1
|
Dabbagh Ohadi MA, Aleyasin MS, Samiee R, Bordbar S, Maroufi SF, Bayan N, Hanaei S, Smith TR. Micro RNAs as a Diagnostic Marker between Glioma and Primary CNS Lymphoma: A Systematic Review. Cancers (Basel) 2023; 15:3628. [PMID: 37509289 PMCID: PMC10377645 DOI: 10.3390/cancers15143628] [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: 04/16/2023] [Revised: 07/04/2023] [Accepted: 07/08/2023] [Indexed: 07/30/2023] Open
Abstract
Differentiating glioma from primary central nervous system lymphoma (PCNSL) can be challenging, and current diagnostic measures such as MRI and biopsy are of limited efficacy. Liquid biopsies, which detect circulating biomarkers such as microRNAs (miRs), may provide valuable insights into diagnostic biomarkers for improved discrimination. This review aimed to investigate the role of specific miRs in diagnosing and differentiating glioma from PCNSL. A systematic search was conducted of PubMed, Scopus, Web of Science, and Embase for articles on liquid biopsies as a diagnostic method for glioma and PCNSL. Sixteen dysregulated miRs were identified with significantly different levels in glioma and PCNSL, including miR-21, which was the most prominent miR with higher levels in PCNSL, followed by glioma, including glioblastoma (GBM), and control groups. The lowest levels of miR-16 and miR-205 were observed in glioma, followed by PCNSL and control groups, whereas miR-15b and miR-301 were higher in both tumor groups, with the highest levels observed in glioma patients. The levels of miR-711 were higher in glioma (including GBM) and downregulated in PCNSL compared to the control group. This review suggests that using these six circulating microRNAs as liquid biomarkers with unique changing patterns could aid in better discrimination between glioma, especially GBM, and PCNSL.
Collapse
Affiliation(s)
- Mohammad Amin Dabbagh Ohadi
- Department of Pediatric Neurological Surgery, Children's Medical Center, Tehran University of Medical Sciences, Tehran 1419733151, Iran
- Interdisciplinary Neuroscience Research Program, Tehran University of Medical Sciences, Tehran 1417755331, Iran
| | - Mir Sajjad Aleyasin
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran 1417755331, Iran
| | - Reza Samiee
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran 1417755331, Iran
| | - Sanaz Bordbar
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran 1417755331, Iran
| | - Seyed Farzad Maroufi
- Department of Pediatric Neurological Surgery, Children's Medical Center, Tehran University of Medical Sciences, Tehran 1419733151, Iran
| | - Nikoo Bayan
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran 1417755331, Iran
| | - Sara Hanaei
- Neurosurgery Department, Imam Khomeini Hospital Complex (IKHC), Tehran University of Medical Sciences, Tehran 1419733151, Iran
| | - Timothy R Smith
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA 02115, USA
| |
Collapse
|
2
|
Thomas R, Yang X. Semaphorins in immune cell function, inflammatory and infectious diseases. CURRENT RESEARCH IN IMMUNOLOGY 2023; 4:100060. [PMID: 37645659 PMCID: PMC10461194 DOI: 10.1016/j.crimmu.2023.100060] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 05/08/2023] [Accepted: 05/14/2023] [Indexed: 08/31/2023] Open
Abstract
The Semaphorin family is a group of proteins studied broadly for their functions in nervous systems. They consist of eight subfamilies ubiquitously expressed in vertebrates, invertebrates, and viruses and exist in membrane-bound or secreted forms. Emerging evidence indicates the relevance of semaphorins outside the nervous system, including angiogenesis, cardiogenesis, osteoclastogenesis, tumour progression, and, more recently, the immune system. This review provides a broad overview of current knowledge on the role of semaphorins in the immune system, particularly its involvement in inflammatory and infectious diseases, including chlamydial infections.
Collapse
Affiliation(s)
- Rony Thomas
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Xi Yang
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
3
|
Tota M, Łacwik J, Laska J, Sędek Ł, Gomułka K. The Role of Eosinophil-Derived Neurotoxin and Vascular Endothelial Growth Factor in the Pathogenesis of Eosinophilic Asthma. Cells 2023; 12:cells12091326. [PMID: 37174726 PMCID: PMC10177218 DOI: 10.3390/cells12091326] [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: 03/10/2023] [Revised: 04/23/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Asthma is a chronic complex pulmonary disease characterized by airway inflammation, remodeling, and hyperresponsiveness. Vascular endothelial growth factor (VEGF) and eosinophil-derived neurotoxin (EDN) are two significant mediators involved in the pathophysiology of asthma. In asthma, VEGF and EDN levels are elevated and correlate with disease severity and airway hyperresponsiveness. Diversity in VEGF polymorphisms results in the variability of responses to glucocorticosteroids and leukotriene antagonist treatment. Targeting VEGF and eosinophils is a promising therapeutic approach for asthma. We identified lichochalcone A, bevacizumab, azithromycin (AZT), vitamin D, diosmetin, epigallocatechin gallate, IGFBP-3, Neovastat (AE-941), endostatin, PEDF, and melatonin as putative add-on drugs in asthma with anti-VEGF properties. Further studies and clinical trials are needed to evaluate the efficacy of those drugs. AZT reduces the exacerbation rate and may be considered in adults with persistent symptomatic asthma. However, the long-term effects of AZT on community microbial resistance require further investigation. Vitamin D supplementation may enhance corticosteroid responsiveness. Herein, anti-eosinophil drugs are reviewed. Among them are, e.g., anti-IL-5 (mepolizumab, reslizumab, and benralizumab), anti-IL-13 (lebrikizumab and tralokinumab), anti-IL-4 and anti-IL-13 (dupilumab), and anti-IgE (omalizumab) drugs. EDN over peripheral blood eosinophil count is recommended to monitor the asthma control status and to assess the efficacy of anti-IL-5 therapy in asthma.
Collapse
Affiliation(s)
- Maciej Tota
- Student Scientific Group of Adult Allergology, Clinical Department of Internal Medicine, Pneumology and Allergology, Wroclaw Medical University, 50-369 Wrocław, Poland
| | - Julia Łacwik
- Student Scientific Group of Microbiology and Immunology, Department of Microbiology and Immunology, Zabrze, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Julia Laska
- Student Scientific Group of Microbiology and Immunology, Department of Microbiology and Immunology, Zabrze, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Łukasz Sędek
- Department of Microbiology and Immunology, Zabrze, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Krzysztof Gomułka
- Clinical Department of Internal Medicine, Pneumology and Allergology, Wroclaw Medical University, 50-369 Wrocław, Poland
| |
Collapse
|
4
|
Jarahian M, Marofi F, Maashi MS, Ghaebi M, Khezri A, Berger MR. Re-Expression of Poly/Oligo-Sialylated Adhesion Molecules on the Surface of Tumor Cells Disrupts Their Interaction with Immune-Effector Cells and Contributes to Pathophysiological Immune Escape. Cancers (Basel) 2021; 13:5203. [PMID: 34680351 PMCID: PMC8534074 DOI: 10.3390/cancers13205203] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/28/2022] Open
Abstract
Glycans linked to surface proteins are the most complex biological macromolecules that play an active role in various cellular mechanisms. This diversity is the basis of cell-cell interaction and communication, cell growth, cell migration, as well as co-stimulatory or inhibitory signaling. Our review describes the importance of neuraminic acid and its derivatives as recognition elements, which are located at the outermost positions of carbohydrate chains linked to specific glycoproteins or glycolipids. Tumor cells, especially from solid tumors, mask themselves by re-expression of hypersialylated neural cell adhesion molecule (NCAM), neuropilin-2 (NRP-2), or synaptic cell adhesion molecule 1 (SynCAM 1) in order to protect themselves against the cytotoxic attack of the also highly sialylated immune effector cells. More particularly, we focus on α-2,8-linked polysialic acid chains, which characterize carrier glycoproteins such as NCAM, NRP-2, or SynCam-1. This characteristic property correlates with an aggressive clinical phenotype and endows them with multiple roles in biological processes that underlie all steps of cancer progression, including regulation of cell-cell and/or cell-extracellular matrix interactions, as well as increased proliferation, migration, reduced apoptosis rate of tumor cells, angiogenesis, and metastasis. Specifically, re-expression of poly/oligo-sialylated adhesion molecules on the surface of tumor cells disrupts their interaction with immune-effector cells and contributes to pathophysiological immune escape. Further, sialylated glycoproteins induce immunoregulatory cytokines and growth factors through interactions with sialic acid-binding immunoglobulin-like lectins. We describe the processes, which modulate the interaction between sialylated carrier glycoproteins and their ligands, and illustrate that sialic acids could be targets of novel therapeutic strategies for treatment of cancer and immune diseases.
Collapse
Affiliation(s)
- Mostafa Jarahian
- German Cancer Research Center, Toxicology and Chemotherapy Unit Heidelberg, 69120 Heidelberg, Germany;
| | - Faroogh Marofi
- Department of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 5165665931, Iran;
| | - Marwah Suliman Maashi
- Stem Cells and Regenerative Medicine Unit at King Fahad Medical Research Centre, Jeddah 11211, Saudi Arabia;
| | - Mahnaz Ghaebi
- Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan 4513956184, Iran;
| | - Abdolrahman Khezri
- Department of Biotechnology, Inland Norway University of Applied Sciences, 2418 Hamar, Norway;
| | - Martin R. Berger
- German Cancer Research Center, Toxicology and Chemotherapy Unit Heidelberg, 69120 Heidelberg, Germany;
| |
Collapse
|
5
|
Sopariwala DH, Likhite N, Pei G, Haroon F, Lin L, Yadav V, Zhao Z, Narkar VA. Estrogen-related receptor α is involved in angiogenesis and skeletal muscle revascularization in hindlimb ischemia. FASEB J 2021; 35:e21480. [PMID: 33788962 PMCID: PMC11135633 DOI: 10.1096/fj.202001794rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 12/19/2022]
Abstract
Skeletal muscle ischemia is a major consequence of peripheral arterial disease (PAD) or critical limb ischemia (CLI). Although therapeutic options for resolving muscle ischemia in PAD/CLI are limited, the issue is compounded by poor understanding of the mechanisms driving muscle vascularization. We found that nuclear receptor estrogen-related receptor alpha (ERRα) expression is induced in murine skeletal muscle by hindlimb ischemia (HLI), and in cultured myotubes by hypoxia, suggesting a potential role for ERRα in ischemic response. To test this, we generated skeletal muscle-specific ERRα transgenic (TG) mice. In these mice, ERRα drives myofiber type switch from glycolytic type IIB to oxidative type IIA/IIX myofibers, which are typically associated with more vascular supply in muscle. Indeed, RNA sequencing and functional enrichment analysis of TG muscle revealed that "paracrine angiogenesis" is the top-ranked transcriptional program activated by ERRα in the skeletal muscle. Immunohistochemistry and angiography showed that ERRα overexpression increases baseline capillarity, arterioles and non-leaky blood vessel formation in the skeletal muscles. Moreover, ERRα overexpression facilitates ischemic neo-angiogenesis and perfusion recovery in hindlimb musculature of mice subjected to HLI. Therefore, ERRα is a hypoxia inducible nuclear receptor that is involved in skeletal muscle angiogenesis and could be potentially targeted for treating PAD/CLI.
Collapse
Affiliation(s)
- Danesh H. Sopariwala
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, TX, USA
| | - Neah Likhite
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, TX, USA
| | - Gungsheng Pei
- Center for Precision Medicine, School of Biomedical Informatics, UTHealth, Houston, TX, USA
| | - Fnu Haroon
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, TX, USA
| | - Lisa Lin
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, TX, USA
- Biochemistry and Cell Biology, Rice University, Houston, TX, USA
| | - Vikas Yadav
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, TX, USA
- Current address: Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Zhongming Zhao
- Center for Precision Medicine, School of Biomedical Informatics, UTHealth, Houston, TX, USA
- Human Genetics Center, School of Public Health, UTHealth, Houston, TX, USA
| | - Vihang A. Narkar
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, TX, USA
- Graduate School of Biomedical Sciences, UTHealth, TX, USA
| |
Collapse
|
6
|
Abdel Ghafar MT, Elkhouly RA, Elnaggar MH, Mabrouk MM, Darwish SA, Younis RL, Elkholy RA. Utility of serum neuropilin-1 and angiopoietin-2 as markers of hepatocellular carcinoma. J Investig Med 2021; 69:1222-1229. [PMID: 33833047 DOI: 10.1136/jim-2020-001744] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2021] [Indexed: 12/24/2022]
Abstract
This study aimed to assess the diagnostic value of two serum angiogenetic markers neuropilin-1 (NRP-1) and angiopoietin-2 (ANG-2) in patients with hepatocellular carcinoma (HCC) and their relation to tumor characteristics. 149 subjects were recruited and divided into 50 patients with recently diagnosed HCC, 49 patients with cirrhosis on top of hepatitis C virus infection, and 50 healthy subjects. Serum NRP-1 and ANG-2 were estimated by ELISA. Alpha-fetoprotein (AFP) levels were measured using fluorescence immunoassay. Serum NRP-1 and ANG-2 levels were significantly higher in patients with HCC (2221.8±1056.6 pg/mL and 3018.5±841.4 pg/mL) than healthy subjects (219.3±61.8 pg/mL and 2007.7±904.8 pg/mL) and patients with cirrhosis (1108.9±526.6 pg/mL and 2179.1±599.2 pg/mL), respectively. In multivariate logistic regression analysis, NRP-1 and AFP were the only independent factors of HCC development and correlated positively with each other (r=0.781, p<0.001). Receiver operating characteristic curve analysis showed that the area under the curve (AUC) of NRP-1 was higher than that of ANG-2 in discriminating HCC from patients with cirrhosis (0.801 vs 0.748, p=0.250) and healthy subjects (0.992 vs 0.809, p<0.001). The AUC of NRP-1 was detected to be increased (0.994) when combined estimation with AFP was performed. Elevated serum NRP-1 and ANG-2 levels were detected in patients with HCC with tumor numbers >3, tumor size ≥5 cm, tumor stages B/C according to the Barcelona Clinic Liver Cancer staging system, vascular invasion, and distant metastasis. In conclusion, NRP-1 is a potential serological marker for HCC diagnosis and is better than ANG-2. It is feasible to be estimated in combination with AFP to enhance its diagnostic power. High serum NRP-1 and ANG-2 levels are associated with advanced HCC tumor characteristics.
Collapse
Affiliation(s)
| | - Reham A Elkhouly
- Tropical Medicine, Tanta University Faculty of Medicine, Tanta, Egypt
| | | | - Mohamed M Mabrouk
- Internal Medicine, Tanta University Faculty of Medicine, Tanta, Egypt
| | - Sara A Darwish
- Clinical Oncology and Nuclear Medicine, Tanta University Faculty of Medicine, Tanta, Egypt
| | - Reham L Younis
- Physiology, Tanta University Faculty of Medicine, Tanta, Egypt
| | - Rasha A Elkholy
- Clinical Pathology, Tanta University Faculty of Medicine, Tanta, Egypt
| |
Collapse
|
7
|
Naderi S, Roshan R, Behdani M, Kazemi-Lomedasht F. Inhibition of neovascularisation in human endothelial cells using anti NRP-1 nanobody fused to truncated form of diphtheria toxin as a novel immunotoxin. Immunopharmacol Immunotoxicol 2021; 43:230-238. [PMID: 33657977 DOI: 10.1080/08923973.2021.1888114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Neuropilin-1 (NRP-1) regulates a range of physiological and pathological processes, including angiogenesis. Targeting of NRP1 is considered a significant approach in cancer therapy. In the present study, a novel antiNRP1 immunotoxin (αNRP1 IT) was developed by genetic fusion of a single domain (VHH) anti-NRP-1 antibody fragment to a truncated diphtheria toxin. The αNRP1 IT was expressed into bacterial cells as an inclusion body (IB). Expression of αNRP1 IT was confirmed by SDS-PAGE and western blotting. Recombinant αNRP1 IT was purified using nickel affinity chromatography. Toxicity and antiangiogenesis effect of αNRP1 IT was investigated both in vitro and in vivo. Results showed that αNRP1 IT significantly reduced the viability of human umbilical vein endothelial cell line (HUVEC) (p < .05). The αNRP1 IT significantly inhibited tube formation of HUVEC cells (p < .001). Furthermore, αNRP1 IT inhibited angiogenesis in Chick Chorioallantoic Membrane (CAM) Assay. These data suggest the potential of αNRP1 IT as a novel therapeutic in targeted cancer therapy.
Collapse
Affiliation(s)
- Shamsi Naderi
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Islamic Republic of Iran
| | - Reyhaneh Roshan
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Islamic Republic of Iran
| | - Mahdi Behdani
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Islamic Republic of Iran
| | - Fatemeh Kazemi-Lomedasht
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Islamic Republic of Iran
| |
Collapse
|
8
|
Slavik H, Balik V, Vrbkova J, Rehulkova A, Vaverka M, Hrabalek L, Ehrmann J, Vidlarova M, Gurska S, Hajduch M, Srovnal J. Identification of Meningioma Patients at High Risk of Tumor Recurrence Using MicroRNA Profiling. Neurosurgery 2021; 87:1055-1063. [PMID: 32125436 PMCID: PMC7566524 DOI: 10.1093/neuros/nyaa009] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 12/15/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Meningioma growth rates are highly variable, even within benign subgroups, with some remaining stable, whereas others grow rapidly. OBJECTIVE To identify molecular-genetic markers for more accurate prediction of meningioma recurrence and better-targeted therapy. METHODS Microarrays identified microRNA (miRNA) expression in primary and recurrent meningiomas of all World Health Organization (WHO) grades. Those found to be deregulated were further validated by quantitative real-time polymerase chain reaction in a cohort of 172 patients. Statistical analysis of the resulting dataset revealed predictors of meningioma recurrence. RESULTS Adjusted and nonadjusted models of time to relapse identified the most significant prognosticators to be miR-15a-5p, miR-146a-5p, and miR-331-3p. The final validation phase proved the crucial significance of miR-146a-5p and miR-331-3p, and clinical factors such as type of resection (total or partial) and WHO grade in some selected models. Following stepwise selection in a multivariate model on an expanded cohort, the most predictive model was identified to be that which included lower miR-331-3p expression (hazard ratio [HR] 1.44; P < .001) and partial tumor resection (HR 3.90; P < .001). Moreover, in the subgroup of total resections, both miRNAs remained prognosticators in univariate models adjusted to the clinical factors. CONCLUSION The proposed models might enable more accurate prediction of time to meningioma recurrence and thus determine optimal postoperative management. Moreover, combining this model with current knowledge of molecular processes underpinning recurrence could permit the identification of distinct meningioma subtypes and enable better-targeted therapies.
Collapse
Affiliation(s)
- Hanus Slavik
- Laboratory of Experimental Medicine, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, Czech Republic
| | - Vladimir Balik
- Laboratory of Experimental Medicine, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, Czech Republic.,Department of Neurosurgery, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, Czech Republic
| | - Jana Vrbkova
- Laboratory of Experimental Medicine, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, Czech Republic
| | - Alona Rehulkova
- Laboratory of Experimental Medicine, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, Czech Republic
| | - Miroslav Vaverka
- Department of Neurosurgery, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, Czech Republic
| | - Lumir Hrabalek
- Department of Neurosurgery, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, Czech Republic
| | - Jiri Ehrmann
- Laboratory of Experimental Medicine, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, Czech Republic.,Institute of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, Czech Republic, Czech Republic
| | - Monika Vidlarova
- Laboratory of Experimental Medicine, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, Czech Republic
| | - Sona Gurska
- Laboratory of Experimental Medicine, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, Czech Republic
| | - Marian Hajduch
- Laboratory of Experimental Medicine, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, Czech Republic
| | - Josef Srovnal
- Laboratory of Experimental Medicine, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, Czech Republic
| |
Collapse
|
9
|
Jiao B, Liu S, Tan X, Lu P, Wang D, Xu H. Class-3 semaphorins: Potent multifunctional modulators for angiogenesis-associated diseases. Biomed Pharmacother 2021; 137:111329. [PMID: 33545660 DOI: 10.1016/j.biopha.2021.111329] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/18/2021] [Accepted: 01/25/2021] [Indexed: 11/29/2022] Open
Abstract
Semaphorins, the neuronal guidance cues, were shown to have broad influences on pathophysiological processes such as bone remodeling, immune responses, and angiogenesis. In particular, Class-3 Semaphorins (SEMA3) is considered a vital regulator involved in angiogenesis. Scientific evidence has pointed to the role of angiogenesis in many diseases, and numerous efforts have been made to explore the possibilities of curing those diseases by targeting angiogenesis. Nevertheless, the efficacies are limited owing to the complex mechanisms of angiogenesis. Hence, investigating the mechanisms of SEMA3 in angiogenesis may contribute to novel therapeutics for diseases. Previous reviews mainly focused on the various functions of semaphorins in one particular disease, and the specific angiogenesis mechanism of SEMA3 in diverse diseases has not been well elucidated. Additionally, the role of SEMA3 in angiogenesis remains elusive, as contradicting results have been found in different disease types. Some evidence from recent studies implies that, while most SEMA3 molecules inhibit pathological angiogenesis in different diseases, occasionally SEMA3 may also promote angiogenesis. This review summarizes the specific role of SEMA3 in a variety of angiogenesis-associated diseases, and documents SEMA3 may be a promising therapeutic target for treating angiogenesis-associated diseases.
Collapse
Affiliation(s)
- Bo Jiao
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shiyang Liu
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xi Tan
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Pei Lu
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Danning Wang
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hui Xu
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
10
|
Abstract
Tumor-homing peptides are widely used for improving tumor selectivity of anticancer drugs and imaging agents. The goal is to increase tumor uptake and reduce accumulation at nontarget sites. Here, we describe current approaches for tumor-homing peptide identification and validation, and provide comprehensive overview of classes of tumor-homing peptides undergoing preclinical and clinical development. We focus on unique mechanistic features and applications of a recently discovered class of tumor-homing peptides, tumor-penetrating C-end Rule (CendR) peptides, that can be used for tissue penetrative targeting of extravascular tumor tissue. Finally, we discuss unanswered questions and future directions in the field of development of peptide-guided smart drugs and imaging agents.
Collapse
|
11
|
MiR-146a Regulates Migration and Invasion by Targeting NRP2 in Circulating-Tumor Cell Mimicking Suspension Cells. Genes (Basel) 2020; 12:genes12010045. [PMID: 33396906 PMCID: PMC7824086 DOI: 10.3390/genes12010045] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/28/2020] [Accepted: 12/28/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer metastasis is the primary cause of cancer-related death and metastatic cancer has circulating-tumor cells (CTCs), which circulate in the bloodstream before invading other organs. Thus, understanding the precise role of CTCs may provide new insights into the metastasis process and reduce cancer mortality. However, the molecular characteristics of CTCs are not well understood due to a lack of number of CTCs. Therefore, suspension cells were generated from MDA-MB-468 cells to mimic CTCs, and we investigate the microRNA (miRNA)-dependent molecular networks and their role in suspension cells. Here, we present an integrated analysis of mRNA and miRNA sequencing data for suspension cell lines, through comparison with adherent cells. Among the differentially regulated miRNA–mRNAs axes, we focus on the miR-146a-Neuropilin2 (NRP2) axis, which is known to influence tumor aggressiveness. We show that miR-146a directly regulates NRP2 expression and inhibits Semaphorin3C (SEMA3C) signaling. Functional studies reveal that miR-146a represses SEMA3C-induced invasion and proliferation by targeting NRP2. Finally, high-NRP2 is shown to be associated with poor outcomes in breast cancer patients. This study identifies the key role of the miR-146a–NRP2 signaling axis that is critical for the regulation of migration and invasion in CTC-mimicking cells.
Collapse
|
12
|
Zhao M, Zhang M, Tao Z, Cao J, Wang L, Hu X. miR-331-3p Suppresses Cell Proliferation in TNBC Cells by Downregulating NRP2. Technol Cancer Res Treat 2020; 19:1533033820905824. [PMID: 32174262 PMCID: PMC7076578 DOI: 10.1177/1533033820905824] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Triple-negative breast cancer is characterized by fast progression with high possible for metastasis and poor survival. Dysfunction of microRNAs plays an important role in the initiation and progression of cancer. Our previous microRNA-seq data indicated the downregulation of miR-331-3p in triple-negative breast cancer tissues compared with that of the noncancer tissues. However, the function of miR-331-3p in triple-negative breast cancer remains largely unknown. Herein, the involvement of miR-331-3p in triple-negative breast cancer was investigated and the therapeutic potential of miR-331-3p was also explored. METHODS Real-time quantitative polymerase chain reaction was performed to detect the expression of miR-331-3p in triple-negative breast cancer tissues and cell lines. The cell proliferation was determined by the cell counting kit-8 assay. Apoptosis of triple-negative breast cancer cells was examined by annexin V/propidium iodide staining. miRDB database was used to predict the potential targets of miR-331-3p. Western blot was performed to examine the expression of the target protein. RESULTS miR-331-3p was significantly downregulated in triple-negative breast cancer tissues and cell line. Lower miR-331-3p expression was significantly correlated with the tumor size, TNM stage, and lymph node metastasis of patients with triple-negative breast cancer. Functional experiments showed that the overexpression of miR-331-3p inhibited the proliferation and increased apoptosis of triple-negative breast cancer cells. Neuropilin-2 was identified as a target of miR-331-3p, which harbored binding site of miR-331-3p in its 3'-untranslated region. Overexpression of miR-331-3p decreased the messenger RNA and protein levels of neuropilin-2 in triple-negative breast cancer cells. Restoration of neuropilin-2 partially reversed the inhibitory effects of miR-331-3p on the proliferation of triple-negative breast cancer cells. CONCLUSIONS Our results demonstrated the novel function of miR-331-3p/neuropilin-2 signaling in regulating the malignant behaviors of triple-negative breast cancer cells, which suggested miR-331-3p as a potential target for the treatment of triple-negative breast cancer.
Collapse
Affiliation(s)
- Mingchuan Zhao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Fudan University School of Medicine, Shanghai, People's Republic of China
| | - Mengmeng Zhang
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Zhonghua Tao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Fudan University School of Medicine, Shanghai, People's Republic of China
| | - Jun Cao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Fudan University School of Medicine, Shanghai, People's Republic of China
| | - Leiping Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Fudan University School of Medicine, Shanghai, People's Republic of China
| | - Xichun Hu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Fudan University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
13
|
Chen Z, Gao H, Dong Z, Shen Y, Wang Z, Wei W, Yi J, Wang R, Wu N, Jin S. NRP1 regulates radiation-induced EMT via TGF-β/Smad signaling in lung adenocarcinoma cells. Int J Radiat Biol 2020; 96:1281-1295. [PMID: 32659143 DOI: 10.1080/09553002.2020.1793015] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
PURPOSE Radiation has been shown to promote the epithelial-mesenchymal transition (EMT) in tumor cells, and TGF-β/Smad and PI3K-Akt signaling pathways play an important role in the EMT. In this study, we investigated the effects of neuropilin-1 (NRP1) on radiation-induced TGF-β/Smad and non-classical Smad signaling pathways in lung cancer cells, as well as the effects of NRP1 on invasion and migration. MATERIALS AND METHODS Changes in the expression levels of EMT markers (β-catenin, N-cadherin, and vimentin) and related transcription factors (Twist and ZEB1) in stably transfected cells were detected by Western blotting and qPCR, and changes were assessed by TGF-β/Smad and non-classical Smad signaling. Immunofluorescence was used to detect the expression of the cytoskeletal protein F-actin. Expression of TGF-β1 and CXCL-12 was detected by ELISA. Transwell and scratch assays were used to detect the invasive ability and migration of lung cancer cells, respectively. RESULTS Our results showed that ionizing radiation could induce the EMT as well as morphological changes in lung adenocarcinoma cells (A549); however, the effects were not significant in lung squamous carcinoma cells (SK-MES-1). Moreover, we showed that NRP1 promotes the EMT induced by ionizing radiation in A549 cells, which may be related to the increased expression of EMT-related transcription factors. NRP1 may promote the radiation-induced EMT of A549 cells mainly through TGF-β1/Smad2/3 signaling. NRP1 also enhanced radiation-induced invasion, migration, and CXCL-12 expression in A549 cells. CONCLUSIONS We conclude that NRP1 promotes radiation-induced EMT in lung adenocarcinoma cells via TGF-β1/Smad signaling and not non-classical Smad signaling, and enhances the invasion and migration of lung adenocarcinoma cells.
Collapse
Affiliation(s)
- ZhiYuan Chen
- School of Public Health, NHC Key Laboratory of Radiobiology, Jilin University, Changchun, Jilin, China.,Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Hui Gao
- School of Public Health, NHC Key Laboratory of Radiobiology, Jilin University, Changchun, Jilin, China.,Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Zhuo Dong
- School of Public Health, NHC Key Laboratory of Radiobiology, Jilin University, Changchun, Jilin, China
| | - YanNan Shen
- School of Public Health, NHC Key Laboratory of Radiobiology, Jilin University, Changchun, Jilin, China
| | - ZhiCheng Wang
- School of Public Health, NHC Key Laboratory of Radiobiology, Jilin University, Changchun, Jilin, China
| | - Wei Wei
- School of Public Health, NHC Key Laboratory of Radiobiology, Jilin University, Changchun, Jilin, China
| | - JunXuan Yi
- School of Public Health, NHC Key Laboratory of Radiobiology, Jilin University, Changchun, Jilin, China
| | - Rui Wang
- School of Public Health, NHC Key Laboratory of Radiobiology, Jilin University, Changchun, Jilin, China
| | - Ning Wu
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Shunzi Jin
- School of Public Health, NHC Key Laboratory of Radiobiology, Jilin University, Changchun, Jilin, China
| |
Collapse
|
14
|
Neuropilin1 Expression Acts as a Prognostic Marker in Stomach Adenocarcinoma by Predicting the Infiltration of Treg Cells and M2 Macrophages. J Clin Med 2020; 9:jcm9051430. [PMID: 32408477 PMCID: PMC7290937 DOI: 10.3390/jcm9051430] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/27/2020] [Accepted: 05/09/2020] [Indexed: 12/14/2022] Open
Abstract
Neuropilin1 (NRP1) plays a critical role in tumor progression and immune responses. Although the roles of NRP1 in various tumors have been investigated, the clinical relevance of NRP1 expression in stomach adenocarcinoma (STAD) has not been studied. To investigate the use of NRP1 as a prognostic biomarker of STAD, we analyzed NRP1 mRNA expression and its correlation with patient survival and immune cell infiltration using various databases. NRP1 mRNA expression was significantly higher in STAD than normal tissues, and Kaplan-Meier survival analysis showed that NRP1 expression was significantly associated with poor prognosis in patients with STAD. To elucidate the related mechanism, we analyzed the correlation between NRP1 expression and immune cell infiltration level. In particular, the infiltration of immune-suppressive cells, such as regulatory T (Treg) cells and M2 macrophage, was significantly increased by NRP1 expression. In addition, the expression of interleukin (IL)-35, IL-10, and TGF-β1 was also positively correlated with NRP1 expression, resulting in the immune suppression. Collectively in this study, our integrated analysis using various clinical databases shows that the significant correlation between NRP1 expression and the infiltration of Treg cells and M2 macrophage explains poor prognosis mechanism in STAD, suggesting the clinical relevance of NRP1 expression as a prognostic biomarker for STAD patients.
Collapse
|
15
|
Gamper C, Spenlé C, Boscá S, van der Heyden M, Erhardt M, Orend G, Bagnard D, Heinlein M. Functionalized Tobacco Mosaic Virus Coat Protein Monomers and Oligomers as Nanocarriers for Anti-Cancer Peptides. Cancers (Basel) 2019; 11:cancers11101609. [PMID: 31652529 PMCID: PMC6826726 DOI: 10.3390/cancers11101609] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/14/2019] [Accepted: 10/18/2019] [Indexed: 01/13/2023] Open
Abstract
Components with self-assembly properties derived from plant viruses provide the opportunity to design biological nanoscaffolds for the ordered display of agents of diverse nature and with complementing functions. With the aim of designing a functionalized nanoscaffold to target cancer, the coat protein (CP) of Tobacco mosaic virus (TMV) was tested as nanocarrier for an insoluble, highly hydrophobic peptide that targets the transmembrane domain of the Neuropilin-1 (NRP1) receptor in cancer cells. The resulting construct CPL-K (CP-linker-“Kill”) binds to NRP1 in cancer cells and disrupts NRP1 complex formation with PlexA1 as well as downstream Akt survival signaling. The application of CPL-K also inhibits angiogenesis and cell migration. CP was also fused to a peptide that targets the extracellular domain of NRP1 and this fusion protein (CPL-F, CP-Linker-“Find”) is shown to bind to cultured cancer cells and to inhibit NRP1-dependent angiogenesis as well. CPL-K and CPL-F maintain their anti-angiogenic properties upon co-assembly to oligomers/nanoparticles together with CPL. The observations show that the CP of TMV can be employed to generate a functionalized nanoparticle with biological activity. Remarkably, fusion to CPL allowed us to solubilize the highly insoluble transmembrane NRP1 peptide and to retain its anti-angiogenic effect.
Collapse
Affiliation(s)
- Coralie Gamper
- Institut de Biologie Moléculaire des Plantes (IBMP-CNRS), Université de Strasbourg, 67000 Strasbourg, France.
- INSERM 1119, BMNST Laboratory, Université de Strasbourg, 67000 Strasbourg, France.
- Labex Medalis, Université de Strasbourg, 67000 Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg, FMTS, Université de Strasbourg, 67000 Strasbourg, France.
- INSERM 1109, MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Université de Strasbourg, 67000 Strasbourg, France.
| | - Caroline Spenlé
- Institut de Biologie Moléculaire des Plantes (IBMP-CNRS), Université de Strasbourg, 67000 Strasbourg, France.
- INSERM 1119, BMNST Laboratory, Université de Strasbourg, 67000 Strasbourg, France.
- Labex Medalis, Université de Strasbourg, 67000 Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg, FMTS, Université de Strasbourg, 67000 Strasbourg, France.
- INSERM 1109, MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Université de Strasbourg, 67000 Strasbourg, France.
| | - Sonia Boscá
- Institut de Biologie Moléculaire des Plantes (IBMP-CNRS), Université de Strasbourg, 67000 Strasbourg, France.
| | - Michael van der Heyden
- INSERM 1119, BMNST Laboratory, Université de Strasbourg, 67000 Strasbourg, France.
- Labex Medalis, Université de Strasbourg, 67000 Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg, FMTS, Université de Strasbourg, 67000 Strasbourg, France.
- INSERM 1109, MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Université de Strasbourg, 67000 Strasbourg, France.
| | - Mathieu Erhardt
- Institut de Biologie Moléculaire des Plantes (IBMP-CNRS), Université de Strasbourg, 67000 Strasbourg, France.
| | - Gertraud Orend
- Labex Medalis, Université de Strasbourg, 67000 Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg, FMTS, Université de Strasbourg, 67000 Strasbourg, France.
- INSERM 1109, MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Université de Strasbourg, 67000 Strasbourg, France.
- INSERM 1109, The Tumor Microenvironment Laboratory, Université de Strasbourg, 67000 Strasbourg, France.
| | - Dominique Bagnard
- INSERM 1119, BMNST Laboratory, Université de Strasbourg, 67000 Strasbourg, France.
- Labex Medalis, Université de Strasbourg, 67000 Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg, FMTS, Université de Strasbourg, 67000 Strasbourg, France.
- INSERM 1109, MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Université de Strasbourg, 67000 Strasbourg, France.
| | - Manfred Heinlein
- Institut de Biologie Moléculaire des Plantes (IBMP-CNRS), Université de Strasbourg, 67000 Strasbourg, France.
- University of Strasbourg Institute of Advanced Study (USIAS), 67000 Strasbourg, France.
| |
Collapse
|
16
|
Bartosińska J, Michalak-Stoma A, Kowal M, Raczkiewicz D, Krasowska D, Chodorowska G, Giannopoulos K. Analysis of circulating soluble programmed death 1 (PD-1), neuropilin 1 (NRP-1) and human leukocyte antigen-G (HLA-G) in psoriatic patients. Postepy Dermatol Alergol 2019; 36:167-172. [PMID: 31320849 PMCID: PMC6627263 DOI: 10.5114/ada.2018.73329] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 01/03/2018] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Circulating soluble programmed death 1 (PD-1), neuropilin 1 (NRP-1) and human leukocyte antigen-G (HLA-G) take part in modulating immune tolerance causing disturbances in the molecular mechanisms responsible for maintenance of balance between effector and regulatory components of the immune system. Since their cell-surface expression levels were found to be changed in lesional and/or non-lesional skin of psoriatic patients, analysis of soluble PD-1, NRP-1 and HLA-G concentrations sheds more light on their role in detecting unbalanced immune tolerance in psoriasis. AIM To assess soluble PD-1, NRP-1 and HLA-G concentrations in psoriasis. MATERIAL AND METHODS The study included 57 psoriatic patients and 29 controls. Duration of psoriasis was in the range 1 to 55 years; the median was 19 years. The plasma concentrations of soluble HLA-G (sHLA-G), soluble NRP-1 (sNRP-1) and soluble PD-1 (sPD-1) were examined using the ELISA method. Severity of the skin lesions was assessed by means of Psoriasis Area Severity Index (PASI), body surface area (BSA) and Physician Global Assessment (PGA). RESULTS Psoriasis Area Severity Index in the studied group was in the range 3 to 43; the median was 12. Body surface area was in the range 2-75%; the median was 15%. The median value of PGA was 3. Soluble NRP concentration was significantly higher in the psoriatic patients (median: 1.59 pg/ml; range: 0.67-2.62 pg/ml) than in the control group (median: 1.35 pg/ml; range: 0.05-2.61 pg/ml) (p = 0.010). Soluble PD-1 and sHLA-G concentrations were not significantly different between the studied and control groups (p = 0.094 and p = 0.482, respectively). CONCLUSIONS Increased concentrations of sNRP-1 and unchanged values of sHLA-G and sPD-1 concentrations may be indicative of impaired immune tolerance mechanisms in psoriasis.
Collapse
Affiliation(s)
- Joanna Bartosińska
- Department of Dermatology, Venereology and Paediatric Dermatology, Medical University of Lublin, Lublin, Poland
| | - Anna Michalak-Stoma
- Department of Dermatology, Venereology and Paediatric Dermatology, Medical University of Lublin, Lublin, Poland
| | - Małgorzata Kowal
- Department of Dermatology, Venereology and Paediatric Dermatology, Medical University of Lublin, Lublin, Poland
| | - Dorota Raczkiewicz
- Institute of Statistics and Demography, Warsaw School of Economics, Warsaw, Poland
| | - Dorota Krasowska
- Department of Dermatology, Venereology and Paediatric Dermatology, Medical University of Lublin, Lublin, Poland
| | - Grażyna Chodorowska
- Department of Dermatology, Venereology and Paediatric Dermatology, Medical University of Lublin, Lublin, Poland
| | - Krzysztof Giannopoulos
- Experimental Hematooncology Department, Medical University of Lublin, Lublin, Poland
- Hematology Department, St John’s Cancer Center, Lublin, Poland
| |
Collapse
|
17
|
Angiogenic effect of platelet-rich concentrates on dental pulp stem cells in inflamed microenvironment. Clin Oral Investig 2019; 23:3821-3831. [PMID: 30687907 DOI: 10.1007/s00784-019-02811-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 01/11/2019] [Indexed: 01/04/2023]
Abstract
OBJECTIVE In this study, we aimed to determine the suitable concentrations of human platelet lysate (HPL) and platelet-rich plasma (PRP) for maintaining the in vitro proliferative and angiogenic potential of inflamed dental pulp stem cells. MATERIALS AND METHODS Lipopolysaccharide (LPS)-induced inflamed dental pulp-derived stem cells (iDPSCs) were treated with different concentrations of HPL and PRP (10% and 20%) followed by determination of viability using Alamar Blue assay. Expression of angiogenesis-, adhesion-, and inflammation-regulating genes was also analyzed using RT-qPCR array. Furthermore, expression of growth factors at protein level in the cell culture microenvironment was measured using multiplex assay. RESULTS Viability of iDPSCs was significantly (p < 0.05) higher in 20% HPL-supplemented media compared to iDPSCs. Expression of 10 out of 12 selected angiogenic genes, four out of seven adhesion molecules, and seven out of nine cytokine-producing genes were significantly (p < 0.05) higher in cells maintained in 20% HPL-supplemented media compared to that in FBS-supplemented media. Furthermore, expression of all the selected growth factors was significantly higher (p < 0.05) in the supernatants from 20% HPL media at 12 and 24 h post-incubation. CONCLUSION This study suggests that 20% HPL could be optimum to stimulate angiogenesis-related factors in iDPSCs while maintaining their viability. CLINICAL RELEVANCE This data may suggest the potential use of 20% HPL for expanding DPSCs scheduled for clinical trials for regenerative therapies including dental pulp regeneration.
Collapse
|
18
|
Stallone G, Matteo M, Netti GS, Infante B, Di Lorenzo A, Prattichizzo C, Carlucci S, Trezza F, Gesualdo L, Greco P, Grandaliano G. Semaphorin 3F expression is reduced in pregnancy complicated by preeclampsia. An observational clinical study. PLoS One 2017; 12:e0174400. [PMID: 28350837 PMCID: PMC5370113 DOI: 10.1371/journal.pone.0174400] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 03/08/2017] [Indexed: 12/22/2022] Open
Abstract
Background and objective Preeclampsia is a systemic disorder, affecting 2–10% of pregnancies, characterized by a deregulated pro- and anti-angiogenic balance. Semaphorin 3F is an angiogenesis inhibitor. We aimed to investigate whether semaphorin 3F expression is modulated in preeclampsia. Design, setting, participants, and measurements We performed two observational single center cohort studies between March 2013 and August 2014. In the first we enrolled 110 consecutive women, undergoing an elective cesarean section; in the second we included 150 consecutive women undergoing amniocentesis for routine clinical indications at 16–18 week of gestation. Semaphorin 3F concentration was evaluated in maternal peripheral blood, venous umbilical blood and amniotic fluid, along with its placenta protein expression at the time of delivery in the first study group and in the amniotic fluid at 16–18 weeks of gestation in the second study group. Results In the first study 19 patients presented at delivery with preeclampsia. Semaphorin 3F placenta tissue expression was significantly reduced in preeclampsia. In addition, semaphorin 3F level at delivery was significantly lower in serum, amniotic fluid and venous umbilical blood of preeclamptic patients compared with normal pregnant women. In the prospective cohort study 14 women developed preeclampsia. In this setting, semaphorin 3F amniotic level at 16–18 weeks of gestation was reduced in women who subsequently developed preeclampsia compared to women with a normal pregnancy. ROC curve analysis showed that semaphorin 3F amniotic levels could identify women at higher risk of preeclampsia. Conclusions Semaphorin 3F might represent a predictive biomarker of preeclampsia.
Collapse
Affiliation(s)
- Giovanni Stallone
- Nephrology Dialysis and Transplantation Unit, Dept. of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, Foggia, Italy
| | - Maria Matteo
- Gynaecologic and Obstetric Unit, Dept. of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, Foggia, Italy
| | - Giuseppe Stefano Netti
- Clinical Pathology Unit, Dept. of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, Foggia, Italy
| | - Barbara Infante
- Nephrology Dialysis and Transplantation Unit, Dept. of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, Foggia, Italy
| | - Adelaide Di Lorenzo
- Nephrology Dialysis and Transplantation Unit, Dept. of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, Foggia, Italy
| | - Clelia Prattichizzo
- Nephrology Dialysis and Transplantation Unit, Dept. of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, Foggia, Italy
| | - Stefania Carlucci
- Gynaecologic and Obstetric Unit, Dept. of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, Foggia, Italy
| | - Federica Trezza
- Gynaecologic and Obstetric Unit, Dept. of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, Foggia, Italy
| | - Loreto Gesualdo
- Nephrology Dialysis and Transplantation Unit, Dept. of Emergency and Organ Transplantation, University of Bari “A. Moro”, Piazza G. Cesare 11, Bari, Italy
| | - Pantaleo Greco
- Gynaecologic and Obstetric Unit, Dept. of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, Foggia, Italy
| | - Giuseppe Grandaliano
- Nephrology Dialysis and Transplantation Unit, Dept. of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, Foggia, Italy
- * E-mail:
| |
Collapse
|
19
|
131I-labeled monoclonal antibody targeting neuropilin receptor type-2 for tumor SPECT imaging. Int J Oncol 2016; 50:649-659. [PMID: 28000859 DOI: 10.3892/ijo.2016.3808] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 12/05/2016] [Indexed: 11/05/2022] Open
Abstract
As a co-receptor for vascular endothelial growth factor‑3 (VEGF‑3), neuropilin receptor type‑2 (NRP‑2) plays a central role in lymphangiogenesis and angiogenesis. Recently, mounting data of evidence show that NRP‑2 is overexpressed in several human cancers, and its overexpression is often associated with poor prognosis. Therefore, it is necessary for us to develop an affinity reagent for noninvasive imaging of NRP‑2 expression because it may be possible to provide early cancer diagnosis, more accurate prognosis, and better treatment planning. Due to their high affinity, and specificity, monoclonal antibodies (mAbs) have been considered attractive candidates for targeted cancer therapy and diagnostics. We recently generated and validated a monoclonal antibody that specifically binds NRP‑2 b1b2 domain with no cross‑reactivity to NRP‑1 b1b2 domain, also known to be overexpressed in a variety of cancers. Here, we developed a single photon emission computed tomography (SPECT) probe for imaging NRP‑2- positive tumors. Anti‑NRP‑2 monoclonal antibodies were prepared by hybridomas and were labeled with iodine‑131 by chloramine‑T method. The in vitro physicochemical properties of 131I‑anti‑NRP‑2 mAb was determined. Binding affinity and specificity of 131I‑anti‑NRP‑2 mAb to NRP‑2 were assessed using human lung adenocarcinoma A549 cells. Biodistribution and SPECT studies were performed in mice bearing A549 tumor xenografts to evaluate the in vivo performance of 131I‑anti‑NRP‑2 mAb. The preparation of anti‑NRP‑2 mAb was completed successfully by hybridoma with high purity (>95%) and specific for NRP‑2 b1b2 domain, but not NRP‑1 b1b2 domain. The radiosynthesis of 131I‑anti‑NRP‑2 mAb was completed successfully within 60 min with high labelling efficiency (94.69±3.63%), and radiochemical purity (98.56±0.48%). The resulting probe, 131I‑anti‑NRP‑2 mAb displayed excellent stability in PBS solution during 24-72 h. 131I‑anti‑NRP‑2 mAb showed high binding affinity with A549 cells (96.6±1.44 nM). In vivo biodistribution and SPECT studies demonstrated targeting of A549 glioma xenografts was NRP‑2 specific. The tumor uptake was 5.86±0.27% ID/g at 6 h, and kept at high level of 4.64±0.82% ID/g at 72 h‑post‑injection. The tumor to contralateral muscle ratio (T/NT) was 2.08±0.33 at 6 h, and reached the highest level of 3.83±0.18 at 72 h after injection. SPECT imaging studies revealed that 131I‑anti‑NRP‑2 mAb could clearly identify A549 tumors with good contrast, especially at 48‑72 h after injection. In conclusion, this study demonstrates that 131I‑anti‑NRP‑2 mAb exhibited highly selective uptake in NRP‑2‑expressing tumors, and may provide a promising SPECT probe for imaging NRP‑2 positive tumors.
Collapse
|
20
|
Yang Y, Chen N, Li Z, Wang XJ, Wang SY, Tingwu, Luo FH, Yan JH. Preparation, Purification, and Identification of a Monoclonal Antibody Against NRP2 b1b2 Domain. Monoclon Antib Immunodiagn Immunother 2016; 34:354-9. [PMID: 26492624 DOI: 10.1089/mab.2015.0025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
First identified as a high-affinity kinase-deficient receptor for class-3 semaphorins and vascular endothelial growth factor (VEGF) families, Neuropilin2 (NRP2) is a transmembrane non-tyrosine-kinase glycoprotein that has a vital function in neuronal patterning. Furthermore, NRP2 expression is often upregulated in cancer tissues and correlated with poor prognosis. In the present study, we report the establishment of a monoclonal antibody specific for NRP2b1b2 domain (NRP2 MAb) through hybridoma method. NRP2 MAb is measured to have a titer of 5.12 × 10(5) against NRP2b1b2 in indirect ELISA. Western blotting, flow cytometry, and immunofluorescence analysis indicate that NRP2 MAb can combine full-length NRP2 in LoVo and SW480 cells. Besides helping further understand NRP2-related pathological mechanisms and cell-signaling pathways, NRP2 MAb may act as a therapeutic agent for cancer in the future.
Collapse
Affiliation(s)
- Yun Yang
- Cancer Research Center, Medical College, Xiamen University , Xiamen, Fujian, China
| | - Na Chen
- Cancer Research Center, Medical College, Xiamen University , Xiamen, Fujian, China
| | - Zhe Li
- Cancer Research Center, Medical College, Xiamen University , Xiamen, Fujian, China
| | - Xian-Jiang Wang
- Cancer Research Center, Medical College, Xiamen University , Xiamen, Fujian, China
| | - Sheng-Yu Wang
- Cancer Research Center, Medical College, Xiamen University , Xiamen, Fujian, China
| | - Tingwu
- Cancer Research Center, Medical College, Xiamen University , Xiamen, Fujian, China
| | - Fang-Hong Luo
- Cancer Research Center, Medical College, Xiamen University , Xiamen, Fujian, China
| | - Jiang-Hua Yan
- Cancer Research Center, Medical College, Xiamen University , Xiamen, Fujian, China
| |
Collapse
|
21
|
Loegl J, Nussbaumer E, Hiden U, Majali-Martinez A, Ghaffari-Tabrizi-Wizy N, Cvitic S, Lang I, Desoye G, Huppertz B. Pigment epithelium-derived factor (PEDF): a novel trophoblast-derived factor limiting feto-placental angiogenesis in late pregnancy. Angiogenesis 2016; 19:373-88. [PMID: 27278471 PMCID: PMC4930480 DOI: 10.1007/s10456-016-9513-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 05/13/2016] [Indexed: 12/24/2022]
Abstract
The rapidly expanding feto-placental vasculature needs tight control by paracrine and endocrine mechanisms. Here, we focused on paracrine influence by trophoblast, the placental epithelium. We aimed to identify differences in regulation of feto-placental angiogenesis in early versus late pregnancy. To this end, the effect of conditioned media (CM) from early and late pregnancy human trophoblast was tested on network formation, migration and proliferation of human feto-placental endothelial cells. Only CM of late pregnancy trophoblast reduced network formation and migration. Screening of trophoblast transcriptome for anti-angiogenic candidates identified pigment epithelium-derived factor (PEDF) with higher expression and protein secretion in late pregnancy trophoblast. Addition of a PEDF-neutralizing antibody restored the anti-angiogenic effect of CM from late pregnancy trophoblast. Notably, human recombinant PEDF reduced network formation only in combination with VEGF. Also in the CAM assay, the combination of PEDF with VEGF reduced branching of vessels below control levels. Analysis of phosphorylation of ERK1/2 and FAK, two key players in VEGF-induced proliferation and migration, revealed that PEDF altered VEGF signaling, while PEDF alone did not affect phosphorylation of ERK1/2 and FAK. These data suggest that the trophoblast-derived anti-angiogenic molecule PEDF is involved in restricting growth and expansion of the feto-placental endothelium predominantly in late pregnancy and targets to modulate the intracellular effect of VEGF.
Collapse
Affiliation(s)
- Jelena Loegl
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria.,Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Erika Nussbaumer
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Ursula Hiden
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria.
| | | | | | - Silvija Cvitic
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria.,Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Ingrid Lang
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Gernot Desoye
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Berthold Huppertz
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| |
Collapse
|
22
|
Aberrant regulation of miR-15b in human malignant tumors and its effects on the hallmarks of cancer. Tumour Biol 2015; 37:177-83. [DOI: 10.1007/s13277-015-4269-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 10/15/2015] [Indexed: 12/22/2022] Open
|
23
|
Wittmann P, Grubinger M, Gröger C, Huber H, Sieghart W, Peck-Radosavljevic M, Mikulits W. Neuropilin-2 induced by transforming growth factor-β augments migration of hepatocellular carcinoma cells. BMC Cancer 2015; 15:909. [PMID: 26573807 PMCID: PMC4647494 DOI: 10.1186/s12885-015-1919-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 11/09/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the most common form of liver cancer and the third most lethal cancer worldwide. The epithelial to mesenchymal transition (EMT) describes the transformation of well-differentiated epithelial cells to a de-differentiated phenotype and plays a central role in the invasion and intrahepatic metastasis of HCC cells. Modulation of the transforming growth factor-β (TGF-β) signaling is known to induce various tumor-promoting and EMT-inducing pathways in HCC. The meta-analysis of a panel of EMT gene expression studies revealed that neuropilin 2 (NRP2) is significantly upregulated in cells that have undergone EMT induced by TGF-β. In this study we assessed the functional role of NRP2 in epithelial and mesenchymal-like HCC cells and focused on the molecular interplay between NRP2 and TGF-β/Smad signaling. METHODS NRP2 expression was analyzed in human HCC cell lines and tissue arrays comprising 133 HCC samples. Cell migration was examined by wound healing and Transwell assays in the presence and absence of siRNA against NRP2. NRP2 and TGF-β signaling were analyzed by Western blotting and confocal immunofluorescence microscopy. RESULTS We show that NRP2 is particularly expressed in HCC cell lines with a dedifferentiated, mesenchymal-like phenotype. NRP2 expression is upregulated by the canonical TGF-β/Smad signaling while NRP2 expression has no impact on TGF-β signaling in HCC cells. Reduced expression of NRP2 by knock-down or inhibition of TGF-β signaling resulted in diminished cell migration independently of each other, suggesting that NRP2 fails to collaborate with TGF-β signaling in cell movement. In accordance with these data, elevated levels of NRP2 correlated with a higher tumor grade and less differentiation in a large collection of human HCC specimens. CONCLUSIONS These data suggest that NRP2 associates with a less differentiated, mesenchymal-like HCC phenotype and that NRP2 plays an important role in tumor cell migration upon TGF-β-dependent HCC progression.
Collapse
Affiliation(s)
- Philipp Wittmann
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| | - Markus Grubinger
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| | - Christian Gröger
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| | - Heidemarie Huber
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| | - Wolfgang Sieghart
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria.
| | - Markus Peck-Radosavljevic
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria.
| | - Wolfgang Mikulits
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
24
|
Chen C, Hu Y, Li L. NRP1 is targeted by miR-130a and miR-130b, and is associated with multidrug resistance in epithelial ovarian cancer based on integrated gene network analysis. Mol Med Rep 2015; 13:188-96. [PMID: 26573160 PMCID: PMC4686085 DOI: 10.3892/mmr.2015.4556] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 10/19/2015] [Indexed: 11/29/2022] Open
Abstract
Multidrug resistance (MDR) in epithelial ovarian cancer (EOC) remains a public health issue for women worldwide, and its molecular mechanisms remain to be fully elucidated. The present study aimed to predict the potential genes involved in MDR, and examine the mechanisms underlying MDR in EOC using bioinformatics techniques. In the present study, four public microarray datasets, including GSE41499, GSE33482, GSE15372 and GSE28739, available in Gene Expression Omnibus were downloaded, and 11 microRNAs (miRNA; miRs), including miR-130a, miR-214, let-7i, miR-125b, miR-376c, miR-199a, miR-93, miR-141, miR-130b, miR-193b* and miR-200c, from previously published reports in PubMed were used to perform a comprehensive bioinformatics analysis through gene expression analysis, signaling pathway analysis, literature co-occurrence and miRNA-mRNA interaction networks. The results demonstrated that the expression of neuropilin 1 (NRP1) was upregulated, thereby acting as the most important hub gene in the integrated gene network. NRP1 was targeted by miR-130a and miR-130b at the binding site of chromosome 10: 33466864-3466870, which was involved in the axon guidance signaling pathway. These results suggested that alteration of the gene expression levels of NRP1 expression may contribute to MDR in EOC. These data provide important information for further experimental investigations of the drug resistance-associated functions of NRP1 in EOC.
Collapse
Affiliation(s)
- Changxian Chen
- Department of Gynecological Oncology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yanling Hu
- Department of Bioinformatics, Medical Research Center, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Li Li
- Department of Gynecological Oncology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
25
|
Graziani G, Lacal PM. Neuropilin-1 as Therapeutic Target for Malignant Melanoma. Front Oncol 2015; 5:125. [PMID: 26090340 PMCID: PMC4453476 DOI: 10.3389/fonc.2015.00125] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 05/19/2015] [Indexed: 01/13/2023] Open
Abstract
Neuropilin-1 (NRP-1) is a transmembrane glycoprotein that acts as a co-receptor for various members of the vascular endothelial growth factor (VEGF) family. Its ability to bind or modulate the activity of a number of other extracellular ligands, such as class 3 semaphorins, TGF-β, HGF, FGF, and PDGF, has suggested the involvement of NRP-1 in a variety of physiological and pathological processes. Actually, this co-receptor has been implicated in axon guidance, angiogenesis, and immune responses. NRP-1 is also expressed in a variety of cancers (prostate, lung, pancreatic, or colon carcinoma, melanoma, astrocytoma, glioblastoma, and neuroblastoma), suggesting a critical role in tumor progression. Moreover, a growing amount of evidence indicates that NRP-1 might display important functions independently of other VEGF receptors. In particular, in the absence of VEGFR-1/2, NRP-1 promotes melanoma invasiveness, through the activation of selected integrins, by stimulating VEGF-A and metalloproteinases secretion and modulating specific signal transduction pathways. This review is focused on the role of NRP-1 in melanoma aggressiveness and on the evidence supporting its use as target of therapies for metastatic melanoma.
Collapse
Affiliation(s)
- Grazia Graziani
- Department of Systems Medicine, University of Rome "Tor Vergata" , Rome , Italy
| | - Pedro M Lacal
- Laboratory of Molecular Oncology, "Istituto Dermopatico dell'Immacolata", Istituto di Ricovero e Cura a Carattere Scientifico , Rome , Italy
| |
Collapse
|
26
|
MiR-148a, a microRNA upregulated in the WNT subgroup tumors, inhibits invasion and tumorigenic potential of medulloblastoma cells by targeting Neuropilin 1. Oncoscience 2015; 2:334-48. [PMID: 26097868 PMCID: PMC4468320 DOI: 10.18632/oncoscience.137] [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: 11/17/2014] [Accepted: 02/27/2015] [Indexed: 01/03/2023] Open
Abstract
Medulloblastoma, a common pediatric malignant brain tumor consists of four molecular subgroups viz. WNT, SHH, Group 3 and Group 4. MiR-148a is over-expressed in the WNT subgroup tumors, which have the lowest incidence of metastasis and excellent survival among all medulloblastomas. MiR-148a was expressed either in a transient manner using a synthetic mimic or in a stable doxycycline inducible manner using a lentiviral vector in non-WNT medulloblastoma cell lines. Expression of miR-148a to levels comparable to that in the WNT subgroup tumors was found to inhibit proliferation, clonogenic potential, invasion potential and tumorigenicity of medulloblastoma cells. MiR-148a expression in medulloblastoma cells brought about reduction in the expression of NRP1, a novel miR-148a target. Restoration of NRP1 expression in medulloblastoma cells was found to rescue the reduction in the invasion potential and tumorigenicity brought about by miR-148a expression. NRP1 is known to play role in multiple signaling pathways that promote tumor growth, invasion and metastasis. NRP1 expression in medulloblastomas was found to be associated with poor survival, with little or no expression in majority of the WNT tumors. The tumor suppressive effect of miR-148a expression accompanied by the down-regulation of NRP1 makes miR-148a an attractive therapeutic agent for the treatment of medulloblastomas.
Collapse
|
27
|
Okon IS, Coughlan KA, Zhang C, Moriasi C, Ding Y, Song P, Zhang W, Li G, Zou MH. Protein kinase LKB1 promotes RAB7-mediated neuropilin-1 degradation to inhibit angiogenesis. J Clin Invest 2014; 124:4590-602. [PMID: 25180605 DOI: 10.1172/jci75371] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 07/25/2014] [Indexed: 12/16/2022] Open
Abstract
After internalization, transmembrane receptors (TMRs) are typically recycled back to the cell surface or targeted for degradation. Efficient TMR trafficking is critical for regulation of several processes, including signal transduction pathways, development, and disease. Here, we determined that trafficking of the angiogenic receptor neuropilin-1 (NRP-1) is abrogated by the liver kinase B1 (LKB1), a serine-threonine kinase of the calcium calmodulin family. We found that aberrant NRP-1 expression in tumor cells from patients with lung adenocarcinoma is associated with decreased levels of LKB1. In cultured lung cells, LKB1 accentuated formation of a complex between NRP-1 and RAB7 in late endosomes. LKB1 specifically bound GTP-bound RAB7, but not a dominant-negative GDP-bound form of RAB7, promoting rapid transfer and lysosome degradation of NRP-1. siRNA-mediated depletion of RAB7 disrupted the transfer of NRP-1 to the lysosome, resulting in recovery of the receptor as well as increased tumor growth and angiogenesis. Together, our findings indicate that LKB1 functions as a RAB7 effector and suppresses angiogenesis by promoting the cellular trafficking of NRP-1 from RAB7 vesicles to the lysosome for degradation. Furthermore, these data suggest that LKB1 and NRP-1 have potential as therapeutic targets for limiting tumorigenesis.
Collapse
|
28
|
Mittal K, Ebos J, Rini B. Angiogenesis and the tumor microenvironment: vascular endothelial growth factor and beyond. Semin Oncol 2014; 41:235-51. [PMID: 24787295 DOI: 10.1053/j.seminoncol.2014.02.007] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Our understanding of the dynamic tumor microenvironment (TME) has improved exponentially over the last few decades. In addition to traditional cytotoxic agents, anti-cancer strategies now include numerous molecular-targeted drugs that modulate distinct elements of the TME. Angiogenesis is an underlying promoter of tumor growth, invasion, and metastases. From traditional and emerging angiogenic cytokines and their receptors to novel immune checkpoint inhibitors, regulation of the tumor microenvironment is potentially key in countering tumor progression. In this article, an overview of the architecture of the TME and the orchestration of angiogenesis within the TME is provided. Additionally, traditional and novel angiogenic targets of current interest within the TME are reviewed.
Collapse
Affiliation(s)
- Kriti Mittal
- Cleveland Clinic Taussig Cancer Institute, Case Comprehensive Cancer Center Cleveland, OH 44195.
| | - John Ebos
- Roswell Park Cancer Institute, Buffalo, NY
| | - Brian Rini
- Cleveland Clinic Taussig Cancer Institute, Case Comprehensive Cancer Center Cleveland, OH 44195
| |
Collapse
|
29
|
The type III TGFβ receptor regulates filopodia formation via a Cdc42-mediated IRSp53-N-WASP interaction in epithelial cells. Biochem J 2013; 454:79-89. [PMID: 23750457 DOI: 10.1042/bj20121701] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Cell adhesion and migration are tightly controlled by regulated changes in the actin cytoskeleton. Previously we reported that the TGFβ (transforming growth factor β) superfamily co-receptor, TβRIII (type III TGFβ receptor; also known as βglycan), regulates cell adhesion, migration and invasion, and suppresses cancer progression, in part, through activation of the small GTPase Cdc42 (cell division cycle 42), and Cdc42-dependent alterations to the actin cytoskeleton. In the present study we demonstrate that TβRIII specifically promotes filopodial formation and extension in MCF10A and HMEC (human mammary epithelial cell) mammary epithelial cells. Mechanistically, cell-surface TβRIII and Cdc42 co-localize to filopodial structures and co-complex in a β-arrestin2-dependent, and a TβRI/TβRII-independent manner. The β-arrestin2-mediated interaction between TβRIII and Cdc42 increases complex formation between the Cdc42 effectors IRSp53 with N-WASP (neuronal Wiskott-Aldrich syndrome protein) to increase filopodial formation. We demonstrate a function link between filopodial structures and epithelial cell adhesion as regulated by the TβRIII-Cdc42 interaction. The present studies identify TβRIII as a novel regulator of IRSp53/N-WASP via Cdc42 to regulate filopodial formation and cell adhesion.
Collapse
|
30
|
Dornbusch J, Zacharis A, Meinhardt M, Erdmann K, Wolff I, Froehner M, Wirth MP, Zastrow S, Fuessel S. Analyses of potential predictive markers and survival data for a response to sunitinib in patients with metastatic renal cell carcinoma. PLoS One 2013; 8:e76386. [PMID: 24086736 PMCID: PMC3785463 DOI: 10.1371/journal.pone.0076386] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 08/30/2013] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Patients with metastatic clear cell renal cell carcinoma (ccRCC) are frequently treated with tyrosine kinase inhibitors (TKI) such as sunitinib. It inhibits angiogenic pathways by mainly targeting the receptors of VEGF and PDGF. In ccRCC, angiogenesis is characterized by the inactivation of the von Hippel-Lindau gene (VHL) which in turn leads to the induction of HIF1α target genes such as CA9 and VEGF. Furthermore, the angiogenic phenotype of ccRCC is also reflected by endothelial markers (CD31, CD34) or other tumor-promoting factors like Ki67 or survivin. METHODS Tissue microarrays from primary tumor specimens of 42 patients with metastatic ccRCC under sunitinib therapy were immunohistochemically stained for selected markers related to angiogenesis. The prognostic and predictive potential of theses markers was assessed on the basis of the objective response rate which was evaluated according to the RECIST criteria after 3, 6, 9 months and after last report (12-54 months) of sunitinib treatment. Additionally, VHL copy number and mutation analyses were performed on DNA from cryo-preserved tumor tissues of 20 ccRCC patients. RESULTS Immunostaining of HIF-1α, CA9, Ki67, CD31, pVEGFR1, VEGFR1 and -2, pPDGFRα and -β was significantly associated with the sunitinib response after 6 and 9 months as well as last report under therapy. Furthermore, HIF-1α, CA9, CD34, VEGFR1 and -3 and PDGRFα showed significant associations with progression-free survival (PFS) and overall survival (OS). In multivariate Cox proportional hazards regression analyses high CA9 membrane staining and a response after 9 months were independent prognostic factors for longer OS. Frequently observed copy number loss and mutation of VHL gene lead to altered expression of VHL, HIF-1α, CA9, and VEGF. CONCLUSIONS Immunoexpression of HIF-1α, CA9, Ki67, CD31, pVEGFR1, VEGFR1 and -2, pPDGFRα and -β in the primary tumors of metastatic ccRCC patients might support the prediction of a good response to sunitinib treatment.
Collapse
Affiliation(s)
- Juana Dornbusch
- Department of Urology, Dresden University of Technology, Dresden, Germany
| | | | - Matthias Meinhardt
- Institute of Pathology, Dresden University of Technology, Dresden, Germany
| | - Kati Erdmann
- Department of Urology, Dresden University of Technology, Dresden, Germany
| | - Ingmar Wolff
- Department of Urology, Dresden University of Technology, Dresden, Germany
| | - Michael Froehner
- Department of Urology, Dresden University of Technology, Dresden, Germany
| | - Manfred P. Wirth
- Department of Urology, Dresden University of Technology, Dresden, Germany
| | - Stefan Zastrow
- Department of Urology, Dresden University of Technology, Dresden, Germany
| | - Susanne Fuessel
- Department of Urology, Dresden University of Technology, Dresden, Germany
| |
Collapse
|
31
|
Zheng X, Chopp M, Lu Y, Buller B, Jiang F. MiR-15b and miR-152 reduce glioma cell invasion and angiogenesis via NRP-2 and MMP-3. Cancer Lett 2012; 329:146-54. [PMID: 23142217 DOI: 10.1016/j.canlet.2012.10.026] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 10/23/2012] [Accepted: 10/26/2012] [Indexed: 12/11/2022]
Abstract
We tested invasion and angiogenesis related mRNA expression and miRNA profiles of glioma. Genes with mRNA expression that changed significantly were selected to predict possible miRNAs that regulate mRNA expression, and were then matched with miRNA results. NRP-2 with the matching miRNA miR-15b, and MMP-3 with the matching miRNA miR-152 were selected for further study. Luciferase activity assay confirmed that miR-15b and miR-152 attenuate expression of NRP-2 and MMP-3 protein by binding to NRP-2 and MMP-3 transcript, respectively. In vitro invasion assay data showed that miR-15b and miR-152 significantly decreased 9L cell invasiveness. In vitro tube formation assay data showed that miR-15b reduced tube formation. A preliminary pathway study indicated that miR-15b and miR-152 deactivated the MEK-ERK pathway via NRP-2 and MMP-3 in 9L cells, respectively.
Collapse
Affiliation(s)
- Xuguang Zheng
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | | | | | | | | |
Collapse
|
32
|
Hota PK, Buck M. Plexin structures are coming: opportunities for multilevel investigations of semaphorin guidance receptors, their cell signaling mechanisms, and functions. Cell Mol Life Sci 2012; 69:3765-805. [PMID: 22744749 PMCID: PMC11115013 DOI: 10.1007/s00018-012-1019-0] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 04/09/2012] [Accepted: 04/11/2012] [Indexed: 01/13/2023]
Abstract
Plexin transmembrane receptors and their semaphorin ligands, as well as their co-receptors (Neuropilin, Integrin, VEGFR2, ErbB2, and Met kinase) are emerging as key regulatory proteins in a wide variety of developmental, regenerative, but also pathological processes. The diverse arenas of plexin function are surveyed, including roles in the nervous, cardiovascular, bone and skeletal, and immune systems. Such different settings require considerable specificity among the plexin and semaphorin family members which in turn are accompanied by a variety of cell signaling networks. Underlying the latter are the mechanistic details of the interactions and catalytic events at the molecular level. Very recently, dramatic progress has been made in solving the structures of plexins and of their complexes with associated proteins. This molecular level information is now suggesting detailed mechanisms for the function of both the extracellular as well as the intracellular plexin regions. Specifically, several groups have solved structures for extracellular domains for plexin-A2, -B1, and -C1, many in complex with semaphorin ligands. On the intracellular side, the role of small Rho GTPases has been of particular interest. These directly associate with plexin and stimulate a GTPase activating (GAP) function in the plexin catalytic domain to downregulate Ras GTPases. Structures for the Rho GTPase binding domains have been presented for several plexins, some with Rnd1 bound. The entire intracellular domain structure of plexin-A1, -A3, and -B1 have also been solved alone and in complex with Rac1. However, key aspects of the interplay between GTPases and plexins remain far from clear. The structural information is helping the plexin field to focus on key questions at the protein structural, cellular, as well as organism level that collaboratoria of investigations are likely to answer.
Collapse
Affiliation(s)
- Prasanta K. Hota
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106 USA
| | - Matthias Buck
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106 USA
- Department of Neuroscience, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106 USA
- Department of Pharmacology, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106 USA
- Comprehensive Cancer Center, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106 USA
- Center for Proteomics and Bioinformatics, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106 USA
| |
Collapse
|
33
|
Partanen TA, Vuola P, Jauhiainen S, Lohi J, Salminen P, Pitkäranta A, Häkkinen SK, Honkonen K, Alitalo K, Ylä-Herttuala S. Neuropilin-2 and vascular endothelial growth factor receptor-3 are up-regulated in human vascular malformations. Angiogenesis 2012; 16:137-46. [PMID: 22961441 DOI: 10.1007/s10456-012-9305-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Accepted: 08/28/2012] [Indexed: 01/21/2023]
Abstract
Despite multiple previous studies in the field of vascular anomalies, the mechanism(s) leading to their development, progression and maintenance has remained unclear. In this study, we have characterized the expression levels of vascular endothelial growth factors and their receptors in 33 human vascular anomalies. Analysis with quantitative real-time PCR and gene-specific assays showed higher expression of neuropilin-2 (NRP2) and VEGF-receptor-3 (VEGFR-3) mRNAs in vascular malformations (VascM) as compared to infantile hemangiomas (Hem). In addition, the expression levels of PlGF and VEGF-C mRNA were significantly higher in venous VascM when compared to the other VascM and Hem. Higher expression of NRP2 and VEGFR-3 were confirmed by immunohistochemistry. To further study the importance of NRP2 and VEGFR-3, endothelial cell (EC) cultures were established from vascular anomalies. It was found that NRP2 and VEGFR-3 mRNA levels were significantly higher in some of the VascM ECs as compared to human umbilical vein ECs which were used as control cells in the study. Furthermore, adenoviral delivery of soluble decoy NRP2 prevented the proliferation of ECs isolated from most of the vascular anomalies. Our findings suggest that NRP2 functions as a factor maintaining the pathological vascular network in these anomalies. Thus, NRP2 could become a potential therapeutic target for the diagnosis and treatment of vascular anomalies.
Collapse
Affiliation(s)
- Taina A Partanen
- Department of Surgery, South Karelia Central Hospital, Lappeenranta, Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Soares AR, Reverendo M, Pereira PM, Nivelles O, Pendeville H, Bezerra AR, Moura GR, Struman I, Santos MAS. Dre-miR-2188 targets Nrp2a and mediates proper intersegmental vessel development in zebrafish embryos. PLoS One 2012; 7:e39417. [PMID: 22761789 PMCID: PMC3382224 DOI: 10.1371/journal.pone.0039417] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 05/24/2012] [Indexed: 12/30/2022] Open
Abstract
Background MicroRNAs (miRNAs) are a class of small RNAs that are implicated in the control of eukaryotic gene expression by binding to the 3′UTR of target mRNAs. Several algorithms have been developed for miRNA target prediction however, experimental validation is still essential for the correct identification of miRNA targets. We have recently predicted that Neuropilin2a (Nrp2a), a vascular endothelial growth factor receptor which is essential for normal developmental angiogenesis in zebrafish, is a dre-miR-2188 target. Methodology Here we show that dre-miR-2188 targets the 3′-untranslated region (3′UTR) of Nrp2a mRNA and is implicated in proper intersegmental vessel development in vivo. Over expression of miR-2188 in zebrafish embryos down regulates Nrp2a expression and results in intersegmental vessel disruption, while its silencing increases Nrp2a expression and intersegmental vessel sprouting. An in vivo GFP sensor assay based on a fusion between the GFP coding region and the Nrp2a 3′UTR confirms that miR-2188 binds to the 3′UTR of Nrp2a and inhibits protein translation. Conclusions We demonstrate that miR-2188 targets Nrp2a and affects intersegmental vessel development in zebrafish embryos.
Collapse
Affiliation(s)
- Ana R. Soares
- RNA Biology Laboratory, Department of Biology & CESAM, University of Aveiro, Aveiro, Portugal
| | - Marisa Reverendo
- RNA Biology Laboratory, Department of Biology & CESAM, University of Aveiro, Aveiro, Portugal
| | - Patrícia M. Pereira
- RNA Biology Laboratory, Department of Biology & CESAM, University of Aveiro, Aveiro, Portugal
| | - Olivier Nivelles
- Unit of Molecular Biology and Genetic Engineering, GIGA-Research, University of Liège, Sart Tilman, Liège, Belgium
| | - Hélène Pendeville
- Unit of Molecular Biology and Genetic Engineering, GIGA-Research, University of Liège, Sart Tilman, Liège, Belgium
| | - Ana Rita Bezerra
- RNA Biology Laboratory, Department of Biology & CESAM, University of Aveiro, Aveiro, Portugal
| | - Gabriela R. Moura
- RNA Biology Laboratory, Department of Biology & CESAM, University of Aveiro, Aveiro, Portugal
| | - Ingrid Struman
- Unit of Molecular Biology and Genetic Engineering, GIGA-Research, University of Liège, Sart Tilman, Liège, Belgium
| | - Manuel A. S. Santos
- RNA Biology Laboratory, Department of Biology & CESAM, University of Aveiro, Aveiro, Portugal
- * E-mail:
| |
Collapse
|
35
|
Wild JRL, Staton CA, Chapple K, Corfe BM. Neuropilins: expression and roles in the epithelium. Int J Exp Pathol 2012; 93:81-103. [PMID: 22414290 DOI: 10.1111/j.1365-2613.2012.00810.x] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Initially found expressed in neuronal and then later in endothelial cells, it is well established that the transmembrane glycoproteins neuropilin-1 (NRP1) and neuropilin-2 (NRP2) play essential roles in axonal growth and guidance and in physiological and pathological angiogenesis. Neuropilin expression and function in epithelial cells has received little attention when compared with neuronal and endothelial cells. Overexpression of NRPs is shown to enhance growth, correlate with invasion and is associated with poor prognosis in various tumour types, especially those of epithelial origin. The contribution of NRP and its ligands to tumour growth and metastasis has spurred a strong interest in NRPs as novel chemotherapy drug targets. Given NRP's role as a multifunctional co-receptor with an ability to bind with disparate ligand families, this has sparked new areas of research implicating NRPs in diverse biological functions. Here, we review the growing body of research demonstrating NRP expression and role in the normal and neoplastic epithelium.
Collapse
Affiliation(s)
- Jonathan R L Wild
- Molecular Gastroenterology Research Group, Academic Unit of Surgical Oncology, Department of Oncology, University of Sheffield, The Medical School, Sheffield, UK
| | | | | | | |
Collapse
|
36
|
Boschetti E, Righetti P. Mixed Beds. ADVANCES IN CHROMATOGRAPHY 2012. [DOI: 10.1201/b11636-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
37
|
Abstract
Each organ and pathology has a unique vascular ZIP code that can be targeted with affinity ligands. In vivo peptide phage display can be used for unbiased mapping of the vascular diversity. Remarkably, some of the peptides identified by such screens not only bind to target vessels but also elicit biological responses. Recently identified tissue-penetrating CendR peptides trigger vascular exit and parenchymal spread of a wide range of conjugated and coadministered payloads. This review is designed to serve as a practical guide for researchers interested in setting up ex vivo and in vivo phage display technology. We focus on T7 coliphage platform that our lab prefers to use due to its versatility, physical resemblance of phage particles to clinical nanoparticles, and ease of manipulation.
Collapse
|
38
|
Younan S, Elhoseiny S, Hammam A, Gawdat R, El-Wakil M, Fawzy M. Role of neuropilin-1 and its expression in Egyptian acute myeloid and acute lymphoid leukemia patients. Leuk Res 2011; 36:169-73. [PMID: 21978468 DOI: 10.1016/j.leukres.2011.08.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Revised: 07/25/2011] [Accepted: 08/18/2011] [Indexed: 12/01/2022]
Abstract
Neuropilins are expressed in tumors vasculature and cells. Their expression is thought to be correlated with tumor angiogenesis and progression. In this study, we analyzed NRP-1 expression level in 40 acute leukemia patients [20 acute myeloid leukemia (AML) and 20 acute lymphoblastic leukemia (ALL)] and 10 healthy controls using Real-Time Quantitative Reverse-Transcriptase Polymerase Chain Reaction (RTQ-PCR) aiming to show Neuropilin-1 (NRP-1) gene expression pattern in acute leukemia patients and its role in disease severity and progression. NRP-1 was expressed in 80% and 95% of ALL and AML respectively with levels higher in patients than controls and in ALL than AML patients. NRP-1 levels were significantly correlated with blast percentage and complete remission. We conclude that NRP-1 is significantly associated with acute leukemia and that its level might serve as an indicator for disease severity and progression. NRP-1 signaling may represent a novel therapeutic approach for the treatment of acute leukemia subsets.
Collapse
Affiliation(s)
- Sarah Younan
- Department of Clinical and Chemical Pathology, Kasr El Aini Hospital, Cairo University, El Saraya Street, Infront of El Manial Palace, Cairo 11451, Egypt.
| | | | | | | | | | | |
Collapse
|
39
|
Chiodelli P, Mitola S, Ravelli C, Oreste P, Rusnati M, Presta M. Heparan sulfate proteoglycans mediate the angiogenic activity of the vascular endothelial growth factor receptor-2 agonist gremlin. Arterioscler Thromb Vasc Biol 2011; 31:e116-27. [PMID: 21921258 DOI: 10.1161/atvbaha.111.235184] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Heparan sulfate proteoglycans (HSPGs) modulate the interaction of proangiogenic heparin-binding vascular endothelial growth factors (VEGFs) with signaling VEGF receptor-2 (VEGFR2) and neuropilin coreceptors in endothelial cells (ECs). The bone morphogenic protein antagonist gremlin is a proangiogenic ligand of VEGFR2, distinct from canonical VEGFs. Here we investigated the role of HSPGs in VEGFR2 interaction, signaling, and proangiogenic capacity of gremlin in ECs. METHODS AND RESULTS Surface plasmon resonance demonstrated that gremlin binds heparin and heparan sulfate, but not other glycosaminoglycans, via N-, 2-O, and 6-O-sulfated groups of the polysaccharide. Accordingly, gremlin binds HSPGs of the EC surface and extracellular matrix. Gremlin/HSPG interaction is prevented by free heparin and heparan sulfate digestion or undersulfation following EC treatment with heparinase II or sodium chlorate. However, at variance with canonical heparin-binding VEGFs, gremlin does not interact with neuropilin-1 coreceptor. On the other hand, HSPGs mediate VEGFR2 engagement and autophosphorylation, extracellular signaling-regulated kinase(1/2) and p38 mitogen-activated protein kinase activation, and consequent proangiogenic responses of ECs to gremlin. On this basis, we evaluated the gremlin-antagonist activity of a panel of chemically sulfated derivatives of the Escherichia coli K5 polysaccharide. The results demonstrate that the highly N,O-sulfated derivative K5-N,OS(H) binds gremlin with high potency, thus inhibiting VEGFR2 interaction and angiogenic activity in vitro and in vivo. CONCLUSIONS HSPGs act as functional gremlin coreceptors in ECs, affecting its productive interaction with VEGFR2 and angiogenic activity. This has allowed the identification of the biotechnological K5-N,OS(H) as a novel angiostatic gremlin antagonist.
Collapse
Affiliation(s)
- Paola Chiodelli
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, Italy
| | | | | | | | | | | |
Collapse
|
40
|
Jauhiainen S, Häkkinen SK, Toivanen PI, Heinonen SE, Jyrkkänen HK, Kansanen E, Leinonen H, Levonen AL, Ylä-Herttuala S. Vascular Endothelial Growth Factor (VEGF)-D Stimulates VEGF-A, Stanniocalcin-1, and Neuropilin-2 and Has Potent Angiogenic Effects. Arterioscler Thromb Vasc Biol 2011; 31:1617-24. [DOI: 10.1161/atvbaha.111.225961] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Objective—
The mature form of human vascular endothelial growth factor-D (hVEGF-D
ΔNΔC
) is an efficient angiogenic factor, but its full mechanism of action has remained unclear. We studied the effects of hVEGF-D
ΔNΔC
in endothelial cells using gene array, signaling, cell culture, and in vivo gene transfer techniques.
Methods and Results—
Concomitant with the angiogenic and proliferative responses, hVEGF-D
ΔNΔC
enhanced the phosphorylation of VEGF receptor-2, Akt, and endothelial nitric oxide synthase. Gene arrays, quantitative reverse transcription–polymerase chain reaction, and Western blot revealed increases in VEGF-A, stanniocalcin-1 (STC1), and neuropilin (NRP) 2 expression by hVEGF-D
ΔNΔC
stimulation, whereas induction with hVEGF-A
165
altered the expression of STC1 and NRP1, another coreceptor for VEGFs. The effects of hVEGF-D
ΔNΔC
were seen only under high-serum conditions, whereas for hVEGF-A
165
, the strongest response was observed under low-serum conditions. The hVEGF-D
ΔNΔC
-induced upregulation of STC1 and NRP2 was also evident in vivo in mouse skeletal muscle treated with hVEGF-D
ΔNΔC
by adenoviral gene delivery. The importance of NRP2 in hVEGF-D
ΔNΔC
signaling was further studied with NRP2 small interfering RNA and NRP antagonist, which were able to block hVEGF-D
ΔNΔC
-induced survival of endothelial cells.
Conclusion—
In this study, the importance of serum and upregulation of NRP2 and STC1 for VEGF-D
ΔNΔC
effects were demonstrated. Better knowledge of VEGF-D
ΔNΔC
signaling and regulation is valuable for the development of efficient and safe VEGF-D
ΔNΔC
-based therapeutic applications for cardiovascular diseases.
Collapse
Affiliation(s)
- Suvi Jauhiainen
- From the Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences (S.J., S.-K.H., P.I.T., S.E.H., H.-K.J., E.K., H.L., A.-L.L., S.Y.-H.) and Department of Medicine (S.Y.-H.), University of Eastern Finland, Kuopio, Finland; Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland (S.Y.-H.)
| | - Sanna-Kaisa Häkkinen
- From the Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences (S.J., S.-K.H., P.I.T., S.E.H., H.-K.J., E.K., H.L., A.-L.L., S.Y.-H.) and Department of Medicine (S.Y.-H.), University of Eastern Finland, Kuopio, Finland; Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland (S.Y.-H.)
| | - Pyry I. Toivanen
- From the Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences (S.J., S.-K.H., P.I.T., S.E.H., H.-K.J., E.K., H.L., A.-L.L., S.Y.-H.) and Department of Medicine (S.Y.-H.), University of Eastern Finland, Kuopio, Finland; Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland (S.Y.-H.)
| | - Suvi E. Heinonen
- From the Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences (S.J., S.-K.H., P.I.T., S.E.H., H.-K.J., E.K., H.L., A.-L.L., S.Y.-H.) and Department of Medicine (S.Y.-H.), University of Eastern Finland, Kuopio, Finland; Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland (S.Y.-H.)
| | - Henna-Kaisa Jyrkkänen
- From the Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences (S.J., S.-K.H., P.I.T., S.E.H., H.-K.J., E.K., H.L., A.-L.L., S.Y.-H.) and Department of Medicine (S.Y.-H.), University of Eastern Finland, Kuopio, Finland; Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland (S.Y.-H.)
| | - Emilia Kansanen
- From the Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences (S.J., S.-K.H., P.I.T., S.E.H., H.-K.J., E.K., H.L., A.-L.L., S.Y.-H.) and Department of Medicine (S.Y.-H.), University of Eastern Finland, Kuopio, Finland; Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland (S.Y.-H.)
| | - Hanna Leinonen
- From the Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences (S.J., S.-K.H., P.I.T., S.E.H., H.-K.J., E.K., H.L., A.-L.L., S.Y.-H.) and Department of Medicine (S.Y.-H.), University of Eastern Finland, Kuopio, Finland; Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland (S.Y.-H.)
| | - Anna-Liisa Levonen
- From the Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences (S.J., S.-K.H., P.I.T., S.E.H., H.-K.J., E.K., H.L., A.-L.L., S.Y.-H.) and Department of Medicine (S.Y.-H.), University of Eastern Finland, Kuopio, Finland; Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland (S.Y.-H.)
| | - Seppo Ylä-Herttuala
- From the Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences (S.J., S.-K.H., P.I.T., S.E.H., H.-K.J., E.K., H.L., A.-L.L., S.Y.-H.) and Department of Medicine (S.Y.-H.), University of Eastern Finland, Kuopio, Finland; Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland (S.Y.-H.)
| |
Collapse
|
41
|
Ischemic neurons prevent vascular regeneration of neural tissue by secreting semaphorin 3A. Blood 2011; 117:6024-35. [PMID: 21355092 DOI: 10.1182/blood-2010-10-311589] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The failure of blood vessels to revascularize ischemic neural tissue represents a significant challenge for vascular biology. Examples include proliferative retinopathies (PRs) such as retinopathy of prematurity and proliferative diabetic retinopathy, which are the leading causes of blindness in children and working-age adults. PRs are characterized by initial microvascular degeneration, followed by a compensatory albeit pathologic hypervascularization mounted by the hypoxic retina attempting to reinstate metabolic equilibrium. Paradoxically, this secondary revascularization fails to grow into the most ischemic regions of the retina. Instead, the new vessels are misdirected toward the vitreous, suggesting that vasorepulsive forces operate in the avascular hypoxic retina. In the present study, we demonstrate that the neuronal guidance cue semaphorin 3A (Sema3A) is secreted by hypoxic neurons in the avascular retina in response to the proinflammatory cytokine IL-1β. Sema3A contributes to vascular decay and later forms a chemical barrier that repels neo-vessels toward the vitreous. Conversely, silencing Sema3A expression enhances normal vascular regeneration within the ischemic retina, thereby diminishing aberrant neovascularization and preserving neuroretinal function. Overcoming the chemical barrier (Sema3A) released by ischemic neurons accelerates the vascular regeneration of neural tissues, which restores metabolic supply and improves retinal function. Our findings may be applicable to other neurovascular ischemic conditions such as stroke.
Collapse
|
42
|
Quantification and cell-to-cell variation of vascular endothelial growth factor receptors. Exp Cell Res 2010; 317:955-65. [PMID: 21185287 DOI: 10.1016/j.yexcr.2010.12.014] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 12/03/2010] [Accepted: 12/15/2010] [Indexed: 01/13/2023]
Abstract
The vascular endothelial growth factor receptors (VEGFR) play a significant role in angiogenesis, the formation of new blood vessels from existing vasculature. Systems biology offers promising approaches to better understand angiogenesis by computational modeling the key molecular interactions in this process. Such modeling requires quantitative knowledge of cell surface density of pro-angiogenic receptors versus anti-angiogenic receptors, their regulation, and their cell-to-cell variability. Using quantitative fluorescence, we systematically characterized the endothelial surface density of VEGFRs and neuropilin-1 (NRP1). We also determined the role of VEGF in regulating the surface density of these receptors. Applying cell-by-cell analysis revealed heterogeneity in receptor surface density and VEGF tuning of this heterogeneity. Altogether, we determine inherent differences in the surface expression levels of these receptors and the role of VEGF in regulating the balance of anti-angiogenic or modulatory (VEGFR1) and pro-angiogenic (VEGFR2) receptors.
Collapse
|
43
|
Uniewicz KA, Cross MJ, Fernig DG. Exogenous recombinant dimeric neuropilin-1 is sufficient to drive angiogenesis. J Biol Chem 2010; 286:12-23. [PMID: 20956519 DOI: 10.1074/jbc.m110.190801] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neuropilin-1 (NRP-1) is present on the cell surface of endothelial cells, or as a soluble truncated variant. Membrane NRP-1 is proposed to enhance angiogenesis by promoting the formation of a signaling complex between vascular endothelial growth factor-A(165) (VEGF-A(165)), VEGF receptor-2 (VEGFR-2) and heparan sulfate, whereas the soluble NRP-1 is thought to act as an antagonist of signaling complex formation. We have analyzed the angiogenic potential of a chimera comprising the entire extracellular NRP-1 region dimerized through an Fc IgG domain and a monomeric truncated NRP-1 variant. Both NRP-1 proteins stimulated tubular morphogenesis and cell migration in HDMECs and HUVECs. Fc rNRP-1 was able to induce VEGFR-2 phosphorylation and expression of the VEGFR-2 specific target, regulator of calcineurin-1 (RCAN1.4). siRNA mediated gene silencing of VEGFR-2 revealed that VEGFR-2 was required for Fc rNRP-1 mediated activation of the intracellular signaling proteins PLC-γ, AKT, and MAPK and tubular morphogenesis. The stimulatory activity was independent of VEGF-A(165). This was evidenced by depleting the cell culture of exogenous VEGF-A(165), and using instead for routine culture VEGF-A(121), which does not interact with NRP-1, and by the inability of VEGF-A sequestering antibodies to inhibit the angiogenic activity of the NRP proteins. Analysis of angiogenesis over a period of 6 days in an in vitro fibroblast/endothelial co-culture model revealed that Fc rNRP-1 could induce endothelial cell tubular morphogenesis. Thus, we conclude that soluble Fc rNRP-1 is a VEGF-A(165)-independent agonist of VEGFR-2 and stimulates angiogenesis in endothelial cells.
Collapse
Affiliation(s)
- Katarzyna A Uniewicz
- Department of Chemical and Structural Biology, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, United Kingdom
| | | | | |
Collapse
|
44
|
Wakamatsu A, Imai JI, Watanabe S, Isogai T. Alternative splicing of genes during neuronal differentiation of NT2 pluripotential human embryonal carcinoma cells. FEBS Lett 2010; 584:4041-7. [PMID: 20728443 DOI: 10.1016/j.febslet.2010.08.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Revised: 08/11/2010] [Accepted: 08/13/2010] [Indexed: 11/19/2022]
Abstract
We analyzed the mRNA diversity of genes after inducing neuronal differentiation in human NT2 teratocarcinoma cells using all-trans retinoic acid (RA). DNA microarray analyses of cells treated with RA identified 358 RA-responsive genes. mRNA diversity analysis revealed that 274 genes produced multiple protein-coding transcripts by alternative splicing. Among these 274 genes, we chose 26 genes that showed AS in their C-terminus and 12 transcription factor genes for further analysis. By using transcript-specific primers, we performed quantitative real-time PCR analysis to examine the expression profiles of all the protein-coding transcripts. Consequently, we identified genes which showed different RA-induced changes in the expression of their protein-coding transcripts.
Collapse
Affiliation(s)
- Ai Wakamatsu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | | | | | | |
Collapse
|
45
|
Geretti E, van Meeteren LA, Shimizu A, Dudley AC, Claesson-Welsh L, Klagsbrun M. A mutated soluble neuropilin-2 B domain antagonizes vascular endothelial growth factor bioactivity and inhibits tumor progression. Mol Cancer Res 2010; 8:1063-73. [PMID: 20651020 DOI: 10.1158/1541-7786.mcr-10-0157] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Neuropilins (NRP1 and NRP2) are coreceptors for vascular endothelial growth factor (VEGF) and mediate angiogenesis and tumor progression. VEGF binds to the NRP1 and NRP2 B domains. Previously, it was shown that mutagenesis of the soluble NRP2 B domain (MutB-NRP2) increased affinity to VEGF by 8-fold. Here, we show that MutB-NRP2 inhibited (125)I-VEGF binding to NRP1, NRP2, and VEGFR-2. It antagonized VEGF-induced VEGFR-2/NRP2 complex formation and inhibited VEGF-induced activation of AKT, a mediator of cell survival, without affecting activation of VEGFR-2. In three-dimensional embryoid bodies, a model of VEGF-induced angiogenesis, MutB-NRP2 inhibited VEGF-induced sprouting. When overexpressed in human melanoma cells, MutB-NRP2 inhibited tumor growth compared with control tumors. Avastin (bevacizumab), a monoclonal antibody to VEGF, inhibited VEGF interactions with VEGFR-2, but not with NRPs. The combination of MutB-NRP2 and Avastin resulted in an enhanced inhibition of human melanoma tumor growth compared with MutB-NRP2 treatment only or Avastin treatment only. In conclusion, these results indicate that MutB-NRP2 is a novel antagonist of VEGF bioactivity and tumor progression.
Collapse
Affiliation(s)
- Elena Geretti
- Department of Surgery, Children's Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | |
Collapse
|
46
|
Probst-Kepper M, Buer J. FOXP3 and GARP (LRRC32): the master and its minion. Biol Direct 2010; 5:8. [PMID: 20137067 PMCID: PMC2825496 DOI: 10.1186/1745-6150-5-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Accepted: 02/05/2010] [Indexed: 11/10/2022] Open
Abstract
The transcription factor FOXP3 is essential for the development and function of CD4+CD25hiFOXP3+ regulatory T (T(reg)) cells, but also expressed in activated human helper T cells without acquisition of a regulatory phenotype. This comment focuses on glycoprotein-A repetitions predominant (GARP or LRRC32) recently identified as specific marker of activated human T(reg) cells, which may provide the missing link toward a better molecular definition of the regulatory phenotype.
Collapse
Affiliation(s)
- Michael Probst-Kepper
- Institute for Microbiology, Immunology and Hygiene, Städtisches Klinikum Braunschweig gGmbH, Braunschweig, Germany
| | - Jan Buer
- Institute for Medical Microbiology, University Hospital Essen, Essen, Germany
| |
Collapse
|
47
|
Mouton-Barbosa E, Roux-Dalvai F, Bouyssié D, Berger F, Schmidt E, Righetti PG, Guerrier L, Boschetti E, Burlet-Schiltz O, Monsarrat B, Gonzalez de Peredo A. In-depth exploration of cerebrospinal fluid by combining peptide ligand library treatment and label-free protein quantification. Mol Cell Proteomics 2010; 9:1006-21. [PMID: 20093276 DOI: 10.1074/mcp.m900513-mcp200] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cerebrospinal fluid (CSF) is the biological fluid in closest contact with the brain and thus contains proteins of neural cell origin. Hence, CSF is a biochemical window into the brain and is particularly attractive for the search for biomarkers of neurological diseases. However, as in the case of other biological fluids, one of the main analytical challenges in proteomic characterization of the CSF is the very wide concentration range of proteins, largely exceeding the dynamic range of current analytical approaches. Here, we used the combinatorial peptide ligand library technology (ProteoMiner) to reduce the dynamic range of protein concentration in CSF and unmask previously undetected proteins by nano-LC-MS/MS analysis on an LTQ-Orbitrap mass spectrometer. This method was first applied on a large pool of CSF from different sources with the aim to better characterize the protein content of this fluid, especially for the low abundance components. We were able to identify 1212 proteins in CSF, and among these, 745 were only detected after peptide library treatment. However, additional difficulties for clinical studies of CSF are the low protein concentration of this fluid and the low volumes typically obtained after lumbar puncture, precluding the conventional use of ProteoMiner with large volume columns for treatment of patient samples. The method has thus been optimized to be compatible with low volume samples. We could show that the treatment is still efficient with this miniaturized protocol and that the dynamic range of protein concentration is actually reduced even with small amounts of beads, leading to an increase of more than 100% of the number of identified proteins in one LC-MS/MS run. Moreover, using a dedicated bioinformatics analytical work flow, we found that the method is reproducible and applicable for label-free quantification of series of samples processed in parallel.
Collapse
Affiliation(s)
- Emmanuelle Mouton-Barbosa
- Institut de Pharmacologie et de Biologie Structurale (IPBS), CNRS, 205 route de Narbonne, 31077 Toulouse, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Guidance molecules were first described in the nervous system to control axon outgrowth direction. They are also widely expressed outside the nervous system where they control cell migration, tissue development and establishment of the vascular network. In addition, they are involved in cancer development, tumor angiogenesis and metastasis. This review is primarily focused on their functions in lung cancer and their involvement in lung development is also presented. Five guidance molecule families and their corresponding receptors are described, including the semaphorins/neuropilins/plexins, ephrins and Eph receptors, netrin/DCC/UNC5, Slit/Robo and Notch/Delta. In addition, the possibility to target these molecules as a therapeutic approach in cancer is discussed.
Collapse
Affiliation(s)
- Patrick Nasarre
- Medical University of South Carolina, Division of Hematology/Oncology, Charleston, SC, USA
| | | | | | | |
Collapse
|
49
|
Geretti E, Shimizu A, Klagsbrun M. Neuropilin structure governs VEGF and semaphorin binding and regulates angiogenesis. Angiogenesis 2008; 11:31-9. [DOI: 10.1007/s10456-008-9097-1] [Citation(s) in RCA: 135] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Accepted: 01/28/2008] [Indexed: 12/22/2022]
|