1
|
Bai K, Long Y, Yuan F, Huang X, Liu P, Hou Y, Zou X, Jiang T, Sun J. Hedyotis diffusa injection modulates the ferroptosis in bladder cancer via CAV1/JUN/VEGFA. Int Immunopharmacol 2025; 147:113925. [PMID: 39765005 DOI: 10.1016/j.intimp.2024.113925] [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: 07/21/2024] [Revised: 11/26/2024] [Accepted: 12/19/2024] [Indexed: 01/29/2025]
Abstract
Hedyotis diffusa Willd. (HDW), a traditional Chinese medicinal plant, exhibits a variety of pharmacological effects and has anticancer potential for a wide range of cancer types; Ferroptosis is a non-apoptosis-regulated cell death induced by iron accumulation and subsequent lipid peroxidation; and there is currently an increasing interest in the therapeutic role of ferroptosis in cancer. However, the effects of HDW on bladder cancer and its underlying molecular mechanisms remain largely unknown. In this study, a combination of in vivo and in vitro experiments, network pharmacology and data mining methods were used to investigate the effects of HDW on BLCA. The results showed that HDW exerted its anticancer activity by inducing ferroptosis in bladder cancer cells. Subsequently, we demonstrated for the first time that HDW induced ferroptosis in vitro and in vivo. To further explore the possible targets of HDW-induced ferroptosis in bladder cancer, we performed network pharmacological analyses, transcriptomic analyses, and single-cell analyses; through integrative analyses, we identified three key pivotal genes associated with iron death, CAV1, VEGFA, and JUN.Mechanistically, we showed that CAV1, VEGFA and JUN are key determinants of HDW-induced ferroptosis in BLCA. Knockdown of target genes altered the anticancer effects of HDW in 5637 and T24 cells. In conclusion, our data show for the first time that HDW exerts its anticancer effects on BLCA through CAV1, VEGFA and JUN gene-induced ferroptosis. This is expected to provide a promising compound for bladder cancer therapy.
Collapse
Affiliation(s)
- Kaiping Bai
- Department of Urology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, No. 1678 Dongfang Road, Pudong New Area, Shanghai 200127, China.
| | - Yanxi Long
- Department of Anesthesiology, International Peace Maternity & Child Health Hospital of China Welfare Institute, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Fei Yuan
- Department of Urology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, No. 1678 Dongfang Road, Pudong New Area, Shanghai 200127, China.
| | - Xiaoling Huang
- Department of Urology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, No. 1678 Dongfang Road, Pudong New Area, Shanghai 200127, China.
| | - Pengtao Liu
- Department of Urology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, No. 1678 Dongfang Road, Pudong New Area, Shanghai 200127, China.
| | - Yanping Hou
- Department of Urology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, No. 1678 Dongfang Road, Pudong New Area, Shanghai 200127, China.
| | - Xiangyu Zou
- Department of Urology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, No. 1678 Dongfang Road, Pudong New Area, Shanghai 200127, China.
| | - Tao Jiang
- Department of Andrology and Sexual Medicine, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian, Liaoning 116000, China.
| | - Jie Sun
- Department of Urology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, No. 1678 Dongfang Road, Pudong New Area, Shanghai 200127, China.
| |
Collapse
|
2
|
Chen CH, Hsu SY, Yu WJ, Chiang CS, Yu CF. Distinct roles of small extracellular vesicles from resident and infiltrating macrophages on glioma growth and mobility. J Cancer 2025; 16:969-981. [PMID: 39781357 PMCID: PMC11705045 DOI: 10.7150/jca.103595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/15/2024] [Indexed: 01/12/2025] Open
Abstract
Previous studies revealed that tumor-associated macrophages/microglia (TAMs) promoted glioma invasiveness during tumor progression and after radiotherapy. However, the communication of TAMs with tumor cells remains unclear. This study aimed to examine the role of small extracellular vesicles (sEVs) derived from TAMs in TAMs-mediated brain tumor invasion. This study utilized BV2 and RAW264.7 cell lines representing resident and infiltrating macrophages, respectively, to unveil their effect on tumor cells. Purified sEVs from BV2 and RAW264.7 were validated by nanoparticle track analysis (NTA), transmission electron microscopy (TEM), and western blotting for sEV markers. The effect of sEVs on the murine astrocytoma tumor cell line ALTS1C1 was examined on cell proliferation, migration, and gene expression. The results showed that ALTS1C1 cells effectively engulfed sEVs purified from BV2 and RAW264.7. Only BV2-derived sEVs promoted cell proliferation and were dose-dependent. Further, morphological changes in ALTS1C1 cells were observed after incubation with BV2-derived sEVs, which was associated with enhancing cell migration. BV2-mediated glioma proliferation and mobility were related to the upregulation of vascular endothelial growth factor (VEGF) and downregulation of death effector domain-containing protein (DEDD) gene expression. This study demonstrates the distinct function of sEVs of resident macrophages on glioma cell invasion and reveals the mechanism underlying microglia-mediated tumor progression. These findings suggested resident microglia is the potential therapeutic target for TAMs-induced brain tumor invasiveness.
Collapse
Affiliation(s)
- Chu-Hsuan Chen
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsin-Chu, Taiwan
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsin-Chu, Taiwan
| | - Sheng-Yun Hsu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsin-Chu, Taiwan
| | - Wen-Jui Yu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsin-Chu, Taiwan
| | - Chi-Shiun Chiang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsin-Chu, Taiwan
- Institute of Nuclear Engineering and Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
- The BNCT Research Center, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Ching-Fang Yu
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan, 33302, Taiwan
- Research Center for Radiation Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Radiation Oncology, Chang Gung Memorial Hospital Linkou Branch, Taoyuan 33382, Taiwan
| |
Collapse
|
3
|
Yu D, Lu Z, Chong Y. Integrins as a bridge between bacteria and cells: key targets for therapeutic wound healing. BURNS & TRAUMA 2024; 12:tkae022. [PMID: 39015251 PMCID: PMC11250365 DOI: 10.1093/burnst/tkae022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 12/17/2023] [Accepted: 04/22/2024] [Indexed: 07/18/2024]
Abstract
Integrins are heterodimers composed of α and β subunits that are bonded through non-covalent interactions. Integrins mediate the dynamic connection between extracellular adhesion molecules and the intracellular actin cytoskeleton. Integrins are present in various tissues and organs where these heterodimers participate in diverse physiological and pathological responses at the molecular level in living organisms. Wound healing is a crucial process in the recovery from traumatic diseases and comprises three overlapping phases: inflammation, proliferation and remodeling. Integrins are regulated during the entire wound healing process to enhance processes such as inflammation, angiogenesis and re-epithelialization. Prolonged inflammation may result in failure of wound healing, leading to conditions such as chronic wounds. Bacterial colonization of a wound is one of the primary causes of chronic wounds. Integrins facilitate the infectious effects of bacteria on the host organism, leading to chronic inflammation, bacterial colonization, and ultimately, the failure of wound healing. The present study investigated the role of integrins as bridges for bacteria-cell interactions during wound healing, evaluated the role of integrins as nodes for bacterial inhibition during chronic wound formation, and discussed the challenges and prospects of using integrins as therapeutic targets in wound healing.
Collapse
Affiliation(s)
- Dong Yu
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368 Hanjiang Middle Road, Yangzhou 225000, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368 Hanjiang Middle Road, Yangzhou 225000, Jiangsu, China
| | - Zhaoyu Lu
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368 Hanjiang Middle Road, Yangzhou 225000, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368 Hanjiang Middle Road, Yangzhou 225000, Jiangsu, China
| | - Yang Chong
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368 Hanjiang Middle Road, Yangzhou 225000, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368 Hanjiang Middle Road, Yangzhou 225000, Jiangsu, China
| |
Collapse
|
4
|
Yu D, Lu Z, Nie F, Chong Y. Integrins regulation of wound healing processes: insights for chronic skin wound therapeutics. Front Cell Infect Microbiol 2024; 14:1324441. [PMID: 38505290 PMCID: PMC10949986 DOI: 10.3389/fcimb.2024.1324441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 02/21/2024] [Indexed: 03/21/2024] Open
Abstract
Integrins are heterodimers composed of non-covalently associated alpha and beta subunits that mediate the dynamic linkage between extracellular adhesion molecules and the intracellular actin cytoskeleton. Integrins are present in various tissues and organs and are involved in different physiological and pathological molecular responses in vivo. Wound healing is an important process in the recovery from traumatic diseases and consists of three overlapping phases: inflammation, proliferation, and remodeling. Integrin regulation acts throughout the wound healing process to promote wound healing. Prolonged inflammation may lead to failure of wound healing, such as wound chronicity. One of the main causes of chronic wound formation is bacterial colonization of the wound. In this review, we review the role of integrins in the regulation of wound healing processes such as angiogenesis and re-epithelialization, as well as the role of integrins in mediating bacterial infections during wound chronicity, and the challenges and prospects of integrins as therapeutic targets for infected wound healing.
Collapse
Affiliation(s)
- Dong Yu
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Zhaoyu Lu
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Fengsong Nie
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yang Chong
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
5
|
Li H, Jia C, Li C, Wang Y, Du W, Jiang H. Anthracycline chemicals with anthracyclinone structure exert antitumor effects by inhibiting angiogenesis and lymphangiogenesis in a xenografted gastric tumor model. Gastric Cancer 2023; 26:863-877. [PMID: 37344705 DOI: 10.1007/s10120-023-01412-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/14/2023] [Indexed: 06/23/2023]
Abstract
BACKGROUND It is vital to screen or develop alternative therapeutic drugs with higher curative characteristics and fewer side effects for the clinical treatment of gastric cancer. METHODS Gastric cancer cells were exposed to different auramycin G doses while determining the impact on cell viability, migration, and invasion. Then the antitumor effects of auramycin G, 5-fluorouracil (5-Fu) and their combination were evaluated. Furthermore, the molecular mechanisms of angiogenesis and lymphangiogenesis regulated by auramycin G and its analogs were investigated. RESULTS Auramycin G inhibited cell viability in a dose-dependent manner, with a 50% inhibitory concentration of 23.72 ± 6.36 mg/L and 32.54 ± 5.91 mg/L for AGS and MGC803 cells, respectively. The migration and invasion of gastric cancer cells were significantly inhibited by 10 mg/L auramycin G, which was consistent with the down-regulation of the VEGFR2-VEGFA-pPI3K-pAkt-pErk1 and VEGFR3-VEGFC-pPI3K-pAkt-pmTOR proteins. Notably, the average tumor weights were significantly reduced in both the auramycin G (2.21 ± 0.45 g) of 50 mg/kg body weight and auramycin G + 5-fluorouracil (5-Fu) groups (1.33 ± 0.28 g), compared with the control (3.73 ± 0.56 g). Considering that auramycin G decreased the growth of blood and lymphatic vessels while reducing the degree of tumor malignancy, it effectively suppressed tumors by regulating the angiogenic and lymphangiogenic pathways. CONCLUSION The present study confirmed that auramycin G displayed a prominent antitumor activity in gastric tumor models, both in vitro and in vivo. Moreover, it was confirmed that auramycin G played a specific role in certain gastric cancer cell types, while the mechanism was validated to be associated with angiogenesis- and lymphangiogenesis-related pathway suppression.
Collapse
Affiliation(s)
- Huiying Li
- College of Biological Sciences and Technology, Beijing Key Laboratory of Food Processing and Safety in Forestry, Beijing Forestry University, Beijing, People's Republic of China
| | - Cuicui Jia
- College of Biological Sciences and Technology, Beijing Key Laboratory of Food Processing and Safety in Forestry, Beijing Forestry University, Beijing, People's Republic of China
| | - Chaonan Li
- College of Biological Sciences and Technology, Beijing Key Laboratory of Food Processing and Safety in Forestry, Beijing Forestry University, Beijing, People's Republic of China
| | - Yang Wang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, People's Republic of China
| | - Weimin Du
- College of Biological Sciences and Technology, Beijing Key Laboratory of Food Processing and Safety in Forestry, Beijing Forestry University, Beijing, People's Republic of China
| | - Hongpeng Jiang
- Department of General Surgery, Beijing Key Laboratory of Cancer Invasion and Metastasis Research and National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China.
| |
Collapse
|
6
|
Vofo BN, Chowers I. Suppressing Inflammation for the Treatment of Diabetic Retinopathy and Age-Related Macular Degeneration: Dazdotuftide as a Potential New Multitarget Therapeutic Candidate. Biomedicines 2023; 11:1562. [PMID: 37371657 PMCID: PMC10295757 DOI: 10.3390/biomedicines11061562] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
Diabetic retinopathy (DR) and age-related macular degeneration (AMD) are major causes of blindness globally. The primary treatment option for DME and neovascular AMD (nAMD) is anti-vascular endothelial growth factor (VEGF) compounds, but this treatment modality often yields insufficient results, and monthly injections can place a burden on the health system and patients. Although various inflammatory pathways and mediators have been recognized as key players in the development of DR and AMD, there are limited treatment options targeting these pathways. Molecular pathways that are interlinked, or triggers of multiple inflammatory pathways, could be promising targets for drug development. This review focuses on the role of inflammation in the pathogenesis of DME and AMD and presents current anti-inflammatory compounds, as well as a potential multitarget anti-inflammatory compound (dazdotuftide) that could be a candidate treatment option for the management of DME and AMD.
Collapse
Affiliation(s)
| | - Itay Chowers
- Department of Ophthalmology, Hadassah—Hebrew University Medical Center, Jerusalem 91120, Israel;
| |
Collapse
|
7
|
Bi S, Chen W, Fang Y, Wang Y, Zhang Q, Guo H, Ju H, Liu Y. Cancer Cell-Selective Membrane Receptor Clustering Driven by VEGF Secretion for In Vivo Therapy. J Am Chem Soc 2023; 145:5041-5052. [PMID: 36815672 DOI: 10.1021/jacs.2c10428] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Clustering of cell membrane receptors regulates cell behaviors. Although receptor clustering plans have achieved wide applications in cancer therapy, it still remains challenging to manipulate receptor clustering selectively for cancer cells with little influence on normal cells. Here, we design a Raji cell Selective MAnipulation of Receptor Clustering (SMARC) strategy for CD20, which is driven by endogenous secretion of Raji cells. Retractable DNA nanostrings with repeating hairpin-structured units are anchored to the cell membrane CD20, which contract in response to Raji cell-secreted vascular endothelial growth factor (VEGF) with corresponding CD20 clustering. The contraction of DNA nanostrings is intensified via a VEGF amplifier including DNA cyclic reactions to continuously trigger the foldings of hairpin-structured units in DNA nanostrings. The SMARC strategy shows selective and efficient apoptosis of Raji cells with little interference to normal B cells and demonstrates good in vivo therapeutic efficacy, which provides a promising tool for precise cancer therapy.
Collapse
Affiliation(s)
- Shiyi Bi
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Wei Chen
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing 210008, China
| | - Yanyun Fang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yingfei Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Qing Zhang
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing 210008, China
| | - Hongqian Guo
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing 210008, China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Ying Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.,Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China
| |
Collapse
|
8
|
Patnam M, Dommaraju SR, Masood F, Herbst P, Chang JH, Hu WY, Rosenblatt MI, Azar DT. Lymphangiogenesis Guidance Mechanisms and Therapeutic Implications in Pathological States of the Cornea. Cells 2023; 12:319. [PMID: 36672254 PMCID: PMC9856498 DOI: 10.3390/cells12020319] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/22/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Corneal lymphangiogenesis is one component of the neovascularization observed in several inflammatory pathologies of the cornea including dry eye disease and corneal graft rejection. Following injury, corneal (lymph)angiogenic privilege is impaired, allowing ingrowth of blood and lymphatic vessels into the previously avascular cornea. While the mechanisms underlying pathological corneal hemangiogenesis have been well described, knowledge of the lymphangiogenesis guidance mechanisms in the cornea is relatively scarce. Various signaling pathways are involved in lymphangiogenesis guidance in general, each influencing one or multiple stages of lymphatic vessel development. Most endogenous factors that guide corneal lymphatic vessel growth or regression act via the vascular endothelial growth factor C signaling pathway, a central regulator of lymphangiogenesis. Several exogenous factors have recently been repurposed and shown to regulate corneal lymphangiogenesis, uncovering unique signaling pathways not previously known to influence lymphatic vessel guidance. A strong understanding of the relevant lymphangiogenesis guidance mechanisms can facilitate the development of targeted anti-lymphangiogenic therapeutics for corneal pathologies. In this review, we examine the current knowledge of lymphatic guidance cues, their regulation of inflammatory states in the cornea, and recently discovered anti-lymphangiogenic therapeutic modalities.
Collapse
Affiliation(s)
- Mehul Patnam
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Sunil R. Dommaraju
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Faisal Masood
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Paula Herbst
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Jin-Hong Chang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Wen-Yang Hu
- Department of Urology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Mark I. Rosenblatt
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Dimitri T. Azar
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
9
|
Patel SA, Nilsson MB, Le X, Cascone T, Jain RK, Heymach JV. Molecular Mechanisms and Future Implications of VEGF/VEGFR in Cancer Therapy. Clin Cancer Res 2023; 29:30-39. [PMID: 35969170 DOI: 10.1158/1078-0432.ccr-22-1366] [Citation(s) in RCA: 178] [Impact Index Per Article: 89.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/28/2022] [Accepted: 08/03/2022] [Indexed: 02/06/2023]
Abstract
Angiogenesis, the sprouting of new blood vessels from existing vessels, is one of six known mechanisms employed by solid tumors to recruit blood vessels necessary for their initiation, growth, and metastatic spread. The vascular network within the tumor facilitates the transport of nutrients, oxygen, and immune cells and is regulated by pro- and anti-angiogenic factors. Nearly four decades ago, VEGF was identified as a critical factor promoting vascular permeability and angiogenesis, followed by identification of VEGF family ligands and their receptors (VEGFR). Since then, over a dozen drugs targeting the VEGF/VEGFR pathway have been approved for approximately 20 solid tumor types, usually in combination with other therapies. Initially designed to starve tumors, these agents transiently "normalize" tumor vessels in preclinical and clinical studies, and in the clinic, increased tumor blood perfusion or oxygenation in response to these agents is associated with improved outcomes. Nevertheless, the survival benefit has been modest in most tumor types, and there are currently no biomarkers in routine clinical use for identifying which patients are most likely to benefit from treatment. However, the ability of these agents to reprogram the immunosuppressive tumor microenvironment into an immunostimulatory milieu has rekindled interest and has led to the FDA approval of seven different combinations of VEGF/VEGFR pathway inhibitors with immune checkpoint blockers for many solid tumors in the past 3 years. In this review, we discuss our understanding of the mechanisms of response and resistance to blocking VEGF/VEGFR, and potential strategies to develop more effective therapeutic approaches.
Collapse
Affiliation(s)
- Sonia A Patel
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Monique B Nilsson
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiuning Le
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tina Cascone
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - John V Heymach
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
10
|
Sezer O, Nursal AF, Kuruca N, Yigit S. The effect of a 18 bp deletion/insertion variant of VEGF gene on the FMF development. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2023; 42:296-307. [PMID: 36215175 DOI: 10.1080/15257770.2022.2127766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
Objective: Familial Mediterranean fever (FMF) is one of the most common inherited autoinflammatory diseases. Angiogenesis is a feature of inflammatory activation and part of pathogenic processes in autoimmune diseases. Therefore, this study aimed to investigate the role of the Vascular endothelial growth factor (VEGF) gene insertion/deletion (I/D) functional variant in FMF Turkish patients. Methods: MEFV gene mutations were detected in all patients. The FMF patients (N:105) and the healthy controls (N:100) were genotyped for the VEGF I/D variant using PCR followed by agarose gel electrophoresis. The results were statistically analyzed by calculating the odds ratios (OR) and their 95% confidence intervals (95% CI) using the χ2-tests. Results: The mean age of patients was 25.46 ± 10.09. Fifty-nine patients (56.2%) had two or more MEFV gene mutations. The most common MEFV mutation was M694V/M694V. The VEGF I/D variant genotype distribution exhibited a statistically significant difference between the patients and the controls. VEGF I/D genotype was higher in controls compared to patients, while D/D genotype was higher in patients compared to the controls (p = 0.003, p = 0.013, respectively). When we examined the clinical findings, joint pain was more common in patients with VEGF D/D and I/D genotypes compared to I/I genotype (p = 0.043). Although not statistically significant, the most common genotype in patients with two or more MEFV mutations was VEGF D/D (28.6%). Conclusion: The results provided evidence supporting that the D/D genotype of the VEGF I/D variant is associated with an increased risk of FMF in a group of Turkish populations.
Collapse
Affiliation(s)
- Ozlem Sezer
- Department of Medical Genetics, Samsun Education and Research Hospital, University of Health Sciences, Samsun, Turkey
| | - Ayse Feyda Nursal
- Department of Medical Genetics, Hitit University, Faculty of Medicine, Corum, Turkey
| | - Nilufer Kuruca
- Department of Pathology, Ondokuz Mayis University, Faculty of Veterinary, Samsun, Turkey
| | - Serbulent Yigit
- Department of Genetics, Ondokuz Mayis University, Faculty of Veterinary, Samsun, Turkey
| |
Collapse
|
11
|
Abstract
The term "lectin" is derived from the Latin word lego- (aggregate) (Boyd & Shapleigh, 1954). Indeed, lectins' folds can flexibly alter their pocket structures just like Lego blocks, which enables them to grab a wide-variety of substances. Thus, this useful fold is well-conserved among various organisms. Through evolution, prototypic soluble lectins acquired transmembrane regions and signaling motifs to become C-type lectin receptors (CLRs). While CLRs seem to possess certain intrinsic affinity to self, some CLRs adapted to efficiently recognize glycoconjugates present in pathogens as pathogen-associated molecular patterns (PAMPs) and altered self. CLRs further extended their diversity to recognize non-glycosylated targets including pathogens and self-derived molecules. Thus, CLRs seem to have developed to monitor the internal/external stresses to maintain homeostasis by sensing various "unfamiliar" targets. In this review, we will summarize recent advances in our understanding of CLRs, their ligands and functions and discuss future perspectives.
Collapse
Affiliation(s)
- Carla Guenther
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan; Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Masamichi Nagae
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan; Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Sho Yamasaki
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan; Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan.
| |
Collapse
|
12
|
Pérez-García J, Martel-Martel A, García-Vallés P, Corchete LA, García JL, Gestoso-Uzal N, Vidal-Tocino R, Blanco Ó, Méndez L, Sánchez-Martín M, Fuentes M, Herrero AB, Holowatyj AN, Perea J, González-Sarmiento R. Recurrent NOMO1 Gene Deletion Is a Potential Clinical Marker in Early-Onset Colorectal Cancer and Is Involved in the Regulation of Cell Migration. Cancers (Basel) 2022; 14:4029. [PMID: 36011023 PMCID: PMC9406593 DOI: 10.3390/cancers14164029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
The incidence of early-onset colorectal cancer (EOCRC; age younger than 50 years) has been progressively increasing over the last decades globally, with causes unexplained. A distinct molecular feature of EOCRC is that compared with cases of late-onset colorectal cancer, in EOCRC cases, there is a higher incidence of Nodal Modulator 1 (NOMO1) somatic deletions. However, the mechanisms of NOMO1 in early-onset colorectal carcinogenesis are currently unknown. In this study, we show that in 30% of EOCRCs with heterozygous deletion of NOMO1, there were pathogenic mutations in this gene, suggesting that NOMO1 can be inactivated by deletion or mutation in EOCRC. To study the role of NOMO1 in EOCRC, CRISPR/cas9 technology was employed to generate NOMO1 knockout HCT-116 (EOCRC) and HS-5 (bone marrow) cell lines. NOMO1 loss in these cell lines did not perturb Nodal pathway signaling nor cell proliferation. Expression microarrays, RNA sequencing, and protein expression analysis by LC-IMS/MS showed that NOMO1 inactivation deregulates other signaling pathways independent of the Nodal pathway, such as epithelial-mesenchymal transition and cell migration. Significantly, NOMO1 loss increased the migration capacity of CRC cells. Additionally, a gut-specific conditional NOMO1 KO mouse model revealed no subsequent tumor development in mice. Overall, these findings suggest that NOMO1 could play a secondary role in early-onset colorectal carcinogenesis because its loss increases the migration capacity of CRC cells. Therefore, further study is warranted to explore other signalling pathways deregulated by NOMO1 loss that may play a significant role in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Jésica Pérez-García
- Institute of Biomedical Research of Salamanca (IBSAL), SACYL-University of Salamanca-CSIC, 37007 Salamanca, Spain
- Molecular Medicine Unit, Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
- Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-CSIC, 37007 Salamanca, Spain
| | - Abel Martel-Martel
- Institute of Biomedical Research of Salamanca (IBSAL), SACYL-University of Salamanca-CSIC, 37007 Salamanca, Spain
- Molecular Medicine Unit, Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
- Medical Oncology Department, Complejo Asistencial Universitario de Salamanca-IBSAL, 37007 Salamanca, Spain
| | - Paula García-Vallés
- Institute of Biomedical Research of Salamanca (IBSAL), SACYL-University of Salamanca-CSIC, 37007 Salamanca, Spain
- Molecular Medicine Unit, Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
| | - Luis A. Corchete
- Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-CSIC, 37007 Salamanca, Spain
- Hematology Department, Complejo Asistencial Universitario de Salamanca-IBSAL, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Juan L. García
- Institute of Biomedical Research of Salamanca (IBSAL), SACYL-University of Salamanca-CSIC, 37007 Salamanca, Spain
- Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-CSIC, 37007 Salamanca, Spain
| | - Nerea Gestoso-Uzal
- Institute of Biomedical Research of Salamanca (IBSAL), SACYL-University of Salamanca-CSIC, 37007 Salamanca, Spain
- Molecular Medicine Unit, Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
| | - Rosario Vidal-Tocino
- Institute of Biomedical Research of Salamanca (IBSAL), SACYL-University of Salamanca-CSIC, 37007 Salamanca, Spain
- Medical Oncology Department, Complejo Asistencial Universitario de Salamanca-IBSAL, 37007 Salamanca, Spain
| | - Óscar Blanco
- Institute of Biomedical Research of Salamanca (IBSAL), SACYL-University of Salamanca-CSIC, 37007 Salamanca, Spain
- Anatomy Pathology Service, University Hospital of Salamanca, 37007 Salamanca, Spain
| | - Lucía Méndez
- Institute of Biomedical Research of Salamanca (IBSAL), SACYL-University of Salamanca-CSIC, 37007 Salamanca, Spain
- Transgenic Service, Nucleus, University of Salamanca, 37007 Salamanca, Spain
| | - Manuel Sánchez-Martín
- Institute of Biomedical Research of Salamanca (IBSAL), SACYL-University of Salamanca-CSIC, 37007 Salamanca, Spain
- Transgenic Service, Nucleus, University of Salamanca, 37007 Salamanca, Spain
| | - Manuel Fuentes
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain
- Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain
| | - Ana B. Herrero
- Institute of Biomedical Research of Salamanca (IBSAL), SACYL-University of Salamanca-CSIC, 37007 Salamanca, Spain
- Molecular Medicine Unit, Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
- Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-CSIC, 37007 Salamanca, Spain
| | - Andreana N. Holowatyj
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - José Perea
- Institute of Biomedical Research of Salamanca (IBSAL), SACYL-University of Salamanca-CSIC, 37007 Salamanca, Spain
| | - Rogelio González-Sarmiento
- Institute of Biomedical Research of Salamanca (IBSAL), SACYL-University of Salamanca-CSIC, 37007 Salamanca, Spain
- Molecular Medicine Unit, Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
- Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-CSIC, 37007 Salamanca, Spain
| |
Collapse
|
13
|
Wang L, Wang X, Guo E, Mao X, Miao S. Emerging roles of platelets in cancer biology and their potential as therapeutic targets. Front Oncol 2022; 12:939089. [PMID: 35936717 PMCID: PMC9355257 DOI: 10.3389/fonc.2022.939089] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/29/2022] [Indexed: 12/15/2022] Open
Abstract
The main role of platelets is to control bleeding and repair vascular damage via thrombosis. They have also been implicated to promote tumor metastasis through platelet-tumor cell interactions. Platelet-tumor cell interactions promote tumor cell survival and dissemination in blood circulation. Tumor cells are known to induce platelet activation and alter platelet RNA profiles. Liquid biopsies based on tumor-educated platelet biomarkers can detect tumors and correlate with prognosis, personalized therapy, treatment monitoring, and recurrence prediction. Platelet-based strategies for cancer prevention and tumor-targeted therapy include developing drugs that target platelet receptors, interfere with the release of platelet particles, inhibit platelet-specific enzymes, and utilize platelet-derived “nano-platelets” as a targeted drug delivery platform for tumor therapy. This review elaborates on platelet-tumor cell interactions and the molecular mechanisms and discusses future research directions for platelet-based liquid biopsy techniques and platelet-targeted anti-tumor strategies.
Collapse
Affiliation(s)
- Lei Wang
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Erliang Guo
- Department of Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xionghui Mao
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
- *Correspondence: Xionghui Mao, ; Susheng Miao,
| | - Susheng Miao
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
- *Correspondence: Xionghui Mao, ; Susheng Miao,
| |
Collapse
|
14
|
Lee SH, Park NR, Kim JE. Bioinformatics of Differentially Expressed Genes in Phorbol 12-Myristate 13-Acetate-Induced Megakaryocytic Differentiation of K562 Cells by Microarray Analysis. Int J Mol Sci 2022; 23:ijms23084221. [PMID: 35457039 PMCID: PMC9031040 DOI: 10.3390/ijms23084221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/31/2022] [Accepted: 04/09/2022] [Indexed: 01/27/2023] Open
Abstract
Megakaryocytes are large hematopoietic cells present in the bone marrow cavity, comprising less than 0.1% of all bone marrow cells. Despite their small number, megakaryocytes play important roles in blood coagulation, inflammatory responses, and platelet production. However, little is known about changes in gene expression during megakaryocyte maturation. Here we identified the genes whose expression was changed during K562 leukemia cell differentiation into megakaryocytes using an Affymetrix GeneChip microarray to determine the multifunctionality of megakaryocytes. K562 cells were differentiated into mature megakaryocytes by treatment for 7 days with phorbol 12-myristate 13-acetate, and a microarray was performed using RNA obtained from both types of cells. The expression of 44,629 genes was compared between K562 cells and mature megakaryocytes, and 954 differentially expressed genes (DEGs) were selected based on a p-value < 0.05 and a fold change >2. The DEGs was further functionally classified using five major megakaryocyte function-associated clusters—inflammatory response, angiogenesis, cell migration, extracellular matrix, and secretion. Furthermore, interaction analysis based on the STRING database was used to generate interactions between the proteins translated from the DEGs. This study provides information on the bioinformatics of the DEGs in mature megakaryocytes after K562 cell differentiation.
Collapse
Affiliation(s)
- Seung-Hoon Lee
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (S.-H.L.); (N.R.P.)
- BK21 Four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, Kyungpook National University, Daegu 41944, Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu 41944, Korea
| | - Na Rae Park
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (S.-H.L.); (N.R.P.)
- Cell and Matrix Research Institute, Kyungpook National University, Daegu 41944, Korea
| | - Jung-Eun Kim
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (S.-H.L.); (N.R.P.)
- BK21 Four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, Kyungpook National University, Daegu 41944, Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu 41944, Korea
- Correspondence: ; Tel.: +82-53-420-4949
| |
Collapse
|
15
|
Wang X, Geng B, Wang H, Wang S, Zhao D, He J, Lu F, An J, Wang C, Xia Y. Fluid shear stress-induced down-regulation of microRNA-140-5p promotes osteoblast proliferation by targeting VEGFA via the ERK5 pathway. Connect Tissue Res 2022; 63:156-168. [PMID: 33588662 DOI: 10.1080/03008207.2021.1891228] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE Fluid shear stress (FSS) plays a critical role in osteoblast proliferation. However, the role of miRNA in osteoblast proliferation induced by FSS and the possible molecular mechanisms remain to be defined. The aim of the present study was to investigate whether miR-140-5p regulates osteoblast proliferation under FSS and its molecular mechanism. MATERIALS AND METHODS miR-140-5p expression was measured by qRT-PCR. Western blot was used to measure the expressions of P-ERK1/2, ERK1/2, P-ERK5 and ERK5. The levels of VEGFA, PCNA, CDK4 and Cyclin D1 were identified through qRT-PCR and western blot, respectively. Cell proliferation was detected by CCK-8 assay and EdU labeling assay. Dual-luciferase reporter assay was used to validate the target of miR-140-5p. RESULTS miR-140-5p was significantly down-regulated when MC3T3-E1 cells were exposed to FSS. We then confirmed that up-regulation of miR-140-5p inhibited and down-regulation of miR-140-5p promoted osteoblast proliferation. In addition, FSS promotes osteoblast proliferation via down-regulating miR-140-5p. Luciferase reporter assay demonstrated that VEGFA is a direct target of miR-140-5p. Furthermore, transfection of mimic-140-5p inhibited the up-regulation of VEGFA protein level induced by FSS, suggesting that FSS regulates VEGFA protein expression via miR-140-5p. Further investigations demonstrated that VEGFA could promote osteoblast proliferation. Lastly, we demonstrated that miR-140-5p regulates osteoblast proliferation and ERK5 activation through VEGFA. CONCLUSIONS Our study demonstrates that FSS-induced the down-regulation of miR-140-5p promotes osteoblast proliferation through activing VEGFA/ERK5 signaling pathway. These findings may provide a novel mechanism of FSS-induced osteoblast proliferation and offer a new avenue to further investigate osteogenesis induced by mechanical loading.
Collapse
Affiliation(s)
- Xingwen Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Bin Geng
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Hong Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Shenghong Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Dacheng Zhao
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Jinwen He
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Fan Lu
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Jiangdong An
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Cuifang Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Yayi Xia
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| |
Collapse
|
16
|
Li J, Zhang Y, Zeng X, Cheng Y, Tang L, Hong D, Yang X. Lycopene ameliorates insulin resistance and increases muscle capillary density in aging via activation of SIRT1. J Nutr Biochem 2021; 99:108862. [PMID: 34530111 DOI: 10.1016/j.jnutbio.2021.108862] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 02/14/2021] [Accepted: 07/25/2021] [Indexed: 11/27/2022]
Abstract
Lycopene (Ly) is a kind of hydrocarbon, which belongs to the family of tetraterpene carotene and exists in red fruits and vegetables. The decrease of capillary density and blood flow with age is a significant reason for the increase of mortality and morbidity. Herein, our study aims to explore the effects of Ly (a bioactive food compound) on vascular aging in vitro and in vivo and its potential mechanisms. The cytological results showed that Ly could promote the proliferation of human umbilical vein endothelial cell (HUVECs) and enhance the ability of HUVECs to form capillary-like structures. Furthermore, the expression of SIRT1 in aged HUVECs was up-regulated. In vivo, aging rats showed signs of insulin resistance and blood vessel damage. Additionally, the capillary density and blood flow were reduced during the vascular aging process in both D-gal-induced and naturally aging muscle. However, when Ly was given, these conditions could be reversed. Simultaneously, the contents of ATP, lactic acid and pyruvic acid were determined, and it was found that Ly could promote angiogenesis by increasing the utilization rate of glucose and promoting energy metabolism. Finally, in the insulin resistance cell model, we knocked down the SIRT1 and administrated with Ly, and found that it couldn't restore insulin transdution. In conclusion, all the data in this study demonstrate that Ly could reactivate SIRT1 and improve insulin resistance, which was a reversible cause of vascular aging.
Collapse
Affiliation(s)
- Jing Li
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, P. R. China; Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, P. R. China
| | - Yingjiang Zhang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, P. R. China
| | - Xin Zeng
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, P. R. China
| | - Yahong Cheng
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, P. R. China
| | - Liu Tang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, P. R. China
| | - Ding Hong
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, P. R. China.
| | - Xiaolong Yang
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, P. R. China.
| |
Collapse
|
17
|
Gao B, Feng C, Chai F, Wei S, Hong N, Ye Y, Wang Y, Cheng J. CT-detected extramural venous invasion-related gene signature for the overall survival prediction in patients with gastric cancer. Cancer Med 2021; 10:7816-7830. [PMID: 34510798 PMCID: PMC8559479 DOI: 10.1002/cam4.4266] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 07/25/2021] [Accepted: 09/01/2021] [Indexed: 12/17/2022] Open
Abstract
Background Computed tomography (CT)‐detected extramural venous invasion (EMVI) has been identified as an independent factor that can be used for risk stratification and prediction of prognosis in patients with gastric cancer (GC). Overall survival (OS) is identified as the most important prognostic indicator for GC patients. However, the molecular mechanism of EMVI development and its potential relationship with OS in GC are not fully understood. In this radiogenomics‐based study, we sought to investigate the molecular mechanism underlying CT‐detected EMVI in patients with GC, and aimed to construct a genomic signature based on EMVI‐related genes with the goal of using this signature to predict the OS. Materials and Methods Whole mRNA genome sequencing of frozen tumor samples from 13 locally advanced GC patients was performed to identify EMVI‐related genes. EMVI‐prognostic hub genes were selected based on overlapping EMVI‐related differentially expressed genes and OS‐related genes, using a training cohort of 176 GC patients who were included in The Cancer Genome Atlas database. Another 174 GC patients from this database comprised the external validation cohort. A risk stratification model using a seven‐gene signature was constructed through the use of a least absolute shrinkage and selection operator Cox regression model. Results Patients with high risk score showed significantly reduced OS (training cohort, p = 1.143e‐04; validation cohort, p = 2.429e‐02). Risk score was an independent predictor of OS in multivariate Cox regression analyses (training cohort, HR = 2.758; 95% CI: 1.825–4.169; validation cohort, HR = 2.173; 95% CI: 1.347–3.505; p < 0.001 for both). Gene functions/pathways of the seven‐gene signature mainly included cell proliferation, cell adhesion, regulation of metal ion transport, and epithelial to mesenchymal transition. Conclusions A CT‐detected EMVI‐related gene model could be used to predict the prognosis in GC patients, potentially providing clinicians with additional information regarding appropriate therapeutic strategy and medical decision‐making.
Collapse
Affiliation(s)
- Bo Gao
- Department of General Surgery, Peking University People's Hospital, Beijing, China
| | - Caizhen Feng
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Fan Chai
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Shengcai Wei
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Nan Hong
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Yingjiang Ye
- Department of Gastrointestinal Surgery, Peking University People's Hospital, Beijing, China
| | - Yi Wang
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Jin Cheng
- Department of Radiology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
18
|
Xu S, Zhang T, Cao Z, Zhong W, Zhang C, Li H, Song J. Integrin-α9β1 as a Novel Therapeutic Target for Refractory Diseases: Recent Progress and Insights. Front Immunol 2021; 12:638400. [PMID: 33790909 PMCID: PMC8005531 DOI: 10.3389/fimmu.2021.638400] [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: 12/06/2020] [Accepted: 02/26/2021] [Indexed: 12/12/2022] Open
Abstract
Integrins refer to heterodimers consisting of subunits α and β. They serve as receptors on cell membranes and interact with extracellular ligands to mediate intracellular molecular signals. One of the least-studied members of the integrin family is integrin-α9β1, which is widely distributed in various human tissues and organs. Integrin-α9β1 regulates the physiological state of cells through a variety of complex signaling pathways to participate in the specific pathological processes of some intractable diseases. In recent years, an increasing amount of research has focused on the role of α9β1 in the molecular mechanisms of different refractory diseases and its promising potential as a therapeutic target. Accordingly, this review introduces and summarizes recent research related to integrin-α9β1, describes the synergistic functions of α9β1 and its corresponding ligands in cancer, autoimmune diseases, nerve injury and thrombosis and, more importantly, highlights the potential of α9β1 as a distinctive target for the treatment of these intractable diseases.
Collapse
Affiliation(s)
- Shihan Xu
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Tingwei Zhang
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Zhengguo Cao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Periodontology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Wenjie Zhong
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Chuangwei Zhang
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Han Li
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jinlin Song
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| |
Collapse
|
19
|
Pugazhendhi A, Hubbell M, Jairam P, Ambati B. Neovascular Macular Degeneration: A Review of Etiology, Risk Factors, and Recent Advances in Research and Therapy. Int J Mol Sci 2021; 22:1170. [PMID: 33504013 PMCID: PMC7866170 DOI: 10.3390/ijms22031170] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/11/2021] [Accepted: 01/19/2021] [Indexed: 12/14/2022] Open
Abstract
Neovascular age-related macular degeneration (exudative or wet AMD) is a prevalent, progressive retinal degenerative macular disease that is characterized by neovascularization of the choroid, mainly affecting the elderly population causing gradual vision impairment. Risk factors such as age, race, genetics, iris color, smoking, drinking, BMI, and diet all play a part in nvAMD's progression, with anti-vascular endothelial growth factor (anti-VEGF) therapy being the mainstay of treatment. Current therapeutic advancements slow the progression of the disease but do not cure or reverse its course. Newer therapies such as gene therapies, Rho-kinase inhibitors, and levodopa offer potential new targets for treatment.
Collapse
Affiliation(s)
- Arunbalaji Pugazhendhi
- Knights Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR 97403, USA; (A.P.); (M.H.)
| | - Margaret Hubbell
- Knights Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR 97403, USA; (A.P.); (M.H.)
| | - Pooja Jairam
- Vagelos College of Physicians & Surgeons, Columbia Irving Medical Center, Columbia University, New York, NY 10032, USA;
| | - Balamurali Ambati
- Knights Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR 97403, USA; (A.P.); (M.H.)
| |
Collapse
|
20
|
García-Romero N, Palacín-Aliana I, Madurga R, Carrión-Navarro J, Esteban-Rubio S, Jiménez B, Collazo A, Pérez-Rodríguez F, Ortiz de Mendivil A, Fernández-Carballal C, García-Duque S, Diamantopoulos-Fernández J, Belda-Iniesta C, Prat-Acín R, Sánchez-Gómez P, Calvo E, Ayuso-Sacido A. Bevacizumab dose adjustment to improve clinical outcomes of glioblastoma. BMC Med 2020; 18:142. [PMID: 32564774 PMCID: PMC7310142 DOI: 10.1186/s12916-020-01610-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 04/29/2020] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM) is one of the most aggressive and vascularized brain tumors in adults, with a median survival of 20.9 months. In newly diagnosed and recurrent GBM, bevacizumab demonstrated an increase in progression-free survival, but not in overall survival. METHODS We conducted an in silico analysis of VEGF expression, in a cohort of 1082 glioma patients. Then, to determine whether appropriate bevacizumab dose adjustment could increase the anti-angiogenic response, we used in vitro and in vivo GBM models. Additionally, we analyzed VEGFA expression in tissue, serum, and plasma in a cohort of GBM patients before and during bevacizumab treatment. RESULTS We identified that 20% of primary GBM did not express VEGFA suggesting that these patients would probably not respond to bevacizumab therapy as we proved in vitro and in vivo. We found that a specific dose of bevacizumab calculated based on VEGFA expression levels increases the response to treatment in cell culture and serum samples from mice bearing GBM tumors. Additionally, in a cohort of GBM patients, we observed a correlation of VEGFA levels in serum, but not in plasma, with bevacizumab treatment performance. CONCLUSIONS Our data suggest that bevacizumab dose adjustment could improve clinical outcomes in Glioblastoma treatment.
Collapse
Affiliation(s)
- N García-Romero
- Fundación de Investigación HM Hospitales, HM Hospitales, Madrid, Spain
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, 28223, Spain
| | - I Palacín-Aliana
- Fundación de Investigación HM Hospitales, HM Hospitales, Madrid, Spain
- Atrys Health, Barcelona, 08025, Spain
| | - R Madurga
- Fundación de Investigación HM Hospitales, HM Hospitales, Madrid, Spain
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, 28223, Spain
| | - J Carrión-Navarro
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, 28223, Spain
- Fundación Vithas, Vithas Hospitals, Madrid, 28043, Spain
- Formerly: Fundación de Investigación HM Hospitales, HM Hospitales, Madrid, Spain
| | - S Esteban-Rubio
- Formerly: Facultad de Medicina (IMMA), Universidad San Pablo-CEU, Madrid, Spain
| | - B Jiménez
- Fundación de Investigación HM Hospitales, HM Hospitales, Madrid, Spain
| | - A Collazo
- Fundación de Investigación HM Hospitales, HM Hospitales, Madrid, Spain
| | - F Pérez-Rodríguez
- Fundación de Investigación HM Hospitales, HM Hospitales, Madrid, Spain
| | | | - C Fernández-Carballal
- Servicio de Neurocirugía, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - S García-Duque
- Fundación de Investigación HM Hospitales, HM Hospitales, Madrid, Spain
| | | | - C Belda-Iniesta
- Fundación de Investigación HM Hospitales, HM Hospitales, Madrid, Spain
| | - R Prat-Acín
- Departamento de Neurocirugía, Hospital Universitario la Fe, Valencia, Spain
| | - P Sánchez-Gómez
- Neuro-oncology Unit, Instituto de Salud Carlos III-UFIEC, Madrid, Spain
| | - E Calvo
- Fundación de Investigación HM Hospitales, HM Hospitales, Madrid, Spain
- START Madrid-CIOCC, Centro Integral Oncológico Clara Campal, Madrid, Spain
| | - A Ayuso-Sacido
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, 28223, Spain.
- Fundación Vithas, Vithas Hospitals, Madrid, 28043, Spain.
- Formerly: Fundación de Investigación HM Hospitales, HM Hospitales, Madrid, Spain.
- Formerly: Facultad de Medicina (IMMA), Universidad San Pablo-CEU, Madrid, Spain.
- Formerly: Facultad de Medicina (IMMA), Universidad San Pablo-CEU, Madrid, Spain.
| |
Collapse
|
21
|
McGraw MD, Kim SY, White CW, Veress LA. Acute cytotoxicity and increased vascular endothelial growth factor after in vitro nitrogen mustard vapor exposure. Ann N Y Acad Sci 2020; 1479:223-233. [PMID: 32408394 DOI: 10.1111/nyas.14367] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 04/09/2020] [Accepted: 04/20/2020] [Indexed: 12/20/2022]
Abstract
Nitrogen mustard (NM) is a highly toxic alkylating agent. Inhalation exposure can cause acute and chronic lung injury. This study's aims were to develop an in vitro coculture model of mustard-induced airway injury and to identify growth factors contributing to airway pathology. Primary human bronchial epithelial cells cultured with pulmonary endothelial cells were exposed to NM (25, 50, 100, 250, or 500 μM) or PBS (control) for 1 hour. Lactate dehydrogenase (LDH) and transepithelial electrical resistance (TEER) were measured before and 24 h after NM exposure. Fixed cultures were stained for hematoxylin and eosin or live/dead staining. Culture media were analyzed for 11 growth factors. A 1-h vapor exposure to greater than or equal to 50 μM NM increased supernatant LDH, decreased TEER, and caused airway epithelial cell detachment. Endothelial cell death occurred at 500 μM NM. Vascular endothelial growth factor A (VEGF-A) and placental growth factor (PlGF) expression increased in 500 μM NM-exposed cultures compared with PBS-exposed control cultures. NM vapor exposure causes differential cytotoxicity to airway epithelial and endothelial injury in culture. Increased VEGF-A and PlGF expression occurred acutely in airway cocultures. Future studies are required to validate the role of VEGF signaling in mustard-induced airway pathology.
Collapse
Affiliation(s)
- Matthew D McGraw
- Department of Pediatric Pulmonology, University of Rochester Medical Center, Rochester, New York.,Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| | - So-Young Kim
- Department of Pediatric Pulmonology, University of Rochester Medical Center, Rochester, New York
| | - Carl W White
- Department of Pediatrics, Pulmonology Section, Pediatric Airway Research Center, University of Colorado Denver, Aurora, Colorado
| | - Livia A Veress
- Department of Pediatrics, Pulmonology Section, Pediatric Airway Research Center, University of Colorado Denver, Aurora, Colorado
| |
Collapse
|
22
|
The Role of Rho GTPases in VEGF Signaling in Cancer Cells. Anal Cell Pathol (Amst) 2020; 2020:2097214. [PMID: 32377503 PMCID: PMC7182966 DOI: 10.1155/2020/2097214] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 12/11/2022] Open
Abstract
Vascular endothelial growth factors (VEGFs) consist of five molecules (VEGFA through D as well as placental growth factor) which are crucial for regulating key cellular and tissue functions. The role of VEGF and its intracellular signaling and downstream molecular pathways have been thoroughly studied. Activation of VEGF signal transduction can be initiated by the molecules' binding to two classes of transmembrane receptors: (1) the VEGF tyrosine kinase receptors (VEGF receptors 1 through 3) and (2) the neuropilins (NRP1 and 2). The involvement of Rho GTPases in modulating VEGFA signaling in both cancer cells and endothelial cells has also been well established. Additionally, different isoforms of Rho GTPases, namely, RhoA, RhoC, and RhoG, have been shown to regulate VEGF expression as well as blood vessel formation. This review article will explore how Rho GTPases modulate VEGF signaling and the consequences of such interaction on cancer progression.
Collapse
|
23
|
Atienza-Roca P, Kieser DC, Cui X, Bathish B, Ramaswamy Y, Hooper GJ, Clarkson AN, Rnjak-Kovacina J, Martens PJ, Wise LM, Woodfield TBF, Lim KS. Visible light mediated PVA-tyramine hydrogels for covalent incorporation and tailorable release of functional growth factors. Biomater Sci 2020; 8:5005-5019. [DOI: 10.1039/d0bm00603c] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PVA-Tyr hydrogel facilitated covalent incorporation can control release of pristine growth factors while retaining their native bioactivity.
Collapse
Affiliation(s)
- Pau Atienza-Roca
- Department of Orthopaedic Surgery
- University of Otago Christchurch
- Christchurch 8011
- New Zealand
| | - David C. Kieser
- Department of Orthopaedic Surgery
- University of Otago Christchurch
- Christchurch 8011
- New Zealand
| | - Xiaolin Cui
- Department of Orthopaedic Surgery
- University of Otago Christchurch
- Christchurch 8011
- New Zealand
| | - Boushra Bathish
- Department of Orthopaedic Surgery
- University of Otago Christchurch
- Christchurch 8011
- New Zealand
| | - Yogambha Ramaswamy
- School of Biomedical Engineering
- University of Sydney
- Sydney 2006
- Australia
| | - Gary J. Hooper
- Department of Orthopaedic Surgery
- University of Otago Christchurch
- Christchurch 8011
- New Zealand
| | - Andrew N. Clarkson
- Department of Anatomy
- Brain Health Research Centre and Brain Research New Zealand
- University of Otago
- Dunedin 9054
- New Zealand
| | | | - Penny J. Martens
- Graduate School of Biomedical Engineering
- UNSW Sydney
- Sydney 2052
- Australia
| | - Lyn M. Wise
- Department of Pharmacology and Toxicology
- University of Otago
- New Zealand
| | - Tim B. F. Woodfield
- Department of Orthopaedic Surgery
- University of Otago Christchurch
- Christchurch 8011
- New Zealand
| | - Khoon S. Lim
- Department of Orthopaedic Surgery
- University of Otago Christchurch
- Christchurch 8011
- New Zealand
| |
Collapse
|
24
|
Buettmann EG, McKenzie JA, Migotsky N, Sykes DA, Hu P, Yoneda S, Silva MJ. VEGFA From Early Osteoblast Lineage Cells (Osterix+) Is Required in Mice for Fracture Healing. J Bone Miner Res 2019; 34:1690-1706. [PMID: 31081125 PMCID: PMC6744295 DOI: 10.1002/jbmr.3755] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/23/2019] [Accepted: 05/01/2019] [Indexed: 12/21/2022]
Abstract
Bone formation via intramembranous and endochondral ossification is necessary for successful healing after a wide range of bone injuries. The pleiotropic cytokine, vascular endothelial growth factor A (VEGFA) has been shown, via nonspecific pharmacologic inhibition, to be indispensable for angiogenesis and ossification following bone fracture and cortical defect repair. However, the importance of VEGFA expression by different cell types during bone healing is not well understood. We sought to determine the role of VEGFA from different osteoblast cell subsets following clinically relevant models of bone fracture and cortical defect. Ubiquitin C (UBC), Osterix (Osx), or Dentin matrix protein 1 (Dmp1) Cre-ERT2 mice (male and female) containing floxed VEGFA alleles (VEGFAfl/fl ) were either given a femur full fracture, ulna stress fracture, or tibia cortical defect at 12 weeks of age. All mice received tamoxifen continuously starting 2 weeks before bone injury and throughout healing. UBC Cre-ERT2 VEGFAfl/fl (UBC cKO) mice, which were used to mimic nonspecific inhibition, had minimal bone formation and impaired angiogenesis across all bone injury models. UBC cKO mice also exhibited impaired periosteal cell proliferation during full fracture, but not stress fracture repair. Osx Cre-ERT2 VEGFAfl/fl (Osx cKO) mice, but not Dmp1 Cre-ERT2 VEGFAfl/fl (Dmp1 cKO) mice, showed impaired periosteal bone formation and angiogenesis in models of full fracture and stress fracture. Neither Osx cKO nor Dmp1 cKO mice demonstrated significant impairments in intramedullary bone formation and angiogenesis following cortical defect. These data suggest that VEGFA from early osteolineage cells (Osx+), but not mature osteoblasts/osteocytes (Dmp1+), is critical at the time of bone injury for rapid periosteal angiogenesis and woven bone formation during fracture repair. Whereas VEGFA from another cell source, not from the osteoblast cell lineage, is necessary at the time of injury for maximum cortical defect intramedullary angiogenesis and osteogenesis. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Evan G Buettmann
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Jennifer A McKenzie
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Nicole Migotsky
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - David Aw Sykes
- Department of Biology, Washington University in St. Louis, St. Louis, MO, USA
| | - Pei Hu
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Susumu Yoneda
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Matthew J Silva
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
25
|
Yang H, Jung DH, Lee HW. Therapeutic effect of Cnidium officinale Makino extract on ovariectomized hind-limb ischemic mice. Integr Med Res 2019; 8:107-115. [PMID: 31193479 PMCID: PMC6529788 DOI: 10.1016/j.imr.2019.04.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 04/17/2019] [Accepted: 04/29/2019] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Cnidium officinale Makino (COM) has been used traditionally to treat female sexual disorders, such as amenorrhea, hypomenorrhea and oligomenorrhea, by improving blood circulation. METHODS The present study aimed to investigate the alleviating effect of COM extracts on surgical injury-induced ischemia in the hind-limb of mice. In this study, female C57BL/6 mice were ovariectomized, and the vessels of the hind-limb were excised after ligation by surgical silk (6-0). The mice were orally administered with COM (150 or 300 mg/kg/day) for 3 weeks, and the blood flow rate of hind-limbs was evaluated by using a laser Doppler system after hind-limb ischemic surgery in an in vivo study. Additionally, the migration and tube formation of human umbilical vein endothelial cells (HUVECs) were evaluated in an in vitro study. RESULTS The blood flow rate was synchronized to the nonischemic lesion of the hind-limb, and its elevation compared to the vehicle was observed at 14 and 21 days after hind-limb ischemic surgery in COM-treated groups. The number of capillaries increase in a dose-dependent manner in the COM-treated groups (150 and 300 mg/kg). In HUVECs, the activities of cell migration were significantly increased by 50 and 75 μg/mL for the COM-treated groups. In addition, the number of tubule branches and junctions was also increased by doses of COM (50 and/or 75 μg/mL). CONCLUSION The results of our study suggested that the COM extract may have therapeutic application for the treatment of hind-limb ischemic damage, which is due to the improvement of the peripheral angiogenetic system.
Collapse
Affiliation(s)
| | | | - Hye Won Lee
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon, Republic of Korea
| |
Collapse
|
26
|
Wu CH, Ko JL, Pan HH, Chiu LY, Kang YT, Hsiao YP. Ni-induced TGF-β signaling promotes VEGF-a secretion via integrin β3 upregulation. J Cell Physiol 2019; 234:22093-22102. [PMID: 31066035 DOI: 10.1002/jcp.28772] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 04/11/2019] [Accepted: 04/17/2019] [Indexed: 12/20/2022]
Abstract
Nickel compounds are associated with lung and skin cancer incidence increase and accumulation of nickel in the body contributes to carcinogenesis. Upregulation of certain integrins in the primary tumor is associated with cancer metastasis and poor prognosis. However, the molecular mechanisms of nickel-induced cancer metastasis are still unclear. The purpose of the present study was to investigate the effects of nickel chloride (NiCl2 ) on the progression of cancer during metastasis. The results of showed that NiCl2 induces the expression of integrin β3 mRNA and protein in a dose- and time-dependent manner. Inhibition of integrin αvβ3 activation by ITGB3 ligand mimetics and GR144053, as well as downregulation of ITGB3 by lentiviral shRNA gene silencing, diminished NiCl2 -induced secretion of vascular endothelial growth factor-a (VEGF-a). Furthermore, pretreatment with type I TGF-β receptor inhibitor, SB525334, suppressed the expression of ITGB3 at cell surface and secretion of VEGF-a in NiCl2 -treated cells. In conclusion, NiCl2 induces the expression of ITGB3 through TGF-β signaling activation, followed by increasing VEGF-a secretion, revealing a novel role for ITGB3 in nickel compound-induced cancer metastasis and tumor angiogenesis.
Collapse
Affiliation(s)
- Chih-Hsien Wu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Jiunn-Liang Ko
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Dermatology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Hui-Hsien Pan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Pediatrics, Institute of Allergy, Immunology, and Rheumatology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ling-Yen Chiu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yu-Ting Kang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yu-Ping Hsiao
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Dermatology, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
27
|
Strubl S, Schubert U, Kühnle A, Rebl A, Ahmadvand N, Fischer S, Preissner KT, Galuska SP. Polysialic acid is released by human umbilical vein endothelial cells (HUVEC) in vitro. Cell Biosci 2018; 8:64. [PMID: 30555678 PMCID: PMC6288938 DOI: 10.1186/s13578-018-0262-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 12/04/2018] [Indexed: 12/27/2022] Open
Abstract
Background Sialic acids represent common terminal residues on numerous mammalian glycoconjugates, thereby influencing e.g. lumen formation in developing blood vessels. Interestingly, besides monosialylated also polysialylated glycoconjugates are produced by endothelial cells. Polysialic acid (polySia) is formed in several organs during embryonal and postnatal development influencing, for instance, cell migration processes. Furthermore, the function of cytokines like basic fibroblast growth factor (bFGF) is modulated by polySia. Results In this study, we demonstrated that human umbilical vein endothelial cells (HUVEC) also secrete polysialylated glycoconjugates. Furthermore, an interaction between polySia and vascular endothelial growth factor (VEGF) was observed. VEGF modulates like bFGF the migration of HUVEC. Since both growth factors interact with polySia, we examined, if polySia modulates the migration of HUVEC. To this end scratch assays were performed showing that the migration of HUVEC is stimulated, when polySia was degraded. Conclusions Since polySia can interact with bFGF as well as VEGF and the degradation of polySia resulted in an increased cell migration capacity in the applied scratch assay, we propose that polySia may trap these growth factors influencing their biological activity. Thus, polySia might also contribute to the fine regulation of physiological processes in endothelial cells.
Collapse
Affiliation(s)
- Sebastian Strubl
- 1Institute of Biochemistry, Faculty of Medicine, Justus-Liebig-University, Friedrichstrasse 24, 35392 Giessen, Germany.,2Department II of Internal Medicine, Center for Molecular Medicine Cologne, University Cologne, Kerpener Str. 62, 50931 Cologne, Germany
| | - Uwe Schubert
- 1Institute of Biochemistry, Faculty of Medicine, Justus-Liebig-University, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Andrea Kühnle
- 3Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Alexander Rebl
- 4Institute of Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Negah Ahmadvand
- 1Institute of Biochemistry, Faculty of Medicine, Justus-Liebig-University, Friedrichstrasse 24, 35392 Giessen, Germany.,Excellence Cluster Cardio Pulmonary System (ECCPS), Aulweg 130, 35392 Giessen, Germany
| | - Silvia Fischer
- 1Institute of Biochemistry, Faculty of Medicine, Justus-Liebig-University, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Klaus T Preissner
- 1Institute of Biochemistry, Faculty of Medicine, Justus-Liebig-University, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Sebastian P Galuska
- 1Institute of Biochemistry, Faculty of Medicine, Justus-Liebig-University, Friedrichstrasse 24, 35392 Giessen, Germany.,3Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| |
Collapse
|
28
|
Kim J, Piao HL, Kim BJ, Yao F, Han Z, Wang Y, Xiao Z, Siverly AN, Lawhon SE, Ton BN, Lee H, Zhou Z, Gan B, Nakagawa S, Ellis MJ, Liang H, Hung MC, You MJ, Sun Y, Ma L. Long noncoding RNA MALAT1 suppresses breast cancer metastasis. Nat Genet 2018; 50:1705-1715. [PMID: 30349115 PMCID: PMC6265076 DOI: 10.1038/s41588-018-0252-3] [Citation(s) in RCA: 548] [Impact Index Per Article: 78.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 09/07/2018] [Indexed: 12/17/2022]
Abstract
MALAT1 has previously been described as a metastasis-promoting long noncoding RNA (lncRNA). We show here, however, that targeted inactivation of the Malat1 gene in a transgenic mouse model of breast cancer, without altering the expression of its adjacent genes, promotes lung metastasis, and that this phenotype can be reversed by genetic add-back of Malat1. Similarly, knockout of MALAT1 in human breast cancer cells induces their metastatic ability, which is reversed by re-expression of Malat1. Conversely, overexpression of Malat1 suppresses breast cancer metastasis in transgenic, xenograft, and syngeneic models. Mechanistically, the MALAT1 lncRNA binds and inactivates the prometastatic transcription factor TEAD, preventing TEAD from associating with its co-activator YAP and target gene promoters. Moreover, MALAT1 levels inversely correlate with breast cancer progression and metastatic ability. These findings demonstrate that MALAT1 is a metastasis-suppressing lncRNA rather than a metastasis promoter in breast cancer, calling for rectification of the model for this highly abundant and conserved lncRNA.
Collapse
Affiliation(s)
- Jongchan Kim
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hai-Long Piao
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Scientific Research Center for Translational Medicine, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Beom-Jun Kim
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Fan Yao
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhenbo Han
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yumeng Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhenna Xiao
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Ashley N Siverly
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sarah E Lawhon
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Baochau N Ton
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hyemin Lee
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhicheng Zhou
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shinichi Nakagawa
- RNA Biology Laboratory, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Matthew J Ellis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Han Liang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
| | - M James You
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yutong Sun
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Li Ma
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
| |
Collapse
|
29
|
Phenotypic and Expressional Heterogeneity in the Invasive Glioma Cells. Transl Oncol 2018; 12:122-133. [PMID: 30292065 PMCID: PMC6172486 DOI: 10.1016/j.tranon.2018.09.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 09/21/2018] [Accepted: 09/24/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND: Tumor cell invasion is a hallmark of glioblastoma (GBM) and a major contributing factor for treatment failure, tumor recurrence, and the poor prognosis of GBM. Despite this, our understanding of the molecular machinery that drives invasion is limited. METHODS: Time-lapse imaging of patient-derived GBM cell invasion in a 3D collagen gel matrix, analysis of both the cellular invasive phenotype and single cell invasion pattern with microarray expression profiling. RESULTS: GBM invasion was maintained in a simplified 3D-milieue. Invasion was promoted by the presence of the tumorsphere graft. In the absence of this, the directed migration of cells subsided. The strength of the pro-invasive repulsive signaling was specific for a given patient-derived culture. In the highly invasive GBM cultures, the majority of cells had a neural progenitor-like phenotype, while the less invasive cultures had a higher diversity in cellular phenotypes. Microarray expression analysis of the non-invasive cells from the tumor core displayed a higher GFAP expression and a signature of genes containing VEGFA, hypoxia and chemo-repulsive signals. Cells of the invasive front expressed higher levels of CTGF, TNFRSF12A and genes involved in cell survival, migration and cell cycle pathways. A mesenchymal gene signature was associated with increased invasion. CONCLUSION: The GBM tumorsphere core promoted invasion, and the invasive front was dominated by a phenotypically defined cell population expressing genes regulating traits found in aggressive cancers. The detected cellular heterogeneity and transcriptional differences between the highly invasive and core cells identifies potential targets for manipulation of GBM invasion.
Collapse
|
30
|
Integrin α9 Suppresses Hepatocellular Carcinoma Metastasis by Rho GTPase Signaling. J Immunol Res 2018; 2018:4602570. [PMID: 29951557 PMCID: PMC5989280 DOI: 10.1155/2018/4602570] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 04/10/2018] [Indexed: 12/12/2022] Open
Abstract
Integrin subunit alpha 9 (ITGA9) mediates cell-cell and cell-matrix adhesion, cell migration, and invasion through binding different kinds of extracellular matrix (ECM) components. However, its potential role and underlying molecular mechanisms remain unclear in hepatocellular carcinoma (HCC). Here, we found that ITGA9 expression was obviously decreased in patients with HCC, which was negatively correlated with HCC growth and metastasis. ITGA9 overexpression significantly inhibited cell proliferation and migration in vitro as well as tumor growth and metastasis in vivo. Our data demonstrated that the inhibitory effect of ITGA9 on HCC cell motility was associated with reduced phosphorylation of focal adhesion kinase (FAK) and c-Src tyrosine kinase (Src), disrupted focal adhesion reorganization, and decreased Rac1 and RhoA activity. Our data suggest ITGA9, as a suppressor of HCC, prevents tumor cell migration and invasiveness through FAK/Src-Rac1/RhoA signaling.
Collapse
|
31
|
Shimizu A, Zankov DP, Kurokawa-Seo M, Ogita H. Vascular Endothelial Growth Factor-A Exerts Diverse Cellular Effects via Small G Proteins, Rho and Rap. Int J Mol Sci 2018; 19:ijms19041203. [PMID: 29659486 PMCID: PMC5979568 DOI: 10.3390/ijms19041203] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 04/03/2018] [Accepted: 04/12/2018] [Indexed: 12/18/2022] Open
Abstract
Vascular endothelial growth factors (VEGFs) include five molecules (VEGF-A, -B, -C, -D, and placental growth factor), and have various roles that crucially regulate cellular functions in many kinds of cells and tissues. Intracellular signal transduction induced by VEGFs has been extensively studied and is usually initiated by their binding to two classes of transmembrane receptors: receptor tyrosine kinase VEGF receptors (VEGF receptor-1, -2 and -3) and neuropilins (NRP1 and NRP2). In addition to many established results reported by other research groups, we have previously identified small G proteins, especially Ras homologue gene (Rho) and Ras-related protein (Rap), as important mediators of VEGF-A-stimulated signaling in cancer cells as well as endothelial cells. This review article describes the VEGF-A-induced signaling pathways underlying diverse cellular functions, including cell proliferation, migration, and angiogenesis, and the involvement of Rho, Rap, and their related molecules in these pathways.
Collapse
Affiliation(s)
- Akio Shimizu
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan.
| | - Dimitar P Zankov
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan.
| | - Misuzu Kurokawa-Seo
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan.
| | - Hisakazu Ogita
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan.
| |
Collapse
|
32
|
Das A, Huang GX, Bonkowski MS, Longchamp A, Li C, Schultz MB, Kim LJ, Osborne B, Joshi S, Lu Y, Treviño-Villarreal JH, Kang MJ, Hung TT, Lee B, Williams EO, Igarashi M, Mitchell JR, Wu LE, Turner N, Arany Z, Guarente L, Sinclair DA. Impairment of an Endothelial NAD +-H 2S Signaling Network Is a Reversible Cause of Vascular Aging. Cell 2018; 173:74-89.e20. [PMID: 29570999 PMCID: PMC5884172 DOI: 10.1016/j.cell.2018.02.008] [Citation(s) in RCA: 335] [Impact Index Per Article: 47.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/25/2017] [Accepted: 01/31/2018] [Indexed: 11/26/2022]
Abstract
A decline in capillary density and blood flow with age is a major cause of mortality and morbidity. Understanding why this occurs is key to future gains in human health. NAD precursors reverse aspects of aging, in part, by activating sirtuin deacylases (SIRT1-SIRT7) that mediate the benefits of exercise and dietary restriction (DR). We show that SIRT1 in endothelial cells is a key mediator of pro-angiogenic signals secreted from myocytes. Treatment of mice with the NAD+ booster nicotinamide mononucleotide (NMN) improves blood flow and increases endurance in elderly mice by promoting SIRT1-dependent increases in capillary density, an effect augmented by exercise or increasing the levels of hydrogen sulfide (H2S), a DR mimetic and regulator of endothelial NAD+ levels. These findings have implications for improving blood flow to organs and tissues, increasing human performance, and reestablishing a virtuous cycle of mobility in the elderly.
Collapse
Affiliation(s)
- Abhirup Das
- Paul F. Glenn Center for the Biological Mechanisms of Aging, Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Laboratory for Ageing Research, Department of Pharmacology, School of Medical Sciences, The University of New South Wales, Sydney, NSW 2052, Australia; Paul F. Glenn Center for Science of Aging Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - George X Huang
- Paul F. Glenn Center for the Biological Mechanisms of Aging, Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Bldg. 421, Philadelphia, PA 19104, USA
| | - Michael S Bonkowski
- Paul F. Glenn Center for the Biological Mechanisms of Aging, Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Alban Longchamp
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Catherine Li
- Laboratory for Ageing Research, Department of Pharmacology, School of Medical Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Michael B Schultz
- Paul F. Glenn Center for the Biological Mechanisms of Aging, Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Lynn-Jee Kim
- Laboratory for Ageing Research, Department of Pharmacology, School of Medical Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Brenna Osborne
- Mitochondrial Bioenergetics Laboratory, Department of Pharmacology, School of Medical Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Sanket Joshi
- Mitochondrial Bioenergetics Laboratory, Department of Pharmacology, School of Medical Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Yuancheng Lu
- Paul F. Glenn Center for the Biological Mechanisms of Aging, Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | | | - Myung-Jin Kang
- Laboratory for Ageing Research, Department of Pharmacology, School of Medical Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Tzong-Tyng Hung
- Biological Resources Imaging Laboratory, Mark Wainwright Analytical Centre, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Brendan Lee
- Biological Resources Imaging Laboratory, Mark Wainwright Analytical Centre, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Eric O Williams
- Paul F. Glenn Center for Science of Aging Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Masaki Igarashi
- Paul F. Glenn Center for Science of Aging Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - James R Mitchell
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Lindsay E Wu
- Laboratory for Ageing Research, Department of Pharmacology, School of Medical Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Nigel Turner
- Mitochondrial Bioenergetics Laboratory, Department of Pharmacology, School of Medical Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Zolt Arany
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Bldg. 421, Philadelphia, PA 19104, USA.
| | - Leonard Guarente
- Paul F. Glenn Center for Science of Aging Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - David A Sinclair
- Paul F. Glenn Center for the Biological Mechanisms of Aging, Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Laboratory for Ageing Research, Department of Pharmacology, School of Medical Sciences, The University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
33
|
Pu H, Zhang Q, Zhao C, Shi L, Wang Y, Wang J, Zhang M. VEGFA Involves in the Use of Fluvastatin and Zoledronate Against Breast Cancer. Pathol Oncol Res 2017; 24:557-565. [PMID: 28744693 DOI: 10.1007/s12253-017-0277-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 07/12/2017] [Indexed: 12/19/2022]
Abstract
Our study aimed to identify key genes involved in the use of fluvastatin and zoledronate against breast cancer, as well as to investigate the roles of vascular endothelial growth factor A (VEGFA) in the malignant behaviors of breast cancer cells. The expression data GSE33552 was downloaded from Gene Expression Omnibus database, including mocked-, fluvastatin- and zoledronate-treated MDA-MB-231 cells. Differentially expressed genes (DEGs) were identified in fluvastatin- and zoledronate-treated cells using limma package, respectively. Pathway enrichment analysis and protein-protein interaction (PPI) network analysis were then performed. Then we used shRNA specifically targeting VEGFA (shVEGFA) to knock down the expression of VEGFA in MDA-MB-231 cells. Cell viability assay, scratch wound healing assay, Transwell invasion assay and flow cytometry were performed to explore the effects of VEGFA knockdown on the malignant behaviors of breast cancer cells. VEGFA was up-regulated in both fluvastatin- and zoledronate-treated breast cancer cells. Moreover, VEGFA was a hub node in PPI network. In addition, VEGFA was successfully knocked down in MDA-MB-231 cells by shVEGFA. Suppression of VEGFA promoted the migration and invasion of breast cancer MDA-MB-231 cells. Suppression of VEGFA inhibited the apoptosis of MDA-MB-231 cells. Our results indicate that up-regulation of VEGFA may prevent the progression of breast cancer after fluvastatin and zoledronate treatment via inducing cell apoptosis and inhibiting migration and invasion. VEGFA may serve as a potential prognostic indicator for clinical outcome in the management of breast cancer.
Collapse
Affiliation(s)
- Haihong Pu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Haping Road 150 of Nangang District, Harbin, Heilongjiang Province, 150086, China
| | - Qingyuan Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Haping Road 150 of Nangang District, Harbin, Heilongjiang Province, 150086, China.
| | - Chunbo Zhao
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, 150086, Heilongjiang Province, China
| | - Lei Shi
- Department of Radiation Oncology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yan Wang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Haping Road 150 of Nangang District, Harbin, Heilongjiang Province, 150086, China
| | - Jingxuan Wang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Haping Road 150 of Nangang District, Harbin, Heilongjiang Province, 150086, China
| | - Minghui Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Haping Road 150 of Nangang District, Harbin, Heilongjiang Province, 150086, China
| |
Collapse
|
34
|
Cehofski LJ, Kruse A, Bøgsted M, Magnusdottir SO, Stensballe A, Honoré B, Vorum H. Retinal proteome changes following experimental branch retinal vein occlusion and intervention with ranibizumab. Exp Eye Res 2016; 152:49-56. [PMID: 27619476 DOI: 10.1016/j.exer.2016.09.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 08/07/2016] [Accepted: 09/08/2016] [Indexed: 01/28/2023]
Abstract
Animal models of experimental branch retinal vein occlusion (BRVO) provide a unique opportunity to study protein changes directly in retinal tissue. Results from these experimental models suggest that experimental BRVO is associated with an upregulation of extracellular matrix remodeling and adhesion signaling processes. To study whether these processes could be blocked by inhibition of VEGF-A, a porcine model of experimental BRVO was combined with proteomic analyses. In six Danish Landrace pigs experimental BRVO was induced with argon laser in both eyes. After 24 h an injection of 0.05 mL ranibizumab was given in the right eyes of the animals while left eyes received an injection of 0.05 mL 9 mg/mL sodium chloride water. Retinas were dissected three days after BRVO and the retinal samples were analyzed with label-free quantification as well as tandem mass tag based proteomics. In retinas treated with ranibizumab five proteins exhibited statistically significant changes in content with both proteomic techniques. These five proteins, which were all decreased in content, included integrin β-1, peroxisomal 3-ketoacyl-CoA thiolase, OCIA domain-containing protein 1, calnexin and 40S ribosomal protein S5. As anti-integrin therapies are under development for inhibition of angiogenesis in retinal diseases it is interesting that inhibition of VEGF-A in itself resulted in a small decrease in the content of integrin β-1. The decreased content of integrin β-1 indicates that extracellular matrix remodeling and adhesion processes associated with BRVO are at least partly reversed through inhibition of VEGF-A.
Collapse
Affiliation(s)
- Lasse Jørgensen Cehofski
- Department of Ophthalmology, Aalborg University Hospital, Aalborg, Denmark; Biomedical Research Laboratory, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| | - Anders Kruse
- Department of Ophthalmology, Aalborg University Hospital, Aalborg, Denmark
| | - Martin Bøgsted
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Department of Haematology, Aalborg University Hospital, Aalborg, Denmark
| | | | - Allan Stensballe
- Department of Health Science and Technology, Aalborg University, Denmark
| | - Bent Honoré
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Henrik Vorum
- Department of Ophthalmology, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
35
|
Zeng FC, Zeng MQ, Huang L, Li YL, Gao BM, Chen JJ, Xue RZ, Tang ZY. Downregulation of VEGFA inhibits proliferation, promotes apoptosis, and suppresses migration and invasion of renal clear cell carcinoma. Onco Targets Ther 2016; 9:2131-41. [PMID: 27110129 PMCID: PMC4835130 DOI: 10.2147/ott.s98002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Objective The aim of this study was to investigate the effects of vascular endothelial growth factor A (VEGFA) on cell proliferation, apoptosis, migration, and invasion in renal clear cell carcinoma (RCCC). Methods Between June 2012 and June 2015, RCCC tissues were obtained for the experimental group, and RCCC adjacent tumor-free kidney parenchyma tissues were obtained for the control group. VEGFA mRNA and protein expressions and phosphoinositide 3-kinase, serine/threonine-specific protein kinase (AKT), and phosphorylated-AKT protein expressions were detected. The chemically synthesized specific siRNA using RNA interference technology was used to inhibit VEGFA gene expression in human RCCC 786-O cells. The negative control (NC) group was transfected with NC sequence, and the blank group was transfected with no sequence. Flow cytometry, scratch test, and cell-penetrating experiment were used to detect cell proliferation, apoptosis, migration, and invasion of 786-O cells. Results Positive expression of VEGFA protein was 60.62% in RCCC tissue and 18.34% in adjacent tissue with statistically significant difference (P<0.001). VEGFA protein and mRNA expressions were higher in RCCC tissue than those in adjacent tissue (both P<0.01). VEGF expression in RCCC tissue was associated with Fuhrman grading and American Joint Committee on Cancer staging (both P<0.05). After RCCC 786-O cells transfecting the VEGFA siRNA, the VEGFA mRNA and protein expressions and phosphoinositide 3-kinase and phosphorylated-AKT protein expressions were significantly decreased, cell proliferation was remarkably inhibited, cell apoptotic ratio was obviously increased, and migration distance and invasive cell number were markedly decreased compared to those in the NC group and the blank group (all P<0.05). Conclusion Inhibition of VEGFA inhibited proliferation, promoted apoptosis, and suppressed migration and invasion of RCCC 786-O cells. VEGF has a potential role in diagnosis and therapy of RCCC.
Collapse
Affiliation(s)
- Fan-Chang Zeng
- Department of Urology, Xiangya Hospital, Central South University, Changsha, People's Republic of China; Department of Urology, Hainan General Hospital, Haikou, People's Republic of China
| | - Ming-Qiang Zeng
- Department of Urology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Liang Huang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Yong-Lin Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Ben-Min Gao
- Department of Urology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Jun-Jie Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Rui-Zhi Xue
- Department of Urology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Zheng-Yan Tang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| |
Collapse
|
36
|
Dong KN, Huang X, Xing WY, Guo WW, Feng R. Effect of endoplasmic reticulum stress on gastric cancer cell migration and invasion. Shijie Huaren Xiaohua Zazhi 2016; 24:1485-1491. [DOI: 10.11569/wcjd.v24.i10.1485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIM: To investigate the effect of endoplasmic reticulum stress (ERS) on gastric cancer cell migration and invasion.
METHODS: Gastric cancer BGC823 or SGC7901 cells were treated with tunicamycin (TM) for 0, 12, 24 and 36 h. The expression of glucose-regulated protein 78 (GRP78), matrix metalloproteinase 9 (MMP9) and vascular endothelial growth factor (VEGF) was analyzed using Western blot. Wound healing assay was used to detect the migrating ability of gastric cancer cells at each time point with or without TM treatment. Transwell assay was employed to examine invasive ability of gastric cancer cells at each time point with or without TM treatment.
RESULTS: Compared with baseline values, the expression of GRP78, MMP9 and VEGF in gastric cancer cells was significantly increased after treatment with TM for 12, 24 or 36 h (P < 0.05). Wound healing ability of gastric cancer cells treated with TM for 12, 24 or 36 h was much stronger than that in the 0 h group (P < 0.05). The number of gastric cancer cells invading to the lower chamber increased after TM treatment for 12, 24 or 36 h (P < 0.05).
CONCLUSION: ERS promotes gastric cancer cell migration and invasion probably via upregulation of MMP9 and VEGF expression.
Collapse
|
37
|
Helal-Neto E, Brandão-Costa RM, Saldanha-Gama R, Ribeiro-Pereira C, Midlej V, Benchimol M, Morandi V, Barja-Fidalgo C. Priming Endothelial Cells With a Melanoma-Derived Extracellular Matrix Triggers the Activation of αvβ3/VEGFR2 Axis. J Cell Physiol 2016; 231:2464-73. [DOI: 10.1002/jcp.25358] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 02/22/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Edward Helal-Neto
- Laboratório de Farmacologia Celular e Molecular, IBRAG; Universidade do Estado do Rio de Janeiro; Rio de Janeiro Brazil
- Laboratório de Biologia da Célula Endotelial e da Angiogênese, IBRAG; Universidade do Estado do Rio de Janeiro; Rio de Janeiro Brazil
| | - Renata M. Brandão-Costa
- Laboratório de Farmacologia Celular e Molecular, IBRAG; Universidade do Estado do Rio de Janeiro; Rio de Janeiro Brazil
| | - Roberta Saldanha-Gama
- Laboratório de Farmacologia Celular e Molecular, IBRAG; Universidade do Estado do Rio de Janeiro; Rio de Janeiro Brazil
| | - Cristiane Ribeiro-Pereira
- Laboratório de Farmacologia Celular e Molecular, IBRAG; Universidade do Estado do Rio de Janeiro; Rio de Janeiro Brazil
| | - Victor Midlej
- Instituto de Biofísica Carlos Chagas Filho; Universidade Federal do Rio de Janeiro; Rio de Janeiro Brazil
| | - Marlene Benchimol
- Instituto de Biofísica Carlos Chagas Filho; Universidade Federal do Rio de Janeiro; Rio de Janeiro Brazil
- Unigranrio; Universidade do Grande Rio; Rio de Janeiro Brazil
| | - Verônica Morandi
- Laboratório de Biologia da Célula Endotelial e da Angiogênese, IBRAG; Universidade do Estado do Rio de Janeiro; Rio de Janeiro Brazil
| | - Christina Barja-Fidalgo
- Laboratório de Farmacologia Celular e Molecular, IBRAG; Universidade do Estado do Rio de Janeiro; Rio de Janeiro Brazil
| |
Collapse
|
38
|
Fink DM, Steele MM, Hollingsworth MA. The lymphatic system and pancreatic cancer. Cancer Lett 2015; 381:217-36. [PMID: 26742462 DOI: 10.1016/j.canlet.2015.11.048] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 11/16/2015] [Accepted: 11/30/2015] [Indexed: 02/06/2023]
Abstract
This review summarizes current knowledge of the biology, pathology and clinical understanding of lymphatic invasion and metastasis in pancreatic cancer. We discuss the clinical and biological consequences of lymphatic invasion and metastasis, including paraneoplastic effects on immune responses and consider the possible benefit of therapies to treat tumors that are localized to lymphatics. A review of current techniques and methods to study interactions between tumors and lymphatics is presented.
Collapse
Affiliation(s)
- Darci M Fink
- Eppley Institute, University of Nebraska Medical Center, Omaha, NE 68198-5950, USA
| | - Maria M Steele
- Eppley Institute, University of Nebraska Medical Center, Omaha, NE 68198-5950, USA
| | | |
Collapse
|
39
|
Blandin AF, Renner G, Lehmann M, Lelong-Rebel I, Martin S, Dontenwill M. β1 Integrins as Therapeutic Targets to Disrupt Hallmarks of Cancer. Front Pharmacol 2015; 6:279. [PMID: 26635609 PMCID: PMC4656837 DOI: 10.3389/fphar.2015.00279] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 11/05/2015] [Indexed: 01/11/2023] Open
Abstract
Integrins belong to a large family of αβ heterodimeric transmembrane proteins first recognized as adhesion molecules that bind to dedicated elements of the extracellular matrix and also to other surrounding cells. As important sensors of the cell microenvironment, they regulate numerous signaling pathways in response to structural variations of the extracellular matrix. Biochemical and biomechanical cues provided by this matrix and transmitted to cells via integrins are critically modified in tumoral settings. Integrins repertoire are subjected to expression level modifications, in tumor cells, and in surrounding cancer-associated cells, implicated in tumor initiation and progression as well. As critical players in numerous cancer hallmarks, defined by Hanahan and Weinberg (2011), integrins represent pertinent therapeutic targets. We will briefly summarize here our current knowledge about integrin implications in those different hallmarks focusing primarily on β1 integrins.
Collapse
Affiliation(s)
- Anne-Florence Blandin
- Department "Tumoral Signaling and Therapeutic Targets," Faculty of Pharmacy, UMR7213 Centre National de la Recherche Scientifique, University of Strasbourg Illkirch, France
| | - Guillaume Renner
- Department "Tumoral Signaling and Therapeutic Targets," Faculty of Pharmacy, UMR7213 Centre National de la Recherche Scientifique, University of Strasbourg Illkirch, France
| | - Maxime Lehmann
- Department "Tumoral Signaling and Therapeutic Targets," Faculty of Pharmacy, UMR7213 Centre National de la Recherche Scientifique, University of Strasbourg Illkirch, France
| | - Isabelle Lelong-Rebel
- Department "Tumoral Signaling and Therapeutic Targets," Faculty of Pharmacy, UMR7213 Centre National de la Recherche Scientifique, University of Strasbourg Illkirch, France
| | - Sophie Martin
- Department "Tumoral Signaling and Therapeutic Targets," Faculty of Pharmacy, UMR7213 Centre National de la Recherche Scientifique, University of Strasbourg Illkirch, France
| | - Monique Dontenwill
- Department "Tumoral Signaling and Therapeutic Targets," Faculty of Pharmacy, UMR7213 Centre National de la Recherche Scientifique, University of Strasbourg Illkirch, France
| |
Collapse
|
40
|
Egervari K, Potter G, Guzman-Hernandez ML, Salmon P, Soto-Ribeiro M, Kastberger B, Balla T, Wehrle-Haller B, Kiss JZ. Astrocytes spatially restrict VEGF signaling by polarized secretion and incorporation of VEGF into the actively assembling extracellular matrix. Glia 2015; 64:440-56. [PMID: 26539695 DOI: 10.1002/glia.22939] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 10/13/2015] [Accepted: 10/15/2015] [Indexed: 01/13/2023]
Abstract
The spatial organization of vascular endothelial growth factor (VEGF) signaling is a key determinant of vascular patterning during development and tissue repair. How VEGF signaling becomes spatially restricted and the role of VEGF secreting astrocytes in this process remains poorly understood. Using a VEGF-GFP fusion protein and confocal time-lapse microscopy, we observed the intracellular routing, secretion and immobilization of VEGF in scratch-activated living astrocytes. We found VEGF to be directly transported to cell-extracellular matrix attachments where it is incorporated into fibronectin fibrils. VEGF accumulated at β1 integrin containing fibrillar adhesions and was translocated along the cell surface prior to internalization and degradation. We also found that only the astrocyte-derived, matrix-bound, and not soluble VEGF decreases β1 integrin turnover in fibrillar adhesions. We suggest that polarized VEGF release and ECM remodeling by VEGF secreting cells is key to control the local concentration and signaling of VEGF. Our findings highlight the importance of astrocytes in directing VEGF functions and identify these mechanisms as promising target for angiogenic approaches.
Collapse
Affiliation(s)
| | - Gael Potter
- Department of Neurosciences, University of Geneva, Switzerland
| | - Maria Luisa Guzman-Hernandez
- Section on Molecular Signal Transduction, Program for Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Patrick Salmon
- Department of Neurosciences, University of Geneva, Switzerland
| | | | - Birgit Kastberger
- Department of Cell Physiology and Metabolism, University of Geneva, Switzerland
| | - Tamas Balla
- Section on Molecular Signal Transduction, Program for Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | | | | |
Collapse
|
41
|
Koudelkova P, Weber G, Mikulits W. Liver Sinusoidal Endothelial Cells Escape Senescence by Loss of p19ARF. PLoS One 2015; 10:e0142134. [PMID: 26528722 PMCID: PMC4631446 DOI: 10.1371/journal.pone.0142134] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 10/16/2015] [Indexed: 11/18/2022] Open
Abstract
Liver sinusoidal endothelial cells (LSECs) represent a highly differentiated cell type that lines hepatic sinusoids. LSECs form a discontinuous endothelium due to fenestrations under physiological conditions, which are reduced upon chronic liver injury. Cultivation of rodent LSECs associates with a rapid onset of stress-induced senescence a few days post isolation, which limits genetic and biochemical studies ex vivo. Here we show the establishment of LSECs isolated from p19ARF-/- mice which undergo more than 50 cell doublings in the absence of senescence. Isolated p19ARF-/- LSECs display a cobblestone-like morphology and show the ability of tube formation. Analysis of DNA content revealed a stable diploid phenotype after long-term passaging without a gain of aneuploidy. Notably, p19ARF-/- LSECs express the endothelial markers CD31, vascular endothelial growth factor receptor (VEGFR)-2, VE-cadherin, von Willebrand factor, stabilin-2 and CD146 suggesting that these cells harbor and maintain an endothelial phenotype. In line, treatment with small molecule inhibitors against VEGFR-2 caused cell death, demonstrating the sustained ability of p19ARF-/- LSECs to respond to anti-angiogenic therapeutics. From these data we conclude that loss of p19ARF overcomes senescence of LSECs, allowing immortalization of cells without losing endothelial characteristics. Thus, p19ARF-/- LSECs provide a novel cellular model to study endothelial cell biology.
Collapse
Affiliation(s)
- Petra Koudelkova
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Gerhard Weber
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Mikulits
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- * E-mail:
| |
Collapse
|
42
|
Proksch S, Bittermann G, Vach K, Nitschke R, Tomakidi P, Hellwig E. hMSC-Derived VEGF Release Triggers the Chemoattraction of Alveolar Osteoblasts. Stem Cells 2015; 33:3114-24. [PMID: 26235535 DOI: 10.1002/stem.2119] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 06/30/2015] [Indexed: 12/11/2022]
Abstract
Human mesenchymal stem cells (hMSCs) are promising candidates for regenerative periodontal strategies, due to the broad spectrum of supportive effects on cells and tissues at the site of application. Although positive effects are visible, the understanding of their underlying mechanisms still requires further elucidation. Recently, we have shown that hMSCs are capable to prompt osteogenic differentiation of alveolar osteoblasts, thereby presumably contributing to alveolar bone regeneration. Another issue that is critical in this context is the attraction of hard tissue-forming cells to regeneration sites, but it is an open question whether hMSCs can afford this. In the present manuscript, we show by life cell imaging that in interactive cocultures, hMSCs successfully trigger osteoblast chemotaxis. Gene expression analysis for hMSC-innate chemoattractive biomolecules, orchestrating this process, revealed vascular endothelial growth factor (VEGF), PgE synthase, osteoprotegerin (OPG), monocyte colony-stimulating factor, and transforming growth factor β1, which was confirmed for VEGF and OPG on the protein level. Noteworthy, we showed that only corresponding levels of VEGF but not OPG attracted alveolar osteoblasts similar to hMSC coculture, while VEGF inhibitor abolished both the VEGF and the hMSC-triggered chemoattraction. In summary, we have identified secreted OPG and VEGF proteins as potential chemoattractants, of which further characterization yielded VEGF as a causative for hMSC-directed osteoblast chemotaxis. With respect to the better understanding of potential hMSC-based periodontal regeneration strategies, we propose hMSC-derived VEGF release as a mechanism in the recruitment of hard tissue-forming cells to alveolar bone sites in need of regeneration.
Collapse
Affiliation(s)
- Susanne Proksch
- Department of Operative Dentistry and Periodontology, University Medical Centre Freiburg, Germany
| | - Gido Bittermann
- Department of Oral and Maxillofacial Surgery, University Medical Centre Freiburg, Germany
| | - Kirstin Vach
- Department of Medical Biometry and Medical Informatics, Division of Data Analysis and Modelling, University Medical Centre Freiburg, Germany
| | - Roland Nitschke
- Centre for Biological Systems Analysis, Life Imaging Center, Albert-Ludwigs-University Freiburg, Germany.,BIOSS Centre for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, Germany
| | - Pascal Tomakidi
- Department of Oral Biotechnology, Centre for Dental Medicine, University Medical Centre Freiburg, Germany
| | - Elmar Hellwig
- Department of Operative Dentistry and Periodontology, University Medical Centre Freiburg, Germany
| |
Collapse
|
43
|
Detchokul S, Williams ED, Parker MW, Frauman AG. Tetraspanins as regulators of the tumour microenvironment: implications for metastasis and therapeutic strategies. Br J Pharmacol 2015; 171:5462-90. [PMID: 23731188 DOI: 10.1111/bph.12260] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 05/16/2013] [Accepted: 05/16/2013] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED One of the hallmarks of cancer is the ability to activate invasion and metastasis. Cancer morbidity and mortality are largely related to the spread of the primary, localized tumour to adjacent and distant sites. Appropriate management and treatment decisions based on predicting metastatic disease at the time of diagnosis is thus crucial, which supports better understanding of the metastatic process. There are components of metastasis that are common to all primary tumours: dissociation from the primary tumour mass, reorganization/remodelling of extracellular matrix, cell migration, recognition and movement through endothelial cells and the vascular circulation and lodgement and proliferation within ectopic stroma. One of the key and initial events is the increased ability of cancer cells to move, escaping the regulation of normal physiological control. The cellular cytoskeleton plays an important role in cancer cell motility and active cytoskeletal rearrangement can result in metastatic disease. This active change in cytoskeletal dynamics results in manipulation of plasma membrane and cellular balance between cellular adhesion and motility which in turn determines cancer cell movement. Members of the tetraspanin family of proteins play important roles in regulation of cancer cell migration and cancer-endothelial cell interactions, which are critical for cancer invasion and metastasis. Their involvements in active cytoskeletal dynamics, cancer metastasis and potential clinical application will be discussed in this review. In particular, the tetraspanin member, CD151, is highlighted for its major role in cancer invasion and metastasis. LINKED ARTICLES This article is part of a themed section on Cytoskeleton, Extracellular Matrix, Cell Migration, Wound Healing and Related Topics. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-24.
Collapse
Affiliation(s)
- S Detchokul
- Clinical Pharmacology and Therapeutics Unit, Department of Medicine (Austin Health/Northern Health), The University of Melbourne, Heidelberg, Vic., Australia
| | | | | | | |
Collapse
|
44
|
Valverde A, Peñarando J, Cañas A, López-Sánchez LM, Conde F, Hernández V, Peralbo E, López-Pedrera C, de la Haba-Rodríguez J, Aranda E, Rodríguez-Ariza A. Simultaneous inhibition of EGFR/VEGFR and cyclooxygenase-2 targets stemness-related pathways in colorectal cancer cells. PLoS One 2015; 10:e0131363. [PMID: 26107817 PMCID: PMC4479446 DOI: 10.1371/journal.pone.0131363] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 06/01/2015] [Indexed: 02/06/2023] Open
Abstract
Despite the demonstrated benefits of anti-EGFR/VEGF targeted therapies in metastatic colorectal cancer (mCRC), many patients initially respond, but then show evidence of disease progression. New therapeutic strategies are needed to make the action of available drugs more efficient. Our study aimed to explore whether simultaneous targeting of EGFR/VEGF and cyclooxygenase-2 (COX-2) may aid the treatment and management of mCRC patients. The dual tyrosine kinase inhibitor AEE788 and celecoxib were used to inhibit EGFR/VEGFR and COX-2, respectively, in colorectal cancer cells. COX-2 inhibition with celecoxib augmented the antitumoral and antiangiogenic efficacy of AEE788, as indicated by the inhibition of cell proliferation, induction of apoptosis and G1 cell cycle arrest, down-regulation of VEGF production by cancer cells and reduction of cell migration. These effects were related with a blockade in the EGFR/VEGFR signaling axis. Notably, the combined AEE788/celecoxib treatment prevented β-catenin nuclear accumulation in tumor cells. This effect was associated with a significant downregulation of FOXM1 protein levels and an impairment in the interaction of this transcription factor with β-catenin, which is required for its nuclear localization. Furthermore, the combined treatment also reduced the expression of the stem cell markers Oct 3/4, Nanog, Sox-2 and Snail in cancer cells, and contributed to the diminution of the CSC subpopulation, as indicated by colonosphere formation assays. In conclusion, the combined treatment of AEE788 and celecoxib not only demonstrated enhanced anti-tumoral efficacy in colorectal cancer cells, but also reduced colon CSCs subpopulation by targeting stemness-related pathways. Therefore, the simultaneous targeting of EGFR/VEGF and COX-2 may aid in blocking mCRC progression and improve the efficacy of existing therapies in colorectal cancer.
Collapse
Affiliation(s)
- Araceli Valverde
- Oncology Department, Maimonides Institute of Biomedical Research (IMIBIC), Reina Sofía Hospital, University of Córdoba, Córdoba, Spain
- Spanish Cancer Network (RTICC), Instituto de Salud Carlos III, Madrid, Spain
| | - Jon Peñarando
- Oncology Department, Maimonides Institute of Biomedical Research (IMIBIC), Reina Sofía Hospital, University of Córdoba, Córdoba, Spain
- Spanish Cancer Network (RTICC), Instituto de Salud Carlos III, Madrid, Spain
| | - Amanda Cañas
- Oncology Department, Maimonides Institute of Biomedical Research (IMIBIC), Reina Sofía Hospital, University of Córdoba, Córdoba, Spain
- Spanish Cancer Network (RTICC), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura M. López-Sánchez
- Oncology Department, Maimonides Institute of Biomedical Research (IMIBIC), Reina Sofía Hospital, University of Córdoba, Córdoba, Spain
- Spanish Cancer Network (RTICC), Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco Conde
- Oncology Department, Maimonides Institute of Biomedical Research (IMIBIC), Reina Sofía Hospital, University of Córdoba, Córdoba, Spain
- Spanish Cancer Network (RTICC), Instituto de Salud Carlos III, Madrid, Spain
| | - Vanessa Hernández
- Oncology Department, Maimonides Institute of Biomedical Research (IMIBIC), Reina Sofía Hospital, University of Córdoba, Córdoba, Spain
- Spanish Cancer Network (RTICC), Instituto de Salud Carlos III, Madrid, Spain
| | - Esther Peralbo
- Research Unit, Maimonides Institute of Biomedical Research (IMIBIC), Reina Sofía Hospital, University of Córdoba, Córdoba, Spain
| | - Chary López-Pedrera
- Research Unit, Maimonides Institute of Biomedical Research (IMIBIC), Reina Sofía Hospital, University of Córdoba, Córdoba, Spain
| | - Juan de la Haba-Rodríguez
- Oncology Department, Maimonides Institute of Biomedical Research (IMIBIC), Reina Sofía Hospital, University of Córdoba, Córdoba, Spain
- Spanish Cancer Network (RTICC), Instituto de Salud Carlos III, Madrid, Spain
| | - Enrique Aranda
- Oncology Department, Maimonides Institute of Biomedical Research (IMIBIC), Reina Sofía Hospital, University of Córdoba, Córdoba, Spain
- Spanish Cancer Network (RTICC), Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Rodríguez-Ariza
- Oncology Department, Maimonides Institute of Biomedical Research (IMIBIC), Reina Sofía Hospital, University of Córdoba, Córdoba, Spain
- Spanish Cancer Network (RTICC), Instituto de Salud Carlos III, Madrid, Spain
- * E-mail:
| |
Collapse
|
45
|
Heparin/Heparan sulfate proteoglycans glycomic interactome in angiogenesis: biological implications and therapeutical use. Molecules 2015; 20:6342-88. [PMID: 25867824 PMCID: PMC6272510 DOI: 10.3390/molecules20046342] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 03/31/2015] [Accepted: 04/01/2015] [Indexed: 12/20/2022] Open
Abstract
Angiogenesis, the process of formation of new blood vessel from pre-existing ones, is involved in various intertwined pathological processes including virus infection, inflammation and oncogenesis, making it a promising target for the development of novel strategies for various interventions. To induce angiogenesis, angiogenic growth factors (AGFs) must interact with pro-angiogenic receptors to induce proliferation, protease production and migration of endothelial cells (ECs). The action of AGFs is counteracted by antiangiogenic modulators whose main mechanism of action is to bind (thus sequestering or masking) AGFs or their receptors. Many sugars, either free or associated to proteins, are involved in these interactions, thus exerting a tight regulation of the neovascularization process. Heparin and heparan sulfate proteoglycans undoubtedly play a pivotal role in this context since they bind to almost all the known AGFs, to several pro-angiogenic receptors and even to angiogenic inhibitors, originating an intricate network of interaction, the so called "angiogenesis glycomic interactome". The decoding of the angiogenesis glycomic interactome, achievable by a systematic study of the interactions occurring among angiogenic modulators and sugars, may help to design novel antiangiogenic therapies with implications in the cure of angiogenesis-dependent diseases.
Collapse
|
46
|
Koivisto L, Heino J, Häkkinen L, Larjava H. Integrins in Wound Healing. Adv Wound Care (New Rochelle) 2014; 3:762-783. [PMID: 25493210 DOI: 10.1089/wound.2013.0436] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Indexed: 01/06/2023] Open
Abstract
Significance: Regulation of cell adhesions during tissue repair is fundamentally important for cell migration, proliferation, and protein production. All cells interact with extracellular matrix proteins with cell surface integrin receptors that convey signals from the environment into the nucleus, regulating gene expression and cell behavior. Integrins also interact with a variety of other proteins, such as growth factors, their receptors, and proteolytic enzymes. Re-epithelialization and granulation tissue formation are crucially dependent on the temporospatial function of multiple integrins. This review explains how integrins function in wound repair. Recent Advances: Certain integrins can activate latent transforming growth factor beta-1 (TGF-β1) that modulates wound inflammation and granulation tissue formation. Dysregulation of TGF-β1 function is associated with scarring and fibrotic disorders. Therefore, these integrins represent targets for therapeutic intervention in fibrosis. Critical Issues: Integrins have multifaceted functions and extensive crosstalk with other cell surface receptors and molecules. Moreover, in aberrant healing, integrins may assume different functions, further increasing the complexity of their functionality. Discovering and understanding the role that integrins play in wound healing provides an opportunity to identify the mechanisms for medical conditions, such as excessive scarring, chronic wounds, and even cancer. Future Directions: Integrin functions in acute and chronic wounds should be further addressed in models better mimicking human wounds. Application of any products in acute or chronic wounds will potentially alter integrin functions that need to be carefully considered in the design.
Collapse
Affiliation(s)
- Leeni Koivisto
- Laboratory of Periodontal Biology, Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Jyrki Heino
- Department of Biochemistry, University of Turku, Turku, Finland
| | - Lari Häkkinen
- Laboratory of Periodontal Biology, Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Hannu Larjava
- Laboratory of Periodontal Biology, Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| |
Collapse
|
47
|
Lee E, Lee SJ, Koskimaki JE, Han Z, Pandey NB, Popel AS. Inhibition of breast cancer growth and metastasis by a biomimetic peptide. Sci Rep 2014; 4:7139. [PMID: 25409905 PMCID: PMC4238022 DOI: 10.1038/srep07139] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 11/05/2014] [Indexed: 12/21/2022] Open
Abstract
Metastasis is the main cause of mortality in cancer patients. Though there are many anti-cancer drugs targeting primary tumor growth, anti-metastatic agents are rarely developed. Angiogenesis and lymphangiogenesis are crucial for cancer progression, particularly, lymphangiogenesis is pivotal for metastasis in breast cancer. Here we report that a novel collagen IV derived biomimetic peptide inhibits breast cancer growth and metastasis by blocking angiogenesis and lymphangiogenesis. The peptide inhibits blood and lymphatic endothelial cell viability, migration, adhesion, and tube formation by targeting IGF1R and Met signals. The peptide blocks MDA-MB-231 tumor growth by inhibiting tumor angiogenesis in vivo. Moreover, the peptide inhibits lymphangiogenesis in primary tumors. MDA-MB-231 tumor conditioned media (TCM) was employed to accelerate spontaneous metastasis in tumor xenografts, and the anti-metastatic activity of the peptide was tested in this model. The peptide prevents metastasis to the lungs and lymph nodes by inhibiting TCM-induced lymphangiogenesis and angiogenesis in the pre-metastatic organs. In summary, a novel biomimetic peptide inhibits breast cancer growth and metastasis by blocking angiogenesis and lymphangiogenesis in the pre-metastatic organs as well as primary tumors.
Collapse
Affiliation(s)
- Esak Lee
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Seung Jae Lee
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Jacob E Koskimaki
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Zheyi Han
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Niranjan B Pandey
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Aleksander S Popel
- 1] Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States [2] Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
| |
Collapse
|
48
|
Moreno-Layseca P, Streuli CH. Signalling pathways linking integrins with cell cycle progression. Matrix Biol 2014; 34:144-53. [DOI: 10.1016/j.matbio.2013.10.011] [Citation(s) in RCA: 158] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 10/22/2013] [Accepted: 10/22/2013] [Indexed: 12/30/2022]
|
49
|
Liu Y, Li Z, Song T, Xue Z, Zhang Z. Mechanisms of anti-leukemic activity of the Bcl-2 homology domain-3 mimetic S1. Biomed Pharmacother 2013; 67:583-91. [DOI: 10.1016/j.biopha.2013.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 04/19/2013] [Indexed: 10/26/2022] Open
|
50
|
Larusch GA, Merkulova A, Mahdi F, Shariat-Madar Z, Sitrin RG, Cines DB, Schmaier AH. Domain 2 of uPAR regulates single-chain urokinase-mediated angiogenesis through β1-integrin and VEGFR2. Am J Physiol Heart Circ Physiol 2013; 305:H305-20. [PMID: 23709605 DOI: 10.1152/ajpheart.00110.2013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
How single-chain urokinase (ScuPA) mediates angiogenesis is incompletely understood. ScuPA (≥4 nM) induces phosphorylated (p)ERK1/2 (MAPK44 and MAPK42) and pAkt (Ser(473)) in umbilical vein and dermal microvascular endothelial cells. Activation of pERK1/2 by ScuPA is blocked by PD-98059 or U-0126, and pAkt (Ser(473)) activation is inhibited by wortmannin or LY-294002. ScuPA (32 nM) or protease-inhibited two-chain urokinase stimulates pERK1/2 to the same extent, indicating that signaling is not dependent on enzymatic activity. ScuPA induces pERK1/2, but not pAkt (Ser(473)), in SIN1(-/-) cells, indicating that the two pathways are not identical. Peptides from domain 2 of the urokinase plasminogen activator receptor (uPAR) or domain 5 of high-molecular-weight kininogen compete with ScuPA for the induction of pERK1/2 and pAkt (Ser(473)). A peptide of the integrin-binding site on uPAR, a β1-integrin peptide that binds uPAR, antibody 6S6 to β1-integrin, tyrosine kinase inhibitors AG-1478 or PP3, and small interfering RNA knockdown of VEFG receptor 2, but not HER1-HER4, blocked ScuPA-induced pERK1/2 and pAkt (Ser(473)). ScuPA-induced endothelial cell proliferation was blocked by inhibitors of pERK1/2 and pAkt (Ser(473)), antibody 6S6, and uPAR or kininogen peptides. ScuPA initiated aortic sprouts and Matrigel plug angiogenesis in normal, but not uPAR-deficient, mouse aortae or mice, respectively, but these were blocked by PD-98059, LY-294002, AG-1478, or cleaved high-molecular-weight kininogen. In summary, this investigation indicates a novel, a nonproteolytic signaling pathway initiated by zymogen ScuPA and mediated by domain 2 of uPAR, β1-integrins, and VEGF receptor 2 leading to angiogenesis. Kininogens or peptides from it downregulate this pathway.
Collapse
Affiliation(s)
- Gretchen A Larusch
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | | | | | | | | | |
Collapse
|