were identified, including 1742 upregulated and 54 downregulated DEGs. A subsequent functional analysis showed that the significant DEGs were involved in the angiogenesis process. Immunohistochemical analysis confirmed the reliability of the transcriptomic data. Conclusions These results suggest that angiogenesis is a possible mechanism underlying the tumor-like biological behavior observed during E multilocularis infection. Genes related to this process may play important roles in AE invasion and distant metastasis.
Collapse
Affiliation(s)
- Maiweilidan Yimingjiang
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Abudusalamu Aini
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Talaiti Tuergan
- Department of Hepatic Hydatid and Hepatobiliary Surgery, Digestive and Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Wei Zhang
- Correspondence: Dr. Wei Zhang, Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan Southern Road, Urumqi, Xinjiang 830054, China ( )
| |
Collapse
|
10
|
Hargadon KM, Goodloe TB, Lloyd ND. Oncogenic functions of the FOXC2 transcription factor: a hallmarks of cancer perspective. Cancer Metastasis Rev 2022; 41:833-852. [PMID: 35701636 DOI: 10.1007/s10555-022-10045-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/06/2022] [Indexed: 01/25/2023]
Abstract
Epigenetic regulation of gene expression is a fundamental determinant of molecular and cellular function, and epigenetic reprogramming in the context of cancer has emerged as one of the key enabling characteristics associated with acquisition of the core hallmarks of this disease. As such, there has been renewed interest in studying the role of transcription factors as epigenetic regulators of gene expression in cancer. In this review, we discuss the current state of knowledge surrounding the oncogenic functions of FOXC2, a transcription factor that frequently becomes dysregulated in a variety of cancer types. In addition to highlighting the clinical impact of aberrant FOXC2 activity in cancer, we discuss mechanisms by which this transcription factor becomes dysregulated in both tumor and tumor-associated cells, placing particular emphasis on the ways in which FOXC2 promotes key hallmarks of cancer progression. Finally, we bring attention to important issues related to the oncogenic dysregulation of FOXC2 that must be addressed going forward in order to improve our understanding of FOXC2-mediated cancer progression and to guide prognostic and therapeutic applications of this knowledge in clinical settings.
Collapse
Affiliation(s)
- Kristian M Hargadon
- Hargadon Laboratory, Department of Biology, Hampden-Sydney College, Hampden-Sydney, VA, 23943, USA.
| | - Travis B Goodloe
- Hargadon Laboratory, Department of Biology, Hampden-Sydney College, Hampden-Sydney, VA, 23943, USA
| | - Nathaniel D Lloyd
- Hargadon Laboratory, Department of Biology, Hampden-Sydney College, Hampden-Sydney, VA, 23943, USA
| |
Collapse
|
11
|
Recouvreux MS, Miao J, Gozo MC, Wu J, Walts AE, Karlan BY, Orsulic S. FOXC2 Promotes Vasculogenic Mimicry in Ovarian Cancer. Cancers (Basel) 2022; 14:4851. [PMID: 36230774 PMCID: PMC9564305 DOI: 10.3390/cancers14194851] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
FOXC2 is a forkhead family transcription factor that plays a critical role in specifying mesenchymal cell fate during embryogenesis. FOXC2 expression is associated with increased metastasis and poor survival in various solid malignancies. Using in vitro and in vivo assays in mouse ovarian cancer cell lines, we confirmed the previously reported mechanisms by which FOXC2 could promote cancer growth, metastasis, and drug resistance, including epithelial-mesenchymal transition, stem cell-like differentiation, and resistance to anoikis. In addition, we showed that FOXC2 expression is associated with vasculogenic mimicry in mouse and human ovarian cancers. FOXC2 overexpression increased the ability of human ovarian cancer cells to form vascular-like structures in vitro, while inhibition of FOXC2 had the opposite effect. Thus, we present a novel mechanism by which FOXC2 might contribute to cancer aggressiveness and poor patient survival.
Collapse
Affiliation(s)
- Maria Sol Recouvreux
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Jiangyong Miao
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Maricel C. Gozo
- Women’s Cancer Program, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jingni Wu
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Ann E. Walts
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Beth Y. Karlan
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Sandra Orsulic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
- Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90095, USA
| |
Collapse
|
12
|
Abstract
While most tissues exhibit their greatest growth during development, adipose tissue is capable of additional massive expansion in adults. Adipose tissue expandability is advantageous when temporarily storing fuel for use during fasting, but becomes pathological upon continuous food intake, leading to obesity and its many comorbidities. The dense vasculature of adipose tissue provides necessary oxygen and nutrients, and supports delivery of fuel to and from adipocytes under fed or fasting conditions. Moreover, the vasculature of adipose tissue comprises a major niche for multipotent progenitor cells, which give rise to new adipocytes and are necessary for tissue repair. Given the multiple, pivotal roles of the adipose tissue vasculature, impairments in angiogenic capacity may underlie obesity-associated diseases such as diabetes and cardiometabolic disease. Exciting new studies on the single-cell and single-nuclei composition of adipose tissues in mouse and humans are providing new insights into mechanisms of adipose tissue angiogenesis. Moreover, new modes of intercellular communication involving micro vesicle and exosome transfer of proteins, nucleic acids and organelles are also being recognized to play key roles. This review focuses on new insights on the cellular and signaling mechanisms underlying adipose tissue angiogenesis, and on their impact on obesity and its pathophysiological consequences.
Collapse
|
13
|
Role of Distinct Fat Depots in Metabolic Regulation and Pathological Implications. Rev Physiol Biochem Pharmacol 2022; 186:135-176. [PMID: 35915363 DOI: 10.1007/112_2022_73] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
People suffering from obesity and associated metabolic disorders including diabetes are increasing exponentially around the world. Adipose tissue (AT) distribution and alteration in their biochemical properties play a major role in the pathogenesis of these diseases. Emerging evidence suggests that AT heterogeneity and depot-specific physiological changes are vital in the development of insulin resistance in peripheral tissues like muscle and liver. Classically, AT depots are classified into white adipose tissue (WAT) and brown adipose tissue (BAT); WAT is the site of fatty acid storage, while BAT is a dedicated organ of metabolic heat production. The discovery of beige adipocyte clusters in WAT depots indicates AT heterogeneity has a more central role than hither to ascribed. Therefore, we have discussed in detail the current state of understanding on cellular and molecular origin of different AT depots and their relevance toward physiological metabolic homeostasis. A major focus is to highlight the correlation between altered WAT distribution in the body and metabolic pathogenesis in animal models and humans. We have also underscored the disparity in the molecular (including signaling) changes in various WAT tissues during diabetic pathogenesis. Exercise-mediated beneficial alteration in WAT physiology/distribution that protects against metabolic disorders is evolving. Here we have discussed the depot-specific biochemical adjustments induced by different forms of exercise. A detailed understanding of the molecular details of inter-organ crosstalk via substrate utilization/storage and signaling through chemokines provide strategies to target selected WAT depots to pharmacologically mimic the benefits of exercise countering metabolic diseases including diabetes.
Collapse
|
14
|
Xia LZ, Tao J, Chen YJ, Liang LL, Luo GF, Cai ZM, Wang Z. Factors Affecting the Re-Endothelialization of Endothelial Progenitor Cell. DNA Cell Biol 2021; 40:1009-1025. [PMID: 34061680 DOI: 10.1089/dna.2021.0082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The vascular endothelium, which plays an essential role in maintaining the normal shape and function of blood vessels, is a natural barrier between the circulating blood and the vascular wall tissue. The endothelial damage can cause vascular lesions, such as atherosclerosis and restenosis. After the vascular intima injury, the body starts the endothelial repair (re-endothelialization) to inhibit the neointimal hyperplasia. Endothelial progenitor cell is the precursor of endothelial cells and plays an important role in the vascular re-endothelialization. However, re-endothelialization is inevitably affected in vivo and in vitro by factors, which can be divided into two types, namely, promotion and inhibition, and act on different links of the vascular re-endothelialization. This article reviews these factors and related mechanisms.
Collapse
Affiliation(s)
- Lin-Zhen Xia
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Jun Tao
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Yan-Jun Chen
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Ling-Li Liang
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Gui-Fang Luo
- Department of Gynaecology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Ze-Min Cai
- Pediatrics Department, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Zuo Wang
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
15
|
Sphyris N, King C, Hoar J, Werden SJ, Vijay GV, Miura N, Gaharwar A, Sarkar TR. Carcinoma cells that have undergone an epithelial-mesenchymal transition differentiate into endothelial cells and contribute to tumor growth. Oncotarget 2021; 12:823-844. [PMID: 33889304 PMCID: PMC8057273 DOI: 10.18632/oncotarget.27940] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/25/2021] [Indexed: 12/23/2022] Open
Abstract
Hypoxia stimulates neoangiogenesis, promoting tumor outgrowth, and triggers the epithelial-mesenchymal transition (EMT), which bestows cells with mesenchymal traits and multi-lineage differentiation potential. Here, we investigated whether EMT can confer endothelial attributes upon carcinoma cells, augmenting tumor growth and vascularization. Following orthotopic implantation of MCF-7 human epithelial breast cancer cells into mice, tumors of different sizes were immunostained for markers of hypoxia and EMT. Larger tumors were well-vascularized with CD31-positive cells of human origin. Hypoxic regions, demarcated by HIF-1α staining, exhibited focal areas of E-cadherin loss and elevated levels of vimentin and the EMT-mediator FOXC2. Implantation of MCF-7 cells, co-mixed with human mammary epithelial (HMLE) cells overexpressing the EMT-inducer Snail, markedly potentiated tumor growth and vascularization, compared with MCF-7 cells injected alone or co-mixed with HMLE-vector cells. Intra-tumoral vessels contained CD31-positive cells derived from either donor cell type. FOXC2 knockdown abrogated the potentiating effects of HMLE-Snail cells on MCF-7 tumor growth and vascularization, and compromised endothelial transdifferentiation of mesenchymal cells cultured in endothelial growth medium. Hence, cells that have undergone EMT can promote tumor growth and neovascularization either indirectly, by promoting endothelial transdifferentiation of carcinoma cells, or directly, by acquiring an endothelial phenotype, with FOXC2 playing key roles in these processes.
Collapse
Affiliation(s)
- Nathalie Sphyris
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Present address: Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK.,These authors contributed equally to this work
| | - Cody King
- Department of Biochemistry, Texas A&M University, College Station, TX, USA
| | - Jonathan Hoar
- Department of Biology, Texas A&M University, College Station, TX, USA
| | - Steven J Werden
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Geraldine V Vijay
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naoyuki Miura
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Akhilesh Gaharwar
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Tapasree Roy Sarkar
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Biology, Texas A&M University, College Station, TX, USA.,These authors contributed equally to this work
| |
Collapse
|
16
|
Hargadon KM, Győrffy B, Strong EW. The prognostic significance of FOXC2 gene expression in cancer: A comprehensive analysis of RNA-seq data from the cancer genome atlas. Cancer Genet 2021; 254-255:58-64. [PMID: 33636524 DOI: 10.1016/j.cancergen.2021.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 02/03/2021] [Accepted: 02/08/2021] [Indexed: 11/29/2022]
Abstract
The FOXC2 transcription factor is a key regulator of tumor progression in many cancer types. Known to exhibit an array of oncogenic functions when dysregulated, FOXC2 has emerged as a useful biomarker for predicting disease aggression and patient outcome. In this regard, increased expression and nuclear localization of FOXC2 protein in tumor tissue have become well-established as poor prognostic factors for many cancer types. However, whether FOXC2 gene expression can serve as a similarly useful RNA-level biomarker has remained largely unexplored. Therefore, we conducted a comprehensive analysis of TCGA RNA-seq data to evaluate whether FOXC2 gene expression levels in primary tumor biopsies correlate with patient outcome. We report herein that increased expression of FOXC2 RNA in tumor tissue is a poor prognostic factor for patient survival in many cancer types. Moreover, we also found that FOXC2 gene expression predicts cancer patient response to several commonly prescribed chemotherapeutics. Together, these data highlight FOXC2 RNA expression in tumor tissue as an important biomarker with prognostic significance for solid tumors of diverse origin.
Collapse
Affiliation(s)
- Kristian M Hargadon
- Hargadon Laboratory, Department of Biology, Hampden-Sydney College, Brown Student Center, Box 837, Hampden-Sydney, VA 23943, USA.
| | - Balázs Győrffy
- TTK Cancer Biomarker Research Group, Magyar Tudósok körútja 2., H-1117 Budapest, Hungary; Department of Bioinformatics and 2nd Department of Pediatrics, Semmelweis University, Tuzolto u. 7-9, H-1094 Budapest, Hungary
| | - Elijah W Strong
- Hargadon Laboratory, Department of Biology, Hampden-Sydney College, Brown Student Center, Box 837, Hampden-Sydney, VA 23943, USA
| |
Collapse
|
17
|
Rodriguez-Ayala E, Gallegos-Cabrales EC, Gonzalez-Lopez L, Laviada-Molina HA, Salinas-Osornio RA, Nava-Gonzalez EJ, Leal-Berumen I, Escudero-Lourdes C, Escalante-Araiza F, Buenfil-Rello FA, Peschard VG, Laviada-Nagel A, Silva E, Veloz-Garza RA, Martinez-Hernandez A, Barajas-Olmos FM, Molina-Segui F, Gonzalez-Ramirez L, Espadas-Olivera R, Lopez-Muñoz R, Arjona-Villicaña RD, Hernandez-Escalante VM, Rodriguez-Arellano ME, Gaytan-Saucedo JF, Vaquera Z, Acebo-Martinez M, Cornejo-Barrera J, Jancy Andrea HQ, Castillo-Pineda JC, Murillo-Ramirez A, Diaz-Tena SP, Figueroa-Nuñez B, Valencia-Rendon ME, Garzon-Zamora R, Viveros-Paredes JM, Ángeles-Chimal J, Santa-Olalla Tapia J, Remes-Troche JM, Valdovinos-Chavez SB, Huerta-Avila EE, Lopez-Alvarenga JC, Comuzzie AG, Haack K, Han X, Orozco L, Weintraub S, Kent JW, Cole SA, Bastarrachea RA. Towards precision medicine: defining and characterizing adipose tissue dysfunction to identify early immunometabolic risk in symptom-free adults from the GEMM family study. Adipocyte 2020; 9:153-169. [PMID: 32272872 PMCID: PMC7153654 DOI: 10.1080/21623945.2020.1743116] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/03/2020] [Accepted: 03/10/2020] [Indexed: 12/15/2022] Open
Abstract
Interactions between macrophages and adipocytes are early molecular factors influencing adipose tissue (AT) dysfunction, resulting in high leptin, low adiponectin circulating levels and low-grade metaflammation, leading to insulin resistance (IR) with increased cardiovascular risk. We report the characterization of AT dysfunction through measurements of the adiponectin/leptin ratio (ALR), the adipo-insulin resistance index (Adipo-IRi), fasting/postprandial (F/P) immunometabolic phenotyping and direct F/P differential gene expression in AT biopsies obtained from symptom-free adults from the GEMM family study. AT dysfunction was evaluated through associations of the ALR with F/P insulin-glucose axis, lipid-lipoprotein metabolism, and inflammatory markers. A relevant pattern of negative associations between decreased ALR and markers of systemic low-grade metaflammation, HOMA, and postprandial cardiovascular risk hyperinsulinemic, triglyceride and GLP-1 curves was found. We also analysed their plasma non-coding microRNAs and shotgun lipidomics profiles finding trends that may reflect a pattern of adipose tissue dysfunction in the fed and fasted state. Direct gene differential expression data showed initial patterns of AT molecular signatures of key immunometabolic genes involved in AT expansion, angiogenic remodelling and immune cell migration. These data reinforce the central, early role of AT dysfunction at the molecular and systemic level in the pathogenesis of IR and immunometabolic disorders.
Collapse
Affiliation(s)
- Ernesto Rodriguez-Ayala
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Norte, México City, México
| | | | - Laura Gonzalez-Lopez
- Dirección de Postgrado e Investigación, Universidad del Valle de Atemajac (UNIVA), Zapopan, México
| | | | - Rocio A. Salinas-Osornio
- Dirección de Postgrado e Investigación, Universidad del Valle de Atemajac (UNIVA), Zapopan, México
| | | | - Irene Leal-Berumen
- Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua, México
| | | | - Fabiola Escalante-Araiza
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Norte, México City, México
| | - Fatima A. Buenfil-Rello
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | - Vanessa-Giselle Peschard
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Norte, México City, México
| | - Antonio Laviada-Nagel
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | - Eliud Silva
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Norte, México City, México
| | - Rosa A. Veloz-Garza
- Facultad de Enfermería, Universidad Autónoma de Nuevo León (UANL), Monterrey, México
| | - Angelica Martinez-Hernandez
- Laboratorio de Inmunogenómica y Enfermedades Metabólicas, Instituto Nacional de Medicina Genómica, México City, México
| | - Francisco M. Barajas-Olmos
- Laboratorio de Inmunogenómica y Enfermedades Metabólicas, Instituto Nacional de Medicina Genómica, México City, México
| | | | | | | | - Ricardo Lopez-Muñoz
- Escuela de Ciencias de la Salud, Universidad Marista de Mérida, Yucatán, Mexico
| | | | - Victor M. Hernandez-Escalante
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | | | - Janeth F. Gaytan-Saucedo
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | - Zoila Vaquera
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | | | - Judith Cornejo-Barrera
- Departamento de Enseñanza, Postgrado e Investigación, Hospital Infantil de Tamaulipas, Ciudad, México
| | - Huertas-Quintero Jancy Andrea
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | | | | | - Sara P. Diaz-Tena
- Departamento de Nutrición Humana, Universidad Latina de América, Morelia, México
| | | | | | - Rafael Garzon-Zamora
- Dirección de Postgrado e Investigación, Universidad del Valle de Atemajac (UNIVA), Zapopan, México
| | | | - José Ángeles-Chimal
- Facultad de Medicina, Universidad Autónoma Estado de Morelos, Cuernavaca, México
| | | | - José M. Remes-Troche
- Instituto de Investigaciones Médico-Biológicas, Universidad Veracruzana, Veracruz, México
| | | | - Eira E. Huerta-Avila
- Laboratorio de Inmunogenómica y Enfermedades Metabólicas, Instituto Nacional de Medicina Genómica, México City, México
| | - Juan Carlos Lopez-Alvarenga
- School of Medicine & South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | | | - Karin Haack
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | - Xianlin Han
- Department of Medicine, Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Lorena Orozco
- Laboratorio de Inmunogenómica y Enfermedades Metabólicas, Instituto Nacional de Medicina Genómica, México City, México
| | - Susan Weintraub
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX, USA
| | - Jack W. Kent
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | - Shelley A. Cole
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | - Raul A. Bastarrachea
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| |
Collapse
|
18
|
Pang C, Lim CS, Brookes J, Tsui J, Hamilton G. Emerging importance of molecular pathogenesis of vascular malformations in clinical practice and classifications. Vasc Med 2020; 25:364-377. [PMID: 32568624 DOI: 10.1177/1358863x20918941] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Vascular malformations occur during early vascular development resulting in abnormally formed vessels that can manifest as arterial, venous, capillary or lymphatic lesions, or in combination, and include local tissue overdevelopment. Vascular malformations are largely caused by sporadic somatic gene mutations. This article aims to review and discuss current molecular signaling pathways and therapeutic targets for vascular malformations and to classify vascular malformations according to the molecular pathways involved. A literature review was performed using Embase and Medline. Different MeSH terms were combined for the search strategy, with the aim of encompassing all studies describing the classification, pathogenesis, and treatment of vascular malformations. Major pathways involved in the pathogenesis of vascular malformations are vascular endothelial growth factor (VEGF), Ras/Raf/MEK/ERK, angiopoietin-TIE2, transforming growth factor beta (TGF-β), and PI3K/AKT/mTOR. These pathways are involved in controlling cellular growth, apoptosis, differentiation, and proliferation, and play a central role in endothelial cell signaling and angiogenesis. Many vascular malformations share similar aberrant molecular signaling pathways with cancers and inflammatory disorders. Therefore, selective anticancer agents and immunosuppressants may be beneficial in treating vascular malformations of specific mutations. The current classification systems of vascular malformations, including the International Society of the Study of Vascular Anomalies (ISSVA) classification, are primarily observational and clinical, and are not based on the molecular pathways involved in the pathogenesis of the condition. Several molecular pathways with potential therapeutic targets have been demonstrated to contribute to the development of various vascular anomalies. Classifying vascular malformations based on their molecular pathogenesis may improve treatment by determining the underlying nature of the condition and their potential therapeutic target.
Collapse
Affiliation(s)
- Calver Pang
- Department of Vascular Surgery, Royal Free London NHS Foundation Trust, London, United Kingdom.,Department of Surgical Biotechnology, Division of Surgery & Interventional Science, Faculty of Medical Sciences, University College London, United Kingdom
| | - Chung Sim Lim
- Department of Vascular Surgery, Royal Free London NHS Foundation Trust, London, United Kingdom.,Department of Surgical Biotechnology, Division of Surgery & Interventional Science, Faculty of Medical Sciences, University College London, United Kingdom.,NIHR, University College London Hospitals Biomedical Research Centre, London, United Kingdom
| | - Jocelyn Brookes
- Department of Vascular Surgery, Royal Free London NHS Foundation Trust, London, United Kingdom.,Department of Interventional Radiology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Janice Tsui
- Department of Vascular Surgery, Royal Free London NHS Foundation Trust, London, United Kingdom.,Department of Surgical Biotechnology, Division of Surgery & Interventional Science, Faculty of Medical Sciences, University College London, United Kingdom.,NIHR, University College London Hospitals Biomedical Research Centre, London, United Kingdom
| | - George Hamilton
- Department of Vascular Surgery, Royal Free London NHS Foundation Trust, London, United Kingdom.,Department of Surgical Biotechnology, Division of Surgery & Interventional Science, Faculty of Medical Sciences, University College London, United Kingdom
| |
Collapse
|
19
|
Angiopoietin-2-integrin α5β1 signaling enhances vascular fatty acid transport and prevents ectopic lipid-induced insulin resistance. Nat Commun 2020; 11:2980. [PMID: 32532986 PMCID: PMC7293240 DOI: 10.1038/s41467-020-16795-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 05/22/2020] [Indexed: 02/07/2023] Open
Abstract
Proper storage of excessive dietary fat into subcutaneous adipose tissue (SAT) prevents ectopic lipid deposition-induced insulin resistance, yet the underlying mechanism remains unclear. Here, we identify angiopoietin-2 (Angpt2)–integrin α5β1 signaling as an inducer of fat uptake specifically in SAT. Adipocyte-specific deletion of Angpt2 markedly reduced fatty acid uptake and storage in SAT, leading to ectopic lipid accumulation in glucose-consuming organs including skeletal muscle and liver and to systemic insulin resistance. Mechanistically, Angpt2 activated integrin α5β1 signaling in the endothelium and triggered fatty acid transport via CD36 and FATP3 into SAT. Genetic or pharmacological inhibition of the endothelial integrin α5β1 recapitulated adipocyte-specific Angpt2 knockout phenotypes. Our findings demonstrate the critical roles of Angpt2–integrin α5β1 signaling in SAT endothelium in regulating whole-body fat distribution for metabolic health and highlight adipocyte–endothelial crosstalk as a potential target for prevention of ectopic lipid deposition-induced lipotoxicity and insulin resistance. Fat uptake and storage in subcutaneous adipose tissue (SAT) prevents ectopic fat accumulation and associated metabolic complications, however, the underlying mechanisms are incompletely understood. Here, the authors show that adipose angiopoietin-2 (Angpt2) enhances SAT size via increased endothelial fatty acid transport.
Collapse
|
20
|
Yu X, Ye F. Role of Angiopoietins in Development of Cancer and Neoplasia Associated with Viral Infection. Cells 2020; 9:cells9020457. [PMID: 32085414 PMCID: PMC7072744 DOI: 10.3390/cells9020457] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/09/2020] [Accepted: 02/13/2020] [Indexed: 02/06/2023] Open
Abstract
Angiopoietin/tyrosine protein kinase receptor Tie-2 signaling in endothelial cells plays an essential role in angiogenesis and wound healing. Angiopoietin-1 (Ang-1) is crucial for blood vessel maturation while angiopoietin-2 (Ang-2), in collaboration with vascular endothelial growth factor (VEGF), initiates angiogenesis by destabilizing existing blood vessels. In healthy people, the Ang-1 level is sustained while Ang-2 expression is restricted. In cancer patients, Ang-2 level is elevated, which correlates with poor prognosis. Ang-2 not only drives tumor angiogenesis but also attracts infiltration of myeloid cells. The latter rapidly differentiate into tumor stromal cells that foster tumor angiogenesis and progression, and weaken the host’s anti-tumor immunity. Moreover, through integrin signaling, Ang-2 induces expression of matrix metallopeptidases (MMPs) to promote tumor cell invasion and metastasis. Many oncogenic viruses induce expression of Ang-2 to promote development of neoplasia associated with viral infection. Multiple Ang-2 inhibitors exhibit remarkable anti-tumor activities, further highlighting the importance of Ang-2 in cancer development.
Collapse
Affiliation(s)
- Xiaolan Yu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, China
- Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, China
- Correspondence: (X.Y.); (F.Y.); Tel.: +086-27-88661237 (X.Y.); +216-368-8892 (F.Y.)
| | - Fengchun Ye
- Department of Molecular Biology & Microbiology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
- Correspondence: (X.Y.); (F.Y.); Tel.: +086-27-88661237 (X.Y.); +216-368-8892 (F.Y.)
| |
Collapse
|
21
|
Ji C, Yue S, Gu J, Kong Y, Chen H, Yu C, Sun Z, Zhao M. 2,7-Dibromocarbazole interferes with tube formation in HUVECs by altering Ang2 promoter DNA methylation status. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 697:134156. [PMID: 32380619 DOI: 10.1016/j.scitotenv.2019.134156] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/24/2019] [Accepted: 08/27/2019] [Indexed: 06/11/2023]
Abstract
2,7-Dibromocarbazole (2,7-DBCZ) is one of the most frequently detected polyhalogenated carbazoles (PHCZs) in the environmental media. 2,7-DBCZ has attracted public attention for its potential for dioxin-like toxicity and cardiovascular toxicity. However, researches on the potential mechanism of angiogenesis inhibition by 2,7-DBCZ is still insufficient. Herein, human umbilical vein endothelial cells (HUVECs) were applied to explore the angiogenic effect of 2,7-DBCZ and the potential underlying mechanisms. 2,7-DBCZ significantly inhibited tube formation in HUVECs in the non-toxic concentration range. PCR array showed that 2,7-DBCZ reduced the expression proportion between VEGFs and Ang2, thereby inhibiting tube formation in HUVECs. Then, small RNA interference and DNA methylation assays were adopted to explore the potential mechanisms. It has been found that angiopoietin2 (Ang2)-silencing recovered the tube formation inhibited by 2,7-DBCZ. The DNA methylation status of Ang2 promoter also showed a demethylation tendency after exposure. In conclusion, 2,7-DBCZ could demethylate the Ang2 promoter to potentiate Ang2 expression, thus altering angiogenic phenotype of HUVECs by reducing the proportion between Ang2 and VEGFs. The data presented here can help to guide safety measures on the use of dioxin-like PHCZs for their potential adverse effects and provide a method for identifying the relevant biomarkers to assess their cardiovascular toxicity.
Collapse
Affiliation(s)
- Chenyang Ji
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, China
| | - Siqing Yue
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, China
| | - Jinping Gu
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yuan Kong
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, China
| | - Haofeng Chen
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, China
| | - Chang Yu
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, China
| | - Zhe Sun
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | - Meirong Zhao
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, China.
| |
Collapse
|
22
|
Jiang X, Tian W, Nicolls MR, Rockson SG. The Lymphatic System in Obesity, Insulin Resistance, and Cardiovascular Diseases. Front Physiol 2019; 10:1402. [PMID: 31798464 PMCID: PMC6868002 DOI: 10.3389/fphys.2019.01402] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 10/31/2019] [Indexed: 12/22/2022] Open
Abstract
Obesity, insulin resistance, dyslipidemia, and hypertension are fundamental clinical manifestations of the metabolic syndrome. Studies over the last few decades have implicated chronic inflammation and microvascular remodeling in the development of obesity and insulin resistance. Newer observations, however, suggest that dysregulation of the lymphatic system underlies the development of the metabolic syndrome. This review summarizes recent advances in the field, discussing how lymphatic abnormality promotes obesity and insulin resistance, and, conversely, how the metabolic syndrome impairs lymphatic function. We also discuss lymphatic biology in metabolically dysregulated diseases, including type 2 diabetes, atherosclerosis, and myocardial infarction.
Collapse
Affiliation(s)
- Xinguo Jiang
- VA Palo Alto Health Care System, Palo Alto, CA, United States.,Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Wen Tian
- VA Palo Alto Health Care System, Palo Alto, CA, United States.,Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Mark R Nicolls
- VA Palo Alto Health Care System, Palo Alto, CA, United States.,Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Stanley G Rockson
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
23
|
Barchetta I, Cimini FA, Ciccarelli G, Baroni MG, Cavallo MG. Sick fat: the good and the bad of old and new circulating markers of adipose tissue inflammation. J Endocrinol Invest 2019; 42:1257-1272. [PMID: 31073969 DOI: 10.1007/s40618-019-01052-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 04/29/2019] [Indexed: 01/08/2023]
Abstract
Adipose tissue (AT) is one of the largest endocrine organs contributing to metabolic homeostasis. The functional pleiotropism of AT depends on its ability to secrete a large number of hormones, cytokines, extracellular matrix proteins and growth factors, all influencing many local and systemic physiological and pathophysiological processes. In condition of chronic positive energy balance, adipocyte expansion, hypoxia, apoptosis and stress all lead to AT inflammation and dysfunction, and it has been demonstrated that this sick fat is a main risk factor for many metabolic disorders, such as type 2 diabetes mellitus, fatty liver, cardiovascular disease and cancer. AT dysfunction is tightly associated with aberrant secretion of bioactive peptides, the adipocytokines, and their blood concentrations often reflect the expression in the AT. Despite the existence of an association between AT dysfunction and systemic pro-inflammatory state, most of the circulating molecules detectable in obese and dysmetabolic individuals do not identify specifically the condition of sick fat. Based on this premise, this review provides a concise overview of "classic" and novel promising adipocytokines associated with AT inflammation and discusses possible critical approaches to their interpretation in clinical practice.
Collapse
Affiliation(s)
- I Barchetta
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, 00161, Rome, Italy
| | - F A Cimini
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, 00161, Rome, Italy
| | - G Ciccarelli
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, 00161, Rome, Italy
| | - M G Baroni
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, 00161, Rome, Italy.
| | - M G Cavallo
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, 00161, Rome, Italy.
| |
Collapse
|
24
|
Ramirez-Peña E, Arnold J, Shivakumar V, Joseph R, Vidhya Vijay G, den Hollander P, Bhangre N, Allegakoen P, Prasad R, Conley Z, Matés JM, Márquez J, Chang JT, Vasaikar S, Soundararajan R, Sreekumar A, Mani SA. The Epithelial to Mesenchymal Transition Promotes Glutamine Independence by Suppressing GLS2 Expression. Cancers (Basel) 2019; 11:cancers11101610. [PMID: 31652551 PMCID: PMC6826439 DOI: 10.3390/cancers11101610] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 10/05/2019] [Accepted: 10/18/2019] [Indexed: 12/17/2022] Open
Abstract
Identifying bioenergetics that facilitate the epithelial to mesenchymal transition (EMT) in breast cancer cells may uncover targets to treat incurable metastatic disease. Metastasis is the number one cause of cancer-related deaths; therefore, it is urgent to identify new treatment strategies to prevent the initiation of metastasis. To characterize the bioenergetics of EMT, we compared metabolic activities and gene expression in cells induced to differentiate into the mesenchymal state with their epithelial counterparts. We found that levels of GLS2, which encodes a glutaminase, are inversely associated with EMT. GLS2 down-regulation was correlated with reduced mitochondrial activity and glutamine independence even in low-glucose conditions. Restoration of GLS2 expression in GLS2-negative breast cancer cells rescued mitochondrial activity, enhanced glutamine utilization, and inhibited stem-cell properties. Additionally, inhibition of expression of the transcription factor FOXC2, a critical regulator of EMT in GLS2-negative cells, restored GLS2 expression and glutamine utilization. Furthermore, in breast cancer patients, high GLS2 expression is associated with improved survival. These findings suggest that epithelial cancer cells rely on glutamine and that cells induced to undergo EMT become glutamine independent. Moreover, the inhibition of EMT leads to a GLS2-directed metabolic shift in mesenchymal cancer cells, which may make these cells susceptible to chemotherapies.
Collapse
Affiliation(s)
- Esmeralda Ramirez-Peña
- National Cancer Institute, Cancer Prevention Fellowship Program, Division of Cancer Prevention, Bethesda, MD 20892, USA.
| | - James Arnold
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Vinita Shivakumar
- Wiess School of Natural Sciences, Rice University, Houston, TX 77005, USA.
| | - Robiya Joseph
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| | | | - Petra den Hollander
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Neeraja Bhangre
- Department of Fibrosis Biology, Gilead Sciences, Foster City, CA 94404, USA.
| | - Paul Allegakoen
- Department of Medicine, University of California-San Francisco, San Francisco, CA 94143, USA.
| | - Rishika Prasad
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Zachary Conley
- Center for Science Outreach, Department of Teaching and Learning, Vanderbilt University, Nashville, TN 37235, USA.
| | - José M Matés
- Canceromics Lab, Department of Molecular Biology and Biochemistry, University of Málaga and Instituto de Investigación Biomedica de Málaga (IBIMA), 29071 Málaga, Spain.
| | - Javier Márquez
- Canceromics Lab, Department of Molecular Biology and Biochemistry, University of Málaga and Instituto de Investigación Biomedica de Málaga (IBIMA), 29071 Málaga, Spain.
| | - Jeffrey T Chang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX 77030, USA.
| | - Suhas Vasaikar
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Rama Soundararajan
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Arun Sreekumar
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Sendurai A Mani
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
25
|
Medela AMB, Penton A, Bostrom KI, Saparov A, Jumabay M. Generation of Vascular Networks from Adipocytes In Vitro. INTERNATIONAL JOURNAL OF CELL SCIENCE & MOLECULAR BIOLOGY 2019; 6:555684. [PMID: 33954280 PMCID: PMC8095932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Multipotent cells derived from white mature adipocytes, referred to as dedifferentiated fat (DFAT) cells have the capacity differentiate into endothelial cells. The objective of this study was to modify the isolation method for DFAT cells in order to optimize the endothelial lineage potential. The adipocytes were preincubated for 24 hours, washed, and then incubated for 5 days to allow the generated DFAT cells to remain in proximity to the adipocytes while the cells aggregated into cell clusters. The DFAT cells rapidly differentiated into adipocytes after which endothelial-like cells (ECs) emerged and formed tube-like structure closely associated with the newly differentiated adipocytes. The lipid-filled cells then gradually disappeared whereas the network of tube structure expanded over the course of 3 weeks. ECs accounted for 35-45% of the cells derived from the DFAT cells, as assessed by qPCR, immunofluorescence and fluorescence-activated cell sorting. The DFAT cell-derived ECs could also be further enriched by magnetic sorting, thereby serving as a mouse cell line for further research.
Collapse
Affiliation(s)
| | - Ashley Penton
- Division of Cardiology, David Geffen School of Medicine at UCLA, USA
| | | | - Arman Saparov
- Department of Medicine, Nazarbayev University School of Medicine, Kazakhstan
| | - Medet Jumabay
- Division of Cardiology, David Geffen School of Medicine at UCLA, USA,Corresponding author: Medet Jumabay, Division of Cardiology, David Geffen School of Medicine at UCLA, Box 951679, Los Angeles, CA 90095-1679
| |
Collapse
|
26
|
Aabel P, Utheim TP, Olstad OK, Rask-Andersen H, Dilley RJ, von Unge M. Transcription and microRNA Profiling of Cultured Human Tympanic Membrane Epidermal Keratinocytes. J Assoc Res Otolaryngol 2018; 19:243-260. [PMID: 29623476 DOI: 10.1007/s10162-018-0660-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 02/19/2018] [Indexed: 01/06/2023] Open
Abstract
The human tympanic membrane (TM) has a thin outer epidermal layer which plays an important role in TM homeostasis and ear health. The specialised cells of the TM epidermis have a different physiology compared to normal skin epidermal keratinocytes, displaying a dynamic and constitutive migration that maintains a clear TM surface and assists in regeneration. Here, we characterise and compare molecular phenotypes in keratinocyte cultures from TM and normal skin. TM keratinocytes were isolated by enzymatic digestion and cultured in vitro. We compared global mRNA and microRNA expression of the cultured cells with that of human epidermal keratinocyte cultures. Genes with either relatively higher or lower expression were analysed further using the biostatistical tools g:Profiler and Ingenuity Pathway Analysis. Approximately 500 genes were found differentially expressed. Gene ontology enrichment and Ingenuity analyses identified cellular migration and closely related biological processes to be the most significant functions of the genes highly expressed in the TM keratinocytes. The genes of low expression showed a marked difference in homeobox (HOX) genes of clusters A and C, giving the TM keratinocytes a strikingly low HOX gene expression profile. An in vitro scratch wound assay showed a more individualised cell movement in cells from the tympanic membrane than normal epidermal keratinocytes. We identified 10 microRNAs with differential expression, several of which can also be linked to regulation of cell migration and expression of HOX genes. Our data provides clues to understanding the specific physiological properties of TM keratinocytes, including candidate genes for constitutive migration, and may thus help focus further research.
Collapse
Affiliation(s)
- Peder Aabel
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway. .,Ear, Nose and Throat Department, Division of Surgery, Akershus University Hospital, Lørenskog, Norway. .,Division of Surgery, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Tor Paaske Utheim
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway.,Institute of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | | | | | - Rodney James Dilley
- Ear Science Institute Australia, Perth, Australia.,Ear Sciences Centre and Centre for Cell Therapy and Regenerative Medicine, University of Western Australia, Nedlands, Australia
| | - Magnus von Unge
- Ear, Nose and Throat Department, Division of Surgery, Akershus University Hospital, Lørenskog, Norway.,Division of Surgery, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Centre for Clinical Research, University of Uppsala, Västerås, Sweden
| |
Collapse
|
27
|
Ahn J, Han KS, Heo JH, Bang D, Kang YH, Jin HA, Hong SJ, Lee JH, Ham WS. FOXC2 and CLIP4 : a potential biomarker for synchronous metastasis of ≤7-cm clear cell renal cell carcinomas. Oncotarget 2018; 7:51423-51434. [PMID: 27283491 PMCID: PMC5239485 DOI: 10.18632/oncotarget.9842] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 05/20/2016] [Indexed: 12/15/2022] Open
Abstract
Renal cell carcinomas (RCC) smaller than 7-cm are heterogeneous and exhibit metastatic potential in approximately 15% of cases. Although large-scale characterization of mutations in clear cell RCC (ccRCC), the most common RCC subtype, has been established, the genetic alterations related to ≤7-cm ccRCCs undergoing synchronous metastasis are poorly understood. To discover biomarkers that can be used to estimate the risk of synchronous metastasis in these ccRCC patients, we performed whole exome sequencing on the formalin-fixed paraffin-embedded (FFPE) samples of 10 ccRCC patients with ≤7-cm tumors and synchronous metastasis and expanded our study using The Cancer Genome Atlas (TCGA) ccRCC dataset (n = 201). Recurrent mutations were selected according to functional prediction and statistical significance. Mutations in three candidate genes, RELN (1 out of 10), FOXC2 (1 out of 10), and CLIP4 (2 out of 10) were found in expanded analysis using a TCGA cohort. Furthermore, siRNA-mediated target gene knockdown (FOXC2 and CLIP4) and overexpression (RELN) assays showed that FOXC2 and CLIP4 significantly increased cell migration and viability in ccRCCs. Our study demonstrated that FOXC2 and CLIP4 activity correlates to the presence of ≤7-cm ccRCCs with synchronous metastasis and may be potential molecular predictors of synchronous metastasis of ≤7-cm ccRCCs.
Collapse
Affiliation(s)
- Jinwoo Ahn
- Department of Chemistry, Yonsei University, Seoul, Korea
| | - Kyung Seok Han
- Department of Urology, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| | - Jun Hyeok Heo
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Duhee Bang
- Department of Chemistry, Yonsei University, Seoul, Korea
| | - You Hyun Kang
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Hyun A Jin
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Sung Joon Hong
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Hyun Lee
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Won Sik Ham
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
28
|
Wang T, Zheng L, Wang Q, Hu YW. Emerging roles and mechanisms of FOXC2 in cancer. Clin Chim Acta 2018; 479:84-93. [PMID: 29341903 DOI: 10.1016/j.cca.2018.01.019] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/12/2018] [Accepted: 01/12/2018] [Indexed: 12/20/2022]
Abstract
Forkhead box protein C2 (FOXC2), a transcription factor of the forkhead/winged-helix family, is required for embryonic and prenatal development. FOXC2 acts as a crucial modulator during both angiogenesis and lymphangiogenesis via multiple angiogenic and lymphangiogenic pathways, respectively. Although recent studies have shed light on the emerging role of FOXC2 in cancer, very little is known about the precise underlying mechanisms. The purpose of this review is to summarize the current understanding of FOXC2 and provide potential mechanistic explanations of the relationship between FOXC2 and cancer, as well as discuss the prospect for future research in the promising prognostic value of FOXC2 in cancer.
Collapse
Affiliation(s)
- Teng Wang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Lei Zheng
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Qian Wang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yan-Wei Hu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
29
|
Tsuji M, Morishima M, Shimizu K, Morikawa S, Heglind M, Enerbäck S, Ezaki T, Tamaoki J. Foxc2influences alveolar epithelial cell differentiation during lung development. Dev Growth Differ 2017; 59:501-514. [DOI: 10.1111/dgd.12368] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 04/17/2017] [Accepted: 05/07/2017] [Indexed: 12/30/2022]
Affiliation(s)
- Mayoko Tsuji
- First Department of Medicine; Tokyo Women's Medical University; Tokyo Japan
| | - Masae Morishima
- Department of Anatomy and Developmental Biology; Tokyo Women's Medical University; Tokyo Japan
| | - Kazuhiko Shimizu
- Department of Anatomy and Developmental Biology; Tokyo Women's Medical University; Tokyo Japan
| | - Shunichi Morikawa
- Department of Anatomy and Developmental Biology; Tokyo Women's Medical University; Tokyo Japan
| | - Mikael Heglind
- Department of Medical Biochemistry and Cell Biology; Institute of Biomedicine; University of Gothenburg; Gothenburg Sweden
| | - Sven Enerbäck
- Department of Medical Biochemistry and Cell Biology; Institute of Biomedicine; University of Gothenburg; Gothenburg Sweden
| | - Taichi Ezaki
- Department of Anatomy and Developmental Biology; Tokyo Women's Medical University; Tokyo Japan
| | - Jun Tamaoki
- First Department of Medicine; Tokyo Women's Medical University; Tokyo Japan
| |
Collapse
|
30
|
Breier G, Chavakis T, Hirsch E. Angiogenesis in metabolic-vascular disease. Thromb Haemost 2017; 117:1289-1295. [PMID: 28594427 DOI: 10.1160/th17-05-0325] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 05/29/2017] [Indexed: 12/15/2022]
Abstract
Angiogenesis, literally formation of new blood vessels, is the main process through which the vascular system expands during embryonic and postnatal development. Endothelial cells, which constitute the inner lining of all blood vessels, are typically in a quiescent state in the healthy adult organism. However, in vascular and metabolic diseases, the endothelium becomes unstable and dysfunctional. The resulting tissue hypoxia may thereby induce pathological angiogenesis, which is a hallmark of disease conditions like cancer or diabetic retinopathy. However, recent evidence suggests that angiogenesis is also a major player in the context of further metabolic diseases, especially in obesity. In particular, deregulated angiogenesis is linked with adipose tissue dysfunction and insulin resistance. On the other hand, signalling pathways, such as the PI3K pathway, may regulate metabolic activities in the endothelium. Endothelial cell metabolism emerges as an important regulator of angiogenesis. This review summarises the role of angiogenesis in metabolic-vascular disease, with specific focus on the role of angiogenesis in obesity-related metabolic dysfunction and on signaling pathways, especially PI3K, linking cell metabolism to endothelial function.
Collapse
Affiliation(s)
| | - Triantafyllos Chavakis
- Triantafyllos Chavakis, Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany, E-mail:
| | | |
Collapse
|
31
|
An YA, Sun K, Joffin N, Zhang F, Deng Y, Donzé O, Kusminski CM, Scherer PE. Angiopoietin-2 in white adipose tissue improves metabolic homeostasis through enhanced angiogenesis. eLife 2017; 6. [PMID: 28355132 PMCID: PMC5391203 DOI: 10.7554/elife.24071] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 03/28/2017] [Indexed: 01/12/2023] Open
Abstract
Despite many angiogenic factors playing crucial roles in metabolic homeostasis, effects of angiopoietin-2 (ANG-2) in adipose tissue (AT) remain unclear. Utilizing a doxycycline-inducible AT-specific ANG-2 overexpression mouse model, we assessed the effects of ANG-2 in AT expansion upon a high-fat diet (HFD) challenge. ANG-2 is significantly induced, with subcutaneous white AT (sWAT) displaying the highest ANG-2 expression. ANG-2 overexpressing mice show increased sWAT vascularization and are resistant to HFD-induced obesity. In addition, improved glucose and lipid metabolism are observed. Mechanistically, the sWAT displays a healthier expansion pattern with increased anti-inflammatory macrophage infiltration. Conversely, ANG-2 neutralization in HFD-challenged wild-type mice shows reduced vascularization in sWAT, associated with impaired glucose tolerance and lipid clearance. Blocking ANG-2 causes significant pro-inflammatory and pro-fibrotic changes, hallmarks of an unhealthy AT expansion. In contrast to other pro-angiogenic factors, such as vascular endothelial growth factor-A (VEGF-A), this is achieved without any enhanced beiging of white AT. DOI:http://dx.doi.org/10.7554/eLife.24071.001
Collapse
Affiliation(s)
- Yu A An
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Kai Sun
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States.,Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, United States
| | - Nolwenn Joffin
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Fang Zhang
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States.,Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yingfeng Deng
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | | | - Christine M Kusminski
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States.,Department of Cell Biology, The University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
32
|
Crewe C, An YA, Scherer PE. The ominous triad of adipose tissue dysfunction: inflammation, fibrosis, and impaired angiogenesis. J Clin Invest 2017; 127:74-82. [PMID: 28045400 DOI: 10.1172/jci88883] [Citation(s) in RCA: 509] [Impact Index Per Article: 63.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
There are three dominant contributors to the pathogenesis of dysfunctional adipose tissue (AT) in obesity: unresolved inflammation, inappropriate extracellular matrix (ECM) remodeling and insufficient angiogenic potential. The interactions of these processes during AT expansion reflect both a linear progression as well as feed-forward mechanisms. For example, both inflammation and inadequate angiogenic remodeling can drive fibrosis, which can in turn promote migration of immune cells into adipose depots and impede further angiogenesis. Therefore, the relationship between the members of this triad is complex but important for our understanding of the pathogenesis of obesity. Here we untangle some of these intricacies to highlight the contributions of inflammation, angiogenesis, and the ECM to both "healthy" and "unhealthy" AT expansion.
Collapse
|
33
|
Munger SJ, Geng X, Srinivasan RS, Witte MH, Paul DL, Simon AM. Segregated Foxc2, NFATc1 and Connexin expression at normal developing venous valves, and Connexin-specific differences in the valve phenotypes of Cx37, Cx43, and Cx47 knockout mice. Dev Biol 2016; 412:173-90. [PMID: 26953188 PMCID: PMC4826804 DOI: 10.1016/j.ydbio.2016.02.033] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 02/12/2016] [Accepted: 02/12/2016] [Indexed: 01/25/2023]
Abstract
Venous valves (VVs) are critical for unidirectional blood flow from superficial and deep veins towards the heart. Congenital valve aplasia or agenesis may, in some cases, be a direct cause of vascular disease, motivating an understanding of the molecular mechanisms underlying the development and maintenance of VVs. Three gap junction proteins (Connexins), Cx37, Cx43, and Cx47, are specifically expressed at VVs in a highly polarized fashion. VVs are absent from adult mice lacking Cx37; however it is not known if Cx37 is required for the initial formation of valves. In addition, the requirement of Cx43 and Cx47 for VV development has not been studied. Here, we provide a detailed description of Cx37, Cx43, and Cx47 expression during mouse vein development and show by gene knockout that each Cx is necessary for normal valve development. The valve phenotypes in the knockout lines exhibit Cx-specific differences, however, including whether peripheral or central VVs are affected by gene inactivation. In addition, we show that a Cx47 null mutation impairs peripheral VV development but does not affect lymphatic valve formation, a finding of significance for understanding how some CX47 mutations cause inherited lymphedema in humans. Finally, we demonstrate a striking segregation of Foxc2 and NFATc1 transcription factor expression between the downstream and upstream faces, respectively, of developing VV leaflets and show that this segregation is closely associated with the highly polarized expression of Cx37, Cx43, and Cx47. The partition of Foxc2 and NFATc1 expression at VV leaflets makes it unlikely that these factors directly cooperate during the leaflet elongation stage of VV development.
Collapse
Affiliation(s)
| | - Xin Geng
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
| | - R Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
| | - Marlys H Witte
- Department of Surgery, University of Arizona, Tucson, AZ 85724, USA.
| | - David L Paul
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| | - Alexander M Simon
- Department of Physiology, University of Arizona, Tucson, AZ 85724, USA.
| |
Collapse
|
34
|
Rajput C, Tauseef M, Farazuddin M, Yazbeck P, Amin MR, Avin Br V, Sharma T, Mehta D. MicroRNA-150 Suppression of Angiopoetin-2 Generation and Signaling Is Crucial for Resolving Vascular Injury. Arterioscler Thromb Vasc Biol 2016; 36:380-8. [PMID: 26743170 DOI: 10.1161/atvbaha.115.306997] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 12/17/2015] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Increased vascular permeability is a hallmark of sepsis and acute respiratory distress syndrome. Angiopoietin (Ang2) induces vascular leak, and excess Ang2 generation is associated with patient mortality from these diseases. However, mechanisms dampening Ang2 generation during injury remain unclear. Interestingly, microRNA (miR)-150 levels were decreased in septic patients. miR regulate signaling networks by silencing mRNAs containing complementary sequences. Thus, we hypothesized that miR-150 suppresses Ang2 generation and thereby resolves vascular injury. APPROACH AND RESULTS Wild-type or miR-150(-/-) mice or endothelial cells were exposed to lipopolysaccharide or sepsis, and Ang2 levels, adherens junction reannealing, endothelial barrier function, and mortality were determined. Although Ang2 transiently increased during lipopolysaccharide-induced injury in wild-type endothelial cells and lungs, miR-150 expression was elevated only during recovery from injury. Deletion of miR-150 caused a persistent increase in Ang2 levels and impaired adherens junctions reannealing after injury, resulting thereby in an irreversible increase in vascular permeability. Also, miR-150(-/-) mice died rapidly after sepsis. Rescuing miR-150 expression in endothelial cells prevented Ang2 generation, thereby restoring vascular barrier function in miR-150(-/-) mice. miR-150 terminated Ang2 generation by targeting the transcription factor, early growth response 2. Thus, early growth response 2 or Ang2 depletion in miR-150(-/-) endothelial cells restored junctional reannealing and reinstated barrier function. Importantly, upregulating miR-150 expression by injecting a chemically synthesized miR-150 mimic into wild-type mice vasculature decreased early growth response 2 and Ang2 levels and hence mortality from sepsis. CONCLUSIONS miR-150 is a novel suppressor of Ang2 generation with a key role in resolving vascular injury and reducing mortality resulting from sepsis.
Collapse
Affiliation(s)
- Charu Rajput
- From the Department of Pharmacology and Center for Lung and Vascular Biology, University of Illinois at Chicago
| | - Mohammad Tauseef
- From the Department of Pharmacology and Center for Lung and Vascular Biology, University of Illinois at Chicago
| | - Mohammad Farazuddin
- From the Department of Pharmacology and Center for Lung and Vascular Biology, University of Illinois at Chicago
| | - Pascal Yazbeck
- From the Department of Pharmacology and Center for Lung and Vascular Biology, University of Illinois at Chicago
| | - Md-Ruhul Amin
- From the Department of Pharmacology and Center for Lung and Vascular Biology, University of Illinois at Chicago
| | - Vijay Avin Br
- From the Department of Pharmacology and Center for Lung and Vascular Biology, University of Illinois at Chicago
| | - Tiffany Sharma
- From the Department of Pharmacology and Center for Lung and Vascular Biology, University of Illinois at Chicago
| | - Dolly Mehta
- From the Department of Pharmacology and Center for Lung and Vascular Biology, University of Illinois at Chicago.
| |
Collapse
|
35
|
Zhang X, Tian Y, Zhang H, Kavishwar A, Lynes M, Brownell AL, Sun H, Tseng YH, Moore A, Ran C. Curcumin analogues as selective fluorescence imaging probes for brown adipose tissue and monitoring browning. Sci Rep 2015; 5:13116. [PMID: 26269357 PMCID: PMC4534785 DOI: 10.1038/srep13116] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 07/16/2015] [Indexed: 01/10/2023] Open
Abstract
Manipulation of brown adipose tissue (BAT) and browning of white adipose tissue (WAT) can be promising new approaches to counter metabolic disorder diseases in humans. Imaging probes that could consistently monitor BAT mass and browning of WAT are highly desirable. In the course of our imaging probe screening, we found that BAT could be imaged with curcumin analogues in mice. However, the poor BAT selectivity over WAT and short emissions of the lead probes promoted further lead optimization. Limited uptake mechanism studies suggested that CD36/FAT (fatty acid transporter) probably contributed to the facilitated uptake of the probes. By increasing the stereo-hindrance of the lead compound, we designed CRANAD-29 to extend the emission and increase the facilitated uptake, thus increasing its BAT selectivity. Our data demonstrated that CRANAD-29 had significantly improved selectivity for BAT over WAT, and could be used for imaging BAT mass change in a streptozotocin-induced diabetic mouse model, as well as for monitoring BAT activation under cold exposure. In addition, CRANAD-29 could be used for monitoring the browning of subcutaneous WAT (sWAT) induced by β3-adrenoceptor agonist CL-316, 243.
Collapse
Affiliation(s)
- Xueli Zhang
- 1] Molecular Imaging Laboratory, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital/Harvard Medical School, Boston, MA [2] School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China [3] Department of pharmacy, ZhongDa Hospital, Southeast University, Nanjing 210009, China
| | - Yanli Tian
- 1] Molecular Imaging Laboratory, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital/Harvard Medical School, Boston, MA [2] Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Hongbin Zhang
- Joslin Diabetes Center, Harvard Medical School, and Harvard Stem Cell Institute, Boston, MA 02215
| | - Amol Kavishwar
- Molecular Imaging Laboratory, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital/Harvard Medical School, Boston, MA
| | - Matthew Lynes
- Joslin Diabetes Center, Harvard Medical School, and Harvard Stem Cell Institute, Boston, MA 02215
| | - Anna-Liisa Brownell
- Molecular Imaging Laboratory, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital/Harvard Medical School, Boston, MA
| | - Hongbin Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yu-Hua Tseng
- Joslin Diabetes Center, Harvard Medical School, and Harvard Stem Cell Institute, Boston, MA 02215
| | - Anna Moore
- Molecular Imaging Laboratory, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital/Harvard Medical School, Boston, MA
| | - Chongzhao Ran
- Molecular Imaging Laboratory, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital/Harvard Medical School, Boston, MA
| |
Collapse
|
36
|
Zhou Z, Zhang L, Xie B, Wang X, Yang X, Ding N, Zhang J, Liu Q, Tan G, Feng D, Sun LQ. FOXC2 promotes chemoresistance in nasopharyngeal carcinomas via induction of epithelial mesenchymal transition. Cancer Lett 2015; 363:137-45. [PMID: 25896630 DOI: 10.1016/j.canlet.2015.04.008] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 04/03/2015] [Accepted: 04/08/2015] [Indexed: 10/23/2022]
Abstract
Paclitaxel (Taxol) is currently used as the front-line chemotherapeutic drug for many types of human cancers. However, the emergence of drug resistance has been a major obstacle to the effective treatment of cancers in clinical settings. The transcription factor Forkhead box protein C2 (FOXC2) was recently demonstrated to activate the epithelial-mesenchymal transition (EMT). In this article, we present a novel role of FOXC2 in regulating chemoresistance of nasopharyngeal carcinoma (NPC) through the EMT. Using an EMT PCR array based on the screening of 84 genes, the expression of FOXC2 was notably upregulated in paclitaxel-resistant NPC cells (CNE2/t). We observed that the paclitaxel-resistant cells exhibited characteristic EMT phenotypes. The silencing of FOXC2 expression in the resistant cells can reverse the EMT molecular markers and chemoresistant phenotypes, such as cellular morphology, proliferation and anoikis. In an NPC xenograft mouse model, the downregulation of FOXC2 expression in the resistant NPC cells increased their sensitivity to paclitaxel treatment, resulting in reduced tumor growth. Taken together, our results suggest that FOXC2-mediated EMT may be an alternative mechanism through which cancer cells can initiate and maintain drug resistance. Thus, targeting FOXC2 may provide a novel strategy for overcoming chemoresistance in NPC therapy.
Collapse
Affiliation(s)
- Zhijiao Zhou
- Department of Pathology, Xiangya Hospital and School of Basic Medical Sciences, Central South University, Changsha 410008, China
| | - Lu Zhang
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Bowen Xie
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xiangpu Wang
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xinhui Yang
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Nianhua Ding
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jing Zhang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qingqing Liu
- Department of Respiration, 2nd Xiangya Hospital, Central South University, Changsha 410008, China
| | - Guolin Tan
- Department of Otolaryngology Head and Neck Surgery, 3(rd) Xiangya Hospital, Changsha 410008, China
| | - Deyun Feng
- Department of Pathology, Xiangya Hospital and School of Basic Medical Sciences, Central South University, Changsha 410008, China.
| | - Lun-Quan Sun
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
37
|
Qian S, Huang H, Tang Q. Brown and beige fat: the metabolic function, induction, and therapeutic potential. Front Med 2015; 9:162-72. [DOI: 10.1007/s11684-015-0382-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 10/16/2014] [Indexed: 12/27/2022]
|
38
|
Correlation of Forkhead Box c2 with subtypes and invasive ability of invasive breast cancer. ACTA ACUST UNITED AC 2014; 34:896-901. [DOI: 10.1007/s11596-014-1370-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 08/31/2014] [Indexed: 10/24/2022]
|
39
|
Cui YM, Jiao HL, Ye YP, Chen CM, Wang JX, Tang N, Li TT, Lin J, Qi L, Wu P, Wang SY, He MR, Liang L, Bian XW, Liao WT, Ding YQ. FOXC2 promotes colorectal cancer metastasis by directly targeting MET. Oncogene 2014; 34:4379-90. [PMID: 25381815 DOI: 10.1038/onc.2014.368] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 09/24/2014] [Accepted: 09/30/2014] [Indexed: 12/11/2022]
Abstract
Metastasis is the major cause of death in colorectal cancer (CRC). Although multiple genes have been identified to be responsible for the development of CRC, the molecular changes that enable CRC cells to undergo early local invasion and to form distant metastatic colonies still remain largely unknown. Herein, we investigated the role of Forkhead box protein C2 (FOXC2) and explored the underlying mechanisms in invasion and metastasis of CRC. We show that both high FOXC2 expression and nuclear localization of FOXC2 are significantly correlated with advanced TNM (T=primary tumor; N=regional lymph nodes; M=distant metastasis) stages. FOXC2 enhanced the invasive abilities of CRC cells in vitro and promoted local invasion and distant metastasis in an orthotopic mouse metastatic model of CRC. Microarray analysis revealed that overexpression of FOXC2 increased the proto-oncogene MET tyrosine kinase expression and activated the hepatocyte growth factor (HGF)-MET signaling pathway. Furthermore, luciferase reporter assays and chromatin immunoprecipitation assays revealed that FOXC2 directly associated with MET promoter to increase the transcriptional activity of MET. Inhibition of MET attenuates the invasive phenotype and metastatic potential of FOXC2-overexpressing CRC cells, indicating that MET is a major mediator of FOXC2-promoted metastasis. In addition, FOXC2 expression was positively correlated with MET expression in CRC tissue samples. Our findings suggest that FOXC2 has a crucial role in CRC metastasis by regulating HGF-MET signaling via inducing MET expression, highlighting FOXC2 as a potential therapeutic target for preventing or reducing metastasis in CRC.
Collapse
Affiliation(s)
- Y-M Cui
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - H-L Jiao
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Y-P Ye
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - C-M Chen
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - J-X Wang
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - N Tang
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - T-T Li
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - J Lin
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - L Qi
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - P Wu
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - S-Y Wang
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - M-R He
- Guangdong Provincial Key Laboratory of Gastroenterology and Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - L Liang
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - X-W Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - W-T Liao
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Y-Q Ding
- 1] Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China [3] Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| |
Collapse
|
40
|
Sargent C, Bauer J, Khalil M, Filmore P, Bernas M, Witte M, Pearson MP, Erickson RP. A five generation family with a novel mutation in FOXC2 and lymphedema worsening to hydrops in the youngest generation. Am J Med Genet A 2014; 164A:2802-7. [PMID: 25252123 DOI: 10.1002/ajmg.a.36736] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 07/27/2014] [Indexed: 12/29/2022]
Abstract
We describe a five generation family with dominantly inherited lymphedema, but no distichiasis, in which 3/3 affected offspring in the fifth generation have died of fetal hydrops and related birth defects. Mutational analysis disclosed a novel mutation in FOXC2 (R121C) in affected members. We searched for possible genetic influences on the greater severity of lymphedema (hydrops) in the fifth generation. Karyotypes disclosed an extra band in Xp in one affected fetus, but this was also found in the mother. Copy number variation (CNV) studies on four members of the pedigree (mother of the three severely affected fetuses/infants; one severely affected; a full, and a half, unaffected sibs) did not detect the source of the Xp band or a possible influence on the severe phenotype. However, use of SNP arrays did allow identification of the portion of the maternal proximal Xp shared by a hydrops-affected daughter and son which was not shared by an unaffected daughter from the same sibship.
Collapse
Affiliation(s)
- Carole Sargent
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, UK
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Igniting thermogenesis within white adipose tissue (i.e., promoting expression and activity of the uncoupling protein UCP1) has attracted much interest. Numerous "browning agents" have now been described (gene ablations, transgenes, food components, drugs, environments, etc.). The implied action of browning agents is that they increase UCP1 through this heat production, leading to slimming. Here, we particularly point to the possibility that cause and effect may on occasion be the reverse: browning agents may disrupt, for example, the fur, leading to increased heat loss, increased thermogenic demand to counteract this heat loss, and thus, through sympathetic nervous system activation, to enhanced UCP1 expression in white (and brown) adipose tissues.
Collapse
Affiliation(s)
- Jan Nedergaard
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 91 Stockholm, Sweden.
| | - Barbara Cannon
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 91 Stockholm, Sweden
| |
Collapse
|
42
|
Orekhov AN, Bobryshev YV, Chistiakov DA. The complexity of cell composition of the intima of large arteries: focus on pericyte-like cells. Cardiovasc Res 2014; 103:438-51. [PMID: 25016615 DOI: 10.1093/cvr/cvu168] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Pericytes, which are also known as Rouget cells or perivascular cells, are considered to represent a likely distinct pool of vascular cells that are extremely branched and located mostly in the periphery of the vascular system. The family of pericytes is a heterogeneous cell population that includes pericytes and pericyte-like cells. Accumulated data indicate that networks of pericyte-like cells exist in normal non-atherosclerotic intima, and that pericyte-like cells can be involved in the development of atherosclerotic lesions from the very early stages of disease. The pathogenic role of arterial pericytes and pericyte-like cells also might be important in advanced and complicated atherosclerotic lesions via realizing mechanisms of vascular remodelling, ectopic ossification, intraplaque neovascularization, and probably thrombosis.
Collapse
Affiliation(s)
- Alexander N Orekhov
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| | - Yuri V Bobryshev
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia Faculty of Medicine, School of Medical Sciences, University of New South Wales, Kensington, Sydney, NSW 2052, Australia
| | - Dimitry A Chistiakov
- Department of Medical Nanobiotechnology, Pirogov Russian State Medical University, Moscow, Russia
| |
Collapse
|
43
|
Peirce V, Carobbio S, Vidal-Puig A. The different shades of fat. Nature 2014; 510:76-83. [PMID: 24899307 DOI: 10.1038/nature13477] [Citation(s) in RCA: 343] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 03/26/2014] [Indexed: 01/09/2023]
Abstract
Our understanding of adipose tissue biology has progressed rapidly since the turn of the century. White adipose tissue has emerged as a key determinant of healthy metabolism and metabolic dysfunction. This realization is paralleled only by the confirmation that adult humans have heat-dissipating brown adipose tissue, an important contributor to energy balance and a possible therapeutic target for the treatment of metabolic disease. We propose that the development of successful strategies to target brown and white adipose tissues will depend on investigations that elucidate their developmental origins and cell-type-specific functional regulators.
Collapse
Affiliation(s)
- Vivian Peirce
- University of Cambridge Metabolic Research Laboratories, Level 4, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Cambridge CB2 OQQ, UK
| | - Stefania Carobbio
- 1] University of Cambridge Metabolic Research Laboratories, Level 4, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Cambridge CB2 OQQ, UK. [2] Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Antonio Vidal-Puig
- 1] University of Cambridge Metabolic Research Laboratories, Level 4, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Cambridge CB2 OQQ, UK. [2] Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| |
Collapse
|
44
|
Li D, Wei X, Xie K, Chen K, Li J, Fang J. A novel decoy receptor fusion protein for FGF-2 potently inhibits tumour growth. Br J Cancer 2014; 111:68-77. [PMID: 24874473 PMCID: PMC4090743 DOI: 10.1038/bjc.2014.282] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/29/2014] [Accepted: 04/30/2014] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Antiangiogenic therapies have been proven effective in cancer treatment. Fibroblast growth factor-2 (FGF-2) has been functionally implicated in tumour angiogenesis and is an important target of antiangiogenic therapies. The aim of this work was to develop a novel FGF-2 inhibitor for cancer therapy. METHODS Eleven fusion proteins were developed by fusing various truncated extracellular regions of FGFR1 with the Fc region of IgG1. The optimal decoy receptor fusion protein with the highest binding affinity for FGF-2 was identified by an FGF-2-binding assay and its potential antitumour effects were investigated. RESULTS We obtained a soluble decoy receptor fusion protein with the highest binding activity for FGF-2, named FGF-Trap. Fibroblast growth factor-Trap significantly abolished FGF-2-stimulated activation of FGF signalling as demonstrated by its suppression of FGF-2-mediated phosphorylation of Erk1/2 and Akt, upregulation of cyclins D1 and E and the increase in mRNA levels of vascular endothelial growth factor R1 and R2 (VEGFR1 and VEGFR2). Furthermore, FGF-Trap effectively suppressed FGF-2-induced proliferation and migration of human umbilical vein endothelial cells (HUVECs) in vitro. Most importantly, FGF-Trap potently inhibited tumour growth and angiogenesis in Caki-1 and A549 xenograft models in vivo. CONCLUSIONS Fibroblast growth factor-Trap potently inhibits tumour growth by blocking FGF-2 signalling pathways and could be an effective therapeutic agent for cancer patients.
Collapse
Affiliation(s)
- D Li
- School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - X Wei
- School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - K Xie
- School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - K Chen
- School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - J Li
- School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - J Fang
- School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| |
Collapse
|
45
|
Xia CY, Yu AX, Qi B, Zhou M, Li ZH, Wang WY. Analysis of blood flow and local expression of angiogenesis‑associated growth factors in infected wounds treated with negative pressure wound therapy. Mol Med Rep 2014; 9:1749-54. [PMID: 24584462 DOI: 10.3892/mmr.2014.1997] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 02/19/2014] [Indexed: 11/06/2022] Open
Abstract
Angiogenesis is involved in the wound healing process. Increased angiogenesis and blood flow constitute a major mechanism of negative pressure wound therapy (NPWT), which has been shown to facilitate the healing of infected wounds. However, the effect on the expression of angiogensis‑related growth factor remains unknown. The goal of the current study was to investigate the angiogenic factor levels prior to and following NPWT in infected wounds. A total of 20 patients with infected wounds treated with NPWT were included in the study. Patients acted as their own control; the postoperative measurements of patients were considered as the experimental group, while preoperative measurements were considered as the controlled group. Blood flow was recorded prior to and during NPWT. A total of 10 angiogensis‑related growth factors were detected using a protein biochip array to analyze the change in protein levels prior to NPWT, and on the third day during NPWT. All wounds were successfully reconstructed by skin grafting or using local flaps following NPWT. NPWT resulted in significantly increased blood flow in the wound. There was a significant increase in vascular endothelial growth factor (VEGF), EGF, platelet‑derived growth factor and angiotesin‑2 following NPWT, while basic fibroblast growth factor decreased significantly. NPWT affects the local expression of angiogenesis‑associated growth factors, which represents another mechanism to explain how NPWT accelerates wound healing.
Collapse
Affiliation(s)
- Cheng-Yan Xia
- Department of Micro‑Orthopaedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Ai-Xi Yu
- Department of Micro‑Orthopaedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Baiwen Qi
- Department of Micro‑Orthopaedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Min Zhou
- Department of Micro‑Orthopaedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zong-Huan Li
- Department of Micro‑Orthopaedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Wei-Yang Wang
- Department of Micro‑Orthopaedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
46
|
Hosaka K, Yang Y, Seki T, Nakamura M, Andersson P, Rouhi P, Yang X, Jensen L, Lim S, Feng N, Xue Y, Li X, Larsson O, Ohhashi T, Cao Y. Tumour PDGF-BB expression levels determine dual effects of anti-PDGF drugs on vascular remodelling and metastasis. Nat Commun 2014; 4:2129. [PMID: 23831851 DOI: 10.1038/ncomms3129] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 06/10/2013] [Indexed: 11/09/2022] Open
Abstract
Anti-platelet-derived growth factor (PDGF) drugs are routinely used in front-line therapy for the treatment of various cancers, but the molecular mechanism underlying their dose-dependent impact on vascular remodelling remains poorly understood. Here we show that anti-PDGF drugs significantly inhibit tumour growth and metastasis in high PDGF-BB-producing tumours by preventing pericyte loss and vascular permeability, whereas they promote tumour cell dissemination and metastasis in PDGF-BB-low-producing or PDGF-BB-negative tumours by ablating pericytes from tumour vessels. We show that this opposing effect is due to PDGF-β signalling in pericytes. Persistent exposure of pericytes to PDGF-BB markedly downregulates PDGF-β and inactivation of the PDGF-β signalling decreases integrin α1β1 levels, which impairs pericyte adhesion to extracellular matrix components in blood vessels. Our data suggest that tumour PDGF-BB levels may serve as a biomarker for selection of tumour-bearing hosts for anti-PDGF therapy and unsupervised use of anti-PDGF drugs could potentially promote tumour invasion and metastasis.
Collapse
Affiliation(s)
- Kayoko Hosaka
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 171 77, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Cao Y. Angiogenesis and vascular functions in modulation of obesity, adipose metabolism, and insulin sensitivity. Cell Metab 2013; 18:478-89. [PMID: 24035587 DOI: 10.1016/j.cmet.2013.08.008] [Citation(s) in RCA: 243] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
White and brown adipose tissues are hypervascularized and the adipose vasculature displays phenotypic and functional plasticity to coordinate with metabolic demands of adipocytes. Blood vessels not only supply nutrients and oxygen to nourish adipocytes, they also serve as a cellular reservoir to provide adipose precursor and stem cells that control adipose tissue mass and function. Multiple signaling molecules modulate the complex interplay between the vascular system and the adipocytes. Understanding fundamental mechanisms by which angiogenesis and vasculatures modulate adipocyte functions may provide new therapeutic options for treatment of obesity and metabolic disorders by targeting the adipose vasculature.
Collapse
Affiliation(s)
- Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden; Department of Medicine and Health Sciences, Linköping University, 581 85 Linköping, Sweden.
| |
Collapse
|
48
|
Brown and beige fat: development, function and therapeutic potential. Nat Med 2013; 19:1252-63. [PMID: 24100998 DOI: 10.1038/nm.3361] [Citation(s) in RCA: 1753] [Impact Index Per Article: 146.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Accepted: 08/27/2013] [Indexed: 12/13/2022]
Abstract
Adipose tissue, best known for its role in fat storage, can also suppress weight gain and metabolic disease through the action of specialized, heat-producing adipocytes. Brown adipocytes are located in dedicated depots and express constitutively high levels of thermogenic genes, whereas inducible 'brown-like' adipocytes, also known as beige cells, develop in white fat in response to various activators. The activities of brown and beige fat cells reduce metabolic disease, including obesity, in mice and correlate with leanness in humans. Many genes and pathways that regulate brown and beige adipocyte biology have now been identified, providing a variety of promising therapeutic targets for metabolic disease.
Collapse
|
49
|
Rahman S, Lu Y, Czernik PJ, Rosen CJ, Enerback S, Lecka-Czernik B. Inducible brown adipose tissue, or beige fat, is anabolic for the skeleton. Endocrinology 2013; 154:2687-701. [PMID: 23696565 PMCID: PMC3713216 DOI: 10.1210/en.2012-2162] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
It is known that insulin resistance and type 2 diabetes mellitus are associated with increased fractures and that brown adipose tissue (BAT) counteracts many if not all of the symptoms associated with type 2 diabetes. By the use of FoxC2(AD)(+/Tg) mice, a well-established model for induction of BAT, or beige fat, we present data extending the beneficial action of beige fat to also include a positive effect on bone. FoxC2(AD)(+/Tg) mice are lean and insulin-sensitive and have high bone mass due to increased bone formation associated with high bone turnover. Inducible BAT is linked to activation of endosteal osteoblasts whereas osteocytes have decreased expression of the Sost transcript encoding sclerostin and elevated expression of Rankl. Conditioned media (CM) collected from forkhead box c2 (FOXC2)-induced beige adipocytes activated the osteoblast phenotype and increased levels of phospho-AKT and β-catenin in recipient cells. In osteocytes, the same media decreased Sost expression. Immunodepletion of CM with antibodies against wingless related MMTV integration site 10b (WNT10b) and insulin-like growth factor binding protein 2 (IGFBP2) resulted in the loss of pro-osteoblastic activity, and the loss of increase in the levels of phospho-AKT and β-catenin. Conversely, CM derived from cells overexpressing IGFBP2 or WNT10b restored osteoblastic activity in recipient cells. In conclusion, beige fat secretes endocrine/paracrine activity that is beneficial for the skeleton.
Collapse
Affiliation(s)
- Sima Rahman
- Departments of Orthopaedic Surgery, University ofToledo Health Sciences Campus, Toledo, OH 43614, USA
| | | | | | | | | | | |
Collapse
|
50
|
Tomada N, Tomada I, Botelho F, Pacheco-Figueiredo L, Lopes T, Negrão R, Pestana M, Cruz F. Endothelial function in patients with metabolic syndrome and erectile dysfunction: a question of Angiopoietin imbalance? Andrology 2013; 1:541-8. [DOI: 10.1111/j.2047-2927.2013.00102.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Revised: 03/31/2013] [Accepted: 04/23/2013] [Indexed: 02/04/2023]
Affiliation(s)
| | | | | | | | | | - R. Negrão
- Department of Biochemistry (U38/FCT); Faculty of Medicine of Universidade do Porto; Porto; Portugal
| | - M. Pestana
- Kidney, Urologic and Infectious Diseases Department; Faculty of Medicine of Universidade do Porto; Porto
| | | |
Collapse
|