1
|
Lee C, Kim MJ, Kumar A, Lee HW, Yang Y, Kim Y. Vascular endothelial growth factor signaling in health and disease: from molecular mechanisms to therapeutic perspectives. Signal Transduct Target Ther 2025; 10:170. [PMID: 40383803 PMCID: PMC12086256 DOI: 10.1038/s41392-025-02249-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/09/2025] [Accepted: 04/21/2025] [Indexed: 05/20/2025] Open
Abstract
Vascular endothelial growth factor (VEGF) signaling is a critical regulator of vasculogenesis, angiogenesis, and lymphangiogenesis, processes that are vital for the development of vascular and lymphatic systems, tissue repair, and the maintenance of homeostasis. VEGF ligands and their receptors orchestrate endothelial cell proliferation, migration, and survival, playing a pivotal role in dynamic vascular remodeling. Dysregulated VEGF signaling drives diverse pathological conditions, including tumor angiogenesis, cardiovascular diseases, and ocular disorders. Excessive VEGF activity promotes tumor growth, invasion, and metastasis, while insufficient signaling contributes to impaired wound healing and ischemic diseases. VEGF-targeted therapies, such as monoclonal antibodies and tyrosine kinase inhibitors, have revolutionized the treatment of diseases involving pathological angiogenesis, offering significant clinical benefits in oncology and ophthalmology. These therapies inhibit angiogenesis and slow disease progression, but they often face challenges such as therapeutic resistance, suboptimal efficacy, and adverse effects. To further explore these issues, this review provides a comprehensive overview of VEGF ligands and receptors, elucidating their molecular mechanisms and regulatory networks. It evaluates the latest progress in VEGF-targeted therapies and examines strategies to address current challenges, such as resistance mechanisms. Moreover, the discussion includes emerging therapeutic strategies such as innovative drug delivery systems and combination therapies, highlighting the continuous efforts to improve the effectiveness and safety of VEGF-targeted treatments. This review highlights the translational potential of recent discoveries in VEGF biology for improving patient outcomes.
Collapse
Affiliation(s)
- Chunsik Lee
- Department of R&D, GEMCRO Inc, Seoul, Republic of Korea.
| | - Myung-Jin Kim
- Department of Biological Sciences and Research Institute of Women's Health, Sookmyung Women's University, Seoul, Republic of Korea
| | - Anil Kumar
- Center for Research and Innovations, Adichunchanagiri University, Mandya, Karnataka, India
| | - Han-Woong Lee
- Department of R&D, GEMCRO Inc, Seoul, Republic of Korea
| | - Yunlong Yang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yonghwan Kim
- Department of Biological Sciences and Research Institute of Women's Health, Sookmyung Women's University, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Guo Y, Dupart M, Irondelle M, Peraldi P, Bost F, Mazure NM. YAP1 modulation of primary cilia-mediated ciliogenesis in 2D and 3D prostate cancer models. FEBS Lett 2024; 598:3071-3086. [PMID: 39424416 DOI: 10.1002/1873-3468.15029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/10/2024] [Accepted: 09/16/2024] [Indexed: 10/21/2024]
Abstract
The primary cilium, a non-motile organelle present in most human cells, plays a crucial role in detecting microenvironmental changes and regulating intracellular signaling. Its dysfunction is linked to various diseases, including cancer. We explored the role of ciliated cells in prostate cancer by using Gefitinib and Jasplakinolide compounds to induce ciliated cells in both normal and tumor-like prostate cell lines. We assessed GLI1 and IFT20 expression and investigated YAP1 protein's role, which is implicated in primary cilium regulation. Finally, we examined these compounds in 3D cell models, aiming to simulate in vivo conditions. Our study highlights YAP1 as a potential target for novel genetic models to understand the primary cilium's role in mediating resistance to anticancer treatments.
Collapse
Affiliation(s)
- Yingbo Guo
- INSERM U1065, C3M, Université Côte d'Azur, Nice Cedex 03, France
- Equipe Labellisée Ligue Contre le Cancer, Xxxxx, France
| | - Mathilde Dupart
- INSERM U1065, C3M, Université Côte d'Azur, Nice Cedex 03, France
- Equipe Labellisée Ligue Contre le Cancer, Xxxxx, France
- IRCAN, Université Côte d'Azur, Nice Cedex 02, France
| | - Marie Irondelle
- INSERM U1065, C3M, Université Côte d'Azur, Nice Cedex 03, France
| | - Pascal Peraldi
- INSERM U1065, C3M, Université Côte d'Azur, Nice Cedex 03, France
- Equipe Labellisée Ligue Contre le Cancer, Xxxxx, France
| | - Frederic Bost
- INSERM U1065, C3M, Université Côte d'Azur, Nice Cedex 03, France
- Equipe Labellisée Ligue Contre le Cancer, Xxxxx, France
| | - Nathalie M Mazure
- INSERM U1065, C3M, Université Côte d'Azur, Nice Cedex 03, France
- Equipe Labellisée Ligue Contre le Cancer, Xxxxx, France
| |
Collapse
|
3
|
Ferraretti G, Abondio P, Alberti M, Dezi A, Sherpa PT, Cocco P, Tiriticco M, Di Marcello M, Gnecchi-Ruscone GA, Natali L, Corcelli A, Marinelli G, Peluzzi D, Sarno S, Sazzini M. Archaic introgression contributed to shape the adaptive modulation of angiogenesis and cardiovascular traits in human high-altitude populations from the Himalayas. eLife 2024; 12:RP89815. [PMID: 39513938 PMCID: PMC11548878 DOI: 10.7554/elife.89815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
It is well established that several Homo sapiens populations experienced admixture with extinct human species during their evolutionary history. Sometimes, such a gene flow could have played a role in modulating their capability to cope with a variety of selective pressures, thus resulting in archaic adaptive introgression events. A paradigmatic example of this evolutionary mechanism is offered by the EPAS1 gene, whose most frequent haplotype in Himalayan highlanders was proved to reduce their susceptibility to chronic mountain sickness and to be introduced in the gene pool of their ancestors by admixture with Denisovans. In this study, we aimed at further expanding the investigation of the impact of archaic introgression on more complex adaptive responses to hypobaric hypoxia evolved by populations of Tibetan/Sherpa ancestry, which have been plausibly mediated by soft selective sweeps and/or polygenic adaptations rather than by hard selective sweeps. For this purpose, we used a combination of composite-likelihood and gene network-based methods to detect adaptive loci in introgressed chromosomal segments from Tibetan WGS data and to shortlist those presenting Denisovan-like derived alleles that participate to the same functional pathways and are absent in populations of African ancestry, which are supposed to do not have experienced Denisovan admixture. According to this approach, we identified multiple genes putatively involved in archaic introgression events and that, especially as regards TBC1D1, RASGRF2, PRKAG2, and KRAS, have plausibly contributed to shape the adaptive modulation of angiogenesis and of certain cardiovascular traits in high-altitude Himalayan peoples. These findings provided unprecedented evidence about the complexity of the adaptive phenotype evolved by these human groups to cope with challenges imposed by hypobaric hypoxia, offering new insights into the tangled interplay of genetic determinants that mediates the physiological adjustments crucial for human adaptation to the high-altitude environment.
Collapse
Affiliation(s)
- Giulia Ferraretti
- Laboratory of Molecular Anthropology and Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of BolognaBolognaItaly
| | - Paolo Abondio
- Department of Cultural Heritage, Ravenna Campus, University of BolognaBolognaItaly
| | - Marta Alberti
- Laboratory of Molecular Anthropology and Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of BolognaBolognaItaly
| | - Agnese Dezi
- Department of Emergency and Organ Transplantation, University of Bari Aldo MoroBari Aldo MoroItaly
| | | | - Paolo Cocco
- Explora Nunaat International, Montorio al VomanoTeramoItaly
| | | | | | | | - Luca Natali
- Explora Nunaat International, Montorio al VomanoTeramoItaly
- Italian Institute of Human PaleontologyRomeItaly
| | - Angela Corcelli
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari Aldo MoroBariItaly
| | | | - Davide Peluzzi
- Explora Nunaat International, Montorio al VomanoTeramoItaly
| | - Stefania Sarno
- Laboratory of Molecular Anthropology and Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of BolognaBolognaItaly
| | - Marco Sazzini
- Laboratory of Molecular Anthropology and Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of BolognaBolognaItaly
- Interdepartmental Centre Alma Mater Research Institute on Global Changes and Climate Change, University of BolognaBolognaItaly
| |
Collapse
|
4
|
Bazsó A, Szodoray P, Shoenfeld Y, Kiss E. Biomarkers reflecting the pathogenesis, clinical manifestations, and guide therapeutic approach in systemic sclerosis: a narrative review. Clin Rheumatol 2024; 43:3055-3072. [PMID: 39210206 PMCID: PMC11442557 DOI: 10.1007/s10067-024-07123-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 08/17/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Systemic sclerosis (SSc) is a progressive autoimmune disorder that mainly affects the skin. There are other clinical manifestations as renal, pulmonary, cardiovascular, and gastrointestinal tract involvements. Based on the skin involvement there are two subtypes of SSc, as limited cutaneous SSc (lSSc) which involves the acral part of the body and diffuse cutaneous SSc (dSSc) resulting in significant skin thickening of the body. Despite of the extensive research the pathomechanism is not fully clarified, how Ssc develops, moreover identifying biomarkers to predict the clinical outcome and prognosis still remains challenging. Circulating biomarkers can be crucial to define the diagnosis, to predict the prognosis and monitor the clinical course. However, only some patients are responsive to the therapy in SSc, and there is a need to reach the ideal therapy for any individual to prevent or slow down the progression in early stages of the disease. In this narrative review, our purpose was to summarize the potential biomarkers in Ssc, describe their role in the diagnosis, pathomechanism, clinical course, organ manifestations, as well as the response to the therapy. Biomarkers assessment aids in the evaluation of disease progression, and disease outcome.
Collapse
Affiliation(s)
- Anna Bazsó
- Department of Clinical Immunology, Adult and Paediatric Rheumatology, National Institute of Locomotor System Disorders and Disabilities, Budapest, Hungary.
| | - Péter Szodoray
- Department of Immunology, Oslo University Hospital, Rikshospitalet and University of Oslo, Oslo, Norway
| | - Yehuda Shoenfeld
- Reichmann University, Herzelia, Israel
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, 5265601, Tel-Hashomer, Israel
| | - Emese Kiss
- Department of Clinical Immunology, Adult and Paediatric Rheumatology, National Institute of Locomotor System Disorders and Disabilities, Budapest, Hungary
- Division of Locomotor System and Rheumatology Prevention, Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
5
|
Xie D, Liu M, Lin Y, Liu X, Yan H. Silencing of topical proline hydroxylase domain 2 promotes the healing of rat diabetic wounds by phosphorylating AMPK. PLoS One 2023; 18:e0294566. [PMID: 38039326 PMCID: PMC10691724 DOI: 10.1371/journal.pone.0294566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 11/04/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND For diabetic ulcers, the impaired response to hypoxia is a key feature associated with delayed healing. In the early phase of hypoxia, hypoxic signaling activates the AMPK system through direct phosphorylation of the PHD2 pathway, producing a significant endogenous hypoxic protective effect. METHODS Twenty Sprague-Dawley (SD) rats were randomly divided into two groups: treatment (sh-PHD2) and control (sh-Control). Using lentiviral encapsulation of PHD2-shRNA and transfection, the silencing efficiency of PHD2 expression was verified in rat dermal fibroblasts (RDF) and in rat aortic endothelial cells (RAECs). Changes in the ability of RDF and RAECs to proliferate, migrate, and in the rate of ATP production were observed and then tested after inhibition of AMPK phosphorylation using dorsomorphin. The lentiviral preparation was injected directly into the wounds of rats and wound healing was recorded periodically to calculate the healing rate. Wounded tissues were excised after 14 days and the efficiency of PHD2 silencing, as well as the expression of growth factors, was examined using molecular biology methods. Histological examination was performed to assess CD31 expression and therefore determine effects on angiogenesis. RESULTS Lentiviral-encapsulated PHD2-sh-RNA effectively suppressed PHD2 expression and improved the proliferation, migration, and ATP production rate of RDF and RAEC, which were restored to their previous levels after inhibition of AMPK. The rate of wound healing, vascular growth, and expression of growth factors were significantly improved in diabetic-model rats after local silencing of PHD2 expression. CONCLUSION Silencing of PHD2 promoted wound healing in diabetic-model SD rats by activating AMPK phosphorylation.
Collapse
Affiliation(s)
- Defu Xie
- Southwest Medical University, No. 1 Section 1, Xianglin Road, Luzhou City, Sichuan Province, 646000, China
- National Key Clinical Construction Specialty, Wound Repair & Regeneration Laboratory, Department of Plastic & Burn Surgery, Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou,Sichuan Province, 646000, China
| | - Mengchang Liu
- Southwest Medical University, No. 1 Section 1, Xianglin Road, Luzhou City, Sichuan Province, 646000, China
- National Key Clinical Construction Specialty, Wound Repair & Regeneration Laboratory, Department of Plastic & Burn Surgery, Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou,Sichuan Province, 646000, China
| | - Yingxi Lin
- Southwest Medical University, No. 1 Section 1, Xianglin Road, Luzhou City, Sichuan Province, 646000, China
- National Key Clinical Construction Specialty, Wound Repair & Regeneration Laboratory, Department of Plastic & Burn Surgery, Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou,Sichuan Province, 646000, China
| | - Xingke Liu
- Southwest Medical University, No. 1 Section 1, Xianglin Road, Luzhou City, Sichuan Province, 646000, China
- National Key Clinical Construction Specialty, Wound Repair & Regeneration Laboratory, Department of Plastic & Burn Surgery, Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou,Sichuan Province, 646000, China
| | - Hong Yan
- Southwest Medical University, No. 1 Section 1, Xianglin Road, Luzhou City, Sichuan Province, 646000, China
- National Key Clinical Construction Specialty, Wound Repair & Regeneration Laboratory, Department of Plastic & Burn Surgery, Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou,Sichuan Province, 646000, China
| |
Collapse
|
6
|
Kong C, Chen X. Combined Photodynamic and Photothermal Therapy and Immunotherapy for Cancer Treatment: A Review. Int J Nanomedicine 2022; 17:6427-6446. [PMID: 36540374 PMCID: PMC9760263 DOI: 10.2147/ijn.s388996] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 12/03/2022] [Indexed: 12/23/2022] Open
Abstract
Photoactivation therapy based on photodynamic therapy (PDT) and photothermal therapy (PTT) has been identified as a tumour ablation modality for numerous cancer indications, with photosensitisers and photothermal conversion agents playing important roles in the phototherapy process, especially in recent decades. In addition, the iteration of nanotechnology has strongly promoted the development of phototherapy in tumour treatment. PDT can increase the sensitivity of tumour cells to PTT by interfering with the tumour microenvironment, whereas the heat generated by PTT can increase blood flow, improve oxygen supply and enhance the PDT therapeutic effect. In addition, tumour cell debris generated by phototherapy can serve as tumour-associated antigens, evoking antitumor immune responses. In this review, the research progress of phototherapy, and its research effects in combination with immunotherapy on the treatment of tumours are mainly outlined, and issues that may need continued attention in the future are raised.
Collapse
Affiliation(s)
- Cunqing Kong
- Department of medical imaging center, central hospital affiliated to Shandong first medical university, Jinan, People’s Republic of China
| | - Xingcai Chen
- Department of Human Anatomy and Center for Genomics and Personalized Medicine, Nanning, People’s Republic of China,Correspondence: Xingcai Chen, Email
| |
Collapse
|
7
|
Noohi P, Abdekhodaie MJ, Nekoofar MH, Galler KM, Dummer PMH. Advances in Scaffolds Used for Pulp-Dentine Complex Tissue Engineering - A Narrative Review. Int Endod J 2022; 55:1277-1316. [PMID: 36039729 DOI: 10.1111/iej.13826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 07/28/2022] [Accepted: 08/10/2022] [Indexed: 11/27/2022]
Abstract
Pulp necrosis in immature teeth disrupts root development and predisposes roots to fracture as a consequence of their thin walls and open apices. Regenerative endodontics is a developing treatment modality whereby necrotic pulps are replaced with newly formed healthy tissue inside the root canal. Many clinical studies have demonstrated the potential of this strategy to stimulate root maturation and apical root-end closure. However, clinical outcomes are patient-dependent and unpredictable. The development of predictable clinical protocols is achieved through the interplay of the three classical elements of tissue engineering, namely, stem cells, signaling molecules, and scaffolds. Scaffolds provide structural support for cells to adhere and proliferate and also regulate cell differentiation and metabolism. Hence, designing and fabricating an appropriate scaffold is a crucial step in tissue engineering. In this review, four main classes of scaffolds used to engineer pulp-dentine complexes, including bioceramic-based scaffolds, synthetic polymer-based scaffolds, natural polymer-based scaffolds, and composite scaffolds, are covered. Additionally, recent advances in the design, fabrication, and application of such scaffolds are analysed along with their advantages and limitations. Finally, the importance of vascular network establishment in the success of pulp-dentine complex regeneration and strategies used to create scaffolds to address this challenge are discussed.
Collapse
Affiliation(s)
- Parisa Noohi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Mohammad J Abdekhodaie
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Mohammad H Nekoofar
- Department of Endodontics, School of Dentistry, Tehran University of Medical Sciences Tehran University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Endodontic, Bahçeşehir University School of Dentistry, Istanbul, Turkey
| | - Kerstin M Galler
- Department of Conservative Dentistry and Periodontology, University Hospital Erlangen-Nürnberg, Erlangen, Germany
| | - Paul M H Dummer
- School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| |
Collapse
|
8
|
Hypoxia signaling in human health and diseases: implications and prospects for therapeutics. Signal Transduct Target Ther 2022; 7:218. [PMID: 35798726 PMCID: PMC9261907 DOI: 10.1038/s41392-022-01080-1] [Citation(s) in RCA: 183] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/17/2022] [Accepted: 06/23/2022] [Indexed: 02/07/2023] Open
Abstract
Molecular oxygen (O2) is essential for most biological reactions in mammalian cells. When the intracellular oxygen content decreases, it is called hypoxia. The process of hypoxia is linked to several biological processes, including pathogenic microbe infection, metabolic adaptation, cancer, acute and chronic diseases, and other stress responses. The mechanism underlying cells respond to oxygen changes to mediate subsequent signal response is the central question during hypoxia. Hypoxia-inducible factors (HIFs) sense hypoxia to regulate the expressions of a series of downstream genes expression, which participate in multiple processes including cell metabolism, cell growth/death, cell proliferation, glycolysis, immune response, microbe infection, tumorigenesis, and metastasis. Importantly, hypoxia signaling also interacts with other cellular pathways, such as phosphoinositide 3-kinase (PI3K)-mammalian target of rapamycin (mTOR) signaling, nuclear factor kappa-B (NF-κB) pathway, extracellular signal-regulated kinases (ERK) signaling, and endoplasmic reticulum (ER) stress. This paper systematically reviews the mechanisms of hypoxia signaling activation, the control of HIF signaling, and the function of HIF signaling in human health and diseases. In addition, the therapeutic targets involved in HIF signaling to balance health and diseases are summarized and highlighted, which would provide novel strategies for the design and development of therapeutic drugs.
Collapse
|
9
|
Impact of Zinc on Oxidative Signaling Pathways in the Development of Pulmonary Vasoconstriction Induced by Hypobaric Hypoxia. Int J Mol Sci 2022; 23:ijms23136974. [PMID: 35805984 PMCID: PMC9266543 DOI: 10.3390/ijms23136974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/14/2022] [Accepted: 06/20/2022] [Indexed: 02/04/2023] Open
Abstract
Hypobaric hypoxia is a condition that occurs at high altitudes (>2500 m) where the partial pressure of gases, particularly oxygen (PO2), decreases. This condition triggers several physiological and molecular responses. One of the principal responses is pulmonary vascular contraction, which seeks to optimize gas exchange under this condition, known as hypoxic pulmonary vasoconstriction (HPV); however, when this physiological response is exacerbated, it contributes to the development of high-altitude pulmonary hypertension (HAPH). Increased levels of zinc (Zn2+) and oxidative stress (known as the “ROS hypothesis”) have been demonstrated in the vasoconstriction process. Therefore, the aim of this review is to determine the relationship between molecular pathways associated with altered Zn2+ levels and oxidative stress in HPV in hypobaric hypoxic conditions. The results indicate an increased level of Zn2+, which is related to increasing mitochondrial ROS (mtROS), alterations in nitric oxide (NO), metallothionein (MT), zinc-regulated, iron-regulated transporter-like protein (ZIP), and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-induced protein kinase C epsilon (PKCε) activation in the development of HPV. In conclusion, there is an association between elevated Zn2+ levels and oxidative stress in HPV under different models of hypoxia, which contribute to understanding the molecular mechanism involved in HPV to prevent the development of HAPH.
Collapse
|
10
|
Perkins EJ, Woolard EA, Garcia-Reyero N. Integration of Adverse Outcome Pathways, Causal Networks and ‘Omics to Support Chemical Hazard Assessment. FRONTIERS IN TOXICOLOGY 2022; 4:786057. [PMID: 35399296 PMCID: PMC8987526 DOI: 10.3389/ftox.2022.786057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 02/14/2022] [Indexed: 12/30/2022] Open
Abstract
Several approaches have been used in an attempt to simplify and codify the events that lead to adverse effects of chemicals including systems biology, ‘omics, in vitro assays and frameworks such as the Adverse Outcome Pathway (AOP). However, these approaches are generally not integrated despite their complementary nature. Here we propose to integrate toxicogenomics data, systems biology information and AOPs using causal biological networks to define Key Events in AOPs. We demonstrate this by developing a causal subnetwork of 28 nodes that represents the Key Event of regenerative proliferation – a critical event in AOPs for liver cancer. We then assessed the effects of three chemicals known to cause liver injury and cell proliferation (carbon tetrachloride, aflatoxin B1, thioacetamide) and two with no known cell proliferation effects (diazepam, simvastatin) on the subnetwork using rat liver gene expression data from the toxicogenomic database Open TG-GATEs. Cyclin D1 (Ccnd1), a gene both causally linked to and sufficient to infer regenerative proliferation activity, was overexpressed after exposures to carbon tetrachloride, aflatoxin B1 and thioacetamide, but not in exposures to diazepam and simvastatin. These results were consistent with known effects on rat livers and liver pathology of exposed rats. Using these approaches, we demonstrate that transcriptomics, AOPs and systems biology can be applied to examine the presence and progression of AOPs in order to better understand the hazards of chemical exposure.
Collapse
Affiliation(s)
- Edward J. Perkins
- Environmental Laboratory, US Army Engineering Research and Development Center, Vicksburg, MS, United States
- *Correspondence: Edward J. Perkins,
| | - E. Alice Woolard
- UNC School of Medicine, University of North Carolina Chapel Hill, Chapel Hill, NC, United States
| | - Natàlia Garcia-Reyero
- Environmental Laboratory, US Army Engineering Research and Development Center, Vicksburg, MS, United States
| |
Collapse
|
11
|
Ferrisse TM, de Oliveira AB, Surur AK, Buzo HS, Brighenti FL, Fontana CR. Photodynamic therapy associated with nanomedicine strategies for treatment of human squamous cell carcinoma: A systematic review and meta-analysis. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 40:102505. [PMID: 34902550 DOI: 10.1016/j.nano.2021.102505] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 10/23/2021] [Accepted: 11/13/2021] [Indexed: 12/19/2022]
Abstract
A systematic review and meta-analysis were conducted about photodynamic therapy (PDT) associated with nanomedicine approaches in the treatment of human squamous cell carcinoma (HSSC). Independent reviewers conducted all steps in the systematic review. For evaluating the risk of bias, RoB 2, OHAT and SYRCLE tools were used. Meta-analysis was performed using a random-effect model (α = 0.05). For PDT against HSSC, Protoporphyrin IX was the photosensitizer, and liposomes were the nanomaterial more frequently used. Photosensitizers conjugated with nanoparticles exhibited positive results against HSSC. Tumors treated with PDT in combination with a nanotechnology drug-delivery system had an increased capacity for inhibiting the tumor growth rate (51.93%/P < 0.0001) when compared with PDT only. Thus, the PDT associated with nanomedicine approaches against HSCC could be a significant option for use in future clinical studies, particularly due to improved results in tumor growth inhibition.
Collapse
Affiliation(s)
- Túlio Morandin Ferrisse
- UNESP-São Paulo State University, School of School of Dentistry-Department of Dental Materials and Prosthodontics, Araraquara, São Paulo, Brazil.
| | - Analú Barros de Oliveira
- UNESP-São Paulo State University, School of Dentistry-Department of Orthodontics and Pediatric Dentistry, Araraquara, São Paulo, Brazil
| | - Amanda Koberstain Surur
- UNESP-São Paulo State University, School of Pharmaceutical Sciences-Department of Clinical Analysis, Araraquara, SP, Brazil
| | - Helen Sordi Buzo
- UNESP-São Paulo State University, School of Pharmaceutical Sciences-Department of Clinical Analysis, Araraquara, SP, Brazil
| | - Fernanda Lourenção Brighenti
- UNESP-São Paulo State University, School of Dentistry-Department of Orthodontics and Pediatric Dentistry, Araraquara, São Paulo, Brazil
| | - Carla Raquel Fontana
- UNESP-São Paulo State University, School of Pharmaceutical Sciences-Department of Clinical Analysis, Araraquara, SP, Brazil.
| |
Collapse
|
12
|
Shi M, Zhou X, Cai Y, Li P, Qin D, Yan X, Du M, Li S, Xu D. Inhibition mechanism of hydroxyproline-like small inhibitors to disorder HIF-VHL interaction by molecular dynamic simulations and binding free energy calculations. CHINESE J CHEM PHYS 2021. [DOI: 10.1063/1674-0068/cjcp2110198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Mingsong Shi
- State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Xin Zhou
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Yao Cai
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Penghui Li
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Dengxue Qin
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Xinrong Yan
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Meng Du
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Shuo Li
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Dingguo Xu
- College of Chemistry, Sichuan University, Chengdu 610064, China
- Research Center for Material Genome Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
13
|
Uemura A, Fruttiger M, D'Amore PA, De Falco S, Joussen AM, Sennlaub F, Brunck LR, Johnson KT, Lambrou GN, Rittenhouse KD, Langmann T. VEGFR1 signaling in retinal angiogenesis and microinflammation. Prog Retin Eye Res 2021; 84:100954. [PMID: 33640465 PMCID: PMC8385046 DOI: 10.1016/j.preteyeres.2021.100954] [Citation(s) in RCA: 187] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 02/12/2021] [Accepted: 02/19/2021] [Indexed: 12/13/2022]
Abstract
Five vascular endothelial growth factor receptor (VEGFR) ligands (VEGF-A, -B, -C, -D, and placental growth factor [PlGF]) constitute the VEGF family. VEGF-A binds VEGF receptors 1 and 2 (VEGFR1/2), whereas VEGF-B and PlGF only bind VEGFR1. Although much research has been conducted on VEGFR2 to elucidate its key role in retinal diseases, recent efforts have shown the importance and involvement of VEGFR1 and its family of ligands in angiogenesis, vascular permeability, and microinflammatory cascades within the retina. Expression of VEGFR1 depends on the microenvironment, is differentially regulated under hypoxic and inflammatory conditions, and it has been detected in retinal and choroidal endothelial cells, pericytes, retinal and choroidal mononuclear phagocytes (including microglia), Müller cells, photoreceptor cells, and the retinal pigment epithelium. Whilst the VEGF-A decoy function of VEGFR1 is well established, consequences of its direct signaling are less clear. VEGFR1 activation can affect vascular permeability and induce macrophage and microglia production of proinflammatory and proangiogenic mediators. However the ability of the VEGFR1 ligands (VEGF-A, PlGF, and VEGF-B) to compete against each other for receptor binding and to heterodimerize complicates our understanding of the relative contribution of VEGFR1 signaling alone toward the pathologic processes seen in diabetic retinopathy, retinal vascular occlusions, retinopathy of prematurity, and age-related macular degeneration. Clinically, anti-VEGF drugs have proven transformational in these pathologies and their impact on modulation of VEGFR1 signaling is still an opportunity-rich field for further research.
Collapse
Affiliation(s)
- Akiyoshi Uemura
- Department of Retinal Vascular Biology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan.
| | - Marcus Fruttiger
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK.
| | - Patricia A D'Amore
- Schepens Eye Research Institute of Massachusetts Eye and Ear, 20 Staniford Street, Boston, MA, 02114, USA.
| | - Sandro De Falco
- Angiogenesis Laboratory, Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", Via Pietro Castellino 111, 80131 Naples, Italy; ANBITION S.r.l., Via Manzoni 1, 80123, Naples, Italy.
| | - Antonia M Joussen
- Department of Ophthalmology, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12200 Berlin, and Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Florian Sennlaub
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France.
| | - Lynne R Brunck
- Bayer Consumer Care AG, Pharmaceuticals, Peter-Merian-Strasse 84, CH-4052 Basel, Switzerland.
| | - Kristian T Johnson
- Bayer Consumer Care AG, Pharmaceuticals, Peter-Merian-Strasse 84, CH-4052 Basel, Switzerland.
| | - George N Lambrou
- Bayer Consumer Care AG, Pharmaceuticals, Peter-Merian-Strasse 84, CH-4052 Basel, Switzerland.
| | - Kay D Rittenhouse
- Bayer Consumer Care AG, Pharmaceuticals, Peter-Merian-Strasse 84, CH-4052 Basel, Switzerland.
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Joseph-Stelzmann-Str. 9, 50931, Cologne, Germany.
| |
Collapse
|
14
|
Coutinho FP, Green CR, Acosta ML, Rupenthal ID. Xentry-Gap19 inhibits Connexin43 hemichannel opening especially during hypoxic injury. Drug Deliv Transl Res 2021; 10:751-765. [PMID: 32318976 PMCID: PMC7223318 DOI: 10.1007/s13346-020-00763-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hypoxic injury results in cell death, tissue damage and activation of inflammatory pathways. This is mediated by pathological Connexin43 (Cx43) hemichannel (HC) opening resulting in osmotic and ionic imbalances as well as cytokine production perpetuating the inflammatory environment. Gap19 is an intracellularly acting Cx43 mimetic peptide that blocks HC opening and thus promotes cell survival. However, native Gap19, which must enter the cell in order to function, exhibits low cell permeability. In this study, Gap19 was conjugated to the cell-penetrating peptide, Xentry, to investigate if cellular uptake could be improved while maintaining peptide function. Cellular uptake of Xentry-Gap19 (XG19) was much greater than that of native Gap19 even under normal cell culture conditions. Peptide function was maintained post uptake as shown by reduced ethidium homodimer influx and ATP release due to Cx43 HC block. While XG19 blocked pathologic HC opening though, normal gap junction communication required for cell repair and survival mechanisms was not affected as shown in a dye scrape-load assay. Under hypoxic conditions, increased expression of Syndecan-4, a plasma membrane proteoglycan targeted by Xentry, enabled even greater XG19 uptake leading to higher inhibition of ATP release and greater cell survival. This suggests that XG19, which is targeted specifically to hypoxic cells, can efficiently and safely block Cx43 HC and could therefore be a novel treatment for hypoxic and inflammatory diseases. Graphical abstract ![]()
Collapse
Affiliation(s)
- Frazer P Coutinho
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
- Department of Ophthalmology, New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Colin R Green
- Department of Ophthalmology, New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Monica L Acosta
- School of Optometry and Vision Science, New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Ilva D Rupenthal
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand.
- Department of Ophthalmology, New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
15
|
Afshari AR, Mollazadeh H, Sahebkar A. Minocycline in Treating Glioblastoma Multiforme: Far beyond a Conventional Antibiotic. JOURNAL OF ONCOLOGY 2020; 2020:8659802. [PMID: 33014057 PMCID: PMC7519463 DOI: 10.1155/2020/8659802] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/05/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022]
Abstract
One of the most lethal forms of CNS pathologies is glioblastoma multiforme (GBM) that represents high invasiveness, uncontrolled proliferation, and angiogenic features. Its invasiveness is responsible for the high recurrence even after maximal surgical interventions. Minocycline is a semisynthetic analog of tetracyclines with potential anti-inflammatory and anticancer effects, distinct from its antimicrobial activity. In this review, we highlight the importance and the cytotoxic mechanisms of minocycline on GBM pathophysiology. Considering the role of certain enzymes in autophagy, apoptosis, tumor cell invasion, and metastatic ability, the possible use of tetracyclines for cancer therapy should be investigated, especially GBM. The present study is, therefore, going to cover the main topics in minocycline pharmacology to date, encouraging its consideration as a new treatment approach for cancer and GBM.
Collapse
Affiliation(s)
- Amir R. Afshari
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Hamid Mollazadeh
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
16
|
Rahimi N, Azizi M, Bahari G, Narouie B, Hashemi M. Association of EGLN2 rs10680577 Polymorphism with the Risk and Clinicopathological Features of Patients with Prostate Cancer. Asian Pac J Cancer Prev 2020; 21:1221-1226. [PMID: 32458625 PMCID: PMC7541883 DOI: 10.31557/apjcp.2020.21.5.1221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Indexed: 11/25/2022] Open
Abstract
Several studies have evaluated the association between EGLN2 4-bp insertion/deletion (ins/del) polymorphism (rs10680577) and many cancers. However, up to date, no study has inspected the impact of rs10680577 polymorphism on prostate cancer (PCa) risk. This case-control study was achieved on 170 pathologically confirmed PCa patients and 196 cancer free men to inspect whether rs10680577 variant is related to the risk and clinicopathological features of patients with PCa. Genotyping was performed by mismatched polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). The findings did not support an association between the variant with the risk and clinicopathological characteristics of PCa patients. When we pooled our results with six preceding studies, the findings suggested that rs10680577 variant significantly augmented the risk of overall cancer in heterozygous (OR=1.38, 95 % CI=1.26-1.52, p<0.00001, ins/del vs ins/ins), homozygous (OR=1.66, 95 % CI=1.05-2.61, p=0.029, del/del vs ins/ins), codominant (OR=1.44, 95%CI=1.32-1.58, p<0.00001, ins/del+del/del vs ins/ins), and allele (OR=1.32, 95%CI=1.18-1.49, p<0.00001, del vs ins) genetic models. Additional well designed studies with larger sample sizes are necessary to confirm our findings.
Collapse
Affiliation(s)
- Nahid Rahimi
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mahsa Azizi
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Gholamreza Bahari
- Children and Adolescent Health Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Behzad Narouie
- Urology and Nephrology Research Center, Department of Urology, Shahid Labbafinejad Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hashemi
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.,Genetics of Non-communicable Disease Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
17
|
Liu Y, Liang S, Ding R, Hou Y, Deng F, Ma X, Song T, Yan D. BCG-induced trained immunity in macrophage: reprograming of glucose metabolism. Int Rev Immunol 2020; 39:83-96. [DOI: 10.1080/08830185.2020.1712379] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Yuntong Liu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun City, Jilin Province, China
| | - Shu Liang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun City, Jilin Province, China
| | - Ru Ding
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun City, Jilin Province, China
| | - Yuyang Hou
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun City, Jilin Province, China
| | - Feier Deng
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun City, Jilin Province, China
| | - Xiaohui Ma
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun City, Jilin Province, China
| | - Tiantian Song
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun City, Jilin Province, China
| | - Dongmei Yan
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun City, Jilin Province, China
| |
Collapse
|
18
|
Chen J, Luo Y, Wang S, Zhu H, Li D. Roles and mechanisms of SUMOylation on key proteins in myocardial ischemia/reperfusion injury. J Mol Cell Cardiol 2019; 134:154-164. [PMID: 31344368 DOI: 10.1016/j.yjmcc.2019.07.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/19/2019] [Accepted: 07/21/2019] [Indexed: 12/19/2022]
Abstract
Myocardial ischemia/reperfusion (MI/R) injury has a great influence on the prognosis of patients with acute coronary occlusion. The underlying mechanisms of MI/R injury are complex. While the incidence of MI/R injury is increasing every year, the existing therapies are not satisfactory. Recently, small ubiquitin-related modifier (SUMO), which is a post-translational modification and involved in many cell processes, was found to play remarkable roles in MI/R injury. Several proteins that can be SUMOylated were found to interfere with different mechanisms of MI/R injury. Sarcoplasmic reticulum Ca2+ ATPase pump SUMOylation alleviated calcium overload. Among the histone deacetylase (HDAC) members, SUMOylation of HDAC4 reduced reactive oxygen species generation, whereas Sirt1 played protective roles in the SUMOylated form. Dynamic-related protein 1 modified by different SUMO proteins exerted opposite effects on the function of mitochondria. SUMOylation of hypoxia-inducible factors was fundamental in oxygen homeostasis, while eukaryotic elongation factor 2 SUMOylation induced cardiomyocyte apoptosis. The impact of other SUMOylation substrates in MI/R injury remains unclear. Here we reviewed how these SUMOylated proteins alleviated or exacerbated myocardial impairments by effecting the MI/R injury mechanisms. This may suggest methods for relieving MI/R injury in clinical practice and provide a reference for further study of SUMOylation in MI/R injury.
Collapse
Affiliation(s)
- Jingwen Chen
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Yuanyuan Luo
- Xuzhou Medical University Affiliated Hospital, Xuzhou, Jiangsu, PR China
| | - Shuai Wang
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Hong Zhu
- Xuzhou Medical University Affiliated Hospital, Xuzhou, Jiangsu, PR China
| | - Dongye Li
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Xuzhou Medical University Affiliated Hospital, Xuzhou, Jiangsu, PR China.
| |
Collapse
|
19
|
The essential role of tumor suppressor gene ING4 in various human cancers and non-neoplastic disorders. Biosci Rep 2019; 39:BSR20180773. [PMID: 30643005 PMCID: PMC6356015 DOI: 10.1042/bsr20180773] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 12/19/2018] [Accepted: 01/13/2019] [Indexed: 12/21/2022] Open
Abstract
Inhibitor of growth 4 (ING4), a member of the ING family discovered in 2003, has been shown to act as a tumor suppressor and is frequently down-regulated in various human cancers. Numerous published in vivo and in vitro studies have shown that ING4 is responsible for important cancer hallmarks such as pathologic cell cycle arrest, apoptosis, autophagy, contact inhibition, and hypoxic adaptation, and also affects tumor angiogenesis, invasion, and metastasis. These characteristics are typically associated with regulation through chromatin acetylation by binding histone H3 trimethylated at lysine 4 (H3K4me3) and through transcriptional activity of transcription factor P53 and NF-κB. In addition, emerging evidence has indicated that abnormalities in ING4 expression and function play key roles in non-neoplastic disorders. Here, we provide an overview of ING4-modulated chromosome remodeling and transcriptional function, as well as the functional consequences of different genetic variants. We also present the current understanding concerning the role of ING4 in the development of neoplastic and non-neoplastic diseases. These studies offer inspiration for pursuing novel therapeutics for various cancers.
Collapse
|
20
|
Lakatos D, Somfai E, Méhes E, Czirók A. Soluble VEGFR1 signaling guides vascular patterns into dense branching morphologies. J Theor Biol 2018; 456:261-278. [PMID: 30086288 PMCID: PMC6292526 DOI: 10.1016/j.jtbi.2018.08.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 08/01/2018] [Accepted: 08/03/2018] [Indexed: 01/27/2023]
Abstract
Vascular patterning is a key process during development and disease. The diffusive decoy receptor sVEGFR1 (sFlt1) is a known regulator of endothelial cell behavior, yet the mechanism by which it controls vascular structure is little understood. We propose computational models to shed light on how vascular patterning is guided by self-organized gradients of the VEGF/sVEGFR1 factors. We demonstrate that a diffusive inhibitor can generate structures with a dense branching morphology in models where the activator elicits directed growth. Inadequate presence of the inhibitor leads to compact growth, while excessive production of the inhibitor blocks expansion and stabilizes existing structures. Model predictions were compared with time-resolved experimental data obtained from endothelial sprout kinetics in fibrin gels. In the presence of inhibitory antibodies against VEGFR1 vascular sprout density increases while the speed of sprout expansion remains unchanged. Thus, the rate of secretion and stability of extracellular sVEGFR1 can modulate vascular sprout density.
Collapse
Affiliation(s)
- Dóra Lakatos
- Department of Biological Physics, Eötvös Loránd University, Budapest, Hungary.
| | - Ellák Somfai
- Institute for Solid State Physics and Optics, Wigner Research Center for Physics, Hungarian Academy of Sciences, Budapest, Hungary
| | - Előd Méhes
- Department of Biological Physics, Eötvös Loránd University, Budapest, Hungary
| | - András Czirók
- Department of Biological Physics, Eötvös Loránd University, Budapest, Hungary; Department of Anatomy & Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
21
|
Abdulle AE, Diercks GFH, Feelisch M, Mulder DJ, van Goor H. The Role of Oxidative Stress in the Development of Systemic Sclerosis Related Vasculopathy. Front Physiol 2018; 9:1177. [PMID: 30197602 PMCID: PMC6117399 DOI: 10.3389/fphys.2018.01177] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 08/06/2018] [Indexed: 12/11/2022] Open
Abstract
Systemic sclerosis (SSc) is a rare connective tissue disease characterized by autoimmunity, vasculopathy, and progressive fibrosis typically affecting multiple organs including the skin. SSc often is a lethal disorder, because effective disease-modifying treatment still remains unavailable. Vasculopathy with endothelial dysfunction, perivascular infiltration of mononuclear cells, vascular wall remodeling and rarefaction of capillaries is the hallmark of the disease. Most patients present with vasospastic attacks of the digital arteries referred to as 'Raynaud's phenomenon,' which is often an indication of an underlying widespread vasculopathy. Although autoimmune responses and inflammation are both found to play an important role in the pathogenesis of this vasculopathy, no definite initiating factors have been identified. Recently, several studies have underlined the potential role of oxidative stress in the pathogenesis of SSc vasculopathy thereby proposing a new aspect in the pathogenesis of this disease. For instance, circulating levels of reactive oxygen species (ROS) related markers have been found to correlate with SSc vasculopathy, the formation of fibrosis and the production of autoantibodies. Excess ROS formation is well-known to lead to endothelial cell (EC) injury and vascular complications. Collectively, these findings suggest a potential role of ROS in the initiation and progression of SSc vasculopathy. In this review, we present the background of oxidative stress related processes (e.g., EC injury, autoimmunity, inflammation, and vascular wall remodeling) that may contribute to SSc vasculopathy. Finally, we describe the use of oxidative stress related read-outs as clinical biomarkers of disease activity and evaluate potential anti-oxidative strategies in SSc.
Collapse
Affiliation(s)
- Amaal E. Abdulle
- Department of Internal Medicine, Division of Vascular Medicine, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Gilles F. H. Diercks
- Section Pathology, Department of Pathology and Medical Biology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Martin Feelisch
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Douwe J. Mulder
- Department of Internal Medicine, Division of Vascular Medicine, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Harry van Goor
- Section Pathology, Department of Pathology and Medical Biology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
22
|
Lee SY, Kim HJ, Oh SC, Lee DH. Genipin inhibits the invasion and migration of colon cancer cells by the suppression of HIF-1α accumulation and VEGF expression. Food Chem Toxicol 2018; 116:70-76. [DOI: 10.1016/j.fct.2018.04.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 03/30/2018] [Accepted: 04/03/2018] [Indexed: 12/12/2022]
|
23
|
Li R, Tian J, Du J, Zhao L, Yao Y, Yu Z, Chang W, Shi R, Li J. Manipulation of autophagy: a novelly potential therapeutic strategy for retinal neovascularization. BMC Ophthalmol 2018; 18:110. [PMID: 29703256 PMCID: PMC5924499 DOI: 10.1186/s12886-018-0774-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 04/13/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The relationship between the role of VEGF and autophagy in the process of retinal angiogenesis is still unclear. In this study, we explored this issue by using the mouse retinal vascular endothelial cell (RVEC) as a model. METHODS RVECs were divided into the following groups: control, hypoxia (H), 3-methyladenine (3-MA) + H, VEGF + H, 3-MA + VEGF+H, anti-VEGF antibody + H, 3-MA+ anti-VEGF antibody + H. We then examined activation of autophagy by detecting formation of autophagosomes with transmission electron microscopy (TEM) and by counting the number of green fluorescent protein-positive (GFP+) puncta in RVECs. The turnover of microtubule associated protein 1 light chain 3 B (LC3B) and VEGF were examined by western blot. Cell migratory capacity was measured with wound healing assay and transwell assay. The capillary formation assay was performed to investigate the angiogenic capacity. RESULTS Hypoxia led to an increased number of autophagosome and of the GFP+ puncta, an increased ratio of LC3B-II/I and enhanced migratory and capillary-formation capacities of RVECs. Pre-treatment with 3-MA attenuated activation of autophagy and abrogated the enhanced cell migration and capillary formation under hypoxia. Exposure to VEGF significantly increased migratory and capillary formation capacities of RVECs under hypoxia and 3-MA decreased VEGF-induced angiogenesis without its expression. Formation of autophagosome, the number of GFP+ puncta of RVECs and expression of LC3B-II/I were both elevated in cells treated with anti-VEGF antibody and these effects were partially inhibited by 3-MA pretreatment. CONCLUSION Our present data may identify autophagic response as a novel target for enhancing the therapeutic efficacy of angiogenesis inhibitors.
Collapse
Affiliation(s)
- Rong Li
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi, China.
| | - Jin Tian
- Department of Ophthalmology, the Weinan Central Hospital, Weinan, Shaanxi, China
| | - Junhui Du
- Department of Ophthalmology, Xi'an Ninth Hospital Affiliated to Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Lei Zhao
- Department of Molecular Physiology and Biophysics, Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Yang Yao
- Department of Central laboratory, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi, China
| | - Zhaoxiang Yu
- Department of General surgery, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi, China
| | - Weiping Chang
- Department of General surgery, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi, China
| | - Rui Shi
- Department of Ophthalmology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Jing Li
- Department of Ophthalmology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| |
Collapse
|
24
|
Hashemi M, Danesh H, Bizhani F, Sattarifard H, Hashemi SM, Bahari G. Detection of a 4-bp Insertion/deletion Polymorphism within the Promoter of EGLN2 Using Mismatch PCR-RFLP and Its Association with Susceptibility to Breast Cancer. Asian Pac J Cancer Prev 2018; 19:923-926. [PMID: 29693343 PMCID: PMC6031807 DOI: 10.22034/apjcp.2018.19.4.923] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
It has been shown that a 4-bp insertion/deletion (ins/del) polymorphism of EGLN2 influences the risk of several cancers. However, to date, no study has inspected the impact of the 4-bp ins/del polymorphism on breast cancer (BC) risk. A case-control study, including 134 breast cancer patients and 154 healthy women, was here conducted to examine the possible association between EGLN2 4-bp ins/del polymorphism and BC risk in a southeast Iranian population. A mismatched polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) was designed for genotyping of the variant. Our findings did not support any association between the 4-bp ins/del polymorphism and the risk of BC in the codominant, dominant, recessive and allele inheritance models tested. When links between the EGLN2 4-bp ins/del polymorphism and clinicopathological characteristics of the patients were evaluate the variant was only associated with HER2 status. More studies with larger sample sizes and diverse ethnicities are warranted to verify our finding.
Collapse
Affiliation(s)
- Mohammad Hashemi
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.
| | | | | | | | | | | |
Collapse
|
25
|
Tudisco L, Orlandi A, Tarallo V, De Falco S. Hypoxia activates placental growth factor expression in lymphatic endothelial cells. Oncotarget 2018; 8:32873-32883. [PMID: 28427198 PMCID: PMC5464835 DOI: 10.18632/oncotarget.15861] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 02/08/2017] [Indexed: 01/13/2023] Open
Abstract
Placental growth factor (PlGF), a proangiogenic member of vascular endothelial growth family, is active during pathological conditions like cancer, metastasis formation and hind limb ischemia and in wound healing. Endothelial cells express PlGF and hypoxia positively modulates in vitro its expression. To verify whether hypoxia modulates PlGF expression in different cellular contexts and in vivo, we first analyzed five human and five mouse cancer cell lines showing that in eight of them hypoxia positively modulates PlGF. Next, we analyzed xenograft colorectal cancer tumors showing that human cancer cells were able to express PlGF in hypoxic area of the tumor. Surprisingly, we did not visualize mouse PlGF in CD31 positive tumor vessels, but in low CD31 positive vessels, a characteristic of lymphatic vessels. We found that hypoxia effectively activates PlGF expression in lymphatic endothelial cells as well as in LYVE1 positive tumor vessels. We also investigated two additional mouse angiogenic models, hind limb ischemia and wound healing, and we confirmed that lymphatic vessels of both ischemic muscles and skin express PlGF. These results show for the first time that hypoxia activates PlGF expression in lymphatic endothelial cells, which have to be considered an additional source for PlGF production in pathological contexts.
Collapse
Affiliation(s)
- Laura Tudisco
- Angiogenesis LAB, Institute of Genetics and Biophysics 'Adriano Buzzati-Traverso', CNR, Naples, Italy
| | - Augusto Orlandi
- Department of Biomedicine and Prevention, Anatomic Pathology, University of Tor Vergata, Rome, Italy
| | - Valeria Tarallo
- Angiogenesis LAB, Institute of Genetics and Biophysics 'Adriano Buzzati-Traverso', CNR, Naples, Italy
| | - Sandro De Falco
- Angiogenesis LAB, Institute of Genetics and Biophysics 'Adriano Buzzati-Traverso', CNR, Naples, Italy
| |
Collapse
|
26
|
Pharmacodynamic and pharmacokinetic assessment of pulmonary rehabilitation mixture for the treatment of pulmonary fibrosis. Sci Rep 2017; 7:3458. [PMID: 28615638 PMCID: PMC5471221 DOI: 10.1038/s41598-017-02774-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 02/21/2017] [Indexed: 12/31/2022] Open
Abstract
Pulmonary rehabilitation mixture (PRM), a Chinese herbal medicine formula, has been used to treat pulmonary fibrosis for decades. In this study, we systematically evaluated the pharmacodynamic and pharmacokinetic performance of PRM. The pharmacodynamic results showed that PRM could improve the condition of CoCl2-stimulated human type II alveolar epithelial cells, human pulmonary microvascular endothelial cells, human lung fibroblasts and pulmonary fibrosis rats induced by bleomycin, PRM treatment reduced the expression of platelet-derived growth factor, fibroblast growth factor, toll-like receptor 4, high-mobility group box protein 1 and hypoxia-inducible factor 1α. In the pharmacokinetic study, an accurate and sensitive ultra-high performance liquid chromatography tandem mass spectrometry method was developed and validated for the simultaneous determination of calycosin, calycosin-7-O-glucoside, formononetin, ononin and mangiferin of PRM in the rat plasma for the first time. The method was then successfully applied to the comparative pharmacokinetic study of PRM in normal and pulmonary fibrosis rats. The five constituents could be absorbed in the blood after the oral administration of PRM and exhibited different pharmacokinetic behaviors in normal and pulmonary fibrosis rats. In summary, PRM exhibited a satisfactory pharmacodynamic and pharmacokinetic performance, which highlights PRM as a potential multi-target oral drug for the treatment of pulmonary fibrosis.
Collapse
|
27
|
|
28
|
Hypoxia inducible factor stabilization improves defective ischemia-induced angiogenesis in a rodent model of chronic kidney disease. Kidney Int 2016; 91:616-627. [PMID: 27927598 DOI: 10.1016/j.kint.2016.09.028] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 08/24/2016] [Accepted: 09/15/2016] [Indexed: 12/17/2022]
Abstract
Chronic kidney disease (CKD) is associated with increased risk and worse prognosis of cardiovascular disease, including peripheral artery disease. An impaired angiogenic response to ischemia may contribute to poor outcomes of peripheral artery disease in patients with CKD. Hypoxia inducible factors (HIF) are master regulators of angiogenesis and therefore represent a promising target for therapeutic intervention. To test this we induced hind-limb ischemia in rats with CKD caused by 5/6 nephrectomy and administered two different treatments known to stabilize HIF protein in vivo: carbon monoxide and a pharmacological inhibitor of prolyl hydroxylation 2-(1-chloro-4- hydroxyisoquinoline-3-carboxamido) acetate (ICA). Expression levels of pro-angiogenic HIF target genes (Vegf, Vegf-r1, Vegf-r2, Ho-1) were measured by qRT-PCR. Capillary density was measured by CD31 immunofluorescence staining and HIF expression was evaluated by immunohistochemistry. Capillary density in ischemic skeletal muscle was significantly lower in CKD animals compared to sham controls. Rats with CKD showed significantly lower expression of HIF and all measured pro-angiogenic HIF target genes, including VEGF. Both HIF stabilizing treatments rescued HIF target gene expression in animals with CKD and led to significantly higher ischemia-induced capillary sprouting compared to untreated controls. ICA was effective regardless of whether it was administered before or after induction of ischemia and led to a HIF expression in skeletal muscle. Thus, impaired ischemia-induced angiogenesis in rats with CKD can be improved by HIF stabilization, even if started after onset of ischemia.
Collapse
|
29
|
Burr SP, Costa ASH, Grice GL, Timms RT, Lobb IT, Freisinger P, Dodd RB, Dougan G, Lehner PJ, Frezza C, Nathan JA. Mitochondrial Protein Lipoylation and the 2-Oxoglutarate Dehydrogenase Complex Controls HIF1α Stability in Aerobic Conditions. Cell Metab 2016; 24:740-752. [PMID: 27923773 PMCID: PMC5106373 DOI: 10.1016/j.cmet.2016.09.015] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 08/11/2016] [Accepted: 09/24/2016] [Indexed: 11/01/2022]
Abstract
Hypoxia-inducible transcription factors (HIFs) control adaptation to low oxygen environments by activating genes involved in metabolism, angiogenesis, and redox homeostasis. The finding that HIFs are also regulated by small molecule metabolites highlights the need to understand the complexity of their cellular regulation. Here we use a forward genetic screen in near-haploid human cells to identify genes that stabilize HIFs under aerobic conditions. We identify two mitochondrial genes, oxoglutarate dehydrogenase (OGDH) and lipoic acid synthase (LIAS), which when mutated stabilize HIF1α in a non-hydroxylated form. Disruption of OGDH complex activity in OGDH or LIAS mutants promotes L-2-hydroxyglutarate formation, which inhibits the activity of the HIFα prolyl hydroxylases (PHDs) and TET 2-oxoglutarate dependent dioxygenases. We also find that PHD activity is decreased in patients with homozygous germline mutations in lipoic acid synthesis, leading to HIF1 activation. Thus, mutations affecting OGDHC activity may have broad implications for epigenetic regulation and tumorigenesis.
Collapse
Affiliation(s)
- Stephen P Burr
- Department of Medicine, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK
| | - Ana S H Costa
- Hutchinson MRC Cancer Unit, University of Cambridge, Cambridge, CB2 0XZ, UK
| | - Guinevere L Grice
- Department of Medicine, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK
| | - Richard T Timms
- Department of Medicine, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK
| | - Ian T Lobb
- Department of Medicine, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK
| | | | - Roger B Dodd
- Department of Medicine, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK
| | - Gordon Dougan
- Department of Medicine, University of Cambridge, Cambridge, CB2 0XY, UK; Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, CB10 1SA, UK
| | - Paul J Lehner
- Department of Medicine, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK
| | - Christian Frezza
- Hutchinson MRC Cancer Unit, University of Cambridge, Cambridge, CB2 0XZ, UK
| | - James A Nathan
- Department of Medicine, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK.
| |
Collapse
|
30
|
Higashi K, Yamagishi T, Ueda Y, Ishigaki Y, Shimasaki M, Nakamura Y, Oguchi M, Takegami T, Sagawa M, Tonami H. Correlation of HIF-1α/HIF-2α expression with FDG uptake in lung adenocarcinoma. Ann Nucl Med 2016; 30:708-715. [PMID: 27663442 DOI: 10.1007/s12149-016-1116-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/14/2016] [Indexed: 01/01/2023]
Abstract
OBJECTIVES Hypoxia is a key element involved in the development and progression of tumors. HIF-1α may transiently induce and mediate the response to acute and severe hypoxia, while HIF-2α may induce a longer response and may control the response to chronic and moderate hypoxia. Hypoxia increases the cellular uptake of FDG. Therefore, HIF may play an important role in the process of the cellular uptake of FDG. The aim of this study was to compare HIF-1α/HIF-2α expression with FDG uptake, Glut-1 expression, and prognosis in the patients with lung adenocarcinoma and to investigate the role of HIF-1α/HIF-2α in the uptake of FDG in lung adenocarcinoma. METHODS In the current work, we compared the immunohistochemical expression of HIF-1α and HIF-2α in surgical specimens of 44 patients with lung adenocarcinoma. The relationships between HIF-α expression and Glut-1 expression, FDG uptake, and clinicopathological factors, including prognosis, were analyzed. RESULTS There was a marginal association between HIF-1α and HIF-2α expressions (P = 0.076). We found a significant correlation between HIF-2α expression and FDG uptake (P = 0.0001). HIF-1α expression showed a marginal association with FDG uptake (P = 0.066). FDG uptake correlated more significantly with HIF-2α expression than with HIF-1α expression. A significant correlation was noticed between Glut-1 expression and both HIF-1α and HIF-2α expressions (P = 0.005 and P = 0.003, respectively). Univariate analysis of disease-free survival demonstrated that FDG uptake and HIF-2α expression, but not HIF-1α expression, were related to recurrence (P < 0.0001). CONCLUSION FDG uptake correlated more significantly with HIF-2α expression than with HIF-1α expression, and both FDG uptake and HIF-2α expression, but not HIF-1α expression was correlated with post-operative recurrence in the patients with lung adenocarcinoma. These results suggest that both FDG uptake and HIF-2α expression may represent a more aggressive phenotype and that HIF-2α may play a more important role than HIF-1α in the uptake of FDG in lung adenocarcinoma.
Collapse
Affiliation(s)
- Kotaro Higashi
- Department of Radiology, Asanogawa General Hospital, 83 Kosakamachi-Naka, Kanazawa, Ishikawa, 920-8621, Japan.
| | | | - Yoshimichi Ueda
- Department of Pathology, Kanazawa Medical University, Ishikawa, Japan
| | - Yasuhito Ishigaki
- Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan
| | - Miyako Shimasaki
- Department of Pathology, Kanazawa Medical University, Ishikawa, Japan
| | - Yuka Nakamura
- Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan
| | - Manabu Oguchi
- Department of Radiation Therapy, Public Central Hospital of Matto Ishikawa, Ishikawa, Japan
| | - Tsutomu Takegami
- Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan
| | - Motoyasu Sagawa
- Department of Thoracic Surgery, Kanazawa Medical University, Ishikawa, Japan
| | - Hisao Tonami
- Department of Radiology, Kanazawa Medical University, Ishikawa, Japan
| |
Collapse
|
31
|
Park KJ, Yu MO, Park DH, Park JY, Chung YG, Kang SH. Role of vincristine in the inhibition of angiogenesis in glioblastoma. Neurol Res 2016; 38:871-9. [PMID: 27472259 DOI: 10.1080/01616412.2016.1211231] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Vincristine, a microtubule-destabilizing drug, was found to exhibit anti-angiogenic effects and anti-tumoral activity. However, the precise mechanism by which vincristine inhibits angiogenesis in glioblastomas is not well understood. Our aim was to investigate whether vincristine affects vascular endothelial growth factor (VEGF) expression in glioblastoma cells and determine whether it is mediated by the downregulation of hypoxia-inducible factor-1α (HIF-1α). METHODS We investigated the expression of HIF-1α in glioblastoma tissues resected from patients and in human glioblastoma cell lines using immunohistochemistry, Western blot analysis, and immunocytochemistry. In addition to an MTT assay assessing the effect of vincristine on cell proliferation and viability, the effects of vincristine on VEGF mRNA expression and HIF-1α protein were examined using real-time RT-PCR and Western blot analysis under 1% O2 (hypoxia). RESULTS HIF-1α was expressed in the majority of glioblastoma tissues and was detected mainly in the nucleus. Strong immunoreactivity for HIF- 1 α was found often in the hypercellular zones. Under hypoxic conditions, HIF-1α protein levels in the glioblastoma cell lines increased, primarily localizing into the nucleus similar to glioblastoma tissues. Exposure of glioblastoma cells to vincristine resulted in enrichment of the G2-M fraction of the cell cycle, which suggests that vincristine-mediated growth inhibition of glioblastoma is correlated with mitotic inhibition. Using doses lower than those found to reduce the viability and proliferation of cells by 50% (IC50), vincristine decreased both the expression of VEGF mRNA and the level of HIF-1α protein in hypoxic glioblastoma cells. In addition, following exposure to vincristine, the expression of VEGF mRNA was correlated with HIF-1α protein levels. CONCLUSIONS Our results suggest that the mechanism by which vincristine elicits an anti-angiogenic effect in glioblastomas under hypoxic conditions might be mediated, in part, by HIF-1α inhibition.
Collapse
Affiliation(s)
- Kyung-Jae Park
- a Department of Neurosurgery, College of Medicine , Korea University Medical Center, Korea University , Seoul , Korea
| | - Mi Ok Yu
- a Department of Neurosurgery, College of Medicine , Korea University Medical Center, Korea University , Seoul , Korea
| | - Dong-Hyuk Park
- a Department of Neurosurgery, College of Medicine , Korea University Medical Center, Korea University , Seoul , Korea
| | - Jung-Yul Park
- a Department of Neurosurgery, College of Medicine , Korea University Medical Center, Korea University , Seoul , Korea
| | - Yong-Gu Chung
- a Department of Neurosurgery, College of Medicine , Korea University Medical Center, Korea University , Seoul , Korea
| | - Shin-Hyuk Kang
- a Department of Neurosurgery, College of Medicine , Korea University Medical Center, Korea University , Seoul , Korea
| |
Collapse
|
32
|
Wang L, Zheng JN, Pei DS. The emerging roles of Jab1/CSN5 in cancer. Med Oncol 2016; 33:90. [DOI: 10.1007/s12032-016-0805-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 07/04/2016] [Indexed: 12/13/2022]
|
33
|
Oh ET, Kim CW, Kim SJ, Lee JS, Hong SS, Park HJ. Docetaxel induced-JNK2/PHD1 signaling pathway increases degradation of HIF-1α and causes cancer cell death under hypoxia. Sci Rep 2016; 6:27382. [PMID: 27263528 PMCID: PMC4893693 DOI: 10.1038/srep27382] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 05/18/2016] [Indexed: 12/29/2022] Open
Abstract
HIF-1 (hypoxia-inducible factor-1) regulates the expression of more than 70 genes involved in angiogenesis, tumor growth, metastasis, chemoresistance, and radioresistance. Thus, there is growing interest in using HIF-1 inhibitors as anticancer drugs. Docetaxel, a Food and Drug Administration-approved anticancer drug, is reported to enhance HIF-1α degradation. Here, we investigated the molecular mechanism underlying docetaxel-induced HIF-1α degradation and cancer cell death under hypoxic conditions. Docetaxel pretreatment enhanced the polyubiquitination and proteasome-mediated degradation of HIF-1α, and increased cancer cell death under hypoxic conditions. Docetaxel also activated the prolyl hydroxylase, PHD1, in hypoxia, and pharmacological inhibition or siRNA-mediated knockdown of PHD1 prevented docetaxel-induced HIF-1α degradation and cancer cell death. Additionally, siRNA-mediated JNK2 knockdown blocked docetaxel-induced HIF-1α degradation and cancer cell death by inhibiting PHD1 activation. A luciferase reporter assay revealed that inhibition of the JNK2/PHD1 signaling pathway significantly increased the transcriptional activity of HIF-1 in docetaxel-treated cancer cells under hypoxia. Consistent with these results, docetaxel-treated JNK2-knockdown tumors grew much faster than control tumors through inhibition of docetaxel-induced PHD1 activation and degradation of HIF-1α. Our results collectively show that, under hypoxic conditions, docetaxel induces apoptotic cell death through JNK2/PHD1 signaling-mediated HIF-1α degradation.
Collapse
Affiliation(s)
- Eun-Taex Oh
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon 22212, Republic of Korea.,Hypoxia-related Disease Research Center, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Chan Woo Kim
- Department of Microbiology, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Soo Jung Kim
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Jae-Seon Lee
- Hypoxia-related Disease Research Center, College of Medicine, Inha University, Incheon 22212, Republic of Korea.,Department of Molecular Medicine, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Soon-Sun Hong
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon 22212, Republic of Korea.,Hypoxia-related Disease Research Center, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Heon Joo Park
- Hypoxia-related Disease Research Center, College of Medicine, Inha University, Incheon 22212, Republic of Korea.,Department of Microbiology, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| |
Collapse
|
34
|
Li R, Du J, Chang Y. Role of Autophagy in Hypoxia-Induced Angiogenesis of RF/6A Cells in vitro. Curr Eye Res 2016; 41:1566-1570. [PMID: 27259688 DOI: 10.3109/02713683.2016.1145234] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE Autophagy plays a role in the pathogenesis of tumor angiogenesis and cardiovascular diseases. The autophagy level in mammalian cells was found to increase in the state of hypoxia or ischemia reperfusion. However, the role of autophagy in ocular angiogenesis has not yet been elucidated. The aim of this study was to investigate the effects of autophagy on hypoxia-induced choroidal and retinal angiogenesis in vitro using a rhesus macaque choroid-retinal endothelial (RF/6A) cell line. MATERIALS AND METHODS RF/6A cells were cultured and randomly divided into three groups according to the different culture media: control, hypoxia model[adding 125μMcobalt chloride (CoCl2) to the culture medium], and hypoxia with an autophagy inhibition group [pretreatment with 5 mmol/L 3-methyladenine (3-MA) for 1.5 h and then adding125μmol/LCoCl2 to the culture medium]. The impact of 3-MA's effect on the level of autophagy proteins (Beclin-1 and LC3) was tested by Western blot analysis. The cell proliferation was assessed using the chromogenicmethylthiazol tetrazolium bromide (MTT) dye after 24 and 48 hours. Cell migration was investigated by wound assay. The tube formation was measured on Matrigel. RESULTS Under chemical hypoxia conditions, Beclin-1 and LC3 levels increased and this change can be inhibited by 3-MA. Cell viability was decreased in cells treated with CoCl2 for 24 and 48 h compared with the control, and pretreatment with 3-MA slightly promoted CoCl2-inhibited cell proliferation. Cell migration and tube formation were increased in cells treated with CoCl2 for 24 and 48 h compared with the control. Pretreatment with 3-MA significantly inhibited CoCl2-induced cell migration and tube formation. CONCLUSIONS Hypoxia-induced autophagy decreased the cell viability and increased the cell migration and tube formation of RF/6A cells. 3-MA can inhibit hypoxia-induced angiogenesis of RF/6A cells in vitro. The present study suggests that autophagy plays a role in retinal and choroidal angiogenesis and the autophagy inhibitor can be a potential candidate for the treatment of choroidal or retinal neovascularization.
Collapse
Affiliation(s)
- Rong Li
- a Department of Ophthalmology , The First Affiliated Hospital, Xi'an Medical University , Xi'an , Shaanxi , China
| | - Junhui Du
- b Department of Ophthalmology , Xi'an Ninth Hospital Affiliated to Medical College of Xi'an Jiaotong University , Xi'an , Shaanxi , China
| | - Yuan Chang
- c Department of Ophthalmology and Otorhinolaryngology , Xi'an Medical University , Xi'an , Shaanxi , China
| |
Collapse
|
35
|
The Clinical Impact of Transcatheter Arterial Chemoembolization (TACE)-Induced c-Met Upregulation on TACE Refractoriness in Hepatocellular Carcinoma. Dig Dis Sci 2016; 61:1572-81. [PMID: 26725068 DOI: 10.1007/s10620-015-4018-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 12/20/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Transcatheter arterial chemoembolization (TACE) is a widely used and well-established treatment for hepatocellular carcinoma (HCC). However, TACE loses its therapeutic efficacy when performed repeatedly, a phenomenon termed TACE refractoriness. c-Met is associated with malignant potential and with resistance to anti-tumor therapies in some kinds of cancers. AIMS The aim of this study is to investigate the clinical impact of TACE on c-Met expression with the aim of understanding the mechanism underlying TACE refractoriness. METHODS The effect of TACE on the c-Met expression level was investigated in vitro in HCC cell lines, and it was shown that c-Met expression is upregulated in HCC cell lines cultured under hypoxia and/or exposed to chemotherapeutic agents. The in vitro results were validated using 82 clinical samples of HCC with and without preoperative TACE treatment. RESULTS c-Met upregulation was observed significantly more frequently in clinical samples of HCC that were treated with preoperative TACE than in samples with no TACE treatment. Increased c-Met expression was significantly associated with poor prognosis. Furthermore, the incidence of c-Met-positive expression was significantly higher in TACE-refractory HCC samples. CONCLUSIONS TACE treatment upregulates c-Met expression in HCC and the upregulated c-Met expression may be responsible for TACE refractoriness.
Collapse
|
36
|
Proteome Profile and Quantitative Proteomic Analysis of Buffalo (Bubalusbubalis) Follicular Fluid during Follicle Development. Int J Mol Sci 2016; 17:ijms17050618. [PMID: 27136540 PMCID: PMC4881444 DOI: 10.3390/ijms17050618] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 04/11/2016] [Accepted: 04/15/2016] [Indexed: 11/17/2022] Open
Abstract
Follicular fluid (FF) accumulates in the antrum of the ovarian follicle and provides the microenvironment for oocyte development. FF plays an important role in follicle growth and oocyte maturation. The FF provides a unique window to investigate the processes occurring during buffalo follicular development. The observed low quality of buffalo oocytes may arise from the poor follicular microenvironment. Investigating proteins found in buffalo FF (BFF) should provide insight into follicular development processes and provide further understanding of intra-follicular maturation and oocytes quality. Here, a proteomic-based approach was used to analyze the proteome of BFF. SDS-PAGE separation combined with mass spectrometry was used to generate the proteomic dataset. In total, 363 proteins were identified and classified by Gene Ontology terms. The proteins were assigned to 153 pathways, including signaling pathways. To evaluate difference in proteins expressed between BFF with different follicle size (small, <4 mm; and large, >8 mm), a quantitative proteomic analysis based on multi-dimensional liquid chromatography pre-fractionation tandem Orbitrap mass spectrometry identification was performed. Eleven differentially expressed proteins (six downregulated and five upregulated in large BFF) were identified and assigned to a variety of functional processes, including serine protease inhibition, oxidation protection and the complement cascade system. Three differentially expressed proteins, Vimentin, Peroxiredoxin-1 and SERPIND1, were verified by Western blotting, consistent with the quantitative proteomics results. Our datasets offers new information about proteins present in BFF and should facilitate the development of new biomarkers. These differentially expressed proteins illuminate the size-dependent protein changes in follicle microenvironment.
Collapse
|
37
|
Hu K, Babapoor-Farrokhran S, Rodrigues M, Deshpande M, Puchner B, Kashiwabuchi F, Hassan SJ, Asnaghi L, Handa JT, Merbs S, Eberhart CG, Semenza GL, Montaner S, Sodhi A. Hypoxia-inducible factor 1 upregulation of both VEGF and ANGPTL4 is required to promote the angiogenic phenotype in uveal melanoma. Oncotarget 2016; 7:7816-28. [PMID: 26761211 PMCID: PMC4884956 DOI: 10.18632/oncotarget.6868] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 01/04/2016] [Indexed: 12/25/2022] Open
Abstract
PURPOSE Expression of the hypoxia-inducible factor (HIF)-1-regulated gene product, vascular endothelial growth factor (VEGF), correlates with tumor vascularity in patients with uveal melanoma (UM). While the relationship between HIF-1 and VEGF in cancer is well-studied, their relative contribution to the angiogenic phenotype in UM has not previously been interrogated. Here we evaluate the contribution of HIF-1, VEGF, and a second HIF-1-regulated gene product, angiopoietin-like 4 (ANGPTL4), to angiogenesis in UM. EXPERIMENTAL DESIGN UM cells were examined for expression of HIF-1α, VEGF, and ANGPTL4. Their contribution to the angiogenic potential of UM cells was assessed using the endothelial cell tubule formation and directed in vivo angiogenesis assays. These results were corroborated in tissue from UM animal models and in tissue from patients with UM. RESULTS Inhibition of VEGF partially reduced tubule formation promoted by conditioned medium from UM cells. Inhibition of ANGPTL4, which was highly expressed in hypoxic UM cells, a UM orthotopic transplant model, a UM tumor array, and vitreous samples from UM patients, inhibited the angiogenic potential of UM cells in vitro and in vivo; this effect was additive to VEGF inhibition. CONCLUSIONS Targeting both ANGPTL4 and VEGF may be required for the effective inhibition of angiogenesis in UM.
Collapse
Affiliation(s)
- Ke Hu
- 1 Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA,2 The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | - Murilo Rodrigues
- 1 Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Monika Deshpande
- 1 Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Brooks Puchner
- 1 Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | - Syed Junaid Hassan
- 1 Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Laura Asnaghi
- 3 Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - James T. Handa
- 1 Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Shannath Merbs
- 1 Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Charles G. Eberhart
- 1 Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA,3 Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Gregg L. Semenza
- 4 Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, Biological Chemistry, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Silvia Montaner
- 5 Department of Oncology and Diagnostic Sciences, Greenebaum Cancer Center, University of Maryland, Baltimore, MD, USA
| | - Akrit Sodhi
- 1 Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
38
|
He L, Xu J, Chen L, Li L. Apelin/APJ signaling in hypoxia-related diseases. Clin Chim Acta 2015; 451:191-8. [PMID: 26436483 DOI: 10.1016/j.cca.2015.09.029] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 09/26/2015] [Accepted: 09/29/2015] [Indexed: 12/29/2022]
Abstract
The regulatory peptide apelin is the endogenous ligand for the orphan G protein-coupled receptor APJ. Apelin and APJ exist in a variety of tissues, with special status in the heart, lung and tumors. Consequently, the apelin/APJ system exerts a broad range of activities that affect multiple organ systems. Accumulating evidence indicates that the expressions of apelin and APJ are significantly augmented by hypoxia through the hypoxia-inducible factor-1 alpha (HIF-1α) signaling pathway. Increased apelin promotes cellular proliferation, migration and survival, therefore regulating angiogenesis. In addition, the pre-administration of exogenous apelin is involved in the occurrence and development of hypoxia-induced pathological diseases. The purpose of this article is to review the properties of the apelin/APJ system, which is affected by hypoxic conditions, and the regulation of apelin/APJ signaling in hypoxia-associated disorders. Thus, the apelin/APJ system may be a potential therapeutic target in hypoxia-related diseases.
Collapse
Affiliation(s)
- Lu He
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, University of South China, Hengyang 421001, PR China
| | - Jin Xu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, University of South China, Hengyang 421001, PR China
| | - Linxi Chen
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, University of South China, Hengyang 421001, PR China.
| | - Lanfang Li
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, University of South China, Hengyang 421001, PR China.
| |
Collapse
|
39
|
Abstract
The treatment of center-involving diabetic macular edema (DME) has improved because of the proven efficacy of drugs that inhibit the effects of vascular endothelial growth factor (VEGF). The newest anti-VEGF drug, aflibercept, has recently been approved by the United States Food and Drug Administration for the treatment of center-involving DME and for diabetic retinopathy in eyes with DME. In the pivotal Phase III VISTA and VIVID trials, intravitreal aflibercept 2 mg injections every 4 or 8 weeks (after 5 monthly loading doses) produced superior gains in BCVA compared to laser/sham injections. In the Diabetic Retinopathy Clinical Research Network Protocol T trial, which featured monthly anti-VEGF monotherapy for 6 months, followed by monthly pro re nata anti-VEGF injections with laser rescue therapy from months 6 through 12, aflibercept 2 mg monthly was superior to bevacizumab 1.25 mg and ranibizumab 0.5 mg in eyes with BCVA of 20/50 or worse (aflibercept versus bevacizumab: P<0.001; aflibercept versus ranibizumab: P=0.003), but the three regimens were comparable for eyes with VA of 20/40 or better. Only in the 20/50 or worse subgroup did aflibercept achieve clinical superiority (>5 letter difference) to bevacizumab. Each treatment regimen led to significant macular thinning, with aflibercept being superior to bevacizumab in both visual acuity subgroups (P<0.001 for each), but it was not statistically superior to ranibizumab in either group. In diabetic patients, aflibercept has an excellent safety profile that does not appear to differ from laser/sham or other VEGF inhibitory drugs.
Collapse
Affiliation(s)
- Michael W Stewart
- Department of Ophthalmology, Mayo School of Medicine, Jacksonville, FL, USA
| |
Collapse
|
40
|
Nanoparticle Delivered VEGF-A siRNA Enhances Photodynamic Therapy for Head and Neck Cancer Treatment. Mol Ther 2015; 24:106-16. [PMID: 26373346 DOI: 10.1038/mt.2015.169] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 08/28/2015] [Indexed: 12/25/2022] Open
Abstract
Photodynamic therapy (PDT) is believed to promote hypoxic conditions to tumor cells leading to overexpression of angiogenic markers such as vascular endothelial growth factor (VEGF). In this study, PDT was combined with lipid-calcium-phosphate nanoparticles (LCP NPs) to deliver VEGF-A small interfering RNA (siVEGF-A) to human head and neck squamous cell carcinoma (HNSCC) xenograft models. VEGF-A were significantly decreased for groups treated with siVEGF-A in human oral squamous cancer cell (HOSCC), SCC4 and SAS models. Cleaved caspase-3 and in situ TdT-mediated dUTP nick-end labeling assay showed more apoptotic cells and reduced Ki-67 expression for treated groups compared to phosphate buffered saline (PBS) group. Indeed, the combined therapy showed significant tumor volume decrease to ~70 and ~120% in SCC4 and SAS models as compared with untreated PBS group, respectively. In vivo toxicity study suggests no toxicity of such LCP NP delivered siVEGF-A. In summary, results suggest that PDT combined with targeted VEGF-A gene therapy could be a potential therapeutic modality to achieve enhanced therapeutic outcome for HNSCC.
Collapse
|
41
|
Lee SH, Jeong D, Han YS, Baek MJ. Pivotal role of vascular endothelial growth factor pathway in tumor angiogenesis. Ann Surg Treat Res 2015; 89:1-8. [PMID: 26131438 PMCID: PMC4481026 DOI: 10.4174/astr.2015.89.1.1] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 02/06/2015] [Accepted: 02/27/2015] [Indexed: 12/18/2022] Open
Abstract
The shaping of new blood vessels is a significant event in cancer growth and metastasis. Therefore, the molecular system of cancer angiogenesis has garnered considerable interest in cancer research. The vascular endothelial growth factor (VEGF) and VEGF receptor pathway are recognized as the key regulators of the angiogenic process. Activation of the VEGF/VEGF-receptor pathway initiates signaling cascades that promote endothelial cell growth, migration, and differentiation. Recently, VEGF was shown to play a role in the recruitment of bone marrow-derived endothelial progenitor cells to neovascularization sites. The role of VEGF in promoting tumor angiogenesis and the occurrence of human cancers has led to the rational design and development of agents that selectively target this pathway. Moreover, these anti-VEGF/VEGF receptor agents show therapeutic potential by inhibition of angiogenesis and tumor growth in preclinical models. In this review, we summarize the role of the VEGF pathway during tumor angiogenesis.
Collapse
Affiliation(s)
- Sang Hun Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea. ; Department of Biochemistry, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Dongjun Jeong
- Department of Pathology, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Yong-Seok Han
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Moo Jun Baek
- Department of Surgery, Soonchunhyang University College of Medicine, Cheonan, Korea
| |
Collapse
|
42
|
Ryou MG, Choudhury GR, Li W, Winters A, Yuan F, Liu R, Yang SH. Methylene blue-induced neuronal protective mechanism against hypoxia-reoxygenation stress. Neuroscience 2015; 301:193-203. [PMID: 26047733 DOI: 10.1016/j.neuroscience.2015.05.064] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 05/13/2015] [Accepted: 05/27/2015] [Indexed: 12/17/2022]
Abstract
UNLABELLED Brain ischemia and reperfusion (I/R) injury occurs in various pathological conditions, but there is no effective treatment currently available in clinical practice. Methylene blue (MB) is a century-old drug with a newly discovered protective function in the ischemic stroke model. In the current investigation we studied the MB-induced neuroprotective mechanism focusing on stabilization and activation of hypoxia-inducible factor-1α (HIF-1α) in an in vitro oxygen and glucose deprivation (OGD)-reoxygenation model. METHODS HT22 cells were exposed to OGD (0.1% O2, 6h) and reoxygenation (21% O2, 24h). Cell viability was determined with the calcein AM assay. The dynamic change of intracellular O2 concentration was monitored by fluorescence lifetime imaging microscopy (FLTIM). Glucose uptake was quantified using the 2-[N-(7-Nitrobenz-2-Oxa-1,3-Diazol-4-yl)Amino]-2-Deoxy-d-Glucose (2-NBDG) assay. ATP concentration and glycolytic enzyme activity were examined by spectrophotometry. Protein content changes were measured by immunoblot: HIF-1α, prolyl hydroxylase 2 (PHD2), erythropoietin (EPO), Akt, mTOR, and PIP5K. The contribution of HIF-1α activation in the MB-induced neuroprotective mechanism was confirmed by blocking HIF-1α activation with 2-methoxyestradiol-2 (2-MeOE2) and by transiently transfecting constitutively active HIF-1α. RESULTS MB increases cell viability by about 50% vs. OGD control. Compared to the corresponding control, MB increases intracellular O2 concentration and glucose uptake as well as the activities of hexokinase and G-6-PDH, and ATP concentration. MB activates the EPO signaling pathway with a corresponding increase in HIF-1α. Phosphorylation of Akt was significantly increased with MB treatment followed by activation of the mTOR pathway. Importantly, we observed, MB increased nuclear translocation of HIF-1α vs. control (about three folds), which was shown by a ratio of nuclear:cytoplasmic HIF-1α protein content. CONCLUSION We conclude that MB protects the hippocampus-derived neuronal cells against OGD-reoxygenation injury by enhancing energy metabolism and increasing HIF-1α protein content accompanied by an activation of the EPO signaling pathway.
Collapse
Affiliation(s)
- M-G Ryou
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA; Cardiovascular Research Institute, University of North Texas Health Science Center, Fort Worth, TX, USA.
| | - G R Choudhury
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - W Li
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - A Winters
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - F Yuan
- Department of Neurosurgery, Beijing Tiantan Hospital, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100050, China
| | - R Liu
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - S-H Yang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA; Cardiovascular Research Institute, University of North Texas Health Science Center, Fort Worth, TX, USA; Department of Neurosurgery, Beijing Tiantan Hospital, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100050, China.
| |
Collapse
|
43
|
Li G, Shan C, Liu L, Zhou T, Zhou J, Hu X, Chen Y, Cui H, Gao N. Tanshinone IIA inhibits HIF-1α and VEGF expression in breast cancer cells via mTOR/p70S6K/RPS6/4E-BP1 signaling pathway. PLoS One 2015; 10:e0117440. [PMID: 25659153 PMCID: PMC4320086 DOI: 10.1371/journal.pone.0117440] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 12/23/2014] [Indexed: 01/08/2023] Open
Abstract
Hypoxia-inducible factor 1α (HIF-1α) and vascular endothelial growth factor (VEGF) play important roles in angiogenesis and tumor growth. Tanshinone IIA (T2A) is a novel antiangiogenic agent with promising antitumor effects; however, the molecular mechanism underlying the antiangiogenic effects of T2A remains unclear. In the present study, we provided evidence showing that T2A inhibited angiogenesis and breast cancer growth by down-regulating VEGF expression. Specifically, T2A repressed HIF-1α expression at the translational level and inhibited the transcriptional activity of HIF-1α, which led to the down-regulation of VEGF expression. Suppression of HIF-1α synthesis by T2A correlated with strong dephosphorylation of mammalian target of rapamycin (mTOR) and its effectors ribosomal protein S6 kinase (p70S6K) and eukaryotic initiation factor 4E-binding protein-1 (4E-BP1), a pathway regulating HIF-1α expression at the translational level. In addition, we also found that T2A inhibited the angiogenesis and growth of human breast cancer xenografts in nude mice through suppression of HIF-1α and VEGF. Our study provides novel perspectives and potential targets for the treatment of human breast cancer.
Collapse
MESH Headings
- Abietanes/pharmacology
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Antineoplastic Agents, Phytogenic
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cell Cycle Proteins
- Cell Line, Tumor
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis
- Mice
- Mice, Nude
- Neoplasm Proteins/metabolism
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Phosphoproteins/metabolism
- Ribosomal Protein S6/metabolism
- Ribosomal Protein S6 Kinases, 70-kDa/metabolism
- Signal Transduction/drug effects
- TOR Serine-Threonine Kinases/metabolism
- Vascular Endothelial Growth Factor A/biosynthesis
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Guobing Li
- College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Changyu Shan
- College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Lei Liu
- College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Ting Zhou
- College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Jing Zhou
- College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Xiaoye Hu
- College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yibiao Chen
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Ning Gao
- College of Pharmacy, Third Military Medical University, Chongqing, China
| |
Collapse
|
44
|
Grace FM, Herbert P, Ratcliffe JW, New KJ, Baker JS, Sculthorpe NF. Age related vascular endothelial function following lifelong sedentariness: positive impact of cardiovascular conditioning without further improvement following low frequency high intensity interval training. Physiol Rep 2015; 3:3/1/e12234. [PMID: 25626864 PMCID: PMC4387763 DOI: 10.14814/phy2.12234] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Aging is associated with diffuse impairments in vascular endothelial function and traditional
aerobic exercise is known to ameliorate these changes. High intensity interval training (HIIT) is
effective at improving vascular function in aging men with existing disease, but its effectiveness
remains to be demonstrated in otherwise healthy sedentary aging. However, the frequency of commonly
used HIIT protocols may be poorly tolerated in older cohorts. Therefore, the present study
investigated the effectiveness of lower frequency HIIT (LfHIIT) on
vascular function in a cohort of lifelong sedentary (SED; n
=22, age 62.7 ± 5.2 years) men compared with a positive
control group of lifelong exercisers (LEX; n = 17, age 61.1 ± 5.4
years). The study consisted of three assessment phases; enrolment to the study (Phase A), following
6 weeks of conditioning exercise in SED (Phase B) and following 6 weeks of low frequency HIIT in
both SED and LEX (LfHIIT; Phase C). Conditioning exercise improved FMD
in SED (3.4 ± 1.5% to 4.9 ± 1.1%; P
<0.01) such that the difference between groups on enrolment (3.4
± 1.5% vs. 5.3 ± 1.4%; P <0.01) was abrogated. This was maintained but not further improved following
LfHIIT in SED whilst FMD remained unaffected by
LfHIIT in LEX. In conclusion, LfHIIT is
effective at maintaining improvements in vascular function achieved during conditioning exercise in
SED. LfHIIT is a well‐tolerated and effective exercise mode for
reducing cardiovascular risk and maintaining but does not improve vascular function beyond that
achieved by conditioning exercise in aging men, irrespective of fitness level. The effects of low frequency high intensity interval training (HIIT) on vascular endothelial
function in lifelong sedentary men remains currently unknown. The present study examined the impact
of low frequency HIIT following conditioning exercise on low determinants of vascular endothelial
function and angiogenic biomarkers in aging men compared with a positive control group of similarly
aged. The major findings of this study indicate that low frequency HIIT is a well‐tolerated
and effective exercise mode for reducing cardiovascular risk and maintaining but not improving
endothelial function beyond that achieved by conditioning exercise in aging men, irrespective of
fitness level.
Collapse
Affiliation(s)
- Fergal M Grace
- Institute of Clinical Exercise & Health Sciences, School of Science and Sport, University of the West of Scotland, Hamilton, UK
| | - Peter Herbert
- Institute of Clinical Exercise & Health Sciences, School of Science and Sport, University of the West of Scotland, Hamilton, UK University of Wales Trinity Saint David, CamarthenWales, UK
| | - John W Ratcliffe
- Institute of Clinical Exercise & Health Sciences, School of Science and Sport, University of the West of Scotland, Hamilton, UK
| | - Karl J New
- School of Health Sport and Professional Practice, University of South Wales, Wales, UK
| | - Julien S Baker
- Institute of Clinical Exercise & Health Sciences, School of Science and Sport, University of the West of Scotland, Hamilton, UK
| | - Nicholas F Sculthorpe
- Institute of Clinical Exercise & Health Sciences, School of Science and Sport, University of the West of Scotland, Hamilton, UK
| |
Collapse
|
45
|
Park SY, Lee SW, Kim HY, Lee WS, Hong KW, Kim CD. HMGB1 induces angiogenesis in rheumatoid arthritis via HIF-1α activation. Eur J Immunol 2015; 45:1216-27. [DOI: 10.1002/eji.201444908] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 11/05/2014] [Accepted: 12/18/2014] [Indexed: 01/02/2023]
Affiliation(s)
- So Youn Park
- Medical Research Center for Ischemic Tissue Regeneration; Pusan National University; Gyeongnam Republic of Korea
| | - Sung Won Lee
- Department of Internal Medicine; College of Medicine; Dong-A University; Busan Republic of Korea
| | - Hye Young Kim
- Medical Research Center for Ischemic Tissue Regeneration; Pusan National University; Gyeongnam Republic of Korea
| | - Won Suk Lee
- Medical Research Center for Ischemic Tissue Regeneration; Pusan National University; Gyeongnam Republic of Korea
- Department of Pharmacology; School of Medicine; Pusan National University; Gyeongnam Republic of Korea
| | - Ki Whan Hong
- Medical Research Center for Ischemic Tissue Regeneration; Pusan National University; Gyeongnam Republic of Korea
| | - Chi Dae Kim
- Medical Research Center for Ischemic Tissue Regeneration; Pusan National University; Gyeongnam Republic of Korea
- Department of Pharmacology; School of Medicine; Pusan National University; Gyeongnam Republic of Korea
| |
Collapse
|
46
|
de Laplanche E, Boudria A, Dacheux E, Vincent A, Gadot N, Assade F, Le Corf K, Leroy X, Mège Lechevallier F, Eymin B, Dalla Venezia N, Simonnet H. Low glucose microenvironment of normal kidney cells stabilizes a subset of messengers involved in angiogenesis. Physiol Rep 2015; 3:3/1/e12253. [PMID: 25602014 PMCID: PMC4387757 DOI: 10.14814/phy2.12253] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
As glucose is a mandatory nutrient for cell proliferation and renewal, it is suspected that glucose microenvironment is sensed by all cell types to regulate angiogenesis. Several glucose-sensing components have been partially described to respond to high glucose levels. However, little is known about the response to low glucose. Here, we used well-differentiated isolated normal rat renal tubules under normal oxygenation conditions to assess the angiogenic response to low glucose. In apparent paradox, but confirming observations made separately in other models, high glucose but also low glucose increased mRNA level of vascular endothelial growth factor A (VEGFA). A subset of mRNAs including hypoxia-inducible factor 1A (HIF1A), angiopoietin receptor (TIE-2), and VEGF receptor 2 (FLK1) were similarly glucose-sensitive and responded to low glucose by increased stability independently of HIF1A and HIF2A proteins. These results contribute to gain some insights as to how normal cells response to low glucose may play a role in the tumor microenvironment.
Collapse
Affiliation(s)
- Elodie de Laplanche
- Université de Lyon, Lyon, F-69000, France Université Lyon 1, Lyon, F-69000, France Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, F-69000, France CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, F-69000, France
| | - Asma Boudria
- Institut Albert Bonniot Equipe 2 Bases Moléculaires de la Progression des Cancers du Poumon, INSERM U823/Université Joseph Fourier, Grenoble, F-38000, France
| | - Estelle Dacheux
- Université de Lyon, Lyon, F-69000, France Université Lyon 1, Lyon, F-69000, France Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, F-69000, France CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, F-69000, France
| | - Anne Vincent
- Université de Lyon, Lyon, F-69000, France Université Lyon 1, Lyon, F-69000, France Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, F-69000, France CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, F-69000, France
| | - Nicolas Gadot
- Université de Lyon, Lyon, F-69000, France Université Lyon 1, Lyon, F-69000, France Department of Pathology, Hôpital Edouard Herriot, Lyon, F-69000, France
| | - Fouzia Assade
- Université de Lyon, Lyon, F-69000, France Université Lyon 1, Lyon, F-69000, France Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, F-69000, France CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, F-69000, France
| | - Katy Le Corf
- Université de Lyon, Lyon, F-69000, France Université Lyon 1, Lyon, F-69000, France Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, F-69000, France CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, F-69000, France
| | - Xavier Leroy
- Institut de Pathologie, CHRU, Faculté de Médecine, Université de Lille, Lille, F-59000, France
| | | | - Béatrice Eymin
- Institut Albert Bonniot Equipe 2 Bases Moléculaires de la Progression des Cancers du Poumon, INSERM U823/Université Joseph Fourier, Grenoble, F-38000, France
| | - Nicole Dalla Venezia
- Université de Lyon, Lyon, F-69000, France Université Lyon 1, Lyon, F-69000, France Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, F-69000, France CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, F-69000, France
| | - Hélène Simonnet
- Université de Lyon, Lyon, F-69000, France Université Lyon 1, Lyon, F-69000, France Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, F-69000, France CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, F-69000, France
| |
Collapse
|
47
|
Du M, Huang K, Huang D, Yang L, Gao L, Wang X, Huang D, Li X, Wang C, Zhang F, Wang Y, Cheng M, Tong Q, Qin G, Huang K, Wang L. Renalase is a novel target gene of hypoxia-inducible factor-1 in protection against cardiac ischaemia–reperfusion injury. Cardiovasc Res 2014; 105:182-91. [DOI: 10.1093/cvr/cvu255] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
48
|
Manresa MC, Godson C, Taylor CT. Hypoxia-sensitive pathways in inflammation-driven fibrosis. Am J Physiol Regul Integr Comp Physiol 2014; 307:R1369-80. [PMID: 25298511 DOI: 10.1152/ajpregu.00349.2014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tissue injury can occur for a variety of reasons, including physical damage, infection, and ischemia. The ability of tissues to effectively recover from injury is a cornerstone of human health. The healing response in tissues is conserved across organs and typically involves distinct but overlapping inflammatory, proliferative, and maturation/resolution phases. If the inflammatory phase is not successfully controlled and appropriately resolved, an excessive healing response characterized by scar formation can lead to tissue fibrosis, a major clinical complication in disorders such as Crohn's disease (CD). As a result of enhanced metabolic and inflammatory processes during chronic inflammation, profound changes in tissue oxygen levels occur leading to localized tissue hypoxia. Therefore, inflammation, fibrosis, and hypoxia are coincidental events during inflammation-driven fibrosis. Our current understanding of the mechanism(s) underpinning fibrosis is limited as are the therapeutic options available. In this review, we discuss what is known about the cellular and molecular mechanisms underpinning inflammation-driven fibrosis and how hypoxia may play a role in shaping this process.
Collapse
Affiliation(s)
- Mario C Manresa
- School of Medicine and Medical Science and the Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Catherine Godson
- School of Medicine and Medical Science and the Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Cormac T Taylor
- School of Medicine and Medical Science and the Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
49
|
Wada H, Yamamoto H, Kim C, Uemura M, Akita H, Tomimaru Y, Hama N, Kawamoto K, Kobayashi S, Eguchi H, Umeshita K, Doki Y, Mori M, Nagano H. Association between ephrin-A1 mRNA expression and poor prognosis after hepatectomy to treat hepatocellular carcinoma. Int J Oncol 2014; 45:1051-8. [PMID: 24969670 DOI: 10.3892/ijo.2014.2519] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 04/17/2014] [Indexed: 11/06/2022] Open
Abstract
Hypoxia regulates the expression of genes that promote tumor growth, angiogenesis and invasion. We previously studied hypoxic tumor cells in vitro and from hepatic metastases of colorectal cancer and determined several potential prognostic factors for hepatocellular carcinoma (HCC). In this study, we evaluated the prognostic impact of the expression of ephrin-A1 (EFNA1) and its receptor, EPHA2, in patients with HCC after curative resection. Samples from a total of 139 HCC patients were analyzed by either microarray alone (n=86) or by microarray and quantitative PCR (n=53). There was no correlation between EFNA1 expression and clinicopathological factors. EPHA2 expression was not significantly correlated with any clinicopathological factors, except for microscopic portal invasion. EFNA1 was an independent prognostic factor for HCC (p=0.0277). These findings suggest that EFNA1 expression may be a useful marker for predicting high risk of recurrence in patients who have undergone curative resection for HCC.
Collapse
Affiliation(s)
- Hiroshi Wada
- Department of Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hirofumi Yamamoto
- Department of Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Chiwan Kim
- Department of Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Mamoru Uemura
- Department of Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hirofumi Akita
- Department of Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yoshito Tomimaru
- Department of Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Naoki Hama
- Department of Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Koichi Kawamoto
- Department of Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shogo Kobayashi
- Department of Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hidetoshi Eguchi
- Department of Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Koji Umeshita
- Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yuichiro Doki
- Department of Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Masaki Mori
- Department of Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hiroaki Nagano
- Department of Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
50
|
Polymorphism in PHD1 gene and risk of non-small cell lung cancer in a Chinese population. Tumour Biol 2014; 35:8921-5. [DOI: 10.1007/s13277-014-2112-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Accepted: 05/15/2014] [Indexed: 02/06/2023] Open
|