1
|
Liu Z, Pan C, Huang H. The role of axon guidance molecules in the pathogenesis of epilepsy. Neural Regen Res 2025; 20:1244-1257. [PMID: 39075893 PMCID: PMC11624883 DOI: 10.4103/nrr.nrr-d-23-01620] [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: 09/27/2023] [Revised: 12/06/2023] [Accepted: 02/21/2024] [Indexed: 07/31/2024] Open
Abstract
Current treatments for epilepsy can only manage the symptoms of the condition but cannot alter the initial onset or halt the progression of the disease. Consequently, it is crucial to identify drugs that can target novel cellular and molecular mechanisms and mechanisms of action. Increasing evidence suggests that axon guidance molecules play a role in the structural and functional modifications of neural networks and that the dysregulation of these molecules is associated with epilepsy susceptibility. In this review, we discuss the essential role of axon guidance molecules in neuronal activity in patients with epilepsy as well as the impact of these molecules on synaptic plasticity and brain tissue remodeling. Furthermore, we examine the relationship between axon guidance molecules and neuroinflammation, as well as the structural changes in specific brain regions that contribute to the development of epilepsy. Ample evidence indicates that axon guidance molecules, including semaphorins and ephrins, play a fundamental role in guiding axon growth and the establishment of synaptic connections. Deviations in their expression or function can disrupt neuronal connections, ultimately leading to epileptic seizures. The remodeling of neural networks is a significant characteristic of epilepsy, with axon guidance molecules playing a role in the dynamic reorganization of neural circuits. This, in turn, affects synapse formation and elimination. Dysregulation of these molecules can upset the delicate balance between excitation and inhibition within a neural network, thereby increasing the risk of overexcitation and the development of epilepsy. Inflammatory signals can regulate the expression and function of axon guidance molecules, thus influencing axonal growth, axon orientation, and synaptic plasticity. The dysregulation of neuroinflammation can intensify neuronal dysfunction and contribute to the occurrence of epilepsy. This review delves into the mechanisms associated with the pathogenicity of axon guidance molecules in epilepsy, offering a valuable reference for the exploration of therapeutic targets and presenting a fresh perspective on treatment strategies for this condition.
Collapse
Affiliation(s)
- Zheng Liu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Chunhua Pan
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Hao Huang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| |
Collapse
|
2
|
Yukawa K. [Elucidating the Pathophysiology of Various Diseases by Investigating the Role of Molecules in Brain Wiring]. YAKUGAKU ZASSHI 2025; 145:133-143. [PMID: 39894482 DOI: 10.1248/yakushi.24-00192] [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: 02/04/2025]
Abstract
Semaphorins and their receptors plexins are axon guidance molecules that navigate axons to their final destinations during neural development. Semaphorins and plexins exert distinct roles in regulating biological functions such as the immune system and bone homeostasis. They also participate in the development and progression of various diseases such as osteoporosis and allergic diseases. This review describes the varied phenotypes revealed by the analysis of semaphorin or plexin knockout mice and discusses the association with pathogenesis and therapy of atherosclerosis, agenesis of the corpus callosum, and neuropsychiatric diseases. The deletion of semaphorin 4D in atherosclerosis-prone Apolipoprotein E-deficient mice mitigated atherosclerotic lesions, indicating its crucial involvement in the progression of atherosclerosis. Semaphorin 4D is also implicated in apoptosis induced by the estrogen-dependent generation of soluble semaphorin 4D and the active form of plexin-B1 in the postnatal vaginal opening in mice. Plexin-A1 knockout BALB/cA mice exhibited the agenesis of corpus callosum. This study indicates the crucial role of plexin-A1 in the midline crossing of callosal pioneer axons projecting from the cerebral cortex during the early phase of callosal formation. Adult plexin-A1-deficient mice exhibit reduced prepulse inhibition deficit, an endophenotype of schizophrenia, in addition to excessive self-grooming. Parvalbumin-expressing interneurons in the medial prefrontal cortex are significantly decreased in plexin-A1 knockout mice. In the parvalbumin neurons, oxidative stress is significantly increased in plexin-A1 knockout mice. Accordingly, plexin-A1 deficiency may augment oxidative stress in parvalbumin neurons, thereby impairing the parvalbumin neuron network and leading to behavioral abnormalities relevant to neuropsychiatric diseases.
Collapse
Affiliation(s)
- Kazunori Yukawa
- Faculty of Pharmacy, Meijo University
- Graduate School of Pharmacy, Meijo University
| |
Collapse
|
3
|
Kume M, Koguchi-Yoshioka H, Nakai S, Matsumura Y, Tanemura A, Yokoi K, Matsuda S, Nakamura Y, Otani N, Taminato M, Tomita K, Kubo T, Wataya-Kaneda M, Kumanogoh A, Fujimoto M, Watanabe R. Downregulation of semaphorin 4A in keratinocytes reflects the features of non-lesional psoriasis. eLife 2024; 13:RP97654. [PMID: 39737847 DOI: 10.7554/elife.97654] [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: 01/01/2025] Open
Abstract
Psoriasis is a multifactorial disorder mediated by IL-17-producing T cells, involving immune cells and skin-constituting cells. Semaphorin 4A (Sema4A), an immune semaphorin, is known to take part in T helper type 1/17 differentiation and activation. However, Sema4A is also crucial for maintaining peripheral tissue homeostasis and its involvement in skin remains unknown. Here, we revealed that while Sema4A expression was pronounced in psoriatic blood lymphocytes and monocytes, it was downregulated in the keratinocytes of both psoriatic lesions and non-lesions compared to controls. Imiquimod application induced more severe dermatitis in Sema4A knockout (KO) mice compared to wild-type (WT) mice. The naïve skin of Sema4A KO mice showed increased T cell infiltration and IL-17A expression along with thicker epidermis and distinct cytokeratin expression compared to WT mice, which are hallmarks of psoriatic non-lesions. Analysis of bone marrow chimeric mice suggested that Sema4A expression in keratinocytes plays a regulatory role in imiquimod-induced dermatitis. The epidermis of psoriatic non-lesion and Sema4A KO mice demonstrated mTOR complex 1 upregulation, and the application of mTOR inhibitors reversed the skewed expression of cytokeratins in Sema4A KO mice. Conclusively, Sema4A-mediated signaling cascades can be triggers for psoriasis and targets in the treatment and prevention of psoriasis.
Collapse
Affiliation(s)
- Miki Kume
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hanako Koguchi-Yoshioka
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Neurocutaneous Medicine, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shuichi Nakai
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
- Research Department, Maruho Co, Ltd., Kyoto, Japan
| | - Yutaka Matsumura
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Atsushi Tanemura
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kazunori Yokoi
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shoichi Matsuda
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
- Research Department, Maruho Co, Ltd., Kyoto, Japan
| | - Yuumi Nakamura
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
- Cutaneous Allergy and Host Defense, Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan
| | - Naoya Otani
- Department of Plastic Surgery, Course of Organ Regulation Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Mifue Taminato
- Department of Plastic Surgery, Course of Organ Regulation Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Koichi Tomita
- Department of Plastic Surgery, Course of Organ Regulation Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Plastic and Reconstructive Surgery, Kindai University, Osaka, Japan
| | - Tateki Kubo
- Department of Plastic Surgery, Course of Organ Regulation Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Mari Wataya-Kaneda
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Neurocutaneous Medicine, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Course of Internal Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Manabu Fujimoto
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Rei Watanabe
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Medicine for Cutaneous Immunological Diseases, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
4
|
Yang L, Zhang S, Zheng L, Kong F, Dang W, Shen S, Li X, Jia L, Zhang X, Lu N. Pan-Cancer Analysis of the Prognostic and Immunological Role of SEMA7A. Int J Gen Med 2024; 17:6443-6461. [PMID: 39735166 PMCID: PMC11682669 DOI: 10.2147/ijgm.s499872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 12/20/2024] [Indexed: 12/31/2024] Open
Abstract
Background Semaphorin7A (SEMA7A) has been found to regulate both nerve and vessel homeostasis, but its specific role in pan-cancer remains uncertain. This research seeks to delve into the function and clinical relevance of SEMA7A in pan-cancer. Methods Through an analysis of gene expression omnibus and the cancer genome atlas datasets, we investigated the impact of SEMA7A on prognosis and immune regulation across 33 types of tumors. Variations in SEMA7A expression were observed between cancerous and adjacent normal tissues, with a notable correlation between SEMA7A levels and patient prognosis. Results Across most cancer types, SEMA7A expression was linked to the infiltration of immune cells, as well as immune checkpoints and other immune regulators. The findings were further confirmed through quantitative real-time polymerase chain reaction analysis of SEMA7A expression in breast cancer. Further, SEMA7A is positively associated with prognosis in different cancers. Additionally, SEMA7A expression was associated with TMB and MSI in some cancer types, while in 15 types of cancer, there was a correlation between SEMA7A expression and DNA methylation. SEMA7A was associated with the expression of multiple immune checkpoint genes and abundance of tumor-infiltrating immune cells across multiple types of cancer. Conclusion This inaugural pan-cancer examination of SEMA7A sheds light on its prognostic and immunological significance in diverse tumor types, suggesting its potential utility as a biomarker for predicting unfavorable outcomes and immune cell infiltration in cancer.
Collapse
Affiliation(s)
- Lixian Yang
- Department of Breast Surgery, Xingtai People’s Hospital, Xingtai, Hebei, 054000, People’s Republic of China
| | - Shiyu Zhang
- Department of Breast Surgery, Xingtai People’s Hospital, Xingtai, Hebei, 054000, People’s Republic of China
| | - Lei Zheng
- Department of Breast Surgery, Xingtai People’s Hospital, Xingtai, Hebei, 054000, People’s Republic of China
| | - Fanting Kong
- Department of Breast Surgery, Xingtai People’s Hospital, Xingtai, Hebei, 054000, People’s Republic of China
| | - Wei Dang
- Medical Research Center, Xingtai Medical College, Xingtai, Hebei, 054000, People’s Republic of China
| | - Shipeng Shen
- Medical Research Center, Xingtai Medical College, Xingtai, Hebei, 054000, People’s Republic of China
| | - Xiaowei Li
- Department of Breast Surgery, Xingtai People’s Hospital, Xingtai, Hebei, 054000, People’s Republic of China
| | - Lining Jia
- Department of Breast Surgery, Xingtai People’s Hospital, Xingtai, Hebei, 054000, People’s Republic of China
| | - Xiaoru Zhang
- Department of Breast Surgery, Xingtai People’s Hospital, Xingtai, Hebei, 054000, People’s Republic of China
| | - Na Lu
- Department of Breast Surgery, Xingtai People’s Hospital, Xingtai, Hebei, 054000, People’s Republic of China
| |
Collapse
|
5
|
Suh SB, Suh JY, Lee H, Cho SB. Human dermal fibroblast-derived secretory proteins for regulating nerve restoration: A bioinformatic approach. Skin Res Technol 2024; 30:e13810. [PMID: 38887125 PMCID: PMC11182777 DOI: 10.1111/srt.13810] [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: 05/19/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Human dermal fibroblasts secrete diverse proteins that regulate wound repair and tissue regeneration. METHODS In this study, dermal fibroblast-conditioned medium (DFCM) proteins potentially regulating nerve restoration were bioinformatically selected among the 337 protein lists identified by quantitative liquid chromatography-tandem mass spectrometry. Using these proteins, protein-protein interaction network analysis was conducted. In addition, the roles of DFCM proteins were reviewed according to their protein classifications. RESULTS Gene Ontology protein classification categorized these 57 DFCM proteins into various classes, including protein-binding activity modulator (N = 11), cytoskeletal protein (N = 8), extracellular matrix protein (N = 6), metabolite interconversion enzyme (N = 5), chaperone (N = 4), scaffold/adapter protein (N = 4), calcium-binding protein (N = 3), cell adhesion molecule (N = 2), intercellular signal molecule (N = 2), protein modifying enzyme (N = 2), transfer/carrier protein (N = 2), membrane traffic protein (N = 1), translational protein (N = 1), and unclassified proteins (N = 6). Further protein-protein interaction network analysis of 57 proteins revealed significant interactions among the proteins that varied according to the settings of confidence score. CONCLUSIONS Our bioinformatic analysis demonstrated that DFCM contains many secretory proteins that form significant protein-protein interaction networks crucial for regulating nerve restoration. These findings underscore DFCM proteins' critical roles in various nerve restoration stages during the wound repair process.
Collapse
Affiliation(s)
| | | | | | - Sung Bin Cho
- Yonsei Seran Dermatology and Laser ClinicSeoulSouth Korea
| |
Collapse
|
6
|
Chen T, Li S, Wang L. Semaphorins in tumor microenvironment: Biological mechanisms and therapeutic progress. Int Immunopharmacol 2024; 132:112035. [PMID: 38603857 DOI: 10.1016/j.intimp.2024.112035] [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: 01/08/2024] [Revised: 03/15/2024] [Accepted: 04/05/2024] [Indexed: 04/13/2024]
Abstract
Hallmark features of the tumor microenvironment include immune cells, stromal cells, blood vessels, and extracellular matrix (ECM), providing a conducive environment for the growth and survival of tumors. Recent advances in the understanding of cancer biology have highlighted the functional role of semaphorins (SEMAs). SEMAs are a large and diverse family of widely expressed secreted and membrane-binding proteins, which were initially implicated in axon guidance and neural development. However, it is now clear that they are widely expressed beyond the nervous system and participate in regulating immune responses and cancer progression. In fact, accumulating evidence disclosed that different SEMAs can either stimulate or restrict tumor progression, some of which act as important regulators of tumor angiogenesis. Conversely, limited information is known about the functional relevance of SEMA signals in TME. In this setting, we systematically elaborate the role SEMAs and their major receptors played in characterized components of TME. Furthermore, we provide a convergent view of current SEMAs pharmacological progress in clinical treatment and also put forward their potential application value and clinical prospects in the future.
Collapse
Affiliation(s)
- Tianyi Chen
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022, PR China
| | - Shazhou Li
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022, PR China
| | - Lufang Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022, PR China.
| |
Collapse
|
7
|
El-Husseiny HM, Mady EA, Doghish AS, Zewail MB, Abdelfatah AM, Noshy M, Mohammed OA, El-Dakroury WA. Smart/stimuli-responsive chitosan/gelatin and other polymeric macromolecules natural hydrogels vs. synthetic hydrogels systems for brain tissue engineering: A state-of-the-art review. Int J Biol Macromol 2024; 260:129323. [PMID: 38242393 DOI: 10.1016/j.ijbiomac.2024.129323] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/30/2023] [Accepted: 01/05/2024] [Indexed: 01/21/2024]
Abstract
Currently, there are no viable curative treatments that can enhance the central nervous system's (CNS) recovery from trauma or illness. Bioengineered injectable smart/stimuli-responsive hydrogels (SSRHs) that mirror the intricacy of the CNS milieu and architecture have been suggested as a way to get around these restrictions in combination with medication and cell therapy. Additionally, the right biophysical and pharmacological stimuli are required to boost meaningful CNS regeneration. Recent research has focused heavily on developing SSRHs as cutting-edge delivery systems that can direct the regeneration of brain tissue. In the present article, we have discussed the pathology of brain injuries, and the applicable strategies employed to regenerate the brain tissues. Moreover, the most promising SSRHs for neural tissue engineering (TE) including alginate (Alg.), hyaluronic acid (HA), chitosan (CH), gelatin, and collagen are used in natural polymer-based hydrogels and thoroughly discussed in this review. The ability of these hydrogels to distribute bioactive substances or cells in response to internal and external stimuli is highlighted with particular attention. In addition, this article provides a summary of the most cutting-edge techniques for CNS recovery employing SSRHs for several neurodegenerative diseases.
Collapse
Affiliation(s)
- Hussein M El-Husseiny
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo 183-8509, Japan; Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya 13736, Egypt.
| | - Eman A Mady
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo 183-8509, Japan; Department of Animal Hygiene, Behavior and Management, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya 13736, Egypt.
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Department of Biochemistry, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, Egypt.
| | - Moataz B Zewail
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo, Badr City, Cairo 11829, Egypt
| | - Amr M Abdelfatah
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mina Noshy
- Clinical Pharmacy Department, Faculty of Pharmacy, King Salman International University (KSIU), South Sinai, Ras Sudr 46612, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo, Badr City, Cairo 11829, Egypt
| |
Collapse
|
8
|
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
|
9
|
Naito M, Kumanogoh A. Group 2 innate lymphoid cells and their surrounding environment. Inflamm Regen 2023; 43:21. [PMID: 36941691 PMCID: PMC10026507 DOI: 10.1186/s41232-023-00272-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/14/2023] [Indexed: 03/23/2023] Open
Abstract
Since the discovery of group 2 innate lymphoid cells (ILC2s) in 2010, subsequent studies have revealed their developmental pathways, mechanisms of activation and regulation, and immunological roles in tissue homeostasis and tissue-specific diseases in various organs. Although ILC2s are known to express tissue-specific features depending on where they reside, how the surrounding environment affects the functions of ILC2s remains to be fully elucidated. Recent histologic analyses revealed that ILC2s resides in specific perivascular regions in peripheral tissues with their function being controlled by the surrounding cells via cytokines, lipid mediators, neurotransmitters, and cell-cell interactions through surface molecules. This review summarizes the interactions between ILC2s and surrounding cells, including epithelial cells, neurons, immune cells, and mesenchymal cells, with the objective of promoting the development of novel diagnostic and therapeutic methods for ILC2-related diseases.
Collapse
Affiliation(s)
- Maiko Naito
- 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(WPI), Immunology Frontier Research Center (IFReC), 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(WPI), Immunology Frontier Research Center (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
|
10
|
Chen Z, Fang Y, Jiang W. Important Cells and Factors from Tumor Microenvironment Participated in Perineural Invasion. Cancers (Basel) 2023; 15:1360. [PMID: 36900158 PMCID: PMC10000249 DOI: 10.3390/cancers15051360] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/09/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
Perineural invasion (PNI) as the fourth way for solid tumors metastasis and invasion has attracted a lot of attention, recent research reported a new point that PNI starts to include axon growth and possible nerve "invasion" to tumors as the component. More and more tumor-nerve crosstalk has been explored to explain the internal mechanism for tumor microenvironment (TME) of some types of tumors tends to observe nerve infiltration. As is well known, the interaction of tumor cells, peripheral blood vessels, extracellular matrix, other non-malignant cells, and signal molecules in TME plays a key role in the occurrence, development, and metastasis of cancer, as to the occurrence and development of PNI. We aim to summarize the current theories on the molecular mediators and pathogenesis of PNI, add the latest scientific research progress, and explore the use of single-cell spatial transcriptomics in this invasion way. A better understanding of PNI may help to understand tumor metastasis and recurrence and will be beneficial for improving staging strategies, new treatment methods, and even paradigm shifts in our treatment of patients.
Collapse
Affiliation(s)
- Zirong Chen
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Anatomy Laboratory of Division of Nose and Cranial Base, Clinical Anatomy Center of Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yan Fang
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Anatomy Laboratory of Division of Nose and Cranial Base, Clinical Anatomy Center of Xiangya Hospital, Central South University, Changsha 410008, China
| | - Weihong Jiang
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Anatomy Laboratory of Division of Nose and Cranial Base, Clinical Anatomy Center of Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
11
|
Liu D, Li J, Qi F, You H. Semaphorins and their receptors in pancreatic cancer: Mechanisms and therapeutic opportunities. Front Oncol 2023; 12:1106762. [PMID: 36713527 PMCID: PMC9880980 DOI: 10.3389/fonc.2022.1106762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 12/23/2022] [Indexed: 01/15/2023] Open
Abstract
Pancreatic cancer (PC) is a malignant tumor with high malignancy that is difficult to diagnose and treat. PC is a major medical problem because of its low early diagnosis rate, high surgical mortality rate, low cure rate, and expensive related testing cost. Therefore, the significance of finding new markers for PC is self-evident. Semaphorins (Semas) have been shown to affect angiogenesis and lymphangiogenesis and can also directly affect the behavior of tumor cells. The expression and related action targets of its family members on PC are summarized in this review.
Collapse
Affiliation(s)
- Dahai Liu
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Jie Li
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Fei Qi
- Department of Pulmonary and Critical Care Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Hua You
- Department of Pediatric Hematology and Oncology, Children’s Hospital of Chongqing Medical University, Chongqing, China,Laboratory for Excellence in Systems Biomedicine of Pediatric Oncology, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China,*Correspondence: Hua You,
| |
Collapse
|
12
|
Naito M, Nakanishi Y, Motomura Y, Takamatsu H, Koyama S, Nishide M, Naito Y, Izumi M, Mizuno Y, Yamaguchi Y, Nojima S, Okuzaki D, Kumanogoh A. Semaphorin 6D-expressing mesenchymal cells regulate IL-10 production by ILC2s in the lung. Life Sci Alliance 2022; 5:5/11/e202201486. [PMID: 36038260 PMCID: PMC9434704 DOI: 10.26508/lsa.202201486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 08/18/2022] [Accepted: 08/18/2022] [Indexed: 11/24/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) have features specific to the niches in which they reside, and we found that semaphorin 6D signaling in the lung niche controls IL-10 production by ILC2s. Group 2 innate lymphoid cells (ILC2s) have been implicated in both physiologic tissue remodeling and allergic pathology, yet the niche signaling required for ILC2 properties is poorly understood. Here, we show that an axonal guidance cue semaphorin 6D (Sema6D) plays critical roles in the maintenance of IL-10–producing ILC2s. Sema6d−/− mice exhibit a severe steady-state reduction in ILC2s in peripheral sites such as the lung, visceral adipose tissue, and mesentery. Interestingly, loss of Sema6D results in suppressed alarmin-driven type 2 cytokine production but increased IL-10 production by lung ILC2s both in vitro and in vivo. Consequently, Sema6d−/− mice are resistant to the development of allergic lung inflammation. We further found that lung mesenchymal cells highly express Sema6D, and that niche-derived Sema6D is responsible for these phenotypes through plexin A1. Collectively, these findings suggest that niche-derived Sema6D is implicated in physiological and pathological characteristics of ILC2s.
Collapse
Affiliation(s)
- Maiko Naito
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan.,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan
| | - Yoshimitsu Nakanishi
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan.,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Japan
| | - Yasutaka Motomura
- Laboratory for Innate Immune Systems, Department for Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Japan.,Laboratory for Innate Immune Systems, WPI, Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan.,Laboratory for Innate Immune Systems, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Hyota Takamatsu
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan.,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan
| | - Shohei Koyama
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan.,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan.,Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, Chiba, Japan
| | - Masayuki Nishide
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan.,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan
| | - Yujiro Naito
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan.,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan
| | - Mayuko Izumi
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan.,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan
| | - Yumiko Mizuno
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan.,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan
| | - Yuta Yamaguchi
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan.,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan
| | - Satoshi Nojima
- Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan.,Department of Pathology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Daisuke Okuzaki
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Japan.,Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Japan.,Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan .,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Japan.,Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Japan
| |
Collapse
|
13
|
Sun T, Zeng H, Fan L, Fei J, Chen G. Semaphorin 6D regulate corralling, hematoma compaction and white matter injury in mice after intracerebral hemorrhage. J Stroke Cerebrovasc Dis 2022; 31:106803. [PMID: 36174325 DOI: 10.1016/j.jstrokecerebrovasdis.2022.106803] [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: 07/27/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES The Semaphorin 6D (SEMA6D) shows important roles in cell guidance and lipid metabolism, but the effects and mechanisms of SEMA6D on tissue repair, white matter injury and the recovery of neurological function after intracerebral hemorrhage have not been well studied. MATERIALS AND METHODS In this study, the autologous whole blood injection model of intracerebral hemorrhage was established in C57 male mice. SEMA6D knockout CRISPR utilized in the study. Assessments included neurological score evaluation and immunofluorescence. RESULTS SEMA6D increased and peaked at 7d after intracerebral hemorrhage, and mainly located in neurons, microglia and astrocytes. SEMA6D knockout CRISPR aggravated neurological function and showed signs of poorer corralling and hematoma resolution, with more compartments of well-established physical barrier and more extensive GFAP positive astrocytic border. Furthermore, SEMA6D can prevent the decrease of NF-H in the peri-hematoma region, while SEMA6D knockout aggravated WMI. CONCLUSIONS Our study suggested that SEMA6D could influence the recovery of neurological function by regulating the corralling, hematoma compaction and WMI in mice after intracerebral hemorrhage.
Collapse
Affiliation(s)
- Ting Sun
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, Zhejiang Province 310009, China.
| | - Hanhai Zeng
- Department of Neurological Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, Zhejiang Province 310009, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China..
| | - Linfeng Fan
- Department of Neurological Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, Zhejiang Province 310009, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China..
| | - Jing Fei
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, Zhejiang Province 310009, China.
| | - Gao Chen
- Department of Neurological Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, Zhejiang Province 310009, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China..
| |
Collapse
|
14
|
Nojima S. Class IV semaphorins in disease pathogenesis. Pathol Int 2022; 72:471-487. [PMID: 36066011 DOI: 10.1111/pin.13270] [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: 05/16/2022] [Accepted: 08/16/2022] [Indexed: 12/01/2022]
Abstract
Semaphorins are a large family of secreted and/or transmembrane proteins, originally identified as proteins that function in axon guidance during neuronal development. However, semaphorins play crucial roles in other physiological and pathological processes, including immune responses, angiogenesis, maintenance of tissue homeostasis, and cancer progression. Class IV semaphorins may be present as transmembrane and soluble forms and are implicated in the pathogenesis of various diseases. This review discusses recent progress on the roles of class IV semaphorins determined by clinical and experimental pathology studies.
Collapse
Affiliation(s)
- Satoshi Nojima
- Department of Pathology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, Japan
| |
Collapse
|
15
|
Sezgin G, Kar F, Uslu S. The effect of nutrition and exercise training on irisin and semaphorin-3E levels in obese patients. Arch Physiol Biochem 2022; 128:558-567. [PMID: 32569480 DOI: 10.1080/13813455.2020.1779310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
We aimed to firstly examine the effects of nutrition and exercise training on irisin, Sema-3E, biochemical and inflammatory parameters in obese patients. This study was conducted using 37 individuals were divided into three groups according to body mass index (BMI) as non-obese, 1nd degree and 2nd degree obese individuals. Nutrition and exercise training were applied to groups for eight weeks. Insulin resistance decreased in non-obese and 1st degree obese subjects. HsCRP values decreased only in the second degree obese individuals. Adiponectin values were significantly decreased in all three groups. There was a negative correlation between serum adiponectin and plasma irisin levels both before and after treatment. Sema-3E levels increased significantly in only the first degree obese individuals, whereas plexin-D1 values did not change significantly in any group. Our findings indicate that nutrition and exercise training we apply improved both anthropometric measurements and biochemical parameters in obese and non-obese individuals.
Collapse
Affiliation(s)
- Gülay Sezgin
- Department of Nutrition and Diet, Istanbul Taksim Training and Research Hospital, İstanbul, Turkey
| | - Fatih Kar
- Department of Medical Biochemistry, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Sema Uslu
- Department of Medical Biochemistry, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| |
Collapse
|
16
|
Morton AB, Jacobsen NL, Segal SS. Functionalizing biomaterials to promote neurovascular regeneration following skeletal muscle injury. Am J Physiol Cell Physiol 2021; 320:C1099-C1111. [PMID: 33852364 PMCID: PMC8285637 DOI: 10.1152/ajpcell.00501.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 03/29/2021] [Accepted: 04/06/2021] [Indexed: 12/18/2022]
Abstract
During embryogenesis, blood vessels and nerves develop with similar branching structure in response to shared signaling pathways guiding network growth. With both systems integral to physiological homeostasis, dual targeting of blood vessels and nerves to promote neurovascular regeneration following injury is an emerging therapeutic approach in biomedical engineering. A limitation to this strategy is that the nature of cross talk between emergent vessels and nerves during regeneration in an adult is poorly understood. Following peripheral nerve transection, intraneural vascular cells infiltrate the site of injury to provide a migratory pathway for mobilized Schwann cells of regenerating axons. As Schwann cells demyelinate, they secrete vascular endothelial growth factor, which promotes angiogenesis. Recent advances point to concomitant restoration of neurovascular architecture and function through simultaneous targeting of growth factors and guidance cues shared by both systems during regeneration. In the context of traumatic injury associated with volumetric muscle loss, we consider the nature of biomaterials used to engineer three-dimensional scaffolds, functionalization of scaffolds with molecular signals that guide and promote neurovascular growth, and seeding scaffolds with progenitor cells. Physiological success is defined by each tissue component of the bioconstruct (nerve, vessel, muscle) becoming integrated with that of the host. Advances in microfabrication, cell culture techniques, and progenitor cell biology hold great promise for engineering bioconstructs able to restore organ function after volumetric muscle loss.
Collapse
Affiliation(s)
- Aaron B Morton
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Nicole L Jacobsen
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Steven S Segal
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, Columbia, Missouri
| |
Collapse
|
17
|
Song Y, Wang L, Zhang L, Huang D. The involvement of semaphorin 7A in tumorigenic and immunoinflammatory regulation. J Cell Physiol 2021; 236:6235-6248. [PMID: 33611799 DOI: 10.1002/jcp.30340] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/12/2021] [Accepted: 02/05/2021] [Indexed: 02/04/2023]
Abstract
Semaphorins, a large group of highly conserved proteins, consist of eight subfamilies that are widely expressed in vertebrates, invertebrates, and viruses and exist in membrane-bound or secreted forms. First described as axon guidance cues during neurogenesis, semaphorins also perform physiological functions in other organ systems, such as bone homeostasis, immune response, and tumor progression. Semaphorin 7A (SEMA7A), also known as CDw108, is an immune semaphorin that modulates diverse immunoinflammatory processes, including immune cell interactions, inflammatory infiltration, and cytokine production. In addition, SEMA7A regulates the proliferation, migration, invasion, lymph formation, and angiogenesis of multiple types of tumor cells, and these effects are mediated by the interaction of SEMA7A with two specific receptors, PLXNC1 and integrins. Thus, SEMA7A is intimately related to the pathogenesis of multiple autoimmune and inflammation-related diseases and tumors. This review focuses on the role of SEMA7A in the pathogenesis of autoimmune disorders, inflammatory diseases, and tumors, as well as the underlying mechanisms. Furthermore, strategies targeting SEMA7A as a potential predictive, diagnostic, and therapeutic agent for these diseases are also addressed.
Collapse
Affiliation(s)
- Yao Song
- State Key Laboratory of Oral Diseases and National Clinical Research Center of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liu Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lan Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Dingming Huang
- State Key Laboratory of Oral Diseases and National Clinical Research Center of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
18
|
Zang Y, Chaudhari K, Bashaw GJ. New insights into the molecular mechanisms of axon guidance receptor regulation and signaling. Curr Top Dev Biol 2021; 142:147-196. [PMID: 33706917 DOI: 10.1016/bs.ctdb.2020.11.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
As the nervous system develops, newly differentiated neurons need to extend their axons toward their synaptic targets to form functional neural circuits. During this highly dynamic process of axon pathfinding, guidance receptors expressed at the tips of motile axons interact with soluble guidance cues or membrane tethered molecules present in the environment to be either attracted toward or repelled away from the source of these cues. As competing cues are often present at the same location and during the same developmental period, guidance receptors need to be both spatially and temporally regulated in order for the navigating axons to make appropriate guidance decisions. This regulation is exerted by a diverse array of molecular mechanisms that have come into focus over the past several decades and these mechanisms ensure that the correct complement of surface receptors is present on the growth cone, a fan-shaped expansion at the tip of the axon. This dynamic, highly motile structure is defined by a lamellipodial network lining the periphery of the growth cone interspersed with finger-like filopodial projections that serve to explore the surrounding environment. Once axon guidance receptors are deployed at the right place and time at the growth cone surface, they respond to their respective ligands by initiating a complex set of signaling events that serve to rearrange the growth cone membrane and the actin and microtubule cytoskeleton to affect axon growth and guidance. In this review, we highlight recent advances that shed light on the rich complexity of mechanisms that regulate axon guidance receptor distribution, activation and downstream signaling.
Collapse
Affiliation(s)
- Yixin Zang
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Karina Chaudhari
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Greg J Bashaw
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
19
|
Hong L, Li F, Tang C, Li L, Sun L, Li X, Zhu L. Semaphorin 7A promotes endothelial to mesenchymal transition through ATF3 mediated TGF-β2/Smad signaling. Cell Death Dis 2020; 11:695. [PMID: 32826874 PMCID: PMC7442651 DOI: 10.1038/s41419-020-02818-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 07/06/2020] [Accepted: 07/09/2020] [Indexed: 12/14/2022]
Abstract
Endothelial to mesenchymal transition (EndMT) is an important pathological change in many diseases. Semaphorin7A (Sema7A) has been reported to regulate nerve and vessel homeostasis, but its role in EndMT remains unclear. Here we investigate the effect of Sema7A on EndMT and the underlying mechanism. Sema7A-overexpressed human umbilical vein endothelial cells (Sema7A-HUVECs) were generated and showed lower levels of endothelial cell markers and higher levels of mesenchymal cell markers indicating the occurrence of EndMT. RNA-sequencing analysis showed a total of 1168 upregulated genes and 886 downregulated genes. Among them, most of the molecules associated with EndMT were upregulated in Sema7A-HUVECs. Mechanistically, Sema7A-HUVECs showed a higher TGF-β2 expression and activated TGF-β/Smad Signaling. Importantly, Sema7A overexpression upregulated activating transcription factor 3 (ATF3) that was found to selectively bind the promotor region of TGF-β2, but not TGF-β1, promoting TGF-β2 transcription, which was further confirmed by ATF3-siRNA knockdown approach. Blocking β1 integrin, a known Sema7A receptor, alleviated the expression of ATF3, TGF-β2, and EndMT in Sema7A-overexpressed HUVECs, implying a role of β1 integrin/ATF3/TGF-β2 axis in mediating Sema7A-induced EndMT. Using Sema7A-deficient mice and the partial carotid artery ligation (PCL) model, we showed that Sema7A deletion attenuated EndMT induced by blood flow disturbance in vivo. In conclusion, Sema7A promotes TGF-β2 secretion by upregulating transcription factor ATF3 in a β1 integrin-dependent manner, and thus facilitates EndMT through TGF/Smad signaling, implying Sema7A as a potential therapeutic target for EndMT-related vascular diseases.
Collapse
Affiliation(s)
- Lei Hong
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Suzhou Key Laboratory of Thrombosis and Vascular Biology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, Jiangsu, China
- Department of Vascular Surgery, Anhui Provincial Hospital, University of Science and Technology of China, Hefei, Anhui, China
| | - Fengchan Li
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Suzhou Key Laboratory of Thrombosis and Vascular Biology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, Jiangsu, China
| | - Chaojun Tang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Suzhou Key Laboratory of Thrombosis and Vascular Biology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, Jiangsu, China
| | - Ling Li
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Suzhou Key Laboratory of Thrombosis and Vascular Biology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, Jiangsu, China
| | - Lili Sun
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Xiaoqiang Li
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China.
| | - Li Zhu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Suzhou Key Laboratory of Thrombosis and Vascular Biology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
20
|
Mu X, Li W, Ze X, Li L, Wang G, Hong F, Ze Y. Molecular mechanism of nanoparticulate TiO 2 induction of axonal development inhibition in rat primary cultured hippocampal neurons. ENVIRONMENTAL TOXICOLOGY 2020; 35:895-905. [PMID: 32329576 DOI: 10.1002/tox.22926] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 03/16/2020] [Accepted: 03/20/2020] [Indexed: 06/11/2023]
Abstract
Numerous studies have demonstrated the in vitro and in vivo neurotoxicity of nanoparticulate titanium dioxide (nano-TiO2 ), a mass-produced material for a large number of commercial and industrial applications. The mechanism of nano-TiO2 -induced inhibition of axonal development, however, is still unclear. In our study, primary cultured hippocampal neurons of 24-hour-old fetal Sprague-Dawley rats were exposed to 5, 15, or 30 μg/mL nano-TiO2 for 6, 12, and 24 hours, and the toxic effects of nano-TiO2 exposure on the axons development were detected and its molecular mechanism investigated. Nano-TiO2 accumulated in hippocampal neurons and inhibited the development of axons as nano-TiO2 concentrations increased. Increasing time in culture resulted in decreasing axon length by 32.5%, 36.6%, and 53.8% at 6 hours, by 49.4%, 53.8%, and 69.5% at 12 hours, and by 44.5%, 58.2%, and 63.6% at 24 hours, for 5, 15, and 30 μg/mL nano-TiO2 , respectively. Furthermore, nano-TiO2 downregulated expression of Netrin-1, growth-associated protein-43, and Neuropilin-1, and promoted an increase of semaphorin type 3A and Nogo-A. These studies suggest that nano-TiO2 inhibited axonal development in rat primary cultured hippocampal neurons and this phenomenon is related to changes in the expression of axon growth-related factors.
Collapse
Affiliation(s)
- Xu Mu
- Department of Biochemistry and Molecular Biology, School of Basic Medical and Biological Sciences, Soochow University, Suzhou, China
| | - Wuyan Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical and Biological Sciences, Soochow University, Suzhou, China
| | - Xiao Ze
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Lingjuan Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical and Biological Sciences, Soochow University, Suzhou, China
| | - Guoqing Wang
- Department of Physiology and Neurobiology, School of Basic Medical and Biological Sciences, Soochow University, Suzhou, China
| | - Fashui Hong
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture and Environmental Protection, Huaiyin Normal University, Huaian, China
- Department of Biotechnology, School of Life Sciences, Huaiyin Normal University, Huaian, China
| | - Yuguan Ze
- Department of Biochemistry and Molecular Biology, School of Basic Medical and Biological Sciences, Soochow University, Suzhou, China
| |
Collapse
|
21
|
Jeon KI, Nehrke K, Huxlin KR. Semaphorin 3A potentiates the profibrotic effects of transforming growth factor-β1 in the cornea. Biochem Biophys Res Commun 2020; 521:333-339. [PMID: 31668808 DOI: 10.1016/j.bbrc.2019.10.107] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 10/12/2019] [Indexed: 11/16/2022]
Abstract
Corneal scarring is a major cause of blindness worldwide with few effective therapeutic options. Finding a treatment would be of tremendous public health benefit, but requires a thorough understanding of the complex interactions that underlie this phenomenon. Here, we tested the hypothesis that the large increase in expression of Semaphorin 3A (SEMA3A) in corneal wounds contributes to the development of stromal fibrosis. We first verified this increased expression in vivo, in a cat model of photorefractive keratectomy-induced corneal wounding. We then examined the impact of adding exogenous SEMA3A to cultured corneal fibroblasts, and assessed how this affected the ability of transforming growth factor-beta1 (TGF-β1) to induce their differentiation into myofibroblasts. Finally, we examined how siRNA knockdown of endogenous SEMA3A affected these same phenomena. We found exogenous SEMA3A to significantly potentiate TGF-β1's profibrotic effects, with only a minimal contribution from cell-intrinsic SEMA3A. Our results suggest a previously unrecognized interaction between SEMA3A and TGF-β1 in the wounded cornea, and a possible contribution of SEMA3A to the regulation of tissue fibrosis and remodeling in this transparent organ.
Collapse
Affiliation(s)
| | - Keith Nehrke
- Department of Medicine, University of Rochester, Rochester, NY, USA
| | | |
Collapse
|
22
|
Ng SY, Lee AYW. Traumatic Brain Injuries: Pathophysiology and Potential Therapeutic Targets. Front Cell Neurosci 2019; 13:528. [PMID: 31827423 PMCID: PMC6890857 DOI: 10.3389/fncel.2019.00528] [Citation(s) in RCA: 449] [Impact Index Per Article: 74.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023] Open
Abstract
Traumatic brain injury (TBI) remains one of the leading causes of morbidity and mortality amongst civilians and military personnel globally. Despite advances in our knowledge of the complex pathophysiology of TBI, the underlying mechanisms are yet to be fully elucidated. While initial brain insult involves acute and irreversible primary damage to the parenchyma, the ensuing secondary brain injuries often progress slowly over months to years, hence providing a window for therapeutic interventions. To date, hallmark events during delayed secondary CNS damage include Wallerian degeneration of axons, mitochondrial dysfunction, excitotoxicity, oxidative stress and apoptotic cell death of neurons and glia. Extensive research has been directed to the identification of druggable targets associated with these processes. Furthermore, tremendous effort has been put forth to improve the bioavailability of therapeutics to CNS by devising strategies for efficient, specific and controlled delivery of bioactive agents to cellular targets. Here, we give an overview of the pathophysiology of TBI and the underlying molecular mechanisms, followed by an update on novel therapeutic targets and agents. Recent development of various approaches of drug delivery to the CNS is also discussed.
Collapse
Affiliation(s)
- Si Yun Ng
- Neurobiology/Ageing Program, Centre for Life Sciences, Department of Physiology, Yong Loo Lin School of Medicine, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Alan Yiu Wah Lee
- Neurobiology/Ageing Program, Centre for Life Sciences, Department of Physiology, Yong Loo Lin School of Medicine, Life Sciences Institute, National University of Singapore, Singapore, Singapore.,School of Pharmacy, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
23
|
Hossain MM, Tsuzuki T, Sakakibara K, Imaizumi F, Ikegaya A, Inagaki M, Takahashi I, Ito T, Takamatsu H, Kumanogoh A, Negishi T, Yukawa K. PlexinA1 is crucial for the midline crossing of callosal axons during corpus callosum development in BALB/cAJ mice. PLoS One 2019; 14:e0221440. [PMID: 31430342 PMCID: PMC6701775 DOI: 10.1371/journal.pone.0221440] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/06/2019] [Indexed: 12/04/2022] Open
Abstract
The corpus callosum (CC) is the biggest commissure that links cerebral hemispheres. Guidepost structures develop in the cortical midline during CC development and express axon guidance molecules that instruct neurons regarding the proper direction of axonal elongation toward and across the cortical midline. Neuropilin-1 (Npn1), a high affinity receptor for class 3 semaphorins (Sema3s) localized on cingulate pioneering axons, plays a crucial role in axon guidance to the midline through interactions with Sema3s. However, it remains unclear which type of Plexin is a component of Sema3 holoreceptors with Npn1 during the guidance of cingulate pioneering axons. To address the role of PlexinA1 in CC development, we examined with immunohistochemistry the localization of PlexinA1, Npn1, and Sema3s using embryonic brains from wild-type (WT) and PlexinA1-deficient (PlexinA1 knock-out (KO)) mice with a BALB/cAJ background. The immunohistochemistry confirmed the expression of PlexinA1 in callosal axons derived from the cingulate and neocortex of the WT mice on embryonic day 17.5 (E17.5) but not in the PlexinA1 KO mice. To examine the role of PlexinA1 in the navigation of callosal axons, the extension of callosal axons toward and across the midline was traced in brains of WT and PlexinA1 KO mice at E17.5. As a result, callosal axons in the PlexinA1 KO brains had a significantly lower incidence of midline crossing at E17.5 compared with the WT brains. To further examine the role of PlexinA1 in CC development, the CC phenotype was examined in PlexinA1 KO mice at postnatal day 0.5 (P0.5). Most of the PlexinA1 KO mice at P0.5 showed agenesis of the CC. These results indicate the crucial involvement of PlexinA1 in the midline crossing of callosal axons during CC development in BALB/cAJ mice.
Collapse
Affiliation(s)
| | - Takamasa Tsuzuki
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Kazuki Sakakibara
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Fumitaka Imaizumi
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Akihiro Ikegaya
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Mami Inagaki
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Ikuko Takahashi
- Radioisotope Center, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Takuji Ito
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Hyota Takamatsu
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Atsushi Kumanogoh
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Takayuki Negishi
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Kazunori Yukawa
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
- * E-mail:
| |
Collapse
|
24
|
Körner S, Thau-Habermann N, Kefalakes E, Bursch F, Petri S. Expression of the axon-guidance protein receptor Neuropilin 1 is increased in the spinal cord and decreased in muscle of a mouse model of amyotrophic lateral sclerosis. Eur J Neurosci 2019; 49:1529-1543. [PMID: 30589468 DOI: 10.1111/ejn.14326] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 12/05/2018] [Accepted: 12/13/2018] [Indexed: 12/11/2022]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a degenerative motor neuron disorder. It is supposed that ALS is at least in part an axonopathy. Neuropilin 1 is an important receptor of the axon repellent Semaphorin 3A and a co-receptor of vascular endothelial growth factor. It is probably involved in neuronal and axonal de-/regeneration and might be of high relevance for ALS pathogenesis and/or disease progression. To elucidate whether the expression of either Neuropilin1 or Semaphorin3A is altered in ALS we investigated these proteins in human brain, spinal cord and muscle tissue of ALS-patients and controls as well as transgenic SOD1G93A and control mice. Neuropilin1 and Semaphorin3A gene and protein expression were assessed by quantitative real-time PCR (qRT-PCR), western blot and immunohistochemistry. Groups were compared using either Student t-test or Mann-Whitney U test. We observed a consistent increase of Neuropilin1 expression in the spinal cord and decrease of Neuropilin1 and Semaphorin3A in muscle tissue of transgenic SOD1G93A mice at the mRNA and protein level. Previous studies have shown that damage of neurons physiologically causes Neuropilin1 and Semaphorin3A increase in the central nervous system and decrease in the peripheral nervous system. Our results indicate that this also occurs in ALS. Pharmacological modulation of expression and function of axon repellents could be a promising future therapeutic option in ALS.
Collapse
Affiliation(s)
- Sonja Körner
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | | | - Ekaterini Kefalakes
- Department of Neurology, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Franziska Bursch
- Department of Neurology, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Susanne Petri
- Department of Neurology, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience (ZSN), Hannover, Germany
| |
Collapse
|
25
|
Sar Shalom H, Goldner R, Golan-Vaishenker Y, Yaron A. Balance between BDNF and Semaphorins gates the innervation of the mammary gland. eLife 2019; 8:41162. [PMID: 30628891 PMCID: PMC6328272 DOI: 10.7554/elife.41162] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 12/07/2018] [Indexed: 01/01/2023] Open
Abstract
The innervation of the mammary gland is controlled by brain-derived neurotrophic factor (BDNF), and sexually dimorphic sequestering of BDNF by the truncated form of TrkB (TrkB.T1) directs male-specific axonal pruning in mice. It is unknown whether other cues modulate these processes. We detected specific, non-dimorphic, expression of Semaphorin family members in the mouse mammary gland, which signal through PlexinA4. PlexinA4 deletion in both female and male embryos caused developmental hyperinnervation of the gland, which could be reduced by genetic co-reduction of BDNF. Moreover, in males, PlexinA4 ablation delayed axonal pruning, independently of the initial levels of innervation. In support of this, in vitro reduction of BDNF induced axonal hypersensitivity to PlexinA4 signaling. Overall, our study shows that precise sensory innervation of the mammary gland is regulated by the balance between trophic and repulsive signaling. Upon inhibition of trophic signaling, these repulsive factors may promote axonal pruning.
Collapse
Affiliation(s)
- Hadas Sar Shalom
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Ron Goldner
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | | | - Avraham Yaron
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
26
|
Lotfi R, Yari K. The Role of Semaphorins and their Receptors in the Immune System and their Relation to Multiple Sclerosis. ACTA ACUST UNITED AC 2018. [DOI: 10.29252/shefa.6.4.75] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
27
|
Semaphorin 3A as an inhibitive factor for migration of olfactory ensheathing cells through cofilin activation is involved in formation of olfactory nerve layer. Mol Cell Neurosci 2018; 92:27-39. [PMID: 29940213 DOI: 10.1016/j.mcn.2018.06.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 05/23/2018] [Accepted: 06/19/2018] [Indexed: 01/27/2023] Open
Abstract
Olfactory ensheathing cells (OECs) migrate from olfactory epithelium towards olfactory bulb (OB), contributing to formation of the presumptive olfactory nerve layer during development. However, it remains unclear that molecular mechanism of regulation of OEC migration in OB. In the present study, we found that OECs highly expressed the receptors of semaphorin 3A (Sema3A) in vitro and in vivo, whereas Sema3A displayed a gradient expression pattern with higher in inner layer of OB and lower in outer layer of OB. Furthermore, the collapse assays, Boyden chamber migration assays and single-cell migration assays showed that Sema3A induced the collapse of leading front of OECs and inhibited OEC migration. Thirdly, the leading front of OECs exhibited adaptation in a protein synthesis-independent manner, and endocytosis-dependent manner during Sema3A-induced OEC migration. Finally, Sema3A-induced collapse of leading front was required the decrease of focal adhesion and a retrograde F-actin flow in a cofilin activation-dependent manner. Taken together, these results demonstrate that Sema3A as an inhibitive migratory factor for OEC migration through cofilin activation is involved in the formation of olfactory nerve layer.
Collapse
|
28
|
Kang S, Nakanishi Y, Kioi Y, Okuzaki D, Kimura T, Takamatsu H, Koyama S, Nojima S, Nishide M, Hayama Y, Kinehara Y, Kato Y, Nakatani T, Shimogori T, Takagi J, Toyofuku T, Kumanogoh A. Semaphorin 6D reverse signaling controls macrophage lipid metabolism and anti-inflammatory polarization. Nat Immunol 2018; 19:561-570. [PMID: 29777213 DOI: 10.1038/s41590-018-0108-0] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 04/04/2018] [Indexed: 12/21/2022]
Abstract
Polarization of macrophages into pro-inflammatory or anti-inflammatory states has distinct metabolic requirements, with mechanistic target of rapamycin (mTOR) kinase signaling playing a critical role. However, it remains unclear how mTOR regulates metabolic status to promote polarization of these cells. Here we show that an mTOR-Semaphorin 6D (Sema6D)-Peroxisome proliferator receptor γ (PPARγ) axis plays critical roles in macrophage polarization. Inhibition of mTOR or loss of Sema6D blocked anti-inflammatory macrophage polarization, concomitant with severe impairments in PPARγ expression, uptake of fatty acids, and lipid metabolic reprogramming. Macrophage expression of the receptor Plexin-A4 is responsible for Sema6D-mediated anti-inflammatory polarization. We found that a tyrosine kinase, c-Abl, which associates with the cytoplasmic region of Sema6D, is required for PPARγ expression. Furthermore, Sema6D is important for generation of intestinal resident CX3CR1hi macrophages and prevents development of colitis. Collectively, these findings highlight crucial roles for Sema6D reverse signaling in macrophage polarization, coupling immunity, and metabolism via PPARγ.
Collapse
Affiliation(s)
- Sujin Kang
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan. .,Department of Respiratory Medicine and Clinical Immunology, Graduate School of medicine, Osaka University, Suita City, Osaka, Japan. .,Department of Immune Regulation, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan.
| | - Yoshimitsu Nakanishi
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Graduate School of medicine, Osaka University, Suita City, Osaka, Japan
| | - Yoshiyuki Kioi
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Graduate School of medicine, Osaka University, Suita City, Osaka, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita City, Osaka, Japan
| | - Tetsuya Kimura
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan
| | - Hyota Takamatsu
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Graduate School of medicine, Osaka University, Suita City, Osaka, Japan
| | - Shohei Koyama
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Graduate School of medicine, Osaka University, Suita City, Osaka, Japan
| | - Satoshi Nojima
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan.,Department of Pathology, Graduate School of Medicine, Osaka University, Suita City, Osaka, Japan
| | - Masayuki Nishide
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Graduate School of medicine, Osaka University, Suita City, Osaka, Japan
| | - Yoshitomo Hayama
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Graduate School of medicine, Osaka University, Suita City, Osaka, Japan
| | - Yuhei Kinehara
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Graduate School of medicine, Osaka University, Suita City, Osaka, Japan
| | - Yasuhiro Kato
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Graduate School of medicine, Osaka University, Suita City, Osaka, Japan
| | - Takeshi Nakatani
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Graduate School of medicine, Osaka University, Suita City, Osaka, Japan
| | - Tomomi Shimogori
- RIKEN Brain Science Institute, Laboratory for Molecular Mechanisms of Thalamus Development, Saitama, Japan
| | - Junichi Takagi
- Laboratory of Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Suita City, Osaka, Japan
| | - Toshihiko Toyofuku
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan.,Department of Immunology and Regenerative Medicine, Graduate School of Medicine, Osaka University, Suita City, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan. .,Department of Respiratory Medicine and Clinical Immunology, Graduate School of medicine, Osaka University, Suita City, Osaka, Japan.
| |
Collapse
|
29
|
Chabrat A, Brisson G, Doucet-Beaupré H, Salesse C, Schaan Profes M, Dovonou A, Akitegetse C, Charest J, Lemstra S, Côté D, Pasterkamp RJ, Abrudan MI, Metzakopian E, Ang SL, Lévesque M. Transcriptional repression of Plxnc1 by Lmx1a and Lmx1b directs topographic dopaminergic circuit formation. Nat Commun 2017; 8:933. [PMID: 29038581 PMCID: PMC5643336 DOI: 10.1038/s41467-017-01042-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 08/15/2017] [Indexed: 12/27/2022] Open
Abstract
Mesodiencephalic dopamine neurons play central roles in the regulation of a wide range of brain functions, including voluntary movement and behavioral processes. These functions are served by distinct subtypes of mesodiencephalic dopamine neurons located in the substantia nigra pars compacta and the ventral tegmental area, which form the nigrostriatal, mesolimbic, and mesocortical pathways. Until now, mechanisms involved in dopaminergic circuit formation remained largely unknown. Here, we show that Lmx1a, Lmx1b, and Otx2 transcription factors control subtype-specific mesodiencephalic dopamine neurons and their appropriate axon innervation. Our results revealed that the expression of Plxnc1, an axon guidance receptor, is repressed by Lmx1a/b and enhanced by Otx2. We also found that Sema7a/Plxnc1 interactions are responsible for the segregation of nigrostriatal and mesolimbic dopaminergic pathways. These findings identify Lmx1a/b, Otx2, and Plxnc1 as determinants of dopaminergic circuit formation and should assist in engineering mesodiencephalic dopamine neurons capable of regenerating appropriate connections for cell therapy.Midbrain dopaminergic neurons (mDAs) in the VTA and SNpc project to different regions and form distinct circuits. Here the authors show that transcription factors Lmx1a, Lmx1b, and Otx2 control the axon guidance of mDAs and the segregation of mesolimbic and nigrostriatal dopaminergic pathways.
Collapse
Affiliation(s)
- Audrey Chabrat
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, Quebec, G1V 0A6, Canada
- CERVO Brain Research Centre, 2601, chemin de la Canardière, Québec, Quebec, Canada, G1J 2G3
| | - Guillaume Brisson
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, Quebec, G1V 0A6, Canada
- CERVO Brain Research Centre, 2601, chemin de la Canardière, Québec, Quebec, Canada, G1J 2G3
| | - Hélène Doucet-Beaupré
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, Quebec, G1V 0A6, Canada
- CERVO Brain Research Centre, 2601, chemin de la Canardière, Québec, Quebec, Canada, G1J 2G3
| | - Charleen Salesse
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, Quebec, G1V 0A6, Canada
- CERVO Brain Research Centre, 2601, chemin de la Canardière, Québec, Quebec, Canada, G1J 2G3
| | - Marcos Schaan Profes
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, Quebec, G1V 0A6, Canada
- CERVO Brain Research Centre, 2601, chemin de la Canardière, Québec, Quebec, Canada, G1J 2G3
| | - Axelle Dovonou
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, Quebec, G1V 0A6, Canada
- CERVO Brain Research Centre, 2601, chemin de la Canardière, Québec, Quebec, Canada, G1J 2G3
| | - Cléophace Akitegetse
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, Quebec, G1V 0A6, Canada
- CERVO Brain Research Centre, 2601, chemin de la Canardière, Québec, Quebec, Canada, G1J 2G3
| | - Julien Charest
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, Quebec, G1V 0A6, Canada
- CERVO Brain Research Centre, 2601, chemin de la Canardière, Québec, Quebec, Canada, G1J 2G3
| | - Suzanne Lemstra
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG, Utrecht, The Netherlands
| | - Daniel Côté
- CERVO Brain Research Centre, 2601, chemin de la Canardière, Québec, Quebec, Canada, G1J 2G3
- Département de Physique, Genie Physique et Optique, Université Laval, Québec, Quebec, G1V 0A6, Canada
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG, Utrecht, The Netherlands
| | - Monica I Abrudan
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
- Faculty of Medicine, School of Public Health, Imperial College, London, W2 1PG, UK
| | - Emmanouil Metzakopian
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Siew-Lan Ang
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Martin Lévesque
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, Quebec, G1V 0A6, Canada.
- CERVO Brain Research Centre, 2601, chemin de la Canardière, Québec, Quebec, Canada, G1J 2G3.
| |
Collapse
|
30
|
Garcia-Areas R, Libreros S, Simoes M, Castro-Silva C, Gazaniga N, Amat S, Jaczewska J, Keating P, Schilling K, Brito M, Wojcikiewicz EP, Iragavarpu-Charyulu V. Suppression of tumor-derived Semaphorin 7A and genetic ablation of host-derived Semaphorin 7A impairs tumor progression in a murine model of advanced breast carcinoma. Int J Oncol 2017; 51:1395-1404. [PMID: 29048670 PMCID: PMC5642386 DOI: 10.3892/ijo.2017.4144] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 07/31/2017] [Indexed: 12/12/2022] Open
Abstract
Solid tumors can generate a plethora of neurogenesis-related molecules that enhance their growth and metastasis. Among them, we have identified axonal guidance molecule Semaphorin 7A (SEMA7A) in breast cancer. The goal of this study was to determine the therapeutic effect of suppressing SEMA7A levels in the 4T1 murine model of advanced breast carcinoma. We used anti-SEMA7A short hairpin RNA (shRNA) to gene silence SEMA7A in 4T1 mammary tumor cells. When implanted into the mammary fat pads of syngeneic mice, SEMA7A shRNA-expressing 4T1 tumors exhibited decreased growth rates, deferred metastasis and reduced mortality. In vitro, SEMA7A shRNA-expressing 4T1 cells had weakened proliferative, migratory and invasive abilities, and decreased levels of mesenchymal factors. Atomic force microscopy studies showed that SEMA7A shRNA-expressing 4T1 cells had an increase in cell stiffness that corresponded with their decreased malignant potential. Genetic ablation of host-derived SEMA7A further enhanced the antitumor effects of SEMA7A shRNA gene silencing in 4T1 cells. Our preclinical findings demonstrate a critical role for SEMA7A in mediating mammary tumor progression.
Collapse
Affiliation(s)
- R Garcia-Areas
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - S Libreros
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - M Simoes
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - C Castro-Silva
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - N Gazaniga
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - S Amat
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - J Jaczewska
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - P Keating
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - K Schilling
- Lynn Women's Health & Wellness Institute, Boca Raton Regional Hospital, Boca Raton, FL 33431, USA
| | - M Brito
- Department of Pathology, Boca Raton Regional Hospital, Boca Raton, FL 33431, USA
| | - E P Wojcikiewicz
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - V Iragavarpu-Charyulu
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| |
Collapse
|
31
|
Pasquini JM, Barrantes FJ, Quintá HR. Normal development of spinal axons in early embryo stages and posterior locomotor function is independent of GAL-1. J Comp Neurol 2017; 525:2861-2875. [DOI: 10.1002/cne.24243] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 05/11/2017] [Accepted: 05/11/2017] [Indexed: 11/06/2022]
Affiliation(s)
- Juana M. Pasquini
- Departamento de Química Biológica; Instituto de Química y Físico Química Biológica, Universidad de Buenos Aires; Buenos Aires Argentina
| | | | - Héctor R. Quintá
- Departamento de Química Biológica; Instituto de Química y Físico Química Biológica, Universidad de Buenos Aires; Buenos Aires Argentina
| |
Collapse
|
32
|
Vanoni MA. Structure-function studies of MICAL, the unusual multidomain flavoenzyme involved in actin cytoskeleton dynamics. Arch Biochem Biophys 2017; 632:118-141. [PMID: 28602956 DOI: 10.1016/j.abb.2017.06.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 05/27/2017] [Accepted: 06/05/2017] [Indexed: 12/11/2022]
Abstract
MICAL (from the Molecule Interacting with CasL) indicates a family of multidomain proteins conserved from insects to humans, which are increasingly attracting attention for their participation in the control of actin cytoskeleton dynamics, and, therefore, in the several related key processes in health and disease. MICAL is unique among actin binding proteins because it catalyzes a NADPH-dependent F-actin depolymerizing reaction. This unprecedented reaction is associated with its N-terminal FAD-containing domain that is structurally related to p-hydroxybenzoate hydroxylase, the prototype of aromatic monooxygenases, but catalyzes a strong NADPH oxidase activity in the free state. This review will focus on the known structural and functional properties of MICAL forms in order to provide an overview of the arguments supporting the current hypotheses on the possible mechanism of action of MICAL in the free and F-actin bound state, on the modulating effect of the CH, LIM, and C-terminal domains that follow the catalytic flavoprotein domain on the MICAL activities, as well as that of small molecules and proteins interacting with MICAL.
Collapse
Affiliation(s)
- Maria Antonietta Vanoni
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133 Milano, Italy.
| |
Collapse
|
33
|
Ito D, Kumanogoh A. The role of Sema4A in angiogenesis, immune responses, carcinogenesis, and retinal systems. Cell Adh Migr 2016; 10:692-699. [PMID: 27736304 PMCID: PMC5160039 DOI: 10.1080/19336918.2016.1215785] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Semaphorins were originally identified as axon guidance cues that regulate the functional activity of axons in the nervous system. In addition, accumulating evidence indicates that semaphorins have multiple functions in physiological and pathogenic processes, including vascular development, tumor progression, and immune responses. Sema4A is a semaphorin expressed in immune cells, and is thus termed an “immune semaphorin.” Sema4A has 4 types of receptors: Plexin D family, Plexin B family, Tim-2, and Nrp-1. Recent studies suggest that Sema4A plays critical roles in many processes including cell–cell interactions, immune-cell activation, differentiation, and migration. In other studies, Sema4A is also associated with carcinogenesis and retinal systems. In this review, we summarize current knowledge regarding the biology of Sema4A in relation to angiogenesis, immune responses, colorectal cancer, and the retina.
Collapse
Affiliation(s)
- Daisuke Ito
- a Department of Respiratory Medicine , Allergy and Rheumatic Disease, Osaka University Graduate School of Medicine , Suita , Osaka , Japan
| | - Atsushi Kumanogoh
- a Department of Respiratory Medicine , Allergy and Rheumatic Disease, Osaka University Graduate School of Medicine , Suita , Osaka , Japan
| |
Collapse
|
34
|
Roh S, Yang D, Jeong S. Differential ligand regulation of PlexB signaling in motor neuron axon guidance in
Drosophila. Int J Dev Neurosci 2016; 55:34-40. [DOI: 10.1016/j.ijdevneu.2016.09.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 09/08/2016] [Accepted: 09/12/2016] [Indexed: 10/21/2022] Open
Affiliation(s)
- Seyun Roh
- Department of Molecular BiologyChonbuk National UniversityJeonjuJeollabukdo54896Republic of Korea
| | - Da‐som Yang
- Department of Molecular BiologyChonbuk National UniversityJeonjuJeollabukdo54896Republic of Korea
| | - Sangyun Jeong
- Department of Molecular BiologyChonbuk National UniversityJeonjuJeollabukdo54896Republic of Korea
| |
Collapse
|
35
|
Black SA, Nelson AC, Gurule NJ, Futscher BW, Lyons TR. Semaphorin 7a exerts pleiotropic effects to promote breast tumor progression. Oncogene 2016; 35:5170-8. [PMID: 27065336 PMCID: PMC5720143 DOI: 10.1038/onc.2016.49] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 12/22/2016] [Accepted: 01/19/2016] [Indexed: 02/05/2023]
Abstract
Understanding what drives breast tumor progression is of utmost importance for blocking tumor metastasis; we have identified that semaphorin 7a is a potent driver of ductal carcinoma in situ (DCIS) progression. Semaphorin 7a is a GPI membrane anchored protein that promotes attachment and spreading in multiple cell types. Here we show that increased expression of SEMA7A occurs in a large percentage of breast cancers and is associated with decreased overall and distant metastasis free survival. In both in vitro and in vivo models, shRNA mediated silencing of SEMA7A reveals roles for semaphorin 7a in the promotion of DCIS growth, motility, and invasion as well as lymphangiogenesis in the tumor microenvironment. Our studies also uncover a relationship between COX-2 and semaphorin 7a expression and suggest that semaphorin 7a promotes tumor cell invasion on collagen and lymphangiogenesis via activation of β1-integrin receptor. Our results suggest that semaphorin 7a, may be novel target for blocking breast tumor progression.
Collapse
Affiliation(s)
- S A Black
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - A C Nelson
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - N J Gurule
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,University of Colorado Cancer Biology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - B W Futscher
- The University of Arizona Cancer Center, The University of Arizona, Tucson, AZ, USA.,Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - T R Lyons
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,University of Colorado Cancer Biology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
36
|
Asan A, Raiders SA, Priess JR. Morphogenesis of the C. elegans Intestine Involves Axon Guidance Genes. PLoS Genet 2016; 12:e1005950. [PMID: 27035721 PMCID: PMC4817974 DOI: 10.1371/journal.pgen.1005950] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 03/01/2016] [Indexed: 11/21/2022] Open
Abstract
Genetic and molecular studies have provided considerable insight into how various tissue progenitors are specified in early embryogenesis, but much less is known about how those progenitors create three-dimensional tissues and organs. The C. elegans intestine provides a simple system for studying how a single progenitor, the E blastomere, builds an epithelial tube of 20 cells. As the E descendants divide, they form a primordium that transitions between different shapes over time. We used cell contours, traced from confocal optical z-stacks, to build a 3D graphic reconstruction of intestine development. The reconstruction revealed several new aspects of morphogenesis that extend and clarify previous observations. The first 8 E descendants form a plane of four right cells and four left cells; the plane arises through oriented cell divisions and VANG-1/Van Gogh-dependent repositioning of any non-planar cells. LIN-12/Notch signaling affects the left cells in the E8 primordium, and initiates later asymmetry in cell packing. The next few stages involve cell repositioning and intercalation events that shuttle cells to their final positions, like shifting blocks in a Rubik’s cube. Repositioning involves breaking and replacing specific adhesive contacts, and some of these events involve EFN-4/Ephrin, MAB-20/semaphorin-2a, and SAX-3/Robo. Once cells in the primordium align along a common axis and in the correct order, cells at the anterior end rotate clockwise around the axis of the intestine. The anterior rotation appears to align segments of the developing lumen into a continuous structure, and requires the secreted ligand UNC-6/netrin, the receptor UNC-40/DCC, and an interacting protein called MADD-2. Previous studies showed that rotation requires a second round of LIN-12/Notch signaling in cells on the right side of the primordium, and we show that MADD-2-GFP appears to be downregulated in those cells. This report uses the intestine of the nematode C. elegans as a model system to address how progenitor cells form a three-dimensional organ. The fully formed intestine is a cylindrical tube of only 20 epithelial cells, and all of these cells are descendants of a single cell, the E blastomere. The E descendants form a primordium that changes shape over time as different E descendants divide and move. Cells in the primordium must continually adhere to each other during these movements to maintain the integrity of the primordium. Here, we generated a 3D graphic reconstruction of the developing intestine in order to analyze these events. We found that the cell movements are highly reproducible, suggesting that they are programmed by asymmetric gene expression in the primordium. In particular, we found that the conserved receptor LIN-12/Notch appears to modulate left-right adhesion in the primordium, leading to the asymmetric packing of cells. One of the most remarkable events in intestinal morphogenesis is the circumferential rotation of a subset of cells. We found that rotation appears to have a role in aligning the developing lumen of the intestine, and involves a conserved, UNC-6/netrin signaling pathway that is best known for its roles in the guided growth of neurons.
Collapse
Affiliation(s)
- Alparsan Asan
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Stephan A. Raiders
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - James R. Priess
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Molecular and Cellular Biology Program, University of Washington, Seattle, Washington, United States of America
- Department of Biology, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
37
|
Masu M. Proteoglycans and axon guidance: a new relationship between old partners. J Neurochem 2016; 139 Suppl 2:58-75. [PMID: 26709493 DOI: 10.1111/jnc.13508] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 12/08/2015] [Accepted: 12/11/2015] [Indexed: 01/12/2023]
Abstract
Neural circuits are formed with great precision during development. Accumulated evidence over the past three decades has demonstrated that growing axons are navigated toward their targets by the combined actions of attractants and repellents together with their receptors. It has long been known that proteoglycans, glycosylated proteins possessing covalently attached glycosaminoglycans, play a critical role in axon guidance; however, the molecular mechanisms by which proteoglycans regulate axon behaviors remain largely unknown. Glycosaminoglycans such as heparan sulfate and chondroitin sulfate are large linear polysaccharides composed of repeating disaccharide units that are highly modified by specific sulfation and epimerization. Recent biochemical and molecular biological studies have identified the enzymes that are involved in the biosynthesis of glycosaminoglycans. Interestingly, many mutants lacking glycosaminoglycan-synthesizing enzymes or proteoglycans in several model organisms show defects in specific nerve tract formation. In parallel, detailed biochemical studies have identified the molecular interactions between axon guidance molecules and glycosaminoglycans that have specific modification in their sugar chains. This review summarizes the structure and function of axon guidance molecules and glycosaminoglycans, and then tries to combine the knowledge from these studies to understand the role of proteoglycans from a new vantage point. Deciphering the sugar code is important for understanding the complicated nature of proteoglycans in axon guidance. Neural circuits are formed by the combined actions of axon guidance molecules. Proteoglycans play critical roles in regulating axon guidance through the interaction between signaling molecules and glycosaminoglycan chains attached to the core protein. This paper summarizes the structure and functions of axon guidance molecules and glycosaminoglycans and reviews the molecular mechanisms by which proteoglycans regulate axon guidance from a new vantage point. This article is part of the 60th Anniversary special issue.
Collapse
Affiliation(s)
- Masayuki Masu
- Department of Molecular Neurobiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, Japan.
| |
Collapse
|
38
|
Sang C, Zhang Y, Chen F, Huang P, Qi J, Wang P, Zhou Q, Kang H, Cao X, Guo L. Tumor necrosis factor alpha suppresses osteogenic differentiation of MSCs by inhibiting semaphorin 3B via Wnt/β-catenin signaling in estrogen-deficiency induced osteoporosis. Bone 2016; 84:78-87. [PMID: 26723579 DOI: 10.1016/j.bone.2015.12.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 11/14/2015] [Accepted: 12/20/2015] [Indexed: 12/18/2022]
Abstract
The proinflammatory cytokines, especially tumor necrosis factor alpha (TNF-α), have been shown to inhibit osteogenic differentiation of mesenchymal stem cells (MSCs) and bone formation in estrogen-deficiency-induced osteoporosis, but the mechanisms of TNF-α impaired bone formation remain poorly understood. Semaphorins have been shown to regulate cell growth, cell migration, and cell differentiation in a variety of tissues, including bone tissue. Here, we identified a novel mechanism whereby TNF-α, suppressing Semaphorin3B expression contributes to estrogen-deficiency-induced osteoporosis. In this study, we found that TNF-α could decrease Semaphorin3B expression in osteogenic differentiation of MSCs. Overexpression of Semaphorin3B in MSCs attenuated the inhibitory effects of TNF-α on MSCs proliferation and osteoblastic differentiation. Mechanistically, activation of the Wnt/β-catenin signaling markedly rescued TNF-α-inhibited Semaphorin3B expression, suggesting that Wnt/β-catenin signaling was involved in the regulation of Semaphorin3B expression by TNF-α. Taken together, our results revealed a novel function for Semaphorin3B and suggested that suppressed Semaphorin3B may contribute to impaired bone formation by elevated TNF-α in estrogen-deficiency-induced osteoporosis. This study may indicate a therapeutic target gene of Semaphorin3B for osteoporosis.
Collapse
Affiliation(s)
- Chenglin Sang
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Orthopaedics and Traumatology, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Orthopaedics, Second Military Medical University's Jinan Clinical Medicine College, Jinan, China; Department of Orthaopedics, General Hospital of Jinan Military Command, Jinan 250031, Shandong, China
| | - Yongxian Zhang
- Department of Orthopaedics, Second Military Medical University's Jinan Clinical Medicine College, Jinan, China; Department of Orthaopedics, General Hospital of Jinan Military Command, Jinan 250031, Shandong, China
| | - Fangjing Chen
- Department of Orthopaedics, Second Military Medical University's Jinan Clinical Medicine College, Jinan, China; Department of Orthaopedics, General Hospital of Jinan Military Command, Jinan 250031, Shandong, China
| | - Ping Huang
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Orthopaedics and Traumatology, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jin Qi
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Orthopaedics and Traumatology, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Pingshan Wang
- Department of Orthopaedics, Second Military Medical University's Jinan Clinical Medicine College, Jinan, China; Department of Orthaopedics, General Hospital of Jinan Military Command, Jinan 250031, Shandong, China
| | - Qi Zhou
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Orthopaedics and Traumatology, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hui Kang
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Orthopaedics and Traumatology, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xuecheng Cao
- Department of Orthopaedics, Second Military Medical University's Jinan Clinical Medicine College, Jinan, China; Department of Orthaopedics, General Hospital of Jinan Military Command, Jinan 250031, Shandong, China.
| | - Lei Guo
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Orthopaedics and Traumatology, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
39
|
Leung C, Liu Y, Lu X, Kim M, Drysdale TA, Feng Q. Rac1 Signaling Is Required for Anterior Second Heart Field Cellular Organization and Cardiac Outflow Tract Development. J Am Heart Assoc 2015; 5:e002508. [PMID: 26722124 PMCID: PMC4859369 DOI: 10.1161/jaha.115.002508] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 11/18/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND The small GTPase Rac1 regulates diverse cellular functions, including both apicobasal and planar cell polarity pathways; however, its role in cardiac outflow tract (OFT) development remains unknown. In the present study, we aimed to examine the role of Rac1 in the anterior second heart field (SHF) splanchnic mesoderm and subsequent OFT development during heart morphogenesis. METHODS AND RESULTS Using the Cre/loxP system, mice with an anterior SHF-specific deletion of Rac1 (Rac1(SHF)) were generated. Embryos were collected at various developmental time points for immunostaining and histological analysis. Intrauterine echocardiography was also performed to assess aortic valve blood flow in embryos at embryonic day 18.5. The Rac1(SHF) splanchnic mesoderm exhibited disruptions in SHF progenitor cellular organization and proliferation. Consequently, this led to a spectrum of OFT defects along with aortic valve defects in Rac1(SHF) embryos. Mechanistically, it was found that the ability of the Rac1(SHF) OFT myocardial cells to migrate into the proximal OFT cushion was severely reduced. In addition, expression of the neural crest chemoattractant semaphorin 3c was decreased. Lineage tracing showed that anterior SHF contribution to the OFT myocardium and aortic valves was deficient in Rac1(SHF) hearts. Furthermore, functional analysis with intrauterine echocardiography at embryonic day 18.5 showed aortic valve regurgitation in Rac1(SHF) hearts, which was not seen in control hearts. CONCLUSIONS Disruptions of Rac1 signaling in the anterior SHF results in aberrant progenitor cellular organization and defects in OFT development. Our data show Rac1 signaling to be a critical regulator of cardiac OFT formation during embryonic heart development.
Collapse
Affiliation(s)
- Carmen Leung
- Departments of Physiology and Pharmacology, Medicine and PediatricsSchulich School of Medicine and DentistryCollaborative Program in Developmental BiologyChildren's Health Research InstituteUniversity of Western OntarioLondonOntarioCanada
| | - Yin Liu
- Departments of Physiology and Pharmacology, Medicine and PediatricsSchulich School of Medicine and DentistryCollaborative Program in Developmental BiologyChildren's Health Research InstituteUniversity of Western OntarioLondonOntarioCanada
| | - Xiangru Lu
- Departments of Physiology and Pharmacology, Medicine and PediatricsSchulich School of Medicine and DentistryCollaborative Program in Developmental BiologyChildren's Health Research InstituteUniversity of Western OntarioLondonOntarioCanada
| | - Mella Kim
- Departments of Physiology and Pharmacology, Medicine and PediatricsSchulich School of Medicine and DentistryCollaborative Program in Developmental BiologyChildren's Health Research InstituteUniversity of Western OntarioLondonOntarioCanada
| | - Thomas A. Drysdale
- Departments of Physiology and Pharmacology, Medicine and PediatricsSchulich School of Medicine and DentistryCollaborative Program in Developmental BiologyChildren's Health Research InstituteUniversity of Western OntarioLondonOntarioCanada
| | - Qingping Feng
- Departments of Physiology and Pharmacology, Medicine and PediatricsSchulich School of Medicine and DentistryCollaborative Program in Developmental BiologyChildren's Health Research InstituteUniversity of Western OntarioLondonOntarioCanada
| |
Collapse
|
40
|
Uchida Y, James JM, Suto F, Mukouyama YS. Class 3 semaphorins negatively regulate dermal lymphatic network formation. Biol Open 2015; 4:1194-205. [PMID: 26319580 PMCID: PMC4582121 DOI: 10.1242/bio.012302] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The development of a patterned lymphatic vascular network is essential for proper lymphatic functions during organ development and homeostasis. Here we report that class 3 semaphorins (SEMA3s), SEMA3F and SEMA3G negatively regulate lymphatic endothelial cell (LEC) growth and sprouting to control dermal lymphatic network formation. Neuropilin2 (NRP2) functions as a receptor for SEMA3F and SEMA3G, as well as vascular endothelial growth factor C (VEGFC). In culture, Both SEMA3F and SEMA3G inhibit VEGFC-mediated sprouting and proliferation of human dermal LECs. In the developing mouse skin, Sema3f is expressed in the epidermis and Sema3g expression is restricted to arteries, whereas their receptor Nrp2 is preferentially expressed by lymphatic vessels. Both Sema3f;Sema3g double mutants and Nrp2 mutants exhibit increased LEC growth in the skin. In contrast, Sema3f;Sema3g double mutants display increased lymphatic branching, while Nrp2 mutants exhibit reduced lymphatic branching. A targeted mutation in PlexinA1 or PlexinA2, signal transducers forming a receptor complex with NRP2 for SEMA3s, exhibits an increase in LEC growth and lymphatic branching as observed in Sema3f;Sema3g double mutants. Our results provide the first evidence that SEMA3F and SEMA3G function as a negative regulator for dermal lymphangiogenesis in vivo. The reciprocal phenotype in lymphatic branching between Sema3f;Sema3g double mutants and Nrp2 mutants suggest a complex NRP2 function that regulates LEC behavior both positively and negatively, through a binding with VEGFC or SEMA3s.
Collapse
Affiliation(s)
- Yutaka Uchida
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Building 10/6C103, 10 Center Drive, Bethesda, MD 20892, USA
| | - Jennifer M James
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Building 10/6C103, 10 Center Drive, Bethesda, MD 20892, USA
| | - Fumikazu Suto
- Department of Ultrastructural Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo 187-8502, Japan
| | - Yoh-Suke Mukouyama
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Building 10/6C103, 10 Center Drive, Bethesda, MD 20892, USA
| |
Collapse
|
41
|
Teixeira FG, Panchalingam KM, Anjo SI, Manadas B, Pereira R, Sousa N, Salgado AJ, Behie LA. Do hypoxia/normoxia culturing conditions change the neuroregulatory profile of Wharton Jelly mesenchymal stem cell secretome? Stem Cell Res Ther 2015. [PMID: 26204925 PMCID: PMC4533943 DOI: 10.1186/s13287-015-0124-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Introduction The use of human umbilical cord Wharton Jelly-derived mesenchymal stem cells (hWJ-MSCs) has been considered a new potential source for future safe applications in regenerative medicine. Indeed, the application of hWJ-MSCs into different animal models of disease, including those from the central nervous system, has shown remarkable therapeutic benefits mostly associated with their secretome. Conventionally, hWJ-MSCs are cultured and characterized under normoxic conditions (21 % oxygen tension), although the oxygen levels within tissues are typically much lower (hypoxic) than these standard culture conditions. Therefore, oxygen tension represents an important environmental factor that may affect the performance of mesenchymal stem cells in vivo. However, the impact of hypoxic conditions on distinct mesenchymal stem cell characteristics, such as the secretome, still remains unclear. Methods In the present study, we have examined the effects of normoxic (21 % O2) and hypoxic (5 % O2) conditions on the hWJ-MSC secretome. Subsequently, we address the impact of the distinct secretome in the neuronal cell survival and differentiation of human neural progenitor cells. Results The present data indicate that the hWJ-MSC secretome collected from normoxic and hypoxic conditions displayed similar effects in supporting neuronal differentiation of human neural progenitor cells in vitro. However, proteomic analysis revealed that the use of hypoxic preconditioning led to the upregulation of several proteins within the hWJ-MSC secretome. Conclusions Our results suggest that the optimization of parameters such as hypoxia may lead to the development of strategies that enhance the therapeutic effects of the secretome for future regenerative medicine studies and applications.
Collapse
Affiliation(s)
- Fábio G Teixeira
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal. .,ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Krishna M Panchalingam
- Pharmaceutical Production Research Facility (PPRF), Schulich School of Engineering, University of Calgary, Calgary, AB, Canada.
| | - Sandra Isabel Anjo
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal. .,Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal.
| | - Bruno Manadas
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal. .,Biocant - Biotechnology Innovation Center, Cantanhede, Portugal.
| | - Ricardo Pereira
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal. .,ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal. .,ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal. .,ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Leo A Behie
- Pharmaceutical Production Research Facility (PPRF), Schulich School of Engineering, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
42
|
Ito D, Nojima S, Nishide M, Okuno T, Takamatsu H, Kang S, Kimura T, Yoshida Y, Morimoto K, Maeda Y, Hosokawa T, Toyofuku T, Ohshima J, Kamimura D, Yamamoto M, Murakami M, Morii E, Rakugi H, Isaka Y, Kumanogoh A. mTOR Complex Signaling through the SEMA4A-Plexin B2 Axis Is Required for Optimal Activation and Differentiation of CD8+ T Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:934-43. [PMID: 26116513 DOI: 10.4049/jimmunol.1403038] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 05/22/2015] [Indexed: 12/19/2022]
Abstract
Mammalian target of rapamycin (mTOR) plays crucial roles in activation and differentiation of diverse types of immune cells. Although several lines of evidence have demonstrated the importance of mTOR-mediated signals in CD4(+) T cell responses, the involvement of mTOR in CD8(+) T cell responses is not fully understood. In this study, we show that a class IV semaphorin, SEMA4A, regulates CD8(+) T cell activation and differentiation through activation of mTOR complex (mTORC) 1. SEMA4A(-/-) CD8(+) T cells exhibited impairments in production of IFN-γ and TNF-α and induction of the effector molecules granzyme B, perforin, and FAS-L. Upon infection with OVA-expressing Listeria monocytogenes, pathogen-specific effector CD8(+) T cell responses were significantly impaired in SEMA4A(-/-) mice. Furthermore, SEMA4A(-/-) CD8(+) T cells exhibited reduced mTORC1 activity and elevated mTORC2 activity, suggesting that SEMA4A is required for optimal activation of mTORC1 in CD8(+) T cells. IFN-γ production and mTORC1 activity in SEMA4A(-/-) CD8(+) T cells were restored by administration of recombinant Sema4A protein. In addition, we show that plexin B2 is a functional receptor of SEMA4A in CD8(+) T cells. Collectively, these results not only demonstrate the role of SEMA4A in CD8(+) T cells, but also reveal a novel link between a semaphorin and mTOR signaling.
Collapse
Affiliation(s)
- Daisuke Ito
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan; Department of Geriatric Medicine and Nephrology, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan; Department of Respiratory Medicine, Allergy, and Rheumatic Disease, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Suita City, Osaka 565-0871, Japan
| | - Satoshi Nojima
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Suita City, Osaka 565-0871, Japan; Department of Pathology, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan;
| | - Masayuki Nishide
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan; Department of Respiratory Medicine, Allergy, and Rheumatic Disease, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Suita City, Osaka 565-0871, Japan
| | - Tatsusada Okuno
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan; Department of Neurology, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan
| | - Hyota Takamatsu
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan; Department of Respiratory Medicine, Allergy, and Rheumatic Disease, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Suita City, Osaka 565-0871, Japan
| | - Sujin Kang
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan; Department of Respiratory Medicine, Allergy, and Rheumatic Disease, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Suita City, Osaka 565-0871, Japan; Department of Clinical Application of Biologics, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan
| | - Tetsuya Kimura
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan; Department of Respiratory Medicine, Allergy, and Rheumatic Disease, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Suita City, Osaka 565-0871, Japan
| | - Yuji Yoshida
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan; Department of Respiratory Medicine, Allergy, and Rheumatic Disease, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Suita City, Osaka 565-0871, Japan
| | - Keiko Morimoto
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan; Department of Respiratory Medicine, Allergy, and Rheumatic Disease, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Suita City, Osaka 565-0871, Japan
| | - Yohei Maeda
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan; Department of Otorhinolaryngology-Head and Neck Surgery, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan
| | - Takashi Hosokawa
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan; Department of Respiratory Medicine, Allergy, and Rheumatic Disease, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Suita City, Osaka 565-0871, Japan
| | - Toshihiko Toyofuku
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Suita City, Osaka 565-0871, Japan; Department of Immunology and Regenerative Medicine, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan
| | - Jun Ohshima
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita City, Osaka 565-0871, Japan; Laboratory of Immunoparasitology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan; and
| | - Daisuke Kamimura
- Department of Molecular Neuroimmunology, Institute for Genetic Medicine, Hokkaido University Graduate School of Medicine, Sapporo City, Hokkaido 060-0815, Japan
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita City, Osaka 565-0871, Japan; Laboratory of Immunoparasitology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan; and
| | - Masaaki Murakami
- Department of Molecular Neuroimmunology, Institute for Genetic Medicine, Hokkaido University Graduate School of Medicine, Sapporo City, Hokkaido 060-0815, Japan
| | - Eiichi Morii
- Department of Pathology, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan
| | - Hiromi Rakugi
- Department of Geriatric Medicine and Nephrology, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan
| | - Yoshitaka Isaka
- Department of Geriatric Medicine and Nephrology, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan
| | - Atsushi Kumanogoh
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan; Department of Respiratory Medicine, Allergy, and Rheumatic Disease, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Suita City, Osaka 565-0871, Japan;
| |
Collapse
|
43
|
Plexin-B1 and semaphorin 4D cooperate to promote cutaneous squamous cell carcinoma cell proliferation, migration and invasion. J Dermatol Sci 2015; 79:127-36. [PMID: 26051877 DOI: 10.1016/j.jdermsci.2015.05.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 04/21/2015] [Accepted: 05/07/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND OBJECTIVES Semaphorin 4D (Sema4D) and its receptor, Plexin-B1, are involved in the pathogenesis of squamous cell carcinoma (SCC) by mediating angiogenesis or perineural invasion through the interaction between Sema4D expression on SCC cells and Plexin-B1 expression on endothelial cells or nerves. Plexin-B1 was also recently found to be expressed on SCC cells. Plexin-B1 expression on several types of tumor cells could mediate various, and occasionally opposing, effects, including tumor cell survival, proliferation, angiogenesis, invasion, and metastasis. However, whether Sema4D exerts paracrine or autocrine effects on SCC via Plexin-B1 remains unclear. OBJECTIVES The aim of this study is to explore the effects of Sema4D/Plexin-B1 interaction on SCC via Plexin-B1 expressed on the tumor cells. METHODS In the present study, we detected the expression of Plexin-B1 and Sema4D in cutaneous SCC (cSCC) tissues and in the cSCC cell line A431 and analyzed the effects of the Sema4D/Plexin-B1 interaction on cSCC cell proliferation, migration, and invasion, as well as on the signaling pathway downstream of Plexin-B1. RESULTS We observed significantly increased Plexin-B1 and Sema4D expression in keratinocytes in cSCC lesions and in A431 cells compared with that in normal skin tissue and in non-malignant keratinocytes. Plexin-B1 silencing reduced the growth, proliferation, migration, and invasion of A431 cells and inhibited the phosphorylation of Akt and extracellular signal-regulated protein kinase (Erk). Soluble recombinant Sema4D promoted the growth, proliferation, migration, and invasion of A431 cells; Akt and Erk phosphorylation is also involved in these processes with a Plexin-B1 dependent manner. CONCLUSION Plexin-B1 induces cSCC cell proliferation, migration, and invasion by interacting with Sema4D. Plexin-B1 might serve as a useful biomarker and/or as a novel therapeutic target for cSCC.
Collapse
|
44
|
Laurenzana A, Fibbi G, Chillà A, Margheri G, Del Rosso T, Rovida E, Del Rosso M, Margheri F. Lipid rafts: integrated platforms for vascular organization offering therapeutic opportunities. Cell Mol Life Sci 2015; 72:1537-57. [PMID: 25552244 PMCID: PMC11113367 DOI: 10.1007/s00018-014-1814-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 12/12/2014] [Accepted: 12/19/2014] [Indexed: 02/07/2023]
Abstract
Research on the nanoscale membrane structures known as lipid rafts is relevant to the fields of cancer biology, inflammation and ischaemia. Lipid rafts recruit molecules critical to signalling and regulation of the invasion process in malignant cells, the leukocytes that provide immunity in inflammation and the endothelial cells that build blood and lymphatic vessels, as well as the patterning of neural networks. As angiogenesis is a common denominator, regulation of receptors and signalling molecules critical to angiogenesis is central to the design of new approaches aimed at reducing, promoting or normalizing the angiogenic process. The goal of this review is to highlight some of the key issues that indicate the involvement of endothelial cell lipid rafts at each step of so-called 'sprouting angiogenesis', from stimulation of the vascular endothelial growth factor to the choice of tip cells, activation of migratory and invasion pathways, recruitment of molecules that guide axons in vascular patterning and maturation of blood vessels. Finally, the review addresses opportunities for future studies to define how these lipid domains (and their constituents) may be manipulated to stimulate the so-called 'normalization' of vascular networks within tumors, and be identified as the main target, enabling the development of more efficient chemotherapeutics and cancer immunotherapies.
Collapse
Affiliation(s)
- Anna Laurenzana
- Section of Experimental Pathology and Oncology, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy
| | - Gabriella Fibbi
- Section of Experimental Pathology and Oncology, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy
| | - Anastasia Chillà
- Section of Experimental Pathology and Oncology, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy
| | - Giancarlo Margheri
- Institute of Complex Systems (ISC), Consiglio Nazionale delle Ricerche (CNR), Florence, Italy
| | - Tommaso Del Rosso
- Department of Physics, Pontificia Universidade Catolica do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elisabetta Rovida
- Section of Experimental Pathology and Oncology, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy
| | - Mario Del Rosso
- Section of Experimental Pathology and Oncology, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy
- Istituto Toscano Tumori, Florence, Italy
| | - Francesca Margheri
- Section of Experimental Pathology and Oncology, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy
| |
Collapse
|
45
|
Wang JS, Jing CQ, Shan KS, Chen YZ, Guo XB, Cao ZX, Mu LJ, Peng LP, Zhou ML, Li LP. Semaphorin 4D and hypoxia-inducible factor-1α overexpression is related to prognosis in colorectal carcinoma. World J Gastroenterol 2015; 21:2191-2198. [PMID: 25717256 PMCID: PMC4326158 DOI: 10.3748/wjg.v21.i7.2191] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Revised: 09/17/2014] [Accepted: 11/11/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate semaphorin 4D (Sema4D) and hypoxia-inducible factor-1α (HIF-1α) expression in colorectal carcinoma and evaluate their clinicopathological and prognostic significance.
METHODS: Eighty-six curatively resected colorectal carcinoma patients at different stages of disease were randomly selected from the group of patients who underwent surgery, and none of them received preoperative radiochemotherapy. Normal proximal adjacent bowel tissue, which served as an internal control, was obtained from 52 randomly selected patients. Immunohistochemistry was performed to analyze the expression of Sema4D and the tumor angiogenesis-related protein HIF-1α in normal colorectal tissues and colorectal carcinoma tissues. The relationships between the expression and clinical characters and prognosis were analyzed.
RESULTS: HIF-1α and Sema4D were positively expressed in 58% and 60% of colorectal carcinoma tissues, respectively. Significantly lower expression levels were observed in normal mucosa (8% and 12%, respectively). HIF-1α and Sema4D expression was closely correlated with histological tumor type, tumor-node-metastasis (TNM) stage, and lymphatic metastasis (P < 0.05), but not with age or tumor size (P > 0.05). HIF-1α and Sema4D protein expression was significantly correlated with prognosis of colorectal carcinoma, as determined by Spearman rank correlation analysis (r = 0.567; P < 0.01). Multivariate Cox analysis revealed that only Sema4D expression played a significant role in predicting patient prognosis (P < 0.05).
CONCLUSION: These findings suggest that HIF-1α and Sema4D expression correlates with histological tumor type, TNM stage, and lymphatic metastasis in colorectal carcinoma and that Sema4D is a prognostic indicator of colorectal carcinoma.
Collapse
|
46
|
Developmental gene expression profile of axon guidance cues in Purkinje cells during cerebellar circuit formation. THE CEREBELLUM 2014; 13:307-17. [PMID: 24550128 DOI: 10.1007/s12311-014-0548-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The establishment of precise neural circuits during development involves a variety of contact-mediated and secreted guidance molecules that are expressed in a complementary fashion by different cell types. To build a functional circuit, each cell type must first trigger an intrinsic genetic program that is led by their environment at a key time point. It is therefore essential to identify the different cell-specific and stage-specific transcriptional profiles expressed by neurons. However, very few studies have been done to address this issue thus far. Herein, we have carried out a large-scale quantitative real-time PCR analysis of all classical axon guidance molecules (i.e., Semaphorins, Netrins, Ephrins, and Slits) and their receptors expressed by Purkinje cells (PCs) at specific stages of postnatal cerebellar development in vivo. Most cerebellar connections are setup in a well-characterized sequential manner during postnatal development and lead to the fine regulation of the PC, the sole output of the structure. Our analysis of the relative expression of these guidance cues has uncovered a dynamic expression pattern corresponding to specific stages of cerebellar development, thus providing a starting point for studying the role of these axon guidance molecules in cerebellar wiring.
Collapse
|
47
|
Ko JA, Hirata J, Yamane K, Sonoda KH, Kiuchi Y. Up-regulation of semaphorin 4A expression in human retinal pigment epithelial cells by PACAP released from cocultured neural cells. Cell Biochem Funct 2014; 33:29-36. [PMID: 25515530 DOI: 10.1002/cbf.3082] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 11/04/2014] [Accepted: 11/05/2014] [Indexed: 01/01/2023]
Abstract
Development and homeostasis of multicellular organisms require interactions between neighbouring cells. We recently established an in vitro model of cell-cell interaction based on a collagen vitrigel membrane. We have now examined the role of neural cells in retinal homeostasis by coculture of human retinal pigment epithelial (RPE) cells and neural cells on opposite sides of such a membrane. The neural cells (differentiated PC12 cells) induced up-regulation of semaphorin 4A (Sema4A), a member of the semaphorin family of neural guidance proteins, in RPE (ARPE19) cells. This effect of the neural cells was mimicked by the neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) and was abolished by the PACAP antagonist PACAP(6-38). Coculture with neural cells or stimulation with PACAP also induced the phosphorylation of extracellular-signal-regulated kinase in ARPE19 cells, and this effect of the neural cells was inhibited by PACAP(6-38). Finally, among various cytokines examined, only the amount of interleukin-6 released by cocultures of ARPE19 and neural cells differed from that released by ARPE19 cells cultured alone. Interleukin-6 was not detected in culture supernatants of neural cells, and the reduction in the amount of interleukin-6 released by the cocultures compared with that released by ARPE19 cells alone was prevented by PACAP(6-38). Our findings suggest that PACAP released from retinal neural cells (photoreceptors or optic nerve cells) may regulate Sema4A expression in RPE cells and thereby contribute to the maintenance of retinal structure and function. Development and homeostasis of multicellular organisms require interactions between neighbouring cells. With the use of a coculture system based on a collagen vitrigel membrane, we have now shown that neural cells induce up-regulation of the neural guidance protein Sema4A in RPE cells. This effect of neural cells appears to be mediated by the neuropeptide PACAP. PACAP released from retinal neural cells (photoreceptors or optic nerve cells) may thus regulate Sema4A expression in RPE cells and thereby contribute to the maintenance of retinal structure and function.
Collapse
Affiliation(s)
- Ji-Ae Ko
- Department of Ophthalmology, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima City, Hiroshima, Japan
| | | | | | | | | |
Collapse
|
48
|
Ma B, Herzog EL, Lee CG, Peng X, Lee CM, Chen X, Rockwell S, Koo JS, Kluger H, Herbst RS, Sznol M, Elias JA. Role of chitinase 3-like-1 and semaphorin 7a in pulmonary melanoma metastasis. Cancer Res 2014; 75:487-96. [PMID: 25511377 DOI: 10.1158/0008-5472.can-13-3339] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The prototypic chitinase-like protein Chi3l1 is induced in cancers and portends a poor prognosis, but whether it contributes to cancer progression is unknown. To address this gap in knowledge, we investigated the production of Chi3l1 in melanoma lung metastases. We found that Chi3l1 was induced during pulmonary melanoma metastasis and that this induction was regulated by the semaphorin Sema7a, interacting in stimulatory or inhibitory ways with its β1 integrin or Plexin C1 receptors, respectively. In mouse strains with genetic deletions of Chi3l1 or Sema7a, there was a significant reduction in pulmonary metastasis. Notably, antiserum raised against Chi3l1 or Sema7a phenocopied the reduction produced by genetic deletions. Melanoma lung metastasis was also decreased in the absence of IL13Rα2, a recently identified receptor for Chi3l1, consistent with a key role for Chi3l1 in melanoma spread. We confirmed roles for Sema7a and Chi3l1 in pulmonary metastasis of EMT6 breast cancer cells. Taken together, our studies establish a novel pathway through which Sem7a and its receptors regulate Chi3l1, revealing a host axis involving IL13Rα2 that plays a critical role in generating a pulmonary microenvironment that is critical to license metastasis.
Collapse
Affiliation(s)
- Bing Ma
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island
| | - Erica L Herzog
- Section of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island
| | - Xueyan Peng
- Section of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Chang-Min Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island
| | - Xiaosong Chen
- Section of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Sara Rockwell
- Department of Therapeutic Radiology, Yale Cancer Center, New Haven, Connecticut
| | - Ja Seok Koo
- Section of Medical Oncology, Department of Internal Medicine, Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| | - Harriet Kluger
- Section of Medical Oncology, Department of Internal Medicine, Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| | - Roy S Herbst
- Section of Medical Oncology, Department of Internal Medicine, Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| | - Mario Sznol
- Section of Medical Oncology, Department of Internal Medicine, Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| | - Jack A Elias
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island. Section of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut.
| |
Collapse
|
49
|
Sanchez-Castro M, Pichon O, Briand A, Poulain D, Gournay V, David A, Caignec CL. Disruption of theSEMA3DGene in a Patient with Congenital Heart Defects. Hum Mutat 2014; 36:30-3. [DOI: 10.1002/humu.22702] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 09/11/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Marta Sanchez-Castro
- INSERM; UMR1087; l'institut du thorax; Nantes France
- Université de Nantes; Nantes France
| | - Olivier Pichon
- CHU Nantes; Service de Génétique Médicale; Nantes France
| | - Annaig Briand
- CHU Nantes; Service de Génétique Médicale; Nantes France
| | - Damien Poulain
- CHU Nantes; Service de Génétique Médicale; Nantes France
| | | | - Albert David
- CHU Nantes; Service de Génétique Médicale; Nantes France
| | - Cédric Le Caignec
- INSERM; UMR1087; l'institut du thorax; Nantes France
- Université de Nantes; Nantes France
- CHU Nantes; Service de Génétique Médicale; Nantes France
| |
Collapse
|
50
|
Andermatt I, Wilson NH, Bergmann T, Mauti O, Gesemann M, Sockanathan S, Stoeckli ET. Semaphorin 6B acts as a receptor in post-crossing commissural axon guidance. Development 2014; 141:3709-20. [PMID: 25209245 DOI: 10.1242/dev.112185] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Semaphorins are a large family of axon guidance molecules that are known primarily as ligands for plexins and neuropilins. Although class-6 semaphorins are transmembrane proteins, they have been implicated as ligands in different aspects of neural development, including neural crest cell migration, axon guidance and cerebellar development. However, the specific spatial and temporal expression of semaphorin 6B (Sema6B) in chick commissural neurons suggested a receptor role in axon guidance at the spinal cord midline. Indeed, in the absence of Sema6B, post-crossing commissural axons lacked an instructive signal directing them rostrally along the contralateral floorplate border, resulting in stalling at the exit site or even caudal turns. Truncated Sema6B lacking the intracellular domain was unable to rescue the loss-of-function phenotype, confirming a receptor function of Sema6B. In support of this, we demonstrate that Sema6B binds to floorplate-derived plexin A2 (PlxnA2) for navigation at the midline, whereas a cis-interaction between PlxnA2 and Sema6B on pre-crossing commissural axons may regulate the responsiveness of axons to floorplate-derived cues.
Collapse
Affiliation(s)
- Irwin Andermatt
- Institute of Molecular Life Sciences and Neuroscience Center Zurich, Winterthurerstrasse 190, Zurich 8057, Switzerland
| | - Nicole H Wilson
- Institute of Molecular Life Sciences and Neuroscience Center Zurich, Winterthurerstrasse 190, Zurich 8057, Switzerland
| | - Timothy Bergmann
- Institute of Molecular Life Sciences and Neuroscience Center Zurich, Winterthurerstrasse 190, Zurich 8057, Switzerland
| | - Olivier Mauti
- Institute of Molecular Life Sciences and Neuroscience Center Zurich, Winterthurerstrasse 190, Zurich 8057, Switzerland
| | - Matthias Gesemann
- Institute of Molecular Life Sciences and Neuroscience Center Zurich, Winterthurerstrasse 190, Zurich 8057, Switzerland
| | - Shanthini Sockanathan
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Esther T Stoeckli
- Institute of Molecular Life Sciences and Neuroscience Center Zurich, Winterthurerstrasse 190, Zurich 8057, Switzerland
| |
Collapse
|