1
|
Khan A, Sharma P, Dahiya S, Sharma B. Plexins: Navigating through the neural regulation and brain pathology. Neurosci Biobehav Rev 2025; 169:105999. [PMID: 39756719 DOI: 10.1016/j.neubiorev.2024.105999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 12/21/2024] [Accepted: 12/30/2024] [Indexed: 01/07/2025]
Abstract
Plexins are a family of transmembrane receptors known for their diverse roles in neural development, axon guidance, neuronal migration, synaptogenesis, and circuit formation. Semaphorins are a class of secreted and membrane proteins that act as primary ligands for plexin receptors. Semaphorins play a crucial role in central nervous system (CNS) development by regulating processes such as axonal growth, neuronal positioning, and synaptic connectivity. Various types of semaphorins like sema3A, sema4A, sema4C, sema4D, and many more have a crucial role in developing brain diseases. Likewise, various evidence suggests that plexin receptors are of four types: plexin A, plexin B, plexin C, and plexin D. Plexins have emerged as crucial regulators of neurogenesis and neuronal development and connectivity. When bound to semaphorins, these receptors trigger two major networking cascades, namely Rho and Ras GTPase networks. Dysregulation of plexin networking has been implicated in a myriad of brain disorders, including autism spectrum disorder (ASD), Schizophrenia, Alzheimer's disease (AD), Parkinson's disease (PD), and many more. This review synthesizes findings from molecular, cellular, and animal model studies to elucidate the mechanisms by which plexins contribute to the pathogenesis of various brain diseases.
Collapse
Affiliation(s)
- Ariba Khan
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Poonam Sharma
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India; Lloyd Institute of Management and Technology, Plot No.-11, Knowledge Park-II, Greater Noida, 201306 Uttar Pradesh, India.
| | - Sarthak Dahiya
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Bhupesh Sharma
- Department of Pharmaceutical Sciences, Faculty of Life Sciences, Gurugram University (A State Govt. University), Gurugram, Haryana, India.
| |
Collapse
|
2
|
Sajib MS, Zahra FT, Lamprou M, Akwii RG, Park JH, Osorio M, Tullar P, Doci CL, Zhang C, Huveneers S, Van Buul JD, Wang MH, Markiewski MM, Srivastava SK, Zheng Y, Gutkind JS, Hu J, Bickel U, Maeda DY, Zebala JA, Lionakis MS, Trasti S, Mikelis CM. Tumor-induced endothelial RhoA activation mediates tumor cell transendothelial migration and metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.22.614304. [PMID: 39372784 PMCID: PMC11451620 DOI: 10.1101/2024.09.22.614304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
The endothelial barrier plays an active role in transendothelial tumor cell migration during metastasis, however, the endothelial regulatory elements of this step remain obscure. Here we show that endothelial RhoA activation is a determining factor during this process. Breast tumor cell-induced endothelial RhoA activation is the combined outcome of paracrine IL-8-dependent and cell-to-cell contact β 1 integrin-mediated mechanisms, with elements of this pathway correlating with clinical data. Endothelial-specific RhoA blockade or in vivo deficiency inhibited the transendothelial migration and metastatic potential of human breast tumor and three murine syngeneic tumor cell lines, similar to the pharmacological blockade of the downstream RhoA pathway. These findings highlight endothelial RhoA as a potent, universal target in the tumor microenvironment for anti-metastatic treatment of solid tumors.
Collapse
|
3
|
Cui K, Tang X, Yang B, Fan M, Hu A, Wu P, Yang F, Lin J, Kong H, Lu X, Yu S, Xu Y, Liang X. Sema4D Knockout Attenuates Choroidal Neovascularization by Inhibiting M2 Macrophage Polarization Via Regulation of the RhoA/ROCK Pathway. Invest Ophthalmol Vis Sci 2024; 65:34. [PMID: 38913005 PMCID: PMC11204059 DOI: 10.1167/iovs.65.6.34] [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: 02/27/2024] [Accepted: 06/01/2024] [Indexed: 06/25/2024] Open
Abstract
Purpose The aim of this study was to elucidate the role of Sema4D in the pathogenesis of senescence-associated choroidal neovascularization (CNV) and to explore its underlying mechanisms. Methods In this study, we utilized a model of laser-induced CNV in both young (3 months old) and old (18 months old) mice, including those with or without Sema4D knockout. The expression and localization of Sema4D in CNV were assessed using PCR, Western blot, and immunostaining. Subsequently, the morphological and imaging examinations were used to evaluate the size of CNV and vascular leakage. Finally, the expression of M2 markers, senescence-related markers, and molecules involved in the RhoA/ROCK pathway was detected. Results We found that Sema4D was predominantly expressed in macrophages within CNV lesions, and both the mRNA and protein levels of Sema4D progressively increased following laser photocoagulation, a trend more pronounced in old mice. Moreover, Sema4D knockout markedly inhibited M2 polarization in senescent macrophages and reduced the size and leakage of CNV, particularly in aged mice. Mechanistically, aging was found to upregulate RhoA/ROCK signaling, and knockout of Sema4D effectively suppressed the activation of this pathway, with more significant effects observed in aged mice. Conclusions Our findings revealed that the deletion of Sema4D markedly inhibited M2 macrophage polarization through the suppression of the RhoA/ROCK pathway, ultimately leading to the attenuation of senescence-associated CNV. These data indicate that targeting Sema4D could offer a promising approach for gene editing therapy in patients with neovascular age-related macular degeneration.
Collapse
Affiliation(s)
- Kaixuan Cui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xiaoyu Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Boyu Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Matthew Fan
- Yale College, Yale University, New Haven, Connecticut, United States
| | - Andina Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Peiqi Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Fengmei Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jicheng Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Haolin Kong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xi Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Shanshan Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yue Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xiaoling Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
4
|
Field SE, Curle AJ, Barker RA. Inflammation and Huntington's disease - a neglected therapeutic target? Expert Opin Investig Drugs 2024; 33:451-467. [PMID: 38758356 DOI: 10.1080/13543784.2024.2348738] [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: 12/19/2023] [Accepted: 04/24/2024] [Indexed: 05/18/2024]
Abstract
INTRODUCTION Huntington's Disease (HD) is a genetic neurodegenerative disease for which there is currently no disease-modifying treatment. One of several underlying mechanisms proposed to be involved in HD pathogenesis is inflammation; there is now accumulating evidence that the immune system may play an integral role in disease pathology and progression. As such, modulation of the immune system could be a potential therapeutic target for HD. AREAS COVERED To date, the number of trials targeting immune aspects of HD has been limited. However, targeting it, may have great advantages over other therapeutic areas, given that many drugs already exist that have actions in this system coupled to the fact that inflammation can be measured both peripherally and, to some extent, centrally using CSF and PET imaging. In this review, we look at evidence that the immune system and the newly emerging area of the microbiome are altered in HD patients, and then present and discuss clinical trials that have targeted different parts of the immune system. EXPERT OPINION We then conclude by discussing how this field might develop going forward, focusing on the role of imaging and other biomarkers to monitor central immune activation and response to novel treatments in HD.
Collapse
Affiliation(s)
- Sophie E Field
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, and MRC-WT Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Annabel J Curle
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, and MRC-WT Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Roger A Barker
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, and MRC-WT Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
5
|
Cui Y, Jiang X, Yang M, Yuan Y, Zhou Z, Gao X, Jia G, Cao L, Li D, Zhao Y, Zhang X, Zhao G. SEMA4D/VEGF surface enhances endothelialization by diminished-glycolysis-mediated M2-like macrophage polarization. Mater Today Bio 2023; 23:100832. [PMID: 38024840 PMCID: PMC10630656 DOI: 10.1016/j.mtbio.2023.100832] [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: 06/27/2023] [Revised: 09/20/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
Cardiovascular disease remains the leading cause of death and morbidity worldwide. Inflammatory responses after percutaneous coronary intervention led to neoathrosclerosis and in-stent restenosis and thus increase the risk of adverse clinical outcomes. In this work, a metabolism reshaped surface is engineered, which combines the decreased glycolysis promoting, M2-like macrophage polarization, and rapid endothelialization property. Anionic heparin plays as a linker and mediates cationic SEMA4D and VEGF to graft electronically onto PLL surfaces. The system composed by anticoagulant heparin, immunoregulatory SEMA4D and angiogenic VEGF endows the scaffold with significant inhibition of platelets, fibrinogen and anti-thrombogenic properties, also noteworthy immunometabolism reprogram, anti-inflammation M2-like polarization and finally leading to rapid endothelializaiton performances. Our research indicates that the immunometabolism method can accurately reflect the immune state of modified surfaces. It is envisioned immunometabolism study will open an avenue to the surface engineering of vascular implants for better clinical outcomes.
Collapse
Affiliation(s)
- Yuanyuan Cui
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Xiaomei Jiang
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Maozhu Yang
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yinglin Yuan
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Zili Zhou
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Xiang Gao
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Guiqing Jia
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Lvzhou Cao
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Danni Li
- Department of Pharmacy, Longquanyi District of Chengdu Maternity & Child Health Care Hospital, Chengdu, 610072, China
| | - Yanshuang Zhao
- Department of Pharmacy, The People's Hospital of Leshan, Leshan, China
| | - Xin Zhang
- School of Mechanical and Electrical Engineering, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Gaoping Zhao
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| |
Collapse
|
6
|
Mraz V, Funch AB, Jee MH, Gadsbøll ASØ, Weber JF, Yeung K, Lohmann RKD, Hawkes A, Ødum N, Woetmann A, McKay D, Witherden D, Geisler C, Bonefeld CM. CD100 boosts the inflammatory response in the challenge phase of allergic contact dermatitis in mice. Contact Dermatitis 2023; 89:442-452. [PMID: 37700557 DOI: 10.1111/cod.14414] [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: 06/24/2023] [Revised: 08/18/2023] [Accepted: 08/29/2023] [Indexed: 09/14/2023]
Abstract
BACKGROUND Allergic contact dermatitis (ACD) is an inflammatory disease with a complex pathophysiology in which epidermal-resident memory CD8+ T (TRM ) cells play a key role. The mechanisms involved in the activation of CD8+ TRM cells during allergic flare-up responses are not understood. METHODS The expression of CD100 and its ligand Plexin B2 on CD8+ TRM cells and keratinocytes before and after allergen exposure was determined by flow cytometry and RT-qPCR. The role of CD100 in the inflammatory response during the challenge phase of ACD was determined in a model of ACD in CD100 knockout and wild-type mice. RESULTS We show that CD8+ TRM cells express CD100 during homeostatic conditions and up-regulate it following re-exposure of allergen-experienced skin to the experimental contact allergen 1-fluoro-2,4-dinitrobenzene (DNFB). Furthermore, Plexin B2 is up-regulated on keratinocytes following exposure to some contact allergens. We show that loss of CD100 results in a reduced inflammatory response to DNFB with impaired production of IFNγ, IL-17A, CXCL1, CXCL2, CXCL5, and IL-1β and decreased recruitment of neutrophils to the epidermis. CONCLUSION Our study demonstrates that CD100 is expressed on CD8+ TRM cells and is required for full activation of CD8+ TRM cells and the flare-up response of ACD.
Collapse
Affiliation(s)
- Veronika Mraz
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, The University of Copenhagen, Copenhagen, Denmark
| | - Anders B Funch
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, The University of Copenhagen, Copenhagen, Denmark
- Department of Dermatology and Allergy, National Allergy Research Center, Copenhagen University Hospital Gentofte, Hellerup, Denmark
| | - Mia H Jee
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, The University of Copenhagen, Copenhagen, Denmark
| | - Anne-Sofie Ø Gadsbøll
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, The University of Copenhagen, Copenhagen, Denmark
| | - Julie F Weber
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, The University of Copenhagen, Copenhagen, Denmark
| | - Kelvin Yeung
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, The University of Copenhagen, Copenhagen, Denmark
- Department of Dermatology and Allergy, National Allergy Research Center, Copenhagen University Hospital Gentofte, Hellerup, Denmark
| | - Rebecca K D Lohmann
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, The University of Copenhagen, Copenhagen, Denmark
| | - Alana Hawkes
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Niels Ødum
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, The University of Copenhagen, Copenhagen, Denmark
| | - Anders Woetmann
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, The University of Copenhagen, Copenhagen, Denmark
| | - Dianne McKay
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Deborah Witherden
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Carsten Geisler
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, The University of Copenhagen, Copenhagen, Denmark
| | - Charlotte M Bonefeld
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, The University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
7
|
Lieffrig SA, Gyimesi G, Mao Y, Finnemann SC. Clearance phagocytosis by the retinal pigment epithelial during photoreceptor outer segment renewal: Molecular mechanisms and relation to retinal inflammation. Immunol Rev 2023; 319:81-99. [PMID: 37555340 PMCID: PMC10615845 DOI: 10.1111/imr.13264] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/18/2023] [Indexed: 08/10/2023]
Abstract
Mammalian photoreceptor outer segment renewal is a highly coordinated process that hinges on timed cell signaling between photoreceptor neurons and the adjacent retinal pigment epithelial (RPE). It is a strictly rhythmic, synchronized process that underlies in part circadian regulation. We highlight findings from recently developed methods that quantify distinct phases of outer segment renewal in retinal tissue. At light onset, outer segments expose the conserved "eat-me" signal phosphatidylserine exclusively at their distal, most aged tip. A coordinated two-receptor efferocytosis process follows, in which ligands bridge outer segment phosphatidylserine with the RPE receptors αvβ5 integrin, inducing cytosolic signaling toward Rac1 and focal adhesion kinase/MERTK, and with MERTK directly, additionally inhibiting RhoA/ROCK and thus enabling F-actin dynamics favoring outer segment fragment engulfment. Photoreceptors and RPE persist for life with each RPE cell in the eye servicing dozens of overlying photoreceptors. Thus, RPE cells phagocytose more often and process more material than any other cell type. Mutant mice with impaired outer segment renewal largely retain functional photoreceptors and retinal integrity. However, when anti-inflammatory signaling in the RPE via MERTK or the related TYRO3 is lacking, catastrophic inflammation leads to immune cell infiltration that swiftly destroys the retina causing blindness.
Collapse
Affiliation(s)
- Stephanie A. Lieffrig
- Center for Cancer, Genetic Diseases and Gene Regulation, Department of Biological Sciences, Fordham University, Bronx, NY
| | - Gavin Gyimesi
- Center for Cancer, Genetic Diseases and Gene Regulation, Department of Biological Sciences, Fordham University, Bronx, NY
| | | | - Silvia C. Finnemann
- Center for Cancer, Genetic Diseases and Gene Regulation, Department of Biological Sciences, Fordham University, Bronx, NY
| |
Collapse
|
8
|
Al Turkestani N, Zhang Z, Nör JE. Semaphorin 4D Induces Vasculogenic Differentiation of Dental Pulp Stem Cells. Dent J (Basel) 2023; 11:160. [PMID: 37504226 PMCID: PMC10378119 DOI: 10.3390/dj11070160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/25/2023] [Accepted: 06/23/2023] [Indexed: 07/29/2023] Open
Abstract
This work aimed to evaluate the effect of Semaphorin 4D (SEMA4D) signaling through Plexin B1 on the vasculogenic differentiation of dental pulp stem cells. We assessed the protein expression of SEMA4D and Plexin B1 in dental pulp stem cells (DPSC) from permanent human teeth and stem cells from human exfoliated deciduous (SHED) teeth using Western blots. Their expression in human dental pulp tissues and DPSC-engineered dental pulps was determined using immunofluorescence. We then exposed dental pulp stem cells to recombinant human SEMA4D (rhSEMA4D), evaluated the expression of endothelial cell differentiation markers, and assessed the vasculogenic potential of rhSEMA4D using an in vitro sprouting assay. Lastly, Plexin B1 was silenced to ascertain its role in SEMA4D-mediated vasculogenic differentiation. We found that SEMA4D and Plexin B1 are expressed in DPSC, SHED, and human dental pulp tissues. rhSEMA4D (25-100 ng/mL) induced the expression of endothelial markers, i.e., vascular endothelial growth factor receptor (VEGFR)-2, cluster of differentiation (CD)-31, and tyrosine kinase with immunoglobulin-like and EGF-like domains (Tie)-2, in dental pulp stem cells and promoted capillary-like sprouting in vitro (p < 0.05). Furthermore, Plexin B1 silencing abrogated the vasculogenic differentiation of dental pulp stem cells and significantly inhibited capillary sprouting upon exposure to rhSEMA4D. Collectively, these data provide evidence that SEMA4D induces vasculogenic differentiation of dental pulp stem cells through Plexin B1 signaling.
Collapse
Affiliation(s)
- Najla Al Turkestani
- Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA; (N.A.T.); (Z.Z.)
- Department of Restorative and Aesthetic Dentistry, Faculty of Dentistry, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Zhaocheng Zhang
- Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA; (N.A.T.); (Z.Z.)
| | - Jacques Eduardo Nör
- Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA; (N.A.T.); (Z.Z.)
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI 48109, USA
- Department of Otolaryngology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| |
Collapse
|
9
|
Zauderer M, Evans EE. Conclusions of the SIGNAL study in Huntington and implications for treatment of other slowly progressive neurodegenerative diseases. Clin Transl Med 2023; 13:e1169. [PMID: 36710564 PMCID: PMC9885077 DOI: 10.1002/ctm2.1169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 01/31/2023] Open
|
10
|
Britto DD, He J, Misa JP, Chen W, Kakadia PM, Grimm L, Herbert CD, Crosier KE, Crosier PS, Bohlander SK, Hogan BM, Hall CJ, Torres-Vázquez J, Astin JW. Plexin D1 negatively regulates zebrafish lymphatic development. Development 2022; 149:dev200560. [PMID: 36205097 PMCID: PMC9720674 DOI: 10.1242/dev.200560] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 09/23/2022] [Indexed: 06/16/2023]
Abstract
Lymphangiogenesis is a dynamic process that involves the directed migration of lymphatic endothelial cells (LECs) to form lymphatic vessels. The molecular mechanisms that underpin lymphatic vessel patterning are not fully elucidated and, to date, no global regulator of lymphatic vessel guidance is known. In this study, we identify the transmembrane cell signalling receptor Plexin D1 (Plxnd1) as a negative regulator of both lymphatic vessel guidance and lymphangiogenesis in zebrafish. plxnd1 is expressed in developing lymphatics and is required for the guidance of both the trunk and facial lymphatic networks. Loss of plxnd1 is associated with misguided intersegmental lymphatic vessel growth and aberrant facial lymphatic branches. Lymphatic guidance in the trunk is mediated, at least in part, by the Plxnd1 ligands, Semaphorin 3AA and Semaphorin 3C. Finally, we show that Plxnd1 normally antagonises Vegfr/Erk signalling to ensure the correct number of facial LECs and that loss of plxnd1 results in facial lymphatic hyperplasia. As a global negative regulator of lymphatic vessel development, the Sema/Plxnd1 signalling pathway is a potential therapeutic target for treating diseases associated with dysregulated lymphatic growth.
Collapse
Affiliation(s)
- Denver D. Britto
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Jia He
- Skirball Institute of Biomolecular Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - June P. Misa
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Wenxuan Chen
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Purvi M. Kakadia
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand
- Leukaemia and Blood Cancer Research Unit, Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1023, New Zealand
| | - Lin Grimm
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne 3010, Australia
- Department of Anatomy and Physiology, University of Melbourne, Melbourne 3010, Australia
| | - Caitlin D. Herbert
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Kathryn E. Crosier
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Philip S. Crosier
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Stefan K. Bohlander
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand
- Leukaemia and Blood Cancer Research Unit, Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1023, New Zealand
| | - Benjamin M. Hogan
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne 3010, Australia
- Department of Anatomy and Physiology, University of Melbourne, Melbourne 3010, Australia
| | - Christopher J. Hall
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Jesús Torres-Vázquez
- Skirball Institute of Biomolecular Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Jonathan W. Astin
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand
| |
Collapse
|
11
|
Feigin A, Evans EE, Fisher TL, Leonard JE, Smith ES, Reader A, Mishra V, Manber R, Walters KA, Kowarski L, Oakes D, Siemers E, Kieburtz KD, Zauderer M. Pepinemab antibody blockade of SEMA4D in early Huntington's disease: a randomized, placebo-controlled, phase 2 trial. Nat Med 2022; 28:2183-2193. [PMID: 35941373 PMCID: PMC9361919 DOI: 10.1038/s41591-022-01919-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 06/27/2022] [Indexed: 12/18/2022]
Abstract
SIGNAL is a multicenter, randomized, double-blind, placebo-controlled phase 2 study (no. NCT02481674) established to evaluate pepinemab, a semaphorin 4D (SEMA4D)-blocking antibody, for treatment of Huntington's disease (HD). The trial enrolled a total of 265 HD gene expansion carriers with either early manifest (EM, n = 179) or late prodromal (LP, n = 86) HD, randomized (1:1) to receive 18 monthly infusions of pepinemab (n = 91 EM, 41 LP) or placebo (n = 88 EM, 45 LP). Pepinemab was generally well tolerated, with a relatively low frequency of serious treatment-emergent adverse events of 5% with pepinemab compared to 9% with placebo, including both EM and LP participants. Coprimary efficacy outcome measures consisted of assessments within the EM cohort of (1) a two-item HD cognitive assessment family comprising one-touch stockings of Cambridge (OTS) and paced tapping (PTAP) and (2) clinical global impression of change (CGIC). The differences between pepinemab and placebo in mean change (95% confidence interval) from baseline at month 17 for OTS were -1.98 (-4.00, 0.05) (one-sided P = 0.028), and for PTAP 1.43 (-0.37, 3.23) (one-sided P = 0.06). Similarly, because a significant treatment effect was not observed for CGIC, the coprimary endpoint, the study did not meet its prespecified primary outcomes. Nevertheless, a number of other positive outcomes and post hoc subgroup analyses-including additional cognitive measures and volumetric magnetic resonance imaging and fluorodeoxyglucose-positron-emission tomography imaging assessments-provide rationale and direction for the design of a phase 3 study and encourage the continued development of pepinemab in patients diagnosed with EM HD.
Collapse
Affiliation(s)
- Andrew Feigin
- New York University Langone Health and The Marlene and Paolo Fresco Institute for Parkinson's and Movement Disorders, New York, NY, USA
| | | | | | | | | | | | | | | | | | - Lisa Kowarski
- WCG Statistics Collaborative, Inc., Washington, DC, USA
| | - David Oakes
- University of Rochester Medical Center, Rochester, NY, USA
| | | | | | | |
Collapse
|
12
|
Kerhervé M, Rosińska S, Trillet K, Zeinaty A, Feyeux M, Nedellec S, Gavard J. Neuropilin-1 modulates the 3D invasive properties of glioblastoma stem-like cells. Front Cell Dev Biol 2022; 10:981583. [PMID: 36204684 PMCID: PMC9530787 DOI: 10.3389/fcell.2022.981583] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a rare, yet devastating, primary brain tumor in adults. Current treatments remain generally ineffective and GBM almost invariably recurs, resulting in median survival of 15 months. This high malignancy sources notably from the resilience and invasive capabilities of tumor cells. Within GBM, exists a population of self-sustaining transformed cells with stem-like properties (GSCs), which are thought to be responsible for tumor initiation, growth, and invasion, as well as recurrence. In the tumor microenvironment, GSCs might be found in the vicinity of brain endothelial cells, which provide a protective habitat. Likewise, these resistant, quiescent GSCs may accumulate in hypoxic zones, away from the perivascular niche, or travel towards the healthy brain parenchyma, by eminently co-opting neuro-vascular tracks. Herein, we established an ex vivo model to explore GSC invasive behavior. We found that patient-derived cells massively invade the collagen matrix. In addition, we described that the glycoprotein Neuropilin-1 (NRP1) contributes to GSC spreading and invasion. Indeed, both RNA interference-mediated silencing and CRISPR-mediated gene editing deletion of NRP1 strongly impaired the 3D invasive properties of patient-derived GSCs and their close localization to the brain blood vessels. Of note, other typical features of GSCs, such as expansion and self-renewal were maintained. From a mechanistic standpoint, this biological effect might rely on the expression of the β3 subunit integrin cell-extracellular matrix adhesive receptor. Our data, therefore, propose a reliable approach to explore invasive properties of patient glioma cells ex vivo and identify NRP1 as a mediator in this malignant process.
Collapse
Affiliation(s)
- Mathilde Kerhervé
- Team SOAP, CRCI2NA, Nantes Université, Inserm, CNRS, Université D’Angers, Nantes, France
- Equipe Labellisée Ligue Contre le Cancer, Nantes, France
| | - Sara Rosińska
- Team SOAP, CRCI2NA, Nantes Université, Inserm, CNRS, Université D’Angers, Nantes, France
- Equipe Labellisée Ligue Contre le Cancer, Nantes, France
| | - Kilian Trillet
- Team SOAP, CRCI2NA, Nantes Université, Inserm, CNRS, Université D’Angers, Nantes, France
- Equipe Labellisée Ligue Contre le Cancer, Nantes, France
| | - Alya Zeinaty
- Team SOAP, CRCI2NA, Nantes Université, Inserm, CNRS, Université D’Angers, Nantes, France
- Equipe Labellisée Ligue Contre le Cancer, Nantes, France
| | - Magalie Feyeux
- Nantes Université, CHU Nantes, CNRS, Inserm, BioCore, US16, SFR Bonamy, Nantes, France
| | - Steven Nedellec
- Nantes Université, CHU Nantes, CNRS, Inserm, BioCore, US16, SFR Bonamy, Nantes, France
| | - Julie Gavard
- Team SOAP, CRCI2NA, Nantes Université, Inserm, CNRS, Université D’Angers, Nantes, France
- Equipe Labellisée Ligue Contre le Cancer, Nantes, France
- Institut de Cancérologie de L’Ouest (ICO), Angers, France
- *Correspondence: Julie Gavard,
| |
Collapse
|
13
|
Evans EE, Mishra V, Mallow C, Gersz EM, Balch L, Howell A, Reilly C, Smith ES, Fisher TL, Zauderer M. Semaphorin 4D is upregulated in neurons of diseased brains and triggers astrocyte reactivity. J Neuroinflammation 2022; 19:200. [PMID: 35933420 PMCID: PMC9356477 DOI: 10.1186/s12974-022-02509-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 06/01/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The close interaction and interdependence of astrocytes and neurons allows for the possibility that astrocyte dysfunction contributes to and amplifies neurodegenerative pathology. Molecular pathways that trigger reactive astrocytes may represent important targets to preserve normal homeostatic maintenance and modify disease progression. METHODS Semaphorin 4D (SEMA4D) expression in the context of disease-associated neuropathology was assessed in postmortem brain sections of patients with Huntington's (HD) and Alzheimer's disease (AD), as well as in mouse models of HD (zQ175) and AD (CVN; APPSwDI/NOS2-/-) by immunohistochemistry. Effects of SEMA4D antibody blockade were assessed in purified astrocyte cultures and in the CVN mouse AD model. CVN mice were treated weekly from 26 to 38 weeks of age; thereafter mice underwent cognitive assessment and brains were collected for histopathology. RESULTS We report here that SEMA4D is upregulated in neurons during progression of neurodegenerative diseases and is a trigger of reactive astrocytes. Evidence of reactive astrocytes in close proximity to neurons expressing SEMA4D is detected in brain sections of patients and mouse models of HD and AD. We further report that SEMA4D-blockade prevents characteristic loss of GABAergic synapses and restores spatial memory and learning in CVN mice, a disease model that appears to reproduce many features of AD-like pathology including neuroinflammation. In vitro mechanistic studies demonstrate that astrocytes express cognate receptors for SEMA4D and that ligand binding triggers morphological variations, and changes in expression of key membrane receptors and enzymes characteristic of reactive astrocytes. These changes include reductions in EAAT-2 glutamate transporter and glutamine synthetase, key enzymes in neurotransmitter recycling, as well as reduced GLUT-1 glucose and MCT-4 lactate transporters, that allow astrocytes to couple energy metabolism with synaptic activity. Antibody blockade of SEMA4D prevented these changes and reversed functional deficits in glucose uptake. CONCLUSIONS Collectively, these results suggest that SEMA4D blockade may ameliorate disease pathology by preserving normal astrocyte function and reducing the negative consequences of reactive astrogliosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Alan Howell
- Vaccinex, Inc., Research, Rochester, NY, USA
| | | | | | | | - Maurice Zauderer
- Vaccinex, Inc., Research, Rochester, NY, USA.
- Department of Neurology, Center for Health and Technology (SMD), University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
14
|
Liu L, Yang L, Liu X, Liu M, Liu J, Feng X, Nie Z, Luo J. SEMA4D/PlexinB1 promotes AML progression via activation of PI3K/Akt signaling. Lab Invest 2022; 20:304. [PMID: 35794581 PMCID: PMC9258142 DOI: 10.1186/s12967-022-03500-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 06/24/2022] [Indexed: 11/10/2022]
Abstract
Abstract
Background
Acute myeloid leukemia (AML) is the most common type of acute leukemia in adults. SEMA4D is a 150 kDa transmembrane protein that belongs to the IV class of the subfamily of semaphorin family. Previous studies have reported that SEMA4D is a multifunctional target in many solid tumors, involving multiple physiological systems, and there are emerging therapies to target these pathways. The role of SEMA4D in AML has not yet been explored.
Methods
The SEMA4D expression prolile, clinical data and potential prognostic analysis were acquired via the cBioPortal and GEPIA databases. SEMA4D expression was measured using real-time quantitative PCR and western blot. Cell counting kit-8 (CCK8) and flow cytometry were used to evaluate the malignant biological characteristics.
Results
We observed that SEMA4D was increased in AML patients and correlated with risk stratification and prognosis. Moreover, SEMA4D promotes the proliferation and inhibits apoptosis of AML cells by binding to its receptor, PlexinB1, and reduces the sensitivity of AML cells to daunorubicin. In addition, SEMA4D/PlexinB1 promotes the proliferation and survival of AML cells by activating the PI3K/Akt signaling pathway. VX15/2503, an anti-SEMA4D antibody, can inhibit the proliferation of AML cells in xenograft mouse models, thereby inhibiting the development of AML.
Conclusion
SEMA4D will serve as a unique predictive biomarker and a possible therapeutic target in AML.
Collapse
|
15
|
Bonetti G, Paolacci S, Samaja M, Maltese PE, Michelini S, Michelini S, Michelini S, Ricci M, Cestari M, Dautaj A, Medori MC, Bertelli M. Low Efficacy of Genetic Tests for the Diagnosis of Primary Lymphedema Prompts Novel Insights into the Underlying Molecular Pathways. Int J Mol Sci 2022; 23:ijms23137414. [PMID: 35806420 PMCID: PMC9267137 DOI: 10.3390/ijms23137414] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/16/2022] [Accepted: 06/29/2022] [Indexed: 02/07/2023] Open
Abstract
Lymphedema is a chronic inflammatory disorder caused by ineffective fluid uptake by the lymphatic system, with effects mainly on the lower limbs. Lymphedema is either primary, when caused by genetic mutations, or secondary, when it follows injury, infection, or surgery. In this study, we aim to assess to what extent the current genetic tests detect genetic variants of lymphedema, and to identify the major molecular pathways that underlie this rather unknown disease. We recruited 147 individuals with a clinical diagnosis of primary lymphedema and used established genetic tests on their blood or saliva specimens. Only 11 of these were positive, while other probands were either negative (63) or inconclusive (73). The low efficacy of such tests calls for greater insight into the underlying mechanisms to increase accuracy. For this purpose, we built a molecular pathways diagram based on a literature analysis (OMIM, Kegg, PubMed, Scopus) of candidate and diagnostic genes. The PI3K/AKT and the RAS/MAPK pathways emerged as primary candidates responsible for lymphedema diagnosis, while the Rho/ROCK pathway appeared less critical. The results of this study suggest the most important pathways involved in the pathogenesis of lymphedema, and outline the most promising diagnostic and candidate genes to diagnose this disease.
Collapse
Affiliation(s)
- Gabriele Bonetti
- MAGI’s LAB, 38068 Rovereto, Italy; (S.P.); (P.E.M.); (A.D.); (M.C.M.); (M.B.)
- Correspondence: ; Tel.: +39-0365-62-061
| | - Stefano Paolacci
- MAGI’s LAB, 38068 Rovereto, Italy; (S.P.); (P.E.M.); (A.D.); (M.C.M.); (M.B.)
| | | | | | - Sandro Michelini
- Vascular Diagnostics and Rehabilitation Service, Marino Hospital, ASL Roma 6, 00047 Marino, Italy;
| | - Serena Michelini
- Unit of Physical Medicine, “Sapienza” University of Rome, 00185 Rome, Italy;
| | | | - Maurizio Ricci
- Division of Rehabilitation Medicine, Azienda Ospedaliero-Universitaria, Ospedali Riuniti di Ancona, 60126 Ancona, Italy;
| | - Marina Cestari
- Study Centre Pianeta Linfedema, 05100 Terni, Italy;
- Lymphology Sector of the Rehabilitation Service, USLUmbria2, 05100 Terni, Italy
| | - Astrit Dautaj
- MAGI’s LAB, 38068 Rovereto, Italy; (S.P.); (P.E.M.); (A.D.); (M.C.M.); (M.B.)
| | - Maria Chiara Medori
- MAGI’s LAB, 38068 Rovereto, Italy; (S.P.); (P.E.M.); (A.D.); (M.C.M.); (M.B.)
| | - Matteo Bertelli
- MAGI’s LAB, 38068 Rovereto, Italy; (S.P.); (P.E.M.); (A.D.); (M.C.M.); (M.B.)
- MAGI Group, 25010 San Felice del Benaco, Italy;
- MAGI Euregio, 39100 Bolzano, Italy
| |
Collapse
|
16
|
Roberto GM, Emery G. Directing with restraint: Mechanisms of protrusion restriction in collective cell migrations. Semin Cell Dev Biol 2022; 129:75-81. [PMID: 35397972 DOI: 10.1016/j.semcdb.2022.03.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/23/2022] [Accepted: 03/29/2022] [Indexed: 01/15/2023]
Abstract
Cell migration is necessary for morphogenesis, tissue homeostasis, wound healing and immune response. It is also involved in diseases. In particular, cell migration is inherent in metastasis. Cells can migrate individually or in groups. To migrate efficiently, cells need to be able to organize into a leading front that protrudes by forming membrane extensions and a trailing edge that contracts. This organization is scaled up at the group level during collective cell movements. If a cell or a group of cells is unable to limit its leading edge and hence to restrict the formation of protrusions to the front, directional movements are impaired or abrogated. Here we summarize our current understanding of the mechanisms restricting protrusion formation in collective cell migration. We focus on three in vivo examples: the neural crest cell migration, the rotatory migration of follicle cells around the Drosophila egg chamber and the border cell migration during oogenesis.
Collapse
Affiliation(s)
- Gabriela Molinari Roberto
- Vesicular Trafficking and Cell Signalling Research Unit, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, P.O. Box 6128, Downtown station, Montréal, Québec H3C 3J7, Canada
| | - Gregory Emery
- Vesicular Trafficking and Cell Signalling Research Unit, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, P.O. Box 6128, Downtown station, Montréal, Québec H3C 3J7, Canada; Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montréal, Québec H3C 3J7, Canada.
| |
Collapse
|
17
|
Jiang H, Tang J, Qiu L, Zhang Z, Shi S, Xue L, Kui L, Huang T, Nan W, Zhou B, Zhao C, Yu M, Sun Q. Semaphorin 4D is a potential biomarker in pediatric leukemia and promotes leukemogenesis by activating PI3K/AKT and ERK signaling pathways. Oncol Rep 2021. [PMID: 33649851 DOI: 10.3892/or.2021.8021/html] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2023] Open
Abstract
Semaphorin 4D (Sema4D) is highly expressed in a variety of tumors and is associated with high invasion, poor prognosis and poor therapeutic response. However, the expression and role of Sema4D in leukemia remains unclear. The present study investigated the expression of Sema4D in pediatric leukemia and its effects in leukemia cells. The results demonstrated that Sema4D protein was highly expressed in peripheral blood mononuclear cells of patients with pediatric leukemia, and high levels of soluble Sema4D were also observed in the plasma of these patients. Sema4D knockdown induced cell cycle arrest in G0/G1 phase, inhibited proliferation and promoted apoptosis in BALL‑1 cells, while Sema4D overexpression exhibited the opposite effect. In Jurkat cells, Sema4D knockdown inhibited proliferation and promoted apoptosis, while Sema4D overexpression decreased the abundance of the cells in the G0/G1 phase of the cell cycle and promoted proliferation. Sema4D overexpression also increased the migratory capacity of Jurkat cells and the invasive capacity of BALL‑1 cells. The phosphorylation level of PI3K was decreased in both Sema4D knocked‑down Jurkat and BALL‑1 cells, and the phosphorylation level of ERK was decreased in Sema4D knocked‑down BALL‑1 cells. The phosphorylation levels of PI3K, ERK and AKT were elevated in patients with pediatric leukemia, and were correlated to the increased Sema4D expression. Sema4D overexpression was associated with a shorter overall survival in patients with acute myeloid leukemia. Overall, the results of the present study indicated that Sema4D serves an important role in leukemia development by activating PI3K/AKT and ERK signaling, and it may be used as a potential target for the diagnosis and treatment of leukemia.
Collapse
MESH Headings
- Adolescent
- Antigens, CD/biosynthesis
- Antigens, CD/blood
- Biomarkers, Tumor/biosynthesis
- Biomarkers, Tumor/blood
- Case-Control Studies
- Cell Line, Tumor
- Cell Proliferation/physiology
- Child
- Child, Preschool
- Female
- Humans
- Infant
- Jurkat Cells
- Leukemia, Myeloid, Acute/blood
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukocytes, Mononuclear/metabolism
- MAP Kinase Signaling System
- Male
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphorylation
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
- Proto-Oncogene Proteins c-akt/metabolism
- Semaphorins/biosynthesis
- Semaphorins/blood
Collapse
Affiliation(s)
- Hongchao Jiang
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Jiaolian Tang
- Institute of Pediatrics, Children's Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650228, P.R. China
| | - Lijuan Qiu
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Zhen Zhang
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Shulan Shi
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Li Xue
- Institute of Medicine, Dali University, Dali, Yunnan 671000, P.R. China
| | - Liyue Kui
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Tilong Huang
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Weiwei Nan
- Institute of Medicine, Dali University, Dali, Yunnan 671000, P.R. China
| | - Bailing Zhou
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Canchun Zhao
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Ming Yu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Biomedical Engineering Research Center, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Qiangming Sun
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan 650118, P.R. China
| |
Collapse
|
18
|
Jiang H, Tang J, Qiu L, Zhang Z, Shi S, Xue L, Kui L, Huang T, Nan W, Zhou B, Zhao C, Yu M, Sun Q. Semaphorin 4D is a potential biomarker in pediatric leukemia and promotes leukemogenesis by activating PI3K/AKT and ERK signaling pathways. Oncol Rep 2021; 45:1. [PMID: 33649851 PMCID: PMC7877000 DOI: 10.3892/or.2021.7952] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 12/18/2020] [Indexed: 12/18/2022] Open
Abstract
Semaphorin 4D (Sema4D) is highly expressed in a variety of tumors and is associated with high invasion, poor prognosis and poor therapeutic response. However, the expression and role of Sema4D in leukemia remains unclear. The present study investigated the expression of Sema4D in pediatric leukemia and its effects in leukemia cells. The results demonstrated that Sema4D protein was highly expressed in peripheral blood mononuclear cells of patients with pediatric leukemia, and high levels of soluble Sema4D were also observed in the plasma of these patients. Sema4D knockdown induced cell cycle arrest in G0/G1 phase, inhibited proliferation and promoted apoptosis in BALL-1 cells, while Sema4D overexpression exhibited the opposite effect. In Jurkat cells, Sema4D knockdown inhibited proliferation and promoted apoptosis, while Sema4D overexpression decreased the abundance of the cells in the G0/G1 phase of the cell cycle and promoted proliferation. Sema4D overexpression also increased the migratory capacity of Jurkat cells and the invasive capacity of BALL-1 cells. The phosphorylation level of PI3K was decreased in both Sema4D knocked-down Jurkat and BALL-1 cells, and the phosphorylation level of ERK was decreased in Sema4D knocked-down BALL-1 cells. The phosphorylation levels of PI3K, ERK and AKT were elevated in patients with pediatric leukemia, and were correlated to the increased Sema4D expression. Sema4D overexpression was associated with a shorter overall survival in patients with acute myeloid leukemia. Overall, the results of the present study indicated that Sema4D serves an important role in leukemia development by activating PI3K/AKT and ERK signaling, and it may be used as a potential target for the diagnosis and treatment of leukemia.
Collapse
Affiliation(s)
- Hongchao Jiang
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Jiaolian Tang
- Institute of Pediatrics, Children's Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650228, P.R. China
| | - Lijuan Qiu
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Zhen Zhang
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Shulan Shi
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Li Xue
- Institute of Medicine, Dali University, Dali, Yunnan 671000, P.R. China
| | - Liyue Kui
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Tilong Huang
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Weiwei Nan
- Institute of Medicine, Dali University, Dali, Yunnan 671000, P.R. China
| | - Bailing Zhou
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Canchun Zhao
- Institute of Pediatrics, The Kunming Children's Hospital, Kunming, Yunnan 650228, P.R. China
| | - Ming Yu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Biomedical Engineering Research Center, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Qiangming Sun
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan 650118, P.R. China
| |
Collapse
|
19
|
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
|
20
|
Chen W, Xia P, Wang H, Tu J, Liang X, Zhang X, Li L. The endothelial tip-stalk cell selection and shuffling during angiogenesis. J Cell Commun Signal 2019; 13:291-301. [PMID: 30903604 DOI: 10.1007/s12079-019-00511-z] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 02/25/2019] [Indexed: 12/17/2022] Open
Abstract
Angiogenesis is a critical, fine-tuned, multi-staged biological process. Tip-stalk cell selection and shuffling are the building blocks of sprouting angiogenesis. Accumulated evidences show that tip-stalk cell selection and shuffling are regulated by a variety of physical, chemical and biological factors, especially the interaction among multiple genes, their products and environments. The classic Notch-VEGFR, Slit-Robo, ECM-binding integrin, semaphorin and CCN family play important roles in tip-stalk cell selection and shuffling. In this review, we outline the progress and prospect in the mechanism and the roles of the various molecules and related signaling pathways in endothelial tip-stalk cell selection and shuffling. In the future, the regulators of tip-stalk cell selection and shuffling would be the potential markers and targets for angiogenesis.
Collapse
Affiliation(s)
- Wenqi Chen
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Peng Xia
- Department of Anesthesia, Jilin Provincial People's Hospital, Changchun, China
| | - Heping Wang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, China
| | - Jihao Tu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xinyue Liang
- The First Hospital of Jilin University, Changchun, China
| | - Xiaoling Zhang
- The First Hospital of Jilin University, Changchun, China. .,Institute of Immunology, Jilin University, Changchun, China.
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China.
| |
Collapse
|
21
|
Xia Y, Cai XY, Fan JQ, Zhang LL, Ren JH, Li ZY, Zhang RG, Zhu F, Wu G. The role of sema4D in vasculogenic mimicry formation in non-small cell lung cancer and the underlying mechanisms. Int J Cancer 2018; 144:2227-2238. [PMID: 30374974 DOI: 10.1002/ijc.31958] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 10/06/2018] [Accepted: 10/24/2018] [Indexed: 12/11/2022]
Abstract
Vasculogenic mimicry (VM) is a special vascular pattern in malignant tumors, which is composed of highly aggressive tumor cells. This tumor cell-mediated blood supply pattern is closely associated with a poor prognosis in cancer patients. The interaction of axon guidance factor Sema4D and its high affinity receptor plexinB1 could activate small GTPase RhoA and its downstream ROCKs; this process has an active role in the migration of endothelial cells and tumor angiogenesis. Here, we have begun to uncover the role of this pathway in VM formation in non-small cell lung cancer (NSCLC). First, we confirmed this special form of vasculature in NSCLC tissues and found the existence of VM channels in tumor tissues was correlated with Sema4D expression. Further, we found that inhibition of Sema4D in the human NSCLC cells H1299 and HCC827 reduces VM formation both in vitro and in vivo. Moreover, we demonstrated that downregulating the expression of plexinB1 by siRNA expressing vectors and inhibiting the RhoA/ROCK signaling pathway using fasudil can reduce VM formation of H1299 and HCC827 cells. Finally, we found that suppression of Sema4D leads to less stress fibers and depleted the motility of H1299 and HCC827 cells. Collectively, our study implicates Sema4D plays an important role in the process of VM formation in NSCLC through activating the RhoA/ROCK pathway and regulating tumor cell plasticity and migration. Modulation of the Sema4D/plexinB1 and downstream RhoA/ROCK pathway may prevent the tumor blood supply through the VM pattern, which may eventually halt growth and metastasis of NSCLC.
Collapse
Affiliation(s)
- Yun Xia
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xian-Yi Cai
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ji-Quan Fan
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li-Ling Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing-Hua Ren
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhen-Yu Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui-Guang Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Zhu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
22
|
Clavijo PE, Friedman J, Robbins Y, Moore EC, Smith E, Zauderer M, Evans EE, Allen CT. Semaphorin4D Inhibition Improves Response to Immune-Checkpoint Blockade via Attenuation of MDSC Recruitment and Function. Cancer Immunol Res 2018; 7:282-291. [PMID: 30514791 DOI: 10.1158/2326-6066.cir-18-0156] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 09/20/2018] [Accepted: 11/27/2018] [Indexed: 01/15/2023]
Abstract
Tumor infiltration by immunosuppressive myeloid cells, such as myeloid-derived suppressor cells (MDSCs), causes resistance to immunotherapy. Semaphorin4D, originally characterized for its axonal guidance properties, also contributes to endothelial cell migration and survival and modulates global immune cytokine profiles and myeloid cell polarization within the tumor microenvironment. Here, we show how a therapeutic murine Sema4D mAb improves responses to immune-checkpoint blockade (ICB) in two murine carcinoma models. Treatment of tumor-bearing mice with Sema4D mAb abrogated Ly6Ghi PMN-MDSC recruitment through reducing MAPK-dependent chemokine production by tumor cells in Murine oral cancer-1 (MOC1) tumors. PMN-MDSC suppressive capacity was reduced through inhibition of Sema4D-driven arginase expression. These changes led to enhanced tumor infiltration by CD8+ TIL and activation of tumor-draining lymph node T lymphocytes in response to tumor antigen. Sema4D mAb in combination with either CTLA-4 or PD-1 blockade enhanced rejection of tumors or tumor growth delay, resulting in prolonged survival with either treatment. This function of Sema4D mAb provides a rationale for its evaluation in combination with ICB to treat tumors with immunosuppressive myeloid infiltration.
Collapse
Affiliation(s)
- Paul E Clavijo
- Translational Tumor Immunology Program, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland
| | - Jay Friedman
- Translational Tumor Immunology Program, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland
| | - Yvette Robbins
- Translational Tumor Immunology Program, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland
| | - Ellen C Moore
- Translational Tumor Immunology Program, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland
| | | | | | | | - Clint T Allen
- Translational Tumor Immunology Program, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland. .,Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
23
|
Duarte A, André‐Grégoire G, Trillet K, Thys A, Bidère N, Ribeiro‐Silva A, Gavard J. Inhibition of mTOR in head and neck cancer cells alters endothelial cell morphology in a paracrine fashion. Mol Carcinog 2018; 58:161-168. [DOI: 10.1002/mc.22911] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 09/04/2018] [Accepted: 09/11/2018] [Indexed: 01/04/2023]
Affiliation(s)
- Andressa Duarte
- CRCINATeam SOAPInsermCNRSUniversité de NantesUniversité d'AngersNantesFrance
- Laboratory of Cancer PathologyRibeirao Preto Medical SchoolUniversity of Sao PauloSao PauloBrazil
| | - Gwennan André‐Grégoire
- CRCINATeam SOAPInsermCNRSUniversité de NantesUniversité d'AngersNantesFrance
- Institut de Cancérologie de l'OuestSite René GauducheauSaint HerblainFrance
| | - Kilian Trillet
- CRCINATeam SOAPInsermCNRSUniversité de NantesUniversité d'AngersNantesFrance
| | - An Thys
- CRCINATeam SOAPInsermCNRSUniversité de NantesUniversité d'AngersNantesFrance
| | - Nicolas Bidère
- CRCINATeam SOAPInsermCNRSUniversité de NantesUniversité d'AngersNantesFrance
| | - Alfredo Ribeiro‐Silva
- Laboratory of Cancer PathologyRibeirao Preto Medical SchoolUniversity of Sao PauloSao PauloBrazil
| | - Julie Gavard
- CRCINATeam SOAPInsermCNRSUniversité de NantesUniversité d'AngersNantesFrance
- Institut de Cancérologie de l'OuestSite René GauducheauSaint HerblainFrance
| |
Collapse
|
24
|
Liu BS, Dai XY, Xia HW, Xu HJ, Tang QL, Gong QY, Nie YZ, Bi F. Geranylgeranyl transferase 1 inhibitor GGTI‑298 enhances the anticancer effect of gefitinib. Mol Med Rep 2018; 18:4023-4029. [PMID: 30106149 DOI: 10.3892/mmr.2018.9371] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 05/11/2018] [Indexed: 02/05/2023] Open
Abstract
Dysregulation of epidermal growth factor receptor (EGFR) signaling is responsible for the resistance to EGFR tyrosine kinase inhibitors (TKIs), such as gefitinib and erlotinib, and is thereby associated with the progression of tumors in non‑small cell lung cancers (NSCLCs). Immunoblotting results revealed that geranylgeranyl transferase 1 inhibitor (GGTI)‑298, a geranylgeranyl transferase 1 inhibitor with potential antitumor effects, effectively inhibited the phosphorylation of EGFR and its downstream target protein kinase B (AKT). A combination of gefitinib and GGTI‑298 amplified the inhibition of the EGFR‑AKT signaling pathway. In addition, GGTI‑298 treatment produced a synergistic effect on the inhibition of proliferation as indicated by the combination index values of <1 when combined with gefitinib in the NSCLC cell lines HCC827 and A549. These synergistic effects were also observed to induce apoptosis and migration inhibition. Further mechanistic studies demonstrated that GGTI‑298 inhibited the activity of Ras homolog family member A (RhoA), and downregulation of RhoA with small interfering RNA impaired the phosphorylation of EGFR, which suggested that EGFR inhibition by GGTI‑298 may be exerted mainly through RhoA mediation. These results presented a novel, promising therapeutic strategy involving a combination of two drugs for targeting EGFR signaling in lung cancer.
Collapse
Affiliation(s)
- Bi-Sheng Liu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xin-Yu Dai
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hong-Wei Xia
- Laboratory of Molecular Targeted Therapy in Oncology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Huan-Ji Xu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Qiu-Lin Tang
- Laboratory of Molecular Targeted Therapy in Oncology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Qi-Yong Gong
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yong-Zhan Nie
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digest Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Feng Bi
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
25
|
Crosstalk between cancer cells and endothelial cells: implications for tumor progression and intervention. Arch Pharm Res 2018; 41:711-724. [DOI: 10.1007/s12272-018-1051-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/26/2018] [Indexed: 02/07/2023]
|
26
|
Cui Y, Zhou F, Bai L, Wei L, Tan J, Zeng Z, Song Q, Chen J, Huang N. SEMA4D-heparin Complexes Immobilized on Titanium Surfaces Have Anticoagulant, Cell-Migration-Promoting, and Immunoregulatory Effects. ACS Biomater Sci Eng 2018; 4:1598-1608. [PMID: 33445317 DOI: 10.1021/acsbiomaterials.8b00098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Soluble semaphorin 4D (SEMA4D) is a 120 kDa transmembrane protein, which belongs to the semaphorin family of axon guidance molecules that act primarily axonal repellents. SEMA4D elicits its migration-promoting and immunomodulatory effects through activation of PLXNB1 and CD72, respectively. In this study, SEMA4D combined with heparin were adsorbed onto cationic surfaces. The biocompatibility evaluation results indicated that the SEMA4D-heparin-modified surfaces displayed less platelet adhesion and activation, prolonged activated partial thromboplastin time (APTT), prothrombin time (PT) and thrombin time (TT) and reduced fibrinogen gamma chain (FGG) exposure and fibrinogen adhesion. Additionally, endothelial cells (ECs) showed improved adhesion density and proliferation activity on the SEMA4D-heparin-modified surfaces. Chemotactic and haptotaxis assays indicated a highly guided migration for ECs on the modified surfaces. The immunological tests revealed that the SEMA4D-heparin complexes had a positive immunomodulatory effect on macrophages and promoted macrophages polarization into M2 phenotypes. Overall, the results suggested that the SEMA4D-heparin complexes can be a potential therapeutic agent to promote tissue healing and accelerate in situ endothelialization with minimal side effects and positive immunomodulatory effect.
Collapse
Affiliation(s)
| | - Feng Zhou
- Institute of Aeronautics and Astronautics, University of Electronic Science and Technology of China, Chengdu 611731, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Derakhshandeh R, Sanadhya S, Lee Han K, Chen H, Goloubeva O, Webb TJ, Younis RH. Semaphorin 4D in human head and neck cancer tissue and peripheral blood: A dense fibrotic peri-tumoral stromal phenotype. Oncotarget 2018. [PMID: 29541402 PMCID: PMC5834246 DOI: 10.18632/oncotarget.24277] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The search for stromal biomarkers in carcinoma patients is a challenge in the field. Semaphorin 4D (Sema4D), known for its various developmental, physiological and pathological effects, plays a role in pro and anti-inflammatory responses. It is expressed in many epithelial tumors including head and neck squamous cell carcinoma (HNSCC). Recently, we found that HNSCC-associated Sema4D modulates an immune-suppressive, tumor-permissible environment by inducing the expansion of myeloid derived suppressor cells. The purpose of this study was to determine the value of Sema4D as a biomarker for the peri-tumoral stromal phenotype in human HNSCC. Our data showed Sema4D+ve/high tumor cells in 34% of the studied cohort with positive correlation to Stage III (p=0.0001). Sema4D+ve/high tumor cells correlated directly with dense fibrotic peri-tumoral stroma (p=0.0001) and inversely with infiltrate of Sema4D+ve/high tumor-associated inflammatory cells (TAIs) (p=0.01). Most of the Sema4D+ve/high TAIs were co-positive for the macrophage biomarker CD163. Knockdown of Sema4D in WSU-HN6 cells inhibited collagen production by fibroblasts, and decreased activated TGF-β1 levels in culture medium of HNSCC cell lines. In a stratification model of HNSCC using combined Sema4D and the programmed death ligand 1 (PDL-1), Sema4D+ve/high tumor cells represented a phenotype distinct from the PDL-1 positive tumors. Finally,Sema4D was detected in plasma of HNC patients at significantly higher levels (115.44, ± 39.37) compared to healthy donors (38.60± 12.73) (p <0.0001). In conclusion, we present a novel HNSCC tumor stratification model, based on the expression of the biomarker Sema4D. This model opens new avenues to novel targeted therapeutic strategies.
Collapse
Affiliation(s)
- Roshanak Derakhshandeh
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland Baltimore, Baltimore, Maryland, USA.,Department of Microbiology and Immunology, School of Medicine, University of Maryland Baltimore, Baltimore, Maryland, USA
| | - Sonia Sanadhya
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland Baltimore, Baltimore, Maryland, USA
| | - Kyu Lee Han
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland Baltimore, Baltimore, Maryland, USA
| | - Haiyan Chen
- Department of Dental Public Health, School of Dentistry, University of Maryland Baltimore, Baltimore, Maryland, USA
| | - Olga Goloubeva
- Department of Epidemiology and Public Health, School of Medicine, University of Maryland Baltimore, Baltimore, Maryland, USA.,The Marlene and Stewart Greenebaum Cancer Center, University of Maryland Baltimore, Baltimore, Maryland, USA
| | - Tonya J Webb
- Department of Microbiology and Immunology, School of Medicine, University of Maryland Baltimore, Baltimore, Maryland, USA.,The Marlene and Stewart Greenebaum Cancer Center, University of Maryland Baltimore, Baltimore, Maryland, USA
| | - Rania H Younis
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland Baltimore, Baltimore, Maryland, USA.,Oral Pathology Consultants, School of Dentistry, University of Maryland Baltimore, Baltimore, Maryland, USA.,Department of oral Pathology, Faculty of Dentistry, Alexandria University, Alexandria, Egypt.,The Marlene and Stewart Greenebaum Cancer Center, University of Maryland Baltimore, Baltimore, Maryland, USA
| |
Collapse
|
28
|
Neuronal expression patterns of the PlexinA family during zebrafish development. Gene Expr Patterns 2017; 27:56-66. [PMID: 29107805 DOI: 10.1016/j.gep.2017.10.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 10/23/2017] [Accepted: 10/25/2017] [Indexed: 12/15/2022]
Abstract
Plexins (Plxns) and Semaphorins (Semas) are key signaling molecules that regulate many aspects of development. Plxns are a family of transmembrane protein receptors that are activated upon extracellular binding by Semas. Activated Plxns trigger intracellular signaling cascades, which regulate a range of developmental processes, including axon guidance, neuronal positioning and vasculogenesis. Semas are a large family of both transmembrane and secreted signaling molecules, and show subtype specific binding to different Plxn family members. Each Plxn can play different roles in development, and so tightly regulated temporal and spatial expression of receptor subtypes is critical to ensure appropriate signaling. Here we elucidate the expression profiles of the plxnA family, plxnA1a, A1b, A2, A3 and A4 at 18, 24, 36, 48, 60 and 72 h post fertilization in the developing zebrafish. We show that PlxnA family members are expressed in neuronal tissues during zebrafish development, but exhibit key differences in expression within these tissues. We also highlight that plxnA1 has two genes in zebrafish, A1a and A1b, which show divergences in expression patterns during early development.
Collapse
|
29
|
Liu R, Lo L, Lay AJ, Zhao Y, Ting KK, Robertson EN, Sherrah AG, Jarrah S, Li H, Zhou Z, Hambly BD, Richmond DR, Jeremy RW, Bannon PG, Vadas MA, Gamble JR. ARHGAP18 Protects Against Thoracic Aortic Aneurysm Formation by Mitigating the Synthetic and Proinflammatory Smooth Muscle Cell Phenotype. Circ Res 2017; 121:512-524. [DOI: 10.1161/circresaha.117.310692] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 07/06/2017] [Accepted: 07/11/2017] [Indexed: 12/11/2022]
Abstract
Rationale:
Thoracic aortic aneurysm (TAA) is a potentially lethal condition, which can affect individuals of all ages. TAA may be complicated by the sudden onset of life-threatening dissection or rupture. The underlying mechanisms leading to TAA formation, particularly in the nonsyndromal idiopathic group of patients, are not well understood. Thus, identification of new genes and targets that are involved in TAA pathogenesis are required to help prevent and reverse the disease phenotype.
Objective:
Here we explore the role of ARHGAP18, a novel Rho GAP expressed by smooth muscle cells (SMCs), in the pathogenesis of TAA.
Methods and Results:
Using human and mouse aortic samples, we report that ARHGAP18 levels were significantly reduced in the SMC layer of aortic aneurysms.
Arhgap18
global knockout (
Arhgap18
−/
−
) mice exhibited a highly synthetic, proteolytic, and proinflammatory smooth muscle phenotype under basal conditions and when challenged with angiotensin II, developed TAA with increased frequency and severity compared with littermate controls. Chromatin immunoprecipitation studies revealed this phenotype is partly associated with strong enrichment of H3K4me3 and depletion of H3K27me3 at the
MMP2
and
TNF-α
promoters in
Arhgap18
-deficient SMC. We further show that TAA formation in the
Arhgap18
−/−
mice is associated with loss of Akt activation. The abnormal SMC phenotype observed in the
Arhgap18
−/−
mice can be partially rescued by pharmacological treatment with the mTORC1 inhibitor rapamycin, which reduces the synthetic and proinflammatory phenotype of
Arhgap18
-deficient SMC.
Conclusion:
We have identified
ARHGAP18
as a novel protective gene against TAA formation and define an additional target for the future development of treatments to limit TAA pathogenesis.
Collapse
Affiliation(s)
- Renjing Liu
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Lisa Lo
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Angelina J. Lay
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Yang Zhao
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Ka Ka Ting
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Elizabeth N. Robertson
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Andrew G. Sherrah
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Sorour Jarrah
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Haibo Li
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Zhaoxiong Zhou
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Brett D. Hambly
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - David R. Richmond
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Richmond W. Jeremy
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Paul G. Bannon
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Mathew A. Vadas
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Jennifer R. Gamble
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| |
Collapse
|
30
|
Ke Y, Dang E, Shen S, Zhang T, Qiao H, Chang Y, Liu Q, Wang G. Semaphorin4D Drives CD8 + T-Cell Lesional Trafficking in Oral Lichen Planus via CXCL9/CXCL10 Upregulations in Oral Keratinocytes. J Invest Dermatol 2017; 137:2396-2406. [PMID: 28760660 DOI: 10.1016/j.jid.2017.07.818] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/12/2017] [Accepted: 07/10/2017] [Indexed: 12/26/2022]
Abstract
Chemokine-mediated CD8+ T-cell recruitment is an essential but not well-established event for the persistence of oral lichen planus (OLP). Semaphorin 4D (Sema4D)/CD100 is implicated in immune dysfunction, chemokine modulation, and cell migration, which are critical aspects for OLP progression, but its implication in OLP pathogenesis has not been determined. In this study, we sought to explicate the effect of Sema4D on human oral keratinocytes and its capacity to drive CD8+ T-cell lesional trafficking via chemokine modulation. We found that upregulations of sSema4D in OLP tissues and blood were positively correlated with disease severity and activity. In vitro observation revealed that Sema4D induced C-X-C motif chemokine ligand 9/C-X-C motif chemokine ligand 10 production by binding to plexin-B1 via protein kinase B-NF-κB cascade in human oral keratinocytes, which elicited OLP CD8+ T-cell migration. We also confirmed using clinical samples that elevated C-X-C motif chemokine ligand 9/C-X-C motif chemokine ligand 10 levels were positively correlated with sSema4D levels in OLP lesions and serum. Notably, we determined matrix metalloproteinase-9 as a new proteolytic enzyme for the cleavage of sSema4D from the T-cell surface, which may contribute to the high levels of sSema4D in OLP lesions and serum. Our findings conclusively revealed an amplification feedback loop involving T cells, chemokines, and Sema4D-dependent signal that promotes OLP progression.
Collapse
Affiliation(s)
- Yao Ke
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China; Department of Oral Medicine, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Erle Dang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Shengxian Shen
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Tongmei Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hongjiang Qiao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yuqian Chang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Qing Liu
- Department of Oral Medicine, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
31
|
Carrasco P, Zuazo-Gaztelu I, Casanovas O. Sprouting strategies and dead ends in anti-angiogenic targeting of NETs. J Mol Endocrinol 2017; 59:R77-R91. [PMID: 28469004 DOI: 10.1530/jme-17-0029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 05/03/2017] [Indexed: 01/13/2023]
Abstract
Neuroendocrine tumors (NETs) are a heterogeneous group of neoplasms that arise from cells of the neuroendocrine system. NETs are characterized by being highly vascularized tumors that produce large amounts of proangiogenic factors. Due to their complexity and heterogeneity, progress in the development of successful therapeutic approaches has been limited. For instance, standard chemotherapy-based therapies have proven to be poorly selective for tumor cells and toxic for normal tissues. Considering the urge to develop an efficient therapy to treat NET patients, vascular targeting has been proposed as a new approach to block tumor growth. This review provides an update of the mechanisms regulating different components of vessels and their contribution to tumor progression in order to develop new therapeutic drugs. Following the description of classical anti-angiogenic therapies that target VEGF pathway, new angiogenic targets such as PDGFs, EGFs, FGFs and semaphorins are further explored. Based on recent research in the field, the combination of therapies that target multiple and different components of vessel formation would be the best approach to specifically target NETs and inhibit tumor growth.
Collapse
Affiliation(s)
- Patricia Carrasco
- Tumor Angiogenesis GroupProCURE, Catalan Institute of Oncology - IDIBELL, Barcelona, Spain
| | - Iratxe Zuazo-Gaztelu
- Tumor Angiogenesis GroupProCURE, Catalan Institute of Oncology - IDIBELL, Barcelona, Spain
| | - Oriol Casanovas
- Tumor Angiogenesis GroupProCURE, Catalan Institute of Oncology - IDIBELL, Barcelona, Spain
| |
Collapse
|
32
|
Abstract
During bone remodelling, osteoclasts induce chemotaxis of osteoblasts and yet maintain spatial segregation. We show that osteoclasts express the repulsive guidance factor Semaphorin 4D and induce contact inhibition of locomotion (CIL) in osteoblasts through its receptor Plexin-B1. To examine causality and elucidate how localized Plexin-B1 stimulation may spatiotemporally coordinate its downstream targets in guiding cell migration, we develop an optogenetic tool for Plexin-B1 designated optoPlexin. Precise optoPlexin activation at the leading edge of migrating osteoblasts readily induces local retraction and, unexpectedly, distal protrusions to steer cells away. These morphological changes are accompanied by reorganization of Myosin II, PIP3, adhesion and active Cdc42. We attribute the resultant repolarization to RhoA/ROCK-mediated redistribution of β-Pix, which activates Cdc42 and promotes protrusion. Thus, our data demonstrate a causal role of Plexin-B1 for CIL in osteoblasts and reveals a previously unknown effect of Semaphorin signalling on spatial distribution of an activator of cell migration.
Collapse
|
33
|
Bulloj A, Maminishkis A, Mizui M, Finnemann SC. Semaphorin4D-PlexinB1 Signaling Attenuates Photoreceptor Outer Segment Phagocytosis by Reducing Rac1 Activity of RPE Cells. Mol Neurobiol 2017. [PMID: 28624895 DOI: 10.1007/s12035-017-0649-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Semaphorins form a family of secreted and membrane-bound molecules that were identified originally as axonal guidance factors during neuronal development. Given their wide distribution in many including mature tissues, semaphorin 4D (sema4D) and its main functional receptor plexin B1 (plxnB1) are expected to fulfill additional functions that remain to be uncovered. A main characteristic of the plexin receptor family is its ability to reorganize the cytoskeleton. PlxnB1 specifically may directly interact with Rho family GTPases to regulate F-actin driven pathways in cells in culture. Diurnal clearance phagocytosis by the retinal pigment epithelium (RPE) of photoreceptor outer segment fragments (POS) is critical for photoreceptor function and longevity. In this process, rearrangement of RPE cytoskeletal F-actin via activation of the Rho family GTPase Rac1 is essential for POS internalization. Here, we show a novel role in POS phagocytosis by RPE cells in culture and in vivo for plexin B1 and its ligand sema4D. Exogenous sema4D abolishes POS internalization (but not binding) by differentiated RPE cells in culture by decreasing the GTP load of Rac1. In the rat eye, sema4D localizes to retinal photoreceptors, while PlxnB1 is expressed by neighboring RPE cells. At the peak of diurnal retinal phagocytosis after light onset, plxnB1 phosphorylation and sema4D levels are reduced in wild-type rat retina in situ but not in mutant RCS rat retina in which the RPE lacks phagocytic activity. Finally, increased POS phagosome content after light onset is observed in the RPE in situ of mice with either plxnB1 or sema4D gene deletion. Altogether, our results demonstrate a novel physiological function for sema4D/plxnB1 signaling in RPE phagocytosis serving as attenuating brake prior to light onset whose release enables the diurnal phagocytic burst.
Collapse
Affiliation(s)
- Ayelen Bulloj
- Department of Biological Sciences Center for Cancer, Genetic Diseases, and Gene Regulation, Fordham University, Larkin Hall, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Arvydas Maminishkis
- Section on Epithelial and Retinal Physiology and Disease, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Masayuki Mizui
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Silvia C Finnemann
- Department of Biological Sciences Center for Cancer, Genetic Diseases, and Gene Regulation, Fordham University, Larkin Hall, 441 East Fordham Road, Bronx, NY, 10458, USA.
| |
Collapse
|
34
|
LaGanke C, Samkoff L, Edwards K, Jung Henson L, Repovic P, Lynch S, Stone L, Mattson D, Galluzzi A, Fisher TL, Reilly C, Winter LA, Leonard JE, Zauderer M. Safety/tolerability of the anti-semaphorin 4D Antibody VX15/2503 in a randomized phase 1 trial. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2017. [PMID: 28642891 PMCID: PMC5473956 DOI: 10.1212/nxi.0000000000000367] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Objective: To evaluate the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of VX15/2503 in a randomized, single-dose, dose-escalation, double-blind, placebo-controlled study enrolling adult patients with MS. Methods: Single IV doses of VX15/2503 or placebo were administered. Ten patients each were randomized (4:1 randomization ratio) into 5 ascending dose cohorts of 1, 3, 6, 10, or 20 mg/kg. Safety, immunogenicity, PK/PD, MRI, ECG, and lymphocyte subset levels were evaluated. A Dose Escalation Safety Committee (DESC) approved each dose escalation. Results: VX15/2503 was well tolerated, and all participants completed the study. Antibody treatment–related adverse events were primarily grade 1 or 2 and included urinary tract infection (12.5%) and muscle weakness, contusion, and insomnia (each 7.5%). No dose-limiting toxicities were observed, and no maximum tolerated dose was determined. One subject (20 mg/kg) experienced disease relapse 3 months before study entry and exhibited a grade 3 (nonserious) increase in brain lesions by day 29, possibly related to VX15/2503. Twenty-nine patients exhibited human anti-humanized antibody responses; 5 with titer ≥100. No anti-VX15/2503 antibody responses were fully neutralizing. VX15/2503 Cmax, area under the time-concentration curve, and mean half-life increased with dose level; at 20 mg/kg, the T1/2 was 20 days. Cellular SEMA4D saturation occurred at serum antibody concentrations ≤0.3 μg/mL, resulting in decreased cSEMA4D expression. At 20 mg/kg, cSEMA4D saturation persisted for ≥155 days. Total sSEMA4D levels increased with dose level and declined with antibody clearance. Conclusions: These results support the continued investigation of VX15/2503 in neurodegenerative diseases. ClinicalTrials.gov identifier: NCT01764737. Classification of evidence: This study provides Class III evidence that anti-semaphorin 4D antibody VX15/2503 at various doses was safe and well tolerated vs placebo, although an increase in treatment-emergent adverse events in the treatment group could not be excluded (risk difference −0.7%, 95% CI −28.0% to 32.7%).
Collapse
Affiliation(s)
| | | | - Keith Edwards
- Author affiliations are provided at the end of the article
| | | | - Pavle Repovic
- Author affiliations are provided at the end of the article
| | - Sharon Lynch
- Author affiliations are provided at the end of the article
| | - Lael Stone
- Author affiliations are provided at the end of the article
| | - David Mattson
- Author affiliations are provided at the end of the article
| | - Aaron Galluzzi
- Author affiliations are provided at the end of the article
| | | | | | | | - John E Leonard
- Author affiliations are provided at the end of the article
| | | |
Collapse
|
35
|
Niu Y, Xia Y, Wang J, Shi X. O-GlcNAcylation promotes migration and invasion in human ovarian cancer cells via the RhoA/ROCK/MLC pathway. Mol Med Rep 2017; 15:2083-2089. [PMID: 28259907 PMCID: PMC5364967 DOI: 10.3892/mmr.2017.6244] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 11/11/2016] [Indexed: 02/06/2023] Open
Abstract
O-GlcNAcylation is a dynamic and reversible post-translational modification associated with the regulation of multiple cellular functions. The addition and removal of O-Linked β-N-acetylglucosamine (O-GlcNAc) on target proteins is catalyzed by O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), respectively. Accumulating evidence suggests that O-GlcNAcylation is associated with the malignancy of several types of human cancer. To investigate the effect of O-GlcNAcylation on ovarian cancer phenotypes, global O-GlcNAc levels were decreased by OGT silencing through RNA interference and increased by inhibiting OGA activity with Thiamet-G. Transwell assay results demonstrated that OGT silencing inhibited the migration and invasion of SKOV3 and 59M ovarian cells in vitro, while Thiamet-G treatment promoted migration and invasion. Furthermore, a pull-down assay and western blot analysis demonstrated that Thiamet-G treatment enhanced RhoA activity and the phosphorylation of the Rho-associated protein kinase (ROCK) substrate, myosin light chain (MLC), while OGT silencing attenuated RhoA activity and MLC phosphorylation. In addition, RhoA silencing via RNA interference and inhibition of ROCK activity with Y-27632 prevented Thiamet-G-induced increases in cell migration and invasion. These data suggest that O-GlcNAcylation augments the motility of ovarian cancer cells via the RhoA/ROCK/MLC signaling pathway. Therefore, O-GlcNAcylation may be a potential target for the diagnosis and treatment of ovarian cancer.
Collapse
Affiliation(s)
- Yichao Niu
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200126, P.R. China
| | - Ye Xia
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200126, P.R. China
| | - Jingyun Wang
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200126, P.R. China
| | - Xiaofei Shi
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200126, P.R. China
| |
Collapse
|
36
|
Abstract
Demyelination of central nervous system axons, associated with traumatic injury and demyelinating diseases such as multiple sclerosis, causes impaired neural transmission and ultimately axon degeneration. Consequently, extensive research has focused on signaling systems that promote myelinating activity of oligodendrocytes or promote production of new oligodendrocytes from oligodendrocyte progenitor cells. Many receptor systems, notably including growth factor receptors and G protein-coupled receptors, control myelination. A number of recent clinical trials target these receptor signaling pathways.
Collapse
Affiliation(s)
- Mark Bothwell
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington 98195;
| |
Collapse
|
37
|
Neufeld G, Mumblat Y, Smolkin T, Toledano S, Nir-Zvi I, Ziv K, Kessler O. The role of the semaphorins in cancer. Cell Adh Migr 2016; 10:652-674. [PMID: 27533782 PMCID: PMC5160032 DOI: 10.1080/19336918.2016.1197478] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Revised: 05/19/2016] [Accepted: 05/30/2016] [Indexed: 12/16/2022] Open
Abstract
The semaphorins were initially characterized as axon guidance factors, but have subsequently been implicated also in the regulation of immune responses, angiogenesis, organ formation, and a variety of additional physiological and developmental functions. The semaphorin family contains more then 20 genes divided into 7 subfamilies, all of which contain the signature sema domain. The semaphorins transduce signals by binding to receptors belonging to the neuropilin or plexin families. Additional receptors which form complexes with these primary semaphorin receptors are also frequently involved in semaphorin signaling. Recent evidence suggests that semaphorins also fulfill important roles in the etiology of multiple forms of cancer. Some semaphorins have been found to function as bona-fide tumor suppressors and to inhibit tumor progression by various mechanisms while other semaphorins function as inducers and promoters of tumor progression.
Collapse
Affiliation(s)
- Gera Neufeld
- Cancer Research and Vascular Biology Center, The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Yelena Mumblat
- Cancer Research and Vascular Biology Center, The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Tatyana Smolkin
- Cancer Research and Vascular Biology Center, The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Shira Toledano
- Cancer Research and Vascular Biology Center, The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Inbal Nir-Zvi
- Cancer Research and Vascular Biology Center, The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Keren Ziv
- Cancer Research and Vascular Biology Center, The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Ofra Kessler
- Cancer Research and Vascular Biology Center, The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
38
|
Neufeld G, Mumblat Y, Smolkin T, Toledano S, Nir-Zvi I, Ziv K, Kessler O. The semaphorins and their receptors as modulators of tumor progression. Drug Resist Updat 2016; 29:1-12. [DOI: 10.1016/j.drup.2016.08.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 07/31/2016] [Accepted: 08/23/2016] [Indexed: 12/16/2022]
|
39
|
Ikeya T, Maeda K, Nagahara H, Shibutani M, Iseki Y, Hirakawa K. The combined expression of Semaphorin4D and PlexinB1 predicts disease recurrence in colorectal cancer. BMC Cancer 2016; 16:525. [PMID: 27456345 PMCID: PMC4960918 DOI: 10.1186/s12885-016-2577-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 07/18/2016] [Indexed: 01/15/2023] Open
Abstract
Background Binding to Sema4D and PlexinB1 induce angiogenesis and invasive growth in colorectal cancer (CRC). The expression of Semaphorin4D (Sema4D) and PlexinB1 has been shown to be related to the prognosis of patients with various malignancies. However, the correlation between the expression of Sema4D and PlexinB1 and the relapse-free survival in patients with colorectal cancer remains controversial. Methods The study population included patients who underwent surgery for colorectal cancer (n = 226). The expression of Sema4D and PlexinB1 were analyzed by immunohistochemistry in tissue of stage I, II, and III colon cancers. Results The immunohistochemical staining of colorectal cancer tissue specimens revealed that 95 (42 %) and 105 (46.4 %) of the specimens were positive for Sema4D and PlexinB1. The expression of Sema4D and PlexinB1 respectively were both found to be significantly related to stage, depth of tumor invasion, lymph node metastasis, lymphatic invasion, and venous invasion, respectively. Sixty-three patients (27.9 %) expressed both Sema4D and PlexinB1. The positive expression of both Sema4D and PlexinB1 was found to be an independent risk factor for a worse survival (HR 1.079, CI 1.013–2.868; P = 0.044). Conclusion The combination of Sema4D and PlexinB1 protein detected by immunohistochemistry was therefore useful for predicting disease recurrence in CRC patients.
Collapse
Affiliation(s)
- Tetsuro Ikeya
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Abeno-ku, Osaka, Japan.
| | - Kiyoshi Maeda
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Abeno-ku, Osaka, Japan
| | - Hisashi Nagahara
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Abeno-ku, Osaka, Japan
| | - Masatsune Shibutani
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Abeno-ku, Osaka, Japan
| | - Yasuhito Iseki
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Abeno-ku, Osaka, Japan
| | - Kosei Hirakawa
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Abeno-ku, Osaka, Japan
| |
Collapse
|
40
|
Yao J, Gao P, Xu Y, Li Z. α-TEA inhibits the growth and motility of human colon cancer cells via targeting RhoA/ROCK signaling. Mol Med Rep 2016; 14:2534-40. [PMID: 27432222 PMCID: PMC4991732 DOI: 10.3892/mmr.2016.5525] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 05/03/2016] [Indexed: 01/01/2023] Open
Abstract
Colon or colorectal cancer is a common type of human cancer, which originates in the intestine crassum or the rectum. In the United States, colorectal cancer has one of the highest rates of cancer-related mortality. Investigating novel chemotherapeutic approaches is significant in the treatment of cancers, such as colorectal cancer. α-tocopherol ether-linked acetic acid (α-TEA) is a potent anticancer agent in multiple types of human cancer. However, its effect remains to be determined in colon cancer. In this study, HCT116 and SW480 human colon cancer cells were used to investigate the anticancer role of α-TEA. It was demonstrated that α-TEA inhibited cell proliferation, migration and invasion in colon cancer cells. Furthermore, it was shown that α-TEA downregulated the activity of RhoA and phosphorylated Rho-associated protein kinase (ROCK) substrate myosin light chain (MLC) using a pull-down assay and western blotting, respectively, implying that the RhoA/ROCK pathway is involved in α-TEA-mediated cell growth and motility inhibition. In order to confirm this hypothesis a RhoA inhibitor (clostridium botulinum C3 exoenzyme), a ROCK inhibitor (Y27632) and RhoA small interfering (si)RNA were applied to block RhoA/ROCK signaling. This resulted in the attenuation of MLC phosphorylation, and augmentation of α-TEA-mediated growth and motility inhibition in colon cancer cells. In conclusion, these results indicate that α-TEA inhibits growth and motility in colon cancer cells possibly by targeting RhoA/ROCK signaling. Moreover, combined with RhoA or ROCK inhibitors, α-TEA may exhibit a more effective inhibitory role in colon cancer.
Collapse
Affiliation(s)
- Jialin Yao
- Department of Pediatric Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Peng Gao
- Department of Pediatric Surgery, The Harbin Children's Hospital, Harbin, Heilongjiang 150001, P.R. China
| | - Yang Xu
- Department of Oncology, The Third Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Zhaozhu Li
- Department of Pediatric Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
41
|
Song GL, Jin CC, Zhao W, Tang Y, Wang YL, Li M, Xiao M, Li X, Li QS, Lin X, Chen WW, Kuang J. Regulation of the RhoA/ROCK/AKT/β-catenin pathway by arginine-specific ADP-ribosytransferases 1 promotes migration and epithelial-mesenchymal transition in colon carcinoma. Int J Oncol 2016; 49:646-56. [PMID: 27277835 DOI: 10.3892/ijo.2016.3539] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 04/05/2016] [Indexed: 11/06/2022] Open
Abstract
Arginine-specific ADP-ribosytransferases 1 (ART1) is able to modify the arginine of specific proteins by mono-ADP-ribosylation. We previously reported that the expression of ART1 in human colon adenocarcinoma tissues was higher than in adjacent tissues. Herein, we primarily revealed that ART1 could regulate the epithelial-mesenchymal transition (EMT) and, therefore, the development of colon carcinoma. In CT26 cells, which overexpressed ART1 by lentiviral transfection, the following were promoted: alterations of spindle-like non-polarization, expression of EMT inducers and mesenchymal markers, migration, invasion and adhesion. However, epithelial marker expression was decreased. Correspondingly, knockdown of ART1 in CT26 cells had the opposite effects. The effect of ART1 on EMT and carcinoma metastasis was also verified in a liver metastasis model of BALB/c mice. To further explore the molecular mechanism of ART1 in EMT, CT26 cells were treated with several specific inhibitors and gene silencing. Our data suggest that ART1 could regulate EMT by regulating the RhoA/ROCK1/AKT/β-catenin pathway and its downstream factors (snail1, vimentin, N-cadherin and E-cadherin) and that it therefore plays an important role in the progression of colon carcinoma.
Collapse
Affiliation(s)
- Guang-Lin Song
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Cong-Cong Jin
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Wei Zhao
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yi Tang
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Ya-Lan Wang
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Ming Li
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Ming Xiao
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xian Li
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Qing-Shu Li
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xiao Lin
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Wen-Wen Chen
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jing Kuang
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
42
|
Battistini C, Tamagnone L. Transmembrane semaphorins, forward and reverse signaling: have a look both ways. Cell Mol Life Sci 2016; 73:1609-22. [PMID: 26794845 PMCID: PMC11108563 DOI: 10.1007/s00018-016-2137-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 01/07/2016] [Accepted: 01/11/2016] [Indexed: 01/06/2023]
Abstract
Semaphorins are signaling molecules playing pivotal roles not only as axon guidance cues, but are also involved in the regulation of a range of biological processes, such as immune response, angiogenesis and invasive tumor growth. The main functional receptors for semaphorins are plexins, which are large single-pass transmembrane molecules. Semaphorin signaling through plexins-the "classical" forward signaling-affects cytoskeletal remodeling and integrin-dependent adhesion, consequently influencing cell migration. Intriguingly, semaphorins and plexins can interact not only in trans, but also in cis, leading to differentiated and highly regulated signaling outputs. Moreover, transmembrane semaphorins can also mediate a so-called "reverse" signaling, by acting not as ligands but rather as receptors, and initiate a signaling cascade through their own cytoplasmic domains. Semaphorin reverse signaling has been clearly demonstrated in fruit fly Sema1a, which is required to control motor axon defasciculation and target recognition during neuromuscular development. Sema1a invertebrate semaphorin is most similar to vertebrate class-6 semaphorins, and examples of semaphorin reverse signaling in mammalians have been described for these family members. Reverse signaling is also reported for other vertebrate semaphorin subsets, e.g. class-4 semaphorins, which bear potential PDZ-domain interaction motifs in their cytoplasmic regions. Therefore, thanks to their various signaling abilities, transmembrane semaphorins can play multifaceted roles both in developmental processes and in physiological as well as pathological conditions in the adult.
Collapse
Affiliation(s)
- Chiara Battistini
- Department of Oncology, University of Torino c/o IRCCS, Str. Prov. 142, 10060, Candiolo (TO), Italy
- Candiolo Cancer Institute, IRCCS-FPO, Str. Prov. 142, 10060, Candiolo (TO), Italy
| | - Luca Tamagnone
- Department of Oncology, University of Torino c/o IRCCS, Str. Prov. 142, 10060, Candiolo (TO), Italy.
- Candiolo Cancer Institute, IRCCS-FPO, Str. Prov. 142, 10060, Candiolo (TO), Italy.
| |
Collapse
|
43
|
Plexin-B1 indirectly affects glioma invasiveness and angiogenesis by regulating the RhoA/αvβ3 signaling pathway and SRPK1. Tumour Biol 2016; 37:11225-36. [PMID: 26944058 DOI: 10.1007/s13277-016-4849-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 01/13/2016] [Indexed: 01/15/2023] Open
Abstract
Gliomas are one of the most common primary brain tumors in adults. They display aggressive invasiveness, are highly vascular, and have a poor prognosis. Plexin-B1 is involved in numerous cellular processes, especially cellular migration and angiogenesis. However, the role and regulatory mechanisms of Plexin-B1 in gliomas are not understood and were thus investigated in this study. By using multiple and diverse experimental techniques, we investigated cell apoptosis, mitochondrial membrane potential, cell migration and invasion, angiogenesis, PI3K and Akt phosphorylation, and also the levels of SRPK1 and αvβ3 in glioma cells and animal glioma tissues. The results indicated that Plexin-B1 expression in glioma cell lines is increased compared to normal human astrocytes. Plexin-B1 mediates RhoA/integrin αvβ3 involved in the PI3K/Akt pathway and SRPK1 to influence the growth of glioma cell, angiogenesis, and motility in vitro and in vivo. Thus, Plexin-B1 signaling regulates the Rho/αvβ3/PI3K/Akt pathway and SRPK1, which are involved in glioma invasiveness and angiogenesis. Therefore, the new drug research should focus on Plexin-B1 as a target for the treatment of glioma invasion and angiogenesis.
Collapse
|
44
|
Yang YH, Buhamrah A, Schneider A, Lin YL, Zhou H, Bugshan A, Basile JR. Semaphorin 4D Promotes Skeletal Metastasis in Breast Cancer. PLoS One 2016; 11:e0150151. [PMID: 26910109 PMCID: PMC4766104 DOI: 10.1371/journal.pone.0150151] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 02/10/2016] [Indexed: 01/15/2023] Open
Abstract
Bone density is controlled by interactions between osteoclasts, which resorb bone, and osteoblasts, which deposit it. The semaphorins and their receptors, the plexins, originally shown to function in the immune system and to provide chemotactic cues for axon guidance, are now known to play a role in this process as well. Emerging data have identified Semaphorin 4D (Sema4D) as a product of osteoclasts acting through its receptor Plexin-B1 on osteoblasts to inhibit their function, tipping the balance of bone homeostasis in favor of resorption. Breast cancers and other epithelial malignancies overexpress Sema4D, so we theorized that tumor cells could be exploiting this pathway to establish lytic skeletal metastases. Here, we use measurements of osteoblast and osteoclast differentiation and function in vitro and a mouse model of skeletal metastasis to demonstrate that both soluble Sema4D and protein produced by the breast cancer cell line MDA-MB-231 inhibits differentiation of MC3T3 cells, an osteoblast cell line, and their ability to form mineralized tissues, while Sema4D-mediated induction of IL-8 and LIX/CXCL5, the murine homologue of IL-8, increases osteoclast numbers and activity. We also observe a decrease in the number of bone metastases in mice injected with MDA-MB-231 cells when Sema4D is silenced by RNA interference. These results are significant because treatments directed at suppression of skeletal metastases in bone-homing malignancies usually work by arresting bone remodeling, potentially leading to skeletal fragility, a significant problem in patient management. Targeting Sema4D in these cancers would not affect bone remodeling and therefore could elicit an improved therapeutic result without the debilitating side effects.
Collapse
Affiliation(s)
- Ying-Hua Yang
- Department of Oncology and Diagnostic Sciences, University of Maryland Dental School, Baltimore, Maryland, United States of America
| | - Asma Buhamrah
- Department of Oncology and Diagnostic Sciences, University of Maryland Dental School, Baltimore, Maryland, United States of America
| | - Abraham Schneider
- Department of Oncology and Diagnostic Sciences, University of Maryland Dental School, Baltimore, Maryland, United States of America
- Greenebaum Cancer Center, Baltimore, Maryland, United States of America
| | - Yi-Ling Lin
- Division of Diagnostic and Surgical Sciences, School of Dentistry, University of California Los Angeles, Los Angeles, California, United States of America
| | - Hua Zhou
- Department of Oncology and Diagnostic Sciences, University of Maryland Dental School, Baltimore, Maryland, United States of America
| | - Amr Bugshan
- Department of Oncology and Diagnostic Sciences, University of Maryland Dental School, Baltimore, Maryland, United States of America
| | - John R. Basile
- Department of Oncology and Diagnostic Sciences, University of Maryland Dental School, Baltimore, Maryland, United States of America
- Greenebaum Cancer Center, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
45
|
Meltzer S, Yadav S, Lee J, Soba P, Younger SH, Jin P, Zhang W, Parrish J, Jan LY, Jan YN. Epidermis-Derived Semaphorin Promotes Dendrite Self-Avoidance by Regulating Dendrite-Substrate Adhesion in Drosophila Sensory Neurons. Neuron 2016; 89:741-55. [PMID: 26853303 PMCID: PMC4760923 DOI: 10.1016/j.neuron.2016.01.020] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 05/27/2015] [Accepted: 01/11/2016] [Indexed: 01/15/2023]
Abstract
Precise patterning of dendritic arbors is critical for the wiring and function of neural circuits. Dendrite-extracellular matrix (ECM) adhesion ensures that the dendrites of Drosophila dendritic arborization (da) sensory neurons are properly restricted in a 2D space, and thereby facilitates contact-mediated dendritic self-avoidance and tiling. However, the mechanisms regulating dendrite-ECM adhesion in vivo are poorly understood. Here, we show that mutations in the semaphorin ligand sema-2b lead to a dramatic increase in self-crossing of dendrites due to defects in dendrite-ECM adhesion, resulting in a failure to confine dendrites to a 2D plane. Furthermore, we find that Sema-2b is secreted from the epidermis and signals through the Plexin B receptor in neighboring neurons. Importantly, we find that Sema-2b/PlexB genetically and physically interacts with TORC2 complex, Tricornered (Trc) kinase, and integrins. These results reveal a novel role for semaphorins in dendrite patterning and illustrate how epidermal-derived cues regulate neural circuit assembly.
Collapse
Affiliation(s)
- Shan Meltzer
- Howard Hughes Medical Institute, Departments of Physiology, Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Smita Yadav
- Howard Hughes Medical Institute, Departments of Physiology, Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jiae Lee
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Peter Soba
- Center for Molecular Neurobiology (ZMNH), University of Hamburg Medical School, Falkenried 94, 20251 Hamburg, Germany
| | - Susan H Younger
- Howard Hughes Medical Institute, Departments of Physiology, Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Peng Jin
- Howard Hughes Medical Institute, Departments of Physiology, Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Wei Zhang
- Howard Hughes Medical Institute, Departments of Physiology, Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jay Parrish
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Lily Yeh Jan
- Howard Hughes Medical Institute, Departments of Physiology, Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yuh-Nung Jan
- Howard Hughes Medical Institute, Departments of Physiology, Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
46
|
Andrews WD, Davidson K, Tamamaki N, Ruhrberg C, Parnavelas JG. Altered proliferative ability of neuronal progenitors in PlexinA1 mutant mice. J Comp Neurol 2015; 524:518-34. [PMID: 25975775 PMCID: PMC4737253 DOI: 10.1002/cne.23806] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 04/30/2015] [Accepted: 04/30/2015] [Indexed: 12/12/2022]
Abstract
Cortical interneurons are generated predominantly in the medial ganglionic eminence (MGE) and migrate through the ventral and dorsal telencephalon before taking their final positions within the developing cortical plate. Previously we demonstrated that interneurons from Robo1 knockout (Robo1(-/-)) mice contain reduced levels of neuropilin 1 (Nrp1) and PlexinA1 receptors, rendering them less responsive to the chemorepulsive actions of semaphorin ligands expressed in the striatum and affecting their course of migration (Hernandez-Miranda et al. [2011] J. Neurosci. 31:6174-6187). Earlier studies have highlighted the importance of Nrp1 and Nrp2 in interneuron migration, and here we assess the role of PlexinA1 in this process. We observed significantly fewer cells expressing the interneuron markers Gad67 and Lhx6 in the cortex of PlexinA1(-/-) mice compared with wild-type littermates at E14.5 and E18.5. Although the level of apoptosis was similar in the mutant and control forebrain, proliferation was significantly reduced in the former. Furthermore, progenitor cells in the MGE of PlexinA1(-/-) mice appeared to be poorly anchored to the ventricular surface and showed reduced adhesive properties, which may account for the observed reduction in proliferation. Together our data uncover a novel role for PlexinA1 in forebrain development.
Collapse
Affiliation(s)
- William D Andrews
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, United Kingdom
| | - Kathryn Davidson
- Division of Visual Science and Molecular Genetics, Institute of Ophthalmology, University College London, London, WC1E 6BT, United Kingdom
| | - Nobuaki Tamamaki
- Department of Morphological Neural Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860 0862, Japan
| | - Christiana Ruhrberg
- Division of Visual Science and Molecular Genetics, Institute of Ophthalmology, University College London, London, WC1E 6BT, United Kingdom
| | - John G Parnavelas
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, United Kingdom
| |
Collapse
|
47
|
PRUNE2 is a human prostate cancer suppressor regulated by the intronic long noncoding RNA PCA3. Proc Natl Acad Sci U S A 2015; 112:8403-8. [PMID: 26080435 DOI: 10.1073/pnas.1507882112] [Citation(s) in RCA: 203] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Prostate cancer antigen 3 (PCA3) is the most specific prostate cancer biomarker but its function remains unknown. Here we identify PRUNE2, a target protein-coding gene variant, which harbors the PCA3 locus, thereby classifying PCA3 as an antisense intronic long noncoding (lnc)RNA. We show that PCA3 controls PRUNE2 levels via a unique regulatory mechanism involving formation of a PRUNE2/PCA3 double-stranded RNA that undergoes adenosine deaminase acting on RNA (ADAR)-dependent adenosine-to-inosine RNA editing. PRUNE2 expression or silencing in prostate cancer cells decreased and increased cell proliferation, respectively. Moreover, PRUNE2 and PCA3 elicited opposite effects on tumor growth in immunodeficient tumor-bearing mice. Coregulation and RNA editing of PRUNE2 and PCA3 were confirmed in human prostate cancer specimens, supporting the medical relevance of our findings. These results establish PCA3 as a dominant-negative oncogene and PRUNE2 as an unrecognized tumor suppressor gene in human prostate cancer, and their regulatory axis represents a unique molecular target for diagnostic and therapeutic intervention.
Collapse
|
48
|
Families of microRNAs Expressed in Clusters Regulate Cell Signaling in Cervical Cancer. Int J Mol Sci 2015; 16:12773-90. [PMID: 26057746 PMCID: PMC4490472 DOI: 10.3390/ijms160612773] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Revised: 05/28/2015] [Accepted: 05/29/2015] [Indexed: 01/15/2023] Open
Abstract
Tumor cells have developed advantages to acquire hallmarks of cancer like apoptosis resistance, increased proliferation, migration, and invasion through cell signaling pathway misregulation. The sequential activation of genes in a pathway is regulated by miRNAs. Loss or gain of miRNA expression could activate or repress a particular cell axis. It is well known that aberrant miRNA expression is well recognized as an important step in the development of cancer. Individual miRNA expression is reported without considering that miRNAs are grouped in clusters and may have similar functions, such as the case of clusters with anti-oncomiRs (23b~27b~24-1, miR-29a~29b-1, miR-29b-2~29c, miR-99a~125b-2, miR-99b~125a, miR-100~125b-1, miR-199a-2~214, and miR-302s) or oncomiRs activity (miR-1-1~133a-2, miR-1-2~133a-1, miR-133b~206, miR-17~92, miR-106a~363, miR183~96~182, miR-181a-1~181b-1, and miR-181a-2~181b-2), which regulated mitogen-activated protein kinases (MAPK), phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K), NOTCH, proteasome-culling rings, and apoptosis cell signaling. In this work we point out the pathways regulated by families of miRNAs grouped in 20 clusters involved in cervical cancer. Reviewing how miRNA families expressed in cluster-regulated cell path signaling will increase the knowledge of cervical cancer progression, providing important information for therapeutic, diagnostic, and prognostic methodology design.
Collapse
|
49
|
Plexin-B1 and semaphorin 4D cooperate to promote cutaneous squamous cell carcinoma cell proliferation, migration and invasion. J Dermatol Sci 2015; 79:127-36. [PMID: 26051877 DOI: 10.1016/j.jdermsci.2015.05.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 04/21/2015] [Accepted: 05/07/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND OBJECTIVES Semaphorin 4D (Sema4D) and its receptor, Plexin-B1, are involved in the pathogenesis of squamous cell carcinoma (SCC) by mediating angiogenesis or perineural invasion through the interaction between Sema4D expression on SCC cells and Plexin-B1 expression on endothelial cells or nerves. Plexin-B1 was also recently found to be expressed on SCC cells. Plexin-B1 expression on several types of tumor cells could mediate various, and occasionally opposing, effects, including tumor cell survival, proliferation, angiogenesis, invasion, and metastasis. However, whether Sema4D exerts paracrine or autocrine effects on SCC via Plexin-B1 remains unclear. OBJECTIVES The aim of this study is to explore the effects of Sema4D/Plexin-B1 interaction on SCC via Plexin-B1 expressed on the tumor cells. METHODS In the present study, we detected the expression of Plexin-B1 and Sema4D in cutaneous SCC (cSCC) tissues and in the cSCC cell line A431 and analyzed the effects of the Sema4D/Plexin-B1 interaction on cSCC cell proliferation, migration, and invasion, as well as on the signaling pathway downstream of Plexin-B1. RESULTS We observed significantly increased Plexin-B1 and Sema4D expression in keratinocytes in cSCC lesions and in A431 cells compared with that in normal skin tissue and in non-malignant keratinocytes. Plexin-B1 silencing reduced the growth, proliferation, migration, and invasion of A431 cells and inhibited the phosphorylation of Akt and extracellular signal-regulated protein kinase (Erk). Soluble recombinant Sema4D promoted the growth, proliferation, migration, and invasion of A431 cells; Akt and Erk phosphorylation is also involved in these processes with a Plexin-B1 dependent manner. CONCLUSION Plexin-B1 induces cSCC cell proliferation, migration, and invasion by interacting with Sema4D. Plexin-B1 might serve as a useful biomarker and/or as a novel therapeutic target for cSCC.
Collapse
|
50
|
Xia Y, Cai XY, Fan JQ, Zhang LL, Ren JH, Chen J, Li ZY, Zhang RG, Zhu F, Wu G. Rho Kinase Inhibitor Fasudil Suppresses the Vasculogenic Mimicry of B16 Mouse Melanoma Cells Both In Vitro and In Vivo. Mol Cancer Ther 2015; 14:1582-90. [PMID: 25934709 DOI: 10.1158/1535-7163.mct-14-0523] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 04/24/2015] [Indexed: 11/16/2022]
Abstract
The aim of this study was to investigate the biologic role of the Rho kinase inhibitor fasudil in the vasculogenic mimicry (VM) of B16 mouse melanoma cells. It was previously reported that RhoA plays a critical role in angiogenesis by coordinating endothelial cell cytoskeleton remodeling and promoting endothelial cell motility. Although RhoA has been implicated in the regulation of angiogenesis, little has been described regarding its control of these tumor cell-lined channels. In this study, we established an in vitro model of VM using 3-dimensional cell culturing of mouse B16 melanoma cells and studied VM in vivo by transplanting B16 cells into C57/BL mice. Next, we explored the effect of RhoA and Rho-associated, coiled-coil containing protein kinase (ROCK) on VM formation using the Rho kinase inhibitor fasudil. We provide direct evidence that fasudil leads to reduced vascular-like channels in Matrigel. Additional experiments suggested that fasudil prevents both initial cellular architecture changes and cell migration in vitro. Finally, we provide in-depth evidence for the underlying mechanisms of fasudil-induced VM destruction using the Rho-GTPase agonist lysophosphatidic acid. In vivo studies revealed that fasudil reduced B16 melanoma cell xenograft tumor growth without causing significant toxicity in mice. Fasudil-treated tumors also displayed fewer VM channels. These results suggest that fasudil may be an emerging therapeutic option for targeting cancer VM.
Collapse
Affiliation(s)
- Yun Xia
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xian-Yi Cai
- Department of Orthopaedic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ji-Quan Fan
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li-Ling Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing-Hua Ren
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhen-Yu Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui-Guang Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Zhu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|