1
|
Murakami K, Nakayama Y, Iwata S, Emori S, Yorozu S, Fujii T, Miyamoto K. Rheumatoid meningitis in the absence of active synovitis: A potential association of semaphorin 4A. J Neuroimmunol 2025; 400:578548. [PMID: 39946854 DOI: 10.1016/j.jneuroim.2025.578548] [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: 11/17/2024] [Revised: 12/20/2024] [Accepted: 02/05/2025] [Indexed: 03/03/2025]
Abstract
Rheumatoid meningitis (RM) is a rare complication that can develop even in patients with inactive rheumatoid arthritis (RA). Currently, there are no reliable indicators that reflect the disease activity of RM, and its pathogenesis remains poorly understood. Herein, we presented three cases of RM without active synovitis and investigated the possible association between RM and semaphorin 4A (Sema4A). Two of the three patients with RM developed sudden onset of seizures, while one patient exhibited a slowly progressive gait disturbance and cognitive impairment. All the patients had inactive synovitis, positive anti-cyclic citrullinated peptide antibodies in the serum, high-intensity lesions on the cerebral surface on head magnetic resonance imaging, or a favorable response to glucocorticoids. Serum and cerebrospinal fluid (CSF) Sema4A levels in patients with RM were elevated during the acute phase compared to those in the remission phase. Serum Sema4A was significantly increased in patients with RM than in RA controls (23.8 ng/ml versus 7.48 ng/ml, p = 0.014), although there were no significant differences in RA disease activity between the two groups. Sema4A was expressed in a few infiltrating cells and stromal tissues of the RM leptomeninges. This is the first report to demonstrate that serum and CSF Sema4A levels correlate with the disease activity of RM sine active synovitis. The expression of Sema4A in the leptomeninges may be associated with RM pathogenesis.
Collapse
Affiliation(s)
- Keishu Murakami
- Department of Neurology, Wakayama Medical University, Wakayama, Japan.
| | - Yoshiaki Nakayama
- Department of Neurology, Wakayama Medical University, Wakayama, Japan
| | - Shigeru Iwata
- Department of Rheumatology and Clinical Immunology, Wakayama Medical University, Wakayama, Japan
| | - Seiji Emori
- Department of Neurology, Wakayama Medical University, Wakayama, Japan
| | - Shoko Yorozu
- Department of Neurology, Wakayama Medical University, Wakayama, Japan
| | - Takao Fujii
- Department of Rheumatology and Clinical Immunology, Wakayama Medical University, Wakayama, Japan
| | | |
Collapse
|
2
|
Zhang Y, Shi H, Dai X, Shen J, Yin J, Xu T, Yue G, Guo H, Liang R, Chen Q, Gao S, Wang L, Zhang D. Semaphorin 3A on Osteoporosis: An Overreview of the Literature. Calcif Tissue Int 2025; 116:43. [PMID: 39985619 DOI: 10.1007/s00223-025-01350-4] [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: 08/23/2024] [Accepted: 01/31/2025] [Indexed: 02/24/2025]
Abstract
Semaphorin 3A (Sema3A) is a signaling protein that has attracted increasing attention in recent years for its important role in regulating bone metabolism. In this review, we searched different databases with various combinations of keywords to analyze the effects of Sema3A on osteoporosis. Sema3A promotes bone formation and inhibits bone resorption by directly affecting the osteoblast and osteoclast or indirectly targeting the nervous system. The sympathetic nervous system may be the main link between the central nervous system and bone metabolism for Sema3A. In the peripheral nervous system, Sema3A may improve bone quality via sensory nervous innervation. In addition, estrogen is found to regulate Sema3A levels to improve bone homeostasis. Lots of Sema3A agonists have been documented to exhibit anti-osteoporotic potential in preclinical investigations. Therefore, Sema3A can be considered a novel therapeutic target for preserving bone mass, highlighting an alternative strategy for the development of anti-osteoporosis drugs.
Collapse
Affiliation(s)
- Yueyi Zhang
- Traditional Chinese Medicine School, Diabetes Research Center, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Hanfen Shi
- Traditional Chinese Medicine School, Diabetes Research Center, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xuan Dai
- Traditional Chinese Medicine School, Diabetes Research Center, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jin Shen
- Traditional Chinese Medicine School, Diabetes Research Center, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jiyuan Yin
- Traditional Chinese Medicine School, Diabetes Research Center, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Tianshu Xu
- Traditional Chinese Medicine School, Diabetes Research Center, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Gaiyue Yue
- Traditional Chinese Medicine School, Diabetes Research Center, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Haochen Guo
- Traditional Chinese Medicine School, Diabetes Research Center, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ruiqiong Liang
- Traditional Chinese Medicine School, Diabetes Research Center, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Qishuang Chen
- Traditional Chinese Medicine School, Diabetes Research Center, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Sihua Gao
- Traditional Chinese Medicine School, Diabetes Research Center, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Lili Wang
- Department of TCM Pharmacology, Chinese Material Medica School, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Dongwei Zhang
- Traditional Chinese Medicine School, Diabetes Research Center, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
3
|
Gimenez GA, Romijn M, van den Herik J, Meijer W, Eggers R, Hobo B, De Zeeuw CI, Canto CB, Verhaagen J, Carulli D. A Study on Potential Sources of Perineuronal Net-Associated Sema3A in Cerebellar Nuclei Reveals Toxicity of Non-Invasive AAV-Mediated Cre Expression in the Central Nervous System. Int J Mol Sci 2025; 26:819. [PMID: 39859534 PMCID: PMC11765860 DOI: 10.3390/ijms26020819] [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: 12/19/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Semaphorin 3A (Sema3A) is an axon guidance molecule, which is also abundant in the adult central nervous system (CNS), particularly in perineuronal nets (PNNs). PNNs are extracellular matrix structures that restrict plasticity. The cellular sources of Sema3A in PNNs are unknown. Most Sema3A-bearing neurons do not express Sema3A mRNA, suggesting that Sema3A may be released from other neurons. Another potential source of Sema3A is the choroid plexus. To identify sources of PNN-associated Sema3A, we focused on the cerebellar nuclei, which contain Sema3A+ PNNs. Cerebellar nuclei neurons receive prominent input from Purkinje cells (PCs), which express high levels of Sema3A mRNA. By using a non-invasive viral vector approach, we overexpressed Cre in PCs, the choroid plexus, or throughout the CNS of Sema3Afl/fl mice. Knocking out Sema3A in PCs or the choroid plexus was not sufficient to decrease the amount of PNN-associated Sema3A. Alternatively, knocking out Sema3A throughout the CNS induced a decrease in PNN-associated Sema3A. However, motor deficits, microgliosis, and neurodegeneration were observed, which were due to Cre toxicity. Our study represents the first attempt to unravel cellular sources of PNN-associated Sema3A and shows that non-invasive viral-mediated Cre expression throughout the CNS could lead to toxicity, complicating the interpretation of Cre-mediated Sema3A knock-out.
Collapse
Affiliation(s)
- Geoffrey-Alexander Gimenez
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (G.-A.G.); (M.R.); (J.v.d.H.); (W.M.); (R.E.); (B.H.); (J.V.)
- Department of Cerebellar Coordination & Cognition, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (C.I.D.Z.); (C.B.C.)
| | - Maurits Romijn
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (G.-A.G.); (M.R.); (J.v.d.H.); (W.M.); (R.E.); (B.H.); (J.V.)
| | - Joëlle van den Herik
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (G.-A.G.); (M.R.); (J.v.d.H.); (W.M.); (R.E.); (B.H.); (J.V.)
| | - Wouter Meijer
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (G.-A.G.); (M.R.); (J.v.d.H.); (W.M.); (R.E.); (B.H.); (J.V.)
| | - Ruben Eggers
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (G.-A.G.); (M.R.); (J.v.d.H.); (W.M.); (R.E.); (B.H.); (J.V.)
| | - Barbara Hobo
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (G.-A.G.); (M.R.); (J.v.d.H.); (W.M.); (R.E.); (B.H.); (J.V.)
| | - Chris I. De Zeeuw
- Department of Cerebellar Coordination & Cognition, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (C.I.D.Z.); (C.B.C.)
- Department of Neuroscience, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Cathrin B. Canto
- Department of Cerebellar Coordination & Cognition, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (C.I.D.Z.); (C.B.C.)
| | - Joost Verhaagen
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (G.-A.G.); (M.R.); (J.v.d.H.); (W.M.); (R.E.); (B.H.); (J.V.)
- Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Daniela Carulli
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (G.-A.G.); (M.R.); (J.v.d.H.); (W.M.); (R.E.); (B.H.); (J.V.)
| |
Collapse
|
4
|
Ahn EH, Park JB. Molecular Mechanisms of Alzheimer's Disease Induced by Amyloid-β and Tau Phosphorylation Along with RhoA Activity: Perspective of RhoA/Rho-Associated Protein Kinase Inhibitors for Neuronal Therapy. Cells 2025; 14:89. [PMID: 39851517 PMCID: PMC11764136 DOI: 10.3390/cells14020089] [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: 11/26/2024] [Revised: 12/30/2024] [Accepted: 01/06/2025] [Indexed: 01/26/2025] Open
Abstract
Amyloid-β peptide (Aβ) is a critical cause of Alzheimer's disease (AD). It is generated from amyloid precursor protein (APP) through cleavages by β-secretase and γ-secretase. γ-Secretase, which includes presenilin, is regulated by several stimuli. Tau protein has also been identified as a significant factor in AD. In particular, Tau phosphorylation is crucial for neuronal impairment, as phosphorylated Tau detaches from microtubules, leading to the formation of neurofibrillary tangles and the destabilization of the microtubule structure. This instability in microtubules damages axons and dendrites, resulting in neuronal impairment. Notably, Aβ is linked to Tau phosphorylation. Another crucial factor in AD is neuroinflammation, primarily occurring in the microglia. Microglia possess several receptors that bind with Aβ, triggering the expression and release of an inflammatory factor, although their main physiological function is to phagocytose debris and pathogens in the brain. NF-κB activation plays a major role in neuroinflammation. Additionally, the production of reactive oxygen species (ROS) in the microglia contributes to this neuroinflammation. In microglia, superoxide is produced through NADPH oxidase, specifically NOX2. Rho GTPases play an essential role in regulating various cellular processes, including cytoskeletal rearrangement, morphology changes, migration, and transcription. The typical function of Rho GTPases involves regulating actin filament formation. Neurons, with their complex processes and synapse connections, rely on cytoskeletal dynamics for structural support. Other brain cells, such as astrocytes, microglia, and oligodendrocytes, also depend on specific cytoskeletal structures to maintain their unique cellular architectures. Thus, the aberrant regulation of Rho GTPases activity can disrupt actin filaments, leading to altered cell morphology, including changes in neuronal processes and synapses, and potentially contributing to brain diseases such as AD.
Collapse
Affiliation(s)
- Eun Hee Ahn
- Department of Physiology, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea;
- Department of Neurology, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea
| | - Jae-Bong Park
- Department of Biochemistry, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea
- Institute of Cell Differentiation and Aging, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea
- ELMED Co., Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea
| |
Collapse
|
5
|
Koppel CJ, De Henau CMS, Vreeken D, DeRuiter MC, Jongbloed MRM, van Gils JM. The Role of the Axonal Guidance Cue Semaphorin 3A in Innervation of the Postnatal Heart in Health and Disease. Can J Cardiol 2024:S0828-282X(24)01327-8. [PMID: 39746509 DOI: 10.1016/j.cjca.2024.12.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 12/04/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025] Open
Abstract
During cardiac development, the heart is innervated by the autonomous nervous system. After development, neurons of the autonomic nervous system have limited capacity for growth and regeneration. However, in recent decades, it has become clear that cardiac nerves can regenerate after cardiac damage. Excessive reinnervation, so-called sympathetic hyperinnervation, may render patients vulnerable to ventricular arrhythmias and heart failure. Several studies have investigated axonal guidance cues as mediators of cardiac innervation. Axonal guidance cues direct neuronal growth of the axon and play a significant role in the regeneration and remodelling of cardiac autonomic innervation after cardiac damage. This review focusses on the current literature regarding the axonal guidance cue group of semaphorins and their function in the healthy and diseased postnatal heart. In view of cardiac innervation, most studies have focussed on semaphorin 3A (SEMA3A), whereas less is known about the function of the other semaphorin classes. SEMA3A is a neuronal repellent and is associated with a decrease in the density of sympathetic neurons in the heart. Its decline in expression after myocardial infarction plays a role in the development of sympathetic hyperinnervation and the subsequent increased risk of ventricular arrhythmias. In congestive heart failure, the opposite occurs: an increase in SEMA3A expression underlies decreased nerve density that may also serve as a substrate for ventricular arrhythmias. Although the literature on their role in cardiac innervation is still relatively scarce, semaphorins, especially SEMA3A, seem worthwhile to consider when exploring options to modulate pathologic innervation patterns in cardiovascular disease.
Collapse
Affiliation(s)
- Claire J Koppel
- Centre for Congenital Heart Disease Amsterdam-Leiden, Leiden University Medical Centre, Leiden, The Netherlands; Department of Cardiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Charlotte M S De Henau
- Department of Anatomy & Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Dianne Vreeken
- Department of Cardiology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Marco C DeRuiter
- Centre for Congenital Heart Disease Amsterdam-Leiden, Leiden University Medical Centre, Leiden, The Netherlands; Department of Anatomy & Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Monique R M Jongbloed
- Centre for Congenital Heart Disease Amsterdam-Leiden, Leiden University Medical Centre, Leiden, The Netherlands; Department of Cardiology, Leiden University Medical Centre, Leiden, The Netherlands; Department of Anatomy & Embryology, Leiden University Medical Centre, Leiden, The Netherlands.
| | - Janine M van Gils
- Department of Anatomy & Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
6
|
Reichlmeir M, Duecker RP, Röhrich H, Key J, Schubert R, Abell K, Possemato AP, Stokes MP, Auburger G. The ataxia-telangiectasia disease protein ATM controls vesicular protein secretion via CHGA and microtubule dynamics via CRMP5. Neurobiol Dis 2024; 203:106756. [PMID: 39615799 DOI: 10.1016/j.nbd.2024.106756] [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: 06/24/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 12/16/2024] Open
Abstract
The autosomal recessive disease ataxia-telangiectasia (A-T) presents with cerebellar degeneration, immunodeficiency, radiosensitivity, capillary dilatations, and pulmonary infections. Most symptoms outside the nervous system can be explained by failures of the disease protein ATM as a Ser/Thr-kinase to coordinate DNA damage repair. However, ATM in adult neurons has cytoplasmic localization and vesicle association, where its roles remain unclear. Here, we defined novel ATM protein targets in human neuroblastoma cells, and filtered initial pathogenesis events in ATM-null mouse cerebellum. Profiles of global proteome and phosphoproteomics - both direct ATM/ATR substrates and overall phosphorylation changes - confirmed previous findings for NBN, MRE11, MDC1, CHEK1, EIF4EBP1, AP3B2, PPP2R5C, SYN1 and SLC2A1. Even stronger downregulation of ATM/ATR substrate phosphopeptides after ATM-depletion was documented for CHGA, EXPH5, NBEAL2 and CHMP6 as key factors of protein secretion and endosome dynamics, as well as for CRMP5, DISP2, PHACTR1, PLXNC1, INA and TPX2 as neurite extension factors. Prominent effects on semaphorin-CRMP5-microtubule signals and ATM association with CRMP5 were validated. As a functional consequence, microtubules were stabilized, and neurite retraction ensued. The impact of ATM on secretory granules confirms previous ATM-null cerebellar transcriptome findings. This study provides the first link of A-T neural atrophy to growth cone collapse and aberrant microtubule dynamics.
Collapse
Affiliation(s)
- Marina Reichlmeir
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany.
| | - Ruth Pia Duecker
- Division for Allergy, Pneumatology and Cystic Fibrosis, Department for Children and Adolescence, Goethe-University, Frankfurt am Main, Germany.
| | - Hanna Röhrich
- Institute for Experimental Pediatric Hematology and Oncology, Medical Faculty, Goethe-University Frankfurt, Komturstrasse 3a, 60528 Frankfurt am Main, Germany.
| | - Jana Key
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany.
| | - Ralf Schubert
- Division for Allergy, Pneumatology and Cystic Fibrosis, Department for Children and Adolescence, Goethe-University, Frankfurt am Main, Germany.
| | - Kathryn Abell
- Cell Signaling Technology, Inc., Danvers, MA 01923, USA.
| | | | | | - Georg Auburger
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany.
| |
Collapse
|
7
|
Lin L, Zou J, Pei S, Huang W, Zhang Y, Zhao Z, Ding Y, Xiao C. Germinal center B-cell subgroups in the tumor microenvironment cannot be overlooked: Their involvement in prognosis, immunotherapy response, and treatment resistance in head and neck squamous carcinoma. Heliyon 2024; 10:e37726. [PMID: 39391510 PMCID: PMC11466559 DOI: 10.1016/j.heliyon.2024.e37726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 09/09/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
Background More than 60 % of patients with head and neck squamous carcinoma (HNSCC) are diagnosed at advanced stages and miss radical treatment. This has prompted the need to find new biomarkers to achieve early diagnosis and predict early recurrence and metastasis of tumors. Methods Single-cell RNA sequencing (scRNA-seq) data from HNSCC tissues and peripheral blood samples were obtained through the Gene Expression Omnibus (GEO) database (GSE164690) to characterize the B-cell subgroups, differentiation trajectories, and intercellular communication networks in HNSCC and to construct a prognostic model of the associated risks. In addition, this study analyzed the differences in clinical features, immune cell infiltration, functional enrichment, tumor mutational burden (TMB), and drug sensitivity between the high- and low-risk groups. Results Using scRNA-seq of HNSCC, we classified B and plasma cells into a total of four subgroups: naive B cells (NBs), germinal center B cells (GCBs), memory B cells (MBs), and plasma cells (PCs). Pseudotemporal trajectory analysis revealed that NBs and GCBs were at the early stage of B cell differentiation, while MBs and PCs were at the end. Cellular communication revealed that GCBs acted on tumor cells through the CD99 and SEMA4 signaling pathways. The independent prognostic value, immune cell infiltration, TMB and drug sensitivity assays were validated for the MEF2B+ GCB score groups. Conclusions We identified GCBs as B cell-specific prognostic biomarkers for the first time. The MEF2B+ GCB score fills the research gap in the genetic prognostic prediction model of HNSCC and is expected to provide a theoretical basis for finding new therapeutic targets for HNSCC.
Collapse
Affiliation(s)
- Li Lin
- Department of Stomatology, the First Affiliated Hospital of Soochow University, 188 Shi Zi Rd, Suzhou, 215006, China
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Rd, Shanghai, 200011, China
| | - Jiani Zou
- China Eastern Airlines, Comprehensive Management Department, Aviation Health Department, China
| | - Shengbin Pei
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenyi Huang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Rd, Shanghai, 200011, China
| | - Yichi Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Rd, Shanghai, 200011, China
| | - Zhijie Zhao
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Rd, Shanghai, 200011, China
| | - Yantao Ding
- Institute of Dermatology and Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui, 230032, China
- China bKey Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui, 230032, China
| | - Can Xiao
- Department of Stomatology, the First Affiliated Hospital of Soochow University, 188 Shi Zi Rd, Suzhou, 215006, China
| |
Collapse
|
8
|
Kidd T, Evans T. Analysis of Axon Guidance in the Drosophila Embryo. Cold Spring Harb Protoc 2024; 2024:pdb.top108109. [PMID: 37419653 DOI: 10.1101/pdb.top108109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2023]
Abstract
The establishment of neural connectivity is a major part of neural development. The central nervous system (CNS) midline is the most characterized axon guidance choice point, and work in Drosophila has played a pivotal role in understanding the molecular mechanisms responsible. Axons respond to attractive cues such as Netrin via the Frazzled receptor, and repulsive cues such as Slit via Robo receptors. Both signals are expressed at the CNS midline, affect pioneer axons, and have dramatic effects on the axon scaffold as a whole. Here, we focus on previous research analyzing classic mutants in the Slit/Robo pathway, which can readily be detected with a dissecting microscope. We also discuss analyzing these mutants in a teaching lab situation. The combination of sophisticated genetics and reliable axonal markers in Drosophila allows phenotypic analysis to be performed at the single-cell level. The elaborate architecture of neurons is very sensitive to disruption by genetic mutations, allowing the effects of novel mutations to be easily detected and assessed.
Collapse
Affiliation(s)
- Thomas Kidd
- Department of Biology, University of Nevada, Reno, Nevada 89557, USA
| | - Timothy Evans
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas 72701, USA
| |
Collapse
|
9
|
Moragas N, Fernandez-Nogueira P, Recalde-Percaz L, Inman JL, López-Plana A, Bergholtz H, Noguera-Castells A, Del Burgo PJ, Chen X, Sorlie T, Gascón P, Bragado P, Bissell M, Carbó N, Fuster G. The SEMA3F-NRP1/NRP2 axis is a key factor in the acquisition of invasive traits in in situ breast ductal carcinoma. Breast Cancer Res 2024; 26:122. [PMID: 39138514 PMCID: PMC11320849 DOI: 10.1186/s13058-024-01871-0] [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: 02/15/2024] [Accepted: 07/12/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND A better understanding of ductal carcinoma in situ (DCIS) is urgently needed to identify these preinvasive lesions as distinct clinical entities. Semaphorin 3F (SEMA3F) is a soluble axonal guidance molecule, and its coreceptors Neuropilin 1 (NRP1) and NRP2 are strongly expressed in invasive epithelial BC cells. METHODS We utilized two cell line models to represent the progression from a healthy state to the mild-aggressive or ductal carcinoma in situ (DCIS) stage and, ultimately, to invasive cell lines. Additionally, we employed in vivo models and conducted analyses on patient databases to ensure the translational relevance of our results. RESULTS We revealed SEMA3F as a promoter of invasion during the DCIS-to-invasive ductal carcinoma transition in breast cancer (BC) through the action of NRP1 and NRP2. In epithelial cells, SEMA3F activates epithelialmesenchymal transition, whereas it promotes extracellular matrix degradation and basal membrane and myoepithelial cell layer breakdown. CONCLUSIONS Together with our patient database data, these proof-of-concept results reveal new SEMA3F-mediated mechanisms occurring in the most common preinvasive BC lesion, DCIS, and represent potent and direct activation of its transition to invasion. Moreover, and of clinical and therapeutic relevance, the effects of SEMA3F can be blocked directly through its coreceptors, thus preventing invasion and keeping DCIS lesions in the preinvasive state.
Collapse
MESH Headings
- Humans
- Neuropilin-1/metabolism
- Neuropilin-1/genetics
- Female
- Breast Neoplasms/pathology
- Breast Neoplasms/metabolism
- Breast Neoplasms/genetics
- Neuropilin-2/metabolism
- Neuropilin-2/genetics
- Neoplasm Invasiveness
- Carcinoma, Intraductal, Noninfiltrating/metabolism
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Carcinoma, Intraductal, Noninfiltrating/genetics
- Cell Line, Tumor
- Nerve Tissue Proteins/metabolism
- Nerve Tissue Proteins/genetics
- Epithelial-Mesenchymal Transition/genetics
- Animals
- Membrane Proteins/metabolism
- Membrane Proteins/genetics
- Mice
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/genetics
- Gene Expression Regulation, Neoplastic
- Signal Transduction
Collapse
Affiliation(s)
- Núria Moragas
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona (UB), 08028, Barcelona, Spain
- Institute of Biomedicine of the Universitat de Barcelona (IBUB), Barcelona, Spain
| | - Patricia Fernandez-Nogueira
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona (UB), 08028, Barcelona, Spain
- Institute of Biomedicine of the Universitat de Barcelona (IBUB), Barcelona, Spain
- Department of Biomedicine, School of Medicine, Universitat de Barcelona (UB), 08036, Barcelona, Spain
| | - Leire Recalde-Percaz
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona (UB), 08028, Barcelona, Spain
- Institute of Biomedicine of the Universitat de Barcelona (IBUB), Barcelona, Spain
| | - Jamie L Inman
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Rd., Berkeley, CA, 94720, USA
| | - Anna López-Plana
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona (UB), 08028, Barcelona, Spain
- Institute of Biomedicine of the Universitat de Barcelona (IBUB), Barcelona, Spain
| | - Helga Bergholtz
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, 0450, Oslo, Norway
| | - Aleix Noguera-Castells
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona (UB), 08028, Barcelona, Spain
- Institute of Biomedicine of the Universitat de Barcelona (IBUB), Barcelona, Spain
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Catalonia, Spain
- Centro de Investigacion Biomedica en Red Cancer (CIBERONC), Madrid, Spain
- Department of Biosciences, Faculty of Science, Technology and Engineering, University of Vic - Central University of Catalonia (UVic-UCC), Vic, Barcelona, Catalonia, Spain
| | - Pedro J Del Burgo
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona (UB), 08028, Barcelona, Spain
| | - Xieng Chen
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona (UB), 08028, Barcelona, Spain
| | - Therese Sorlie
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, 0450, Oslo, Norway
| | - Pere Gascón
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona (UB), 08028, Barcelona, Spain
| | - Paloma Bragado
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Universidad Complutense de Madrid, Health Research Institute of the Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - Mina Bissell
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Rd., Berkeley, CA, 94720, USA
| | - Neus Carbó
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona (UB), 08028, Barcelona, Spain
- Institute of Biomedicine of the Universitat de Barcelona (IBUB), Barcelona, Spain
| | - Gemma Fuster
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona (UB), 08028, Barcelona, Spain.
- Institute of Biomedicine of the Universitat de Barcelona (IBUB), Barcelona, Spain.
- Tissue Repair and Regeneration Laboratory (TR2Lab), Institute of Research and Innovation of Life Sciences and Health, Catalunya Central (IRIS-CC), UVIC-UCC, Vic, Spain.
| |
Collapse
|
10
|
Yang Y, Guan W, Sheng XM, Gu HJ. Role of Semaphorin 3A in common psychiatric illnesses such as schizophrenia, depression, and anxiety. Biochem Pharmacol 2024; 226:116358. [PMID: 38857830 DOI: 10.1016/j.bcp.2024.116358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024]
Abstract
With societal development and an ageing population, psychiatric disorders have become a common cause of severe and long-term disability and socioeconomic burdens worldwide. Semaphorin 3A (Sema-3A) is a secreted glycoprotein belonging to the semaphorin family. Sema-3A is well known as an axon guidance factor in the neuronal system and a potent immunoregulator at all stages of the immune response. It is reported to have various biological functions and is involved in many human diseases, including autoimmune diseases, angiocardiopathy, osteoporosis, and tumorigenesis. The signals of sema-3A involved in the pathogenesis of these conditions, are transduced through its cognate receptors and diverse downstream signalling pathways. An increasing number of studies show that sema-3A plays important roles in synaptic and dendritic development, which are closely associated with the pathophysiological mechanisms of psychiatric disorders, including schizophrenia, depression, and autism, suggesting the involvement of sema-3A in the pathogenesis of mental diseases. This indicates that mutations in sema-3A and alterations in its receptors and signalling may compromise neurodevelopment and predispose patients to these disorders. However, the role of sema-3A in psychiatric disorders, particularly in regulating neurodevelopment, remains elusive. In this review, we summarise the recent progress in understanding sema-3A in the pathogenesis of mental diseases and highlight sema-3A as a potential target for the prevention and treatment of these diseases.
Collapse
Affiliation(s)
- Yang Yang
- Department of Pharmacy, Affiliated Tumor Hospital of Nantong University/Nantong Tumor Hospital, China
| | - Wei Guan
- Department of Pharmacology, Pharmacy College, Nantong University, China
| | - Xiao-Ming Sheng
- Department of Trauma Center, Affiliated Hospital of Nantong University, China
| | - Hai-Juan Gu
- Department of Pharmacy, Affiliated Tumor Hospital of Nantong University/Nantong Tumor Hospital, China.
| |
Collapse
|
11
|
Nourisanami F, Sobol M, Li Z, Horvath M, Kowalska K, Kumar A, Vlasak J, Koupilova N, Luginbuhl DJ, Luo L, Rozbesky D. Molecular mechanisms of proteoglycan-mediated semaphorin signaling in axon guidance. Proc Natl Acad Sci U S A 2024; 121:e2402755121. [PMID: 39042673 PMCID: PMC11295036 DOI: 10.1073/pnas.2402755121] [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: 02/08/2024] [Accepted: 06/20/2024] [Indexed: 07/25/2024] Open
Abstract
The precise assembly of a functional nervous system relies on axon guidance cues. Beyond engaging their cognate receptors and initiating signaling cascades that modulate cytoskeletal dynamics, guidance cues also bind components of the extracellular matrix, notably proteoglycans, yet the role and mechanisms of these interactions remain poorly understood. We found that Drosophila secreted semaphorins bind specifically to glycosaminoglycan (GAG) chains of proteoglycans, showing a preference based on the degree of sulfation. Structural analysis of Sema2b unveiled multiple GAG-binding sites positioned outside canonical plexin-binding site, with the highest affinity binding site located at the C-terminal tail, characterized by a lysine-rich helical arrangement that appears to be conserved across secreted semaphorins. In vivo studies revealed a crucial role of the Sema2b C-terminal tail in specifying the trajectory of olfactory receptor neurons. We propose that secreted semaphorins tether to the cell surface through interactions with GAG chains of proteoglycans, facilitating their presentation to cognate receptors on passing axons.
Collapse
Affiliation(s)
- Farahdokht Nourisanami
- Department of Cell Biology, Faculty of Science, Charles University, Prague 128 43, Czechia
- Laboratory of Structural Neurobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague142 20, Czechia
| | - Margarita Sobol
- Department of Cell Biology, Faculty of Science, Charles University, Prague 128 43, Czechia
- Laboratory of Structural Neurobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague142 20, Czechia
| | - Zhuoran Li
- HHMI, Department of Biology, Stanford University, Stanford, CA94305
| | - Matej Horvath
- Department of Cell Biology, Faculty of Science, Charles University, Prague 128 43, Czechia
- Laboratory of Structural Neurobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague142 20, Czechia
| | - Karolina Kowalska
- Department of Cell Biology, Faculty of Science, Charles University, Prague 128 43, Czechia
- Laboratory of Structural Neurobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague142 20, Czechia
| | - Atul Kumar
- Department of Cell Biology, Faculty of Science, Charles University, Prague 128 43, Czechia
- Laboratory of Structural Neurobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague142 20, Czechia
| | - Jonas Vlasak
- Department of Cell Biology, Faculty of Science, Charles University, Prague 128 43, Czechia
- Laboratory of Structural Neurobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague142 20, Czechia
| | - Nicola Koupilova
- Department of Cell Biology, Faculty of Science, Charles University, Prague 128 43, Czechia
- Laboratory of Structural Neurobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague142 20, Czechia
| | | | - Liqun Luo
- HHMI, Department of Biology, Stanford University, Stanford, CA94305
| | - Daniel Rozbesky
- Department of Cell Biology, Faculty of Science, Charles University, Prague 128 43, Czechia
- Laboratory of Structural Neurobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague142 20, Czechia
| |
Collapse
|
12
|
Liu J, Wang Y, Liu X, Han J, Tian Y. Spatiotemporal changes in Netrin/Dscam1 signaling dictate axonal projection direction in Drosophila small ventral lateral clock neurons. eLife 2024; 13:RP96041. [PMID: 39052321 PMCID: PMC11272162 DOI: 10.7554/elife.96041] [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] [Indexed: 07/27/2024] Open
Abstract
Axon projection is a spatial- and temporal-specific process in which the growth cone receives environmental signals guiding axons to their final destination. However, the mechanisms underlying changes in axonal projection direction without well-defined landmarks remain elusive. Here, we present evidence showcasing the dynamic nature of axonal projections in Drosophila's small ventral lateral clock neurons (s-LNvs). Our findings reveal that these axons undergo an initial vertical projection in the early larval stage, followed by a subsequent transition to a horizontal projection in the early-to-mid third instar larvae. The vertical projection of s-LNv axons correlates with mushroom body calyx expansion, while the s-LNv-expressed Down syndrome cell adhesion molecule (Dscam1) interacts with Netrins to regulate the horizontal projection. During a specific temporal window, locally newborn dorsal clock neurons secrete Netrins, facilitating the transition of axonal projection direction in s-LNvs. Our study establishes a compelling in vivo model to probe the mechanisms of axonal projection direction switching in the absence of clear landmarks. These findings underscore the significance of dynamic local microenvironments in the complementary regulation of axonal projection direction transitions.
Collapse
Affiliation(s)
- Jingjing Liu
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast UniversityNanjingChina
| | - Yuedong Wang
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast UniversityNanjingChina
| | - Xian Liu
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast UniversityNanjingChina
| | - Junhai Han
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast UniversityNanjingChina
- Co-innovation Center of Neuroregeneration, Nantong UniversityNantongChina
| | - Yao Tian
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast UniversityNanjingChina
| |
Collapse
|
13
|
Hou T, Zhang P, Tian H, Luo Y, Li J, Zhang K, Li Y. Semaphorin 4A Maintains Trophoblastic Function via Activating the STAT3 Pathway. Biomolecules 2024; 14:826. [PMID: 39062540 PMCID: PMC11274653 DOI: 10.3390/biom14070826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
The migration, proliferation, and apoptosis of trophoblastic cells play a crucial role in ensuring the effective preservation of pregnancy at the maternal-fetal interface. Any deviations in the structure and function of these cells might potentially result in the development of numerous pregnancy-related disorders, including missed abortion (MA). This study involved the examination of semaphorin 4A (SEMA4A) expression in missed abortion (n = 18) and normal early pregnancy (n = 18) villus. The findings of this study indicate a statistically significant decrease in the expression of SEMA4A in the villi of individuals diagnosed with missed abortion, as compared to the control group. The results of our vitro study showed that SEMA4A promoted the migration and proliferation of trophoblast cells and inhibited their apoptosis. Subsequent studies have shown that SEMA4A may be involved in regulating p-STAT3/STAT3, MMP9, bcl-2, and BAX levels. In summary, the findings of this study indicate a correlation between the decreased level of SEMA4A in chorionic villi and missed abortion. These results offer novel theoretical insights into the proper implantation and development of SEMA4A embryos at the maternal-fetal interface.
Collapse
Affiliation(s)
- Taotao Hou
- Graduate School of Hebei North University, Zhangjiakou 075000, China
- Department of Reproductive Genetics, Hebei General Hospital, Shijiazhuang 050051, China
- Department of Gynecology, Tianjin Beichen Hospital, Tianjin 300400, China
| | - Pingping Zhang
- Department of Reproductive Genetics, Hebei General Hospital, Shijiazhuang 050051, China
| | - Haishen Tian
- Department of Reproductive Genetics, Hebei General Hospital, Shijiazhuang 050051, China
| | - Yan Luo
- Department of Reproductive Genetics, Hebei General Hospital, Shijiazhuang 050051, China
| | - Juan Li
- Department of Reproductive Genetics, Hebei General Hospital, Shijiazhuang 050051, China
| | - Kuo Zhang
- Graduate School of University of Science and Technology Beijing, Beijing 100083, China
| | - Yali Li
- Department of Reproductive Genetics, Hebei General Hospital, Shijiazhuang 050051, China
| |
Collapse
|
14
|
Zhang Y, Zhao X, Ge D, Huang Y, Yao Q. The impact and mechanism of nerve injury on bone metabolism. Biochem Biophys Res Commun 2024; 704:149699. [PMID: 38412668 DOI: 10.1016/j.bbrc.2024.149699] [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: 11/19/2023] [Revised: 01/30/2024] [Accepted: 02/15/2024] [Indexed: 02/29/2024]
Abstract
With an increasing understanding of the mechanisms of fracture healing, it has been found that nerve injury plays a crucial role in the process, but the specific mechanism is yet to be completely revealed. To address this issue and provide novel insights for fracture treatment, we compiled this review. This review aims to study the impact of nerve injury on fracture healing, exploring the role of neurotrophic factors in the healing process. We first revisited the effects of the central nervous system (CNS) and the peripheral nervous system (PNS) on the skeletal system, and further explained the phenomenon of significantly accelerated fracture healing under nerve injury conditions. Then, from the perspective of neurotrophic factors, we delved into the physiological functions and mechanisms of neurotrophic factors, such as nerve growth factor (NGF), Neuropeptides (NPs), and Brain-derived neurotrophic factor (BDNF), in bone metabolism. These effects include direct actions on bone cells, improvement of local blood supply, regulation of bone growth factors, control of cellular signaling pathways, promotion of callus formation and bone regeneration, and synergistic or antagonistic effects with other endocrine factors, such as Sema3A and Transforming Growth Factor β (TGF-β). Finally, we discussed the treatments of fractures with nerve injuries and the future research directions in this review, suggesting that the relationship between nerve injury and fracture healing, as well as the role of nerve injury in other skeletal diseases.
Collapse
Affiliation(s)
- Yongqiang Zhang
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China; Key Lab of Additive Manufacturing Technology, Institute of Digital Medicine, Nanjing Medical University, Nanjing, China; Research Center of Digital Medicine and 3D Printing Technology of Jiangsu Province, Nanjing, China
| | - Xiao Zhao
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China; Key Lab of Additive Manufacturing Technology, Institute of Digital Medicine, Nanjing Medical University, Nanjing, China; Research Center of Digital Medicine and 3D Printing Technology of Jiangsu Province, Nanjing, China
| | - Dawei Ge
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China; Key Lab of Additive Manufacturing Technology, Institute of Digital Medicine, Nanjing Medical University, Nanjing, China; Research Center of Digital Medicine and 3D Printing Technology of Jiangsu Province, Nanjing, China
| | - Yang Huang
- International Innovation Center for Forest Chemicals & Materials and Jiangsu Co-Innovation Center of Efficient Processing & Utilization of Forest Resources, Nanjing Forestry University, Nanjing, China
| | - Qingqiang Yao
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China; Key Lab of Additive Manufacturing Technology, Institute of Digital Medicine, Nanjing Medical University, Nanjing, China; Research Center of Digital Medicine and 3D Printing Technology of Jiangsu Province, Nanjing, China.
| |
Collapse
|
15
|
Yao H, Shen Y, Song Z, Han A, Chen X, Zhang Y, Hu B. Rab11 promotes single Mauthner cell axon regeneration in vivo through axon guidance molecule Ntng2b. Exp Neurol 2024; 374:114715. [PMID: 38325655 DOI: 10.1016/j.expneurol.2024.114715] [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: 10/26/2023] [Revised: 01/23/2024] [Accepted: 02/03/2024] [Indexed: 02/09/2024]
Abstract
Effective axon regeneration within the central nervous system (CNS) is pivotal for achieving functional recovery following spinal cord injury (SCI). Numerous extrinsic and intrinsic factors exert influences on the axon regeneration. While prior studies have demonstrated crucial involvement of specific members the Rab protein family in axon regeneration in the peripheral nervous system (PNS), the precise function of Rab11 in CNS axon regeneration in vivo remains elusive. Thus, our study aimed to elucidate the impact of Rab11 on the axon regeneration of Mauthner cells (M-cells) in zebrafish larvae. Our findings demonstrated that overexpression of Rab11bb via single-cell electroporation significantly promoted axon regeneration in individual M-cells. Conversely, knockdown of Rab11bb inhibited the axon regeneration of M-cells. RNA-seq analysis revealed an upregulation of ntng2b following Rab11bb overexpression. As we hypothesized, overexpression of Ntng2b markedly enhanced axon regeneration, while Ntng2b knockdown in the context of Rab11bb pro-regeneration substantially hindered axon regrowth. In conclusion, our study demonstrated that Rab11 promotes axon regeneration of single M-cell in the CNS through the Rab11/axon guidance/Ntng2b pathway.
Collapse
Affiliation(s)
- Huaitong Yao
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Yueru Shen
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Zheng Song
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Along Han
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Xinghan Chen
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Yawen Zhang
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Bing Hu
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China.
| |
Collapse
|
16
|
Nagy GN, Zhao XF, Karlsson R, Wang K, Duman R, Harlos K, El Omari K, Wagner A, Clausen H, Miller RL, Giger RJ, Jones EY. Structure and function of Semaphorin-5A glycosaminoglycan interactions. Nat Commun 2024; 15:2723. [PMID: 38548715 PMCID: PMC10978931 DOI: 10.1038/s41467-024-46725-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 03/07/2024] [Indexed: 04/01/2024] Open
Abstract
Integration of extracellular signals by neurons is pivotal for brain development, plasticity, and repair. Axon guidance relies on receptor-ligand interactions crosstalking with extracellular matrix components. Semaphorin-5A (Sema5A) is a bifunctional guidance cue exerting attractive and inhibitory effects on neuronal growth through the interaction with heparan sulfate (HS) and chondroitin sulfate (CS) glycosaminoglycans (GAGs), respectively. Sema5A harbors seven thrombospondin type-1 repeats (TSR1-7) important for GAG binding, however the underlying molecular basis and functions in vivo remain enigmatic. Here we dissect the structural basis for Sema5A:GAG specificity and demonstrate the functional significance of this interaction in vivo. Using x-ray crystallography, we reveal a dimeric fold variation for TSR4 that accommodates GAG interactions. TSR4 co-crystal structures identify binding residues validated by site-directed mutagenesis. In vitro and cell-based assays uncover specific GAG epitopes necessary for TSR association. We demonstrate that HS-GAG binding is preferred over CS-GAG and mediates Sema5A oligomerization. In vivo, Sema5A:GAG interactions are necessary for Sema5A function and regulate Plexin-A2 dependent dentate progenitor cell migration. Our study rationalizes Sema5A associated developmental and neurological disorders and provides mechanistic insights into how multifaceted guidance functions of a single transmembrane cue are regulated by proteoglycans.
Collapse
Affiliation(s)
- Gergely N Nagy
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.
- Department of Applied Biotechnology and Food Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary.
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary.
| | - Xiao-Feng Zhao
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Richard Karlsson
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen-N, Denmark
| | - Karen Wang
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ramona Duman
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, UK
| | - Karl Harlos
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Kamel El Omari
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, UK
| | - Armin Wagner
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, UK
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen-N, Denmark
| | - Rebecca L Miller
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen-N, Denmark.
| | - Roman J Giger
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Neurology, Ann Arbor, MI, USA.
| | - E Yvonne Jones
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
17
|
Wainberg M, Forde NJ, Mansour S, Kerrebijn I, Medland SE, Hawco C, Tripathy SJ. Genetic architecture of the structural connectome. Nat Commun 2024; 15:1962. [PMID: 38438384 PMCID: PMC10912129 DOI: 10.1038/s41467-024-46023-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 02/12/2024] [Indexed: 03/06/2024] Open
Abstract
Myelinated axons form long-range connections that enable rapid communication between distant brain regions, but how genetics governs the strength and organization of these connections remains unclear. We perform genome-wide association studies of 206 structural connectivity measures derived from diffusion magnetic resonance imaging tractography of 26,333 UK Biobank participants, each representing the density of myelinated connections within or between a pair of cortical networks, subcortical structures or cortical hemispheres. We identify 30 independent genome-wide significant variants after Bonferroni correction for the number of measures studied (126 variants at nominal genome-wide significance) implicating genes involved in myelination (SEMA3A), neurite elongation and guidance (NUAK1, STRN, DPYSL2, EPHA3, SEMA3A, HGF, SHTN1), neural cell proliferation and differentiation (GMNC, CELF4, HGF), neuronal migration (CCDC88C), cytoskeletal organization (CTTNBP2, MAPT, DAAM1, MYO16, PLEC), and brain metal transport (SLC39A8). These variants have four broad patterns of spatial association with structural connectivity: some have disproportionately strong associations with corticothalamic connectivity, interhemispheric connectivity, or both, while others are more spatially diffuse. Structural connectivity measures are highly polygenic, with a median of 9.1 percent of common variants estimated to have non-zero effects on each measure, and exhibited signatures of negative selection. Structural connectivity measures have significant genetic correlations with a variety of neuropsychiatric and cognitive traits, indicating that connectivity-altering variants tend to influence brain health and cognitive function. Heritability is enriched in regions with increased chromatin accessibility in adult oligodendrocytes (as well as microglia, inhibitory neurons and astrocytes) and multiple fetal cell types, suggesting that genetic control of structural connectivity is partially mediated by effects on myelination and early brain development. Our results indicate pervasive, pleiotropic, and spatially structured genetic control of white-matter structural connectivity via diverse neurodevelopmental pathways, and support the relevance of this genetic control to healthy brain function.
Collapse
Affiliation(s)
- Michael Wainberg
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada.
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada.
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.
| | - Natalie J Forde
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Salim Mansour
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Isabel Kerrebijn
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Sarah E Medland
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Psychology, University of Queensland, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Colin Hawco
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada.
| | - Shreejoy J Tripathy
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada.
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.
- Department of Physiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
18
|
Sun G, Kropp KA, Kirchner M, Plückebaum N, Selich A, Serrero M, Dhingra A, Cabrera JR, Ritter B, Bauerfeind R, Wyler E, Landthaler M, Schambach A, Sodeik B, Mertins P, Viejo-Borbolla A. Herpes simplex virus type 1 modifies the protein composition of extracellular vesicles to promote neurite outgrowth and neuroinfection. mBio 2024; 15:e0330823. [PMID: 38275838 PMCID: PMC10865794 DOI: 10.1128/mbio.03308-23] [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: 12/13/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
The highly prevalent herpes simplex virus type 1 (HSV-1) causes a range of diseases, including cold sores, blinding keratitis, and life-threatening encephalitis. HSV-1 initially replicates in epithelial cells, enters the peripheral nervous system via neurites, and establishes lifelong infection in the neuronal cell bodies. Neurites are highly dynamic structures that grow or retract in response to attractive or repulsive cues, respectively. Here, we show that infection with HSV-1, but not with a mutant virus lacking glycoprotein G (gG), reduced the repulsive effect of epithelial cells on neurite outgrowth and facilitated HSV-1 invasion of neurons. HSV-1 gG was required and sufficient to induce neurite outgrowth by modifying the protein composition of extracellular vesicles, increasing the amount of neurotrophic and neuroprotective proteins, including galectin-1. Antibodies directed against galectin-1 neutralized the capacity of extracellular vesicles released from HSV-1-infected cells to promote neurite outgrowth. Our study provides new insights into the neurotropism of HSV-1 and identifies a viral protein that modifies the protein composition of extracellular vesicles to stimulate neurite outgrowth and invasion of the nervous system.IMPORTANCEHerpes simplex virus type 1 (HSV-1) must infect neurites (or nerve endings) to establish a chronic infection in neurons. Neurites are highly dynamic structures that retract or grow in the presence of repulsive or attractive proteins. Some of these proteins are released by epithelial cells in extracellular vesicles and act upon interaction with their receptor present on neurites. We show here that HSV-1 infection of epithelial cells modulated their effect on neurites, increasing neurite growth. Mechanistically, HSV-1 glycoprotein G (gG) modifies the protein composition of extracellular vesicles released by epithelial cells, increasing the amount of attractive proteins that enhance neurite outgrowth and facilitate neuronal infection. These results could inform of therapeutic strategies to block HSV-1 induction of neurite outgrowth and, thereby, neuronal infection.
Collapse
Affiliation(s)
- Guorong Sun
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | | | - Marieluise Kirchner
- Proteomics platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC) and Berlin Institute of Health (BIH), Berlin, Germany
| | - Nina Plückebaum
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Anton Selich
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Manutea Serrero
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Akshay Dhingra
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Jorge Rubén Cabrera
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas—Universidad Autónoma de Madrid, Madrid, Spain
| | - Birgit Ritter
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Rudolf Bauerfeind
- Research Core Unit for Laser Microscopy, Hannover Medical School, Hannover, Germany
| | - Emanuel Wyler
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Markus Landthaler
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Institute for Biology, Humboldt University of Berlin, Berlin, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Beate Sodeik
- Institute of Virology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence-Resolving Infection Susceptibility (RESIST, EXC 2155), Hannover Medical School, Hannover, Germany
| | - Philipp Mertins
- Proteomics platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC) and Berlin Institute of Health (BIH), Berlin, Germany
| | - Abel Viejo-Borbolla
- Institute of Virology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence-Resolving Infection Susceptibility (RESIST, EXC 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
19
|
Naito M, Kumanogoh A. The role of semaphorins in allergic diseases. Allergol Int 2024; 73:31-39. [PMID: 37635021 DOI: 10.1016/j.alit.2023.08.006] [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: 03/12/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 08/29/2023] Open
Abstract
Semaphorins were originally identified as guidance molecules in neural development. However, accumulating evidence indicates that 'immune semaphorins' are critically involved in regulating immune cell activation, differentiation, mobility and migration. Semaphorins are also intimately associated with the pathogenesis of allergic diseases including asthma, allergic rhinitis, atopic dermatitis, allergic conjunctivitis, and eosinophilic chronic rhinosinusitis. Interestingly, reflecting their function in positive or negative regulation of immune cells, levels of some semaphorins are increased while others are decreased in patients with allergic diseases. This review presents the pathogenic functions of immune semaphorins in allergic inflammation and discusses the potential use of these molecules as therapeutic targets for allergic diseases.
Collapse
Affiliation(s)
- Maiko Naito
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan; Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan; Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, Japan; Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Osaka, Japan; Japan Agency for Medical Research and Development - Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Osaka, Japan; Center for Advanced Modalities and DDS (CAMaD), Osaka University, Osaka, Japan.
| |
Collapse
|
20
|
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
|
21
|
Mengis T, Herger N, Heggli I, Devan J, Spirig JM, Laux CJ, Brunner F, Farshad M, Distler O, Dudli S. Bone marrow stromal cells in Modic type 1 changes promote neurite outgrowth. Front Cell Dev Biol 2023; 11:1286280. [PMID: 37965581 PMCID: PMC10641389 DOI: 10.3389/fcell.2023.1286280] [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: 08/31/2023] [Accepted: 10/10/2023] [Indexed: 11/16/2023] Open
Abstract
The pain in patients with Modic type 1 changes (MC1) is often due to vertebral body endplate pain, which is linked to abnormal neurite outgrowth in the vertebral body and adjacent endplate. The aim of this study was to understand the role of MC1 bone marrow stromal cells (BMSCs) in neurite outgrowth. BMSCs can produce neurotrophic factors, which have been shown to be pro-fibrotic in MC1, and expand in the perivascular space where sensory vertebral nerves are located. The study involved the exploration of the BMSC transcriptome in MC1, co-culture of MC1 BMSCs with the neuroblastoma cell line SH-SY5Y, analysis of supernatant cytokines, and analysis of gene expression changes in co-cultured SH-SY5Y. Transcriptomic analysis revealed upregulated brain-derived neurotrophic factor (BDNF) signaling-related pathways. Co-cultures of MC1 BMSCs with SH-SY5Y cells resulted in increased neurite sprouting compared to co-cultures with control BMSCs. The concentration of BDNF and other cytokines supporting neuron growth was increased in MC1 vs. control BMSC co-culture supernatants. Taken together, these findings show that MC1 BMSCs provide strong pro-neurotrophic cues to nearby neurons and could be a relevant disease-modifying treatment target.
Collapse
Affiliation(s)
- Tamara Mengis
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zürich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Zürich, Switzerland
| | - Nick Herger
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zürich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Zürich, Switzerland
| | - Irina Heggli
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zürich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Zürich, Switzerland
| | - Jan Devan
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zürich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Zürich, Switzerland
| | - José Miguel Spirig
- Department of Orthopedics, Balgrist University Hospital, University of Zurich, Zürich, Switzerland
| | - Christoph J. Laux
- Department of Orthopedics, Balgrist University Hospital, University of Zurich, Zürich, Switzerland
| | - Florian Brunner
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Zürich, Switzerland
| | - Mazda Farshad
- Department of Orthopedics, Balgrist University Hospital, University of Zurich, Zürich, Switzerland
| | - Oliver Distler
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zürich, Switzerland
| | - Stefan Dudli
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zürich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Zürich, Switzerland
| |
Collapse
|
22
|
Li D, Li X, Wang J, Li H, Shen H, Xu X, Chen G. Cleavage of semaphorin 4 C interferes with the neuroprotective effect of the semaphorin 4 C/Plexin B2 pathway on experimental intracerebral hemorrhage in rats. J Chem Neuroanat 2023; 132:102318. [PMID: 37482144 DOI: 10.1016/j.jchemneu.2023.102318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 07/13/2023] [Accepted: 07/20/2023] [Indexed: 07/25/2023]
Abstract
Semaphorin 4 C (SEMA4C) and its cognate receptor Plexin B2 are important regulators of axon guidance and are involved in many neurological diseases, in which SEMA4C acts not only as a ligand ("forward" mode) but also as a signaling receptor ("reverse" mode). However, the role of SEMA4C/Plexin B2 in intracerebral hemorrhage (ICH) remains unclear. In this study, ICH in adult male Sprague-Dawley rats was induced by autologous blood injection in the right basal ganglia. In vitro, cultured primary neurons were subjected to OxyHb to imitate ICH injury. Recombinant SEMA4C (rSEMA4C) and overexpressing lentiviruses encoding full-length SEMA4C or secretory SEMA4C (sSEMA4C) were administered to rats by intraventricular injection. First, we found that elevated levels of sSEMA4C in the cerebrospinal fluid (CSF) of clinical patients were associated with poor prognosis. Both SEMA4C and sSEMA4C were increased in brain tissue around the hematoma after ICH in rats. Overexpression of SEMA4C attenuated neuronal apoptosis, neurosis, and neurologic impairment after ICH. However, treatment with rSEMA4C or sSEMA4C overexpression exacerbated neuronal injury. In addition, when treated with SEMA4C overexpression, the forward mode downstream protein RhoA and the reverse mode downstream ID1/3 transcriptional factors of SEMA4C/Plexin B2 signaling were all activated. Nevertheless, when exposed to rSEMA4C or sSEMA4C overexpression, only the forward mode was activated. Thus, sSEMA4C may be a novel molecular biomarker to predict the prognosis of patients with ICH, and the prevention of SEMA4C cleavage is expected to be a promising therapeutic target.
Collapse
Affiliation(s)
- Dong Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou 215006, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou 215006, China
| | - Jiahe Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou 215006, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou 215006, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou 215006, China
| | - Xiang Xu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou 215006, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou 215006, China.
| |
Collapse
|
23
|
Martínez-Mármol R, Muhaisen A, Cotrufo T, Roselló-Busquets C, Ros O, Hernaiz-Llorens M, Pérez-Branguli F, Andrés RM, Parcerisas A, Pascual M, Ulloa F, Soriano E. Syntaxin-1 is necessary for UNC5A-C/Netrin-1-dependent macropinocytosis and chemorepulsion. Front Mol Neurosci 2023; 16:1253954. [PMID: 37829513 PMCID: PMC10565356 DOI: 10.3389/fnmol.2023.1253954] [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: 07/06/2023] [Accepted: 09/04/2023] [Indexed: 10/14/2023] Open
Abstract
Introduction Brain connectivity requires correct axonal guidance to drive axons to their appropriate targets. This process is orchestrated by guidance cues that exert attraction or repulsion to developing axons. However, the intricacies of the cellular machinery responsible for the correct response of growth cones are just being unveiled. Netrin-1 is a bifunctional molecule involved in axon pathfinding and cell migration that induces repulsion during postnatal cerebellar development. This process is mediated by UNC5 homolog receptors located on external granule layer (EGL) tracts. Methods Biochemical, imaging and cell biology techniques, as well as syntaxin-1A/B (Stx1A/B) knock-out mice were used in primary cultures and brain explants. Results and discussion Here, we demonstrate that this response is characterized by enhanced membrane internalization through macropinocytosis, but not clathrin-mediated endocytosis. We show that UNC5A, UNC5B, and UNC5C receptors form a protein complex with the t-SNARE syntaxin-1. By combining botulinum neurotoxins, an shRNA knock-down strategy and Stx1 knock-out mice, we demonstrate that this SNARE protein is required for Netrin1-induced macropinocytosis and chemorepulsion, suggesting that Stx1 is crucial in regulating Netrin-1-mediated axonal guidance.
Collapse
Affiliation(s)
- Ramón Martínez-Mármol
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology and Institute of Neurosciences, Universitat de Barcelona (UB), Barcelona, Spain
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Ashraf Muhaisen
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology and Institute of Neurosciences, Universitat de Barcelona (UB), Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED-CIBER), ISCIII, Madrid, Spain
| | - Tiziana Cotrufo
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology and Institute of Neurosciences, Universitat de Barcelona (UB), Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED-CIBER), ISCIII, Madrid, Spain
| | - Cristina Roselló-Busquets
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology and Institute of Neurosciences, Universitat de Barcelona (UB), Barcelona, Spain
| | - Oriol Ros
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology and Institute of Neurosciences, Universitat de Barcelona (UB), Barcelona, Spain
| | - Marc Hernaiz-Llorens
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology and Institute of Neurosciences, Universitat de Barcelona (UB), Barcelona, Spain
| | - Francesc Pérez-Branguli
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology and Institute of Neurosciences, Universitat de Barcelona (UB), Barcelona, Spain
- IZKF Junior Research Group and BMBF Research Group Neuroscience, IZKF, Friedrich-Alexander-Universitaet Erlangen-Nuernberg, Erlangen, Germany
| | - Rosa Maria Andrés
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology and Institute of Neurosciences, Universitat de Barcelona (UB), Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED-CIBER), ISCIII, Madrid, Spain
| | - Antoni Parcerisas
- Tissue Repair and Regeneration Laboratory (TR2Lab), Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central (IRIS-CC), Vic, Spain
- Biosciences Department, Faculty of Sciences, Technology and Engineerings, University of Vic - Central University of Catalonia (UVic-UCC), Vic, Spain
| | - Marta Pascual
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology and Institute of Neurosciences, Universitat de Barcelona (UB), Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED-CIBER), ISCIII, Madrid, Spain
| | - Fausto Ulloa
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology and Institute of Neurosciences, Universitat de Barcelona (UB), Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED-CIBER), ISCIII, Madrid, Spain
| | - Eduardo Soriano
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology and Institute of Neurosciences, Universitat de Barcelona (UB), Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED-CIBER), ISCIII, Madrid, Spain
| |
Collapse
|
24
|
Sang Y, Tsuji K, Nakanoh H, Fukushima K, Kitamura S, Wada J. Role of Semaphorin 3A in Kidney Development and Diseases. Diagnostics (Basel) 2023; 13:3038. [PMID: 37835781 PMCID: PMC10572269 DOI: 10.3390/diagnostics13193038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
Kidney diseases are worldwide public health problems affecting millions of people. However, there are still limited therapeutic options against kidney diseases. Semaphorin 3A (SEMA3A) is a secreted and membrane-associated protein, which regulates diverse functions, including immune regulation, cell survival, migration and angiogenesis, thus involving in the several pathogeneses of diseases, including eyes and neurons, as well as kidneys. SEMA3A is expressed in podocytes and tubular cells in the normal adult kidney, and recent evidence has revealed that excess SEMA3A expression and the subsequent signaling pathway aggravate kidney injury in a variety of kidney diseases, including nephrotic syndrome, diabetic nephropathy, acute kidney injury, and chronic kidney disease. In addition, several reports have demonstrated that the inhibition of SEMA3A ameliorated kidney injury via a reduction in cell apoptosis, fibrosis and inflammation; thus, SEMA3A may be a potential therapeutic target for kidney diseases. In this review article, we summarized the current knowledge regarding the role of SEMA3A in kidney pathophysiology and their potential use in kidney diseases.
Collapse
Affiliation(s)
- Yizhen Sang
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama 700-8558, Japan; (Y.S.)
- Department of Rheumatology and Immunology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Kenji Tsuji
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama 700-8558, Japan; (Y.S.)
| | - Hiroyuki Nakanoh
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama 700-8558, Japan; (Y.S.)
| | - Kazuhiko Fukushima
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama 700-8558, Japan; (Y.S.)
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Shinji Kitamura
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama 700-8558, Japan; (Y.S.)
- Department of Nursing Science, Faculty of Health and Welfare Science, Okayama Prefectural University, Okayama 719-1197, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama 700-8558, Japan; (Y.S.)
| |
Collapse
|
25
|
Toledano S, Neufeld G. Plexins as Regulators of Cancer Cell Proliferation, Migration, and Invasivity. Cancers (Basel) 2023; 15:4046. [PMID: 37627074 PMCID: PMC10452846 DOI: 10.3390/cancers15164046] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/24/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
Plexins are a family of nine single-pass transmembrane receptors with a conserved GTPase activating protein (GAP) domain. The plexin family is divided into four subfamilies: Type-A, type-B, type-C, and type-D plexins. Plexins function as receptors for axon guidance factors of the semaphorin family. The semaphorin gene family contains 22 genes that are divided into eight subclasses of which subclasses three to seven represent vertebrate semaphorins. The plexins and their semaphorin ligands have important roles as regulators of angiogenesis, cancer proliferation, and metastasis. Class 3 semaphorins, with the exception of sema3E, are the only semaphorins that do not bind directly to plexins. In order to transduce their signals, they bind instead to complexes consisting of receptors of the neuropilin family and various plexins. Some plexins also form complexes with tyrosine-kinase receptors such as the epidermal growth factor receptor ErbB2, the mesenchymal epithelial transition factor receptor (MET), and the Vascular endothelial growth factor receptor 2 (VEGFR2) and, as a result, can modulate cell proliferation and tumor progression. This review focuses on the roles of the different plexins in the control of cancer cell proliferation and invasiveness. Plexins also affect tumor progression and tumor metastasis by indirect mechanisms, such as modulation of angiogenesis and immune responses. However, these topics are not covered in the present review.
Collapse
Affiliation(s)
| | - Gera Neufeld
- The Cancer Research Center, The Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109602, Israel;
| |
Collapse
|
26
|
Mizuno Y, Nakanishi Y, Kumanogoh A. Pathophysiological functions of semaphorins in the sympathetic nervous system. Inflamm Regen 2023; 43:30. [PMID: 37291626 DOI: 10.1186/s41232-023-00281-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/30/2023] [Indexed: 06/10/2023] Open
Abstract
Upon exposure to external stressors, the body senses them and activates the sympathetic nervous system (SNS) to maintain the homeostasis, which is known as the "fight-or-flight" response. Recent studies have revealed that the SNS also plays pivotal roles in regulating immune responses, such as hematopoiesis, leukocyte mobilization, and inflammation. Indeed, overactivation of the SNS causes many inflammatory diseases, including cardiovascular diseases, metabolic disorders, and autoimmune diseases. However, the molecular basis essential for SNS-mediated immune regulation is not completely understood. In this review, we focus on axon guidance cues, semaphorins, which play multifaceted roles in neural and immune systems. We summarize the functions of semaphorins in the crosstalk between the SNS and the immune system, exploring its pathophysiological roles.
Collapse
Affiliation(s)
- Yumiko Mizuno
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Immunopathology, World Premier International Research Center Initiative Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Osaka, Japan
| | - Yoshimitsu Nakanishi
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Immunopathology, World Premier International Research Center Initiative Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Osaka, Japan
- Department of Advanced Clinical and Translational Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
- Department of Immunopathology, World Premier International Research Center Initiative Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Osaka, Japan.
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan.
- Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Osaka, Japan.
- Japan Agency for Medical Research and Development - Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Suita, Osaka, Japan.
- Center for Advanced Modalities and DDS (CAMaD), Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
27
|
Thomas R, Yang X. Semaphorins in immune cell function, inflammatory and infectious diseases. CURRENT RESEARCH IN IMMUNOLOGY 2023; 4:100060. [PMID: 37645659 PMCID: PMC10461194 DOI: 10.1016/j.crimmu.2023.100060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 05/08/2023] [Accepted: 05/14/2023] [Indexed: 08/31/2023] Open
Abstract
The Semaphorin family is a group of proteins studied broadly for their functions in nervous systems. They consist of eight subfamilies ubiquitously expressed in vertebrates, invertebrates, and viruses and exist in membrane-bound or secreted forms. Emerging evidence indicates the relevance of semaphorins outside the nervous system, including angiogenesis, cardiogenesis, osteoclastogenesis, tumour progression, and, more recently, the immune system. This review provides a broad overview of current knowledge on the role of semaphorins in the immune system, particularly its involvement in inflammatory and infectious diseases, including chlamydial infections.
Collapse
Affiliation(s)
- Rony Thomas
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Xi Yang
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
28
|
Liu S, Zhao Y, Zhang J, Liu Z. Application of single-cell RNA sequencing analysis of novel breast cancer phenotypes based on the activation of ferroptosis-related genes. Funct Integr Genomics 2023; 23:173. [PMID: 37212877 DOI: 10.1007/s10142-023-01086-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/23/2023]
Abstract
Ferroptosis is distinct from classic apoptotic cell death characterized by the accumulation of reactive oxygen species (ROS) and lipid peroxides on the cell membrane. Increasing findings have demonstrated that ferroptosis plays an important role in cancer development, but the exploration of ferroptosis in breast cancer is limited. In our study, we aimed to establish a ferroptosis activation-related model based on the differentially expressed genes between a group exhibiting high ferroptosis activation and a group exhibiting low ferroptosis activation. By using machine learning to establish the model, we verified the accuracy and efficiency of our model in The Cancer Genome Atlas Breast Invasive Carcinoma (TCGA-BRCA) set and gene expression omnibus (GEO) dataset. Additionally, our research innovatively utilized single-cell RNA sequencing data to systematically reveal the microenvironment in the high and low FeAS groups, which demonstrated differences between the two groups from comprehensive aspects, including the activation condition of transcription factors, cell pseudotime features, cell communication, immune infiltration, chemotherapy efficiency, and potential drug resistance. In conclusion, different ferroptosis activation levels play a vital role in influencing the outcome of breast cancer patients and altering the tumor microenvironment in different molecular aspects. By analyzing differences in ferroptosis activation levels, our risk model is characterized by a good prognostic capacity in assessing the outcome of breast cancer patients, and the risk score can be used to prompt clinical treatment to prevent potential drug resistance. By identifying the different tumor microenvironment landscapes between the high- and low-risk groups, our risk model provides molecular insight into ferroptosis in breast cancer patients.
Collapse
Affiliation(s)
- Shuochuan Liu
- Department of Breast Disease, Henan Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Dongming Road, Zhengzhou, 450008, Henan Province, China
| | - Yajie Zhao
- Department of Breast Disease, Henan Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Dongming Road, Zhengzhou, 450008, Henan Province, China
| | - Jiao Zhang
- Department of Breast Disease, Henan Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Dongming Road, Zhengzhou, 450008, Henan Province, China
| | - Zhenzhen Liu
- Department of Breast Disease, Henan Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Dongming Road, Zhengzhou, 450008, Henan Province, China.
| |
Collapse
|
29
|
Tang M, Xiong T. MiR-146b-5p/SEMA3G regulates epithelial-mesenchymal transition in clear cell renal cell carcinoma. Cell Div 2023; 18:4. [PMID: 36882799 PMCID: PMC9993666 DOI: 10.1186/s13008-023-00083-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 01/09/2023] [Indexed: 03/09/2023] Open
Abstract
OBJECTIVE The primary purpose was to unveil how the miR-146b-5p/SEMA3G axis works in clear cell renal cell carcinoma (ccRCC). METHODS ccRCC dataset was acquired from TCGA database, and target miRNA to be studied was further analyzed using survival analysis. We performed miRNA target gene prediction through the database, and those predicted miRNAs were intersected with differential mRNAs. After calculating the correlation between miRNAs and mRNAs, we completed the GSEA pathway enrichment analysis on mRNAs. MiRNA and mRNA expression was examined by qRT-PCR. Western blot was introduced to detect SEMA3G, MMP2, MMP9 expression, epithelial-mesenchymal transition (EMT) marker proteins, and Notch/TGF-β signaling pathway-related proteins. Targeted relationship between miRNA and mRNA was validated using a dual-luciferase test. Transwell assay was employed to assess cell migration and invasion. Wound healing assay was adopted for evaluation of migration ability. The effect of different treatments on cell morphology was observed by a microscope. RESULTS In ccRCC cells, miR-146b-5p was remarkably overexpressed, yet SEMA3G was markedly less expressed. MiR-146b-5p was capable of stimulating ccRCC cell invasion, migration and EMT, and promoting the transformation of ccRCC cell morphology to mesenchymal state. SEMA3G was targeted and inhibited via miR-146b-5p. MiR-146b-5p facilitated ccRCC cell migration, invasion, morphology transforming to mesenchymal state and EMT process by targeting SEMA3G and regulating Notch and TGF-β signaling pathways. CONCLUSION MiR-146b-5p regulated Notch and TGF-β signaling pathway by suppressing SEMA3G expression, thus promoting the growth of ccRCC cells, which provides a possible target for ccRCC therapy and prognosis prediction.
Collapse
Affiliation(s)
- Mengxi Tang
- Urinary Surgery, The People's Hospital of Rongchang District, Chongqing, 402460, China
| | - Tao Xiong
- Urinary Surgery, The People's Hospital of Rongchang District, No.3, North Square Road, Changyuan Subdistrict, Chongqing, 402460, China.
| |
Collapse
|
30
|
Komatsu K, Ko JA, Shimizu A, Okumichi H, Kiuchi Y. Functional Analysis of Semaphorin 3A in Retinal Ganglion Cells under Hypoxia In Vitro. BIOL BULL+ 2023. [DOI: 10.1134/s1062359022700017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
|
31
|
Asada N, Katayama Y. A mysterious triangle of blood, bones, and nerves. J Bone Miner Metab 2023; 41:404-414. [PMID: 36752904 DOI: 10.1007/s00774-023-01402-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 01/13/2023] [Indexed: 02/09/2023]
Abstract
The relationship between bone tissue and bone marrow, which is responsible for hematopoiesis, is inseparable. Osteoblasts and osteocytes, which produce and consist of bone tissue, regulate the function of hematopoietic stem cells (HSC), the ancestors of all hematopoietic cells in the bone marrow. The peripheral nervous system finely regulates bone remodeling in bone tissue and modulates HSC function within the bone marrow, either directly or indirectly via modification of the HSC niche function. Peripheral nerve signals also play an important role in the development and progression of malignant tumors (including hematopoietic tumors) and normal tissues, and peripheral nerve control is emerging as a potential new therapeutic target. In this review, we summarize recent findings on the linkage among blood system, bone tissue, and peripheral nerves.
Collapse
Affiliation(s)
- Noboru Asada
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikata-Cho, Kita-ku, Okayama, 700-8558, Japan.
| | - Yoshio Katayama
- Division of Hematology, Department of Medicine, Kobe University Hospital, 7-5-2 Kusunoki-Cho, Chuo-ku, Kobe, 650-0017, Japan.
| |
Collapse
|
32
|
Su Y, Wang X, Yang Y, Chen L, Xia W, Hoi KK, Li H, Wang Q, Yu G, Chen X, Wang S, Wang Y, Xiao L, Verkhratsky A, Fancy SPJ, Yi C, Niu J. Astrocyte endfoot formation controls the termination of oligodendrocyte precursor cell perivascular migration during development. Neuron 2023; 111:190-201.e8. [PMID: 36384142 PMCID: PMC9922530 DOI: 10.1016/j.neuron.2022.10.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 09/14/2022] [Accepted: 10/22/2022] [Indexed: 11/17/2022]
Abstract
Oligodendrocyte precursor cells (OPCs) undergo an extensive and coordinated migration in the developing CNS, using the pre-formed scaffold of developed blood vessels as their physical substrate for migration. While OPC association with vasculature is critical for dispersal, equally important for permitting differentiation and proper myelination of target axons is their appropriate and timely detachment, but regulation of this process remains unclear. Here we demonstrate a correlation between the developmental formation of astrocytic endfeet on vessels and the termination of OPC perivascular migration. Ex vivo and in vivo live imaging shows that astrocyte endfeet physically displace OPCs from vasculature, and genetic abrogation of endfoot formation hinders both OPC detachment from vessels and subsequent differentiation. Astrocyte-derived semaphorins 3a and 6a act to repel OPCs from blood vessels at the cessation of their perivascular migration and, in so doing, permit subsequent OPC differentiation by insulating them from a maturation inhibitory endothelial niche.
Collapse
Affiliation(s)
- Yixun Su
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China; Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| | - Xiaorui Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China
| | - Yujian Yang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China; Department of Ophthalmology, Army Specialty Medical Center, Third Military Medical University, Chongqing 400042, China
| | - Liang Chen
- Department of Orthopedics, Army Specialty Medical Center, Third Military Medical University, Chongqing 400042, China
| | - Wenlong Xia
- Department of Neurology, Department of Pediatrics, Division of Neuroimmunology and Glial Biology, Newborn Brain Research Institute, University of California at San Francisco, San Francisco, CA 94158, USA
| | - Kimberly K Hoi
- Department of Neurology, Department of Pediatrics, Division of Neuroimmunology and Glial Biology, Newborn Brain Research Institute, University of California at San Francisco, San Francisco, CA 94158, USA
| | - Hui Li
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China; Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| | - Qi Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China; Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| | - Guangdan Yu
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China
| | - Xiaoying Chen
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China
| | - Shouyu Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China
| | - Yuxin Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China
| | - Lan Xiao
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M139PL, UK; Achucarro Center for Neuroscience, IKERBASQUE, Bilbao 48011, Spain
| | - Stephen P J Fancy
- Department of Neurology, Department of Pediatrics, Division of Neuroimmunology and Glial Biology, Newborn Brain Research Institute, University of California at San Francisco, San Francisco, CA 94158, USA.
| | - Chenju Yi
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China.
| | - Jianqin Niu
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
33
|
Dumoulin A, Stoeckli ET. Looking for Guidance - Models and Methods to Study Axonal Navigation. Neuroscience 2023; 508:30-39. [PMID: 35940454 DOI: 10.1016/j.neuroscience.2022.08.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/29/2022] [Accepted: 08/01/2022] [Indexed: 01/17/2023]
Abstract
The molecular mechanisms of neural circuit formation have been of interest to Santiago Ramón y Cajal and thousands of neuroscientists sharing his passion for neural circuits ever since. Cajal was a brilliant observer and taught us about the connections and the morphology of neurons in the adult and developing nervous system. Clearly, we will not learn about molecular mechanisms by just looking at brain sections or cells in culture. Technically, we had to come a long way to today's possibilities that allow us to perturb target gene expression and watch the consequences of our manipulations on navigating axons in situ. In this review, we summarize landmark steps towards modern live-imaging approaches used to study the molecular basis of axon guidance.
Collapse
Affiliation(s)
- Alexandre Dumoulin
- Department of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Esther T Stoeckli
- Department of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| |
Collapse
|
34
|
Sullivan KG, Bashaw GJ. Intracellular Trafficking Mechanisms that Regulate Repulsive Axon Guidance. Neuroscience 2023; 508:123-136. [PMID: 35863679 PMCID: PMC9839465 DOI: 10.1016/j.neuroscience.2022.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/01/2022] [Accepted: 07/13/2022] [Indexed: 01/17/2023]
Abstract
Friedrich Bonhoeffer made seminal contributions to the study of axon guidance in the developing nervous system. His discoveries of key cellular and molecular mechanisms that dictate wiring specificity laid the foundation for countless investigators who have followed in his footsteps. Perhaps his most significant contribution was the cloning and characterization of members of the conserved ephrin family of repulsive axon guidance cues. In this review, we highlight the major contributions that Bonhoeffer and his colleagues made to the field of axon guidance, and discuss ongoing investigations into the diverse array of mechanisms that ensure that axon repulsion is precisely regulated to allow for accurate pathfinding. Specifically, we focus our discussion on the post-translational regulation of two major families of repulsive axon guidance factors: ephrin ligands and their Eph receptors, and slit ligands and their Roundabout (Robo) receptors. We will give special emphasis to the ways in which regulated endocytic trafficking events allow navigating axons to adjust their responses to repellant signals and how these trafficking events are intimately related to receptor signaling. By highlighting parallels and differences between the regulation of these two important repulsive axon guidance pathways, we hope to identify key outstanding questions for future investigation.
Collapse
Affiliation(s)
- Kelly G Sullivan
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, United States
| | - Greg J Bashaw
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, United States.
| |
Collapse
|
35
|
Li M, Jin E, Zhu L, Ren C, Liang Z, Zhao M, Qu J. Semaphorin 3A Inhibits Endoplasmic Reticulum Stress Induced by High Glucose in Müller Cells. Curr Eye Res 2023; 48:70-79. [PMID: 36271834 DOI: 10.1080/02713683.2022.2139849] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE This study aimed to explore the effect of the Semaphorin3A (Sema3A)/Neuropilin-1 (Nrp-1) pathway on Müller cell activities and endoplasmic reticulum (ER) stress induced by high glucose (HG) in vitro. METHODS The primary Müller cells of C57BL/6J mice were isolated and cultured in normal or high glucose medium. The expression of endogenous Sema3A and its coreceptor Nrp-1 was measured by Western blot. Müller cells were incubated with exogenous recombinant Sema3A protein or transfected with lentiviral vectors expressing small hairpin RNA (shRNA) to knock down the expression of endogenous Sema3A. The proliferation of Müller cells was detected by CCK-8 assay and EdU staining. The migratory ability was detected by the Transwell migration assay. The level of endoplasmic reticulum (ER) stress was analyzed through the detection of GRP78/BiP, IRE1α, phosphorylated IRE1αS724 (p-IRE1αS724), and the splicing rate of XBP1 (XBP1s/XBP1) by using immunofluorescence, Western blot or quantitative polymerase chain reaction (qPCR). RESULTS HG induced the upregulation of endogenous Sema3A and Nrp-1 receptors in Müller cells. The expression of GRP78/BiP and IRE1α was upregulated by HG, with an increased splicing rate of XBP1. Exogenous Sema3A inhibited HG-induced Müller cell proliferation, migration, and GRP78/BiP-IRE1α-XBP1 axis activation. Knockdown of Sema3A promoted proliferation, migration, and ER stress induced by high glucose in Müller cells. CONCLUSION Sema3A inhibited the increased proliferative and migratory activities induced by high glucose by attenuating ER stress in Müller cells.
Collapse
Affiliation(s)
- Mengyang Li
- Department of Ophthalmology, Peking University People's Hospital, Beijing, China.,Eye Diseases and Optometry Institute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, China
| | - Enzhong Jin
- Department of Ophthalmology, Peking University People's Hospital, Beijing, China.,Eye Diseases and Optometry Institute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, China
| | - Li Zhu
- Department of Ophthalmology, Peking University People's Hospital, Beijing, China.,Eye Diseases and Optometry Institute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, China
| | - Chi Ren
- Department of Ophthalmology, Peking University People's Hospital, Beijing, China.,Eye Diseases and Optometry Institute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, China
| | - Zhiqiao Liang
- Department of Ophthalmology, Peking University People's Hospital, Beijing, China.,Eye Diseases and Optometry Institute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, China
| | - Mingwei Zhao
- Department of Ophthalmology, Peking University People's Hospital, Beijing, China.,Eye Diseases and Optometry Institute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, China
| | - Jinfeng Qu
- Department of Ophthalmology, Peking University People's Hospital, Beijing, China.,Eye Diseases and Optometry Institute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, China
| |
Collapse
|
36
|
Ros O, Nicol X. Axon pathfinding and targeting: (R)evolution of insights from in vitro assays. Neuroscience 2023; 508:110-122. [PMID: 36096337 DOI: 10.1016/j.neuroscience.2022.09.006] [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: 03/31/2022] [Revised: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 01/17/2023]
Abstract
Investigating axonal behaviors while neurons are connecting with each other has been a challenge since the early studies on nervous system development. While molecule-driven axon pathfinding has been theorized by observing neurons at different developmental stages in vivo, direct observation and measurements of axon guidance behaviors required the invention of in vitro systems enabling to test the impact of molecules or cellular extracts on axons growing in vitro. With time, the development of novel in vivo approaches has confirmed the mechanisms highlighted in culture and has led in vitro systems to be adapted for cellular processes that are still inaccessible in intact organisms. We here review the evolution of these in vitro assays, which started with crucial contributions from the Bonhoeffer lab.
Collapse
Affiliation(s)
- Oriol Ros
- Universitat de Barcelona, Department of Cell Biology, Physiology and Immunology, Avinguda Diagonal 643, 08028 Barcelona, Catalonia, Spain
| | - Xavier Nicol
- Sorbonne Université, Inserm, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France.
| |
Collapse
|
37
|
Wang C, Song D, Huang Q, Liu Q. Advances in SEMA3F regulation of clinically high-incidence cancers. Cancer Biomark 2023; 38:131-142. [PMID: 37599522 DOI: 10.3233/cbm-230085] [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] [Indexed: 08/22/2023]
Abstract
Cancer has become a leading cause of morbidity and mortality in recent years. Its high prevalence has had a severe impact on society. Researchers have achieved fruitful results in the causative factors, pathogenesis, treatment strategies, and cancer prevention. Semaphorin 3F (SEMA3F), a member of the signaling family, was initially reported in the literature to inhibit the growth, invasion, and metastasis of cancer cells in lung cancer. Later studies showed it has cancer-inhibiting effects in malignant tumors such as breast, colorectal, ovarian, oral squamous cell carcinoma, melanoma, and head and neck squamous carcinoma. In contrast, recent studies have reported that SEMA3F is expressed more in hepatocellular carcinoma than in normal tissue and promotes metastasis of hepatocellular carcinoma. We chose lung, breast, colorectal, and hepatocellular carcinomas with high clinical prevalence to review the roles and molecular mechanisms of SEMA3F in these four carcinomas. We concluded with an outlook on clinical interventions for patients targeting SEMA3F.
Collapse
Affiliation(s)
- Chaofeng Wang
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, China
| | - Dezhi Song
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, China
| | - Qian Huang
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Qian Liu
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
38
|
Vieira JR, Shah B, Dupraz S, Paredes I, Himmels P, Schermann G, Adler H, Motta A, Gärtner L, Navarro-Aragall A, Ioannou E, Dyukova E, Bonnavion R, Fischer A, Bonanomi D, Bradke F, Ruhrberg C, Ruiz de Almodóvar C. Endothelial PlexinD1 signaling instructs spinal cord vascularization and motor neuron development. Neuron 2022; 110:4074-4089.e6. [PMID: 36549270 PMCID: PMC9796814 DOI: 10.1016/j.neuron.2022.12.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 11/04/2022] [Accepted: 12/03/2022] [Indexed: 12/24/2022]
Abstract
How the vascular and neural compartment cooperate to achieve such a complex and highly specialized structure as the central nervous system is still unclear. Here, we reveal a crosstalk between motor neurons (MNs) and endothelial cells (ECs), necessary for the coordinated development of MNs. By analyzing cell-to-cell interaction profiles of the mouse developing spinal cord, we uncovered semaphorin 3C (Sema3C) and PlexinD1 as a communication axis between MNs and ECs. Using cell-specific knockout mice and in vitro assays, we demonstrate that removal of Sema3C in MNs, or its receptor PlexinD1 in ECs, results in premature and aberrant vascularization of MN columns. Those vascular defects impair MN axon exit from the spinal cord. Impaired PlexinD1 signaling in ECs also causes MN maturation defects at later stages. This study highlights the importance of a timely and spatially controlled communication between MNs and ECs for proper spinal cord development.
Collapse
Affiliation(s)
- José Ricardo Vieira
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany; Faculty of Biosciences, Heidelberg University, Im Neuenheimer 234, 69120 Heidelberg, Germany
| | - Bhavin Shah
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany
| | - Sebastian Dupraz
- Institute for Neurovascular Cell Biology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Isidora Paredes
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany; Faculty of Biosciences, Heidelberg University, Im Neuenheimer 234, 69120 Heidelberg, Germany
| | - Patricia Himmels
- Faculty of Biosciences, Heidelberg University, Im Neuenheimer 234, 69120 Heidelberg, Germany
| | - Géza Schermann
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany; Institute for Neurovascular Cell Biology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Heike Adler
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany
| | - Alessia Motta
- San Raffaele Scientific Institute, Division of Neuroscience, via Olgettina 60, 20132 Milan, Italy
| | - Lea Gärtner
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany
| | - Ariadna Navarro-Aragall
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, EC1V 9EL London, UK
| | - Elena Ioannou
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, EC1V 9EL London, UK
| | - Elena Dyukova
- Max-Planck-Institute for Heart and Lung Research, Ludwigstr. 43, 61231 Bad Nauheim, Germany
| | - Remy Bonnavion
- Max-Planck-Institute for Heart and Lung Research, Ludwigstr. 43, 61231 Bad Nauheim, Germany
| | - Andreas Fischer
- Department of Clinical Chemistry, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany; Division Vascular Signaling and Cancer, German Cancer Research Center Heidelberg, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Dario Bonanomi
- San Raffaele Scientific Institute, Division of Neuroscience, via Olgettina 60, 20132 Milan, Italy
| | - Frank Bradke
- Laboratory of Axon Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, EC1V 9EL London, UK
| | - Carmen Ruiz de Almodóvar
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany; Institute for Neurovascular Cell Biology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany; Schlegel Chair for Neurovascular Cell Biology, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany.
| |
Collapse
|
39
|
McFalls AJ, Imperio CG, Woodward E, Krikorian C, Stoltsfus B, Wronowski B, Grigson PS, Freeman WM, Vrana KE. An RNA-seq study of the mPFC of rats with different addiction phenotypes. Brain Res Bull 2022; 191:107-120. [DOI: 10.1016/j.brainresbull.2022.09.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 09/23/2022] [Accepted: 09/28/2022] [Indexed: 11/15/2022]
|
40
|
Ferretti G, Romano A, Sirabella R, Serafini S, Maier TJ, Matrone C. An increase in Semaphorin 3A biases the axonal direction and induces an aberrant dendritic arborization in an in vitro model of human neural progenitor differentiation. Cell Biosci 2022; 12:182. [DOI: 10.1186/s13578-022-00916-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/17/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Semaphorins (Sema) belong to a large family of repellent guidance cues instrumental in guiding axons during development. In particular, Class 3 Sema (Sema 3) is among the best characterized Sema family members and the only produced as secreted proteins in mammals, thereby exerting both autocrine and paracrine functions. Intriguingly, an increasing number of studies supports the crucial role of the Sema 3A in hippocampal and cortical neurodevelopment. This means that alterations in Sema 3A signaling might compromise hippocampal and cortical circuits and predispose to disorders such as autism and schizophrenia. Consistently, increased Sema 3A levels have been detected in brain of patients with schizophrenia and many polymorphisms in Sema 3A or in the Sema 3A receptors, Neuropilins (Npn 1 and 2) and Plexin As (Plxn As), have been associated to autism.
Results
Here we present data indicating that when overexpressed, Sema 3A causes human neural progenitors (NP) axonal retraction and an aberrant dendritic arborization. Similarly, Sema 3A, when overexpressed in human microglia, triggers proinflammatory processes that are highly detrimental to themselves as well as NP. Indeed, NP incubated in microglia overexpressing Sema 3A media retract axons within an hour and then start suffering and finally die. Sema 3A mediated retraction appears to be related to its binding to Npn 1 and Plxn A2 receptors, thus activating the downstream Fyn tyrosine kinase pathway that promotes the threonine-serine kinase cyclin-dependent kinase 5, CDK5, phosphorylation at the Tyr15 residue and the CDK5 processing to generate the active fragment p35.
Conclusions
All together this study identifies Sema 3A as a critical regulator of human NP differentiation. This may imply that an insult due to Sema 3A overexpression during the early phases of neuronal development might compromise neuronal organization and connectivity and make neurons perhaps more vulnerable to other insults across their lifespan.
Collapse
|
41
|
Tanaka T, Ekimoto T, Nagatomo M, Neyazaki M, Shimoji E, Yamane T, Kanagawa S, Oi R, Mihara E, Takagi J, Akashi S, Ikeguchi M, Nogi T. Hybrid in vitro/in silico analysis of low-affinity protein-protein interactions that regulate signal transduction by Sema6D. Protein Sci 2022; 31:e4452. [PMID: 36156831 PMCID: PMC9601788 DOI: 10.1002/pro.4452] [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: 07/09/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/09/2022]
Abstract
Semaphorins constitute a large family of secreted and membrane-bound proteins that signal through cell-surface receptors, plexins. Semaphorins generally use low-affinity protein-protein interactions to bind with their specific plexin(s) and regulate distinct cellular processes such as neurogenesis, immune response, and organogenesis. Sema6D is a membrane-bound semaphorin that interacts with class A plexins. Sema6D exhibited differential binding affinities to class A plexins in prior cell-based assays, but the molecular mechanism underlying this selectivity is not well understood. Therefore, we performed hybrid in vitro/in silico analysis to examine the binding mode of Sema6D to class A plexins and to identify residues that give rise to the differential affinities and thus contribute to the selectivity within the same class of semaphorins. Our biophysical binding analysis indeed confirmed that Sema6D has a higher affinity for Plexin-A1 than for other class A plexins, consistent with the binding selectivity observed in the previous cell-based assays. Unexpectedly, our present crystallographic analysis of the Sema6D-Plexin-A1 complex showed that the pattern of polar interactions is not interaction-specific because it matches the pattern in the prior structure of the Sema6A-Plexin-A2 complex. Thus, we performed in silico alanine scanning analysis and discovered hotspot residues that selectively stabilized the Sema6D-Plexin-A1 pair via Van der Waals interactions. We then validated the contribution of these hotspot residues to the variation in binding affinity with biophysical binding analysis and molecular dynamics simulations on the mutants. Ultimately, our present results suggest that shape complementarity in the binding interfaces is a determinant for binding selectivity.
Collapse
Affiliation(s)
- Tsubasa Tanaka
- Graduate School of Medical Life ScienceYokohama City UniversityYokohamaKanagawaJapan
| | - Toru Ekimoto
- Graduate School of Medical Life ScienceYokohama City UniversityYokohamaKanagawaJapan
| | - Meri Nagatomo
- Graduate School of Medical Life ScienceYokohama City UniversityYokohamaKanagawaJapan
| | - Makiko Neyazaki
- Graduate School of Medical Life ScienceYokohama City UniversityYokohamaKanagawaJapan
| | - Erena Shimoji
- Graduate School of Medical Life ScienceYokohama City UniversityYokohamaKanagawaJapan
| | - Tsutomu Yamane
- Center for Computational Science, RIKENYokohamaKanagawaJapan
| | - Sakura Kanagawa
- Graduate School of Medical Life ScienceYokohama City UniversityYokohamaKanagawaJapan
| | - Rika Oi
- Graduate School of Medical Life ScienceYokohama City UniversityYokohamaKanagawaJapan
| | - Emiko Mihara
- Institute for Protein Research, Osaka UniversitySuitaOsakaJapan
| | - Junichi Takagi
- Institute for Protein Research, Osaka UniversitySuitaOsakaJapan
| | - Satoko Akashi
- Graduate School of Medical Life ScienceYokohama City UniversityYokohamaKanagawaJapan
| | - Mitsunori Ikeguchi
- Graduate School of Medical Life ScienceYokohama City UniversityYokohamaKanagawaJapan
- Center for Computational Science, RIKENYokohamaKanagawaJapan
| | - Terukazu Nogi
- Graduate School of Medical Life ScienceYokohama City UniversityYokohamaKanagawaJapan
| |
Collapse
|
42
|
Vela-Alcantara AM, Rios-Ramirez A, Santiago-Garcia J, Rodriguez-Alba JC, Tamariz Domínguez E. Modulation of DRG neurons response to semaphorin 3A via substrate stiffness. Cells Dev 2022; 171:203800. [PMID: 35717026 DOI: 10.1016/j.cdev.2022.203800] [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: 12/13/2021] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 01/25/2023]
Abstract
Semaphorin 3A (Sema3a) is a chemotropic protein that acts as a neuronal guidance cue and plays a major role in dorsal root ganglion (DRG) sensory neurons projection during embryo development. The present study evaluated the impact of stiffness in the repulsive response of DRG neurons to Sema3a when cultured over substrates of variable stiffness. Stiffness modified DRG neurons morphology and regulated their response to Sema3a, reducing the collapse of growth cones when they were cultured on softer substrates. Sema3a receptors expression was also regulated by stiffness, neuropilin-1 was overexpressed and plexin A4 mRNA was downregulated in stiffer substrates. Cytoskeleton distribution was also modified by stiffness. In softer substrates, βIII-tubulin and actin co-localized up to the leading edge of the growth cones, and as the substrate became stiffer, βIII-tubulin was confined to the transition and peripheral domains of the growth cone. Moreover, a decrease in the α-actinin adaptor protein was also observed in softer substrates. Our results show that substrate stiffness plays an important role in regulating the collapse response to Sema3a and that the modulation of cytoskeleton distribution and Sema3a receptors expression are related to the differential collapse responses of the growth cones.
Collapse
Affiliation(s)
- Ana Monserrat Vela-Alcantara
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Av. Luis Castelazo Ayala s/n, 91190 Xalapa, Veracruz, Mexico; Maestría y Doctorado en Ciencias de la Salud, Instituto de Ciencias de la Salud, Universidad Veracruzana, Mexico.
| | - Ariadna Rios-Ramirez
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Blvd. Juriquilla #3001, 76230 Juriquilla, Querétaro, Mexico.
| | - Juan Santiago-Garcia
- Instituto de Investigaciones Biológicas, Universidad Veracruzana, Av. Luis Castelazo Ayala s/n, 91190 Xalapa, Veracruz, Mexico.
| | - Juan Carlos Rodriguez-Alba
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Av. Luis Castelazo Ayala s/n, 91190 Xalapa, Veracruz, Mexico.
| | - Elisa Tamariz Domínguez
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Av. Luis Castelazo Ayala s/n, 91190 Xalapa, Veracruz, Mexico.
| |
Collapse
|
43
|
Neuroimmune crosstalk in the cornea: The role of immune cells in corneal nerve maintenance during homeostasis and inflammation. Prog Retin Eye Res 2022; 91:101105. [PMID: 35868985 DOI: 10.1016/j.preteyeres.2022.101105] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 12/29/2022]
Abstract
In the cornea, resident immune cells are in close proximity to sensory nerves, consistent with their important roles in the maintenance of nerves in both homeostasis and inflammation. Using in vivo confocal microscopy in humans, and ex vivo immunostaining and fluorescent reporter mice to visualize corneal sensory nerves and immune cells, remarkable progress has been made to advance our understanding of the physical and functional interactions between corneal nerves and immune cells. In this review, we summarize and discuss recent studies relating to corneal immune cells and sensory nerves, and their interactions in health and disease. In particular, we consider how disrupted corneal nerve axons can induce immune cell activity, including in dendritic cells, macrophages and other infiltrating cells, directly and/or indirectly by releasing neuropeptides such as substance P and calcitonin gene-related peptide. We summarize growing evidence that the role of corneal intraepithelial immune cells is likely different in corneal wound healing versus other inflammatory-dominated conditions. The role of different types of macrophages is also discussed, including how stromal macrophages with anti-inflammatory phenotypes communicate with corneal nerves to provide neuroprotection, while macrophages with pro-inflammatory phenotypes, along with other infiltrating cells including neutrophils and CD4+ T cells, can be inhibitory to corneal re-innervation. Finally, this review considers the bidirectional interactions between corneal immune cells and corneal nerves, and how leveraging this interaction could represent a potential therapeutic approach for corneal neuropathy.
Collapse
|
44
|
Kissoondoyal A, Crawford DA. Prostaglandin E2 Increases Neurite Length and the Formation of Axonal Loops, and Regulates Cone Turning in Differentiating NE4C Cells Via PKA. Cell Mol Neurobiol 2022; 42:1385-1397. [PMID: 33389417 PMCID: PMC11421704 DOI: 10.1007/s10571-020-01029-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/09/2020] [Indexed: 12/13/2022]
Abstract
Prostaglandin E2 (PGE2) is a membrane-derived lipid signaling molecule important in neuronal development. Abnormal levels of PGE2, due to environmental insults prenatal development, have been linked to brain pathologies. We have previously shown that the addition of PGE2 to neuroectodermal (NE4C) stem cells affects early stages of neuronal differentiation (day 0-8) including increased stem cell motility, accelerated formation of neurospheres, and elevated calcium levels in growth cones. In this study, we further examine whether PGE2 can influence actin-dependent neuronal morphology in later stages (day 8-12) of NE4C cell differentiation. We show that exposure to PGE2 from the initiation of differentiation increased neurite length and the proportion of neurites that formed axonal loops. We also observed changes in the proportion of turning growth cones as the differentiation progressed, with a reduced likelihood of observing turning (or asymmetrical) growth cones on day 8 and increased odds on days 10 and 12. Moreover, we showed for the first time that the observed changes in cytoskeletal morphology were PGE2/PKA dependent. Interestingly, we also found that PGE2 decreased the total protein levels of the actin-bound form of spinophilin and increased levels of unbound PKA-phosphorylated ser94-spinophilin. Hence, we propose that exposure to PGE2 can destabilize the actin cytoskeleton at various stages of neuronal differentiation due to dissociation of ser94-spinophilin causing changes in neuronal morphology.
Collapse
Affiliation(s)
- Ashby Kissoondoyal
- School of Kinesiology and Health Science, York University, Toronto, ON, M3J 1P3, Canada
- Neuroscience Graduate Diploma Program, York University, Toronto, ON, M3J 1P3, Canada
| | - Dorota A Crawford
- School of Kinesiology and Health Science, York University, Toronto, ON, M3J 1P3, Canada.
- Neuroscience Graduate Diploma Program, York University, Toronto, ON, M3J 1P3, Canada.
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada.
| |
Collapse
|
45
|
Sheng J, Xu J, Geng K, Liu D. Sema6D Regulates Zebrafish Vascular Patterning and Motor Neuronal Axon Growth in Spinal Cord. Front Mol Neurosci 2022; 15:854556. [PMID: 35465091 PMCID: PMC9021825 DOI: 10.3389/fnmol.2022.854556] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/02/2022] [Indexed: 12/22/2022] Open
Abstract
Vessels and nerves are closely associated in anatomy as well as functions. Accumulating evidences have demonstrated that axon-guiding signals may affect endothelial cells migration and path finding, which is crucial for the patterning of both the complex vascular network and neural system. However, studies regarding the functional overlap between vascular and neuronal orchestrating are still incomplete. Semaphorin6D (Sema6D) belongs to the Semaphorin family and has been identified as an important regulating factor in diverse biological processes. Its roles in vascular development are still unclear. Here, we confirmed that sema6D is enriched in neural system and blood vessels of zebrafish embryos by in situ hybridization. Then, the deficiency of sema6D caused by specific antisense morpholino-oligonucleotides (MO) led to dramatic path finding defects in both intersegmental vessels (ISVs) and primary motor neurons (PMNs) of spinal cord in zebrafish embryos. Furthermore, these defective phenotypes were confirmed in F0 generation of sema6D knockouts and rescue experiments by overexpression of sema6D mRNA in sema6D morphants. These data collectively indicate that sema6D regulates zebrafish vascular patterning and motor neuronal axon growth in the spinal cord, which might be of great therapeutical use to regulate vessel and nerve guidance in the relevant diseases that affect both systems.
Collapse
Affiliation(s)
- Jiajing Sheng
- Nantong Laboratory of Development and Diseases, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, School of Life Science, Second Affiliated Hospital, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Jiehuan Xu
- Medical School, Nantong University, Nantong, China
| | - Kaixi Geng
- Nantong Laboratory of Development and Diseases, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, School of Life Science, Second Affiliated Hospital, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Dong Liu
- Nantong Laboratory of Development and Diseases, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, School of Life Science, Second Affiliated Hospital, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- *Correspondence: Dong Liu
| |
Collapse
|
46
|
Du H, Xu Y, Zhu L. Role of Semaphorins in Ischemic Stroke. Front Mol Neurosci 2022; 15:848506. [PMID: 35350431 PMCID: PMC8957939 DOI: 10.3389/fnmol.2022.848506] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/17/2022] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke is one of the major causes of neurological morbidity and mortality in the world. Although the management of ischemic stroke has been improved significantly, it still imposes a huge burden on the health and property. The integrity of the neurovascular unit (NVU) is closely related with the prognosis of ischemic stroke. Growing evidence has shown that semaphorins, a family of axon guidance cues, play a pivotal role in multiple pathophysiological processes in NVU after ischemia, such as regulating the immune system, angiogenesis, and neuroprotection. Modulating the NVU function via semaphorin signaling has a potential to develop a novel therapeutic strategy for ischemic stroke. We, therefore, review recent progresses on the role of semphorin family members in neurons, glial cells and vasculature after ischemic stroke.
Collapse
Affiliation(s)
- Huaping Du
- Department of Neurology, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, China
| | - Yuan Xu
- Department of Neurology, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, China
| | - Li Zhu
- Department of Neurology, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, China
- Suzhou Key Laboratory of Thrombosis and Vascular Biology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Collaborative Innovation Center of Hematology of Jiangsu Province, National Clinical Research Center for Hematologic Diseases, Cyrus Tang Medical Institute, Soochow University, Suzhou, China
- *Correspondence: Li Zhu,
| |
Collapse
|
47
|
Ramšak Ž, Modic V, Li RA, vom Berg C, Zupanic A. From Causal Networks to Adverse Outcome Pathways: A Developmental Neurotoxicity Case Study. FRONTIERS IN TOXICOLOGY 2022; 4:815754. [PMID: 35295214 PMCID: PMC8915909 DOI: 10.3389/ftox.2022.815754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/31/2022] [Indexed: 11/15/2022] Open
Abstract
The last decade has seen the adverse outcome pathways (AOP) framework become one of the most powerful tools in chemical risk assessment, but the development of new AOPs remains a slow and manually intensive process. Here, we present a faster approach for AOP generation, based on manually curated causal toxicological networks. As a case study, we took a recently published zebrafish developmental neurotoxicity network, which contains causally connected molecular events leading to neuropathologies, and developed two new adverse outcome pathways: Inhibition of Fyna (Src family tyrosine kinase A) leading to increased mortality via decreased eye size (AOP 399 on AOP-Wiki) and GSK3beta (Glycogen synthase kinase 3 beta) inactivation leading to increased mortality via defects in developing inner ear (AOP 410). The approach consists of an automatic separation of the toxicological network into candidate AOPs, filtering the AOPs according to available evidence and length as well as manual development of new AOPs and weight-of-evidence evaluation. The semiautomatic approach described here provides a new opportunity for fast and straightforward AOP development based on large network resources.
Collapse
Affiliation(s)
- Živa Ramšak
- Department of Biotechnology and Systems Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Vid Modic
- Department of Biotechnology and Systems Biology, National Institute of Biology, Ljubljana, Slovenia
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Roman A. Li
- Department of Environmental Toxicology, Eawag—Swiss Federal Institute of Aquatic Science and Technology, Duebendorf, Switzerland
| | - Colette vom Berg
- Department of Environmental Toxicology, Eawag—Swiss Federal Institute of Aquatic Science and Technology, Duebendorf, Switzerland
| | - Anze Zupanic
- Department of Biotechnology and Systems Biology, National Institute of Biology, Ljubljana, Slovenia
- *Correspondence: Anze Zupanic,
| |
Collapse
|
48
|
Molecular mechanisms regulating the spatial configuration of neurites. Semin Cell Dev Biol 2022; 129:103-114. [PMID: 35248463 DOI: 10.1016/j.semcdb.2022.02.015] [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: 12/14/2021] [Revised: 02/13/2022] [Accepted: 02/17/2022] [Indexed: 02/08/2023]
Abstract
Precise neural networks, composed of axons and dendrites, are the structural basis for information processing in the brain. Therefore, the correct formation of neurites is critical for accurate neural function. In particular, the three-dimensional structures of dendrites vary greatly among neuron types, and the unique shape of each dendrite is tightly linked to specific synaptic connections with innervating axons and is correlated with its information processing. Although many systems are involved in neurite formation, the developmental mechanisms that control the orientation, size, and arborization pattern of neurites definitively defines their three-dimensional structure in tissues. In this review, we summarize these regulatory mechanisms that establish proper spatial configurations of neurites, especially dendrites, in invertebrates and vertebrates.
Collapse
|
49
|
Lotfi R, Zamanimehr N. Semaphorin-3A: a promising therapeutic tool in allergic rhinitis. Immunol Res 2022; 70:135-142. [PMID: 35031951 DOI: 10.1007/s12026-022-09264-1] [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: 11/08/2021] [Accepted: 01/10/2022] [Indexed: 11/28/2022]
Abstract
Semaphorin-3A (Sema-3A), a secreted member of the semaphorin family, is well known for playing regulatory functions at all stages of the immune response. Sema-3A transduces signals by binding to its cognate receptors, namely, class A plexins (Plxns A1 to A4) and neuropilin-1 (Nrp-1). The downstream diverse signaling pathways induced by connecting Sema-3A to its receptors were found to be involved in the pathogenesis of different immunological disorders, ranging from cancer to autoimmunity and allergies. Recent studies have demonstrated that Sema-3A expression is diminished in the murine models and patients with allergic rhinitis (AR; a chronic inflammatory disorder of the nasal mucosa), suggesting the involvement of Sema-3A in AR pathogenesis. Investigations also revealed that treatment of these mice with exogenous Sema-3A protein alleviates the clinical symptom scores of AR, thereby compensating for the reduced expression of Sema-3A in AR. Indeed, Sema-3A treatment could suppress allergic responses in AR via inhibiting Th2/Th17 responses and boosting Th1/Treg responses. Also, Sema-3A could diminish dendritic cell (DC) maturation and T cell proliferation. Since it is implicated in the pathogenesis of AR; thus, Sema-3A turns to be a promising tool of therapy to be studied and utilized in this disease. This review intends to highlight the recent evidence on the role of Sema-3A in AR pathogenesis and summarizes the recent findings regarding the expression status of Sema-3A, as well as its therapeutic potential for treating this disease. HIGHLIGHTS: Sema-3A plays regulatory functions at all stages of the immune response. Sema-3A receptors are the class A plexins (A1-A4) and neuropilin-1 (Nrp-1). Sema-3A expression is reduced in murine models and patients with allergic rhinitis. Connecting Sema-3A to Nrp-1 increases Foxp3 expression in Treg cells. Injecting Sema-3A protein exerts therapeutic effects in mouse models of allergic diseases. Sema-3A shows promise as a therapeutic tool for the treatment of allergic rhinitis.
Collapse
Affiliation(s)
- Ramin Lotfi
- Clinical Research Development Center, Tohid Hospital, Kurdistan University of Medical Sciences, Sanandaj, Iran. .,Lung Diseases and Allergy Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, 6617713446, Sanandaj, Iran.
| | - Nahid Zamanimehr
- Clinical Research Development Center, Tohid Hospital, Kurdistan University of Medical Sciences, Sanandaj, Iran.,Department of Emergency Medicine, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
50
|
Abstract
The Tabula Gallus is a proposed project that aims to create a map of every cell type in the chicken body and chick embryos. Chickens (Gallus gallus) are one of the most recognized model animals that recapitulate the development and physiology of mammals. The Tabula Gallus will generate a compendium of single-cell transcriptome data from Gallus gallus, characterize each cell type, and provide tools for the study of the biology of this species, similar to other ongoing cell atlas projects (Tabula Muris and Tabula Sapiens/Human Cell Atlas for mice and humans, respectively). The Tabula Gallus will potentially become an international collaboration between many researchers. This project will be useful for the basic scientific study of Gallus gallus and other birds (e.g., cell biology, molecular biology, developmental biology, neuroscience, physiology, oncology, virology, behavior, ecology, and evolution). It will eventually be beneficial for a better understanding of human health and diseases.
Collapse
|