1
|
Nakayama H, Murakami A, Nishida-Fukuda H, Fukuda S, Matsugi E, Nakahara M, Kusumoto C, Kamei Y, Higashiyama S. Semaphorin 3F inhibits breast cancer metastasis by regulating the Akt-mTOR and TGFβ signaling pathways via neuropilin-2. Sci Rep 2025; 15:7394. [PMID: 40033046 PMCID: PMC11876635 DOI: 10.1038/s41598-025-91559-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/21/2025] [Indexed: 03/05/2025] Open
Abstract
Class 3 semaphorins are axon guidance factors implicated in tumor and vascular biology, including invasive activity. Recent studies indicate that semaphorin 3F (SEMA3F) is a potent inhibitor of metastasis; however, its functional role in breast cancer is not fully understood. We found that exogenous SEMA3F inhibited phosphorylation of Akt and mTOR downstream kinase S6K in MDA-MB-231 and MCF7 cells via neuropilin-2 (NRP2) receptor. We also examined the effect of SEMA3F on breast cancer progression in vivo allograft model. The mouse 4T1 breast cancer cells or 4T1 cells overexpressing SEMA3F (4T1-SEMA3F) were implanted into mammary fat pads of Balb/c mice. We found that tumor growth was significantly inhibited in 4T1-SEMA3F injected mice compared to controls. Immunostaining revealed a remarkable reduction in the expression of vimentin, a mesenchymal cell marker, in 4T1-SEMA3F tumors. We also observed that mice injected with 4T1-SEMA3F cells had minimal metastasis to the liver and lungs, compared to controls. As a novel feature, SEMA3F suppressed TGFβ-induced Smad2 phosphorylation, resulting in the inhibition of cell invasiveness and epithelial-to-mesenchymal transition (EMT) in breast cancer. Consistently, a significant correlation between reduced expression of SEMA3F and poor outcome in patients with breast cancer. We conclude that SEMA3F acts as a dual inhibitor of the Akt-mTOR and TGFβ signaling pathways; thus, it has the potential to treat metastatic breast cancer.
Collapse
Affiliation(s)
- Hironao Nakayama
- Department of Medical Science and Technology, Hiroshima International University, Higashi-hiroshima, 739-2695, Hiroshima, Japan.
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center (PROS), Ehime University, Toon, 791-0295, Ehime, Japan.
| | - Akari Murakami
- Breast Center, Ehime University Hospital, Toon, 791-0295, Ehime, Japan
| | - Hisayo Nishida-Fukuda
- Department of Biochemistry, School of Dentistry, Aichi Gakuin University, Nagoya, 464-8650, Aichi, Japan
| | - Shinji Fukuda
- Department of Biochemistry, School of Dentistry, Aichi Gakuin University, Nagoya, 464-8650, Aichi, Japan
| | - Erina Matsugi
- Department of Medical Science and Technology, Hiroshima International University, Higashi-hiroshima, 739-2695, Hiroshima, Japan
| | - Masako Nakahara
- Department of Medical Science and Technology, Hiroshima International University, Higashi-hiroshima, 739-2695, Hiroshima, Japan
| | - Chiaki Kusumoto
- Department of Medical Science and Technology, Hiroshima International University, Higashi-hiroshima, 739-2695, Hiroshima, Japan
| | - Yoshiaki Kamei
- Breast Center, Ehime University Hospital, Toon, 791-0295, Ehime, Japan
| | - Shigeki Higashiyama
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center (PROS), Ehime University, Toon, 791-0295, Ehime, Japan.
- Department of Oncogenesis and Growth Regulation, Osaka International Cancer Institute, Chuo-ku, Osaka, 541-8567, Japan.
| |
Collapse
|
2
|
Joo SY, Min H, Kim JA, Kim SJ, Jang SH, Lee H, Kim KM, Seong JK, Choi JY, Jung J, Bok J, Gee HY. Biallelic variants of SEMA3F are associated with nonsyndromic hearing loss. Mol Cells 2025; 48:100190. [PMID: 39909336 PMCID: PMC11879669 DOI: 10.1016/j.mocell.2025.100190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 01/22/2025] [Accepted: 01/30/2025] [Indexed: 02/07/2025] Open
Abstract
It is crucial to manage hearing loss and its associated public health impacts. In this study, we aimed to understand the role of Sema3f in the development and maintenance of the auditory system. Inner ear-specific Sema3f knockout mice exhibited hearing loss at 8 weeks with an elevated threshold for auditory brainstem response and an absent threshold for distortion product optoacoustic emission tests. Additionally, an increased number of outer hair cells and abnormal patterns of spiral ganglion neuron projections in the outer hair cell regions were observed. Through the analyses of sequencing data from 558 families with hearing loss, we identified biallelic variants of SEMA3F, which encodes semaphorin-3F, in one of the families. In the family, the proband showed profound progressive nonsyndromic hearing loss with congenital onset. In vitro analysis revealed that the identified missense variants decreased the furin-mediated processing of SEMA3F and abolished the cellular abilities of SEMA3F, which collapsed the filamentous actin cytoskeleton in human umbilical vein-derived endothelial cells. Our data suggest that SEMA3F is essential for normal hearing and is associated with nonsyndromic hearing loss in humans.
Collapse
Affiliation(s)
- Sun Young Joo
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Won Sang Institute for Hearing Loss, Seoul 03722, Republic of Korea
| | - Hyehyun Min
- Department of Anatomy, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jung Ah Kim
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Won Sang Institute for Hearing Loss, Seoul 03722, Republic of Korea
| | - Se Jin Kim
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Won Sang Institute for Hearing Loss, Seoul 03722, Republic of Korea
| | - Seung Hyun Jang
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Won Sang Institute for Hearing Loss, Seoul 03722, Republic of Korea
| | - Ho Lee
- Graduate School of Cancer Science and Policy, National Cancer Center, Goyang-si 10408, Republic of Korea
| | - Kyu Min Kim
- Won Sang Institute for Hearing Loss, Seoul 03722, Republic of Korea; Laboratory of Developmental Biology and Genomics, BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Jae Young Choi
- Won Sang Institute for Hearing Loss, Seoul 03722, Republic of Korea; Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jinsei Jung
- Won Sang Institute for Hearing Loss, Seoul 03722, Republic of Korea; Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| | - Jinwoong Bok
- Won Sang Institute for Hearing Loss, Seoul 03722, Republic of Korea; Department of Anatomy, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| | - Heon Yung Gee
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Won Sang Institute for Hearing Loss, Seoul 03722, Republic of Korea; Woo Choo Lee Institute for Precision Drug Development, Seoul 03722, Republic of Korea.
| |
Collapse
|
3
|
Kawai Y, Nagayama A, Miyao K, Takeuchi M, Yokoe T, Kameyama T, Wang X, Seki T, Takahashi M, Hayashida T, Kitagawa Y. A genome-wide CRISPR/Cas9 knockout screen identifies SEMA3F gene for resistance to cyclin-dependent kinase 4 and 6 inhibitors in breast cancer. Breast Cancer 2025; 32:120-131. [PMID: 39352623 DOI: 10.1007/s12282-024-01641-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/26/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Palbociclib is a cell-cycle targeted small molecule agent used as one of the standards of care in combination with endocrine therapy for patients with hormone receptor (HR)-positive, human epidermal growth factor receptor 2 (HER2)-negative advanced breast cancer. Although several gene alterations such as loss of Rb gene and amplification of p16 gene are known to be conventional resistance mechanisms to cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitors, the comprehensive landscape of resistance is not yet fully elucidated. The purpose of this study is to identify the novel resistant genes to the CDK4/6 inhibitors in HR-positive HER2-negative breast cancer. METHODS The whole genome knockout screen using CRISPR/Cas9 genome editing was conducted in MCF7 to identify resistant genes to palbociclib. The candidate genes for resistance were selected by NGS analysis and GSEA analysis and validated by cell viability assay and mouse xenograft models. RESULTS We identified eight genes including RET, TIRAP, GNRH1, SEMA3F, SEMA5A, GATA4, NOD1, SSTR1 as candidate genes from the whole genome knockout screen. Among those, knockdown of SEMA3F by siRNA significantly and consistently increased the cell viability in the presence of CDK4/6 inhibitors in vitro and in vivo. Furthermore, the level of p-Rb was maintained in the palbociclib treated SEMA3F-downregulated cells, indicating that the resistance is driven by increased activity of cyclin kinases. CONCLUSION Our observation provided the first evidence of SEMA3F as a regulator of sensitivity to CDK4/6 inhibitors in breast cancer. The detailed mechanisms of resistance deserve further functional studies to develop the better strategy to overcome resistance in CDK4/6 inhibitors.
Collapse
Affiliation(s)
- Yuko Kawai
- Department of Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Aiko Nagayama
- Department of Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan.
| | - Kazuhiro Miyao
- Department of Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Makoto Takeuchi
- Department of Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Takamichi Yokoe
- Department of Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Tomoe Kameyama
- Department of Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Xinyue Wang
- Department of Surgery, Keio University Graduate School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Tomoko Seki
- Department of Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Maiko Takahashi
- Department of Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Tetsu Hayashida
- Department of Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| |
Collapse
|
4
|
Lee J, Shin DY, Jang Y, Han JP, Cho EM, Seo YR. Cadmium-induced Carcinogenesis in Respiratory Organs and the Prostate: Insights from Three Perspectives on Toxicogenomic Approach. J Cancer Prev 2023; 28:150-159. [PMID: 38205367 PMCID: PMC10774485 DOI: 10.15430/jcp.2023.28.4.150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Cadmium (Cd) exposure primarily occurs through inhalation, either by smoking or occupational exposure to contaminated air. Upon inhalation, Cd ultimately reaches the prostate through the bloodstream. In this review, we investigate the carcinogenic potential of Cd in both respiratory organs and the prostate. Specifically, this review examines cellular metabolism, comprehensive toxicity, and carcinogenic mechanisms by exploring gene ontology, biological networks, and adverse outcome pathways. In the respiratory organs, Cd induces lung cancer by altering the expression of IL1B and FGF2, causing DNA damage, reducing cell junction integrity, and promoting apoptosis. In the prostate, Cd induces prostate cancer by modifying the expression of EDN1 and HMOX1, leading to abnormal protein activities and maturation, suppressing tumor suppressors, and inducing apoptosis. Collectively, this review provides a comprehensive understanding of the carcinogenic mechanisms of Cd in two different organs by adopting toxicogenomic approaches. These insights can serve as a foundation for further research on cadmium-induced cancer, contributing to the establishment of future cancer prevention strategies.
Collapse
Affiliation(s)
- Jun Lee
- Department of Life Science, Institute of Environmental Medicine for Green Chemistry, Dongguk University Biomedi Campus, Goyang, Korea
| | - Dong Yeop Shin
- Department of Life Science, Institute of Environmental Medicine for Green Chemistry, Dongguk University Biomedi Campus, Goyang, Korea
| | - Yujin Jang
- Department of Life Science, Institute of Environmental Medicine for Green Chemistry, Dongguk University Biomedi Campus, Goyang, Korea
| | - Jun Pyo Han
- Department of Life Science, Institute of Environmental Medicine for Green Chemistry, Dongguk University Biomedi Campus, Goyang, Korea
| | - Eun-Min Cho
- Department of Nano, Chemical & Biological Engineering, College of Natural Science and Engineering, Seokyeong University, Seoul, Korea
| | - Young Rok Seo
- Department of Life Science, Institute of Environmental Medicine for Green Chemistry, Dongguk University Biomedi Campus, Goyang, Korea
| |
Collapse
|
5
|
Bergsten E, Mestivier D, Donnadieu F, Pedron T, Barau C, Meda LT, Mettouchi A, Lemichez E, Gorgette O, Chamaillard M, Vaysse A, Volant S, Doukani A, Sansonetti PJ, Sobhani I, Nigro G. Parvimonas micra, an oral pathobiont associated with colorectal cancer, epigenetically reprograms human colonocytes. Gut Microbes 2023; 15:2265138. [PMID: 37842920 PMCID: PMC10580862 DOI: 10.1080/19490976.2023.2265138] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/20/2023] [Indexed: 10/17/2023] Open
Abstract
Recently, an intestinal dysbiotic microbiota with enrichment in oral cavity bacteria has been described in colorectal cancer (CRC) patients. Here, we characterize and investigate one of these oral pathobionts, the Gram-positive anaerobic coccus Parvimonas micra. We identified two phylotypes (A and B) exhibiting different phenotypes and adhesion capabilities. We observed a strong association of phylotype A with CRC, with its higher abundance in feces and in tumoral tissue compared with the normal homologous colonic mucosa, which was associated with a distinct methylation status of patients. By developing an in vitro hypoxic co-culture system of human primary colonic cells with anaerobic bacteria, we show that P. micra phylotype A alters the DNA methylation profile promoters of key tumor-suppressor genes, oncogenes, and genes involved in epithelial-mesenchymal transition. In colonic mucosa of CRC patients carrying P. micra phylotype A, we found similar DNA methylation alterations, together with significant enrichment of differentially expressed genes in pathways involved in inflammation, cell adhesion, and regulation of actin cytoskeleton, providing evidence of P. micra's possible role in the carcinogenic process.
Collapse
Affiliation(s)
- Emma Bergsten
- Unité de Pathogénie Microbienne Moléculaire, INSERM U1202, Institut Pasteur, Paris, France
- Équipe universitaire EC2M3-EA7375, Université Paris- Est (UPEC), Créteil, France
| | - Denis Mestivier
- Équipe universitaire EC2M3-EA7375, Université Paris- Est (UPEC), Créteil, France
- Plateforme de Bio-informatique, Institut Mondor de Recherche Biomédicale (IMRB/INSERM U955), Université Paris-Est, Créteil, France
| | - Francoise Donnadieu
- Unité de Pathogénie Microbienne Moléculaire, INSERM U1202, Institut Pasteur, Paris, France
| | - Thierry Pedron
- Unité de Pathogénie Microbienne Moléculaire, INSERM U1202, Institut Pasteur, Paris, France
- Unité Bactériophage, Bactérie, Hôte, Institut Pasteur, Paris, France
| | - Caroline Barau
- Plateforme de Ressources Biologiques, CHU Henri Mondor Assistance Publique Hôpitaux de Paris (APHP), Créteil, France
| | - Landry Tsoumtsa Meda
- Unité des Toxines Bactériennes, Université Paris Cité, CNRS UMR6047, INSERM U1306, Institut Pasteur, Paris, France
| | - Amel Mettouchi
- Unité des Toxines Bactériennes, Université Paris Cité, CNRS UMR6047, INSERM U1306, Institut Pasteur, Paris, France
| | - Emmanuel Lemichez
- Unité des Toxines Bactériennes, Université Paris Cité, CNRS UMR6047, INSERM U1306, Institut Pasteur, Paris, France
| | - Olivier Gorgette
- Plateforme de Bio-Imagerie Ultrastructurale, Institut Pasteur, Université Paris Cité, Paris, France
| | - Mathias Chamaillard
- Laboratory of Cell Physiology, INSERM U1003, University of Lille, Lille, France
| | - Amaury Vaysse
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, Paris, France
| | - Stevenn Volant
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, Paris, France
| | - Abiba Doukani
- Sorbonne Université, Inserm, Unité Mixte de Service Production et Analyse de données en Sciences de la Vie et en Santé, Paris, France
| | - Philippe J Sansonetti
- Unité de Pathogénie Microbienne Moléculaire, INSERM U1202, Institut Pasteur, Paris, France
- Chaire de Microbiologie et Maladies Infectieuses, Collège de France, Paris, France
| | - Iradj Sobhani
- Équipe universitaire EC2M3-EA7375, Université Paris- Est (UPEC), Créteil, France
- Service de Gastroentérologie, CHU Henri Mondor Assistance Publique Hôpitaux de Paris (APHP), Créteil, France
| | - Giulia Nigro
- Unité de Pathogénie Microbienne Moléculaire, INSERM U1202, Institut Pasteur, Paris, France
- Microenvironment and Immunity Unit, INSERM U1224, Institut Pasteur, Paris, France
| |
Collapse
|
6
|
Pienkowski T, Kowalczyk T, Cysewski D, Kretowski A, Ciborowski M. Glioma and post-translational modifications: A complex relationship. Biochim Biophys Acta Rev Cancer 2023; 1878:189009. [PMID: 37913943 DOI: 10.1016/j.bbcan.2023.189009] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 10/17/2023] [Accepted: 10/17/2023] [Indexed: 11/03/2023]
Abstract
Post-translational modifications (PTMs) are common covalent processes in biochemical pathways that alter protein function and activity. These modifications occur through proteolytic cleavage or attachment of modifying groups, such as phosphoryl, methyl, glycosyl, or acetyl groups, with one or more amino acid residues of a single protein. Some PTMs also present crosstalk abilities that affect both protein functionality and structure, creating new proteoforms. Any alteration in organism homeostasis may be a cancer hallmark. Cataloging PTMs and consequently, emerging proteoforms, present new therapeutic targets, approaches, and opportunities to discover additional discriminatory biomarkers in disease diagnostics. In this review, we focus on experimentally confirmed PTMs and their potential crosstalk in glioma research to introduce new opportunities for this tumor type, which emerge within the PTMomics area.
Collapse
Affiliation(s)
- Tomasz Pienkowski
- Clinical Research Centre, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Tomasz Kowalczyk
- Clinical Research Centre, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland; Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland
| | - Dominik Cysewski
- Clinical Research Centre, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Adam Kretowski
- Clinical Research Centre, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland; Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Michal Ciborowski
- Clinical Research Centre, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland.
| |
Collapse
|
7
|
Dai L, Shen KF, Zhang CQ. Plexin-mediated neuronal development and neuroinflammatory responses in the nervous system. Histol Histopathol 2023; 38:1239-1248. [PMID: 37170703 DOI: 10.14670/hh-18-625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Plexins are a large family of single-pass transmembrane proteins that mediate semaphorin signaling in multiple systems. Plexins were originally characterized for their role modulating cytoskeletal activity to regulate axon guidance during nervous system development. Thereafter, different semaphorin-plexin complexes were identified in the nervous system that have diverse functions in neurons, astrocytes, glia, oligodendrocytes, and brain derived-tumor cells, providing unexpected but meaningful insights into the biological activities of this protein family. Here, we review the overall structure and relevant downstream signaling cascades of plexins. We consider the current knowledge regarding the function of semaphorin-plexin cascades in the nervous system, including the most recent data regarding their roles in neuronal development, neuroinflammation, and glioma.
Collapse
Affiliation(s)
- Lu Dai
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, China
| | - Kai-Feng Shen
- Department of Neurosurgery, Epilepsy Research Center of PLA, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Chun-Qing Zhang
- Department of Neurosurgery, Epilepsy Research Center of PLA, Xinqiao Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
8
|
Abstract
The endothelium is a dynamic, semipermeable layer lining all blood vessels, regulating blood vessel formation and barrier function. Proper composition and function of the endothelial barrier are required for fluid homeostasis, and clinical conditions characterized by barrier disruption are associated with severe morbidity and high mortality rates. Endothelial barrier properties are regulated by cell-cell junctions and intracellular signaling pathways governing the cytoskeleton, but recent insights indicate an increasingly important role for integrin-mediated cell-matrix adhesion and signaling in endothelial barrier regulation. Here, we discuss diseases characterized by endothelial barrier disruption, and provide an overview of the composition of endothelial cell-matrix adhesion complexes and associated signaling pathways, their crosstalk with cell-cell junctions, and with other receptors. We further present recent insights into the role of cell-matrix adhesions in the developing and mature/adult endothelium of various vascular beds, and discuss how the dynamic regulation and turnover of cell-matrix adhesions regulates endothelial barrier function in (patho)physiological conditions like angiogenesis, inflammation and in response to hemodynamic stress. Finally, as clinical conditions associated with vascular leak still lack direct treatment, we focus on how understanding of endothelial cell-matrix adhesion may provide novel targets for treatment, and discuss current translational challenges and future perspectives.
Collapse
Affiliation(s)
- Jurjan Aman
- Department of Pulmonology, Amsterdam University Medical Center, the Netherlands (J.A.)
| | - Coert Margadant
- Department of Medical Oncology, Amsterdam University Medical Center, the NetherlandsInstitute of Biology, Leiden University, the Netherlands (C.M.)
| |
Collapse
|
9
|
Lambrinos G, Cristofaro V, Pelton K, Bigger-Allen A, Doyle C, Vasquez E, Bielenberg DR, Sullivan MP, Adam RM. Neuropilin 2 Is a Novel Regulator of Distal Colon Contractility. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1592-1603. [PMID: 35985479 PMCID: PMC9667714 DOI: 10.1016/j.ajpath.2022.07.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 06/20/2022] [Accepted: 07/25/2022] [Indexed: 06/05/2023]
Abstract
Appropriate coordination of smooth muscle contraction and relaxation is essential for normal colonic motility. The impact of perturbed motility ranges from moderate, in conditions such as colitis, to potentially fatal in the case of pseudo-obstruction. The mechanisms underlying aberrant motility and the extent to which they can be targeted pharmacologically are incompletely understood. This study identified colonic smooth muscle as a major site of expression of neuropilin 2 (Nrp2) in mice and humans. Mice with inducible smooth muscle-specific knockout of Nrp2 had an increase in evoked contraction of colonic rings in response to carbachol at 1 and 4 weeks following initiation of deletion. KCl-induced contractions were also increased at 4 weeks. Colonic motility was similarly enhanced, as evidenced by faster bead expulsion in Nrp2-deleted mice versus Nrp2-intact controls. In length-tension analysis of the distal colon, passive tension was similar in Nrp2-deficient and Nrp2-intact mice, but at low strains, active stiffness was greater in Nrp2-deficient animals. Consistent with the findings in conditional Nrp2 mice, Nrp2-null mice showed increased contractility in response to carbachol and KCl. Evaluation of selected proteins implicated in smooth muscle contraction revealed no significant differences in the level of α-smooth muscle actin, myosin light chain, calponin, or RhoA. Together, these findings identify Nrp2 as a novel regulator of colonic contractility that may be targetable in conditions characterized by dysmotility.
Collapse
Affiliation(s)
- George Lambrinos
- Urological Diseases Research Center, Boston Children's Hospital, Boston, Massachusetts
| | - Vivian Cristofaro
- Department of Surgery, Harvard Medical School, Boston, Massachusetts; Division of Urology, VA Boston Healthcare System, Boston, Massachusetts
| | - Kristine Pelton
- Urological Diseases Research Center, Boston Children's Hospital, Boston, Massachusetts
| | - Alexander Bigger-Allen
- Urological Diseases Research Center, Boston Children's Hospital, Boston, Massachusetts; Biological and Biomedical Sciences Program, Division of Medical Sciences, Harvard Medical School, Boston, Massachusetts
| | - Claire Doyle
- Urological Diseases Research Center, Boston Children's Hospital, Boston, Massachusetts
| | - Evalynn Vasquez
- Urological Diseases Research Center, Boston Children's Hospital, Boston, Massachusetts
| | - Diane R Bielenberg
- Department of Surgery, Harvard Medical School, Boston, Massachusetts; Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts
| | - Maryrose P Sullivan
- Department of Surgery, Harvard Medical School, Boston, Massachusetts; Division of Urology, VA Boston Healthcare System, Boston, Massachusetts.
| | - Rosalyn M Adam
- Urological Diseases Research Center, Boston Children's Hospital, Boston, Massachusetts; Department of Surgery, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
10
|
Circular RNA circPGD contributes to gastric cancer progression via the sponging miR-16-5p/ABL2 axis and encodes a novel PGD-219aa protein. Cell Death Dis 2022; 8:384. [PMID: 36104322 PMCID: PMC9472197 DOI: 10.1038/s41420-022-01177-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/26/2022]
Abstract
CircRNAs have critical effects on tumor development and progression. However, circPGD effect on gastric cancer (GC) is still elusive. Nuclear and cytoplasmic RNA fractionation, and RNA-FISH assay examined the localization of circPGD in MGC-803 cells. qRT-PCR was conducted to detect the expression and prognostic significance of circPGD, miR-16-5p, and ABL2 within GC tissues. Meanwhile, qRT-PCR, luciferase reporter assays, rescue, and western blotting assays confirmed the interactions between circPGD, miR-16-5p, and ABL2. Transwell, wound healing, and colony-formation assays, as well as CCK-8 and cell apoptosis assays, analyzed the functions of circPGD, miR-16-5p, ABL2, as well as PGD-219aa within GC cells. Western blotting and cell immunofluorescence experiments detected the differences in the expression of the related proteins. Finally, xenograft and metastatic mouse models were used to investigate circPGD function in vivo. Mass spectrometry was used to detect the existence of PGD-219aa in MGC-803 cells. CircPGD was localized in the cytoplasm and nucleus of MGC-803 cells. Compared with the control, circPGD and ABL2 expression increased within GC tissues and cells, and the miR-16-5p level was decreased. Functionally, circPGD promoted cell proliferation, migration and suppressed apoptosis in vitro. Mechanistically, circPGD sponged miR-16-5p for relieving miR-16-5p suppression on the corresponding target ABL2 via the SMAD2/3 and YAP signaling pathways. In addition, circPGD encodes a novel PGD-219aa protein that can enhance the growth and migration of GC cells, while inhibiting GC cells apoptosis via the SMAD2/3 and YAP signaling pathways. Furthermore, circPGD overexpression enhanced tumor aggressiveness, while circPGD knockdown inhibited tumor growth. Overall, circPGD has a novel oncogenic effect on GC cells, indicating the potential of circPGD as the tumorigenic factor and a promising diagnostic marker for GC.
Collapse
|
11
|
β-Arrestin2 Is Critically Involved in the Differential Regulation of Phosphosignaling Pathways by Thyrotropin-Releasing Hormone and Taltirelin. Cells 2022; 11:cells11091473. [PMID: 35563779 PMCID: PMC9103620 DOI: 10.3390/cells11091473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 12/17/2022] Open
Abstract
In recent years, thyrotropin-releasing hormone (TRH) and its analogs, including taltirelin (TAL), have demonstrated a range of effects on the central nervous system that represent potential therapeutic agents for the treatment of various neurological disorders, including neurodegenerative diseases. However, the molecular mechanisms of their actions remain poorly understood. In this study, we investigated phosphosignaling dynamics in pituitary GH1 cells affected by TRH and TAL and the putative role of β-arrestin2 in mediating these effects. Our results revealed widespread alterations in many phosphosignaling pathways involving signal transduction via small GTPases, MAP kinases, Ser/Thr- and Tyr-protein kinases, Wnt/β-catenin, and members of the Hippo pathway. The differential TRH- or TAL-induced phosphorylation of numerous proteins suggests that these ligands exhibit some degree of biased agonism at the TRH receptor. The different phosphorylation patterns induced by TRH or TAL in β-arrestin2-deficient cells suggest that the β-arrestin2 scaffold is a key factor determining phosphorylation events after TRH receptor activation. Our results suggest that compounds that modulate kinase and phosphatase activity can be considered as additional adjuvants to enhance the potential therapeutic value of TRH or TAL.
Collapse
|
12
|
Luo S, Wang F, Chen S, Chen A, Wang Z, Gao X, Kong X, Zuo G, Zhou W, Gu Y, Ge Z, Zhang J. NRP2 promotes atherosclerosis by upregulating PARP1 expression and enhancing low shear stress-induced endothelial cell apoptosis. FASEB J 2022; 36:e22079. [PMID: 35028975 DOI: 10.1096/fj.202101250rr] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/16/2021] [Accepted: 11/18/2021] [Indexed: 01/13/2023]
Abstract
Atherosclerosis-related cardiovascular diseases are leading causes of mortality worldwide, characterized by the development of endothelial cell dysfunction, increased oxidized low-density lipoprotein uptake by macrophages, and the ensuing formation of atherosclerotic plaque. Local blood flow patterns cause uneven atherosclerotic lesion distribution, and endothelial dysfunction caused by disturbed flow is an early step in the development of atherosclerosis. The present research aims to elucidate the mechanism underlying the regulation of Neuropilin 2 (NRP2) under low shear stress (LSS) in the atheroprone phenotype of endothelial cells. We observed that NRP2 expression was significantly upregulated in LSS-stimulated human umbilical vein endothelial cells (HUVECs) and in mouse aortic endothelial cells. Knockdown of NRP2 in HUVECs significantly ameliorated cell apoptosis induced by LSS. Conversely, overexpression of NRP2 had the opposite effect on HUVEC apoptosis. Animal experiments suggest that NRP2 knockdown markedly mitigated the development of atherosclerosis in Apoe-/- mice. Mechanistically, NRP2 knockdown and overexpression regulated PARP1 protein expression in the condition of LSS, which in turn affected the expression of apoptosis-related genes. Moreover, the upstream transcription factor GATA2 was found to regulate NRP2 expression in the progression of atherosclerosis. These findings suggest that NRP2 plays an essential proatherosclerotic role through the regulation of cell apoptosis, and the results reveal that NRP2 is a promising therapeutic target for the treatment of atherosclerotic disorders.
Collapse
Affiliation(s)
- Shuai Luo
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Feng Wang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Siyu Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Aiqun Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Zhimei Wang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiaofei Gao
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Cardiology, Nanjing Heart Centre, Nanjing, China
| | - Xiangquan Kong
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Guangfeng Zuo
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Wenying Zhou
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yue Gu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Zhen Ge
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Junjie Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Cardiology, Nanjing Heart Centre, Nanjing, China
| |
Collapse
|
13
|
Watterston C, Halabi R, McFarlane S, Childs SJ. Endothelial Semaphorin 3fb regulates Vegf pathway-mediated angiogenic sprouting. PLoS Genet 2021; 17:e1009769. [PMID: 34424892 PMCID: PMC8412281 DOI: 10.1371/journal.pgen.1009769] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 09/02/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022] Open
Abstract
Vessel growth integrates diverse extrinsic signals with intrinsic signaling cascades to coordinate cell migration and sprouting morphogenesis. The pro-angiogenic effects of Vascular Endothelial Growth Factor (VEGF) are carefully controlled during sprouting to generate an efficiently patterned vascular network. We identify crosstalk between VEGF signaling and that of the secreted ligand Semaphorin 3fb (Sema3fb), one of two zebrafish paralogs of mammalian Sema3F. The sema3fb gene is expressed by endothelial cells in actively sprouting vessels. Loss of sema3fb results in abnormally wide and stunted intersegmental vessel artery sprouts. Although the sprouts initiate at the correct developmental time, they have a reduced migration speed. These sprouts have persistent filopodia and abnormally spaced nuclei suggesting dysregulated control of actin assembly. sema3fb mutants show simultaneously higher expression of pro-angiogenic (VEGF receptor 2 (vegfr2) and delta-like 4 (dll4)) and anti-angiogenic (soluble VEGF receptor 1 (svegfr1)/ soluble Fms Related Receptor Tyrosine Kinase 1 (sflt1)) pathway components. We show increased phospho-ERK staining in migrating angioblasts, consistent with enhanced Vegf activity. Reducing Vegfr2 kinase activity in sema3fb mutants rescues angiogenic sprouting. Our data suggest that Sema3fb plays a critical role in promoting endothelial sprouting through modulating the VEGF signaling pathway, acting as an autocrine cue that modulates intrinsic growth factor signaling.
Collapse
Affiliation(s)
- Charlene Watterston
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Rami Halabi
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Sarah McFarlane
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Canada
| | - Sarah J. Childs
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
| |
Collapse
|
14
|
Leung CCT, Wong CKC. Effects of stanniocalcin-1 overexpressing hepatocellular carcinoma cells on macrophage migration. PLoS One 2020; 15:e0241932. [PMID: 33156861 PMCID: PMC7647456 DOI: 10.1371/journal.pone.0241932] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/22/2020] [Indexed: 01/24/2023] Open
Abstract
Human stanniocalcin-1 (STC1) is a glycoprotein known to participate in inflammation and tumor progression. However, its role in cancer-macrophage interaction at the tumor environment is not known. In this study, the co-culture of the human metastatic hepatocellular carcinoma cell line (MHCC97L) stably transfected with a control vector (MHCC97L/P), or STC1-overexpressing vector (MHCC97L/S1) with human leukemia monocytic cell line (THP-1) was conducted. We reported that MHCC97L/S1 suppressed the migratory activity of THP-1. Real-time PCR analysis revealed the downregulation of the pro-migratory factors, monocyte-chemoattractant protein receptors, CCR2 and CCR4, and macrophage-migratory cytokine receptor, CSF-1R. Transcriptomic analysis of the THP-1 cells co-cultured with either MHCC97L/P or MHCC97L/S1, detected 1784 differentially expressed genes. The Ingenuity Canonical Pathway analysis predicted that RhoA signaling was associated with the inhibition of the cell migration. Western blot analysis revealed a significant reduction of Ser19-phosphorylation on MLC2, a Rho-A downstream target, in the THP-1 cells. Xenograft tumors derived from MHCC97/S1 in mice showed a remarkable decrease in infiltrating macrophages. Collectively, this is the first report to demonstrate the inhibitory effect of STC1-overexpressing cancer cells on macrophage migration/infiltration. Our data support further investigations on the relationship between tumor STC1 level and macrophage infiltration.
Collapse
Affiliation(s)
- Cherry C. T. Leung
- Croucher Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University, Hong Kong SAR, China
| | - Chris K. C. Wong
- Croucher Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University, Hong Kong SAR, China
- * E-mail:
| |
Collapse
|
15
|
The role of semaphorins in small vessels of the eye and brain. Pharmacol Res 2020; 160:105044. [PMID: 32590102 DOI: 10.1016/j.phrs.2020.105044] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/19/2020] [Accepted: 06/19/2020] [Indexed: 12/20/2022]
Abstract
Small vessel diseases, such as ischemic retinopathy and cerebral small vessel disease (CSVD), are increasingly recognized in patients with diabetes, dementia and cerebrovascular disease. The mechanisms of small vessel diseases are poorly understood, but the latest studies suggest a role for semaphorins. Initially identified as axon guidance cues, semaphorins are mainly studied in neuronal morphogenesis, neural circuit assembly, and synapse assembly and refinement. In recent years, semaphorins have been found to play important roles in regulating vascular growth and development and in many pathophysiological processes, including atherosclerosis, angiogenesis after stroke and retinopathy. Growing evidence indicates that semaphorins affect the occurrence, perfusion and regression of both the macrovasculature and microvasculature by regulating the proliferation, apoptosis, migration, barrier function and inflammatory response of endothelial cells, vascular smooth muscle cells (VSMCs) and pericytes. In this review, we concentrate on the regulatory effects of semaphorins on the cell components of the vessel wall and their potential roles in microvascular diseases, especially in the retina and cerebral small vessel. Finally, we discuss potential molecular approaches in targeting semaphorins as therapies for microvascular disorders in the eye and brain.
Collapse
|
16
|
Plant T, Eamsamarng S, Sanchez-Garcia MA, Reyes L, Renshaw SA, Coelho P, Mirchandani AS, Morgan JM, Ellett FE, Morrison T, Humphries D, Watts ER, Murphy F, Raffo-Iraolagoitia XL, Zhang A, Cash JL, Loynes C, Elks PM, Van Eeden F, Carlin LM, Furley AJ, Whyte MK, Walmsley SR. Semaphorin 3F signaling actively retains neutrophils at sites of inflammation. J Clin Invest 2020; 130:3221-3237. [PMID: 32191647 PMCID: PMC7259996 DOI: 10.1172/jci130834] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 03/11/2020] [Indexed: 12/18/2022] Open
Abstract
Neutrophilic inflammation is central to disease pathogenesis, for example, in chronic obstructive pulmonary disease, yet the mechanisms that retain neutrophils within tissues remain poorly understood. With emerging evidence that axon guidance factors can regulate myeloid recruitment and that neutrophils can regulate expression of a class 3 semaphorin, SEMA3F, we investigated the role of SEMA3F in inflammatory cell retention within inflamed tissues. We observed that neutrophils upregulate SEMA3F in response to proinflammatory mediators and following neutrophil recruitment to the inflamed lung. In both zebrafish tail injury and murine acute lung injury models of neutrophilic inflammation, overexpression of SEMA3F delayed inflammation resolution with slower neutrophil migratory speeds and retention of neutrophils within the tissues. Conversely, constitutive loss of sema3f accelerated egress of neutrophils from the tail injury site in fish, whereas neutrophil-specific deletion of Sema3f in mice resulted in more rapid neutrophil transit through the airways, and significantly reduced time to resolution of the neutrophilic response. Study of filamentous-actin (F-actin) subsequently showed that SEMA3F-mediated retention is associated with F-actin disassembly. In conclusion, SEMA3F signaling actively regulates neutrophil retention within the injured tissues with consequences for neutrophil clearance and inflammation resolution.
Collapse
Affiliation(s)
- Tracie Plant
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Suttida Eamsamarng
- Department of Infection, Immunity and Cardiovascular Disease and
- Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Manuel A. Sanchez-Garcia
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Leila Reyes
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Stephen A. Renshaw
- Department of Infection, Immunity and Cardiovascular Disease and
- Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Patricia Coelho
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Ananda S. Mirchandani
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Jessie-May Morgan
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Felix E. Ellett
- Department of Infection, Immunity and Cardiovascular Disease and
- Bateson Centre, University of Sheffield, Sheffield, United Kingdom
- BioMEMS Resource Centre, Division of Surgery, Innovation and Bioengineering, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Burn Care, Shriners Hospitals for Children — Boston, Boston, Massachusetts, USA
| | - Tyler Morrison
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Duncan Humphries
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Emily R. Watts
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Fiona Murphy
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Ailiang Zhang
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Jenna L. Cash
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Catherine Loynes
- Department of Infection, Immunity and Cardiovascular Disease and
- Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Philip M. Elks
- Department of Infection, Immunity and Cardiovascular Disease and
- Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Freek Van Eeden
- Department of Infection, Immunity and Cardiovascular Disease and
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Leo M. Carlin
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Andrew J.W. Furley
- Bateson Centre, University of Sheffield, Sheffield, United Kingdom
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Moira K.B. Whyte
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease and
| | - Sarah R. Walmsley
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease and
| |
Collapse
|
17
|
The Role of Rho GTPases in VEGF Signaling in Cancer Cells. Anal Cell Pathol (Amst) 2020; 2020:2097214. [PMID: 32377503 PMCID: PMC7182966 DOI: 10.1155/2020/2097214] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 12/11/2022] Open
Abstract
Vascular endothelial growth factors (VEGFs) consist of five molecules (VEGFA through D as well as placental growth factor) which are crucial for regulating key cellular and tissue functions. The role of VEGF and its intracellular signaling and downstream molecular pathways have been thoroughly studied. Activation of VEGF signal transduction can be initiated by the molecules' binding to two classes of transmembrane receptors: (1) the VEGF tyrosine kinase receptors (VEGF receptors 1 through 3) and (2) the neuropilins (NRP1 and 2). The involvement of Rho GTPases in modulating VEGFA signaling in both cancer cells and endothelial cells has also been well established. Additionally, different isoforms of Rho GTPases, namely, RhoA, RhoC, and RhoG, have been shown to regulate VEGF expression as well as blood vessel formation. This review article will explore how Rho GTPases modulate VEGF signaling and the consequences of such interaction on cancer progression.
Collapse
|
18
|
Zhang H, Vreeken D, Junaid A, Wang G, Sol WMPJ, de Bruin RG, van Zonneveld AJ, van Gils JM. Endothelial Semaphorin 3F Maintains Endothelial Barrier Function and Inhibits Monocyte Migration. Int J Mol Sci 2020; 21:ijms21041471. [PMID: 32098168 PMCID: PMC7073048 DOI: 10.3390/ijms21041471] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 12/16/2022] Open
Abstract
In normal physiology, endothelial cells (ECs) form a vital barrier between the blood and underlying tissue controlling leukocyte diapedesis and vascular inflammation. Emerging data suggest that neuronal guidance cues, typically expressed during development, have roles outside the nervous system in vascular biology and immune responses. In particular, Class III semaphorins have been reported to affect EC migration and angiogenesis. While ECs express high levels of semaphorin 3F (SEMA3F), little is known about its function in mature ECs. Here we show that SEMA3F expression is reduced by inflammatory stimuli and increased by laminar flow. Endothelial cells exposed to laminar flow secrete SEMA3F, which subsequently binds to heparan sulfates on the surface of ECs. However, under pro-inflammatory conditions, reduced levels of SEMA3F make ECs more prone to monocyte diapedesis and display impaired barrier function as measured with an electric cell-substrate impedance sensing system and a microfluidic system. In addition, we demonstrate that SEMA3F can directly inhibit the migration of activated monocytes. Taken together, our data suggest an important homeostatic function for EC-expressed SEMA3F, serving as a mediator of endothelial quiescence.
Collapse
|
19
|
The Role of Rho GTPases in Motility and Invasion of Glioblastoma Cells. Anal Cell Pathol (Amst) 2020; 2020:9274016. [PMID: 32089990 PMCID: PMC7013281 DOI: 10.1155/2020/9274016] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 01/14/2020] [Accepted: 01/22/2020] [Indexed: 12/27/2022] Open
Abstract
Astrocytomas are primary malignant brain tumors that originate from astrocytes. Grade IV astrocytoma or glioblastoma is a highly invasive tumor that occur within the brain parenchyma. The Rho family of small GTPases, which includes Rac1, Cdc42, and RhoA, is an important family whose members are key regulators of the invasion and migration of glioblastoma cells. In this review, we describe the role played by the Rho family of GTPases in the regulation of the invasion and migration of glioblastoma cells. Specifically, we focus on the role played by RhoA, Rac1, RhoG, and Cdc42 in cell migration through rearrangement of actin cytoskeleton, cell adhesion, and invasion. Finally, we highlight the importance of potentially targeting Rho GTPases in the treatment of glioblastoma.
Collapse
|
20
|
Anosmin-1 activates vascular endothelial growth factor receptor and its related signaling pathway for olfactory bulb angiogenesis. Sci Rep 2020; 10:188. [PMID: 31932617 PMCID: PMC6957483 DOI: 10.1038/s41598-019-57040-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 12/20/2019] [Indexed: 12/11/2022] Open
Abstract
Anosmin-1 is a secreted glycoprotein encoded by the ANOS1 gene, and its loss of function causes Kallmann syndrome (KS), which is characterized by anosmia and hypogonadism due to olfactory bulb (OB) dysfunction. However, the physiological function of anosmin-1 remains to be elucidated. In KS, disordered angiogenesis is observed in OB, resulting in its hypoplasia. In this study, we examined the involvement of anosmin-1 in angiogenic processes. Anosmin-1 was detected on the vessel-like structure in OB of chick embryos, and promoted the outgrowth of vascular sprouts as shown by assays of OB tissue culture. Cell migration, proliferation, and tube formation of endothelial cells were induced by treatment with anosmin-1 as well as vascular endothelial growth factor-A (VEGF-A), and further enhanced by treatment with both of them. We newly identified that anosmin-1 activated VEGF receptor-2 (VEGFR2) by binding directly to it, and its downstream signaling molecules, phospholipase Cγ1 (PLCγ1) and protein kinase C (PKC). These results suggest that anosmin-1 plays a key role in the angiogenesis of developing OB through the VEGFR2–PLCγ1–PKC axis by enhancing the VEGF function.
Collapse
|
21
|
Dziobek K, Opławski M, Grabarek B, Zmarzły N, Kiełbasiński R, Leśniak E, Januszyk P, Januszyk K, Adwent I, Dąbruś D, Kieszkowski P, Kiełbasiński K, Kuś-Kierach A, Boroń D. Changes in Expression Pattern of SEMA3F Depending on Endometrial Cancer Grade - Pilot Study. Curr Pharm Biotechnol 2020; 20:727-732. [PMID: 31215376 PMCID: PMC7046987 DOI: 10.2174/1389201020666190619145655] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 04/24/2019] [Accepted: 05/08/2019] [Indexed: 01/18/2023]
Abstract
Background: In the course of neoplastic diseases, a reduction in SEMA3F expression is observed, which translates into an increase in the proliferative and proangiogenic potential of cells forming the tumor and the surrounding microenvironment. Objective: The aim of this study was to determine the changes in SEMA3F level in endometrial cancer depending on its grade. Methods: The study material consisted of tissue samples: 15 without neoplastic changes (control group) and 45 with endometrial cancer (G1, 17; G2, 15; G3, 13; study group). SEMA3F expression was assessed using the immune-histochemical method. Results: The expression of SEMA3F was observed in the control group (Me = 159.38) and in the study group (G1, Me = 121.32; G2, Me = 0; G3, Me = 130.37). Differences between each grade and control and between individual grades were statistically significant. There were no significant correlations between SEMA3F expression and weight and Body Mass Index (BMI). The reduced SEMA3F expression in tumor tissue compared to healthy tissue indicates that this protein plays key roles in proliferation and angiogenesis. Conclusion: We found that depending on the severity of the disease, cancer adopts different survival strategies, where SEMA3F plays an important role. As a molecular marker, SEMA3F is not sensitive to weight and BMI.
Collapse
Affiliation(s)
- Konrad Dziobek
- Center of Oncology, M. Sklodowska-Curie Memorial Institute, Cracow Branch, Cracow, Poland
| | - Marcin Opławski
- Department of Gynecology and Obstetrics with Gynecologic Oncology, Ludwik Rydygier Memorial Specialized Hospital, Krakow, Poland
| | - Beniamin Grabarek
- Department of Molecular Biology, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia in Katowice, Katowice, Poland
| | - Nikola Zmarzły
- Department of Molecular Biology, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia in Katowice, Katowice, Poland
| | - Robert Kiełbasiński
- Department of Obstetrics and Gynaecology ward, Health Center in Mikolow, Mikolow, Poland
| | - Ewa Leśniak
- Department of Obstetrics and Gynaecology ward, Health Center in Mikolow, Mikolow, Poland
| | - Piotr Januszyk
- Faculty of Health Science, Public Higher Medical Professional School in Opole, Opole, Poland
| | - Krzysztof Januszyk
- Faculty of Health Science, Public Higher Medical Professional School in Opole, Opole, Poland
| | - Iwona Adwent
- Faculty of Health Science, Public Higher Medical Professional School in Opole, Opole, Poland
| | - Dariusz Dąbruś
- Faculty of Health Science, Public Higher Medical Professional School in Opole, Opole, Poland
| | | | | | - Agnieszka Kuś-Kierach
- Faculty of Health Science, Public Higher Medical Professional School in Opole, Opole, Poland
| | - Dariusz Boroń
- Center of Oncology, M. Sklodowska-Curie Memorial Institute, Cracow Branch, Cracow, Poland.,Department of Gynecology and Obstetrics with Gynecologic Oncology, Ludwik Rydygier Memorial Specialized Hospital, Krakow, Poland.,Faculty of Health Science, Public Higher Medical Professional School in Opole, Opole, Poland.,Department of Histology and Cell Pathology, School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia in Katowice, Katowice, Poland.,Katowice School of Technology, The University of Science and Art in Katowice, Katowice, Poland
| |
Collapse
|
22
|
Woda CB, Bruneau S, Mak AL, Haskova Z, Liu K, Ghosh CC, Briscoe DM. Calcineurin inhibitors augment endothelial-to-mesenchymal transition by enhancing proliferation in association with cytokine-mediated activation. Biochem Biophys Res Commun 2019; 519:667-673. [PMID: 31542230 PMCID: PMC7119266 DOI: 10.1016/j.bbrc.2019.09.043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 09/11/2019] [Indexed: 12/21/2022]
Abstract
Calcineurin Inhibitors (CNIs) are routinely used for immunosuppression following solid organ transplantation. However, the prolonged use of these agents lead to organ fibrosis which limits their efficacy. CNIs induce TGFβ expression, which is reported to augment endothelial-to-mesenchymal transition (EndMT), but their role in this process is not known. In these studies, we find that the CNIs FK506 and cyclosporine (CsA) are potent to increase endothelial cell (EC) proliferation using established in vitro assays (P < 0.05). Furthermore, using phosphokinase arrays, we find that each CNI activates the MAPK and Akt/mTOR signaling pathways, and that pharmacological inhibition of each pathway targets CNI-induced proliferative responses (P < 0.001). EndMT was evaluated by FACS for N-cadherin and CD31 expression and by qPCR for the expression of α-smooth muscle actin, N-cadherin and Snail. We find that CNIs do not directly induce dedifferentiation, while TGFβ and hypoxia induce EndMT in small numbers of EC. In contrast, the treatment of EC with the inflammatory cytokine TNFα was potent to elicit an EndMT response, and its effects were most notably in EC following proliferation/doubling. Taken together, these observations suggest that CNIs elicit proliferative responses, which enhance EndMT in association with local inflammation. The clinical implications of these findings are that anti-proliferative therapeutics have high potential to target the initiation of this EndMT response.
Collapse
Affiliation(s)
- Craig B Woda
- The Transplant Research Program and the Division of Nephrology, Boston Children's Hospital, Boston, MA, 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Sarah Bruneau
- The Transplant Research Program and the Division of Nephrology, Boston Children's Hospital, Boston, MA, 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Anne Linde Mak
- The Transplant Research Program and the Division of Nephrology, Boston Children's Hospital, Boston, MA, 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Zdenka Haskova
- The Transplant Research Program and the Division of Nephrology, Boston Children's Hospital, Boston, MA, 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Kaifeng Liu
- The Transplant Research Program and the Division of Nephrology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Chandra C Ghosh
- The Transplant Research Program and the Division of Nephrology, Boston Children's Hospital, Boston, MA, 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - David M Briscoe
- The Transplant Research Program and the Division of Nephrology, Boston Children's Hospital, Boston, MA, 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
23
|
Zelina P, Pasterkamp RJ. Axons Navigate Noise with 190RhoGAP. Neuron 2019; 102:512-514. [PMID: 31071282 DOI: 10.1016/j.neuron.2019.04.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
In this issue of Neuron, Bonanomi et al. (2019) investigate how navigating axons ignore irrelevant guidance signals. They show that the binding of p190RhoGAP to DCC suppresses inappropriate responses to Netrin-1, allowing motor axons to exit the embryonic spinal cord.
Collapse
Affiliation(s)
- Pavol Zelina
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht 3584 CG, the Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht 3584 CG, the Netherlands.
| |
Collapse
|
24
|
Tan G. Inhibitory effects of Semaphorin 3F as an alternative candidate to anti-VEGF monoclonal antibody on angiogenesis. In Vitro Cell Dev Biol Anim 2019; 55:756-765. [DOI: 10.1007/s11626-019-00392-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 07/29/2019] [Indexed: 12/31/2022]
|
25
|
Multifaceted Functional Role of Semaphorins in Glioblastoma. Int J Mol Sci 2019; 20:ijms20092144. [PMID: 31052281 PMCID: PMC6539029 DOI: 10.3390/ijms20092144] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/19/2019] [Accepted: 04/24/2019] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma (GBM) is the most malignant tumor type affecting the adult central nervous system. Despite advances in therapy, the prognosis for patients with GBM remains poor, with a median survival of about 15 months. To date, few treatment options are available and recent trials based on the molecular targeting of some of the GBM hallmark pathways (e.g., angiogenesis) have not produced any significant improvement in overall survival. The urgent need to develop more efficacious targeted therapies has led to a better molecular characterization of GBM, revealing an emerging role of semaphorins in GBM progression. Semphorins are a wide group of membrane-bound and secreted proteins, originally identified as axon guidance cues, signaling through their receptors, neuropilins, and plexins. A number of semaphorin signals involved in the control of axonal growth and navigation during development have been found to furthermore participate in crosstalk with different dysfunctional GBM pathways, controlling tumor cell proliferation, migration, and invasion, as well as tumor angiogenesis or immune response. In this review, we summarize the regulatory activities mediated by semaphorins and their receptors on the oncogenic pathways implicated in GBM growth and invasive/metastatic progression.
Collapse
|
26
|
Héraud C, Pinault M, Lagrée V, Moreau V. p190RhoGAPs, the ARHGAP35- and ARHGAP5-Encoded Proteins, in Health and Disease. Cells 2019; 8:cells8040351. [PMID: 31013840 PMCID: PMC6523970 DOI: 10.3390/cells8040351] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/05/2019] [Accepted: 04/09/2019] [Indexed: 12/30/2022] Open
Abstract
Small guanosine triphosphatases (GTPases) gathered in the Rat sarcoma (Ras) superfamily represent a large family of proteins involved in several key cellular mechanisms. Within the Ras superfamily, the Ras homolog (Rho) family is specialized in the regulation of actin cytoskeleton-based mechanisms. These proteins switch between an active and an inactive state, resulting in subsequent inhibiting or activating downstream signals, leading finally to regulation of actin-based processes. The On/Off status of Rho GTPases implicates two subsets of regulators: GEFs (guanine nucleotide exchange factors), which favor the active GTP (guanosine triphosphate) status of the GTPase and GAPs (GTPase activating proteins), which inhibit the GTPase by enhancing the GTP hydrolysis. In humans, the 20 identified Rho GTPases are regulated by over 70 GAP proteins suggesting a complex, but well-defined, spatio-temporal implication of these GAPs. Among the quite large number of RhoGAPs, we focus on p190RhoGAP, which is known as the main negative regulator of RhoA, but not exclusively. Two isoforms, p190A and p190B, are encoded by ARHGAP35 and ARHGAP5 genes, respectively. We describe here the function of each of these isoforms in physiological processes and sum up findings on their role in pathological conditions such as neurological disorders and cancers.
Collapse
Affiliation(s)
- Capucine Héraud
- INSERM, UMR1053 Bordeaux Research In Translational Oncology, BaRITOn, F-33000 Bordeaux, France.
- University of Bordeaux, UMR1053 Bordeaux Research In Translational Oncology, BaRITOn, Bordeaux F-33000, France.
- Equipe Labellisée Fondation pour la Recherche Médicale (FRM) 2018, 75007 Paris, France.
| | - Mathilde Pinault
- INSERM, UMR1053 Bordeaux Research In Translational Oncology, BaRITOn, F-33000 Bordeaux, France.
- University of Bordeaux, UMR1053 Bordeaux Research In Translational Oncology, BaRITOn, Bordeaux F-33000, France.
- Equipe Labellisée Fondation pour la Recherche Médicale (FRM) 2018, 75007 Paris, France.
| | - Valérie Lagrée
- INSERM, UMR1053 Bordeaux Research In Translational Oncology, BaRITOn, F-33000 Bordeaux, France.
- University of Bordeaux, UMR1053 Bordeaux Research In Translational Oncology, BaRITOn, Bordeaux F-33000, France.
- Equipe Labellisée Fondation pour la Recherche Médicale (FRM) 2018, 75007 Paris, France.
| | - Violaine Moreau
- INSERM, UMR1053 Bordeaux Research In Translational Oncology, BaRITOn, F-33000 Bordeaux, France.
- University of Bordeaux, UMR1053 Bordeaux Research In Translational Oncology, BaRITOn, Bordeaux F-33000, France.
- Equipe Labellisée Fondation pour la Recherche Médicale (FRM) 2018, 75007 Paris, France.
| |
Collapse
|
27
|
Nakayama H, Kusumoto C, Nakahara M, Fujiwara A, Higashiyama S. Semaphorin 3F and Netrin-1: The Novel Function as a Regulator of Tumor Microenvironment. Front Physiol 2018; 9:1662. [PMID: 30532711 PMCID: PMC6265511 DOI: 10.3389/fphys.2018.01662] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 11/05/2018] [Indexed: 01/13/2023] Open
Abstract
Axon guidance molecules play an important role in regulating proper neuronal networking during neuronal development. They also have non-neuronal properties, which include angiogenesis, inflammation, and tumor development. Semaphorin 3F (SEMA3F), a member of the class 3 semaphorins, was initially identified as an axon guidance factor, that repels axons and collapses growth cones. However, SEMA3F has similar effects on endothelial cells (ECs) and tumor cells. In this review, we discuss the novel molecular mechanisms underlying SEMA3F activity in vascular and tumor biology. Recent evidence suggests that SEMA3F functions as a PI3K-Akt-mTOR inhibitor in mammalian cells, including T cells, ECs, and tumor cells. Therefore, SEMA3F may have broad therapeutic implications. We also discuss the key role of axon guidance molecules as regulators of the tumor microenvironment. Netrin-1, a chemoattractant factor in the neuronal system, promotes tumor progression by enhancing angiogenesis and metastasis. Moreover, our recent studies demonstrate that netrin-1/neogenin interactions augment CD4+ T cell chemokinesis and elicit pro-inflammatory responses, suggesting that netrin-1 plays a key role in modulating the function of a tumor and its surrounding cells in the tumor microenvironment. Overall, this review focuses on SEMA3F and netrin-1 signaling mechanisms to understand the diverse biological functions of axon guidance molecules.
Collapse
Affiliation(s)
- Hironao Nakayama
- Department of Medical Science and Technology, Hiroshima International University, Higashihiroshima, Japan.,Division of Cell Growth and Tumor Regulation, Proteo-Science Center, Ehime University, Toon, Japan
| | - Chiaki Kusumoto
- Department of Medical Science and Technology, Hiroshima International University, Higashihiroshima, Japan
| | - Masako Nakahara
- Department of Medical Science and Technology, Hiroshima International University, Higashihiroshima, Japan
| | - Akira Fujiwara
- Department of Medical Science and Technology, Hiroshima International University, Higashihiroshima, Japan
| | - Shigeki Higashiyama
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center, Ehime University, Toon, Japan
| |
Collapse
|
28
|
Nakayama H, Higashiyama S. [Novel function of axon guidance molecule as a regulator of tumor microenvironment]. Nihon Yakurigaku Zasshi 2018; 150:286-292. [PMID: 29225291 DOI: 10.1254/fpj.150.286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
29
|
Shimizu A, Zankov DP, Kurokawa-Seo M, Ogita H. Vascular Endothelial Growth Factor-A Exerts Diverse Cellular Effects via Small G Proteins, Rho and Rap. Int J Mol Sci 2018; 19:ijms19041203. [PMID: 29659486 PMCID: PMC5979568 DOI: 10.3390/ijms19041203] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 04/03/2018] [Accepted: 04/12/2018] [Indexed: 12/18/2022] Open
Abstract
Vascular endothelial growth factors (VEGFs) include five molecules (VEGF-A, -B, -C, -D, and placental growth factor), and have various roles that crucially regulate cellular functions in many kinds of cells and tissues. Intracellular signal transduction induced by VEGFs has been extensively studied and is usually initiated by their binding to two classes of transmembrane receptors: receptor tyrosine kinase VEGF receptors (VEGF receptor-1, -2 and -3) and neuropilins (NRP1 and NRP2). In addition to many established results reported by other research groups, we have previously identified small G proteins, especially Ras homologue gene (Rho) and Ras-related protein (Rap), as important mediators of VEGF-A-stimulated signaling in cancer cells as well as endothelial cells. This review article describes the VEGF-A-induced signaling pathways underlying diverse cellular functions, including cell proliferation, migration, and angiogenesis, and the involvement of Rho, Rap, and their related molecules in these pathways.
Collapse
Affiliation(s)
- Akio Shimizu
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan.
| | - Dimitar P Zankov
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan.
| | - Misuzu Kurokawa-Seo
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan.
| | - Hisakazu Ogita
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan.
| |
Collapse
|
30
|
Nakayama H, Sakaue T, Maekawa M, Fujisaki A, Higashiyama S. Cullin 3 regulates ADAMs-mediated ectodomain shedding of amphiregulin. Biochem Biophys Res Commun 2018; 499:17-23. [DOI: 10.1016/j.bbrc.2018.03.097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 03/13/2018] [Indexed: 01/01/2023]
|
31
|
Meng PP, Li Z, Wang SY, Zhou WW, Samiullah M, Chen N, Luo FH, Wu T, Yan JH. Preparation, Purification, and Identification of a Monoclonal Antibody Against the C-Terminal Domain of Semaphorin3F. Monoclon Antib Immunodiagn Immunother 2018; 37:52-58. [PMID: 29420136 DOI: 10.1089/mab.2017.0040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Class three semaphorins were originally identified as mediators of axon guidance, which repelled axons and collapsed growth cones. As a member of class three semaphorins, semaphorin3F (Sema3F) has been found to have similar effects on tumor cells and endothelial cells and also is implicated in the signaling of tumor metastasis by forming a complex with neuropilins and plexins. In this study, our laboratory produced a monoclonal antibody against the C-terminal domain of Sema3F (Sema3Fc mAb) using the hybridoma method, expecting to explore the potential role of the antibody and its application in the detection of Sema3F. The capture enzyme-linked immunosorbent assay (ELISA) method indicated that mAb belonged to the IgM subclass and purified Sema3Fc mAb had a titer of 5.12 × 105 against Sema3Fc by indirect ELISA. In addition, results showed that the Sema3Fc mAb could be applied in such experiments as Western blotting, flow cytometry, immunofluorescence, and immunocytochemical staining. It indicates the Sema3Fc mAb is available in the detection of Sema3F with specificity and will help further study the role and mechanism of Sema3F among tumor cells.
Collapse
Affiliation(s)
- Pei-Pei Meng
- Cancer Research Center, Medical College, Xiamen University , Xiangan South Road, Xiamen, Fujian, China
| | - Zhe Li
- Cancer Research Center, Medical College, Xiamen University , Xiangan South Road, Xiamen, Fujian, China
| | - Sheng-Yu Wang
- Cancer Research Center, Medical College, Xiamen University , Xiangan South Road, Xiamen, Fujian, China
| | - Wen-Wen Zhou
- Cancer Research Center, Medical College, Xiamen University , Xiangan South Road, Xiamen, Fujian, China
| | - Malik Samiullah
- Cancer Research Center, Medical College, Xiamen University , Xiangan South Road, Xiamen, Fujian, China
| | - Na Chen
- Cancer Research Center, Medical College, Xiamen University , Xiangan South Road, Xiamen, Fujian, China
| | - Fang-Hong Luo
- Cancer Research Center, Medical College, Xiamen University , Xiangan South Road, Xiamen, Fujian, China
| | - Ting Wu
- Cancer Research Center, Medical College, Xiamen University , Xiangan South Road, Xiamen, Fujian, China
| | - Jiang-Hua Yan
- Cancer Research Center, Medical College, Xiamen University , Xiangan South Road, Xiamen, Fujian, China
| |
Collapse
|
32
|
Angelopoulou E, Piperi C. Emerging role of plexins signaling in glioma progression and therapy. Cancer Lett 2018; 414:81-87. [DOI: 10.1016/j.canlet.2017.11.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/07/2017] [Accepted: 11/08/2017] [Indexed: 12/14/2022]
|
33
|
Liu Y, Li R, Yin K, Ren G, Zhang Y. The crucial role of SEMA3F in suppressing the progression of oral squamous cell carcinoma. Cell Mol Biol Lett 2017; 22:32. [PMID: 29299034 PMCID: PMC5745788 DOI: 10.1186/s11658-017-0064-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 12/15/2017] [Indexed: 01/19/2023] Open
Abstract
Background Oral squamous cell carcinoma (OSCC) is one of the most common types of malignancy. Semaphorin 3F (SEMA3F) is highly conserved but present at a lower level in various cancers than in healthy tissues. While it has been reported that SEMA3F is involved in cancer cell proliferation, migration and invasion, its function in OSCC remains unknown. Methods The expression of SEMA3F in OSCC tissues and OSCC-derived cells was analyzed using qRT-PCR and western blotting. Using SAS and HSC2 cells, we also monitored the effect of SEMA3F on OSCC cell proliferation, migration and invasion using MTT, colony formation and transwell assays. The function of SEMA3F in OSCC tumor formation was also assessed in vivo. Results SEMA3F was significantly downregulated in OSCC tissues and OSCC-derived cells. SEMA3F shows growth inhibitory activity in SAS and HSC2 cells and may act as a tumor suppressor. It can inhibit the migration and invasion potential of OSCC cells. Our results also demonstrate that SEMA3F can suppress the growth of OSCC cells in vivo. Conclusions This study revealed that SEMA3F plays a role as a tumor suppressor in OSCC cell proliferation, migration and invasion. Our finding provides new insight into the progression of OSCC. Therapeutically, SEMA3F has some potential as a target for OSCC treatment, given sufficient future research.
Collapse
Affiliation(s)
- Yi Liu
- Department of Stomatology, Tianjin First Center Hospital, Tianjin, 300192 People's Republic of China
| | - Ronghua Li
- Department of Stomatology, Tianjin First Center Hospital, Tianjin, 300192 People's Republic of China
| | - Kai Yin
- Department of Stomatology, Tianjin First Center Hospital, Tianjin, 300192 People's Republic of China
| | - Gang Ren
- Department of Stomatology, Tianjin First Center Hospital, Tianjin, 300192 People's Republic of China
| | - Yongdong Zhang
- Department of Stomatology, Tianjin First Center Hospital, Tianjin, 300192 People's Republic of China
| |
Collapse
|
34
|
Organoids as preclinical models to improve intraperitoneal chemotherapy effectiveness for colorectal cancer patients with peritoneal metastases: Preclinical models to improve HIPEC. Int J Pharm 2017; 531:143-152. [DOI: 10.1016/j.ijpharm.2017.07.084] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 07/31/2017] [Indexed: 02/06/2023]
|
35
|
Yoon J, Kim SB, Ahmed G, Shay JW, Terman JR. Amplification of F-Actin Disassembly and Cellular Repulsion by Growth Factor Signaling. Dev Cell 2017; 42:117-129.e8. [PMID: 28689759 PMCID: PMC5564210 DOI: 10.1016/j.devcel.2017.06.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 04/10/2017] [Accepted: 06/06/2017] [Indexed: 01/09/2023]
Abstract
Extracellular cues that regulate cellular shape, motility, and navigation are generally classified as growth promoting (i.e., growth factors/chemoattractants and attractive guidance cues) or growth preventing (i.e., repellents and inhibitors). Yet, these designations are often based on complex assays and undefined signaling pathways and thus may misrepresent direct roles of specific cues. Here, we find that a recognized growth-promoting signaling pathway amplifies the F-actin disassembly and repulsive effects of a growth-preventing pathway. Focusing on Semaphorin/Plexin repulsion, we identified an interaction between the F-actin-disassembly enzyme Mical and the Abl tyrosine kinase. Biochemical assays revealed Abl phosphorylates Mical to directly amplify Mical Redox-mediated F-actin disassembly. Genetic assays revealed that Abl allows growth factors and Semaphorin/Plexin repellents to combinatorially increase Mical-mediated F-actin disassembly, cellular remodeling, and repulsive axon guidance. Similar roles for Mical in growth factor/Abl-related cancer cell behaviors further revealed contexts in which characterized positive effectors of growth/guidance stimulate such negative cellular effects as F-actin disassembly/repulsion.
Collapse
Affiliation(s)
- Jimok Yoon
- Departments of Neuroscience and Pharmacology, Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sang Bum Kim
- Department of Cell Biology, Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Giasuddin Ahmed
- Departments of Neuroscience and Pharmacology, Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jerry W Shay
- Department of Cell Biology, Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jonathan R Terman
- Departments of Neuroscience and Pharmacology, Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
36
|
Yokota A, Hirai H, Shoji T, Maekawa T, Okuda K. Constitutively active ABL family kinases, TEL/ABL and TEL/ARG, harbor distinct leukemogenic activities in vivo. Leukemia 2017; 31:2742-2751. [PMID: 28386107 DOI: 10.1038/leu.2017.114] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 03/27/2017] [Indexed: 01/10/2023]
Abstract
ABL (ABL1) and ARG (ABL2) are highly homologous to each other in overall domain structure and amino-acid sequence, with the exception of their C termini. As with ABL, translocations that fuse ARG to ETV6/TEL have been identified in patients with leukemia. To assess the in vivo leukemogenic activity of constitutively active ABL and ARG, we generated a bone marrow (BM) transplantation model using the chimeric forms TEL/ABL and TEL/ARG, which have comparable kinase activities. TEL/ABL rapidly induced fatal myeloid leukemia in recipient mice, whereas recipients of TEL/ARG-transduced cells did not develop myeloid leukemia, instead, they succumbed to a long-latency infiltrative mastocytosis that could be adoptively transferred to secondary recipients. Swapping of the C termini of ABL and ARG altered disease latency and phenotypes. In a detailed in vitro study, TEL/ARG strongly promoted mast cell differentiation in response to stem cell factor or interleukin-3, whereas TEL/ABL preferentially induced myeloid differentiation of hematopoietic stem/progenitor cells. These results indicate that ABL and ARG kinase activate distinct differentiation pathways to induce specific diseases in vivo, that is, myeloid leukemia and mastocytosis, respectively. Further elucidation of the differences in their properties should provide important insight into the pathogenic mechanisms of oncogenes of the ABL kinase family.
Collapse
Affiliation(s)
- A Yokota
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, Kyoto, Japan
| | - H Hirai
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, Kyoto, Japan
| | - T Shoji
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, Kyoto, Japan
| | - T Maekawa
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, Kyoto, Japan
| | - K Okuda
- Department of Molecular Diagnostics and Therapeutics, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| |
Collapse
|
37
|
Vasquez E, Cristofaro V, Lukianov S, Burkhard FC, Gheinani AH, Monastyrskaya K, Bielenberg DR, Sullivan MP, Adam RM. Deletion of neuropilin 2 enhances detrusor contractility following bladder outlet obstruction. JCI Insight 2017; 2:e90617. [PMID: 28194441 DOI: 10.1172/jci.insight.90617] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Chronic urethral obstruction and the ensuing bladder wall remodeling can lead to diminished bladder smooth muscle (BSM) contractility and debilitating lower urinary tract symptoms. No effective pharmacotherapy exists to restore BSM contractile function. Neuropilin 2 (Nrp2) is a transmembrane protein that is highly expressed in BSM. Nrp2 deletion in mice leads to increased BSM contraction. We determined whether genetic ablation of Nrp2 could restore BSM contractility following obstruction. Partial bladder outlet obstruction (pBOO) was created by urethral occlusion in mice with either constitutive and ubiquitous, or inducible smooth muscle-specific deletion of Nrp2, and Nrp2-intact littermates. Mice without obstruction served as additional controls. Contractility was measured by isometric tension testing. Nrp2 deletion prior to pBOO increased force generation in BSM 4 weeks following surgery. Deletion of Nrp2 in mice already subjected to pBOO for 4 weeks showed increased contractility of tissues tested 6 weeks after surgery compared with nondeleted controls. Assessment of tissues from patients with urodynamically defined bladder outlet obstruction revealed reduced NRP2 levels in obstructed bladders with compensated compared with decompensated function, relative to asymptomatic controls. We conclude that downregulation of Nrp2 promotes BSM force generation. Neuropilin 2 may represent a novel target to restore contractility following obstruction.
Collapse
Affiliation(s)
- Evalynn Vasquez
- Urological Diseases Research Center, Boston Children's Hospital.,Department of Surgery, Harvard Medical School
| | - Vivian Cristofaro
- Department of Surgery, Harvard Medical School.,Division of Urology, Veterans Affairs Boston Healthcare System, Boston, Massachusetts, USA
| | - Stefan Lukianov
- Urological Diseases Research Center, Boston Children's Hospital
| | - Fiona C Burkhard
- Urology Research Laboratory, Department of Clinical Research, Universität Bern, Bern, Switzerland
| | - Ali Hashemi Gheinani
- Urology Research Laboratory, Department of Clinical Research, Universität Bern, Bern, Switzerland
| | - Katia Monastyrskaya
- Urology Research Laboratory, Department of Clinical Research, Universität Bern, Bern, Switzerland
| | - Diane R Bielenberg
- Department of Surgery, Harvard Medical School.,Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Maryrose P Sullivan
- Department of Surgery, Harvard Medical School.,Division of Urology, Veterans Affairs Boston Healthcare System, Boston, Massachusetts, USA
| | - Rosalyn M Adam
- Urological Diseases Research Center, Boston Children's Hospital.,Department of Surgery, Harvard Medical School
| |
Collapse
|
38
|
The intragraft microenvironment as a central determinant of chronic rejection or local immunoregulation/tolerance. Curr Opin Organ Transplant 2016; 22:55-63. [PMID: 27898465 DOI: 10.1097/mot.0000000000000373] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE OF REVIEW Chronic rejection is associated with persistent mononuclear cell recruitment, endothelial activation and proliferation, local tissue hypoxia and related biology that enhance effector immune responses. In contrast, the tumor microenvironment elicits signals/factors that inhibit effector T cell responses and rather promote immunoregulation locally within the tissue itself. The identification of immunoregulatory check points and/or secreted factors that are deficient within allografts is of great importance in the understanding and prevention of chronic rejection. RECENT FINDINGS The relative deficiency of immunomodulatory molecules (cell surface and secreted) on microvascular endothelial cells within the intragraft microenvironment, is of functional importance in shaping the phenotype of rejection. These regulatory molecules include coinhibitory and/or intracellular regulatory signals/factors that enhance local activation of T regulatory cells. For example, semaphorins may interact with endothelial cells and CD4 T cells to promote local tolerance. Additionally, metabolites and electrolytes within the allograft microenvironment may regulate local effector and regulatory cell responses. SUMMARY Multiple factors within allografts shape the microenvironment either towards local immunoregulation or proinflammation. Promoting the expression of intragraft cell surface or secreted molecules that support immunoregulation will be critical for long-term graft survival and/or alloimmune tolerance.
Collapse
|
39
|
Yin M, Lu Q, Liu X, Wang T, Liu Y, Chen L. Silencing Drp1 inhibits glioma cells proliferation and invasion by RHOA/ ROCK1 pathway. Biochem Biophys Res Commun 2016; 478:663-8. [PMID: 27495873 DOI: 10.1016/j.bbrc.2016.08.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 08/01/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUNDS Dynamin-related protein 1 (Drp1) is a newly discovered therapeutic target for tumor initiation, migration, proliferation, and chemosensitivity. In the present study, we aimed to examine the level of expression and distribution of DRP1 in glioma tissues and explore the concrete mechanism of DRP1 played in glioma. METHODS Expression of DRP1 in glioma tissues was determined by immunohistochemistry staining. The DRP1 gene was knocked down using small interfering RNA, and was overexpressed using plasmids in glioma cells. To assess changes in cell function, in vitro assays for invasion and growth were applied. Protein expression was tested by using Western-blot method. Variation of F-actin in cells was analyzed using immunofluorescence staining. Interactions between proteins were determined by co-immunoprecipitation. RESULTS The protein expression levels of DRP1 were significantly increased in glioma tissues compared to the normal brain tissues. Down-regulation of DRP1 decreased cell proliferation and invasion, and inhibited the formation of pseudopodias and microvillis. Moreover, a possible link between DRP1 and RHOA was confirmed when interactions between these two proteins were observed in the cells. CONCLUSIONS Our results demonstrated that silencing DRP1 regulated the cytoskeleton remodeling through inhibiting RHOA/ROCK1 pathway, and thus decreased the proliferation and invasion of glioma cells.
Collapse
Affiliation(s)
- Maojia Yin
- Department of Neurology, The Second Affiliated Hospital of Chong Qing Medical University, Number 76, LinJiang Road, YuZhong District, 400010, Chong Qing, China
| | - Qin Lu
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University, 54 Youdian Road, 310006, Hangzhou, China
| | - Xi Liu
- Department of Neurology, The Second Affiliated Hospital of Chong Qing Medical University, Number 76, LinJiang Road, YuZhong District, 400010, Chong Qing, China
| | - Teng Wang
- Department of Neurology, The Second Affiliated Hospital of Chong Qing Medical University, Number 76, LinJiang Road, YuZhong District, 400010, Chong Qing, China
| | - Ying Liu
- Department of Neurology, The Second Affiliated Hospital of Chong Qing Medical University, Number 76, LinJiang Road, YuZhong District, 400010, Chong Qing, China
| | - Lifen Chen
- Department of Neurology, The Second Affiliated Hospital of Chong Qing Medical University, Number 76, LinJiang Road, YuZhong District, 400010, Chong Qing, China.
| |
Collapse
|
40
|
Yang WJ, Hu J, Uemura A, Tetzlaff F, Augustin HG, Fischer A. Semaphorin-3C signals through Neuropilin-1 and PlexinD1 receptors to inhibit pathological angiogenesis. EMBO Mol Med 2016. [PMID: 26194913 PMCID: PMC4604683 DOI: 10.15252/emmm.201404922] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Retinopathy of prematurity causes visual impairment due to destructive neoangiogenesis after degeneration of the retinal microvasculature. This study was aimed at analyzing whether local delivery of Semaphorin-3C (Sema3C) suppresses pathological retinal angiogenesis. Sema3C exerted potent inhibiting effects in cellular models of angiogenesis. In an endothelial cell xenotransplantation assay, Sema3C acted primarily on immature microvessels by inducing endothelial cell apoptosis. Intravitreal administration of recombinant Sema3C disrupted endothelial tip cell formation and cell–cell contacts, which led to decreased vascular bed expansion and vessel branching in the growing retinal vasculature of newborn mice, while not affecting mature vessels in the adult retina. Sema3C administration strongly inhibited the formation of pathological pre-retinal vascular tufts during oxygen-induced retinopathy. Mechanistically, Sema3C signaled through the receptors Neuropilin-1 and PlexinD1, which were strongly expressed on vascular tufts, induced VE-cadherin internalization, and abrogated vascular endothelial growth factor (VEGF)-induced activation of the kinases AKT, FAK, and p38MAPK. This disrupted endothelial cell junctions, focal adhesions, and cytoskeleton assembly resulted in decreased cell migration and survival. Thus, this study identified Sema3C as a potent and selective inhibitor of pathological retinal angiogenesis.
Collapse
Affiliation(s)
- Wan-Jen Yang
- Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany Vascular Biology and Tumor Angiogenesis, Medical Faculty Mannheim (CBTM) Heidelberg University, Mannheim, Germany
| | - Junhao Hu
- Vascular Oncology and Metastasis (A190), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Akiyoshi Uemura
- Department of Retinal Vascular Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Fabian Tetzlaff
- Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Hellmut G Augustin
- Vascular Biology and Tumor Angiogenesis, Medical Faculty Mannheim (CBTM) Heidelberg University, Mannheim, Germany Vascular Oncology and Metastasis (A190), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Andreas Fischer
- Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany Vascular Biology and Tumor Angiogenesis, Medical Faculty Mannheim (CBTM) Heidelberg University, Mannheim, Germany Department of Medicine I and Clinical Chemistry, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
41
|
Neuropilin 1 Receptor Is Up-Regulated in Dysplastic Epithelium and Oral Squamous Cell Carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1055-64. [PMID: 26877262 DOI: 10.1016/j.ajpath.2015.11.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 11/11/2015] [Accepted: 11/30/2015] [Indexed: 12/17/2022]
Abstract
Neuropilins are receptors for disparate ligands, including proangiogenic factors such as vascular endothelial growth factor and inhibitory class 3 semaphorin (SEMA3) family members. Differentiated cells in skin epithelium and cutaneous squamous cell carcinoma highly express the neuropilin-1 (NRP1) receptor. We examined the expression of NRP1 in human and mouse oral mucosa. NRP1 was significantly up-regulated in oral epithelial dysplasia and oral squamous cell carcinoma (OSCC). NRP1 receptor localized to the outer suprabasal epithelial layers in normal tongue, an expression pattern similar to the normal skin epidermis. However, dysplastic tongue epithelium and OSCC up-regulated NRP1 in basal and proliferating epithelial layers, a profile unseen in cutaneous squamous cell carcinoma. NRP1 up-regulation is observed in a mouse carcinogen-induced OSCC model and in human tongue OSCC biopsies. Human OSCC cell lines express NRP1 protein in vitro and in mouse tongue xenografts. Sites of capillary infiltration into orthotopic OSCC tumors correlate with high NRP1 expression. HSC3 xenografts, which express the highest NRP1 levels of the cell lines examined, showed massive intratumoral lymphangiogenesis. SEMA3A inhibited OSCC cell migration, suggesting that the NRP1 receptor was bioactive in OSCC. In conclusion, NRP1 is regulated in the oral epithelium and is selectively up-regulated during epithelial dysplasia. NRP1 may function as a reservoir to sequester proangiogenic ligands within the neoplastic compartment, thereby recruiting neovessels toward tumor cells.
Collapse
|
42
|
Ueno N, Shimizu A, Kanai M, Iwaya Y, Ueda S, Nakayama J, Seo MK. Enhanced Expression of Fibroblast Growth Factor Receptor 3 IIIc Promotes Human Esophageal Carcinoma Cell Proliferation. J Histochem Cytochem 2015; 64:7-17. [PMID: 26487184 DOI: 10.1369/0022155415616161] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Accepted: 10/08/2015] [Indexed: 11/22/2022] Open
Abstract
Deregulated expression of fibroblast growth factor receptors (FGFRs) and their ligands plays critical roles in tumorigenesis. The gene expression of an alternatively spliced isoforms of FGFR3, FGFR3IIIc, was analyzed by RT-PCR in samples from patients with esophageal carcinoma (EC), including esophageal squamous cell carcinoma (ESCC) and adenocarcinoma (EAC). The incidence of FGFR3IIIc was higher in EC [12/16 (75%); p=0.073] than in non-cancerous mucosa (NCM) [6/16 (38%)]. Indeed, an immunohistochemical analysis of early-stage ESCC showed that carcinoma cells expressing FGFR3IIIc stained positively with SCC-112, a tumor marker, and Ki67, a cell proliferation marker, suggesting that the expression of FGFR3IIIc promotes cell proliferation. We used EC-GI-10 cells endogenously expressing FGFR3IIIc as a model of ESCC to provide mechanistic insight into the role of FGFR3IIIc in ESCC. The knockdown of endogenous FGFR3 using siRNA treatment significantly abrogated cell proliferation and the overexpression of FGFR3IIIc in cells with enhanced cell proliferation. EC-GI-10 cells and ESCC from patients with EC showed endogenous expression of FGF2, a specific ligand for FGFR3IIIc, suggesting that the upregulated expression of FGFR3IIIc may create autocrine FGF signaling in ESCC. Taken together, FGFR3IIIc may have the potential to be an early-stage tumor marker and a molecular target for ESCC therapy.
Collapse
Affiliation(s)
- Nobuhiro Ueno
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan (NU, AS, MKS)
| | - Akio Shimizu
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan (NU, AS, MKS)
| | - Michiyuki Kanai
- Digestive Disease Center, Hanwasumiyoshi General Hospital, Osaka, Japan (MK)
| | - Yugo Iwaya
- Department of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan (YI)
| | - Shugo Ueda
- Department of Gastroenterological Surgery and Oncology, Kitano Hospital, Osaka, Japan (SU)
| | - Jun Nakayama
- Department of Molecular Pathology, Shinshu University Graduate School of Medicine, Matsumoto, Japan (JN)
| | - Misuzu Kurokawa Seo
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan (NU, AS, MKS)
| |
Collapse
|
43
|
Li X, Parker MW, Vander Kooi CW. Control of cellular motility by neuropilin-mediated physical interactions. Biomol Concepts 2015; 5:157-66. [PMID: 25018786 DOI: 10.1515/bmc-2013-0035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The neuropilin (Nrp) family consists of multifunctional cell surface receptors with critical roles in a number of different cell and tissue types. A core aspect of Nrp function is in ligand-dependent cellular migration, where it controls the multistep process of cellular motility through integration of ligand binding and receptor signaling. At a molecular level, the role of Nrp in migration is intimately connected to the control of adhesive interactions and cytoskeletal reorganization. Here, we review the physiological role of Nrp in cellular adhesion and motility in the cardiovascular and nervous systems. We also discuss the emerging pathological role of Nrp in tumor cell migration and metastasis, providing motivation for continued efforts toward developing Nrp inhibitors.
Collapse
|
44
|
Yoshida A, Shimizu A, Asano H, Kadonosono T, Kondoh SK, Geretti E, Mammoto A, Klagsbrun M, Seo MK. VEGF-A/NRP1 stimulates GIPC1 and Syx complex formation to promote RhoA activation and proliferation in skin cancer cells. Biol Open 2015. [PMID: 26209534 PMCID: PMC4582117 DOI: 10.1242/bio.010918] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Neuropilin-1 (NRP1) has been identified as a VEGF-A receptor. DJM-1, a human skin cancer cell line, expresses endogenous VEGF-A and NRP1. In the present study, the RNA interference of VEGF-A or NRP1 suppressed DJM-1 cell proliferation. Furthermore, the overexpression of the NRP1 wild type restored shNRP1-treated DJM-1 cell proliferation, whereas NRP1 cytoplasmic deletion mutants did not. A co-immunoprecipitation analysis revealed that VEGF-A induced interactions between NRP1 and GIPC1, a scaffold protein, and complex formation between GIPC1 and Syx, a RhoGEF. The knockdown of GIPC1 or Syx reduced active RhoA and DJM-1 cell proliferation without affecting the MAPK or Akt pathway. C3 exoenzyme or Y27632 inhibited the VEGF-A-induced proliferation of DJM-1 cells. Conversely, the overexpression of the constitutively active form of RhoA restored the proliferation of siVEGF-A-treated DJM-1 cells. Furthermore, the inhibition of VEGF-A/NRP1 signaling upregulated p27, a CDK inhibitor. A cell-penetrating oligopeptide that targeted GIPC1/Syx complex formation inhibited the VEGF-A-induced activation of RhoA and suppressed DJM-1 cell proliferation. In conclusion, this new signaling pathway of VEGF-A/NRP1 induced cancer cell proliferation by forming a GIPC1/Syx complex that activated RhoA to degrade the p27 protein.
Collapse
Affiliation(s)
- Ayumi Yoshida
- Division of Engineering (Biotechnology), Graduate School of Engineering, Kyoto Sangyo University, Kyoto 603-8555, Japan
| | - Akio Shimizu
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8047, Japan
| | - Hirotsugu Asano
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8047, Japan
| | - Tetsuya Kadonosono
- Biofunctional Engineering, Graduate School of Bioscience & Biotechnology, Tokyo Institute of Technology, Tokyo 226-8503, Japan
| | - Shinae Kizaka Kondoh
- Biofunctional Engineering, Graduate School of Bioscience & Biotechnology, Tokyo Institute of Technology, Tokyo 226-8503, Japan
| | - Elena Geretti
- Vascular Biology Program, Boston Children's Hospital, Departments of Surgery and Pathology and Harvard Medical School, Boston, MA 02115, USA
| | - Akiko Mammoto
- Vascular Biology Program, Boston Children's Hospital, Departments of Surgery and Pathology and Harvard Medical School, Boston, MA 02115, USA
| | - Michael Klagsbrun
- Vascular Biology Program, Boston Children's Hospital, Departments of Surgery and Pathology and Harvard Medical School, Boston, MA 02115, USA
| | - Misuzu Kurokawa Seo
- Division of Engineering (Biotechnology), Graduate School of Engineering, Kyoto Sangyo University, Kyoto 603-8555, Japan Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8047, Japan
| |
Collapse
|
45
|
Regulation of mTOR Signaling by Semaphorin 3F-Neuropilin 2 Interactions In Vitro and In Vivo. Sci Rep 2015; 5:11789. [PMID: 26156437 PMCID: PMC4496725 DOI: 10.1038/srep11789] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 06/04/2015] [Indexed: 12/16/2022] Open
Abstract
Semaphorin 3F (SEMA3F) provides neuronal guidance cues via its ability to bind neuropilin 2 (NRP2) and Plexin A family molecules. Recent studies indicate that SEMA3F has biological effects in other cell types, however its mechanism(s) of function is poorly understood. Here, we analyze SEMA3F-NRP2 signaling responses in human endothelial, T cell and tumor cells using phosphokinase arrays, immunoprecipitation and Western blot analyses. Consistently, SEMA3F inhibits PI-3K and Akt activity, and responses are associated with the disruption of mTOR/rictor assembly and mTOR-dependent activation of the RhoA GTPase. We also find that the expression of vascular endothelial growth factor, as well as mTOR-inducible cellular activation responses and cytoskeleton stability are inhibited by SEMA3F-NRP2 interactions in vitro. In vivo, local and systemic overproduction of SEMA3F reduces tumor growth in NRP2-expressing xenografts. Taken together, SEMA3F regulates mTOR signaling in diverse human cell types, suggesting that it has broad therapeutic implications.
Collapse
|
46
|
Nakayama H, Huang L, Kelly RP, Oudenaarden CRL, Dagher A, Hofmann NA, Moses MA, Bischoff J, Klagsbrun M. Infantile hemangioma-derived stem cells and endothelial cells are inhibited by class 3 semaphorins. Biochem Biophys Res Commun 2015; 464:126-32. [PMID: 26086095 DOI: 10.1016/j.bbrc.2015.06.087] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Accepted: 06/12/2015] [Indexed: 01/13/2023]
Abstract
Class 3 semaphorins were discovered as a family of axon guidance molecules, but are now known to be involved in diverse biologic processes. In this study, we investigated the anti-angiogenic potential of SEMA3E and SEMA3F (SEMA3E&F) in infantile hemangioma (IH). IH is a common vascular tumor that involves both vasculogenesis and angiogenesis. Our lab has identified and isolated hemangioma stem cells (HemSC), glucose transporter 1 positive (GLUT1(+)) endothelial cells (designated as GLUT1(sel) cells) based on anti-GLUT1 magnetic beads selection and GLUT1-negative endothelial cells (named HemEC). We have shown that these types of cells play important roles in hemangiogenesis. We report here that SEMA3E inhibited HemEC migration and proliferation while SEMA3F was able to suppress the migration and proliferation in all three types of cells. Confocal microscopy showed that stress fibers in HemEC were reduced by SEMA3E&F and that stress fibers in HemSC were decreased by SEMA3F, which led to cytoskeletal collapse and loss of cell motility in both cell types. Additionally, SEMA3E&F were able to inhibit vascular endothelial growth factor (VEGF)-induced sprouts in all three types of cells. Further, SEMA3E&F reduced the level of p-VEGFR2 and its downstream p-ERK in HemEC. These results demonstrate that SEMA3E&F inhibit IH cell proliferation and suppress the angiogenic activities of migration and sprout formation. SEMA3E&F may have therapeutic potential to treat or prevent growth of highly proliferative IH.
Collapse
Affiliation(s)
- Hironao Nakayama
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Division of Cell Growth and Tumor Regulation, Proteo-Science Center, Ehime University, Toon, Ehime 791-0295, Japan
| | - Lan Huang
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ryan P Kelly
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Clara R L Oudenaarden
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Adelle Dagher
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Nicole A Hofmann
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Marsha A Moses
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Joyce Bischoff
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Michael Klagsbrun
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
47
|
Identification of Glioblastoma Phosphotyrosine-Containing Proteins with Two-Dimensional Western Blotting and Tandem Mass Spectrometry. BIOMED RESEARCH INTERNATIONAL 2015; 2015:134050. [PMID: 26090378 PMCID: PMC4450212 DOI: 10.1155/2015/134050] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 08/25/2014] [Accepted: 08/25/2014] [Indexed: 12/24/2022]
Abstract
To investigate the presence of, and the potential biological roles of, protein tyrosine phosphorylation in the glioblastoma pathogenesis, two-dimensional gel electrophoresis- (2DGE-) based Western blotting coupled with liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI-MS/MS) analysis was used to detect and identify the phosphotyrosine immunoreaction-positive proteins in a glioblastoma tissue. MS/MS and Mascot analyses were used to determine the phosphotyrosine sites of each phosphopeptide. Protein domain and motif analysis and systems pathway analysis were used to determine the protein domains/motifs that contained phosphotyrosine residue and signal pathway networks to clarify the potential biological functions of protein tyrosine phosphorylation. A total of 24 phosphotyrosine-containing proteins were identified. Each phosphotyrosine-containing protein contained at least one tyrosine kinase phosphorylation motif and a certain structural and functional domains. Those phosphotyrosine-containing proteins were involved in the multiple signal pathway systems such as oxidative stress, stress response, and cell migration. Those data show 2DGE-based Western blotting, MS/MS, and bioinformatics are a set of effective approaches to detect and identify glioblastoma tyrosine-phosphorylated proteome and to effectively rationalize the biological roles of tyrosine phosphorylation in the glioblastoma biological systems. It provides novel insights regarding tyrosine phosphorylation and its potential role in the molecular mechanism of a glioblastoma.
Collapse
|
48
|
MacKay JL, Kumar S. Simultaneous and independent tuning of RhoA and Rac1 activity with orthogonally inducible promoters. Integr Biol (Camb) 2015; 6:885-94. [PMID: 25044255 DOI: 10.1039/c4ib00099d] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The GTPases RhoA and Rac1 are key regulators of cell spreading, adhesion, and migration, and they exert distinct effects on the actin cytoskeleton. While RhoA classically stimulates stress fiber assembly and contraction, Rac1 promotes branched actin polymerization and membrane protrusion. These competing influences are reinforced by antagonistic crosstalk between RhoA and Rac1, which has complicated efforts to identify the specific mechanisms by which each GTPase regulates cell behavior. We therefore wondered whether RhoA and Rac1 are intrinsically coupled or whether they can be manipulated independently. To address this question, we placed constitutively active (CA) RhoA under a doxycycline-inducible promoter and CA Rac1 under an orthogonal cumate-inducible promoter, and we stably introduced both constructs into glioblastoma cells. We found that doxycycline addition increased RhoA activity without altering Rac1, and similarly cumate addition increased Rac1 activity without altering RhoA. Furthermore, co-expression of both mutants enabled high activation of RhoA and Rac1 simultaneously. When cells were cultured on collagen hydrogels, RhoA activation prevented cell spreading and motility, whereas Rac1 activation stimulated migration and dynamic cell protrusions. Interestingly, high activation of both GTPases induced a third phenotype, in which cells migrated at intermediate speeds similar to control cells but also aggregated into large, contractile clusters. In addition, we demonstrate dynamic and reversible switching between high RhoA and high Rac1 phenotypes. Overall, this approach represents a unique way to access different combinations of RhoA and Rac1 activity levels in a single cell and may serve as a valuable tool for multiplexed dissection and control of mechanobiological signals.
Collapse
Affiliation(s)
- Joanna L MacKay
- Department of Chemical and Biomolecular Engineering, University of California-Berkeley, Berkeley, California 94720, USA
| | | |
Collapse
|
49
|
Loginov VI, Dmitriev AA, Senchenko VN, Pronina IV, Khodyrev DS, Kudryavtseva AV, Krasnov GS, Gerashchenko GV, Chashchina LI, Kazubskaya TP, Kondratieva TT, Lerman MI, Angeloni D, Braga EA, Kashuba VI. Tumor Suppressor Function of the SEMA3B Gene in Human Lung and Renal Cancers. PLoS One 2015; 10:e0123369. [PMID: 25961819 PMCID: PMC4427300 DOI: 10.1371/journal.pone.0123369] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 02/05/2015] [Indexed: 12/26/2022] Open
Abstract
The SEMA3B gene is located in the 3p21.3 LUCA region, which is frequently affected in different types of cancer. The objective of our study was to expand our knowledge of the SEMA3B gene as a tumor suppressor and the mechanisms of its inactivation. In this study, several experimental approaches were used: tumor growth analyses and apoptosis assays in vitro and in SCID mice, expression and methylation assays and other. With the use of the small cell lung cancer cell line U2020 we confirmed the function of SEMA3B as a tumor suppressor, and showed that the suppression can be realized through the induction of apoptosis and, possibly, associated with the inhibition of angiogenesis. In addition, for the first time, high methylation frequencies have been observed in both intronic (32-39%) and promoter (44-52%) CpG-islands in 38 non-small cell lung carcinomas, including 16 squamous cell carcinomas (SCC) and 22 adenocarcinomas (ADC), and in 83 clear cell renal cell carcinomas (ccRCC). Correlations between the methylation frequencies of the promoter and the intronic CpG-islands of SEMA3B with tumor stage and grade have been revealed for SCC, ADC and ccRCC. The association between the decrease of the SEMA3B mRNA level and hypermethylation of the promoter and the intronic CpG-islands has been estimated in renal primary tumors (P < 0.01). Using qPCR, we observed on the average 10- and 14-fold decrease of the SEMA3B mRNA level in SCC and ADC, respectively, and a 4-fold decrease in ccRCC. The frequency of this effect was high in both lung (92-95%) and renal (84%) tumor samples. Moreover, we showed a clear difference (P < 0.05) of the SEMA3B relative mRNA levels in ADC with and without lymph node metastases. We conclude that aberrant expression and methylation of SEMA3B could be suggested as markers of lung and renal cancer progression.
Collapse
MESH Headings
- Animals
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/pathology
- Cell Line, Tumor
- CpG Islands
- DNA Methylation
- Gene Expression Regulation, Neoplastic
- Humans
- Kidney/metabolism
- Kidney/pathology
- Kidney Neoplasms/genetics
- Kidney Neoplasms/pathology
- Lung/metabolism
- Lung/pathology
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Membrane Glycoproteins/genetics
- Mice, SCID
- Neoplasms, Squamous Cell/genetics
- Neoplasms, Squamous Cell/pathology
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/pathology
- Promoter Regions, Genetic
- Semaphorins/genetics
- Small Cell Lung Carcinoma/genetics
- Small Cell Lung Carcinoma/pathology
Collapse
Affiliation(s)
- Vitaly I. Loginov
- Laboratory of Pathogenomics and Transcriptomics, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315, Moscow, Russia
- Laboratory of Molecular Genetics of Complex Inherited Diseases, Research Center of Medical Genetics, Russian Academy of Medical Sciences, 115478, Moscow, Russia
| | - Alexey A. Dmitriev
- Laboratory of Structural and Functional Genomics, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
- Department of Pathomorphology, P.A. Herzen Moscow Cancer Research Institute, Ministry of Healthcare of the Russian Federation, 125284, Moscow, Russia
| | - Vera N. Senchenko
- Laboratory of Structural and Functional Genomics, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
| | - Irina V. Pronina
- Laboratory of Pathogenomics and Transcriptomics, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315, Moscow, Russia
- Laboratory of Molecular Genetics of Complex Inherited Diseases, Research Center of Medical Genetics, Russian Academy of Medical Sciences, 115478, Moscow, Russia
| | - Dmitry S. Khodyrev
- Laboratory of Genetics, Federal Research Clinical Center of Federal Medical and Biological Agency of Russia, 115682, Moscow, Russia
| | - Anna V. Kudryavtseva
- Laboratory of Structural and Functional Genomics, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
- Department of Pathomorphology, P.A. Herzen Moscow Cancer Research Institute, Ministry of Healthcare of the Russian Federation, 125284, Moscow, Russia
| | - George S. Krasnov
- Laboratory of Structural and Functional Genomics, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
- Laboratory of Biotechnology, Mechnikov Research Institute for Vaccines and Sera, Russian Academy of Medical Sciences, 105064, Moscow, Russia
| | - Ganna V. Gerashchenko
- Department of Molecular Oncogenetics, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, 03680, Kiev, Ukraine
| | - Larisa I. Chashchina
- Department of Molecular Oncogenetics, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, 03680, Kiev, Ukraine
| | - Tatiana P. Kazubskaya
- Research Institute of Clinical Oncology, N.N. Blokhin Cancer Research Center, Russian Academy of Medical Sciences, 115478, Moscow, Russia
| | - Tatiana T. Kondratieva
- Research Institute of Clinical Oncology, N.N. Blokhin Cancer Research Center, Russian Academy of Medical Sciences, 115478, Moscow, Russia
| | | | - Debora Angeloni
- The Institute of Life Sciences, Scuola Superiore Sant'Anna, 56127, Pisa, Italy
- Institute of Clinical Physiology, National Research Council, 56124, Pisa, Italy
- Istituto Toscano Tumori, 56124, Pisa, Italy
| | - Eleonora A. Braga
- Laboratory of Pathogenomics and Transcriptomics, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315, Moscow, Russia
- Laboratory of Molecular Genetics of Complex Inherited Diseases, Research Center of Medical Genetics, Russian Academy of Medical Sciences, 115478, Moscow, Russia
- Laboratory of Post Genomic Molecular Genetic Research, Institute of Biochemical Physics, Russian Academy of Sciences, 119334, Moscow, Russia
| | - Vladimir I. Kashuba
- Department of Molecular Oncogenetics, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, 03680, Kiev, Ukraine
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, SE-17177, Stockholm, Sweden
| |
Collapse
|
50
|
Chen X, Cheng Z, Zhang S, Werling D, Wathes DC. Combining Genome Wide Association Studies and Differential Gene Expression Data Analyses Identifies Candidate Genes Affecting Mastitis Caused by Two Different Pathogens in the Dairy Cow. ACTA ACUST UNITED AC 2015. [DOI: 10.4236/ojas.2015.54040] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|