1
|
Smith CEL, Laugel-Haushalter V, Hany U, Best S, Taylor RL, Poulter JA, Wortmann SB, Feichtinger RG, Mayr JA, Al Bahlani S, Nikolopoulos G, Rigby A, Black GC, Watson CM, Mansour S, Inglehearn CF, Mighell AJ, Bloch-Zupan A. Biallelic variants in Plexin B2 ( PLXNB2) cause amelogenesis imperfecta, hearing loss and intellectual disability. J Med Genet 2024; 61:689-698. [PMID: 38458752 PMCID: PMC11228227 DOI: 10.1136/jmg-2023-109728] [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: 11/02/2023] [Accepted: 02/22/2024] [Indexed: 03/10/2024]
Abstract
BACKGROUND Plexins are large transmembrane receptors for the semaphorin family of signalling proteins. Semaphorin-plexin signalling controls cellular interactions that are critical during development as well as in adult life stages. Nine plexin genes have been identified in humans, but despite the apparent importance of plexins in development, only biallelic PLXND1 and PLXNA1 variants have so far been associated with Mendelian genetic disease. METHODS Eight individuals from six families presented with a recessively inherited variable clinical condition, with core features of amelogenesis imperfecta (AI) and sensorineural hearing loss (SNHL), with variable intellectual disability. Probands were investigated by exome or genome sequencing. Common variants and those unlikely to affect function were excluded. Variants consistent with autosomal recessive inheritance were prioritised. Variant segregation analysis was performed by Sanger sequencing. RNA expression analysis was conducted in C57Bl6 mice. RESULTS Rare biallelic pathogenic variants in plexin B2 (PLXNB2), a large transmembrane semaphorin receptor protein, were found to segregate with disease in all six families. The variants identified include missense, nonsense, splicing changes and a multiexon deletion. Plxnb2 expression was detected in differentiating ameloblasts. CONCLUSION We identify rare biallelic pathogenic variants in PLXNB2 as a cause of a new autosomal recessive, phenotypically diverse syndrome with AI and SNHL as core features. Intellectual disability, ocular disease, ear developmental abnormalities and lymphoedema were also present in multiple cases. The variable syndromic human phenotype overlaps with that seen in Plxnb2 knockout mice, and, together with the rarity of human PLXNB2 variants, may explain why pathogenic variants in PLXNB2 have not been reported previously.
Collapse
Affiliation(s)
- Claire E L Smith
- Institute of Medical Research, St James's University Hospital, University of Leeds Faculty of Medicine and Health, Leeds, UK
| | - Virginie Laugel-Haushalter
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258, CNRS-UMR7104, Université de Strasbourg, Strasbourg, France
| | - Ummey Hany
- Institute of Medical Research, St James's University Hospital, University of Leeds Faculty of Medicine and Health, Leeds, UK
| | - Sunayna Best
- Institute of Medical Research, St James's University Hospital, University of Leeds Faculty of Medicine and Health, Leeds, UK
- Yorkshire Regional Genetics Service, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Rachel L Taylor
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
- Division of Evolution and Genomic Sciences, Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, UK
- EMQN CIC, Manchester, UK
| | - James A Poulter
- Institute of Medical Research, St James's University Hospital, University of Leeds Faculty of Medicine and Health, Leeds, UK
| | - Saskia B Wortmann
- Department of Paediatrics, University Children's Hospital, Salzburger Landesklinken (SALK) and Paracelsus Medical University, Salzburg, Austria
- Amalia Children's Hospital, Radboudumc, Nijmegen, The Netherlands
| | - Rene G Feichtinger
- Department of Paediatrics, University Children's Hospital, Salzburger Landesklinken (SALK) and Paracelsus Medical University, Salzburg, Austria
| | - Johannes A Mayr
- Department of Paediatrics, University Children's Hospital, Salzburger Landesklinken (SALK) and Paracelsus Medical University, Salzburg, Austria
| | - Suhaila Al Bahlani
- Dental & OMFS Clinic, Al Nahdha Hospital, Government of Oman Ministry of Health, Muscat, Oman
| | | | - Alice Rigby
- Institute of Medical Research, St James's University Hospital, University of Leeds Faculty of Medicine and Health, Leeds, UK
- School of Dentistry, University of Leeds Faculty of Medicine and Health, Leeds, UK
| | - Graeme C Black
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
- Division of Evolution and Genomic Sciences, Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, UK
| | - Christopher M Watson
- Institute of Medical Research, St James's University Hospital, University of Leeds Faculty of Medicine and Health, Leeds, UK
- North East and Yorkshire Genomic Laboratory Hub, Central Lab, St James's University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Sahar Mansour
- Lymphovascular Research Unit, Molecular and Clinical Sciences Research Institute, St George's Hospital, University of London, London, UK
- SW Thames Regional Centre for Genomics, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Chris F Inglehearn
- Institute of Medical Research, St James's University Hospital, University of Leeds Faculty of Medicine and Health, Leeds, UK
| | - Alan J Mighell
- School of Dentistry, University of Leeds Faculty of Medicine and Health, Leeds, UK
| | - Agnès Bloch-Zupan
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258, CNRS-UMR7104, Université de Strasbourg, Strasbourg, France
- Faculté de Chirurgie Dentaire, Université de Strasbourg, Strasbourg, France
- Centre de référence des maladies rares orales et dentaires O-Rares, Filière Santé Maladies rares TETE COU, European Reference Network CRANIO, Pôle de Médecine et Chirurgie Bucco-dentaires, Hôpital Civil, Hôpitaux Universitaires de Strasbourg (HUS), Strasbourg, France
| |
Collapse
|
2
|
Deng AF, Wang FX, Wang SC, Zhang YZ, Bai L, Su JC. Bone-organ axes: bidirectional crosstalk. Mil Med Res 2024; 11:37. [PMID: 38867330 PMCID: PMC11167910 DOI: 10.1186/s40779-024-00540-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 05/31/2024] [Indexed: 06/14/2024] Open
Abstract
In addition to its recognized role in providing structural support, bone plays a crucial role in maintaining the functionality and balance of various organs by secreting specific cytokines (also known as osteokines). This reciprocal influence extends to these organs modulating bone homeostasis and development, although this aspect has yet to be systematically reviewed. This review aims to elucidate this bidirectional crosstalk, with a particular focus on the role of osteokines. Additionally, it presents a unique compilation of evidence highlighting the critical function of extracellular vesicles (EVs) within bone-organ axes for the first time. Moreover, it explores the implications of this crosstalk for designing and implementing bone-on-chips and assembloids, underscoring the importance of comprehending these interactions for advancing physiologically relevant in vitro models. Consequently, this review establishes a robust theoretical foundation for preventing, diagnosing, and treating diseases related to the bone-organ axis from the perspective of cytokines, EVs, hormones, and metabolites.
Collapse
Affiliation(s)
- An-Fu Deng
- Institute of Translational Medicine, Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Fu-Xiao Wang
- Institute of Translational Medicine, Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Si-Cheng Wang
- Institute of Translational Medicine, Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 200444, China
| | - Ying-Ze Zhang
- Department of Orthopaedics, the Third Hospital of Hebei Medical University, Orthopaedic Research Institution of Hebei Province, NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, 050051, China.
| | - Long Bai
- Institute of Translational Medicine, Organoid Research Center, Shanghai University, Shanghai, 200444, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China.
- School of Medicine, Shanghai University, Shanghai, 200444, China.
- Wenzhou Institute of Shanghai University, Wenzhou, 325000, Zhejiang, China.
| | - Jia-Can Su
- Institute of Translational Medicine, Organoid Research Center, Shanghai University, Shanghai, 200444, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China.
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
3
|
Greengard E, Williams R, Moriarity B, Liu X, Minard CG, Reid JM, Fisher T, Evans E, Pastore DR, Zauderer M, Voss S, Fox E, Weigel BJ. A phase 1/2 study of pepinemab in children, adolescents, or young adults with recurrent or refractory solid tumors: A children's oncology group consortium report (ADVL1614). Pediatr Blood Cancer 2024; 71:e30938. [PMID: 38520670 PMCID: PMC11187758 DOI: 10.1002/pbc.30938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/19/2024] [Accepted: 02/22/2024] [Indexed: 03/25/2024]
Abstract
PURPOSE Pepinemab, a humanized IgG4 monoclonal antibody, targets the SEMA4D (CD100) antigen to inhibit binding to its high-affinity receptors (plexin B1/PLXNB1, plexin B2/PLXNB2) and low-affinity receptor (CD72). SEMA4D blockade leads to increased cytotoxic T-cell infiltration, delayed tumor growth, and durable tumor rejection in murine tumor models. Pepinemab was well tolerated and improved T cell infiltration in clinical studies in adults with refractory tumors. SEMA4D was identified as a strong candidate proto-oncogene in a model of osteosarcoma. Based on these preclinical and clinical data, we conducted a phase 1/2 study to determine the recommended phase 2 dose (RP2D), pharmacokinetics, pharmacodynamics, and immunogenicity, of pepinemab in pediatric patients with recurrent/refractory solid tumors, and activity in osteosarcoma. EXPERIMENTAL DESIGN Pepinemab was administered intravenously on Days 1 and 15 of a 28-day cycle at 20 mg/kg, the adult RP2D. Part A (phase 1) used a Rolling 6 design; Part B (phase 2) used a Simon 2-stage design in patients with osteosarcoma. Pharmacokinetics and target saturation were evaluated in peripheral blood. RESULTS Pepinemab (20 mg/kg) was well tolerated and no dose-limiting toxicities were observed during Part A. There were no objective responses. Two patients with osteosarcoma achieved disease control and prolonged stable disease. Pepinemab pharmacokinetics were similar to adults. CONCLUSIONS Pepinemab (20 mg/kg) is safe, well tolerated and resulted in adequate and sustained target saturation in pediatric patients. Encouraging disease control in two patients with osteosarcoma warrants further investigation with novel combination strategies to modulate the tumor microenvironment and antitumor immune response. CLINICAL TRIAL REGISTRY This trial is registered as NCT03320330 at Clinicaltrials.gov. DISCLAIMER The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
Collapse
MESH Headings
- Adolescent
- Adult
- Child
- Child, Preschool
- Female
- Humans
- Male
- Young Adult
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Drug Resistance, Neoplasm
- Maximum Tolerated Dose
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/pathology
- Neoplasms/drug therapy
- Osteosarcoma/drug therapy
- Osteosarcoma/pathology
Collapse
Affiliation(s)
- Emily Greengard
- University of Minnesota Masonic Cancer Center, Minneapolis, MN
| | - Robin Williams
- University of Minnesota Masonic Cancer Center, Minneapolis, MN
| | | | | | | | | | | | | | | | | | | | | | - Brenda J Weigel
- University of Minnesota Masonic Cancer Center, Minneapolis, MN
| |
Collapse
|
4
|
Hoard TM, Liu K, Cadigan KM, Giger RJ, Allen BL. Semaphorin Receptors Antagonize Wnt Signaling Through Beta-Catenin Degradation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596372. [PMID: 38854152 PMCID: PMC11160715 DOI: 10.1101/2024.05.29.596372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Precise control of morphogen signaling levels is essential for proper development. An outstanding question is: what mechanisms ensure proper morphogen activity and correct cellular responses? Previous work has identified Semaphorin (SEMA) receptors, Neuropilins (NRPs) and Plexins (PLXNs), as positive regulators of the Hedgehog (HH) signaling pathway. Here, we provide evidence that NRPs and PLXNs antagonize Wnt signaling in both fibroblasts and epithelial cells. Further, Nrp1/2 deletion in fibroblasts results in elevated baseline Wnt pathway activity and increased maximal responses to Wnt stimulation. Notably, and in contrast to HH signaling, SEMA receptor-mediated Wnt antagonism is independent of primary cilia. Mechanistically, PLXNs and NRPs act downstream of Dishevelled (DVL) to destabilize β-catenin (CTNNB1) in a proteosome-dependent manner. Further, NRPs, but not PLXNs, act in a GSK3β/CK1-dependent fashion to antagonize Wnt signaling, suggesting distinct repressive mechanisms for these SEMA receptors. Overall, this study identifies SEMA receptors as novel Wnt pathway antagonists that may also play larger roles integrating signals from multiple inputs.
Collapse
Affiliation(s)
- Tyler M Hoard
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Katie Liu
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kenneth M Cadigan
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Roman J Giger
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Benjamin L Allen
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
5
|
Liu ZZ, Liu LY, Zhu LY, Zhu J, Luo JY, Wang YF, Xu HA. Plexin B3 guides axons to cross the midline in vivo. Front Cell Neurosci 2024; 18:1292969. [PMID: 38628398 PMCID: PMC11018898 DOI: 10.3389/fncel.2024.1292969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 03/11/2024] [Indexed: 04/19/2024] Open
Abstract
During the development of neural circuits, axons are guided by a variety of molecular cues to navigate through the brain and establish precise connections with correct partners at the right time and place. Many axon guidance cues have been identified and they play pleiotropic roles in not only axon guidance but also axon fasciculation, axon pruning, and synaptogenesis as well as cell migration, angiogenesis, and bone formation. In search of receptors for Sema3E in axon guidance, we unexpectedly found that Plexin B3 is highly expressed in retinal ganglion cells of zebrafish embryos when retinal axons are crossing the midline to form the chiasm. Plexin B3 has been characterized to be related to neurodevelopmental disorders. However, the investigation of its pathological mechanisms is hampered by the lack of appropriate animal model. We provide evidence that Plexin B3 is critical for axon guidance in vivo. Plexin B3 might function as a receptor for Sema3E while Neuropilin1 could be a co-receptor. The intracellular domain of Plexin B3 is required for Semaphorin signaling transduction. Our data suggest that zebrafish could be an ideal animal model for investigating the role and mechanisms of Sema3E and Plexin B3 in vivo.
Collapse
Affiliation(s)
- Zhi-Zhi Liu
- Institute of Biomedical Innovation, Nanchang University, Nanchang, China
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang, China
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric diseases, Nanchang, China
| | - Ling-Yan Liu
- Institute of Biomedical Innovation, Nanchang University, Nanchang, China
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang, China
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric diseases, Nanchang, China
| | - Lou-Yin Zhu
- Institute of Biomedical Innovation, Nanchang University, Nanchang, China
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang, China
| | - Jian Zhu
- Institute of Biomedical Innovation, Nanchang University, Nanchang, China
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang, China
| | - Jia-Yu Luo
- Institute of Biomedical Innovation, Nanchang University, Nanchang, China
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang, China
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric diseases, Nanchang, China
| | - Ye-Fan Wang
- Institute of Biomedical Innovation, Nanchang University, Nanchang, China
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang, China
| | - Hong A. Xu
- Institute of Biomedical Innovation, Nanchang University, Nanchang, China
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang, China
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric diseases, Nanchang, China
| |
Collapse
|
6
|
Jacob TV, Doshi GM. New Promising Routes in Peptic Ulcers: Toll-like Receptors and Semaphorins. Endocr Metab Immune Disord Drug Targets 2024; 24:865-878. [PMID: 37605412 DOI: 10.2174/1871530323666230821102718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 07/09/2023] [Accepted: 07/12/2023] [Indexed: 08/23/2023]
Abstract
Peptic ulcers (PU) are one of the commonest yet problematic diseases found to be existing in the majority of the population. Today, drugs from a wide range of therapeutic classes are available for the management of the disease. Still, the complications of the condition are difficult to tackle and the side effect profile is quite a concern. The literature indicates that Toll-like receptors (TLRs) and Semaphorins (SEMAs) have been under study for their various pharmacological actions over the past few decades. Both these signalling pathways are found to regulate immunological and inflammatory responses. Moreover, receptors and signalling molecules from the family of TLRs and SEMAs are found to have bacterial recognition and antibacterial properties which are essential in eradicating Helicobacter pylori (H. pylori), one of the major causative agents of PU. Our understanding of SEMAs, a class of proteins involved in cell signalling, is relatively less developed compared to TLRs, another class of proteins involved in the immune response. SEMAs and TLRs play different roles in biological processes, with SEMAs primarily involved in guiding cell migration and axon guidance during development, while TLRs are responsible for recognizing pathogens and initiating an immune response. Here, in this review, we will discuss in detail the signalling cascade of TLRs and SEMAs and thereby understand its association with PU for future therapeutic targeting. The review also aims at providing an overview of the study that has been into exploring the role of these signalling pathways in the management of PU.
Collapse
Affiliation(s)
- Teresa V Jacob
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, 400056, India
| | - Gaurav M Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, 400056, India
| |
Collapse
|
7
|
Wagner W, Ochman B, Wagner W. Semaphorin 6 Family-An Important Yet Overlooked Group of Signaling Proteins Involved in Cancerogenesis. Cancers (Basel) 2023; 15:5536. [PMID: 38067240 PMCID: PMC10705753 DOI: 10.3390/cancers15235536] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 01/05/2025] Open
Abstract
According to recent evidence, some groups of semaphorins (SEMAs) have been associated with cancer progression. These proteins are able to modulate the cellular signaling of particular receptor tyrosine kinases (RTKs) via the stimulation of SEMA-specific coreceptors, namely plexins (plexin-A, -B, -C, -D) and neuropilins (Np1, Np2), which share common domains with RTKs, leading to the coactivation of the latter receptors. MET, ERBB2, VEGFR2, PFGFR, and EGFR, among others, represent acknowledged targets of semaphorins that are often associated with tumor progression or poor prognosis. In particular, higher expression of SEMA6 family proteins in cancer cells and stromal cells of the cancer niche is often associated with enhanced tumor angiogenesis, metastasis, and resistance to anticancer therapy. Notably, high SEMA6 expression in malignant tumor cells such as melanoma, pleural mesothelioma, gastric cancer, lung adenocarcinoma, and glioblastoma may serve as a prognostic biomarker of tumor progression. To date, very few studies have focused on the mechanisms of transmembrane SEMA6-driven tumor progression and its underlying interplay with RTKs within the tumor microenvironment. This review presents the growing evidence in the literature on the complex and shaping role of SEMA6 family proteins in cancer responsiveness to environmental stimuli.
Collapse
Affiliation(s)
- Wiktor Wagner
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-808 Zabrze, Poland; (W.W.); (B.O.)
| | - Błażej Ochman
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-808 Zabrze, Poland; (W.W.); (B.O.)
| | - Waldemar Wagner
- Laboratory of Cellular Immunology, Institute of Medical Biology, Polish Academy of Sciences, 93-232 Łódź, Poland
- Department of Hormone Biochemistry, Medical University of Łódź, 90-752 Łódź, Poland
| |
Collapse
|
8
|
Matrone C, Ferretti G. Semaphorin 3A influences neuronal processes that are altered in patients with autism spectrum disorder: Potential diagnostic and therapeutic implications. Neurosci Biobehav Rev 2023; 153:105338. [PMID: 37524141 DOI: 10.1016/j.neubiorev.2023.105338] [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: 02/16/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/02/2023]
Abstract
Autism spectrum disorder (ASD) is a pervasive disorder that most frequently manifests in early childhood and lasts for their entire lifespan. Several behavioural traits characterise the phenotype of patients with ASD, including difficulties in reciprocal social communication as well as compulsive/repetitive stereotyped verbal and non-verbal behaviours. Although multiple hypotheses have been proposed to explain the aetiology of ASD and many resources have been used to improve our understanding of ASD, several aspects remain largely unexplored. Class 3 semaphorins (SEMA3) are secreted proteins involved in the organisation of structural and functional connectivity in the brain that regulate synaptic and dendritic development. Alterations in brain connectivity and aberrant neuronal development have been described in some patients with ASD. Mutations and polymorphisms in SEMA3A and alterations in its receptors and signalling have been associated with some neurological disorders such as schizophrenia and epilepsy, which are comorbidities in ASD, but also with ASD itself. In addition, SEMA3A is a key regulator of the immune response and neuroinflammatory processes, which have been found to be dysregulated in mothers of children who develop ASD and in affected patients. In this review, we highlight neurodevelopmental-related processes in which SEMA3A is involved, which are altered in ASD, and provide a viewpoint emphasising the development of strategies targeting changes in the SEMA3A signal to identify patterns of anomalies distinctive of ASD or to predict the prognosis of affected patients.
Collapse
Affiliation(s)
- Carmela Matrone
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy.
| | - Gabriella Ferretti
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy
| |
Collapse
|
9
|
Bica C, Tirpe A, Nutu A, Ciocan C, Chira S, Gurzau ES, Braicu C, Berindan-Neagoe I. Emerging roles and mechanisms of semaphorins activity in cancer. Life Sci 2023; 318:121499. [PMID: 36775114 DOI: 10.1016/j.lfs.2023.121499] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/08/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023]
Abstract
Semaphorins are regulatory molecules that are linked to the modulation of several cancer processes, such as angiogenesis, cancer cell invasiveness and metastasis, tumor growth, as well as cancer cell survival. Semaphorin (SEMA) activity depends on the cancer histotypes and their particularities. In broad terms, the effects of SEMAs result from their interaction with specific receptors/co-receptors - Plexins, Neuropilins and Integrins - and the subsequent effects upon the downstream effectors (e.g. PI3K/AKT, MAPK/ERK). The present article serves as an integrative review work, discussing the broad implications of semaphorins in cancer, focusing on cell proliferation/survival, angiogenesis, invasion, metastasis, stemness, and chemo-resistance/response whilst highlighting their heterogeneity as a family. Herein, we emphasized that semaphorins are largely implicated in cancer progression, interacting with the tumor microenvironment components. Whilst some SEMAs (e.g. SEMA3A, SEMA3B) function widely as tumor suppressors, others (e.g. SEMA3C) act as pro-tumor semaphorins. The differences observed in terms of the biological structure of SEMAs and the particularities of each cancer histotypes require that each semaphorin be viewed as a unique entity, and its roles must be researched accordingly. A more in-depth and comprehensive view of the molecular mechanisms that promote and sustain the malignant behavior of cancer cells is of utmost importance.
Collapse
Affiliation(s)
- Cecilia Bica
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania.
| | - Alexandru Tirpe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania; Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania.
| | - Andreea Nutu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania.
| | - Cristina Ciocan
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania.
| | - Sergiu Chira
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania.
| | - Eugen S Gurzau
- Cluj School of Public Health, College of Political, Administrative and Communication Sciences, Babes-Bolyai University, 7 Pandurilor Street, Cluj-Napoca, Romania; Environmental Health Center, 58 Busuiocului Street, 400240 Cluj-Napoca, Romania.
| | - Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania.
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania.
| |
Collapse
|
10
|
Zhang J, Han Y, Yan D, Zhou D, Yuan X, Zhao W, Zhang D. Identification of Key Genes Associated with Risk and Prognosis of Neuroblastoma. J Mol Neurosci 2022; 72:2398-2412. [PMID: 36443552 DOI: 10.1007/s12031-022-02087-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 11/16/2022] [Indexed: 11/30/2022]
Abstract
Neuroblastoma is a childhood malignancy with high morbidity and mortality. We identified key biomarkers associated with neuroblastoma risk and prognosis. The gene modules most associated with neuroblastoma risk were derived by WGCNA. Modular genes were intersected with differentially expressed genes between patients with high-risk (HR) and non-high-risk (NHR) to obtain risk genes, and enrichment analysis was performed. After incorporating risk genes into Cox regression analysis, LASSO algorithm, and K-M survival analysis, key genes were identified and introduced into four external datasets for validation. We performed short time-series expression miner analysis and single-sample genome enrichment analysis. Finally, we evaluated the difference in DNA methylation levels to identify meaningful methylation marks. We identified 5 key genes (ANO6, CPNE2, DST, PLXNC1, SCN3A) for neuroblastoma risk and prognosis, which correlated closely with known neuroblastoma biomarkers. All key genes showed a progressive downregulation trend with increasing risk levels of neuroblastoma. The immune infiltration of 14 immune cells was significantly different between HR-NB and NHR-NB, and most immune cells were negatively correlated with key genes. Furthermore, the expression of ANO6, CPNE2, DST, and PLXNC1 was modified by DNA methylation. This study identified 5 key genes for neuroblastoma risk and prognosis that were potential biomarkers.
Collapse
Affiliation(s)
- Jiao Zhang
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China.
| | - Yahui Han
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Dun Yan
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Diming Zhou
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Xiafei Yuan
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Wei Zhao
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Da Zhang
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| |
Collapse
|
11
|
Hang J, Wang J, Lu M, Xue Y, Qiao J, Tao L. Protein O-mannosylation across kingdoms and related diseases: From glycobiology to glycopathology. Biomed Pharmacother 2022; 148:112685. [PMID: 35149389 DOI: 10.1016/j.biopha.2022.112685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/29/2022] [Accepted: 02/01/2022] [Indexed: 11/18/2022] Open
Abstract
The post-translational glycosylation of proteins by O-linked α-mannose is conserved from bacteria to humans. Due to advances in high-throughput mass spectrometry-based approaches, a variety of glycoproteins are identified to be O-mannosylated. Various proteins with O-mannosylation are involved in biological processes, providing essential necessity for proper growth and development. In this review, we summarize the process and regulation of O-mannosylation. The multi-step O-mannosylation procedures are quite dynamic and complex, especially when considering the structural and functional inspection of the involved enzymes. The widely studied O-mannosylated proteins in human include α-Dystroglycan (α-DG), cadherins, protocadherins, and plexin, and their aberrant O-mannosylation are associated with many diseases. In addition, O-mannosylation also contributes to diverse functions in lower eukaryotes and prokaryotes. Finally, we present the relationship between O-mannosylation and gut microbiota (GM), and elucidate that O-mannosylation in microbiome is of great importance in the dynamic balance of GM. Our study provides an overview of the processes of O-mannosylation in mammalian cells and other organisms, and also associated regulated enzymes and biological functions, which could contribute to the understanding of newly discovered O-mannosylated glycoproteins.
Collapse
Affiliation(s)
- Jing Hang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
| | - Jinpeng Wang
- Department of Orthopedics, First Hospital of China Medical University, Shenyang 110001, China
| | - Minzhen Lu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
| | - Yuchuan Xue
- The First Department of Clinical Medicine, China Medical University, Shenyang 110001, China
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China.
| | - Lin Tao
- Department of Orthopedics, First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
12
|
Celus W, Oliveira AI, Rivis S, Van Acker HH, Landeloos E, Serneels J, Cafarello ST, Van Herck Y, Mastrantonio R, Köhler A, Garg AD, Flamand V, Tamagnone L, Marine JC, Matteo MD, Costa BM, Bechter O, Mazzone M. Plexin-A4 Mediates Cytotoxic T-cell Trafficking and Exclusion in Cancer. Cancer Immunol Res 2021; 10:126-141. [PMID: 34815265 DOI: 10.1158/2326-6066.cir-21-0061] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 09/07/2021] [Accepted: 11/16/2021] [Indexed: 11/16/2022]
Abstract
Cytotoxic T cell (CTL) infiltration of the tumor carries the potential to limit cancer progression, but their exclusion by the immunosuppressive tumor microenvironment hampers the efficiency of immunotherapy. Here, we show that expression of the axon guidance molecule Plexin-A4 (Plxna4) in CTLs, especially in effector/memory CD8+ T cells, is induced upon T-cell activation, sustained in the circulation, but reduced when entering the tumor bed. Therefore, we deleted Plxna4 and observed that Plxna4-deficient CTLs acquired improved homing capacity to the lymph nodes and to the tumor, as well as increased proliferation, both achieved through enhanced Rac1 activation. Mice with stromal or hematopoietic Plxna4 deletion exhibited enhanced CTL infiltration and impaired tumor growth. In a melanoma model, adoptive transfer of CTLs lacking Plxna4 prolonged survival and improved therapeutic outcome, which was even stronger when combined with anti-programmed cell death protein 1 (PD-1) treatment. PLXNA4 abundance in circulating CTLs was augmented in melanoma patients versus healthy volunteers but decreased after the first cycle of anti-PD-1, alone or in combination with anti-cytotoxic T-Lymphocyte Associated Protein 4 (CTLA-4), in those patients showing complete or partial response to the treatment. Altogether, our data suggest that Plxna4 acts as a "checkpoint," negatively regulating CTL migration and proliferation through cell-autonomous mechanisms independent of the interaction with host-derived Plxna4 ligands, semaphorins. These findings pave the way toward Plxna4-centric immunotherapies and propose Plxna4 detection in circulating CTLs as a potential way to monitor the response to immune checkpoint blockade in patients with metastatic melanoma.
Collapse
Affiliation(s)
- Ward Celus
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium. .,Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Ana I Oliveira
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium.,Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, KU Leuven, Leuven, Belgium.,Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, University of Minho, Braga, Portugal
| | - Silvia Rivis
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium.,Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Heleen H Van Acker
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium.,Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Ewout Landeloos
- Laboratory for Molecular Cancer Biology, Center for Cancer Biology, VIB, Leuven, Belgium.,Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Jens Serneels
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium.,Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Sarah Trusso Cafarello
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium.,Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Yannick Van Herck
- Department of General Medical Oncology, University Hospitals Leuven, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Roberta Mastrantonio
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Arnaud Köhler
- Institute for Medical Immunology, ULB-Center for Research in Immunology, Gosselies, Belgium.,Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
| | - Abhishek D Garg
- Laboratory of Cell Stress & Immunity, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Véronique Flamand
- Institute for Medical Immunology, ULB-Center for Research in Immunology, Gosselies, Belgium.,Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
| | - Luca Tamagnone
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, Center for Cancer Biology, VIB, Leuven, Belgium.,Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Mario Di Matteo
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium.,Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Bruno M Costa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, University of Minho, Braga, Portugal
| | - Oliver Bechter
- Department of General Medical Oncology, University Hospitals Leuven, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium. .,Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, KU Leuven, Leuven, Belgium
| |
Collapse
|
13
|
Liang Y, Meng K, Qiu R. Circular RNA Circ_0013958 Functions as a Tumor Promoter in Ovarian Cancer by Regulating miR-637/PLXNB2 Axis. Front Genet 2021; 12:644451. [PMID: 34367233 PMCID: PMC8334736 DOI: 10.3389/fgene.2021.644451] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/30/2021] [Indexed: 01/22/2023] Open
Abstract
Background: Circular RNAs (circRNAs) have emerged as important regulators in diverse human malignancies, including ovarian cancer (OC). This study was performed to explore the function and regulatory mechanism underlying circ_0013958 in OC progression. Methods: Quantitative real-time polymerase chain reaction (qRT-PCR) or Western blot assay was applied to examine the expression of circ_0013958, microRNA-637 (miR-637), and Plexin B2 (PLXNB2). The target relationship between miR-637 and circ_0013958 or PLXNB2 was verified by dual-luciferase reporter assay or RNA immunoprecipitation (RIP) assay. Cell Counting Kit-8 (CCK-8) and colony formation assays were employed to detect cell viability and clonogenicity ability, respectively. Cell migration and invasion were analyzed by Transwell assay. Cell apoptosis was monitored by flow cytometry. The role of circ_0013958 in vivo was determined by xenograft tumor assay. Results: Circ_0013958 and PLXNB2 were upregulated, while miR-637 was downregulated in OC tissues and cells. Circ_0013958 acted as a sponge for miR-637 to regulate the expression of PLXNB2 in OC cells. The repression effects of circ_0013958 knockdown on cell proliferation, migration, invasion, and apoptosis in OC cells were partly attenuated by the miR-637 inhibitor. And miR-637 targeted PLXNB2 to suppress OC cell proliferation, migration, and invasion. Moreover, circ_0013958 silencing blocked OC tumor growth in vivo. Conclusion: Circ_0013958 knockdown impeded OC development through modulating the miR-637/PLXNB2 axis, highlighting a therapeutic target for OC.
Collapse
Affiliation(s)
- Yanfei Liang
- Department of Gynecology, The Second Nanning People's Hospital, Nanning, China
| | - Kaiyi Meng
- Department of Gynecology, The Second Nanning People's Hospital, Nanning, China
| | - Rui Qiu
- Department of Gynecology, The Second Nanning People's Hospital, Nanning, China
| |
Collapse
|
14
|
Zhang YF, Zhang Y, Jia DD, Yang HY, Cheng MD, Zhu WX, Xin H, Li PF, Zhang YF. Insights into the regulatory role of Plexin D1 signalling in cardiovascular development and diseases. J Cell Mol Med 2021; 25:4183-4194. [PMID: 33837646 PMCID: PMC8093976 DOI: 10.1111/jcmm.16509] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/04/2021] [Accepted: 03/22/2021] [Indexed: 12/30/2022] Open
Abstract
Plexin D1 (PLXND1), which was previously thought to mediate semaphorin signalling, belongs to the Plexin family of transmembrane proteins. PLXND1 cooperates mostly with the coreceptor neuropilin and participates in many aspects of axonal guidance. PLXND1 can also act as both a tumour promoter and a tumour suppressor. Emerging evidence suggests that mutations in PLXND1 or Semaphorin 3E, the canonical ligand of PLXND1, can lead to serious cardiovascular diseases, such as congenital heart defects, CHARGE syndrome and systemic sclerosis. Upon ligand binding, PLXND1 can act as a GTPase‐activating protein (GAP) and modulate integrin‐mediated cell adhesion, cytoskeletal dynamics and cell migration. These effects may play regulatory roles in the development of the cardiovascular system and disease. The cardiovascular effects of PLXND1 signalling have gradually been elucidated. PLXND1 was recently shown to detect physical forces and translate them into intracellular biochemical signals in the context of atherosclerosis. Therefore, the role of PLXND1 in cardiovascular development and diseases is gaining research interest because of its potential as a biomarker and therapeutic target. In this review, we describe the cardiac effects, vascular effects and possible molecular mechanisms of PLXND1 signalling.
Collapse
Affiliation(s)
- Yi-Fei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yu Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Dong-Dong Jia
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Hong-Yu Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Meng-Die Cheng
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wen-Xiu Zhu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hui Xin
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Pei-Feng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yin-Feng Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
15
|
Plant T, Eamsamarng S, Sanchez-Garcia MA, Reyes L, Renshaw SA, Coelho P, Mirchandani AS, Morgan JM, Ellett FE, Morrison T, Humphries D, Watts ER, Murphy F, Raffo-Iraolagoitia XL, Zhang A, Cash JL, Loynes C, Elks PM, Van Eeden F, Carlin LM, Furley AJ, Whyte MK, Walmsley SR. Semaphorin 3F signaling actively retains neutrophils at sites of inflammation. J Clin Invest 2020; 130:3221-3237. [PMID: 32191647 PMCID: PMC7259996 DOI: 10.1172/jci130834] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 03/11/2020] [Indexed: 12/18/2022] Open
Abstract
Neutrophilic inflammation is central to disease pathogenesis, for example, in chronic obstructive pulmonary disease, yet the mechanisms that retain neutrophils within tissues remain poorly understood. With emerging evidence that axon guidance factors can regulate myeloid recruitment and that neutrophils can regulate expression of a class 3 semaphorin, SEMA3F, we investigated the role of SEMA3F in inflammatory cell retention within inflamed tissues. We observed that neutrophils upregulate SEMA3F in response to proinflammatory mediators and following neutrophil recruitment to the inflamed lung. In both zebrafish tail injury and murine acute lung injury models of neutrophilic inflammation, overexpression of SEMA3F delayed inflammation resolution with slower neutrophil migratory speeds and retention of neutrophils within the tissues. Conversely, constitutive loss of sema3f accelerated egress of neutrophils from the tail injury site in fish, whereas neutrophil-specific deletion of Sema3f in mice resulted in more rapid neutrophil transit through the airways, and significantly reduced time to resolution of the neutrophilic response. Study of filamentous-actin (F-actin) subsequently showed that SEMA3F-mediated retention is associated with F-actin disassembly. In conclusion, SEMA3F signaling actively regulates neutrophil retention within the injured tissues with consequences for neutrophil clearance and inflammation resolution.
Collapse
Affiliation(s)
- Tracie Plant
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Suttida Eamsamarng
- Department of Infection, Immunity and Cardiovascular Disease and
- Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Manuel A. Sanchez-Garcia
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Leila Reyes
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Stephen A. Renshaw
- Department of Infection, Immunity and Cardiovascular Disease and
- Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Patricia Coelho
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Ananda S. Mirchandani
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Jessie-May Morgan
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Felix E. Ellett
- Department of Infection, Immunity and Cardiovascular Disease and
- Bateson Centre, University of Sheffield, Sheffield, United Kingdom
- BioMEMS Resource Centre, Division of Surgery, Innovation and Bioengineering, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Burn Care, Shriners Hospitals for Children — Boston, Boston, Massachusetts, USA
| | - Tyler Morrison
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Duncan Humphries
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Emily R. Watts
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Fiona Murphy
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Ailiang Zhang
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Jenna L. Cash
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Catherine Loynes
- Department of Infection, Immunity and Cardiovascular Disease and
- Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Philip M. Elks
- Department of Infection, Immunity and Cardiovascular Disease and
- Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Freek Van Eeden
- Department of Infection, Immunity and Cardiovascular Disease and
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Leo M. Carlin
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Andrew J.W. Furley
- Bateson Centre, University of Sheffield, Sheffield, United Kingdom
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Moira K.B. Whyte
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease and
| | - Sarah R. Walmsley
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease and
| |
Collapse
|
16
|
Ho YC, Srinivasan RS. Lymphatic Vasculature in Energy Homeostasis and Obesity. Front Physiol 2020; 11:3. [PMID: 32038308 PMCID: PMC6987243 DOI: 10.3389/fphys.2020.00003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 01/03/2020] [Indexed: 12/14/2022] Open
Abstract
Obesity is a leading cause of cardiovascular diseases and cancer. Body mass is regulated by the balance between energy uptake and energy expenditure. The etiology of obesity is determined by multiple factors including genetics, nutrient absorption, and inflammation. Lymphatic vasculature is starting to be appreciated as a critical modulator of metabolism and obesity. The primary function of lymphatic vasculature is to maintain interstitial fluid homeostasis. Lymphatic vessels absorb fluids that extravasate from blood vessels and return them to blood circulation. In addition, lymphatic vessels absorb digested lipids from the intestine and regulate inflammation. Hence, lymphatic vessels could be an exciting target for treating obesity. In this article, we will review our current understanding regarding the relationship between lymphatic vasculature and obesity, and highlight some open questions.
Collapse
Affiliation(s)
- Yen-Chun Ho
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - R. Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
17
|
Torres-Salido MT, Sanchis M, Solé C, Moliné T, Vidal M, Vidal X, Solà A, Hotter G, Ordi-Ros J, Cortés-Hernández J. Urinary Neuropilin-1: A Predictive Biomarker for Renal Outcome in Lupus Nephritis. Int J Mol Sci 2019; 20:ijms20184601. [PMID: 31533337 PMCID: PMC6769814 DOI: 10.3390/ijms20184601] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/05/2019] [Accepted: 09/10/2019] [Indexed: 12/23/2022] Open
Abstract
At present, Lupus Nephritis (LN) is still awaiting a biomarker to better monitor disease activity, guide clinical treatment, and predict a patient’s long-term outcome. In the last decade, novel biomarkers have been identified to monitor the disease, but none have been incorporated into clinical practice. The transmembrane receptor neuropilin-1 (NRP-1) is highly expressed by mesangial cells and its genetic deletion results in proteinuric disease and glomerulosclerosis. NRP-1 is increased in kidney biopsies of LN. In this work we were interested in determining whether urinary NRP-1 levels could be a biomarker of clinical response in LN. Our results show that patients with active LN have increased levels of urinary NRP-1. When patients were divided according to clinical response, responders displayed higher urinary and tissue NRP-1 levels at the time of renal biopsy. Areas under the receiver operating characteristic curve, comparing baseline creatinine, proteinuria, urinary NRP-1, and VEGFA protein levels, showed NRP-1 to be an independent predictor for clinical response. In addition, in vitro studies suggest that NRP-1could promote renal recovery through endothelial proliferation and migration, mesangial migration and local T cell cytotoxicity. Based on these results, NRP-1 may be used as an early prognostic biomarker in LN.
Collapse
Affiliation(s)
| | - Mireia Sanchis
- Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute (VHIR), Rheumatology research group, Lupus Unit, 08035 Barcelona, Spain.
| | - Cristina Solé
- Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute (VHIR), Rheumatology research group, Lupus Unit, 08035 Barcelona, Spain.
| | - Teresa Moliné
- Hospital Universitari Vall d'Hebron, Department of Renal Pathology, 08035 Barcelona, Spain.
| | - Marta Vidal
- Hospital Universitari Vall d'Hebron, Department of Renal Pathology, 08035 Barcelona, Spain.
| | - Xavier Vidal
- Clinical Pharmacology Service, Department of Pharmacology, Therapeutics and Toxicology, Fundació Institut Català de Farmacologia, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain.
| | - Anna Solà
- Department of Experimental Pathology, IIBB-CSIC-IDIBAPS, 08036 Barcelona, Spain.
- CIBER-BBN, Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08036 Barcelona, Spain.
| | - Georgina Hotter
- Department of Experimental Pathology, IIBB-CSIC-IDIBAPS, 08036 Barcelona, Spain.
- CIBER-BBN, Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08036 Barcelona, Spain.
| | - Josep Ordi-Ros
- Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute (VHIR), Rheumatology research group, Lupus Unit, 08035 Barcelona, Spain.
| | - Josefina Cortés-Hernández
- Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute (VHIR), Rheumatology research group, Lupus Unit, 08035 Barcelona, Spain.
| |
Collapse
|
18
|
Semaphorin 3E regulates apoptosis in the intestinal epithelium during the development of colitis. Biochem Pharmacol 2019; 166:264-273. [PMID: 31170375 DOI: 10.1016/j.bcp.2019.05.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 05/30/2019] [Indexed: 12/14/2022]
Abstract
Semaphorin 3E (SEMA3E) has emerged as an axon-guiding molecule that regulates various biological processes including the immune responses and apoptosis. However, its role in the pathophysiology of colitis remains elusive. We investigated the role of SEMA3E in intestinal epithelial cells (IECs) activation, using biopsies from patients with active ulcerative colitis (UC), a mouse model of UC, and an in-vitro model of intestinal mucosal healing. In this study, we confirmed that the mRNA level of SEMA3E is reduced significantly in patients with UC and demonstrated a negative linear association between SEMA3E mRNA and p53-associated genes. In mice, genetic deletion of Sema3e resulted in an increase onset and severity of colitis, p53-associated genes, apoptosis, and IL-1beta production. Recombinant SEMA3E treatment protected against colitis and decreased these effects. Furthermore, in stimulated epithelial cells, recombinant SEMA3E treatment enhanced wound healing, resistance to oxidative stress and decreased apoptosis and p53-associated genes. Together, these findings identify SEMA3E as a novel regulator in intestinal inflammation that regulates IECs apoptosis and suggest a potential novel approach to treat UC.
Collapse
|
19
|
Höckel M, Behn U. The Order of Cancer: A Theory of Malignant Progression by Inverse Morphogenesis. Front Oncol 2019; 9:416. [PMID: 31192124 PMCID: PMC6548852 DOI: 10.3389/fonc.2019.00416] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 05/02/2019] [Indexed: 01/15/2023] Open
Abstract
Local spread patterns of malignant tumors follow permissive tissue territories, i.e., cancer fields, as shown for cervical and vulvar carcinoma. The cancer fields are associated in reverse order to the mature derivatives of the morphogenetic fields instrumental in the stepwise development of the tissue from which the tumor arose. This suggests that cancer progression may be linked to morphogenesis by inversion of the cellular bauplan sequence. Successive attractor transitions caused by proliferation-associated constraints of topobiological information processing are proposed for both morphogenesis and cancer. In morphogenesis these transitions sequentially activate bauplans with increasing complexity at decreasing plasticity restricting the permissive territories of the progenitor cell populations. Somatic mutations leading to cell proliferation in domains normally reserved for differentiation trigger the inverse cascade of bauplan changes that increase topobiological plasticity at decreased complexity and stepwise enlarge the permissive territory of neoplastic cells consistent with the clinical manifestations of cancer. The order provided by the sequence of attractor transitions and the defined topography of the permissive territories can be exploited for more accurate tumor staging and for locoregional tumor treatment either by surgery or radiotherapy with higher curative potential.
Collapse
Affiliation(s)
- Michael Höckel
- Department of Gynecology, Women's and Children's Center, University of Leipzig, Leipzig, Germany.,Leipzig School of Radical Pelvic Surgery, University of Leipzig, Leipzig, Germany.,Department of Gynecology and Obstetrics, University of Essen, Essen, Germany
| | - Ulrich Behn
- Faculty of Physics and Earth Sciences, Institute of Theoretical Physics, University of Leipzig, Leipzig, Germany
| |
Collapse
|
20
|
Oleari R, Lettieri A, Paganoni A, Zanieri L, Cariboni A. Semaphorin Signaling in GnRH Neurons: From Development to Disease. Neuroendocrinology 2019; 109:193-199. [PMID: 30504719 DOI: 10.1159/000495916] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 12/02/2018] [Indexed: 11/19/2022]
Abstract
In mammals, fertility critically depends on the pulsatile secretion of gonadotropin-releasing hormone (GnRH) by scattered hypothalamic neurons (GnRH neurons). During development, GnRH neurons originate in the nasal placode and migrate first into the nasal compartment and then through the nasal/forebrain junction, before they reach their final position in the hypothalamus. This neurodevelopmental process, which has been extensively studied in mouse models, is regulated by a plethora of factors that might control GnRH neuron migration or survival as well as the fasciculation/targeting of the olfactory/vomeronasal axons along which the GnRH neurons migrate. Defects in GnRH neuron development or release can lead to isolated GnRH deficiency, with the underlying genetic causes still being partially unknown. Recently, semaphorins and their receptors neuropilins and plexins, a large family of molecules implicated in neuronal development and plasticity, are emerging as key regulators of GnRH neuron biology and deficiency. Specifically, semaphorins have been shown to play different roles in GnRH neuron biology by regulating migration and survival during embryonic development as well as secretion in adulthood.
Collapse
Affiliation(s)
- Roberto Oleari
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Antonella Lettieri
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Alyssa Paganoni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Luca Zanieri
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Anna Cariboni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy,
| |
Collapse
|
21
|
Huang S, Li Z, Liu Y, Gao D, Zhang X, Hao J, Yang F. Neural regulation of bone remodeling: Identifying novel neural molecules and pathways between brain and bone. J Cell Physiol 2018; 234:5466-5477. [PMID: 29377116 DOI: 10.1002/jcp.26502] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 01/20/2018] [Indexed: 02/05/2023]
Affiliation(s)
- Shishu Huang
- Department of Orthopaedic Surgery West China Hospital, Sichuan University Chengdu China
| | - Zhenxia Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics West China Hospital of Stomatology, Sichuan University Chengdu China
| | - Yunhui Liu
- The Brain Cognition & Brain Disease Institute, Shenzhen Institutes of Advanced Technology Chinese Academy of Sciences Shenzhen China
| | - Dashuang Gao
- The Brain Cognition & Brain Disease Institute, Shenzhen Institutes of Advanced Technology Chinese Academy of Sciences Shenzhen China
| | - Xinzhou Zhang
- Department of Nephrology Shenzhen People's Hospital, Second Clinical Medical College, Jinan University Shenzhen China
| | - Jin Hao
- Program in Biological Sciences in Dental Medicine, Harvard School of Dental Medicine Boston Massachusetts
| | - Fan Yang
- The Brain Cognition & Brain Disease Institute, Shenzhen Institutes of Advanced Technology Chinese Academy of Sciences Shenzhen China
| |
Collapse
|
22
|
Duke-Cohan JS, Ishikawa Y, Yoshizawa A, Choi YI, Lee CN, Acuto O, Kissler S, Reinherz EL. Regulation of thymocyte trafficking by Tagap, a GAP domain protein linked to human autoimmunity. Sci Signal 2018; 11:11/534/eaan8799. [PMID: 29895617 DOI: 10.1126/scisignal.aan8799] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Multiple autoimmune pathologies are associated with single-nucleotide polymorphisms of the human gene TAGAP, which encodes TAGAP, a guanosine triphosphatase (GTPase)-activating protein. We showed in mice that Tagap-mediated signaling by the sema3E/plexin-D1 ligand-receptor complex attenuates thymocytes' adhesion to the cortex through their β1-containing integrins. By promoting thymocyte detachment within the cortex of the thymus, Tagap-mediated signaling enabled their translocation to the medulla, which is required for continued thymic selection. Tagap physically interacted with the cytoplasmic domain of plexin-D1 and directly stimulated the activity and signaling of the GTPase RhoA. In addition, Tagap indirectly mediated the activation of Cdc42 in response to the binding of sema3E to plexin-D1. Both RhoA and Cdc42 are key mediators of cytoskeletal and integrin dynamics in thymocytes. Knockdown of Tagap in mice suppressed the sema3E- and plexin-D1-mediated release of thymocytes that adhered within the cortex through β1-containing integrins. This suppression led to the impaired translocation of thymocytes from the cortex to the medulla and resulted in the formation of ectopic medullary structures within the thymic cortex. Our results suggest that TAGAP variation modulates the risk of autoimmunity by altering thymocyte migration during thymic selection.
Collapse
Affiliation(s)
- Jonathan S Duke-Cohan
- Laboratory of Immunobiology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA. .,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Yuki Ishikawa
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA.,Immunobiology Section, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Akihiro Yoshizawa
- Laboratory of Immunobiology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Young-Il Choi
- Laboratory of Immunobiology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Chin-Nien Lee
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA.,Immunobiology Section, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Oreste Acuto
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Stephan Kissler
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA. .,Immunobiology Section, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Ellis L Reinherz
- Laboratory of Immunobiology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA. .,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
23
|
Mostafavi S, Gaiteri C, Sullivan SE, White CC, Tasaki S, Xu J, Taga M, Klein HU, Patrick E, Komashko V, McCabe C, Smith R, Bradshaw EM, Root DE, Regev A, Yu L, Chibnik LB, Schneider JA, Young-Pearse TL, Bennett DA, De Jager PL. A molecular network of the aging human brain provides insights into the pathology and cognitive decline of Alzheimer's disease. Nat Neurosci 2018; 21:811-819. [PMID: 29802388 PMCID: PMC6599633 DOI: 10.1038/s41593-018-0154-9] [Citation(s) in RCA: 340] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 04/20/2018] [Indexed: 02/07/2023]
Abstract
There is a need for new therapeutic targets with which to prevent Alzheimer’s disease (AD), a major contributor to aging-related cognitive decline. Here, we report the construction and validation of a molecular network of the aging human frontal cortex. Using RNA sequence data from 478 individuals, we first build a molecular network using modules of coexpressed genes and then relate these modules to AD and its neuropathologic and cognitive endophenotypes. We confirm these associations in two independent AD datasets as well as in epigenomic data. We also illustrate the use of the network in prioritizing amyloid-associated genes for in vitro validation in human neurons and astrocytes. These analyses based on unique cohorts enable us to resolve the role of distinct cortical modules that have a direct effect on the accumulation of AD pathology from those that have a direct effect on cognitive decline, exemplifying a network approach to complex diseases. Systems biology analysis of RNA sequencing data from the aging human cortex identifies a molecular network which prioritizes groups of genes that influence cognitive decline or neuropathology in Alzheimer’s disease.
Collapse
Affiliation(s)
- Sara Mostafavi
- Department of Statistics, Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.,Canadian Institute for Advanced Research, Toronto, ON, Canada
| | - Chris Gaiteri
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Sarah E Sullivan
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Shinya Tasaki
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Jishu Xu
- Broad Institute, Cambridge, MA, USA
| | - Mariko Taga
- Broad Institute, Cambridge, MA, USA.,Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Hans-Ulrich Klein
- Broad Institute, Cambridge, MA, USA.,Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | | | - Vitalina Komashko
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | | | - Robert Smith
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Elizabeth M Bradshaw
- Broad Institute, Cambridge, MA, USA.,Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | | | | | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Lori B Chibnik
- Broad Institute, Cambridge, MA, USA.,Harvard Medical School, Boston, MA, USA.,Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Tracy L Young-Pearse
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA.
| | - Philip L De Jager
- Broad Institute, Cambridge, MA, USA. .,Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
24
|
Abstract
Semaphorins are extracellular signaling proteins that are essential for the development and maintenance of many organs and tissues. The more than 20-member semaphorin protein family includes secreted, transmembrane and cell surface-attached proteins with diverse structures, each characterized by a single cysteine-rich extracellular sema domain, the defining feature of the family. Early studies revealed that semaphorins function as axon guidance molecules, but it is now understood that semaphorins are key regulators of morphology and motility in many different cell types including those that make up the nervous, cardiovascular, immune, endocrine, hepatic, renal, reproductive, respiratory and musculoskeletal systems, as well as in cancer cells. Semaphorin signaling occurs predominantly through Plexin receptors and results in changes to the cytoskeletal and adhesive machinery that regulate cellular morphology. While much remains to be learned about the mechanisms underlying the effects of semaphorins, exciting work has begun to reveal how semaphorin signaling is fine-tuned through different receptor complexes and other mechanisms to achieve specific outcomes in various cellular contexts and physiological systems. These and future studies will lead to a more complete understanding of semaphorin-mediated development and to a greater understanding of how these proteins function in human disease.
Collapse
Affiliation(s)
- Laura Taylor Alto
- Departments of Neuroscience and Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jonathan R Terman
- Departments of Neuroscience and Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
25
|
Angelopoulou E, Piperi C. Emerging role of plexins signaling in glioma progression and therapy. Cancer Lett 2018; 414:81-87. [DOI: 10.1016/j.canlet.2017.11.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/07/2017] [Accepted: 11/08/2017] [Indexed: 12/14/2022]
|
26
|
Wylie T, Garg R, Ridley AJ, Conte MR. Analysis of the interaction of Plexin-B1 and Plexin-B2 with Rnd family proteins. PLoS One 2017; 12:e0185899. [PMID: 29040270 PMCID: PMC5645086 DOI: 10.1371/journal.pone.0185899] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 09/21/2017] [Indexed: 02/03/2023] Open
Abstract
The Rnd family of proteins, Rnd1, Rnd2 and Rnd3, are atypical Rho family GTPases, which bind to but do not hydrolyse GTP. They interact with plexins, which are receptors for semaphorins, and are hypothesised to regulate plexin signalling. We recently showed that each Rnd protein has a distinct profile of interaction with three plexins, Plexin-B1, Plexin-B2 and Plexin-B3, in mammalian cells, although it is unclear which region(s) of these plexins contribute to this specificity. Here we characterise the binary interactions of the Rnd proteins with the Rho-binding domain (RBD) of Plexin-B1 and Plexin-B2 using biophysical approaches. Isothermal titration calorimetry (ITC) experiments for each of the Rnd proteins with Plexin-B1-RBD and Plexin-B2-RBD showed similar association constants for all six interactions, although Rnd1 displayed a small preference for Plexin-B1-RBD and Rnd3 for Plexin-B2-RBD. Furthermore, mutagenic analysis of Rnd3 suggested similarities in its interaction with both Plexin-B1-RBD and Plexin-B2-RBD. These results suggest that Rnd proteins do not have a clear-cut specificity for different Plexin-B-RBDs, possibly implying the contribution of additional regions of Plexin-B proteins in conferring functional substrate selection.
Collapse
Affiliation(s)
- Thomas Wylie
- Randall Division of Cell and Molecular Biophysics, King’s College London, Guy’s Campus, London, United Kingdom
| | - Ritu Garg
- Randall Division of Cell and Molecular Biophysics, King’s College London, Guy’s Campus, London, United Kingdom
| | - Anne J. Ridley
- Randall Division of Cell and Molecular Biophysics, King’s College London, Guy’s Campus, London, United Kingdom
| | - Maria R. Conte
- Randall Division of Cell and Molecular Biophysics, King’s College London, Guy’s Campus, London, United Kingdom
| |
Collapse
|
27
|
Lumb R, Buckberry S, Secker G, Lawrence D, Schwarz Q. Transcriptome profiling reveals expression signatures of cranial neural crest cells arising from different axial levels. BMC DEVELOPMENTAL BIOLOGY 2017; 17:5. [PMID: 28407732 PMCID: PMC5390458 DOI: 10.1186/s12861-017-0147-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 04/03/2017] [Indexed: 01/13/2023]
Abstract
Background Cranial neural crest cells (NCCs) are a unique embryonic cell type which give rise to a diverse array of derivatives extending from neurons and glia through to bone and cartilage. Depending on their point of origin along the antero-posterior axis cranial NCCs are rapidly sorted into distinct migratory streams that give rise to axial specific structures. These migratory streams mirror the underlying segmentation of the brain with NCCs exiting the diencephalon and midbrain following distinct paths compared to those exiting the hindbrain rhombomeres (r). The genetic landscape of cranial NCCs arising at different axial levels remains unknown. Results Here we have used RNA sequencing to uncover the transcriptional profiles of mouse cranial NCCs arising at different axial levels. Whole transcriptome analysis identified over 120 transcripts differentially expressed between NCCs arising anterior to r3 (referred to as r1-r2 migratory stream for simplicity) and the r4 migratory stream. Eight of the genes differentially expressed between these populations were validated by RT-PCR with 2 being further validated by in situ hybridisation. We also explored the expression of the Neuropilins (Nrp1 and Nrp2) and their co-receptors and show that the A-type Plexins are differentially expressed in different cranial NCC streams. Conclusions Our analyses identify a large number of genes differentially regulated between cranial NCCs arising at different axial levels. This data provides a comprehensive description of the genetic landscape driving diversity of distinct cranial NCC streams and provides novel insight into the regulatory networks controlling the formation of specific skeletal elements and the mechanisms promoting migration along different paths. Electronic supplementary material The online version of this article (doi:10.1186/s12861-017-0147-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rachael Lumb
- Centre for Cancer Biology, University of South Australia and SA Pathology, Frome Road, Adelaide, SA, 5000, Australia.,University of Adelaide, Frome Road, Adelaide, SA, 5000, Australia
| | - Sam Buckberry
- Harry Perkins Institute of Medical Research, Perth, WA, 6008, Australia.,Australian Research Council Centre of Excellence in Plant Energy Biology, University of Western Australia, Perth, 6009, WA, Australia
| | - Genevieve Secker
- Centre for Cancer Biology, University of South Australia and SA Pathology, Frome Road, Adelaide, SA, 5000, Australia
| | - David Lawrence
- ACRF Cancer Genomics Facility, Centre for Cancer Biology, SA Pathology, Adelaide, Australia.,School of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
| | - Quenten Schwarz
- Centre for Cancer Biology, University of South Australia and SA Pathology, Frome Road, Adelaide, SA, 5000, Australia.
| |
Collapse
|
28
|
McColl B, Garg R, Riou P, Riento K, Ridley AJ. Rnd3-induced cell rounding requires interaction with Plexin-B2. J Cell Sci 2016; 129:4046-4056. [PMID: 27656111 PMCID: PMC5117210 DOI: 10.1242/jcs.192211] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/02/2016] [Indexed: 12/24/2022] Open
Abstract
Rnd proteins are atypical members of the Rho GTPase family that induce actin cytoskeletal reorganization and cell rounding. Rnd proteins have been reported to bind to the intracellular domain of several plexin receptors, but whether plexins contribute to the Rnd-induced rounding response is not known. Here we show that Rnd3 interacts preferentially with plexin-B2 of the three plexin-B proteins, whereas Rnd2 interacts with all three B-type plexins, and Rnd1 shows only very weak interaction with plexin-B proteins in immunoprecipitations. Plexin-B1 has been reported to act as a GAP for R-Ras and/or Rap1 proteins. We show that all three plexin-B proteins interact with R-Ras and Rap1, but Rnd proteins do not alter this interaction or R-Ras or Rap1 activity. We demonstrate that plexin-B2 promotes Rnd3-induced cell rounding and loss of stress fibres, and enhances the inhibition of HeLa cell invasion by Rnd3. We identify the amino acids in Rnd3 that are required for plexin-B2 interaction, and show that mutation of these amino acids prevents Rnd3-induced morphological changes. These results indicate that plexin-B2 is a downstream target for Rnd3, which contributes to its cellular function.
Collapse
Affiliation(s)
- Brad McColl
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Ritu Garg
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Philippe Riou
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Kirsi Riento
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Anne J Ridley
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| |
Collapse
|
29
|
Xia J, Worzfeld T. Semaphorins and Plexins in Kidney Disease. Nephron Clin Pract 2016; 132:93-100. [PMID: 26844545 DOI: 10.1159/000443645] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 12/17/2015] [Indexed: 11/19/2022] Open
Abstract
Semaphorins are soluble or membrane-bound cues, which control multiple aspects of cell-cell communication, differentiation, morphology and function. Most of their effects are mediated by a family of transmembrane receptors called plexins. Semaphorins and plexins have emerged as central regulators of diverse physiological and pathophysiological processes in various organs. This review summarizes the role of semaphorins and plexins in renal pathophysiology and their potential use as biomarkers of kidney disease.
Collapse
Affiliation(s)
- Jingjing Xia
- Department of Dermatology, Center for Molecular Medicine Cologne, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | | |
Collapse
|
30
|
Lahm H, Schön P, Doppler S, Dreßen M, Cleuziou J, Deutsch MA, Ewert P, Lange R, Krane M. Tetralogy of Fallot and Hypoplastic Left Heart Syndrome - Complex Clinical Phenotypes Meet Complex Genetic Networks. Curr Genomics 2015; 16:141-58. [PMID: 26069455 PMCID: PMC4460219 DOI: 10.2174/1389202916666150303232520] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 02/28/2015] [Accepted: 03/03/2015] [Indexed: 01/06/2023] Open
Abstract
In many cases congenital heart disease (CHD) is represented by a complex phenotype and
an array of several functional and morphological cardiac disorders. These malformations will be
briefly summarized in the first part focusing on two severe CHD phenotypes, hypoplastic left heart
syndrome (HLHS) and tetralogy of Fallot (TOF). In most cases of CHD the genetic origin remains
largely unknown, though the complexity of the clinical picture strongly argues against a dysregulation which can be attributed
to a single candidate gene but rather suggests a multifaceted polygenetic origin with elaborate interactions. Consistent
with this idea, genome-wide approaches using whole exome sequencing, comparative sequence analysis of multiplex
families to identify de novo mutations and global technologies to identify single nucleotide polymorphisms, copy
number variants, dysregulation of the transcriptome and epigenetic variations have been conducted to obtain information
about genetic alterations and potential predispositions possibly linked to the occurrence of a CHD phenotype. In the second
part of this review we will summarize and discuss the available literature on identified genetic alterations linked to
TOF and HLHS.
Collapse
Affiliation(s)
- Harald Lahm
- Department of Cardiovascular Surgery, Division of Experimental Surgery, German Heart Center Munich, Technische Universität München, Munich Heart Alliance, D-80636 Munich, Germany
| | - Patric Schön
- Department of Paediatric Cardiology and Congenital Heart Defects, German Heart Center Munich, Technische Universität München, D-80636 Munich, Germany
| | - Stefanie Doppler
- Department of Cardiovascular Surgery, Division of Experimental Surgery, German Heart Center Munich, Technische Universität München, Munich Heart Alliance, D-80636 Munich, Germany
| | - Martina Dreßen
- Department of Cardiovascular Surgery, Division of Experimental Surgery, German Heart Center Munich, Technische Universität München, Munich Heart Alliance, D-80636 Munich, Germany
| | - Julie Cleuziou
- Department of Cardiovascular Surgery, Division of Experimental Surgery, German Heart Center Munich, Technische Universität München, Munich Heart Alliance, D-80636 Munich, Germany
| | - Marcus-André Deutsch
- Department of Cardiovascular Surgery, Division of Experimental Surgery, German Heart Center Munich, Technische Universität München, Munich Heart Alliance, D-80636 Munich, Germany
| | - Peter Ewert
- Department of Paediatric Cardiology and Congenital Heart Defects, German Heart Center Munich, Technische Universität München, D-80636 Munich, Germany; ; DZHK (German Center for Cardiovascular Research) - partner site Munich Heart Alliance, Munich, Germany
| | - Rüdiger Lange
- Department of Cardiovascular Surgery, Division of Experimental Surgery, German Heart Center Munich, Technische Universität München, Munich Heart Alliance, D-80636 Munich, Germany; ; DZHK (German Center for Cardiovascular Research) - partner site Munich Heart Alliance, Munich, Germany
| | - Markus Krane
- Department of Cardiovascular Surgery, Division of Experimental Surgery, German Heart Center Munich, Technische Universität München, Munich Heart Alliance, D-80636 Munich, Germany; ; DZHK (German Center for Cardiovascular Research) - partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
31
|
Malik MFA, Ye L, Jiang WG. Reduced expression of semaphorin 4D and plexin-B in breast cancer is associated with poorer prognosis and the potential linkage with oestrogen receptor. Oncol Rep 2015; 34:1049-57. [PMID: 26035216 DOI: 10.3892/or.2015.4015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 03/30/2015] [Indexed: 11/06/2022] Open
Abstract
Involvement of semaphorin 4D (Sema4D) and the receptor proteins of the plexins B family (plexin-B1, -B2 and -B3) in solid tumours suggests they play a role in breast cancer. In the present study, the expression of Sema4D and plexin-Bs was examined in a breast cancer cohort. The expression of Sema4D and plexin-Bs was examined in 147 tumours together with 22 normal mammary tissues using quantitative PCR along with clinicopathological patient data, as well as in MCF-7 and MDA-MB-231 cell lines treated with selective oestrogen receptor modulators (SERMs). The expression of Sema4D, plexin-B1 and -B2 was markedly reduced in tumours with local recurrence, compared to the patients that remained disease-free. The reduced Sema4D expression was associated with poorer disease-free survival (median, 111.6 months, 95% CI, 96.5-126.7), compared to the patients with a higher expression (median, 144.0 months; 95% CI, 130.8-157.3; p=0.033). A reduced expression of plexin-B1 was observed in tumours with poorer differentiation and was associated with poorer overall and disease-free survival. No similar association was identified in relation to plexin-B2 and -B3. A higher expression of Sema4D and plexin-B1 was observed in the ERα-positive tumours compared to the ERα-negative tumours. The expression of these molecules was largely regulated in breast cancer cells exposed to SERMs. A decreased expression of Sema4D, plexin-B1 and -B2 was associated with local recurrence and poor prognosis. Response to SERMs indicated potential perspectives of these molecules in clinical assessment and management of diseases.
Collapse
Affiliation(s)
- Muhammad Faraz Arshad Malik
- Metastasis and Angiogenesis Research Group, Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff CF14 4XN, UK
| | - Lin Ye
- Metastasis and Angiogenesis Research Group, Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff CF14 4XN, UK
| | - Wen G Jiang
- Metastasis and Angiogenesis Research Group, Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff CF14 4XN, UK
| |
Collapse
|
32
|
Giacobini P. Shaping the Reproductive System: Role of Semaphorins in Gonadotropin-Releasing Hormone Development and Function. Neuroendocrinology 2015; 102:200-15. [PMID: 25967979 DOI: 10.1159/000431021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 04/28/2015] [Indexed: 11/19/2022]
Abstract
The semaphorin proteins, which contribute to the morphogenesis and homeostasis of a wide range of systems, are among the best-studied families of guidance cues. Much recent research has focused on the role of semaphorins in the development and adult activity of hormone systems and, reciprocally, how circulating reproductive hormones regulate their expression and function. Specifically, several reports have focused on the molecular mechanisms underlying the effects of semaphorins on the migration, survival and structural and functional plasticity of neurons that secrete gonadotropin-releasing hormone (GnRH), essential for the acquisition and maintenance of reproductive competence in mammals. Alterations in the development of this neuroendocrine system lead to anomalous or absent GnRH secretion, resulting in heterogeneous reproductive disorders such as congenital hypogonadotropic hypogonadism (CHH) or other conditions characterized by infertility or subfertility. This review summarizes current knowledge of the role of semaphorins and their receptors on the development, differentiation and plasticity of the GnRH system. In addition, the involvement of genetic deficits in semaphorin signaling in some forms of CHH in humans is discussed.
Collapse
Affiliation(s)
- Paolo Giacobini
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, U1172, School of Medicine, University of Lille, and Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
| |
Collapse
|
33
|
Hadsell DL, Hadsell LA, Olea W, Rijnkels M, Creighton CJ, Smyth I, Short KM, Cox LL, Cox TC. In-silico QTL mapping of postpubertal mammary ductal development in the mouse uncovers potential human breast cancer risk loci. Mamm Genome 2015; 26:57-79. [PMID: 25552398 DOI: 10.1007/s00335-014-9551-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 12/03/2014] [Indexed: 01/02/2023]
Abstract
Genetic background plays a dominant role in mammary gland development and breast cancer (BrCa). Despite this, the role of genetics is only partially understood. This study used strain-dependent variation in an inbred mouse mapping panel, to identify quantitative trait loci (QTL) underlying structural variation in mammary ductal development, and determined if these QTL correlated with genomic intervals conferring BrCa susceptibility in humans. For about half of the traits, developmental variation among the complete set of strains in this study was greater (P < 0.05) than that of previously studied strains, or strains in current common use for mammary gland biology. Correlations were also detected with previously reported variation in mammary tumor latency and metastasis. In-silico genome-wide association identified 20 mammary development QTL (Mdq). Of these, five were syntenic with previously reported human BrCa loci. The most significant (P = 1 × 10(-11)) association of the study was on MMU6 and contained the genes Plxna4, Plxna4os1, and Chchd3. On MMU5, a QTL was detected (P = 8 × 10(-7)) that was syntenic to a human BrCa locus on h12q24.5 containing the genes Tbx3 and Tbx5. Intersection of linked SNP (r(2) > 0.8) with genomic and epigenomic features, and intersection of candidate genes with gene expression and survival data from human BrCa highlighted several for further study. These results support the conclusion that mammary tumorigenesis and normal ductal development are influenced by common genetic factors and that further studies of genetically diverse mice can improve our understanding of BrCa in humans.
Collapse
Affiliation(s)
- Darryl L Hadsell
- Department of Pediatrics, USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, 1100 Bates St. Suite 10072, Mail Stop: BCM-320, Houston, TX, 77030-2600, USA,
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
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
|
35
|
Abstract
Semaphorins are secreted and membrane-associated proteins that regulate many different developmental processes, including neural circuit assembly, bone formation and angiogenesis. Trans and cis interactions between semaphorins and their multimeric receptors trigger intracellular signal transduction networks that regulate cytoskeletal dynamics and influence cell shape, differentiation, motility and survival. Here and in the accompanying poster we provide an overview of the molecular biology of semaphorin signalling within the context of specific cell and developmental processes, highlighting the mechanisms that act to fine-tune, diversify and spatiotemporally control the effects of semaphorins.
Collapse
Affiliation(s)
- Bart C. Jongbloets
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, UMC Utrecht, 3451 PM Utrecht, The Netherlands
| | - R. Jeroen Pasterkamp
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, UMC Utrecht, 3451 PM Utrecht, The Netherlands
| |
Collapse
|
36
|
Migliozzi MT, Mucka P, Bielenberg DR. Lymphangiogenesis and metastasis--a closer look at the neuropilin/semaphorin3 axis. Microvasc Res 2014; 96:68-76. [PMID: 25087623 DOI: 10.1016/j.mvr.2014.07.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 07/10/2014] [Accepted: 07/11/2014] [Indexed: 01/13/2023]
Abstract
Metastasis is the leading cause of cancer-related deaths. Understanding how the lymphatic system responds to its environment and local stimuli may lead to therapies to combat metastasis and other lymphatic-associated diseases. This review compares lymphatic vessels and blood vessels, discusses markers of lymphatic vasculature, and elucidates some of the signaling motifs involved in lymphangiogenesis. Recent progress implicating the neuropilin and semaphorin axes in this process is discussed.
Collapse
Affiliation(s)
- Matthew T Migliozzi
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Patrick Mucka
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Diane R Bielenberg
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
37
|
|
38
|
Laht P, Otsus M, Remm J, Veske A. B-plexins control microtubule dynamics and dendrite morphology of hippocampal neurons. Exp Cell Res 2014; 326:174-84. [PMID: 24954409 DOI: 10.1016/j.yexcr.2014.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 06/09/2014] [Indexed: 11/26/2022]
Abstract
Semaphorins and their receptors plexins are implicated in various processes in the nervous system, but how B-plexins regulate the growth of dendrites remains poorly characterized. We had previously observed that Plexin-B1 and B3 interact with microtubule end-binding proteins (EBs) that are central adapters at growing microtubule tips, and this interaction is involved in neurite growth. Therefore, we hypothesized that plexins regulate microtubule dynamics and through that also dendritogenesis. The role of all three B-plexins was systematically examined in these processes. B-plexins and their ligand Semaphorin-4D influence the dynamics of microtubule tips both EB-dependently and independendently. EB3 as well as Plexin-B1, B2 and B3 turned out to have a significant role in the development of dendritic arbor of rat hippocampal neurons. Our results clearly indicate that semaphorin-plexin-EB pathway is one molecular mechanism how extracellular guidance cues are translated into intracellular mechanics. Taken together, Semaphorin-4D and B-plexins modulate the dynamic behavior of microtubule tips, and are therefore important in neurite growth.
Collapse
Affiliation(s)
- Piret Laht
- Department of Gene Technology, Tallinn University of Technology, Akadeemia tee 15, Tallinn 12618, Estonia; Competence Centre for Cancer Research, Tallinn, Estonia
| | - Maarja Otsus
- Competence Centre for Cancer Research, Tallinn, Estonia
| | - Jaanus Remm
- Institute of Ecology and Earth Sciences, University of Tartu, Tartu, Estonia
| | - Andres Veske
- Department of Gene Technology, Tallinn University of Technology, Akadeemia tee 15, Tallinn 12618, Estonia; Competence Centre for Cancer Research, Tallinn, Estonia.
| |
Collapse
|
39
|
Tufro A. Semaphorin3a signaling, podocyte shape, and glomerular disease. Pediatr Nephrol 2014; 29:751-5. [PMID: 24464477 PMCID: PMC3992269 DOI: 10.1007/s00467-013-2743-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 12/17/2013] [Accepted: 12/18/2013] [Indexed: 12/21/2022]
Abstract
Semaphorin3a (sema3a), a member of class 3 semaphorins, is a guidance protein that regulates angiogenesis, branching morphogenesis, axon growth, and cell migration, and has pleiotropic roles on organogenesis, immune response, and cancer. Sema3a is secreted by podocytes and is required for normal kidney patterning and glomerular filtration barrier development. We recently discovered that after completion of kidney development, Sema3a gain-of-function in podocytes leads to proteinuric glomerular disease in mice. Excess sema3a causes foot process effacement, glomerular basement lamination, and endothelial damage in vivo, and disrupts cell autonomously podocyte shape by down-regulating nephrin and inhibiting αvβ3 integrin. We identified a novel direct interaction between nephrin and plexinA1, the sema3a signaling receptor. Nephrin-plexinA1 interaction links the slit-diaphragm signaling complex to extracellular sema3a signals. Hence, sema3a functions as an extracellular negative regulator of the structure and function of the glomerular filtration barrier.
Collapse
Affiliation(s)
- Alda Tufro
- Department of Pediatrics, Yale University School of Medicine, 333 Cedar Street, PO Box 208064, New Haven, CT, 06520-8064, USA,
| |
Collapse
|
40
|
Abstract
Mammalian plexins constitute a family of transmembrane receptors for semaphorins and represent critical regulators of various processes during development of the nervous, cardiovascular, skeletal, and renal system. In vitro studies have shown that plexins exert their effects via an intracellular R-Ras/M-Ras GTPase-activating protein (GAP) domain or by activation of RhoA through interaction with Rho guanine nucleotide exchange factor proteins. However, which of these signaling pathways are relevant for plexin functions in vivo is largely unknown. Using an allelic series of transgenic mice, we show that the GAP domain of plexins constitutes their key signaling module during development. Mice in which endogenous Plexin-B2 or Plexin-D1 is replaced by transgenic versions harboring mutations in the GAP domain recapitulate the phenotypes of the respective null mutants in the developing nervous, vascular, and skeletal system. We further provide genetic evidence that, unexpectedly, the GAP domain-mediated developmental functions of plexins are not brought about via R-Ras and M-Ras inactivation. In contrast to the GAP domain mutants, Plexin-B2 transgenic mice defective in Rho guanine nucleotide exchange factor binding are viable and fertile but exhibit abnormal development of the liver vasculature. Our genetic analyses uncover the in vivo context-dependence and functional specificity of individual plexin-mediated signaling pathways during development.
Collapse
|
41
|
Leikauf GD, Concel VJ, Bein K, Liu P, Berndt A, Martin TM, Ganguly K, Jang AS, Brant KA, Dopico RA, Upadhyay S, Cario C, Di YPP, Vuga LJ, Kostem E, Eskin E, You M, Kaminski N, Prows DR, Knoell DL, Fabisiak JP. Functional genomic assessment of phosgene-induced acute lung injury in mice. Am J Respir Cell Mol Biol 2013; 49:368-83. [PMID: 23590305 DOI: 10.1165/rcmb.2012-0337oc] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
In this study, a genetically diverse panel of 43 mouse strains was exposed to phosgene and genome-wide association mapping performed using a high-density single nucleotide polymorphism (SNP) assembly. Transcriptomic analysis was also used to improve the genetic resolution in the identification of genetic determinants of phosgene-induced acute lung injury (ALI). We prioritized the identified genes based on whether the encoded protein was previously associated with lung injury or contained a nonsynonymous SNP within a functional domain. Candidates were selected that contained a promoter SNP that could alter a putative transcription factor binding site and had variable expression by transcriptomic analyses. The latter two criteria also required that ≥10% of mice carried the minor allele and that this allele could account for ≥10% of the phenotypic difference noted between the strains at the phenotypic extremes. This integrative, functional approach revealed 14 candidate genes that included Atp1a1, Alox5, Galnt11, Hrh1, Mbd4, Phactr2, Plxnd1, Ptprt, Reln, and Zfand4, which had significant SNP associations, and Itga9, Man1a2, Mapk14, and Vwf, which had suggestive SNP associations. Of the genes with significant SNP associations, Atp1a1, Alox5, Plxnd1, Ptprt, and Zfand4 could be associated with ALI in several ways. Using a competitive electrophoretic mobility shift analysis, Atp1a1 promoter (rs215053185) oligonucleotide containing the minor G allele formed a major distinct faster-migrating complex. In addition, a gene with a suggestive SNP association, Itga9, is linked to transforming growth factor β1 signaling, which previously has been associated with the susceptibility to ALI in mice.
Collapse
Affiliation(s)
- George D Leikauf
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, PA 15219, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Mining the O-mannose glycoproteome reveals cadherins as major O-mannosylated glycoproteins. Proc Natl Acad Sci U S A 2013; 110:21018-23. [PMID: 24101494 DOI: 10.1073/pnas.1313446110] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The metazoan O-mannose (O-Man) glycoproteome is largely unknown. It has been shown that up to 30% of brain O-glycans are of the O-Man type, but essentially only alpha-dystroglycan (α-DG) of the dystrophin-glycoprotein complex is well characterized as an O-Man glycoprotein. Defects in O-Man glycosylation underlie congenital muscular dystrophies and considerable efforts have been devoted to explore this O-glycoproteome without much success. Here, we used our SimpleCell strategy using nuclease-mediated gene editing of a human cell line (MDA-MB-231) to reduce the structural heterogeneity of O-Man glycans and to probe the O-Man glycoproteome. In this breast cancer cell line we found that O-Man glycosylation is primarily found on cadherins and plexins on β-strands in extracellular cadherin and Ig-like, plexin and transcription factor domains. The positions and evolutionary conservation of O-Man glycans in cadherins suggest that they play important functional roles for this large group of cell adhesion glycoproteins, which can now be addressed. The developed O-Man SimpleCell strategy is applicable to most types of cell lines and enables proteome-wide discovery of O-Man protein glycosylation.
Collapse
|
43
|
Shimizu I, Yoshida Y, Moriya J, Nojima A, Uemura A, Kobayashi Y, Minamino T. Semaphorin3E-induced inflammation contributes to insulin resistance in dietary obesity. Cell Metab 2013; 18:491-504. [PMID: 24093674 DOI: 10.1016/j.cmet.2013.09.001] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 07/10/2013] [Accepted: 08/13/2013] [Indexed: 01/02/2023]
Abstract
Semaphorins and their receptors (plexins) are axon-guiding molecules that regulate the development of the nervous system during embryogenesis. Here we describe a previously unknown role of class 3 semaphorin E (Sema3E) in adipose tissue inflammation and insulin resistance. Expression of Sema3E and its receptor plexinD1 was upregulated in the adipose tissue of a mouse model of dietary obesity. Inhibition of the Sema3E-plexinD1 axis markedly reduced adipose tissue inflammation and improved insulin resistance in this model. Conversely, overexpression of Sema3E in adipose tissue provoked inflammation and insulin resistance. Sema3E promoted infiltration of macrophages, and this effect was inhibited by disrupting plexinD1 expression in macrophages. Disruption of adipose tissue p53 expression led to downregulation of Sema3E expression and improved adipose tissue inflammation. These results indicate that Sema3E acts as a chemoattractant for macrophages, with p53-induced upregulation of Sema3E expression provoking adipose tissue inflammation and systemic insulin resistance in association with dietary obesity.
Collapse
Affiliation(s)
- Ippei Shimizu
- Department of Cardiovascular Science and Medicine, 1-8-1 Inohana, Chuo-ku, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | | | | | | | | | | | | |
Collapse
|
44
|
Messina A, Giacobini P. Semaphorin signaling in the development and function of the gonadotropin hormone-releasing hormone system. Front Endocrinol (Lausanne) 2013; 4:133. [PMID: 24065959 PMCID: PMC3779810 DOI: 10.3389/fendo.2013.00133] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Accepted: 09/09/2013] [Indexed: 12/17/2022] Open
Abstract
The semaphorin proteins are among the best-studied families of guidance cues, contributing to morphogenesis and homeostasis in a wide range of tissue types. The major semaphorin receptors are plexins and neuropilins, however other receptors and co-receptors are capable to mediate signaling by semaphorins. These guidance proteins were originally identified as growth cone "collapsing factors" or as inhibitory signals, crucial for nervous system development. Since those seminal discoveries, the list of functions of semaphorins has rapidly grown. Over the past few years, a growing body of data indicates that semaphorins are involved in the regulation of the immune and vascular systems, in tumor growth/cancer cell metastasis and in neural circuit formation. Recently there has been increasing emphasis on research to determine the potential influence of semaphorins on the development and homeostasis of hormone systems and how circulating reproductive hormones regulate their expression and functions. Here, we focus on the emerging role of semaphorins in the development, differentiation and plasticity of unique neurons that secrete gonadotropin-releasing hormone (GnRH), which are essential for the acquisition and maintenance of reproductive competence in all vertebrates. Genetic evidence is also provided showing that insufficient semaphorin signaling contributes to some forms of reproductive disorders in humans, characterized by the reduction or failure of sexual competence. Finally, we will review some studies with the goal of highlighting how the expression of semaphorins and their receptors might be regulated by gonadal hormones in physiological and pathological conditions.
Collapse
Affiliation(s)
- Andrea Messina
- INSERM, Laboratory of Development and Plasticity of the Postnatal Brain, Jean-Pierre Aubert Research Center, Unité 837, Lille, France
- School of Medicine, UDSL, Lille, France
| | - Paolo Giacobini
- INSERM, Laboratory of Development and Plasticity of the Postnatal Brain, Jean-Pierre Aubert Research Center, Unité 837, Lille, France
- School of Medicine, UDSL, Lille, France
- *Correspondence: Paolo Giacobini, INSERM, Laboratory of Development and Plasticity of the Postnatal Brain, Jean-Pierre Aubert Research Center, Unit 837, Place de Verdun, 59045 Lille Cedex, France e-mail:
| |
Collapse
|
45
|
Wanschel A, Seibert T, Hewing B, Ramkhelawon B, Ray TD, van Gils JM, Rayner KJ, Feig JE, O'Brien ER, Fisher EA, Moore KJ. Neuroimmune guidance cue Semaphorin 3E is expressed in atherosclerotic plaques and regulates macrophage retention. Arterioscler Thromb Vasc Biol 2013; 33:886-93. [PMID: 23430613 DOI: 10.1161/atvbaha.112.300941] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The persistence of myeloid-derived cells in the artery wall is a characteristic of advanced atherosclerotic plaques. However, the mechanisms by which these cells are retained are poorly understood. Semaphorins, a class of neuronal guidance molecules, play a critical role in vascular patterning and development, and recent studies suggest that they may also have immunomodulatory functions. The present study evaluates the expression of Semaphorin 3E (Sema3E) in settings relevant to atherosclerosis and its contribution to macrophage accumulation in plaques. APPROACH AND RESULTS Immunofluorescence staining of Sema3E, and its receptor PlexinD1, demonstrated their expression in macrophages of advanced atherosclerotic lesions of Apoe(-/-) mice. Notably, in 2 different mouse models of atherosclerosis regression, Sema3E mRNA was highly downregulated in plaque macrophages, coincident with a reduction in plaque macrophage content and an enrichment in markers of reparative M2 macrophages. In vitro, Sema3E mRNA was highly expressed in inflammatory M1 macrophages and in macrophages treated with physiological drivers of plaque progression and inflammation, such as oxidized low-density lipoprotein and hypoxia. To explore mechanistically how Sema3E affects macrophage behavior, we treated macrophages with recombinant protein in the presence/absence of chemokines, including CCL19, a chemokine implicated in the egress of macrophages from atherosclerotic plaques. Sema3E blocked actin polymerization and macrophage migration stimulated by the chemokines, suggesting that it may immobilize these cells in the plaque. CONCLUSIONS Sema3E is upregulated in macrophages of advanced plaques, is dynamically regulated by multiple atherosclerosis-relevant factors, and acts as a negative regulator of macrophage migration, which may promote macrophage retention and chronic inflammation in vivo.
Collapse
Affiliation(s)
- Amarylis Wanschel
- Marc and Ruti Bell Vascular Biology and Disease Program, Department of Medicine, Leon H. Charney Division of Cardiology, New YorkUniversity School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Roney K, Holl E, Ting J. Immune plexins and semaphorins: old proteins, new immune functions. Protein Cell 2013; 4:17-26. [PMID: 23307780 DOI: 10.1007/s13238-012-2108-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 10/25/2012] [Indexed: 12/24/2022] Open
Abstract
Plexins and semaphorins are a large family of proteins that are involved in cell movement and response. The importance of plexins and semaphorins has been emphasized by their discovery in many organ systems including the nervous (Nkyimbeng-Takwi and Chapoval, 2011; McCormick and Leipzig, 2012; Yaron and Sprinzak, 2012), epithelial (Miao et al., 1999; Fujii et al., 2002), and immune systems (Takamatsu and Kumanogoh, 2012) as well as diverse cell processes including angiogenesis (Serini et al., 2009; Sakurai et al., 2012), embryogenesis (Perala et al., 2012), and cancer (Potiron et al., 2009; Micucci et al., 2010). Plexins and semaphorins are transmembrane proteins that share a conserved extracellular semaphorin domain (Hota and Buck, 2012). The plexins and semaphorins are divided into four and eight subfamilies respectively based on their structural homology. Semaphorins are relatively small proteins containing the extracellular semaphorin domain and short intracellular tails. Plexins contain the semaphorin domain and long intracellular tails (Hota and Buck, 2012). The majority of plexin and semaphorin research has focused on the nervous system, particularly the developing nervous system, where these proteins are found to mediate many common neuronal cell processes including cell movement, cytoskeletal rearrangement, and signal transduction (Choi et al., 2008; Takamatsu et al., 2010). Their roles in the immune system are the focus of this review.
Collapse
Affiliation(s)
- Kelly Roney
- Department of Microbiology and Immunology, 22-004 Lineberger Comprehensive Cancer Center, University of Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | |
Collapse
|
47
|
Giacobini P, Prevot V. Semaphorins in the development, homeostasis and disease of hormone systems. Semin Cell Dev Biol 2012; 24:190-8. [PMID: 23219659 DOI: 10.1016/j.semcdb.2012.11.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 11/02/2012] [Accepted: 11/28/2012] [Indexed: 11/16/2022]
Abstract
Semaphorin proteins are among the best-studied families of guidance cues. Initially characterized as repulsive neuronal guidance cues, during the last decade, significant progress has been made in defining their involvement in the regulation of dynamic changes in the cellular cytoskeleton during embryonic and postnatal neuronal development, under both physiological and pathological conditions. However, semaphorins are not restricted to the nervous system but widely expressed in other tissues, where they play key roles in angiogenesis and organogenesis. In recent years, there has been an increasing emphasis on the potential influence of semaphorins on the development and homeostasis of hormone systems, and conversely, how circulating reproductive hormones regulate semaphorin expression. In this review, we summarize recent studies analyzing the contribution of semaphorin signaling to the morphogenesis, differentiation and plasticity of fundamental neuroendocrine and endocrine systems that regulate key physiological processes, such as reproduction, bone formation and the control of energy homeostasis.
Collapse
Affiliation(s)
- Paolo Giacobini
- Inserm, Jean-Pierre Aubert Research Center, Development and Plasticity of the Postnatal Brain, Unit 837, France.
| | | |
Collapse
|
48
|
Chen Y, Soong J, Mohanty S, Xu L, Scott G. The neural guidance receptor Plexin C1 delays melanoma progression. Oncogene 2012; 32:4941-9. [PMID: 23160370 DOI: 10.1038/onc.2012.511] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 09/18/2012] [Accepted: 09/21/2012] [Indexed: 01/13/2023]
Abstract
Plexin C1 is a type I transmembrane receptor with intrinsic R-Ras GTPase activity, which regulates cytoskeletal remodeling and adhesion in normal human melanocytes. Melanocytes are pigment-producing cells of the epidermis, precursors for melanoma, and express high levels of Plexin C1, which is lost in melanoma in vitro and in vivo. To determine if Plexin C1 is a tumor suppressor for melanoma, we introduced Plexin C1 into a primary human melanoma cell line, and phenotypes including migration, apoptosis, proliferation and tumor growth in mice were analyzed. Complimentary studies in which Plexin C1 was silenced in human melanocytes were performed. Plexin C1 significantly inhibited migration and proliferation in melanoma, whereas in melanocytes, loss of Plexin C1 increased migration and proliferation. In mouse xenografts, Plexin C1 delayed tumor growth of melanoma at early time points, but tumors eventually escaped the suppressive effects of Plexin C1, due to Plexin C1-dependent activation of the pro-survival protein Akt. R-Ras activation stimulates melanoma migration. Plexin C1 lowered R-Ras activity in melanoma and melanocytes, consistent with inhibitory effects of Plexin C1 on migration of melanocytes and melanoma. To determine if R-Ras is expressed in melanocytic lesions in vivo, staining of tissue microarrays of nevi and melanoma were performed. R-Ras expression was highly limited in melanocytic lesions, being essentially confined to primary melanoma, and almost completely absent in nevi and metastatic melanoma. These data suggest that loss of Plexin C1 in melanoma may promote early steps in melanoma progression through suppression of migration and proliferation, but pro-survival effects of Plexin C1 ultimately abrogate the tumor suppressive effects of Plexin C1. In primary melanoma, loss of Plexin C1 may function in early steps of melanoma progression by releasing inhibition of R-Ras activation, and stimulating migration.
Collapse
Affiliation(s)
- Y Chen
- Department of Dermatology, University of Rochester School of Medicine, Rochester, NY, USA
| | | | | | | | | |
Collapse
|
49
|
Hota PK, Buck M. Plexin structures are coming: opportunities for multilevel investigations of semaphorin guidance receptors, their cell signaling mechanisms, and functions. Cell Mol Life Sci 2012; 69:3765-805. [PMID: 22744749 PMCID: PMC11115013 DOI: 10.1007/s00018-012-1019-0] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 04/09/2012] [Accepted: 04/11/2012] [Indexed: 01/13/2023]
Abstract
Plexin transmembrane receptors and their semaphorin ligands, as well as their co-receptors (Neuropilin, Integrin, VEGFR2, ErbB2, and Met kinase) are emerging as key regulatory proteins in a wide variety of developmental, regenerative, but also pathological processes. The diverse arenas of plexin function are surveyed, including roles in the nervous, cardiovascular, bone and skeletal, and immune systems. Such different settings require considerable specificity among the plexin and semaphorin family members which in turn are accompanied by a variety of cell signaling networks. Underlying the latter are the mechanistic details of the interactions and catalytic events at the molecular level. Very recently, dramatic progress has been made in solving the structures of plexins and of their complexes with associated proteins. This molecular level information is now suggesting detailed mechanisms for the function of both the extracellular as well as the intracellular plexin regions. Specifically, several groups have solved structures for extracellular domains for plexin-A2, -B1, and -C1, many in complex with semaphorin ligands. On the intracellular side, the role of small Rho GTPases has been of particular interest. These directly associate with plexin and stimulate a GTPase activating (GAP) function in the plexin catalytic domain to downregulate Ras GTPases. Structures for the Rho GTPase binding domains have been presented for several plexins, some with Rnd1 bound. The entire intracellular domain structure of plexin-A1, -A3, and -B1 have also been solved alone and in complex with Rac1. However, key aspects of the interplay between GTPases and plexins remain far from clear. The structural information is helping the plexin field to focus on key questions at the protein structural, cellular, as well as organism level that collaboratoria of investigations are likely to answer.
Collapse
Affiliation(s)
- Prasanta K. Hota
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106 USA
| | - Matthias Buck
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106 USA
- Department of Neuroscience, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106 USA
- Department of Pharmacology, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106 USA
- Comprehensive Cancer Center, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106 USA
- Center for Proteomics and Bioinformatics, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106 USA
| |
Collapse
|
50
|
Abstract
Semaphorins are key players in the control of neural circuit development. Recent studies have uncovered several exciting and novel aspects of neuronal semaphorin signalling in various cellular processes--including neuronal polarization, topographical mapping and axon sorting--that are crucial for the assembly of functional neuronal connections. This progress is important for further understanding the many neuronal and non-neuronal functions of semaphorins and for gaining insight into their emerging roles in the perturbed neural connectivity that is observed in some diseases. This Review discusses recent advances in semaphorin research, focusing on novel aspects of neuronal semaphorin receptor regulation and previously unexplored cellular functions of semaphorins in the nervous system.
Collapse
|