1
|
Królikowska K, Kurman N, Błaszczak K, Ławicki S, Gudowska-Sawczuk M, Zajkowska M. The Significance of Neuropilins in Gastrointestinal Cancers. Int J Mol Sci 2025; 26:4937. [PMID: 40430075 DOI: 10.3390/ijms26104937] [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: 03/26/2025] [Revised: 05/19/2025] [Accepted: 05/20/2025] [Indexed: 05/29/2025] Open
Abstract
Cancers represent a significant global health concern, being among the most prevalent malignancies and contributing substantially to morbidity and mortality rates. Notably, colorectal, gastric, pancreatic, and liver cancers are the most frequently diagnosed among these malignancies. The pathogenesis of gastrointestinal (GI) cancers is multifactorial, encompassing a complex interplay of genetic predispositions, environmental exposures, and lifestyle choices. Despite advances in diagnostic approaches and therapeutic strategies, existing treatment modalities, particularly in the advanced stages of these cancers, remain ineffective. Recent research efforts have increasingly focused on the identification and characterization of novel biomarkers that could enhance both the detection and treatment of gastrointestinal cancers. One particularly promising area of investigation involves neuropilins (NRPs). NRPs are involved in essential biological processes such as angiogenesis, cellular migration, and tumor cell-microenvironment interactions, all of which promote tumor progression and contribute to the development of treatment resistance. Overexpression of neuropilins has been linked to poor prognosis in patients, implying that they could be useful in diagnosis and serve as targets for molecular treatment. Recent research also suggests that inhibiting neuropilin activity may slow tumor growth and inhibit angiogenic processes, opening up new possibilities for targeted therapeutic techniques in the treatment of gastrointestinal malignancies.
Collapse
Affiliation(s)
- Kinga Królikowska
- Department of Population Medicine and Lifestyle Diseases Prevention, The Faculty of Medicine, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Natalia Kurman
- Department of Population Medicine and Lifestyle Diseases Prevention, The Faculty of Medicine, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Katarzyna Błaszczak
- Department of Population Medicine and Lifestyle Diseases Prevention, The Faculty of Medicine, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Sławomir Ławicki
- Department of Population Medicine and Lifestyle Diseases Prevention, The Faculty of Medicine, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Monika Gudowska-Sawczuk
- Department of Biochemical Diagnostics, Medical University of Bialystok, Waszyngtona 15A St., 15-269 Bialystok, Poland
| | - Monika Zajkowska
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, Waszyngtona 15A St., 15-269 Bialystok, Poland
| |
Collapse
|
2
|
Lee Y, Tukei KL, Fang Y, Kuila S, Liu X, Imoukhuede PI. Integrative analysis of angiogenic signaling in obesity: capillary features and VEGF binding kinetics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.23.630107. [PMID: 39763822 PMCID: PMC11703262 DOI: 10.1101/2024.12.23.630107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
Obesity is a global health crisis, with its prevalence particularly severe in the United States, where over 42% of adults are classified as obese. Obesity is driven by complex molecular and tissue-level mechanisms that remain poorly understood. Among these, angiogenesis-primarily mediated by vascular endothelial growth factor (VEGF-A)-is critical for adipose tissue expansion but presents unique challenges for therapeutic targeting due to its intricate regulation. Systems biology approaches have advanced our understanding of VEGF-A signaling in vascular diseases, but their application to obesity is limited by scattered and sometimes contradictory data. To address this gap, we performed a comprehensive analysis of the existing literature to synthesize key findings, standardize data, and provide a holistic perspective on the adipose vascular microenvironment. The data mining revealed five key findings: (1) obesity increases adipocyte size by 78%; (2) vessel density in adipose tissue decreases by 51% in obese mice, with vessels being 47-58% smaller and 4-9 times denser in comparison with tumor vessels; (3) capillary basement membrane thickness remains similar regardless of obesity; (4) VEGF-A shows the strongest binding affinity for VEGFR1, with four times stronger affinity for VEGFR2 than for NRP1; and (5) binding affinities measured by radioligand binding assay and surface plasmon resonance (SPR) are significantly different. These consolidated findings provide essential parameters for systems biology modeling, new insights into obesity-induced changes in adipose tissue, and a foundation for developing angiogenesis-targeting therapies for obesity.
Collapse
|
3
|
Yang Y, Guan W, Sheng XM, Gu HJ. Role of Semaphorin 3A in common psychiatric illnesses such as schizophrenia, depression, and anxiety. Biochem Pharmacol 2024; 226:116358. [PMID: 38857830 DOI: 10.1016/j.bcp.2024.116358] [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: 04/23/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024]
Abstract
With societal development and an ageing population, psychiatric disorders have become a common cause of severe and long-term disability and socioeconomic burdens worldwide. Semaphorin 3A (Sema-3A) is a secreted glycoprotein belonging to the semaphorin family. Sema-3A is well known as an axon guidance factor in the neuronal system and a potent immunoregulator at all stages of the immune response. It is reported to have various biological functions and is involved in many human diseases, including autoimmune diseases, angiocardiopathy, osteoporosis, and tumorigenesis. The signals of sema-3A involved in the pathogenesis of these conditions, are transduced through its cognate receptors and diverse downstream signalling pathways. An increasing number of studies show that sema-3A plays important roles in synaptic and dendritic development, which are closely associated with the pathophysiological mechanisms of psychiatric disorders, including schizophrenia, depression, and autism, suggesting the involvement of sema-3A in the pathogenesis of mental diseases. This indicates that mutations in sema-3A and alterations in its receptors and signalling may compromise neurodevelopment and predispose patients to these disorders. However, the role of sema-3A in psychiatric disorders, particularly in regulating neurodevelopment, remains elusive. In this review, we summarise the recent progress in understanding sema-3A in the pathogenesis of mental diseases and highlight sema-3A as a potential target for the prevention and treatment of these diseases.
Collapse
Affiliation(s)
- Yang Yang
- Department of Pharmacy, Affiliated Tumor Hospital of Nantong University/Nantong Tumor Hospital, China
| | - Wei Guan
- Department of Pharmacology, Pharmacy College, Nantong University, China
| | - Xiao-Ming Sheng
- Department of Trauma Center, Affiliated Hospital of Nantong University, China
| | - Hai-Juan Gu
- Department of Pharmacy, Affiliated Tumor Hospital of Nantong University/Nantong Tumor Hospital, China.
| |
Collapse
|
4
|
Toussaint K, Appert-Collin A, Vanalderwiert L, Bour C, Terryn C, Spenlé C, Van Der Heyden M, Roumieux M, Maurice P, Romier-Crouzet B, Sartelet H, Duca L, Blaise S, Bennasroune A. Inhibition of neuraminidase-1 sialidase activity by interfering peptides impairs insulin receptor activity in vitro and glucose homeostasis in vivo. J Biol Chem 2024; 300:107316. [PMID: 38663826 PMCID: PMC11167521 DOI: 10.1016/j.jbc.2024.107316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 03/18/2024] [Accepted: 04/09/2024] [Indexed: 06/02/2024] Open
Abstract
Neuraminidases (NEUs) also called sialidases are glycosidases which catalyze the removal of terminal sialic acid residues from glycoproteins, glycolipids, and oligosaccharides. Mammalian NEU-1 participates in regulation of cell surface receptors such as insulin receptor (IR), epithelial growth factor receptor, low-density lipoprotein receptor, and toll-like receptor 4. At the plasma membrane, NEU-1 can be associated with the elastin-binding protein and the carboxypeptidase protective protein/cathepsin A to constitute the elastin receptor complex. In this complex, NEU-1 is essential for elastogenesis, signal transduction through this receptor and for biological effects of the elastin-derived peptides on atherosclerosis, thrombosis, insulin resistance, nonalcoholic steatohepatitis, and cancers. This is why research teams are developing inhibitors targeting this sialidase. Previously, we developed interfering peptides to inhibit the dimerization and the activation of NEU-1. In this study, we investigated the effects of these peptides on IR activation in vitro and in vivo. Using cellular overexpression and endogenous expression models of NEU-1 and IR (COS-7 and HepG2 cells, respectively), we have shown that interfering peptides inhibit NEU-1 dimerization and sialidase activity which results in a reduction of IR phosphorylation. These results demonstrated that NEU-1 positively regulates IR phosphorylation and activation in our conditions. In vivo, biodistribution study showed that interfering peptides are well distributed in mice. Treatment of C57Bl/6 mice during 8 weeks with interfering peptides induces a hyperglycemic effect in our experimental conditions. Altogether, we report here that inhibition of NEU-1 sialidase activity by interfering peptides decreases IR activity in vitro and glucose homeostasis in vivo.
Collapse
Affiliation(s)
- Kevin Toussaint
- Université de Reims Champagne-Ardenne, CNRS, MEDyC, Reims, France
| | | | | | - Camille Bour
- Université de Reims Champagne-Ardenne, CNRS, MEDyC, Reims, France
| | | | - Caroline Spenlé
- UMR7242 Biotechnology and Cell Signalling, Centre National de la Recherche Scientifique, Strasbourg Drug Discovery and Development Institute (IMS), University of Strasbourg, Illkirch-Graffenstaden, France
| | | | | | - Pascal Maurice
- Université de Reims Champagne-Ardenne, CNRS, MEDyC, Reims, France
| | | | - Hervé Sartelet
- Université de Reims Champagne-Ardenne, CNRS, MEDyC, Reims, France
| | - Laurent Duca
- Université de Reims Champagne-Ardenne, CNRS, MEDyC, Reims, France
| | - Sébastien Blaise
- Université de Reims Champagne-Ardenne, CNRS, MEDyC, Reims, France.
| | - Amar Bennasroune
- Université de Reims Champagne-Ardenne, CNRS, MEDyC, Reims, France.
| |
Collapse
|
5
|
Dhupar R, Powers AA, Eisenberg SH, Gemmill RM, Bardawil CE, Udoh HM, Cubitt A, Nangle LA, Soloff AC. Orchestrating Resilience: How Neuropilin-2 and Macrophages Contribute to Cardiothoracic Disease. J Clin Med 2024; 13:1446. [PMID: 38592275 PMCID: PMC10934188 DOI: 10.3390/jcm13051446] [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: 01/16/2024] [Revised: 02/21/2024] [Accepted: 02/24/2024] [Indexed: 04/10/2024] Open
Abstract
Immunity has evolved to balance the destructive nature of inflammation with wound healing to overcome trauma, infection, environmental insults, and rogue malignant cells. The inflammatory response is marked by overlapping phases of initiation, resolution, and post-resolution remodeling. However, the disruption of these events can lead to prolonged tissue damage and organ dysfunction, resulting long-term disease states. Macrophages are the archetypic phagocytes present within all tissues and are important contributors to these processes. Pleiotropic and highly plastic in their responses, macrophages support tissue homeostasis, repair, and regeneration, all while balancing immunologic self-tolerance with the clearance of noxious stimuli, pathogens, and malignant threats. Neuropilin-2 (Nrp2), a promiscuous co-receptor for growth factors, semaphorins, and integrins, has increasingly been recognized for its unique role in tissue homeostasis and immune regulation. Notably, recent studies have begun to elucidate the role of Nrp2 in both non-hematopoietic cells and macrophages with cardiothoracic disease. Herein, we describe the unique role of Nrp2 in diseases of the heart and lung, with an emphasis on Nrp2 in macrophages, and explore the potential to target Nrp2 as a therapeutic intervention.
Collapse
Affiliation(s)
- Rajeev Dhupar
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (R.D.); (H.M.U.)
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Surgical and Research Services, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA
| | - Amy A. Powers
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (R.D.); (H.M.U.)
| | - Seth H. Eisenberg
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (R.D.); (H.M.U.)
| | - Robert M. Gemmill
- Division of Hematology/Oncology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA;
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Charles E. Bardawil
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (R.D.); (H.M.U.)
| | - Hannah M. Udoh
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (R.D.); (H.M.U.)
| | - Andrea Cubitt
- aTyr Pharma, San Diego, CA 92121, USA; (A.C.); (L.A.N.)
| | | | - Adam C. Soloff
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (R.D.); (H.M.U.)
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Surgical and Research Services, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA
| |
Collapse
|
6
|
Yano Y, Morise T, Matsuzaki K. Effects of Gly Residue and Cholesterol on the GXXXG-Mediated Parallel Association of Transmembrane Helices: A Single-Pair FRET Study. Chembiochem 2022; 23:e202200160. [PMID: 36229427 DOI: 10.1002/cbic.202200160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 10/12/2022] [Indexed: 01/25/2023]
Abstract
Small residue-mediated interhelical packing is ubiquitous in helical membrane proteins: however, the lipid dependence of its stability remains unclear. We previously demonstrated that the introduction of a GXXXG sequence in the middle of de novo-designed (AALALAA)3 helices (AALALAA AGLALGA AALALAA) facilitated their dimerization, which was abolished by cholesterol. Here single-pair FRET measurements revealed that a longer GXXXGXXXG segment (AALALAA A GLALGA AAGALAA) promoted helix dimerization in POPC/cholesterol bilayers, but not without cholesterol. The predicted dimer structures and degrees of helix packing suggested that helix dimers with small (∼10°) and large (∼55°) crossing angles were only stabilized in POPC and POPC/cholesterol membranes, respectively. A steric hindrance in the dimer interface and the large flexibility of helices prevented the formation of stable dimers. Therefore, amino acid sequences and lipid compositions distinctively constrain stable dimer structures in membranes.
Collapse
Affiliation(s)
- Yoshiaki Yano
- School of Pharmacy and Pharmaceutical Sciences, Mukogawa Woman's University, Nishinomiya, 663-8179, Japan
| | - Takayuki Morise
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
| | - Katsumi Matsuzaki
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
| |
Collapse
|
7
|
Huang Y, Wang Y, Xu D, Xiao L, Qin W, Liu B, Yuan X. Characterization of the SARS-CoV-2 co-receptor NRP1 expression profiles in healthy people and cancer patients: Implication for susceptibility to COVID-19 disease and potential therapeutic strategy. Front Genet 2022; 13:995736. [PMID: 36338984 PMCID: PMC9627153 DOI: 10.3389/fgene.2022.995736] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/10/2022] [Indexed: 08/03/2023] Open
Abstract
Neuropilin-1 (NRP1) is a transmembrane protein involved in many physiological and pathological processes, and it functions as a co-receptor to facilitate the entry of SARS-CoV-2 into host cells. Therefore, it is critical to predict the susceptibility to SARS-CoV-2 and prognosis after infection among healthy people and cancer patients based on expression of NRP1. In the current study, we analyzed the conservation and isoform of NRP1 using public databases. NRP1 expression landscape in healthy people, COVID-19 patients, and cancer patients at both bulk and single-cell RNA-seq level was also depicted. We also analyzed the relationship between tissue-specific NRP1 expression and overall survival (OS), as well as tumor immune environment at a pan-cancer level, providing a comprehensive insight into the relationship between the vulnerability to SARS-CoV-2 infection and tumorigenesis. In conclusion, we identified NRP1 as a potential biomarker in predicting susceptibility to SARS-CoV-2 infection among healthy people and cancer patients.
Collapse
Affiliation(s)
| | | | | | | | - Wan Qin
- *Correspondence: Wan Qin, ; Bo Liu, ; Xianglin Yuan,
| | - Bo Liu
- *Correspondence: Wan Qin, ; Bo Liu, ; Xianglin Yuan,
| | - Xianglin Yuan
- *Correspondence: Wan Qin, ; Bo Liu, ; Xianglin Yuan,
| |
Collapse
|
8
|
Investigating the role of peptides in effective therapies against cancer. Cancer Cell Int 2022; 22:139. [PMID: 35346211 PMCID: PMC8962089 DOI: 10.1186/s12935-022-02553-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/14/2022] [Indexed: 12/03/2022] Open
Abstract
Early diagnosis and effective treatment of cancer are challenging. To diagnose and treat cancer effectively and to overcome these challenges, fundamental innovations in traditional diagnosis and therapy are necessary. Peptides can be very helpful in this regard due to their potential and diversity. To enhance the therapeutic potential of peptides, their limitations must be properly identified and their structures engineered and modified for higher efficiency. Promoting the bioavailability and stability of peptides is one of the main concerns. Peptides can also be effective in different areas of targeting, alone or with the help of other therapeutic agents. There has been a lot of research in this area, and the potential for variability of peptides will continue to improve this process. Another promising area in which peptides can help treat cancer is peptide vaccines, which are undergoing promising research, and high throughput technologies can lead to fundamental changes in this area. Peptides have been effective in almost all areas of cancer treatment, and some have even gone through clinical phases. However, many barriers need to be overcome to reach the desired point. The purpose of this review is to evaluate the mechanisms associated with peptides in the diagnosis and treatment of cancer. Therefore, related studies in this area will be discussed.
Collapse
|
9
|
Binamé F, Pham-Van LD, Bagnard D. Manipulating oligodendrocyte intrinsic regeneration mechanism to promote remyelination. Cell Mol Life Sci 2021; 78:5257-5273. [PMID: 34019104 PMCID: PMC11073109 DOI: 10.1007/s00018-021-03852-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/14/2021] [Accepted: 05/08/2021] [Indexed: 02/06/2023]
Abstract
In demyelinated lesions, astrocytes, activated microglia and infiltrating macrophages secrete several factors regulating oligodendrocyte precursor cells' behaviour. What appears to be the initiation of an intrinsic mechanism of myelin repair is only leading to partial recovery and inefficient remyelination, a process worsening over the course of the disease. This failure is largely due to the concomitant accumulation of inhibitory cues in and around the lesion sites opposing to growth promoting factors. Here starts a complex game of interactions between the signalling pathways controlling oligodendrocytes migration or differentiation. Receptors of positive or negative cues are modulating Ras, PI3K or RhoGTPases pathways acting on oligodendrocyte cytoskeleton remodelling. From the description of this intricate signalling network, this review addresses the extent to which the modulation of the global response to inhibitory cues may pave the route towards novel therapeutic approaches for myelin repair.
Collapse
Affiliation(s)
- Fabien Binamé
- INSERM U1119, Biopathology of Myelin, Neuroprotection and Therapeutic Strategy (BMNST Lab), Labex Medalis, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Pôle API, Ecole Supérieure de Biotechnologie, 300 Boulevard Sébastien Brant, 67412, Illkirch, France
| | - Lucas D Pham-Van
- INSERM U1119, Biopathology of Myelin, Neuroprotection and Therapeutic Strategy (BMNST Lab), Labex Medalis, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Pôle API, Ecole Supérieure de Biotechnologie, 300 Boulevard Sébastien Brant, 67412, Illkirch, France
| | - Dominique Bagnard
- INSERM U1119, Biopathology of Myelin, Neuroprotection and Therapeutic Strategy (BMNST Lab), Labex Medalis, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Pôle API, Ecole Supérieure de Biotechnologie, 300 Boulevard Sébastien Brant, 67412, Illkirch, France.
| |
Collapse
|
10
|
Chekol Abebe E, Mengie Ayele T, Tilahun Muche Z, Asmamaw Dejenie T. Neuropilin 1: A Novel Entry Factor for SARS-CoV-2 Infection and a Potential Therapeutic Target. Biologics 2021; 15:143-152. [PMID: 33986591 PMCID: PMC8110213 DOI: 10.2147/btt.s307352] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/19/2021] [Indexed: 12/12/2022]
Abstract
The novel coronavirus disease 2019 (COVID-19) pandemic is severely challenging the healthcare systems and economies of the world, which urgently demand vaccine and therapy development to combat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Hence, advancing our understanding of the comprehensive entry mechanisms of SARS-CoV-2, especially the host factors that facilitate viral infection, is crucial for the discovery of effective vaccines and antiviral drugs. SARS-CoV-2 has previously been documented to reach cells by binding with ACE2 and CD147 receptors in host cells that interact with the spike (S) protein of SARS-CoV-2. A novel entry factor, called neuropilin 1(NRP1), has recently been discovered as a co-receptor facilitating the entry of SARS-CoV-2. NRP1 is a single-pass transmembrane glycoprotein widely distributed throughout the tissues of the body and acts as a multifunctional co-receptor to bind with different ligand proteins and play diverse physiological roles as well as pathological and therapeutic roles in different clinical conditions/diseases, including COVID-19. The current review, therefore, briefly provides the overview of SARS-CoV-2 entry mechanisms, the structure of NRP1, and their roles in health and various diseases, as well as extensively discusses the current understanding of the potential implication of NRP1 in SARS-CoV-2 entry and COVID-19 treatment.
Collapse
Affiliation(s)
- Endeshaw Chekol Abebe
- Department of Medical Biochemistry, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Teklie Mengie Ayele
- Department of Pharmacy, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Zelalem Tilahun Muche
- Department of Medical Physiology, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Tadesse Asmamaw Dejenie
- Department of Medical Biochemistry, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
11
|
Perez-Miller S, Patek M, Moutal A, de Haro PD, Cabel CR, Thorne CA, Campos SK, Khanna R. Novel Compounds Targeting Neuropilin Receptor 1 with Potential To Interfere with SARS-CoV-2 Virus Entry. ACS Chem Neurosci 2021; 12:1299-1312. [PMID: 33787218 PMCID: PMC8029449 DOI: 10.1021/acschemneuro.0c00619] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 03/22/2021] [Indexed: 12/15/2022] Open
Abstract
Neuropilin-1 (NRP-1) is a multifunctional transmembrane receptor for ligands that affect developmental axonal growth and angiogenesis. In addition to a role in cancer, NRP-1 is a reported entry point for several viruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causal agent of coronavirus disease 2019 (COVID-19). The furin cleavage product of SARS-CoV-2 Spike protein takes advantage of the vascular endothelial growth factor A (VEGF-A) binding site on NRP-1 which accommodates a polybasic stretch ending in a C-terminal arginine. This site has long been a focus of drug discovery efforts for cancer therapeutics. We recently showed that interruption of the VEGF-A/NRP-1 signaling pathway ameliorates neuropathic pain and hypothesize that interference of this pathway by SARS-CoV-2 Spike protein interferes with pain signaling. Here, we report confirmed hits from a small molecule and natural product screen of nearly 0.5 million compounds targeting the VEGF-A binding site on NRP-1. We identified nine chemical series with lead- or drug-like physicochemical properties. Using ELISA, we demonstrate that six compounds disrupt VEGF-A-NRP-1 binding more effectively than EG00229, a known NRP-1 inhibitor. Secondary validation in cells revealed that all tested compounds inhibited VEGF-A triggered VEGFR2 phosphorylation. Further, two compounds displayed robust inhibition of a recombinant vesicular stomatitis virus protein that utilizes the SARS-CoV-2 Spike for entry and fusion. These compounds represent a first step in a renewed effort to develop small molecule inhibitors of the VEGF-A/NRP-1 signaling for the treatment of neuropathic pain and cancer with the added potential of inhibiting SARS-CoV-2 virus entry.
Collapse
Affiliation(s)
- Samantha Perez-Miller
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ, United States
- The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Marcel Patek
- Bright Rock Path Consulting, LLC, Tucson, Arizona
| | - Aubin Moutal
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ, United States
| | - Paz Duran de Haro
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ, United States
| | - Carly R. Cabel
- Department of Cellular & Molecular Medicine, College of Medicine, The University of Arizona
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona
| | - Curtis A. Thorne
- Department of Cellular & Molecular Medicine, College of Medicine, The University of Arizona
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona
- Bio5 Institute, University of Arizona
| | - Samuel K. Campos
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona
- Bio5 Institute, University of Arizona
- Department of Immunobiology, College of Medicine, University of Arizona
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ, United States
- The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, Arizona, USA
- Regulonix LLC, Tucson, AZ, USA
| |
Collapse
|
12
|
Shao L, Zhang Y, Gong X, Dong Z, Wei W, Sun H, Sun R, Cong L, Cong X, Jin S. Effects of MLL5 and HOXA regulated by NRP1 on radioresistance in A549. Oncol Lett 2021; 21:403. [PMID: 33777226 PMCID: PMC7988706 DOI: 10.3892/ol.2021.12664] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 02/02/2021] [Indexed: 12/17/2022] Open
Abstract
Radiotherapy is widely used in the management of lung cancer, and physicians are aware that the effect of radiotherapy is dependent on radiosensitivity. Although a series of blockers and activators targeting molecules related to radioresistance have been developed as radiation sensitizers, compensatory mechanisms or drug resistance limits their clinical efficacy. The identification of a key molecule related to lung cancer cell radioresistance or an effective molecular target is a challenging but important problem in radiation oncology. A previous study found that neuropilin 1 (NRP1) is related to radioresistance in A549 cells and is associated with VEGF, PI3K-Akt, MAPK-ERK, P38, NF-κβ and TGF-β. Inhibition of NRP1 can increase the radiosensitivity of A549 cells. Therefore, NRP1 may be a molecular target for radiotherapy-sensitizing drugs in lung cancer. The present study investigated the key downstream genes of NRP1, verified their regulation and clarified their roles in regulating lung cancer radioresistance. NRP1 positively regulated the downstream homeobox genes (HOXs) HOXA6, HOXA9 and mixed lineage leukaemia 5 (MLL5) in addition to MLL5-regulated HOXA6 and HOXA9, but these genes did not regulate NRP1. MLL5, HOXA6 and HOXA9 levels were decreased in tumour tissues and positively correlated with NRP1. All of these genes were induced by ionizing radiation in vivo and in vitro. NRP1 expression was significantly lower in squamous cell carcinoma compared with that in adenocarcinoma, and lymph node metastasis occurred more often in patients with lung cancer with high MLL5 and NRP1 expression compared with patients with low MLL5 and NRP1 expression. Collectively, these data confirmed that NRP1 is associated with MLL5 and regulates radioresistance through HOXA6 and HOXA9.
Collapse
Affiliation(s)
- Lihong Shao
- National Health Commission Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin 130000, P.R. China.,Department of Radiation Oncology and Therapy, Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Yuyu Zhang
- National Health Commission Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin 130000, P.R. China.,Department of Radiation Oncology and Therapy, Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Xinkou Gong
- Department Radiology, 2nd Hospital Affiliated to Jilin University, Changchun, Jilin 130000, P.R. China
| | - Zhuo Dong
- National Health Commission Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin 130000, P.R. China
| | - Wei Wei
- National Health Commission Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin 130000, P.R. China
| | - Hongyan Sun
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Ran Sun
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Lele Cong
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Xianling Cong
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Shunzi Jin
- National Health Commission Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin 130000, P.R. China
| |
Collapse
|
13
|
Albrecht C, Kuznetsov AS, Appert-Collin A, Dhaideh Z, Callewaert M, Bershatsky YV, Urban AS, Bocharov EV, Bagnard D, Baud S, Blaise S, Romier-Crouzet B, Efremov RG, Dauchez M, Duca L, Gueroult M, Maurice P, Bennasroune A. Transmembrane Peptides as a New Strategy to Inhibit Neuraminidase-1 Activation. Front Cell Dev Biol 2020; 8:611121. [PMID: 33392200 PMCID: PMC7772355 DOI: 10.3389/fcell.2020.611121] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/27/2020] [Indexed: 12/31/2022] Open
Abstract
Sialidases, or neuraminidases, are involved in several human disorders such as neurodegenerative, infectious and cardiovascular diseases, and cancers. Accumulative data have shown that inhibition of neuraminidases, such as NEU1 sialidase, may be a promising pharmacological target, and selective inhibitors of NEU1 are therefore needed to better understand the biological functions of this sialidase. In the present study, we designed interfering peptides (IntPep) that target a transmembrane dimerization interface previously identified in human NEU1 that controls its membrane dimerization and sialidase activity. Two complementary strategies were used to deliver the IntPep into cells, either flanked to a TAT sequence or non-tagged for solubilization in detergent micelles. Combined with molecular dynamics simulations and heteronuclear nuclear magnetic resonance (NMR) studies in membrane-mimicking environments, our results show that these IntPep are able to interact with the dimerization interface of human NEU1, to disrupt membrane NEU1 dimerization and to strongly decrease its sialidase activity at the plasma membrane. In conclusion, we report here new selective inhibitors of human NEU1 of strong interest to elucidate the biological functions of this sialidase.
Collapse
Affiliation(s)
- Camille Albrecht
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Andrey S Kuznetsov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Higher School of Economics, Moscow, Russia.,Moscow Institute of Physics and Technology, National Research University, Dolgoprudny, Russia
| | - Aline Appert-Collin
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Zineb Dhaideh
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Maïté Callewaert
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7312, Institut de Chimie Moléculaire de Reims, Reims, France
| | - Yaroslav V Bershatsky
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Moscow Institute of Physics and Technology, National Research University, Dolgoprudny, Russia
| | - Anatoly S Urban
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Moscow Institute of Physics and Technology, National Research University, Dolgoprudny, Russia
| | - Eduard V Bocharov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Moscow Institute of Physics and Technology, National Research University, Dolgoprudny, Russia
| | - Dominique Bagnard
- Université de Strasbourg, Strasbourg, France.,INSERM U1119 Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Labex Medalis, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Stéphanie Baud
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Sébastien Blaise
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Béatrice Romier-Crouzet
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Roman G Efremov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Higher School of Economics, Moscow, Russia.,Moscow Institute of Physics and Technology, National Research University, Dolgoprudny, Russia
| | - Manuel Dauchez
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France.,Plateau de Modélisation Moléculaire Multi-échelle, Reims, France
| | - Laurent Duca
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Marc Gueroult
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Pascal Maurice
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Amar Bennasroune
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| |
Collapse
|
14
|
Perez-Miller S, Patek M, Moutal A, Cabel CR, Thorne CA, Campos SK, Khanna R. In silico identification and validation of inhibitors of the interaction between neuropilin receptor 1 and SARS-CoV-2 Spike protein. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.09.22.308783. [PMID: 32995772 PMCID: PMC7523098 DOI: 10.1101/2020.09.22.308783] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Neuropilin-1 (NRP-1) is a multifunctional transmembrane receptor for ligands that affect developmental axonal growth and angiogenesis. In addition to a role in cancer, NRP-1 is a reported entry point for several viruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causal agent of coronavirus disease 2019 (COVID-19). The furin cleavage product of SARS-CoV-2 Spike protein takes advantage of the vascular endothelial growth factor A (VEGF-A) binding site on NRP-1 which accommodates a polybasic stretch ending in a C-terminal arginine. This site has long been a focus of drug discovery efforts for cancer therapeutics. We recently showed that interruption of the VEGF-A/NRP-1 signaling pathway ameliorates neuropathic pain and hypothesize that interference of this pathway by SARS-CoV-2 spike protein interferes with pain signaling. Here, we report hits from a small molecule and natural product screen of nearly 0.5 million compounds targeting the VEGF-A binding site on NRP-1. We identified nine chemical series with lead- or drug-like physico-chemical properties. Using an ELISA, we demonstrate that six compounds disrupt VEGF-A-NRP-1 binding more effectively than EG00229, a known NRP-1 inhibitor. Secondary validation in cells revealed that almost all tested compounds inhibited VEGF-A triggered VEGFR2 phosphorylation. Two compounds displayed robust inhibition of a recombinant vesicular stomatitis virus protein that utilizes the SARS-CoV-2 Spike for entry and fusion. These compounds represent a first step in a renewed effort to develop small molecule inhibitors of the VEGF-A/NRP-1 signaling for the treatment of neuropathic pain and cancer with the added potential of inhibiting SARS-CoV-2 virus entry.
Collapse
Affiliation(s)
- Samantha Perez-Miller
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ, United States
- The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, Arizona 85724, USA
| | - Marcel Patek
- Bright Rock Path Consulting, LLC, Tucson, Arizona
| | - Aubin Moutal
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ, United States
| | - Carly R. Cabel
- Department of Cellular & Molecular Medicine, College of Medicine, The University of Arizona
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona
| | - Curtis A. Thorne
- Department of Cellular & Molecular Medicine, College of Medicine, The University of Arizona
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona
- Bio5 Institute, University of Arizona
| | - Samuel K. Campos
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona
- Bio5 Institute, University of Arizona
- Department of Immunobiology, College of Medicine, University of Arizona
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ, United States
- The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, Arizona 85724, USA
- Regulonix LLC, 1555 E. Entrada Segunda, Tucson, AZ 85718, USA
| |
Collapse
|
15
|
Barrett CT, Dutch RE. Viral Membrane Fusion and the Transmembrane Domain. Viruses 2020; 12:v12070693. [PMID: 32604992 PMCID: PMC7412173 DOI: 10.3390/v12070693] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 01/05/2023] Open
Abstract
Initiation of host cell infection by an enveloped virus requires a viral-to-host cell membrane fusion event. This event is mediated by at least one viral transmembrane glycoprotein, termed the fusion protein, which is a key therapeutic target. Viral fusion proteins have been studied for decades, and numerous critical insights into their function have been elucidated. However, the transmembrane region remains one of the most poorly understood facets of these proteins. In the past ten years, the field has made significant advances in understanding the role of the membrane-spanning region of viral fusion proteins. We summarize developments made in the past decade that have contributed to the understanding of the transmembrane region of viral fusion proteins, highlighting not only their critical role in the membrane fusion process, but further demonstrating their involvement in several aspects of the viral lifecycle.
Collapse
|
16
|
Albrecht C, Appert-Collin A, Bagnard D, Blaise S, Romier-Crouzet B, Efremov RG, Sartelet H, Duca L, Maurice P, Bennasroune A. Transmembrane Peptides as Inhibitors of Protein-Protein Interactions: An Efficient Strategy to Target Cancer Cells? Front Oncol 2020; 10:519. [PMID: 32351895 PMCID: PMC7174899 DOI: 10.3389/fonc.2020.00519] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/23/2020] [Indexed: 12/11/2022] Open
Abstract
Cellular functions are regulated by extracellular signals such as hormones, neurotransmitters, matrix ligands, and other chemical or physical stimuli. Ligand binding on its transmembrane receptor induced cell signaling and the recruitment of several interacting partners to the plasma membrane. Nowadays, it is well-established that the transmembrane domain is not only an anchor of these receptors to the membrane, but it also plays a key role in receptor dimerization and activation. Indeed, interactions between transmembrane helices are associated with specific biological activity of the proteins as cell migration, proliferation, or differentiation. Overexpression or constitutive dimerization (due notably to mutations) of these transmembrane receptors are involved in several physiopathological contexts as cancers. The transmembrane domain of tyrosine kinase receptors as ErbB family proteins (implicated in several cancers as HER2 in breast cancer) or other receptors as Neuropilins has been described these last years as a target to inhibit their dimerization/activation using several strategies. In this review, we will focus on the strategy which consists in using peptides to disturb in a specific manner the interactions between transmembrane domains and the signaling pathways (induced by ligand binding) of these receptors involved in cancer. This approach can be extended to inhibit other transmembrane protein dimerization as neuraminidase-1 (the catalytic subunit of elastin receptor complex), Discoidin Domain Receptor 1 (a tyrosine kinase receptor activated by type I collagen) or G-protein coupled receptors (GPCRs) which are involved in cancer processes.
Collapse
Affiliation(s)
- Camille Albrecht
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Aline Appert-Collin
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Dominique Bagnard
- Université de Strasbourg, Strasbourg, France.,INSERM U1119 Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Labex Medalis, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Sébastien Blaise
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Béatrice Romier-Crouzet
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Roman G Efremov
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Higher School of Economics, Moscow, Russia
| | - Hervé Sartelet
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Laurent Duca
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Pascal Maurice
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Amar Bennasroune
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| |
Collapse
|
17
|
Neuropilin: Handyman and Power Broker in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1223:31-67. [PMID: 32030684 DOI: 10.1007/978-3-030-35582-1_3] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Neuropilin-1 and neuropilin-2 form a small family of transmembrane receptors, which, due to the lack of a cytosolic protein kinase domain, act primarily as co-receptors for various ligands. Performing at the molecular level both the executive and organizing functions of a handyman as well as of a power broker, they are instrumental in controlling the signaling of various receptor tyrosine kinases, integrins, and other molecules involved in the regulation of physiological and pathological angiogenic processes. In this setting, the various neuropilin ligands and interaction partners on various cells of the tumor microenvironment, such as cancer cells, endothelial cells, cancer-associated fibroblasts, and immune cells, are surveyed. The suitability of various neuropilin-targeting substances and the intervention in neuropilin-mediated interactions is considered as a possible building block of tumor therapy.
Collapse
|
18
|
Westerfield JM, Barrera FN. Membrane receptor activation mechanisms and transmembrane peptide tools to elucidate them. J Biol Chem 2019; 295:1792-1814. [PMID: 31879273 DOI: 10.1074/jbc.rev119.009457] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Single-pass membrane receptors contain extracellular domains that respond to external stimuli and transmit information to intracellular domains through a single transmembrane (TM) α-helix. Because membrane receptors have various roles in homeostasis, signaling malfunctions of these receptors can cause disease. Despite their importance, there is still much to be understood mechanistically about how single-pass receptors are activated. In general, single-pass receptors respond to extracellular stimuli via alterations in their oligomeric state. The details of this process are still the focus of intense study, and several lines of evidence indicate that the TM domain (TMD) of the receptor plays a central role. We discuss three major mechanistic hypotheses for receptor activation: ligand-induced dimerization, ligand-induced rotation, and receptor clustering. Recent observations suggest that receptors can use a combination of these activation mechanisms and that technical limitations can bias interpretation. Short peptides derived from receptor TMDs, which can be identified by screening or rationally developed on the basis of the structure or sequence of their targets, have provided critical insights into receptor function. Here, we explore recent evidence that, depending on the target receptor, TMD peptides cannot only inhibit but also activate target receptors and can accommodate novel, bifunctional designs. Furthermore, we call for more sharing of negative results to inform the TMD peptide field, which is rapidly transforming into a suite of unique tools with the potential for future therapeutics.
Collapse
Affiliation(s)
- Justin M Westerfield
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee 37996
| | - Francisco N Barrera
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee 37996.
| |
Collapse
|
19
|
Gamper C, Spenlé C, Boscá S, van der Heyden M, Erhardt M, Orend G, Bagnard D, Heinlein M. Functionalized Tobacco Mosaic Virus Coat Protein Monomers and Oligomers as Nanocarriers for Anti-Cancer Peptides. Cancers (Basel) 2019; 11:cancers11101609. [PMID: 31652529 PMCID: PMC6826726 DOI: 10.3390/cancers11101609] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/14/2019] [Accepted: 10/18/2019] [Indexed: 01/13/2023] Open
Abstract
Components with self-assembly properties derived from plant viruses provide the opportunity to design biological nanoscaffolds for the ordered display of agents of diverse nature and with complementing functions. With the aim of designing a functionalized nanoscaffold to target cancer, the coat protein (CP) of Tobacco mosaic virus (TMV) was tested as nanocarrier for an insoluble, highly hydrophobic peptide that targets the transmembrane domain of the Neuropilin-1 (NRP1) receptor in cancer cells. The resulting construct CPL-K (CP-linker-“Kill”) binds to NRP1 in cancer cells and disrupts NRP1 complex formation with PlexA1 as well as downstream Akt survival signaling. The application of CPL-K also inhibits angiogenesis and cell migration. CP was also fused to a peptide that targets the extracellular domain of NRP1 and this fusion protein (CPL-F, CP-Linker-“Find”) is shown to bind to cultured cancer cells and to inhibit NRP1-dependent angiogenesis as well. CPL-K and CPL-F maintain their anti-angiogenic properties upon co-assembly to oligomers/nanoparticles together with CPL. The observations show that the CP of TMV can be employed to generate a functionalized nanoparticle with biological activity. Remarkably, fusion to CPL allowed us to solubilize the highly insoluble transmembrane NRP1 peptide and to retain its anti-angiogenic effect.
Collapse
Affiliation(s)
- Coralie Gamper
- Institut de Biologie Moléculaire des Plantes (IBMP-CNRS), Université de Strasbourg, 67000 Strasbourg, France.
- INSERM 1119, BMNST Laboratory, Université de Strasbourg, 67000 Strasbourg, France.
- Labex Medalis, Université de Strasbourg, 67000 Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg, FMTS, Université de Strasbourg, 67000 Strasbourg, France.
- INSERM 1109, MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Université de Strasbourg, 67000 Strasbourg, France.
| | - Caroline Spenlé
- Institut de Biologie Moléculaire des Plantes (IBMP-CNRS), Université de Strasbourg, 67000 Strasbourg, France.
- INSERM 1119, BMNST Laboratory, Université de Strasbourg, 67000 Strasbourg, France.
- Labex Medalis, Université de Strasbourg, 67000 Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg, FMTS, Université de Strasbourg, 67000 Strasbourg, France.
- INSERM 1109, MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Université de Strasbourg, 67000 Strasbourg, France.
| | - Sonia Boscá
- Institut de Biologie Moléculaire des Plantes (IBMP-CNRS), Université de Strasbourg, 67000 Strasbourg, France.
| | - Michael van der Heyden
- INSERM 1119, BMNST Laboratory, Université de Strasbourg, 67000 Strasbourg, France.
- Labex Medalis, Université de Strasbourg, 67000 Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg, FMTS, Université de Strasbourg, 67000 Strasbourg, France.
- INSERM 1109, MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Université de Strasbourg, 67000 Strasbourg, France.
| | - Mathieu Erhardt
- Institut de Biologie Moléculaire des Plantes (IBMP-CNRS), Université de Strasbourg, 67000 Strasbourg, France.
| | - Gertraud Orend
- Labex Medalis, Université de Strasbourg, 67000 Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg, FMTS, Université de Strasbourg, 67000 Strasbourg, France.
- INSERM 1109, MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Université de Strasbourg, 67000 Strasbourg, France.
- INSERM 1109, The Tumor Microenvironment Laboratory, Université de Strasbourg, 67000 Strasbourg, France.
| | - Dominique Bagnard
- INSERM 1119, BMNST Laboratory, Université de Strasbourg, 67000 Strasbourg, France.
- Labex Medalis, Université de Strasbourg, 67000 Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg, FMTS, Université de Strasbourg, 67000 Strasbourg, France.
- INSERM 1109, MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Université de Strasbourg, 67000 Strasbourg, France.
| | - Manfred Heinlein
- Institut de Biologie Moléculaire des Plantes (IBMP-CNRS), Université de Strasbourg, 67000 Strasbourg, France.
- University of Strasbourg Institute of Advanced Study (USIAS), 67000 Strasbourg, France.
| |
Collapse
|
20
|
Novel therapeutic interventions in cancer treatment using protein and peptide-based targeted smart systems. Semin Cancer Biol 2019; 69:249-267. [PMID: 31442570 DOI: 10.1016/j.semcancer.2019.08.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/19/2019] [Accepted: 08/20/2019] [Indexed: 01/12/2023]
Abstract
Cancer, being the most prevalent and resistant disease afflicting any gender, age or social status, is the ultimate challenge for the scientific community. The new generation therapeutics for cancer management has shifted the approach to personalized/precision medicine, making use of patient- and tumor-specific markers for specifying the targeted therapies for each patient. Peptides targeting these cancer-specific signatures hold enormous potential for cancer therapy and diagnosis. The rapid advancements in the combinatorial peptide libraries served as an impetus to the development of multifunctional peptide-based materials for targeted cancer therapy. The present review outlines benefits and shortcomings of peptides as cancer therapeutics and the potential of peptide modified nanomedicines for targeted delivery of anticancer agents.
Collapse
|
21
|
A Hydrophobic Target: Using the Paramyxovirus Fusion Protein Transmembrane Domain To Modulate Fusion Protein Stability. J Virol 2019; 93:JVI.00863-19. [PMID: 31217248 DOI: 10.1128/jvi.00863-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 06/12/2019] [Indexed: 12/15/2022] Open
Abstract
Enveloped viruses utilize surface glycoproteins to bind and fuse with a target cell membrane. The zoonotic Hendra virus (HeV), a member of the family Paramyxoviridae, utilizes the attachment protein (G) and the fusion protein (F) to perform these critical functions. Upon triggering, the trimeric F protein undergoes a large, irreversible conformation change to drive membrane fusion. Previously, we have shown that the transmembrane (TM) domain of the F protein, separate from the rest of the protein, is present in a monomer-trimer equilibrium. This TM-TM association contributes to the stability of the prefusion form of the protein, supporting a role for TM-TM interactions in the control of F protein conformational changes. To determine the impact of disrupting TM-TM interactions, constructs expressing the HeV F TM with limited flanking sequences were synthesized. Coexpression of these constructs with HeV F resulted in dramatic reductions in the stability of F protein expression and fusion activity. In contrast, no effects were observed when the HeV F TM constructs were coexpressed with the nonhomologous parainfluenza virus 5 (PIV5) fusion protein, indicating a requirement for specific interactions. To further examine this, a TM peptide homologous to the PIV5 F TM domain was synthesized. Addition of the peptide prior to infection inhibited infection with PIV5 but did not significantly affect infection with human metapneumovirus, a related virus. These results indicate that targeted disruption of TM-TM interactions significantly impact viral fusion protein stability and function, presenting these interactions as a novel target for antiviral development.IMPORTANCE Enveloped viruses require virus-cell membrane fusion to release the viral genome and replicate. The viral fusion protein triggers from the pre- to the postfusion conformation, an essentially irreversible change, to drive membrane fusion. We found that small proteins containing the TM and a limited flanking region homologous to the fusion protein of the zoonotic Hendra virus reduced protein expression and fusion activity. The introduction of exogenous TM peptides may displace a TM domain, disrupting native TM-TM interactions and globally destabilizing the fusion protein. Supporting this hypothesis, we showed that a sequence-specific transmembrane peptide dramatically reduced viral infection in another enveloped virus model, suggesting a broader inhibitory mechanism. Viral fusion protein TM-TM interactions are important for protein function, and disruption of these interactions dramatically reduces protein stability.
Collapse
|
22
|
Niland S, Eble JA. Neuropilins in the Context of Tumor Vasculature. Int J Mol Sci 2019; 20:ijms20030639. [PMID: 30717262 PMCID: PMC6387129 DOI: 10.3390/ijms20030639] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/23/2019] [Accepted: 01/29/2019] [Indexed: 01/09/2023] Open
Abstract
Neuropilin-1 and Neuropilin-2 form a small family of plasma membrane spanning receptors originally identified by the binding of semaphorin and vascular endothelial growth factor. Having no cytosolic protein kinase domain, they function predominantly as co-receptors of other receptors for various ligands. As such, they critically modulate the signaling of various receptor tyrosine kinases, integrins, and other molecules involved in the regulation of physiological and pathological angiogenic processes. This review highlights the diverse neuropilin ligands and interacting partners on endothelial cells, which are relevant in the context of the tumor vasculature and the tumor microenvironment. In addition to tumor cells, the latter contains cancer-associated fibroblasts, immune cells, and endothelial cells. Based on the prevalent neuropilin-mediated interactions, the suitability of various neuropilin-targeted substances for influencing tumor angiogenesis as a possible building block of a tumor therapy is discussed.
Collapse
Affiliation(s)
- Stephan Niland
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany.
| | - Johannes A Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany.
| |
Collapse
|
23
|
Kuchler-Bopp S, Bagnard D, Van-Der-Heyden M, Idoux-Gillet Y, Strub M, Gegout H, Lesot H, Benkirane-Jessel N, Keller L. Semaphorin 3A receptor inhibitor as a novel therapeutic to promote innervation of bioengineered teeth. J Tissue Eng Regen Med 2018; 12:e2151-e2161. [PMID: 29430872 DOI: 10.1002/term.2648] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 12/15/2017] [Accepted: 01/16/2018] [Indexed: 12/21/2022]
Abstract
The sensory innervation of the dental pulp is essential for tooth function and protection. It is mediated by axons originating from the trigeminal ganglia and is spatio-temporally regulated. We have previously shown that the innervation of bioengineered teeth can be achieved only under immunosuppressive conditions. The aim of this study was to develop a model to determine the role of Semaphorin 3A (Sema3A) in the innervation of bioengineered teeth. We first analysed innervation of the dental pulp of mandibular first molars in newborn (postnatal day 0: PN0) mice deficient for Sema3A (Sema3A-/- ), a strong inhibitor of axon growth. While at PN0, axons detected by immunostaining for peripherin and NF200 were restricted to the peridental mesenchyme in Sema3A+/+ mice, they entered the dental pulp in Sema3A-/- mice. Then, we have implanted cultured teeth obtained from embryonic day-14 (E14) molar germs of Sema3A-/- mice together with trigeminal ganglia. The dental pulps of E14 cultured and implanted Sema3A-/- teeth were innervated, whereas the axons did not enter the pulp of E14 Sema3A+/+ cultured and implanted teeth. A "Membrane Targeting Peptide NRP1," suppressing the inhibitory effect of Sema3A, has been previously identified. The injection of this peptide at the site of implantation allowed the innervation of the dental pulp of bioengineered teeth obtained from E14 dental dissociated mesenchymal and epithelial cells reassociations of ICR mice. In conclusion, these data show that inhibition of only one axon repellent molecule, Sema3A, allows for pulp innervation of bioengineered teeth.
Collapse
Affiliation(s)
- Sabine Kuchler-Bopp
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), FMTS, 67000, Strasbourg
| | - Dominique Bagnard
- INSERM, UMR 1119-Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle, Labex Medalis, University of Strasbourg, Strasbourg, France
| | - Michael Van-Der-Heyden
- INSERM, UMR 1119-Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle, Labex Medalis, University of Strasbourg, Strasbourg, France
| | - Ysia Idoux-Gillet
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), FMTS, 67000, Strasbourg.,Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Ste Elisabeth, 67000, Strasbourg, France
| | - Marion Strub
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), FMTS, 67000, Strasbourg.,Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Ste Elisabeth, 67000, Strasbourg, France.,Hôpitaux universitaires de Strasbourg (HUS), Département de Pédodontie, 1 place de l'Hôpital, 67000, Strasbourg
| | - Hervé Gegout
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), FMTS, 67000, Strasbourg.,Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Ste Elisabeth, 67000, Strasbourg, France
| | - Hervé Lesot
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), FMTS, 67000, Strasbourg
| | - Nadia Benkirane-Jessel
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), FMTS, 67000, Strasbourg.,Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Ste Elisabeth, 67000, Strasbourg, France
| | - Laetitia Keller
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), FMTS, 67000, Strasbourg.,Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Ste Elisabeth, 67000, Strasbourg, France
| |
Collapse
|
24
|
Tryptophan-mediated Dimerization of the TssL Transmembrane Anchor Is Required for Type VI Secretion System Activity. J Mol Biol 2018; 430:987-1003. [DOI: 10.1016/j.jmb.2018.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 01/29/2018] [Accepted: 02/11/2018] [Indexed: 11/23/2022]
|
25
|
Jacob L, Sawma P, Garnier N, Meyer LAT, Fritz J, Hussenet T, Spenlé C, Goetz J, Vermot J, Fernandez A, Baumlin N, Aci-Sèche S, Orend G, Roussel G, Crémel G, Genest M, Hubert P, Bagnard D. Inhibition of PlexA1-mediated brain tumor growth and tumor-associated angiogenesis using a transmembrane domain targeting peptide. Oncotarget 2018; 7:57851-57865. [PMID: 27506939 PMCID: PMC5295395 DOI: 10.18632/oncotarget.11072] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 07/21/2016] [Indexed: 11/25/2022] Open
Abstract
The neuropilin-plexin receptor complex regulates tumor cell migration and proliferation and thus is an interesting therapeutic target. High expression of neuropilin-1 is indeed associated with a bad prognosis in glioma patients. Q-RTPCR and tissue-array analyses showed here that Plexin-A1 is highly expressed in glioblastoma and that the highest level of expression correlates with the worse survival of patients. We next identified a developmental and tumor-associated pro-angiogenic role of Plexin-A1. Hence, by using molecular simulations and a two-hybrid like assay in parallel with biochemical and cellular assays we developed a specific Plexin-A1 peptidic antagonist disrupting transmembrane domain-mediated oligomerization of the receptor and subsequent signaling and functional activity. We found that this peptide exhibits anti-tumor activity in vivo on different human glioblastoma models including glioma cancer stem cells. Thus, screening Plexin-A1 expression and targeting Plexin-A1 in glioblastoma patients exhibit diagnostic and therapeutic value.
Collapse
Affiliation(s)
- Laurent Jacob
- MN3T Team, INSERM U1109, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Paul Sawma
- Laboratoire d’Ingénierie des Systèmes Macromoléculaires (LISM), UMR 7255, CNRS-Aix Marseille Université, Marseille, France
| | - Norbert Garnier
- Centre de Biophysique Moléculaire, UPR 4301, CNRS, Affiliated to the University of Orléans, Orléans, France
| | - Lionel A T Meyer
- MN3T Team, INSERM U1109, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Justine Fritz
- MN3T Team, INSERM U1109, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Thomas Hussenet
- MN3T Team, INSERM U1109, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Caroline Spenlé
- MN3T Team, INSERM U1109, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Jacky Goetz
- MN3T Team, INSERM U1109, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.,Institute of Genetics and Molecular and Cellular Biology (IGBMC), CNRS/INSERM/UDS, Illkirch, France
| | - Julien Vermot
- Institute of Genetics and Molecular and Cellular Biology (IGBMC), CNRS/INSERM/UDS, Illkirch, France
| | - Aurore Fernandez
- MN3T Team, INSERM U1109, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Nadège Baumlin
- MN3T Team, INSERM U1109, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Samia Aci-Sèche
- Centre de Biophysique Moléculaire, UPR 4301, CNRS, Affiliated to the University of Orléans, Orléans, France.,Current address: Institut de Chimie Organique et Analytique UMR, Université d'Orléans, Orléans, France
| | - Gertraud Orend
- MN3T Team, INSERM U1109, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Guy Roussel
- MN3T Team, INSERM U1109, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Gérard Crémel
- MN3T Team, INSERM U1109, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Monique Genest
- Centre de Biophysique Moléculaire, UPR 4301, CNRS, Affiliated to the University of Orléans, Orléans, France
| | - Pierre Hubert
- Laboratoire d’Ingénierie des Systèmes Macromoléculaires (LISM), UMR 7255, CNRS-Aix Marseille Université, Marseille, France
| | - Dominique Bagnard
- MN3T Team, INSERM U1109, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| |
Collapse
|
26
|
Arpel A, Gamper C, Spenlé C, Fernandez A, Jacob L, Baumlin N, Laquerriere P, Orend G, Crémel G, Bagnard D. Inhibition of primary breast tumor growth and metastasis using a neuropilin-1 transmembrane domain interfering peptide. Oncotarget 2018; 7:54723-54732. [PMID: 27351129 PMCID: PMC5342376 DOI: 10.18632/oncotarget.10101] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 05/28/2016] [Indexed: 02/01/2023] Open
Abstract
The transmembrane domains (TMD) in membrane receptors play a key role in cell signaling. As previously shown by us a peptide targeting the TMD of neuropilin-1 (MTP-NRP1), blocks cell proliferation, cell migration and angiogenesis in vitro, and decreases glioblastoma growth in vivo. We now explored the clinical potential of MTP-NRP1 on breast cancer models and demonstrate that MTP-NRP1 blocks proliferation of several breast cancer lines including the MDA-MB-231, a triple negative human breast cancer cell line. In models with long term in vivo administration of the peptide, MTP-NRP1 not only reduced tumor volume but also decreased number and size of breast cancer metastases. Strikingly, treating mice before tumors developed protected from metastasis establishment/formation. Overall, our results report that targeting the TMD of NRP1 in breast cancer is a potent new strategy to fight against breast cancer and related metastasis.
Collapse
Affiliation(s)
- Alexia Arpel
- INSERM U 1109, MN3T Laboratory, Labex Medalis, Strasbourg University, Strasbourg, France.,CNRS UMR 7178, Institut Pluridisciplinaire Hubert Curien, Strasbourg University, Strasbourg, France
| | - Coralie Gamper
- INSERM U 1109, MN3T Laboratory, Labex Medalis, Strasbourg University, Strasbourg, France
| | - Caroline Spenlé
- INSERM U 1109, MN3T Laboratory, Labex Medalis, Strasbourg University, Strasbourg, France
| | - Aurore Fernandez
- INSERM U 1109, MN3T Laboratory, Labex Medalis, Strasbourg University, Strasbourg, France
| | - Laurent Jacob
- INSERM U 1109, MN3T Laboratory, Labex Medalis, Strasbourg University, Strasbourg, France
| | - Nadège Baumlin
- INSERM U 1109, MN3T Laboratory, Labex Medalis, Strasbourg University, Strasbourg, France
| | - Patrice Laquerriere
- CNRS UMR 7178, Institut Pluridisciplinaire Hubert Curien, Strasbourg University, Strasbourg, France
| | - Gertraud Orend
- INSERM U 1109, MN3T Laboratory, Labex Medalis, Strasbourg University, Strasbourg, France
| | - Gérard Crémel
- INSERM U 1109, MN3T Laboratory, Labex Medalis, Strasbourg University, Strasbourg, France
| | - Dominique Bagnard
- INSERM U 1109, MN3T Laboratory, Labex Medalis, Strasbourg University, Strasbourg, France
| |
Collapse
|
27
|
Abstract
All known splice isoforms of vascular endothelial growth factor A (VEGF-A) can bind to the receptor tyrosine kinases VEGFR-1 and VEGFR-2. We focus here on VEGF-A121a and VEGF-A165a, two of the most abundant VEGF-A splice isoforms in human tissue 1 , and their ability to bind the Neuropilin co-receptors NRP1 and NRP2. The Neuropilins are key vascular, immune, and nervous system receptors on endothelial cells, neuronal axons, and regulatory T cells respectively. They serve as co-receptors for the Plexins in Semaphorin binding on neuronal and vascular endothelial cells, and for the VEGFRs in VEGF binding on vascular and lymphatic endothelial cells, and thus regulate the initiation and coordination of cell signaling by Semaphorins and VEGFs. 2 There is conflicting evidence in the literature as to whether only heparin-binding VEGF-A isoforms - that is, isoforms with domains encoded by exons 6 and/or 7 plus 8a - bind to Neuropilins on endothelial cells. While it is clear that VEGF-A165a binds to both NRP1 and NRP2, published studies do not all agree on the ability of VEGF-A121a to bind NRPs. Here, we review and attempt to reconcile evidence for and against VEGF-A121a binding to Neuropilins. This evidence suggests that, in vitro, VEGF-A121a can bind to both NRP1 and NRP2 via domains encoded by exons 5 and 8a; in the case of NRP1, VEGF-A121a binds with lower affinity than VEGF-A165a. In in vitro cell culture experiments, both NRP1 and NRP2 can enhance VEGF-A121a-induced phosphorylation of VEGFR2 and downstream signaling including proliferation. However, unlike VEGFA-165a, experiments have shown that VEGF-A121a does not 'bridge' VEGFR2 and NRP1, i.e. it does not bind both receptors simultaneously at their extracellular domain. Thus, the mechanism by which Neuropilins potentiate VEGF-A121a-mediated VEGFR2 signaling may be different from that for VEGF-A165a. We suggest such an alternate mechanism: interactions between NRP1 and VEGFR2 transmembrane (TM) and intracellular (IC) domains.
Collapse
Affiliation(s)
- Sarvenaz Sarabipour
- a Institute for Computational Medicine, Department of Biomedical Engineering, Institute for NanoBioTechnology , Johns Hopkins University , Baltimore , MD , USA
| | - Feilim Mac Gabhann
- a Institute for Computational Medicine, Department of Biomedical Engineering, Institute for NanoBioTechnology , Johns Hopkins University , Baltimore , MD , USA
| |
Collapse
|
28
|
Mittal VK, McDonald JF. De novo assembly and characterization of breast cancer transcriptomes identifies large numbers of novel fusion-gene transcripts of potential functional significance. BMC Med Genomics 2017; 10:53. [PMID: 28851357 PMCID: PMC5575902 DOI: 10.1186/s12920-017-0289-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 08/17/2017] [Indexed: 11/10/2022] Open
Abstract
Background Gene-fusion or chimeric transcripts have been implicated in the onset and progression of a variety of cancers. Massively parallel RNA sequencing (RNA-Seq) of the cellular transcriptome is a promising approach for the identification of chimeric transcripts of potential functional significance. We report here the development and use of an integrated computational pipeline for the de novo assembly and characterization of chimeric transcripts in 55 primary breast cancer and normal tissue samples. Methods An integrated computational pipeline was employed to screen the transcriptome of breast cancer and control tissues for high-quality RNA-sequencing reads. Reads were de novo assembled into contigs followed by reference genome mapping. Chimeric transcripts were detected, filtered and characterized using our R-SAP algorithm. The relative abundance of reads was used to estimate levels of gene expression. Results De novo assembly allowed for the accurate detection of 1959 chimeric transcripts to nucleotide level resolution and facilitated detailed molecular characterization and quantitative analysis. A number of the chimeric transcripts are of potential functional significance including 79 novel fusion-protein transcripts and many chimeric transcripts with alterations in their un-translated leader regions. A number of chimeric transcripts in the cancer samples mapped to genomic regions devoid of any known genes. Several ‘pro-neoplastic’ fusions comprised of genes previously implicated in cancer are expressed at low levels in normal tissues but at high levels in cancer tissues. Conclusions Collectively, our results underscore the utility of deep sequencing technologies and improved bioinformatics workflows to uncover novel and potentially significant chimeric transcripts in cancer and normal somatic tissues. Electronic supplementary material The online version of this article (doi:10.1186/s12920-017-0289-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vinay K Mittal
- Integrated Cancer Research Center, School of Biological Sciences, and Parker H. Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, 315 Ferst Dr, Atlanta, GA, 30332, USA
| | - John F McDonald
- Integrated Cancer Research Center, School of Biological Sciences, and Parker H. Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, 315 Ferst Dr, Atlanta, GA, 30332, USA.
| |
Collapse
|
29
|
Pinskey JM, Franks NE, McMellen AN, Giger RJ, Allen BL. Neuropilin-1 promotes Hedgehog signaling through a novel cytoplasmic motif. J Biol Chem 2017; 292:15192-15204. [PMID: 28667171 DOI: 10.1074/jbc.m117.783845] [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] [Received: 03/08/2017] [Revised: 06/23/2017] [Indexed: 12/30/2022] Open
Abstract
Hedgehog (HH) signaling critically regulates embryonic and postnatal development as well as adult tissue homeostasis, and its perturbation can lead to developmental disorders, birth defects, and cancers. Neuropilins (NRPs), which have well-defined roles in Semaphorin and VEGF signaling, positively regulate HH pathway function, although their mechanism of action in HH signaling remains unclear. Here, using luciferase-based reporter assays, we provide evidence that NRP1 regulates HH signaling specifically at the level of GLI transcriptional activator function. Moreover, we show that NRP1 localization to the primary cilium, a key platform for HH signal transduction, does not correlate with HH signal promotion. Rather, a structure-function analysis suggests that the NRP1 cytoplasmic and transmembrane domains are necessary and sufficient to regulate HH pathway activity. Furthermore, we identify a previously uncharacterized, 12-amino acid region within the NRP1 cytoplasmic domain that mediates HH signal promotion. Overall, our results provide mechanistic insight into NRP1 function within and potentially beyond the HH signaling pathway. These insights have implications for the development of novel modulators of HH-driven developmental disorders and diseases.
Collapse
Affiliation(s)
- Justine M Pinskey
- From the Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Nicole E Franks
- From the Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Alexandra N McMellen
- From the Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Roman J Giger
- From the Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Benjamin L Allen
- From the Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
30
|
Opławski M, Michalski M, Witek A, Michalski B, Zmarzły N, Jęda-Golonka A, Styblińska M, Gola J, Kasprzyk-Żyszczyńska M, Mazurek U, Plewka A. Identification of a gene expression profile associated with the regulation of angiogenesis in endometrial cancer. Mol Med Rep 2017; 16:2547-2555. [PMID: 28656251 PMCID: PMC5547990 DOI: 10.3892/mmr.2017.6868] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 02/20/2017] [Indexed: 12/11/2022] Open
Abstract
The publication of the human genome sequence provided direction in the search for novel diagnostic and therapeutic methods for the treatment of human diseases. The aim of the present study was to investigate the hypothesis that the expression profile of genes involved in the regulation of angiogenesis may be a marker in endometrial cancer that facilitates the diagnosis and prognosis of patients, as well as the identification of novel therapeutic targets. The current study included 36 patients with grade (G) 1 to 3 endometrial cancer, and a control group of patients consisting of females that qualified for the removal of the uterus. Out of these, 28 samples (control, 3; G1, 7; G2, 12; and G3, 6) were selected for microarray analysis. Molecular analysis of the endometrial samples involved the extraction of total RNA, purification of the obtained extracts and subsequent analysis of the gene expression profiles using an oligonucleotide microarray technique (GeneChip® Human Genome U133A plates). The results indicated that the mRNA expression profile of genes involved in the regulation of angiogenesis varies depending on the degree of histological differentiation of endometrial adenocarcinoma. Similar results were obtained from descriptive statistics characterizing the expression profile of 691 mRNAs associated with the regulation of angiogenesis in the groups of patients with endometrial adenocarcinoma. In addition, the results of the present study indicated that neuropilin2 (NRP2) may serve an important role in the activity of endothelial cells, and may affect vascular endothelial growth factor, and potentially plexins and integrins via regulation of their functions. An understanding of how these proteins interact remains to be determined; however, elucidating these interactions may provide an explanation for the mechanisms underlying angiogenesis. In conclusion, the results of the present study suggest that NRP2 may be a valuable target for investigation in future pharmacological studies involving angiogenesis in endometrial cancer.
Collapse
Affiliation(s)
- Marcin Opławski
- Department of Proteomics, School of Pharmacy, Division of Medical Analytics, Medical University of Silesia, Sosnowiec 41‑200, Poland
| | - Mateusz Michalski
- Department of Gynecological Oncology, Gynecology and Obstetrics, Regional Railway Hospital, Katowice 40‑760, Poland
| | - Andrzej Witek
- Department of Gynecology, Obstetrics and Oncologic Gynecology, Medical University of Silesia, Katowice 40‑752, Poland
| | - Bogdan Michalski
- Department of Oncological Gynaecology, School of Health Sciences, Medical University of Silesia, Katowice 40‑752, Poland
| | - Nikola Zmarzły
- Department of Molecular Biology, School of Pharmacy, Division of Medical Analytics, Medical University of Silesia, Sosnowiec 41‑200, Poland
| | - Agnieszka Jęda-Golonka
- Department of Gynecological Oncology, Gynecology and Obstetrics, Regional Railway Hospital, Katowice 40‑760, Poland
| | - Maria Styblińska
- Department of Molecular Biology, School of Pharmacy, Division of Medical Analytics, Medical University of Silesia, Sosnowiec 41‑200, Poland
| | - Joanna Gola
- Department of Molecular Biology, School of Pharmacy, Division of Medical Analytics, Medical University of Silesia, Sosnowiec 41‑200, Poland
| | - Małgorzata Kasprzyk-Żyszczyńska
- Department of Proteomics, School of Pharmacy, Division of Medical Analytics, Medical University of Silesia, Sosnowiec 41‑200, Poland
| | - Urszula Mazurek
- Department of Molecular Biology, School of Pharmacy, Division of Medical Analytics, Medical University of Silesia, Sosnowiec 41‑200, Poland
| | - Andrzej Plewka
- Department of Proteomics, School of Pharmacy, Division of Medical Analytics, Medical University of Silesia, Sosnowiec 41‑200, Poland
| |
Collapse
|
31
|
Zeng X, Wu P, Yao C, Liang J, Zhang S, Yin H. Small Molecule and Peptide Recognition of Protein Transmembrane Domains. Biochemistry 2017; 56:2076-2085. [DOI: 10.1021/acs.biochem.6b00909] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Xianfeng Zeng
- Center
of Basic Molecular Science, Department of Chemistry, Tsinghua University, Beijing 100082, China
| | - Peiyao Wu
- Center
of Basic Molecular Science, Department of Chemistry, Tsinghua University, Beijing 100082, China
| | - Chengbo Yao
- Center
of Basic Molecular Science, Department of Chemistry, Tsinghua University, Beijing 100082, China
| | - Jiaqi Liang
- Center
of Basic Molecular Science, Department of Chemistry, Tsinghua University, Beijing 100082, China
| | - Shuting Zhang
- Center
of Basic Molecular Science, Department of Chemistry, Tsinghua University, Beijing 100082, China
- School
of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, China
| | - Hang Yin
- Center
of Basic Molecular Science, Department of Chemistry, Tsinghua University, Beijing 100082, China
| |
Collapse
|
32
|
Meyer LAT, Fritz J, Pierdant-Mancera M, Bagnard D. Current drug design to target the Semaphorin/Neuropilin/Plexin complexes. Cell Adh Migr 2016; 10:700-708. [PMID: 27906605 PMCID: PMC5160035 DOI: 10.1080/19336918.2016.1261785] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 11/09/2016] [Accepted: 11/14/2016] [Indexed: 12/21/2022] Open
Abstract
The Semaphorin/Neuropilin/Plexin (SNP) complexes control a wide range of biological processes. Consistently, activity deregulation of these complexes is associated with many diseases. The increasing knowledge on SNP had in turn validated these molecular complexes as novel therapeutic targets. Targeting SNP activities by small molecules, antibodies and peptides or by soluble semaphorins have been proposed as new therapeutic approach. This review is focusing on the latest demonstration of this potential and discusses some of the key questions that need to be addressed before translating SNP targeting into clinically relevant approaches.
Collapse
Affiliation(s)
- Lionel A. T. Meyer
- INSERM U1109 – MN3T Lab, Fédération de Médecine Translationnelle, Labex Medalis, University of Strasbourg, France
| | - Justine Fritz
- INSERM U1109 – MN3T Lab, Fédération de Médecine Translationnelle, Labex Medalis, University of Strasbourg, France
| | - Marie Pierdant-Mancera
- INSERM U1109 – MN3T Lab, Fédération de Médecine Translationnelle, Labex Medalis, University of Strasbourg, France
| | - Dominique Bagnard
- INSERM U1109 – MN3T Lab, Fédération de Médecine Translationnelle, Labex Medalis, University of Strasbourg, France
| |
Collapse
|
33
|
Barton R, Khakbaz P, Bera I, Klauda JB, Iovine MK, Berger BW. Interplay of Specific Trans- and Juxtamembrane Interfaces in Plexin A3 Dimerization and Signal Transduction. Biochemistry 2016; 55:4928-38. [PMID: 27508400 DOI: 10.1021/acs.biochem.6b00517] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Plexins are transmembrane proteins that serve as guidance receptors during angiogenesis, lymphangiogenesis, neuronal development, and zebrafish fin regeneration, with a putative role in cancer metastasis. Receptor dimerization or clustering, induced by extracellular ligand binding but modulated in part by the plexin transmembrane (TM) and juxtamembrane (JM) domains, is thought to drive plexin activity. Previous studies indicate that isolated plexin TM domains interact through a conserved, small-x3-small packing motif, and the cytosolic JM region interacts through a hydrophobic heptad repeat; however, the roles and interplay of these regions in plexin signal transduction remain unclear. Using an integrated experimental and simulation approach, we find disruption of the small-x3-small motifs in the Danio rerio Plexin A3 TM domain enhances dimerization of the TM-JM domain by enhancing JM-mediated dimerization. Furthermore, mutations of the cytosolic JM heptad repeat that disrupt dimerization do so even in the presence of TM domain mutations. However, mutations to the small-x3-small TM interfaces also disrupt Plexin A3 signaling in a zebrafish axonal guidance assay, indicating the importance of this TM interface in signal transduction. Collectively, our experimental and simulation results demonstrate that multiple TM and JM interfaces exist in the Plexin A3 homodimer, and these interfaces independently regulate dimerization that is important in Plexin A3 signal transduction.
Collapse
Affiliation(s)
- Rachael Barton
- Department of Chemical and Biomolecular Engineering, Lehigh University , Bethlehem, Pennsylvania 18015, United States
| | - Pouyan Khakbaz
- Department of Chemical and Biomolecular Engineering, University of Maryland , College Park, Maryland 20742, United States
| | - Indrani Bera
- Department of Chemical and Biomolecular Engineering, University of Maryland , College Park, Maryland 20742, United States
| | - Jeffery B Klauda
- Department of Chemical and Biomolecular Engineering, University of Maryland , College Park, Maryland 20742, United States.,Biophysics Program, University of Maryland , College Park, Maryland 20742-2431, United States
| | - M Kathryn Iovine
- Department of Biological Sciences, Lehigh University , Bethlehem, Pennsylvania 18015, United States
| | - Bryan W Berger
- Department of Chemical and Biomolecular Engineering, Lehigh University , Bethlehem, Pennsylvania 18015, United States.,Program in Bioengineering, Lehigh University , Bethlehem, Pennsylvania 18015, United States
| |
Collapse
|
34
|
Stone TA, Deber CM. Therapeutic design of peptide modulators of protein-protein interactions in membranes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1859:577-585. [PMID: 27580024 DOI: 10.1016/j.bbamem.2016.08.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 08/22/2016] [Accepted: 08/24/2016] [Indexed: 12/12/2022]
Abstract
Membrane proteins play the central roles in a variety of cellular processes, ranging from nutrient uptake and signalling, to cell-cell communication. Their biological functions are directly related to how they fold and assemble; defects often lead to disease. Protein-protein interactions (PPIs) within the membrane are therefore of great interest as therapeutic targets. Here we review the progress in the application of membrane-insertable peptides for the disruption or stabilization of membrane-based PPIs. We describe the design and preparation of transmembrane peptide mimics; and of several categories of peptidomimetics used for study, including d-enantiomers, non-natural amino acids, peptoids, and β-peptides. Further aspects of the review describe modifications to membrane-insertable peptides, including lipidation and cyclization via hydrocarbon stapling. These approaches provide a pathway toward the development of metabolically stable, non-toxic, and efficacious peptide modulators of membrane-based PPIs. This article is part of a Special Issue entitled: Lipid order/lipid defects and lipid-control of protein activity edited by Dirk Schneider.
Collapse
Affiliation(s)
- Tracy A Stone
- Division of Molecular Structure & Function, Research Institute, Hospital for Sick Children, Toronto M5G 0A4, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto M5S 1A8, Ontario, Canada
| | - Charles M Deber
- Division of Molecular Structure & Function, Research Institute, Hospital for Sick Children, Toronto M5G 0A4, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto M5S 1A8, Ontario, Canada.
| |
Collapse
|
35
|
Barton R, Driscoll A, Flores S, Mudbhari D, Collins T, Iovine MK, Berger BW. Cysteines in the neuropilin-2 MAM domain modulate receptor homooligomerization and signal transduction. Biopolymers 2016; 104:371-8. [PMID: 25656526 DOI: 10.1002/bip.22619] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 01/14/2015] [Accepted: 01/24/2015] [Indexed: 01/01/2023]
Abstract
Neuropilins (NRPs) are transmembrane receptors involved in angiogenesis, lymphangiogenesis, and neuronal development as well as in cancer metastasis. Previous studies suggest that NRPs exist in heteromeric complexes with vascular endothelial growth factors (VEGFs) and VEGF receptors as well as plexins and semaphorins. We determined via site-directed mutagenesis and bioluminescent resonance energy transfer assays that a conserved cysteine (C711) in the Danio rerio NRP2a MAM (meprin, A-5 protein, and protein tyrosine phosphatase μ) domain modulates NRP2a homomeric interactions. Mutation of this residue also disrupts semaphorin-3F binding in NRP2a-transfected COS-7 cells and prevents the NRP2a overexpression effects in a zebrafish vascular model. Collectively, our results indicate the MAM domain plays an important role in defining the NRP2 homodimer structure, which is important for semaphorin-dependent signal transduction via NRP2.
Collapse
Affiliation(s)
- Rachael Barton
- Department of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, Pennsylvania
| | - Alyssa Driscoll
- Program in Bioengineering, Lehigh University, Bethlehem, Pennsylvania
| | - Samuel Flores
- Department of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania
| | - Durlav Mudbhari
- Department of Mechanical Engineering, Lehigh University, Bethlehem, Pennsylvania
| | - Theresa Collins
- Department of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania
| | - M Kathryn Iovine
- Department of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania
| | - Bryan W Berger
- Department of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, Pennsylvania.,Program in Bioengineering, Lehigh University, Bethlehem, Pennsylvania
| |
Collapse
|
36
|
Roth L, Prahst C, Ruckdeschel T, Savant S, Weström S, Fantin A, Riedel M, Héroult M, Ruhrberg C, Augustin HG. Neuropilin-1 mediates vascular permeability independently of vascular endothelial growth factor receptor-2 activation. Sci Signal 2016; 9:ra42. [PMID: 27117252 DOI: 10.1126/scisignal.aad3812] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Neuropilin-1 (NRP1) regulates developmental and pathological angiogenesis, arteriogenesis, and vascular permeability, acting as a coreceptor for semaphorin 3A (Sema3A) and the 165-amino acid isoform of vascular endothelial growth factor A (VEGF-A165). NRP1 is also the receptor for the CendR peptides, a class of cell- and tissue-penetrating peptides with a specific R-x-x-R carboxyl-terminal motif. Because the cytoplasmic domain of NRP1 lacks catalytic activity, NRP1 is mainly thought to act through the recruitment and binding to other receptors. We report here that the NRP1 intracellular domain mediates vascular permeability. Stimulation with VEGF-A165, a ligand-blocking antibody, and a CendR peptide led to NRP1 accumulation at cell-cell contacts in endothelial cell monolayers, increased cellular permeability in vitro and vascular leakage in vivo. Biochemical analyses, VEGF receptor-2 (VEGFR-2) silencing, and the use of a specific VEGFR blocker established that the effects induced by the CendR peptide and the antibody were independent of VEGFR-2. Moreover, leakage assays in mice expressing a mutant NRP1 lacking the cytoplasmic domain revealed that this domain was required for NRP1-induced vascular permeability in vivo. Hence, these data define a vascular permeability pathway mediated by NRP1 but independent of VEGFR-2 activation.
Collapse
Affiliation(s)
- Lise Roth
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany. Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Claudia Prahst
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany
| | - Tina Ruckdeschel
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany
| | - Soniya Savant
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany
| | - Simone Weström
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany
| | - Alessandro Fantin
- UCL Institute of Ophthalmology, University College London, EC1V 9EL London, UK
| | - Maria Riedel
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany
| | - Mélanie Héroult
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, EC1V 9EL London, UK
| | - Hellmut G Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany. Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany.
| |
Collapse
|
37
|
Zheng JS, He Y, Zuo C, Cai XY, Tang S, Wang ZA, Zhang LH, Tian CL, Liu L. Robust Chemical Synthesis of Membrane Proteins through a General Method of Removable Backbone Modification. J Am Chem Soc 2016; 138:3553-61. [PMID: 26943264 DOI: 10.1021/jacs.6b00515] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chemical protein synthesis can provide access to proteins with post-translational modifications or site-specific labelings. Although this technology is finding increasing applications in the studies of water-soluble globular proteins, chemical synthesis of membrane proteins remains elusive. In this report, a general and robust removable backbone modification (RBM) method is developed for the chemical synthesis of membrane proteins. This method uses an activated O-to-N acyl transfer auxiliary to install in the Fmoc solid-phase peptide synthesis process a RBM group with switchable reactivity toward trifluoroacetic acid. The method can be applied to versatile membrane proteins because the RBM group can be placed at any primary amino acid. With RBM, the membrane proteins and their segments behave almost as if they were water-soluble peptides and can be easily handled in the process of ligation, purification, and mass characterizations. After the full-length protein is assembled, the RBM group can be readily removed by trifluoroacetic acid. The efficiency and usefulness of the new method has been demonstrated by the successful synthesis of a two-transmembrane-domain protein (HCV p7 ion channel) with site-specific isotopic labeling and a four-transmembrane-domain protein (multidrug resistance transporter EmrE). This method enables practical synthesis of small- to medium-sized membrane proteins or membrane protein domains for biochemical and biophysical studies.
Collapse
Affiliation(s)
- Ji-Shen Zheng
- High Magnetic Field Laboratory, Chinese Academy of Sciences, and School of Life Sciences, University of Science and Technology of China , Hefei 230031, China
| | - Yao He
- High Magnetic Field Laboratory, Chinese Academy of Sciences, and School of Life Sciences, University of Science and Technology of China , Hefei 230031, China
| | - Chao Zuo
- Tsinghua-Peking Center for Life Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University , Beijing 100084, China
| | - Xiao-Ying Cai
- High Magnetic Field Laboratory, Chinese Academy of Sciences, and School of Life Sciences, University of Science and Technology of China , Hefei 230031, China
| | - Shan Tang
- Tsinghua-Peking Center for Life Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University , Beijing 100084, China
| | - Zhipeng A Wang
- Tsinghua-Peking Center for Life Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University , Beijing 100084, China
| | - Long-Hua Zhang
- High Magnetic Field Laboratory, Chinese Academy of Sciences, and School of Life Sciences, University of Science and Technology of China , Hefei 230031, China
| | - Chang-Lin Tian
- High Magnetic Field Laboratory, Chinese Academy of Sciences, and School of Life Sciences, University of Science and Technology of China , Hefei 230031, China
| | - Lei Liu
- Tsinghua-Peking Center for Life Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University , Beijing 100084, China
| |
Collapse
|
38
|
Raimondi C, Brash JT, Fantin A, Ruhrberg C. NRP1 function and targeting in neurovascular development and eye disease. Prog Retin Eye Res 2016; 52:64-83. [PMID: 26923176 PMCID: PMC4854174 DOI: 10.1016/j.preteyeres.2016.02.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 02/08/2016] [Accepted: 02/10/2016] [Indexed: 12/19/2022]
Abstract
Neuropilin 1 (NRP1) is expressed by neurons, blood vessels, immune cells and many other cell types in the mammalian body and binds a range of structurally and functionally diverse extracellular ligands to modulate organ development and function. In recent years, several types of mouse knockout models have been developed that have provided useful tools for experimental investigation of NRP1 function, and a multitude of therapeutics targeting NRP1 have been designed, mostly with the view to explore them for cancer treatment. This review provides a general overview of current knowledge of the signalling pathways that are modulated by NRP1, with particular focus on neuronal and vascular roles in the brain and retina. This review will also discuss the potential of NRP1 inhibitors for the treatment for neovascular eye diseases.
Collapse
Affiliation(s)
- Claudio Raimondi
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - James T Brash
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Alessandro Fantin
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK.
| |
Collapse
|
39
|
Abstract
The majority of therapeutics target membrane proteins, accessible on the surface of cells, to alter cellular signaling. Cells use membrane proteins to transduce signals into cells, transport ions and molecules, bind cells to a surface or substrate, and catalyze reactions. Newly devised technologies allow us to drug conventionally "undruggable" regions of membrane proteins, enabling modulation of protein-protein, protein-lipid, and protein-nucleic acid interactions. In this review, we survey the state of the art of high-throughput screening and rational design in drug discovery, and we evaluate the advances in biological understanding and technological capacity that will drive pharmacotherapy forward against unorthodox membrane protein targets.
Collapse
Affiliation(s)
- Hang Yin
- Department of Chemistry and Biochemistry.,BioFrontiers Institute, and.,Center of Basic Molecular Science, Department of Chemistry, Tsinghua University, Beijing 100082, China
| | - Aaron D Flynn
- BioFrontiers Institute, and.,Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado 80309; ,
| |
Collapse
|
40
|
Bi G, Liebrand TWH, Bye RR, Postma J, van der Burgh AM, Robatzek S, Xu X, Joosten MHAJ. SOBIR1 requires the GxxxG dimerization motif in its transmembrane domain to form constitutive complexes with receptor-like proteins. MOLECULAR PLANT PATHOLOGY 2016; 17:96-107. [PMID: 25891985 PMCID: PMC6638328 DOI: 10.1111/mpp.12266] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Receptor-like proteins (RLPs), forming an important group of transmembrane receptors in plants, play roles in development and immunity. RLPs contain extracellular leucine-rich repeats (LRRs) and, in contrast with receptor-like kinases (RLKs), lack a cytoplasmic kinase required for the initiation of downstream signalling. Recent studies have revealed that the RLK SOBIR1/EVR (SUPPRESSOR OF BIR1-1/EVERSHED) specifically interacts with RLPs. SOBIR1 stabilizes RLPs and is required for their function. However, the mechanism by which SOBIR1 associates with RLPs and regulates RLP function remains unknown. The Cf immune receptors of tomato (Solanum lycopersicum), mediating resistance to the fungus Cladosporium fulvum, are RLPs that also interact with SOBIR1. Here, we show that both the LRR and kinase domain of SOBIR1 are dispensable for association with the RLP Cf-4, whereas the highly conserved GxxxGxxxG motif present in the transmembrane domain of SOBIR1 is essential for its interaction with Cf-4 and additional RLPs. Complementation assays in Nicotiana benthamiana, in which endogenous SOBIR1 levels were knocked down by virus-induced gene silencing, showed that the LRR domain as well as the kinase activity of SOBIR1 are required for the Cf-4/Avr4-triggered hypersensitive response (HR). In contrast, the LRRs and kinase activity of SOBIR1 are not required for facilitation of Cf-4 accumulation. Together, these results suggest that, in addition to being a stabilizing scaffold for RLPs, SOBIR1 is also required for the initiation of downstream signalling through its kinase domain.
Collapse
Affiliation(s)
- Guozhi Bi
- College of Horticulture, Northeast Agricultural University, Harbin, 150030, China
- Laboratory of Phytopathology, Wageningen University, Droevendaalsesteeg 1, 6708 PB, Wageningen, the Netherlands
| | - Thomas W H Liebrand
- Laboratory of Phytopathology, Wageningen University, Droevendaalsesteeg 1, 6708 PB, Wageningen, the Netherlands
| | - Ruby R Bye
- Laboratory of Phytopathology, Wageningen University, Droevendaalsesteeg 1, 6708 PB, Wageningen, the Netherlands
| | - Jelle Postma
- The Sainsbury Laboratory, Norwich Research Park, Norwich, NR4 7UH, UK
| | - Aranka M van der Burgh
- Laboratory of Phytopathology, Wageningen University, Droevendaalsesteeg 1, 6708 PB, Wageningen, the Netherlands
| | - Silke Robatzek
- The Sainsbury Laboratory, Norwich Research Park, Norwich, NR4 7UH, UK
| | - Xiangyang Xu
- College of Horticulture, Northeast Agricultural University, Harbin, 150030, China
| | - Matthieu H A J Joosten
- Laboratory of Phytopathology, Wageningen University, Droevendaalsesteeg 1, 6708 PB, Wageningen, the Netherlands
| |
Collapse
|
41
|
Li X, Parker MW, Vander Kooi CW. Control of cellular motility by neuropilin-mediated physical interactions. Biomol Concepts 2015; 5:157-66. [PMID: 25018786 DOI: 10.1515/bmc-2013-0035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The neuropilin (Nrp) family consists of multifunctional cell surface receptors with critical roles in a number of different cell and tissue types. A core aspect of Nrp function is in ligand-dependent cellular migration, where it controls the multistep process of cellular motility through integration of ligand binding and receptor signaling. At a molecular level, the role of Nrp in migration is intimately connected to the control of adhesive interactions and cytoskeletal reorganization. Here, we review the physiological role of Nrp in cellular adhesion and motility in the cardiovascular and nervous systems. We also discuss the emerging pathological role of Nrp in tumor cell migration and metastasis, providing motivation for continued efforts toward developing Nrp inhibitors.
Collapse
|
42
|
Abstract
Transmembrane (TM) helices of integral membrane proteins can facilitate strong and specific noncovalent protein-protein interactions. Mutagenesis and structural analyses have revealed numerous examples in which the interaction between TM helices of single-pass membrane proteins is dependent on a GxxxG or (small)xxx(small) motif. It is therefore tempting to use the presence of these simple motifs as an indicator of TM helix interactions. In this Current Topic review, we point out that these motifs are quite common, with more than 50% of single-pass TM domains containing a (small)xxx(small) motif. However, the actual interaction strength of motif-containing helices depends strongly on sequence context and membrane properties. In addition, recent studies have revealed several GxxxG-containing TM domains that interact via alternative interfaces involving hydrophobic, polar, aromatic, or even ionizable residues that do not form recognizable motifs. In multipass membrane proteins, GxxxG motifs can be important for protein folding, and not just oligomerization. Our current knowledge thus suggests that the presence of a GxxxG motif alone is a weak predictor of protein dimerization in the membrane.
Collapse
Affiliation(s)
- Mark G Teese
- Lehrstuhl für Chemie der Biopolymere, Technische Universität München , 85354 Freising, Germany.,Center for Integrated Protein Science Munich (CIPSM) , 81377 Munich, Germany
| | - Dieter Langosch
- Lehrstuhl für Chemie der Biopolymere, Technische Universität München , 85354 Freising, Germany.,Center for Integrated Protein Science Munich (CIPSM) , 81377 Munich, Germany
| |
Collapse
|
43
|
Abstract
Neuropilins (NRPs) are highly conserved transmembrane glycoproteins that possess pleiotropic functions. Neuropilin-1 (NRP1) and its homologue neuropilin-2 interact as coreceptors with both class 3 semaphorins and vascular endothelial growth factor and are involved in neuronal guidance and angiogenesis, respectively. The contribution of NRPs to tumor angiogenesis has been highlighted in previous studies, leading to the development of NRP antagonists as novel anti-angiogenesis therapies. However, more recent studies have demonstrated that NRPs have a much broader spectrum of activity in the integration of different pathways in physiological and pathological conditions. A few studies investigated the role of NRPs in both malignant and non-neoplastic liver diseases. In normal liver, NRP1 is expressed in hepatic stellate cells and liver sinusoidal endothelial cells. NRP1 expression in hepatocytes has been associated with malignant transformation and may play an important role in tumor behavior. A contribution of NRPs in sinusoidal remodeling during liver regeneration has been also noted. Studies in chronic liver diseases have indicated that, besides its influence on angiogenesis, NRP1 might contribute to the progression of liver fibrosis owing to its effects on other growth factors, including transforming growth factor β1. As a result, NRP1 has been identified as a promising therapeutic target for future antifibrotic therapies based on the simultaneous blockade of multiple growth factor signaling pathways. In this review, the structure of NRPs and their interactions with various ligands and associated cell surface receptors are described briefly. The current understanding of the roles of the NRPs in liver diseases including tumors, regeneration and fibrogenesis, are also summarized.
Collapse
|
44
|
Bondeva T, Wolf G. Role of Neuropilin-1 in Diabetic Nephropathy. J Clin Med 2015; 4:1293-311. [PMID: 26239560 PMCID: PMC4485001 DOI: 10.3390/jcm4061293] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 05/28/2015] [Accepted: 06/09/2015] [Indexed: 02/07/2023] Open
Abstract
Diabetic nephropathy (DN) often develops in patients suffering from type 1 or type 2 diabetes mellitus. DN is characterized by renal injury resulting in proteinuria. Neuropilin-1 (NRP-1) is a single-pass transmembrane receptor protein devoid of enzymatic activity. Its large extracellular tail is structured in several domains, thereby allowing the molecule to interact with multiple ligands linking NRP-1 to different pathways through its signaling co-receptors. NRP-1’s role in nervous system development, immunity, and more recently in cancer, has been extensively investigated. Although its relation to regulation of apoptosis and cytoskeleton organization of glomerular vascular endothelial cells was reported, its function in diabetes mellitus and the development of DN is less clear. Several lines of evidence demonstrate a reduced NRP-1 expression in glycated-BSA cultured differentiated podocytes as well as in glomeruli from db/db mice (a model of type 2 Diabetes) and in diabetic patients diagnosed with DN. In vitro studies of podocytes implicated NRP-1 in the regulation of podocytes’ adhesion to extracellular matrix proteins, cytoskeleton reorganization, and apoptosis via not completely understood mechanisms. However, the exact role of NRP-1 during the onset of DN is not yet understood. This review intends to shed more light on NRP-1 and to present a link between NRP-1 and its signaling complexes in the development of DN.
Collapse
Affiliation(s)
- Tzvetanka Bondeva
- Department of Internal Medicine III, University Hospital Jena, Jena, 07747, Germany.
| | - Gunter Wolf
- Department of Internal Medicine III, University Hospital Jena, Jena, 07747, Germany.
| |
Collapse
|
45
|
Hara T, Huang Y, Ito A, Kawakami T, Hojo H, Murata M. Trifluoroethanol-containing RP-HPLC mobile phases for the separation of transmembrane peptides human glycophorin-A, integrin alpha-1, and p24: analysis and prevention of potential side reactions due to formic acid. J Pept Sci 2014; 21:61-70. [DOI: 10.1002/psc.2717] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 10/19/2014] [Accepted: 10/23/2014] [Indexed: 11/10/2022]
Affiliation(s)
- Toshiaki Hara
- Department of Chemistry, Graduate School of Science; Osaka University; Toyonaka Osaka 560-0043 Japan
- JST ERATO, Lipid Active Structure Project; Osaka University; Toyonaka Osaka 560-0043 Japan
- Project Research Centre for Fundamental Science; Osaka University; 1-1 Machikaneyama Toyonaka Osaka 560-0043 Japan
| | - Yue Huang
- Department of Chemistry, Graduate School of Science; Osaka University; Toyonaka Osaka 560-0043 Japan
| | - Akihiro Ito
- Department of Chemistry, Graduate School of Science; Osaka University; Toyonaka Osaka 560-0043 Japan
| | - Toru Kawakami
- Institute for Protein Research; Osaka University; 3-2 Yamadaoka Suita Osaka 565-0871 Japan
| | - Hironobu Hojo
- Institute for Protein Research; Osaka University; 3-2 Yamadaoka Suita Osaka 565-0871 Japan
| | - Michio Murata
- Department of Chemistry, Graduate School of Science; Osaka University; Toyonaka Osaka 560-0043 Japan
- JST ERATO, Lipid Active Structure Project; Osaka University; Toyonaka Osaka 560-0043 Japan
- Project Research Centre for Fundamental Science; Osaka University; 1-1 Machikaneyama Toyonaka Osaka 560-0043 Japan
| |
Collapse
|
46
|
Sawma P, Roth L, Blanchard C, Bagnard D, Crémel G, Bouveret E, Duneau JP, Sturgis JN, Hubert P. Evidence for new homotypic and heterotypic interactions between transmembrane helices of proteins involved in receptor tyrosine kinase and neuropilin signaling. J Mol Biol 2014; 426:4099-4111. [PMID: 25315821 DOI: 10.1016/j.jmb.2014.10.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 09/26/2014] [Accepted: 10/08/2014] [Indexed: 12/31/2022]
Abstract
Signaling in eukaryotic cells frequently relies on dynamic interactions of single-pass membrane receptors involving their transmembrane (TM) domains. To search for new such interactions, we have developed a bacterial two-hybrid system to screen for both homotypic and heterotypic interactions between TM helices. We have explored the dimerization of TM domains from 16 proteins involved in both receptor tyrosine kinase and neuropilin signaling. This study has revealed several new interactions. We found that the TM domain of Mucin-4, a putative intramembrane ligand for erbB2, dimerizes not only with erbB2 but also with all four members of the erbB family. In the Neuropilin/Plexin family of receptors, we showed that the TM domains of Neuropilins 1 and 2 dimerize with themselves and also with Plexin-A1, Plexin-B1, and L1CAM, but we were unable to observe interactions with several other TM domains notably those of members of the VEGF receptor family. The potentially important Neuropilin 1/Plexin-A1 interaction was confirmed using a surface plasmon resonance assay. This work shows that TM domain interactions can be highly specific. Exploring further the propensities of TM helix-helix association in cell membrane should have important practical implications related to our understanding of the structure-function of bitopic proteins' assembly and subsequent function, especially in the regulation of signal transduction.
Collapse
Affiliation(s)
- Paul Sawma
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires, UMR 7255, Centre National de la Recherche Scientifique and Aix-Marseille University, 31 Chemin Joseph Aiguier, 13402 Marseille Cedex 20, France
| | - Lise Roth
- INSERM U 1109 and University of Strasbourg, 3 Avenue Molière, 67200 Strasbourg, France
| | - Cécile Blanchard
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires, UMR 7255, Centre National de la Recherche Scientifique and Aix-Marseille University, 31 Chemin Joseph Aiguier, 13402 Marseille Cedex 20, France
| | - Dominique Bagnard
- INSERM U 1109 and University of Strasbourg, 3 Avenue Molière, 67200 Strasbourg, France
| | - Gérard Crémel
- INSERM U 1109 and University of Strasbourg, 3 Avenue Molière, 67200 Strasbourg, France
| | - Emmanuelle Bouveret
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires, UMR 7255, Centre National de la Recherche Scientifique and Aix-Marseille University, 31 Chemin Joseph Aiguier, 13402 Marseille Cedex 20, France
| | - Jean-Pierre Duneau
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires, UMR 7255, Centre National de la Recherche Scientifique and Aix-Marseille University, 31 Chemin Joseph Aiguier, 13402 Marseille Cedex 20, France
| | - James N Sturgis
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires, UMR 7255, Centre National de la Recherche Scientifique and Aix-Marseille University, 31 Chemin Joseph Aiguier, 13402 Marseille Cedex 20, France
| | - Pierre Hubert
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires, UMR 7255, Centre National de la Recherche Scientifique and Aix-Marseille University, 31 Chemin Joseph Aiguier, 13402 Marseille Cedex 20, France.
| |
Collapse
|
47
|
Nasarre P, Gemmill RM, Drabkin HA. The emerging role of class-3 semaphorins and their neuropilin receptors in oncology. Onco Targets Ther 2014; 7:1663-87. [PMID: 25285016 PMCID: PMC4181631 DOI: 10.2147/ott.s37744] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The semaphorins, discovered over 20 years ago, are a large family of secreted or transmembrane and glycophosphatidylinositol -anchored proteins initially identified as axon guidance molecules crucial for the development of the nervous system. It has now been established that they also play important roles in organ development and function, especially involving the immune, respiratory, and cardiovascular systems, and in pathological disorders, including cancer. During tumor progression, semaphorins can have both pro- and anti-tumor functions, and this has created complexities in our understanding of these systems. Semaphorins may affect tumor growth and metastases by directly targeting tumor cells, as well as indirectly by interacting with and influencing cells from the micro-environment and vasculature. Mechanistically, semaphorins, through binding to their receptors, neuropilins and plexins, affect pathways involved in cell adhesion, migration, invasion, proliferation, and survival. Importantly, neuropilins also act as co-receptors for several growth factors and enhance their signaling activities, while class 3 semaphorins may interfere with this. In this review, we focus on the secreted class 3 semaphorins and their neuropilin co-receptors in cancer, including aspects of their signaling that may be clinically relevant.
Collapse
Affiliation(s)
- Patrick Nasarre
- Division of Hematology-Oncology, The Hollings Cancer Center and Medical University of South Carolina, Charleston, SC, USA
| | - Robert M Gemmill
- Division of Hematology-Oncology, The Hollings Cancer Center and Medical University of South Carolina, Charleston, SC, USA
| | - Harry A Drabkin
- Division of Hematology-Oncology, The Hollings Cancer Center and Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
48
|
Aci-Sèche S, Sawma P, Hubert P, Sturgis JN, Bagnard D, Jacob L, Genest M, Garnier N. Transmembrane recognition of the semaphorin co-receptors neuropilin 1 and plexin A1: coarse-grained simulations. PLoS One 2014; 9:e97779. [PMID: 24858828 PMCID: PMC4032258 DOI: 10.1371/journal.pone.0097779] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 04/23/2014] [Indexed: 12/13/2022] Open
Abstract
The cancer associated class 3 semaphorins require direct binding to neuropilins and association to plexins to trigger cell signaling. Here, we address the role of the transmembrane domains of neuropilin 1 and plexin A1 for the dimerization of the two receptors by characterizing the assembly in lipid bilayers using coarse-grained molecular dynamics simulations. From experimental evidence using a two-hybrid system showing the biochemical association of the two receptors transmembrane domains, we performed molecular simulations in DOPC and POPC demonstrating spontaneously assembly to form homodimers and heterodimers with a very high propensity for right-handed packing of the helices. Inversely, left-handed packing was observed with a very low propensity. This mode of packing was observed uniquely when the plexin A1 transmembrane domain was involved in association. Potential of mean force calculations were used to predict a hierarchy of self-association for the monomers: the two neuropilin 1 transmembrane domains strongly associated, neuropilin 1 and plexin A1 transmembrane domains associated less and the two plexin A1 transmembrane domains weakly but significantly associated. We demonstrated that homodimerization and heterodimerization are driven by GxxxG motifs, and that the sequence context modulates the packing mode of the plexin A1 transmembrane domains. This work presents major advances towards our understanding of membrane signaling platforms assembly through membrane domains and provides exquisite information for the design of antagonist drugs defining a novel class of therapeutic agents.
Collapse
Affiliation(s)
- Samia Aci-Sèche
- Centre de Biophysique Moléculaire, Centre National de la Recherche Scientifique UPR, Université d’Orléans, Orléans, France
| | - Paul Sawma
- Laboratoire d’Ingénierie des Systèmes Macromoléculaires UMR, Université d’Aix-Marseille, Marseille, France
| | - Pierre Hubert
- Laboratoire d’Ingénierie des Systèmes Macromoléculaires UMR, Université d’Aix-Marseille, Marseille, France
| | - James N. Sturgis
- Laboratoire d’Ingénierie des Systèmes Macromoléculaires UMR, Université d’Aix-Marseille, Marseille, France
| | - Dominique Bagnard
- Institut National de la Santé et de la Recherche Médicale, Labex Medalis, Université de Strasbourg, Strasbourg, France
| | - Laurent Jacob
- Institut National de la Santé et de la Recherche Médicale, Labex Medalis, Université de Strasbourg, Strasbourg, France
| | - Monique Genest
- Centre de Biophysique Moléculaire, Centre National de la Recherche Scientifique UPR, Université d’Orléans, Orléans, France
- * E-mail: (NG); (MG)
| | - Norbert Garnier
- Centre de Biophysique Moléculaire, Centre National de la Recherche Scientifique UPR, Université d’Orléans, Orléans, France
- * E-mail: (NG); (MG)
| |
Collapse
|
49
|
Sharonov GV, Bocharov EV, Kolosov PM, Astapova MV, Arseniev AS, Feofanov AV. Point mutations in dimerization motifs of the transmembrane domain stabilize active or inactive state of the EphA2 receptor tyrosine kinase. J Biol Chem 2014; 289:14955-64. [PMID: 24733396 DOI: 10.1074/jbc.m114.558783] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The EphA2 receptor tyrosine kinase plays a central role in the regulation of cell adhesion and guidance in many human tissues. The activation of EphA2 occurs after proper dimerization/oligomerization in the plasma membrane, which occurs with the participation of extracellular and cytoplasmic domains. Our study revealed that the isolated transmembrane domain (TMD) of EphA2 embedded into the lipid bicelle dimerized via the heptad repeat motif L(535)X3G(539)X2A(542)X3V(546)X2L(549) rather than through the alternative glycine zipper motif A(536)X3G(540)X3G(544) (typical for TMD dimerization in many proteins). To evaluate the significance of TMD interactions for full-length EphA2, we substituted key residues in the heptad repeat motif (HR variant: G539I, A542I, G553I) or in the glycine zipper motif (GZ variant: G540I, G544I) and expressed YFP-tagged EphA2 (WT, HR, and GZ variants) in HEK293T cells. Confocal microscopy revealed a similar distribution of all EphA2-YFP variants in cells. The expression of EphA2-YFP variants and their kinase activity (phosphorylation of Tyr(588) and/or Tyr(594)) and ephrin-A3 binding were analyzed with flow cytometry on a single cell basis. Activation of any EphA2 variant is found to occur even without ephrin stimulation when the EphA2 content in cells is sufficiently high. Ephrin-A3 binding is not affected in mutant variants. Mutations in the TMD have a significant effect on EphA2 activity. Both ligand-dependent and ligand-independent activities are enhanced for the HR variant and reduced for the GZ variant compared with the WT. These findings allow us to suggest TMD dimerization switching between the heptad repeat and glycine zipper motifs, corresponding to inactive and active receptor states, respectively, as a mechanism underlying EphA2 signal transduction.
Collapse
Affiliation(s)
- George V Sharonov
- From the Department of Structural Biology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of RAS, 117997 Moscow, Russia, the Faculty of Medicine, Moscow State University, 119992 Moscow, Russia
| | - Eduard V Bocharov
- From the Department of Structural Biology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of RAS, 117997 Moscow, Russia
| | - Peter M Kolosov
- the Department of Molecular Neurobiology, Institute of Higher Nervous Activity and Neurophysiology of RAS, 117485 Moscow, Russia, and
| | - Maria V Astapova
- From the Department of Structural Biology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of RAS, 117997 Moscow, Russia
| | - Alexander S Arseniev
- From the Department of Structural Biology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of RAS, 117997 Moscow, Russia
| | - Alexey V Feofanov
- From the Department of Structural Biology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of RAS, 117997 Moscow, Russia, the Biological Faculty, Moscow State University, 119992 Moscow, Russia
| |
Collapse
|
50
|
Chaudhary B, Khaled YS, Ammori BJ, Elkord E. Neuropilin 1: function and therapeutic potential in cancer. Cancer Immunol Immunother 2014; 63:81-99. [PMID: 24263240 PMCID: PMC11028473 DOI: 10.1007/s00262-013-1500-0] [Citation(s) in RCA: 170] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 11/10/2013] [Indexed: 12/15/2022]
Abstract
Neuropilin 1 (NRP1) is a transmembrane glycoprotein that acts as a co-receptor for a number of extracellular ligands including class III/IV semaphorins, certain isoforms of vascular endothelial growth factor and transforming growth factor beta. An exact understanding of the role of NRP1 in the immune system has been obscured by the differences in NRP1 expression observed between mice and humans. In mice, NRP1 is selectively expressed on thymic-derived Tregs and greatly enhances immunosuppressive function. In humans, NRP1 is expressed on plasmacytoid dendritic cells (pDCs) where it aids in priming immune responses and on a subset of T regulatory cells (Tregs) isolated from secondary lymph nodes. Preliminary studies that show NRP1 expression on T cells confers enhanced immunosuppressive activity. However, the mechanism by which this activity is mediated remains unclear. NRP1 expression has also been identified on activated T cells and Tregs isolated from inflammatory microenvironments, suggesting NRP1 might represent a novel T cell activation marker. Of clinical interest, NRP1 may enhance Treg tumour infiltration and a decrease in NRP1+ Tregs correlates with successful chemotherapy, suggesting a specific role for NRP1 in cancer pathology. As a therapeutic target, NRP1 allows simultaneous targeting of NRP1-expressing tumour vasculature, NRP1+ Tregs and pDCs. With the development of anti-NRP1 monoclonal antibodies and cell-penetrating peptides, NRP1 represents a promising new target for cancer therapies. This paper reviews current knowledge on the role and function of NRP1 in Tregs and pDCs, both in physiological and cancer settings, as well as its potential as a therapeutic target in cancer.
Collapse
Affiliation(s)
- Belal Chaudhary
- Biomedical Research Centre, School of Environment and Life Sciences, University of Salford, The Crescent, Peel Building G25, Manchester, M5 4WT UK
| | - Yazan S. Khaled
- Biomedical Research Centre, School of Environment and Life Sciences, University of Salford, The Crescent, Peel Building G25, Manchester, M5 4WT UK
- Institutes of Cancer, Inflammation & Repair, University of Manchester, Manchester, UK
| | - Basil J. Ammori
- Biomedical Research Centre, School of Environment and Life Sciences, University of Salford, The Crescent, Peel Building G25, Manchester, M5 4WT UK
- Institutes of Cancer, Inflammation & Repair, University of Manchester, Manchester, UK
| | - Eyad Elkord
- Biomedical Research Centre, School of Environment and Life Sciences, University of Salford, The Crescent, Peel Building G25, Manchester, M5 4WT UK
- Institutes of Cancer, Inflammation & Repair, University of Manchester, Manchester, UK
- College of Medicine and Health Sciences, United Arab Emirates University, PO Box 17666, Al Ain, United Arab Emirates
| |
Collapse
|