1
|
Al‐kuraishy HM, Sulaiman GM, Mohammed HA, Dawood RA, Albuhadily AK, Al‐Gareeb AI, Abomughaid MM, Klionsky DJ. Alterations in the Processing of Platelet APP (Amyloid Beta Precursor Protein) in Alzheimer Disease: The Possible Nexus. Neuropsychopharmacol Rep 2025; 45:e12525. [PMID: 39757022 PMCID: PMC11702489 DOI: 10.1002/npr2.12525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/03/2024] [Accepted: 12/26/2024] [Indexed: 01/07/2025] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease associated with the development of dementia. The hallmarks of AD neuropathology are accumulations of amyloid peptide (Aβ) and neurofibrillary tangles (NFTs). Aβ is derived from the processing of APP (amyloid beta precursor protein) by BACE1 (beta-secretase 1) and γ-secretase through an amyloidogenic pathway. However, processing of APP by ADAM10/α-secretase (ADAM metallopeptidase domain 10) enzymes through a non-amyloidogenic pathway produces soluble APP alpha (sAPPα), which has a neuroprotective effect. It has been shown that activated platelets are implicated in the pathogenesis of AD, which also increases platelet activation. Under physiological conditions, platelets regulate synaptic plasticity and increase neuronal differentiation by regulation of the inflammatory response. However, overactivated platelets contribute to the pathogenesis of AD. Activated platelets represent the main source of circulating APP and Aβ that may be involved in AD neuropathology. Therefore, there is a close relationship between AD neuropathology and activated platelets. This review discusses the potential role of platelets in the pathogenesis of AD, and how targeting of activated platelets may reduce AD neuropathology.
Collapse
Affiliation(s)
- Hayder M. Al‐kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineMustansiriyah UniversityBaghdadIraq
| | - Ghassan M. Sulaiman
- Division of Biotechnology, Department of Applied SciencesUniversity of TechnologyBaghdadIraq
| | - Hamdoon A. Mohammed
- Department of Medicinal Chemistry and Pharmacognosy, College of PharmacyQassim UniversityBuraydahQassimSaudi Arabia
| | - Retaj A. Dawood
- Department of Biology, College of ScienceAl‐Mustaqbal UniversityHillahIraq
| | - Ali K. Albuhadily
- Department of Clinical Pharmacology and Medicine, College of MedicineMustansiriyah UniversityBaghdadIraq
| | | | - Mosleh M. Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical SciencesUniversity of BishaBishaSaudi Arabia
| | | |
Collapse
|
2
|
Whately KM, Sengottuvel N, Edatt L, Srivastava S, Woods AT, Tsai YS, Porrello A, Zimmerman MP, Chack AC, Jefferys SR, Yacovone G, Kim DJ, Dudley AC, Amelio AL, Pecot CV. Spon1+ inflammatory monocytes promote collagen remodeling and lung cancer metastasis through lipoprotein receptor 8 signaling. JCI Insight 2024; 9:e168792. [PMID: 38716730 PMCID: PMC11141919 DOI: 10.1172/jci.insight.168792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/21/2024] [Indexed: 05/12/2024] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths in the world, and non-small cell lung cancer (NSCLC) is the most common subset. We previously found that infiltration of tumor inflammatory monocytes (TIMs) into lung squamous carcinoma (LUSC) tumors is associated with increased metastases and poor survival. To further understand how TIMs promote metastases, we compared RNA-Seq profiles of TIMs from several LUSC metastatic models with inflammatory monocytes (IMs) of non-tumor-bearing controls. We identified Spon1 as upregulated in TIMs and found that Spon1 expression in LUSC tumors corresponded with poor survival and enrichment of collagen extracellular matrix signatures. We observed SPON1+ TIMs mediate their effects directly through LRP8 on NSCLC cells, which resulted in TGF-β1 activation and robust production of fibrillar collagens. Using several orthogonal approaches, we demonstrated that SPON1+ TIMs were sufficient to promote NSCLC metastases. Additionally, we found that Spon1 loss in the host, or Lrp8 loss in cancer cells, resulted in a significant decrease of both high-density collagen matrices and metastases. Finally, we confirmed the relevance of the SPON1/LRP8/TGF-β1 axis with collagen production and survival in patients with NSCLC. Taken together, our study describes how SPON1+ TIMs promote collagen remodeling and NSCLC metastases through an LRP8/TGF-β1 signaling axis.
Collapse
Affiliation(s)
| | - Nisitha Sengottuvel
- UNC Lineberger Comprehensive Cancer Center and
- Department of Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Lincy Edatt
- UNC Lineberger Comprehensive Cancer Center and
| | - Sonal Srivastava
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Allison T. Woods
- UNC Lineberger Comprehensive Cancer Center and
- Department of Cell Biology and Physiology and
| | - Yihsuan S. Tsai
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Matthew P. Zimmerman
- UNC Lineberger Comprehensive Cancer Center and
- Department of Cell Biology and Physiology and
| | - Aaron C. Chack
- UNC Lineberger Comprehensive Cancer Center and
- Department of Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | | | - Dae Joong Kim
- Department of Microbiology, Immunology, and Cancer Biology and
| | - Andrew C. Dudley
- Department of Microbiology, Immunology, and Cancer Biology and
- UVA Comprehensive Cancer Center, The University of Virginia, Charlottesville, Virginia, USA
| | - Antonio L. Amelio
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
- Department of Head and Neck-Endocrine Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Chad V. Pecot
- UNC Lineberger Comprehensive Cancer Center and
- Division of Oncology and
- RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
3
|
Miyakawa R, Kobayashi M, Sugimoto K, Endo Y, Kojima M, Kobayashi Y, Furukawa S, Honda T, Watanabe T, Asano S, Soeda S, Hashimoto Y, Fujimori K, Chiba H. SPON1 is an independent prognostic biomarker for ovarian cancer. J Ovarian Res 2023; 16:95. [PMID: 37179355 PMCID: PMC10182672 DOI: 10.1186/s13048-023-01180-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND Ovarian cancer has the worst outcome among gynecological malignancies; therefore, biomarkers that could contribute to the early diagnosis and/or prognosis prediction are urgently required. In the present study, we focused on the secreted protein spondin-1 (SPON1) and clarified the prognostic relevance in ovarian cancer. METHODS We developed a monoclonal antibody (mAb) that selectively recognizes SPON1. Using this specific mAb, we determined the expression of SPON1 protein in the normal ovary, serous tubal intraepithelial carcinoma (STIC), and ovarian cancer tissues, as well as in various normal adult tissues by immunohistochemistry, and verified its clinicopathological significance in ovarian cancer. RESULTS The normal ovarian tissue was barely positive for SPON1, and no immunoreactive signals were detected in other healthy tissues examined, which was in good agreement with data obtained from gene expression databases. By contrast, upon semi-quantification, 22 of 242 ovarian cancer cases (9.1%) exhibited high SPON1 expression, whereas 64 (26.4%), 87 (36.0%), and 69 (28.5%) cases, which were designated as SPON1-low, possessed the moderate, weak, and negative SPON1 expression, respectively. The STIC tissues also possessed SPON1-positive signals. The 5-year recurrence-free survival (RFS) rate in the SPON1-high group (13.6%) was significantly lower than that in the SPON1-low group (51.2%). In addition, high SPON1 expression was significantly associated with several clinicopathological variables. Multivariable analysis revealed that high SPON1 was an independent prognostic factor for RFS of ovarian cancer. CONCLUSIONS SPON1 represents a prognostic biomarker for ovarian cancer, and the anti-SPON1 mAb could be valuable as an outcome predictor.
Collapse
Affiliation(s)
- Ryoya Miyakawa
- Department of Basic Pathology, Fukushima Medical University School of Medicine, Fukushima, 960- 1295, Japan
| | - Makoto Kobayashi
- Department of Basic Pathology, Fukushima Medical University School of Medicine, Fukushima, 960- 1295, Japan.
| | - Kotaro Sugimoto
- Department of Basic Pathology, Fukushima Medical University School of Medicine, Fukushima, 960- 1295, Japan
| | - Yuta Endo
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
- Department of Regional Gynecologic Oncology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Manabu Kojima
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
- Department of Regional Gynecologic Oncology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Yasuyuki Kobayashi
- Department of Diagnostic Pathology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Shigenori Furukawa
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
- Department of Regional Medical Support for Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Tsuyoshi Honda
- Department of Regional Medical Support for Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
- Department of Obstetrics and Gynecology, Iwaki City Medical Center, Iwaki, 973-8555, Japan
| | - Takafumi Watanabe
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
- Department of Regional Gynecologic Oncology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Shigeyuki Asano
- Department of Pathology, Iwaki City Medical Center, Iwaki, 973-8555, Japan
| | - Shu Soeda
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
- Department of Regional Gynecologic Oncology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Yuko Hashimoto
- Department of Diagnostic Pathology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Keiya Fujimori
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
- Department of Regional Gynecologic Oncology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
- Department of Regional Medical Support for Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Hideki Chiba
- Department of Basic Pathology, Fukushima Medical University School of Medicine, Fukushima, 960- 1295, Japan.
| |
Collapse
|
4
|
Yang L, Yue W, Zhang H, Gao Y, Yang L, Li L. The role of roof plate-specific spondins in liver homeostasis and disease. LIVER RESEARCH 2022; 6:139-145. [PMID: 39958194 PMCID: PMC11791806 DOI: 10.1016/j.livres.2022.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/05/2022] [Accepted: 09/02/2022] [Indexed: 02/16/2023]
Abstract
As evolutionarily conserved signals, roof plate-specific spondins (R-spondins; RSPOs) are a family with four members (RSPO1-4) exerting distinctly different functions. RSPOs have five receptors and correlate with different signaling pathways through these receptors and then perform various functions. Moreover, their best-known molecular function is the capacity to enhance WNT signaling pathways, which play critical roles in several processes. A recent study shows that RSPOs not only potentiate the WNT/beta (β)-catenin signaling pathway but are also involved in the WNT/planar cell polarity signaling pathway. RSPOs influence liver homeostasis and the development of multiple liver diseases. RSPO1 increases cell proliferation, protects hepatocytes from injury, improves liver regenerative potential, and affects liver metabolic zonation. RSPO2 not only regulates proliferation-associated genes and promotes differentiation in the liver but also participates in liver fibrosis through the WNT/β-catenin signaling pathway. RSPO3 is a key determinant of proper liver function, such as promoting hepatocyte regeneration and maintaining liver zonation. RSPO3 is upregulated in liver fibrosis and livers of patients with non-alcoholic steatohepatitis. Besides, RSPO2 and RSPO3 are confirmed as oncogenes and involved in the occurrence of liver cancer. The role of RSPO4 in the liver remains unclear. In this review, the structural and biochemical properties of RSPOs and their receptors and their roles in liver homeostasis and disease are summarized.
Collapse
Affiliation(s)
- Le Yang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Wenhui Yue
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Hang Zhang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Yue Gao
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Lin Yang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Liying Li
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Fernandez S, Burnham SC, Milicic L, Savage G, Maruff P, Peretti M, Sohrabi HR, Lim YY, Weinborn M, Ames D, Masters CL, Martins RN, Rainey-Smith S, Rowe CC, Salvado O, Groth D, Verdile G, Villemagne VL, Porter T, Laws SM. SPON1 Is Associated with Amyloid-β and APOE ε4-Related Cognitive Decline in Cognitively Normal Adults. J Alzheimers Dis Rep 2021; 5:111-120. [PMID: 33782664 PMCID: PMC7990462 DOI: 10.3233/adr-200246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstract.
Collapse
Affiliation(s)
- Shane Fernandez
- Australian Alzheimer's Research Foundation, Nedlands, Western Australia.,Collaborative Genomics and Translation Group, Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Samantha C Burnham
- Collaborative Genomics and Translation Group, Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,CSIRO Health and Biosecurity, Parkville, Victoria, Australia
| | - Lidija Milicic
- Collaborative Genomics and Translation Group, Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Greg Savage
- ARC Centre of Excellence in Cognition and its Disorders, Department of Psychology, Macquarie University, North Ryde, NSW, Australia
| | - Paul Maruff
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia.,CogState Ltd., Melbourne, Victoria, Australia
| | - Madeline Peretti
- Collaborative Genomics and Translation Group, Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Hamid R Sohrabi
- Australian Alzheimer's Research Foundation, Nedlands, Western Australia.,Centre for Healthy Ageing, Murdoch University, Murdoch, Western Australia, Australia.,Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia.,Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Yen Ying Lim
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Victoria, Australia
| | - Michael Weinborn
- Australian Alzheimer's Research Foundation, Nedlands, Western Australia.,Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,School of Psychological Science, University of Western Australia, Crawley, Western Australia, Australia
| | - David Ames
- Academic Unit for Psychiatry of Old Age, St. Vincent's Health, The University of Melbourne, Kew, Victoria, Australia.,National Ageing Research Institute, Parkville, Victoria, Australia
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Ralph N Martins
- Australian Alzheimer's Research Foundation, Nedlands, Western Australia.,Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia.,Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Stephanie Rainey-Smith
- Australian Alzheimer's Research Foundation, Nedlands, Western Australia.,Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Christopher C Rowe
- Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, Victoria, Australia.,Department of Medicine, Austin Health, The University of Melbourne, Heidelberg, Victoria, Australia
| | - Olivier Salvado
- CSIRO Health and Biosecurity/Australian e-Health Research Centre, Herston, Queensland, Australia
| | - David Groth
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Giuseppe Verdile
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Victor L Villemagne
- Collaborative Genomics and Translation Group, Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, Victoria, Australia
| | - Tenielle Porter
- Collaborative Genomics and Translation Group, Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Simon M Laws
- Collaborative Genomics and Translation Group, Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| |
Collapse
|
6
|
Schubert D. A Brief History of Adherons: The Discovery of Brain Exosomes. Int J Mol Sci 2020; 21:ijms21207673. [PMID: 33081326 PMCID: PMC7590140 DOI: 10.3390/ijms21207673] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/12/2020] [Accepted: 10/15/2020] [Indexed: 12/12/2022] Open
Abstract
Although exosomes were first described in reticulocytes in 1983, many people do not realize that similar vesicles had been studied in the context of muscle and nerve, beginning in 1980. At the time of their discovery, these vesicles were named adherons, and they were found to play an important role in both cell–substrate and cell–cell adhesion. My laboratory described several molecules that are present in adherons, including heparan sulfate proteoglycans (HSPGs) and purpurin. HSPGs have since been shown to play a variety of key roles in brain physiology. Purpurin has a number of important functions in the retina, including a role in nerve cell differentiation and regeneration. In this review, I discuss the discovery of adherons and how that led to continuing studies on their role in the brain with a particular focus on HSPGs.
Collapse
Affiliation(s)
- David Schubert
- Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| |
Collapse
|
7
|
Patterns of spon1b:GFP expression during early zebrafish brain development. BMC Res Notes 2020; 13:14. [PMID: 31910899 PMCID: PMC6945593 DOI: 10.1186/s13104-019-4876-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 12/24/2019] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE F-spondin is part of a group of evolutionarily conserved extracellular matrix proteins in vertebrates. It is highly expressed in the embryonic floor plate, and it can bind to the ECM and promote neuronal outgrowth. A characterization of F-spondin expression patterns in the adult zebrafish brain was previously reported by our group. However, given its importance during development, we aimed to obtain a detailed description of green fluorescent protein (GFP) expression driven by the spon1b promotor, in the developing zebrafish brain of the transgenic Tg(spon1b:GFP) line, using light sheet fluorescence microscopy (LSFM). RESULTS Images obtained in live embryos from 22 to 96 h post fertilization confirmed our earlier reports on the presence of spon1b:GFP expressing cells in the telencephalon and diencephalon (olfactory bulbs, habenula, optic tectum, nuclei of the medial longitudinal fasciculus), and revealed new spon1b:GFP populations in the pituitary anlage, dorso-rostral cluster, and ventro-rostral cluster. LSFM made it possible to follow the dynamics of cellular migration patterns during development. CONCLUSIONS spon1b:GFP larval expression patterns starts in early development in specific neuronal structures of the developing brain associated with sensory-motor modulation. LSFM evaluation of the transgenic Tg(spon1b:GFP) line provides an effective approach to characterize GFP expression patterns in vivo.
Collapse
|
8
|
The Matricellular Protein R-Spondin 2 Promotes Midbrain Dopaminergic Neurogenesis and Differentiation. Stem Cell Reports 2018; 11:651-664. [PMID: 30146491 PMCID: PMC6135723 DOI: 10.1016/j.stemcr.2018.07.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 07/28/2018] [Accepted: 07/29/2018] [Indexed: 12/11/2022] Open
Abstract
The development of midbrain dopaminergic (mDA) neurons is controlled by multiple morphogens and transcription factors. However, little is known about the role of extracellular matrix proteins in this process. Here we examined the function of roof plate-specific spondins (RSPO1-4) and the floor plate-specific, spondin 1 (SPON1). Only RSPO2 and SPON1 were expressed at high levels during mDA neurogenesis, and the receptor LGR5 was expressed by midbrain floor plate progenitors. Surprisingly, RSPO2, but not SPON1, specifically promoted the differentiation of mDA neuroblasts into mDA neurons in mouse primary cultures and embryonic stem cells (ESCs). In addition, RSPO2 was found to promote not only mDA differentiation, but also mDA neurogenesis in human ESCs. Our results thus uncover an unexpected function of the matricellular protein RSPO2 and suggest an application to improve mDA neurogenesis and differentiation in human stem cell preparations destined to cell replacement therapy or drug discovery for Parkinson disease. Rspo2 is dynamically expressed during midbrain dopaminergic neuron development RSPO2 promotes the dopaminergic differentiation of mouse neurons in culture RSPO2 increases dopaminergic neurogenesis and differentiation of human ESCs
Collapse
|
9
|
Alowolodu O, Johnson G, Alashwal L, Addou I, Zhdanova IV, Uversky VN. Intrinsic disorder in spondins and some of their interacting partners. INTRINSICALLY DISORDERED PROTEINS 2016; 4:e1255295. [PMID: 28232900 DOI: 10.1080/21690707.2016.1255295] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 10/22/2016] [Accepted: 10/27/2016] [Indexed: 12/28/2022]
Abstract
Spondins, which are proteins that inhibit and promote adherence of embryonic cells so as to aid axonal growth are part of the thrombospondin-1 family. Spondins function in several important biological processes, such as apoptosis, angiogenesis, etc. Spondins constitute a thrombospondin subfamily that includes F-spondin, a protein that interacts with Aβ precursor protein and inhibits its proteolytic processing; R-spondin, a 4-membered group of proteins that regulates Wnt pathway and have other functions, such as regulation of kidney proliferation, induction of epithelial proliferation, the tumor suppressant action; M-spondin that mediates mechanical linkage between the muscles and apodemes; and the SCO-spondin, a protein important for neuronal development. In this study, we investigated intrinsic disorder status of human spondins and their interacting partners, such as members of the LRP family, LGR family, Frizzled family, and several other binding partners in order to establish the existence and importance of disordered regions in spondins and their interacting partners by conducting a detailed analysis of their sequences, finding disordered regions, and establishing a correlation between their structure and biological functions.
Collapse
Affiliation(s)
- Oluwole Alowolodu
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida , Tampa, FL, USA
| | - Gbemisola Johnson
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida , Tampa, FL, USA
| | - Lamis Alashwal
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida , Tampa, FL, USA
| | - Iqbal Addou
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida , Tampa, FL, USA
| | - Irina V Zhdanova
- Department of Anatomy & Neurobiology, Boston University School of Medicine , Boston, MA, USA
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA; USF Health Byrd Alzheimer Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA; Laboratory of Structural Dynamics, Stability and Folding of Proteins, Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
10
|
Chai X, Frotscher M. How does Reelin signaling regulate the neuronal cytoskeleton during migration? NEUROGENESIS 2016; 3:e1242455. [PMID: 28265585 DOI: 10.1080/23262133.2016.1242455] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/15/2016] [Accepted: 09/25/2016] [Indexed: 01/17/2023]
Abstract
Neuronal migration is an essential step in the formation of laminated brain structures. In the developing cerebral cortex, pyramidal neurons migrate toward the Reelin-containing marginal zone. Reelin is an extracellular matrix protein synthesized by Cajal-Retzius cells. In this review, we summarize our recent results and hypotheses on how Reelin might regulate neuronal migration by acting on the actin and microtubule cytoskeleton. By binding to ApoER2 receptors on the migrating neurons, Reelin induces stabilization of the leading processes extending toward the marginal zone, which involves Dab1 phosphorylation, adhesion molecule expression, cofilin phosphorylation and inhibition of tau phosphorylation. By binding to VLDLR and integrin receptors, Reelin interacts with Lis1 and induces nuclear translocation, accompanied by the ubiquitination of phosphorylated Dab1. Eventually Reelin induces clustering of its receptors resulting in the endocytosis of a Reelin/receptor complex (particularly VLDLR). The resulting decrease in Reelin contributes to neuronal arrest at the marginal zone.
Collapse
Affiliation(s)
- Xuejun Chai
- Institute for Structural Neurobiology, Center for Molecular Neurobiology Hamburg (ZMNH) , Hamburg, Germany
| | - Michael Frotscher
- Institute for Structural Neurobiology, Center for Molecular Neurobiology Hamburg (ZMNH) , Hamburg, Germany
| |
Collapse
|
11
|
Majd S, Power JH, Grantham HJM. Neuronal response in Alzheimer's and Parkinson's disease: the effect of toxic proteins on intracellular pathways. BMC Neurosci 2015; 16:69. [PMID: 26499115 PMCID: PMC4619058 DOI: 10.1186/s12868-015-0211-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 10/13/2015] [Indexed: 01/09/2023] Open
Abstract
Accumulation of protein aggregates is the leading cause of cellular dysfunction in neurodegenerative disorders. Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease, Prion disease and motor disorders such as amyotrophic lateral sclerosis, present with a similar pattern of progressive neuronal death, nervous system deterioration and cognitive impairment. The common characteristic is an unusual misfolding of proteins which is believed to cause protein deposition and trigger degenerative signals in the neurons. A similar clinical presentation seen in many neurodegenerative disorders suggests the possibility of shared neuronal responses in different disorders. Despite the difference in core elements of deposits in each neurodegenerative disorder, the cascade of neuronal reactions such as activation of glycogen synthase kinase-3 beta, mitogen-activated protein kinases, cell cycle re-entry and oxidative stress leading to a progressive neurodegeneration are surprisingly similar. This review focuses on protein toxicity in two neurodegenerative diseases, AD and PD. We reviewed the activated mechanisms of neurotoxicity in response to misfolded beta-amyloid and α-synuclein, two major toxic proteins in AD and PD, leading to neuronal apoptosis. The interaction between the proteins in producing an overlapping pathological pattern will be also discussed.
Collapse
Affiliation(s)
- Shohreh Majd
- Centre for Neuroscience and Paramedic Unit, School of Medicine, Flinders University of South Australia, Adelaide, SA, 5042, Australia.
| | - John H Power
- Department of Human Physiology, School of Medicine, Flinders University of South Australia, Adelaide, SA, 5042, Australia.
| | - Hugh J M Grantham
- Centre for Neuroscience and Paramedic Unit, School of Medicine, Flinders University of South Australia, Adelaide, SA, 5042, Australia.
| |
Collapse
|
12
|
Hu H, Xin N, Liu J, Liu M, Wang Z, Wang W, Zhang Q, Qi J. Characterization of F-spondin in Japanese flounder (Paralichthys olivaceus) and its role in the nervous system development of teleosts. Gene 2015; 575:623-31. [PMID: 26390814 DOI: 10.1016/j.gene.2015.09.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 09/16/2015] [Indexed: 12/21/2022]
Abstract
F-spondin was originally isolated from the developing embryonic floor plate of vertebrates, secreting numerous kinds of neuron-related molecules. The protein performs a positive function in nervous system development, which is attributed to the high conservation of F-spondin protein, an extracellular matrix (ECM) protein in several species. However, its precise function remains unknown, especially in marine fish. In this study, the F-spondin of Japanese flounder (Paralichthys olivaceus). was cloned, and its expression pattern and structural characteristics were analyzed. The 2421bp-long cDNA ORF of PoF-spondin was obtained and divided into 14 exons spread over 61,496bp of the genomic sequence. Phylogenetic analysis showed that PoF-spondin was actually the ortholog of the human spon1 gene and shared high identities with other teleost spon1a genes. Quantitative RT-PCR analysis showed that PoF-spondin was maternally expressed, and transcripts were present from one-cell stage to hatching stage, peaking at tailbud stage. Tissue distribution analysis indicated that PoF-spondin was detectable mainly in the gonads (especially in the ovary) and the brain. Whole mount in situ hybridization analysis revealed that the PoF-spondin transcription distributed throughout the cleavage of the ball in the early stage and expressed at a high level in the floor plate of the trunk at tailbud and pre-hatching stages. Furthermore, the expression of genes related to nervous system development (spon1b, foxo3b, and foxj1a) was significantly increased after the injection of PoF-spondin into the embryos of wild-type zebrafish. Furthermore, PoF-spondin significantly suppressed the expression of the chordamesoderm marker gene ntl, increased the expression of otx2/krox20, ectoderm mark genes, and left the expression of dorsal mesodermal marker gene gsc unaffected at 50% epiboly stage in zebrafish. In short, our results suggest that PoF-spondin functions in the development of the teleost nervous system.
Collapse
Affiliation(s)
- Hongshuang Hu
- Key Laboratory of Marine Genetics and Breeding, Ocean University of China, 266003 Qingdao, Shandong, China
| | - Nian Xin
- Key Laboratory of Marine Genetics and Breeding, Ocean University of China, 266003 Qingdao, Shandong, China
| | - Jinxiang Liu
- Key Laboratory of Marine Genetics and Breeding, Ocean University of China, 266003 Qingdao, Shandong, China
| | - Mengmeng Liu
- Key Laboratory of Marine Genetics and Breeding, Ocean University of China, 266003 Qingdao, Shandong, China
| | - Zhenwei Wang
- Key Laboratory of Marine Genetics and Breeding, Ocean University of China, 266003 Qingdao, Shandong, China
| | - Wenji Wang
- Key Laboratory of Marine Genetics and Breeding, Ocean University of China, 266003 Qingdao, Shandong, China
| | - Quanqi Zhang
- Key Laboratory of Marine Genetics and Breeding, Ocean University of China, 266003 Qingdao, Shandong, China
| | - Jie Qi
- Key Laboratory of Marine Genetics and Breeding, Ocean University of China, 266003 Qingdao, Shandong, China.
| |
Collapse
|
13
|
Genome-wide scan of healthy human connectome discovers SPON1 gene variant influencing dementia severity. Proc Natl Acad Sci U S A 2013; 110:4768-73. [PMID: 23471985 DOI: 10.1073/pnas.1216206110] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Aberrant connectivity is implicated in many neurological and psychiatric disorders, including Alzheimer's disease and schizophrenia. However, other than a few disease-associated candidate genes, we know little about the degree to which genetics play a role in the brain networks; we know even less about specific genes that influence brain connections. Twin and family-based studies can generate estimates of overall genetic influences on a trait, but genome-wide association scans (GWASs) can screen the genome for specific variants influencing the brain or risk for disease. To identify the heritability of various brain connections, we scanned healthy young adult twins with high-field, high-angular resolution diffusion MRI. We adapted GWASs to screen the brain's connectivity pattern, allowing us to discover genetic variants that affect the human brain's wiring. The association of connectivity with the SPON1 variant at rs2618516 on chromosome 11 (11p15.2) reached connectome-wide, genome-wide significance after stringent statistical corrections were enforced, and it was replicated in an independent subsample. rs2618516 was shown to affect brain structure in an elderly population with varying degrees of dementia. Older people who carried the connectivity variant had significantly milder clinical dementia scores and lower risk of Alzheimer's disease. As a posthoc analysis, we conducted GWASs on several organizational and topological network measures derived from the matrices to discover variants in and around genes associated with autism (MACROD2), development (NEDD4), and mental retardation (UBE2A) significantly associated with connectivity. Connectome-wide, genome-wide screening offers substantial promise to discover genes affecting brain connectivity and risk for brain diseases.
Collapse
|
14
|
Grondona JM, Hoyo-Becerra C, Visser R, Fernández-Llebrez P, López-Ávalos MD. The subcommissural organ and the development of the posterior commissure. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2012; 296:63-137. [PMID: 22559938 DOI: 10.1016/b978-0-12-394307-1.00002-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Growing axons navigate through the developing brain by means of axon guidance molecules. Intermediate targets producing such signal molecules are used as guideposts to find distal targets. Glial, and sometimes neuronal, midline structures represent intermediate targets when axons cross the midline to reach the contralateral hemisphere. The subcommissural organ (SCO), a specialized neuroepithelium located at the dorsal midline underneath the posterior commissure, releases SCO-spondin, a large glycoprotein belonging to the thrombospondin superfamily that shares molecular domains with axonal pathfinding molecules. Several evidences suggest that the SCO could be involved in the development of the PC. First, both structures display a close spatiotemporal relationship. Second, certain mutants lacking an SCO present an abnormal PC. Third, some axonal guidance molecules are expressed by SCO cells. Finally, SCO cells, the Reissner's fiber (the aggregated form of SCO-spondin), or synthetic peptides from SCO-spondin affect the neurite outgrowth or neuronal aggregation in vitro.
Collapse
Affiliation(s)
- Jesús M Grondona
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Spain.
| | | | | | | | | |
Collapse
|
15
|
Akle V, Guelin E, Yu L, Brassard-Giordano H, Slack BE, Zhdanova IV. F-spondin/spon1b expression patterns in developing and adult zebrafish. PLoS One 2012; 7:e37593. [PMID: 22768035 PMCID: PMC3387172 DOI: 10.1371/journal.pone.0037593] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 04/26/2012] [Indexed: 01/24/2023] Open
Abstract
F-spondin, an extracellular matrix protein, is an important player in embryonic morphogenesis and CNS development, but its presence and role later in life remains largely unknown. We generated a transgenic zebrafish in which GFP is expressed under the control of the F-spondin (spon1b) promoter, and used it in combination with complementary techniques to undertake a detailed characterization of the expression patterns of F-spondin in developing and adult brain and periphery. We found that F-spondin is often associated with structures forming long neuronal tracts, including retinal ganglion cells, the olfactory bulb, the habenula, and the nucleus of the medial longitudinal fasciculus (nMLF). F-spondin expression coincides with zones of adult neurogenesis and is abundant in CSF-contacting secretory neurons, especially those in the hypothalamus. Use of this new transgenic model also revealed F-spondin expression patterns in the peripheral CNS, notably in enteric neurons, and in peripheral tissues involved in active patterning or proliferation in adults, including the endoskeleton of zebrafish fins and the continuously regenerating pharyngeal teeth. Moreover, patterning of the regenerating caudal fin following fin amputation in adult zebrafish was associated with F-spondin expression in the blastema, a proliferative region critical for tissue reconstitution. Together, these findings suggest major roles for F-spondin in the CNS and periphery of the developing and adult vertebrate.
Collapse
Affiliation(s)
- Veronica Akle
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Emmanuel Guelin
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Lili Yu
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Helena Brassard-Giordano
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Barbara E. Slack
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Irina V. Zhdanova
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
16
|
Cheng YC, Chen TA, Chen CY, Liang CM, Liang SM. 3'poly-G-tailed ODNs inhibit F-spondin to induce cell death and neurite retraction in rat embryonic neurons. Mol Neurobiol 2012; 45:536-49. [PMID: 22592270 DOI: 10.1007/s12035-012-8275-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2012] [Accepted: 05/01/2012] [Indexed: 10/28/2022]
Abstract
The effects and mechanism of action of oligodeoxyribonucleotides containing CpG motif (CpG-ODNs) on neuron cells are largely unexamined. Here, we found that CpG-A ODNs but not other types of CpG-ODNs induced neurite retraction and cell apoptosis of rat embryonic neurons in a TLR9-independent manner. These effects of CpG-A ODNs were primarily due to the poly-guanosine at the 3' terminus (3'G-ODNs). Pull-down analysis showed that 3'G-ODNs associated with transcription factor Y-BOX1 (YB-1) to facilitate the translocation of YB-1 into the nucleus via the nuclear localizing sequence of YB-1. YB-1 then interacted with the promoter of F-spondin directly at -45 and -1,375 sites as demonstrated by chromatin immunoprecipitation (ChIP) analysis. Binding of YB-1 to F-spondin promoter resulted in downregulation of F-spondin expression. Overexpression of F-spondin rescued the cell death and neurite retraction induced by 3'G-ODNs in embryonic neuron cells. Taken together, these findings suggest that 3'G-ODNs enhance nucleus YB-1 to inhibit F-spondin leading to cell death and neurite retraction of embryonic neuron cells.
Collapse
Affiliation(s)
- Yung-Chih Cheng
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
17
|
Herrera F, Maher P, Schubert D. c-Jun N-terminal kinase controls a negative loop in the regulation of glial fibrillary acidic protein expression by retinoic acid. Neuroscience 2012; 208:143-9. [PMID: 22387108 DOI: 10.1016/j.neuroscience.2012.02.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 01/31/2012] [Accepted: 02/08/2012] [Indexed: 11/26/2022]
Abstract
Glial fibrillary acidic protein (GFAP) is a protein widely used as a molecular marker for astroglial differentiation and mature astrocytes. We and others have shown previously that retinoic acid and specific cytokines induce the expression of GFAP in neural precursor cells by activating the phosphatidylinositol-4,5-bisphosphate-3-kinase (PI3K) phosphorylation pathway. Here, we extend our previous work and show that retinoic acid also activates specifically the c-Jun N-terminal kinase (JNK) phosphorylation pathway, which in turn inhibits GFAP expression. Our results suggest the existence of a negative self-regulatory loop in the phosphorylation pathways that regulates GFAP expression. This loop is constitutively repressed by the PI3K pathway. Our results could be relevant for disorders involving sustained GFAP overexpression in precursor cells, such as glioblastoma and Alexander disease.
Collapse
Affiliation(s)
- F Herrera
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
18
|
Tan K, Lawler J. The structure of the Ca²+-binding, glycosylated F-spondin domain of F-spondin - A C2-domain variant in an extracellular matrix protein. BMC STRUCTURAL BIOLOGY 2011; 11:22. [PMID: 21569239 PMCID: PMC3117680 DOI: 10.1186/1472-6807-11-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Accepted: 05/10/2011] [Indexed: 11/12/2022]
Abstract
Background F-spondin is a multi-domain extracellular matrix (ECM) protein and a contact-repellent molecule that directs axon outgrowth and cell migration during development. The reelin_N domain and the F-spondin domain (FS domain) comprise a proteolytic fragment that interacts with the cell membrane and guides the projection of commissural axons to floor plate. The FS domain is found in F-spondins, mindins, M-spondin and amphiF-spondin. Results We present the crystal structure of human F-spondin FS domain at 1.95Å resolution. The structure reveals a Ca2+-binding C2 domain variant with an 8-stranded antiparallel β-sandwich fold. Though the primary sequences of the FS domains of F-spondin and mindin are less than 36% identical, their overall structures are very similar. The unique feature of F-spondin FS domain is the presence of three disulfide bonds associated with the N- and C-termini of the domain and a highly conserved N-linked glycosylation site. The integrin-binding motif found in mindin is not conserved in the F-spondin FS domain. Conclusion The structure of the F-spondin FS domain completes the structural studies of the multiple-domain ECM molecule. The homology of its core structure to a common Ca2+- and lipid-binding C2 domain suggests that the F-spondin FS domain may be responsible for part of the membrane targeting of F-spondin in its regulation of axon development. The structural properties of the FS domain revealed in this study pave the way for further exploration into the functions of F-spondin.
Collapse
Affiliation(s)
- Kemin Tan
- Midwest Center for Structural Genomics and Structural Biology Center, Biosciences Division, Argonne National Laboratory, Argonne, IL 60439, USA.
| | | |
Collapse
|
19
|
Peterziel H, Sackmann T, Strelau J, Kuhn PH, Lichtenthaler SF, Marom K, Klar A, Unsicker K. F-spondin regulates neuronal survival through activation of disabled-1 in the chicken ciliary ganglion. Mol Cell Neurosci 2010; 46:483-97. [PMID: 21145970 DOI: 10.1016/j.mcn.2010.12.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Revised: 11/19/2010] [Accepted: 12/02/2010] [Indexed: 01/06/2023] Open
Abstract
The extracellular membrane-associated protein F-spondin has been implicated in cell-matrix and cell-cell adhesion and plays an important role in axonal pathfinding. We report here that F-spondin is expressed in non-neuronal cells in the embryonic chicken ciliary ganglion (CG) and robustly promotes survival of cultured CG neurons. Using deletion constructs of F-spondin we found that the amino-terminal Reelin/Spondin domain cooperates with thrombospondin type 1 repeat (TSR) 6, a functional TGFβ-activation domain. In ovo treatment with blocking antibodies raised against the Reelin/Spondin domain or the TSR-domains caused increased apoptosis of CG neurons during the phase of programmed cell death and loss of about 30% of the neurons compared to controls. The Reelin/Spondin domain receptor - APP and its downstream signalling molecule disabled-1 are expressed in CG neurons. F-spondin induced rapid phosphorylation of disabled-1. Moreover, both blocking the central APP domain and interference with disabled-1 signalling disrupted the survival promoting effect of F-spondin. Taken together, our data suggest that F-spondin can promote neuron survival by a mechanism involving the Reelin/Spondin and the TSR domains.
Collapse
Affiliation(s)
- H Peterziel
- Neuroanatomy & Interdisciplinary Center for Neurosiences (IZN), University of Heidelberg, Heidelberg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Herrera F, Chen Q, Schubert D. Synergistic effect of retinoic acid and cytokines on the regulation of glial fibrillary acidic protein expression. J Biol Chem 2010; 285:38915-22. [PMID: 20876578 DOI: 10.1074/jbc.m110.170274] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Glial fibrillary acidic protein (GFAP) is the main astroglial marker during astrogliogenesis, but it is also expressed in other cell types, including neural stem cells and old neurons. Activation of the JAK/STAT pathway by the IL-6 family of cytokines is the canonical pathway regulating GFAP expression, whereas retinoic acid is thought to be the only inducer of GFAP to operate independently of this pathway. Here, we show that retinoic acid receptor α not only links retinoic acid signaling to the canonical cytokine-stimulated pathway leading to GFAP expression but that it also plays a key role in the synergistic actions of retinoic acid and cytokines on this pathway. Cytokines both potentiate retinoic acid receptor α expression and enhance its binding to DNA and to the Stat3-p300/CBP-Smad transcriptional complex, the cornerstone of the canonical pathway. PI3K is upstream to all the key events leading to the expression of GFAP. Our results give new insights about the role of retinoic acid signaling in GFAP expression.
Collapse
Affiliation(s)
- Federico Herrera
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | | | | |
Collapse
|
21
|
Bataillé L, Delon I, Da Ponte JP, Brown NH, Jagla K. Downstream of identity genes: muscle-type-specific regulation of the fusion process. Dev Cell 2010; 19:317-28. [PMID: 20708593 PMCID: PMC3852356 DOI: 10.1016/j.devcel.2010.07.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2009] [Revised: 04/10/2010] [Accepted: 05/18/2010] [Indexed: 11/20/2022]
Abstract
In all metazoan organisms, the diversification of cell types involves determination of cell fates and subsequent execution of specific differentiation programs. During Drosophila myogenesis, identity genes specify the fates of founder myoblasts, from which derive all individual larval muscles. Here, to understand how cell fate information residing within founders is translated during differentiation, we focus on three identity genes, eve, lb, and slou, and how they control the size of individual muscles by regulating the number of fusion events. They achieve this by setting expression levels of Mp20, Pax, and mspo, three genes that regulate actin dynamics and cell adhesion and, as we show here, modulate the fusion process in a muscle-specific manner. Thus, these data show how the identity information implemented by transcription factors is translated via target genes into cell-type-specific programs of differentiation.
Collapse
Affiliation(s)
- L. Bataillé
- GReD - INSERM U931, CNRS UMR6247, Clermont Université, France
| | - I. Delon
- Gurdon Institute and Dept of Physiology, Development and Neuroscience, University of Cambridge, UK
| | - JP. Da Ponte
- GReD - INSERM U931, CNRS UMR6247, Clermont Université, France
| | - N. H. Brown
- Gurdon Institute and Dept of Physiology, Development and Neuroscience, University of Cambridge, UK
| | - K. Jagla
- GReD - INSERM U931, CNRS UMR6247, Clermont Université, France
| |
Collapse
|
22
|
Du J, Takeuchi H, Leonhard-Melief C, Shroyer KR, Dlugosz M, Haltiwanger RS, Holdener BC. O-fucosylation of thrombospondin type 1 repeats restricts epithelial to mesenchymal transition (EMT) and maintains epiblast pluripotency during mouse gastrulation. Dev Biol 2010; 346:25-38. [PMID: 20637190 DOI: 10.1016/j.ydbio.2010.07.008] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Revised: 06/09/2010] [Accepted: 07/07/2010] [Indexed: 10/19/2022]
Abstract
Thrombospondin type 1 repeat (TSR) superfamily members regulate diverse biological activities ranging from cell motility to inhibition of angiogenesis. In this study, we verified that mouse protein O-fucosyltransferase-2 (POFUT2) specifically adds O-fucose to TSRs. Using two Pofut2 gene-trap lines, we demonstrated that O-fucosylation of TSRs was essential for restricting epithelial to mesenchymal transition in the primitive streak, correct patterning of mesoderm, and localization of the definitive endoderm. Although Pofut2 mutant embryos established anterior/posterior polarity, they underwent extensive mesoderm differentiation at the expense of maintaining epiblast pluripotency. Moreover, mesoderm differentiation was biased towards the vascular endothelial cell lineage. Localization of Foxa2 and Cer1 expressing cells within the interior of Pofut2 mutant embryos suggested that POFUT2 activity was also required for the displacement of the primitive endoderm by definitive endoderm. Notably, Nodal, BMP4, Fgf8, and Wnt3 expression were markedly elevated and expanded in Pofut2 mutants, providing evidence that O-fucose modification of TSRs was essential for modulation of growth factor signaling during gastrulation. The ability of Pofut2 mutant embryos to form teratomas comprised of tissues from all three germ layer origins suggested that defects in Pofut2 mutant embryos resulted from abnormalities in the extracellular environment. This prediction is consistent with the observation that POFUT2 targets are constitutive components of the extracellular matrix (ECM) or associate with the ECM. For this reason, the Pofut2 mutants represent a valuable tool for studying the role of O-fucosylation in ECM synthesis and remodeling, and will be a valuable model to study how post-translational modification of ECM components regulates the formation of tissue boundaries, cell movements, and signaling.
Collapse
Affiliation(s)
- Jianguang Du
- Department of Biochemistry and Cell Biology, Institute for Cell and Developmental Biology, Center for Developmental Genetics, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Cheng YC, Liang CM, Chen YP, Tsai IH, Kuo CC, Liang SM. F-spondin plays a critical role in murine neuroblastoma survival by maintaining IL-6 expression. J Neurochem 2009; 110:947-55. [PMID: 19549008 DOI: 10.1111/j.1471-4159.2009.06186.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
F-spondin is associated with the regulation of axonal growth and the development of the nervous system. Its mechanism of action, however, is not clearly understood. In this study, we found that murine neuroblastoma Neuro-2a cells expressed a significant level of IL-6, but only trace amounts of IL-12, tumor necrosis factor alpha and nitric oxide. Knock-down of F-spondin mRNA in murine neuroblastoma NB41A3 and Neuro-2a cells using small interfering RNAs led to decreased IL-6 levels along with lower resistance to serum starvation and cytotoxic amyloid beta(1-42) (Abeta(1-42)) peptide. Restoring decline of F-spondin or IL-6 induced by F-spondin knock-down through adding exogenous F-spondin, IL-6 or over-expressing F-spondin reversed the cell death induced by Abeta(1-42) peptide or serum starvation. The decrease of IL-6 level was positively correlated with decrease of NF-kappaB and inhibition of p38 mitogen-activated protein kinase (MAPK). Over-expressing MEKK, a kinase activator of the p38 MAPK pathway, increased IL-6 production, restored the decrease of p38 induced by F-spondin knock-down, and rescued the cells from death caused by Abeta(1-42) peptide. Taken together, these results suggest that F-spondin may play a critical role in murine neuroblastoma survival under adverse conditions by maintaining IL-6 level via a MEKK/p38 MAPK/NF-kappaB-dependent pathway.
Collapse
Affiliation(s)
- Yung-Chih Cheng
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
24
|
Herrera F, Chen Q, Fischer WH, Maher P, Schubert D. Synaptojanin-1 plays a key role in astrogliogenesis: possible relevance for Down's syndrome. Cell Death Differ 2009; 16:910-20. [PMID: 19282871 PMCID: PMC2807404 DOI: 10.1038/cdd.2009.24] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
There is increasing interest in gliogenesis as the relevance of glia to both brain development and pathology becomes better understood. However, little is known about this process. The use of multidimensional protein identification technology (MudPIT) to identify changes in phosphoprotein levels in rat neural precursor cells treated with cytokines or retinoic acid showed that phosphorylation of the catalytic subunit of phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K p110alpha) and dephosphorylation of the inositol phosphatase synaptojanin-1 were common to the gliogenic stimuli. Although PI3K was found to be involved in both neuro- and astrogliogenesis, synaptojanin-1 was specifically involved in astrogliogenesis of neural precursor cells. The role of synaptojanin-1 in astrogliogenesis was further confirmed by analysis of neuron- and glia-specific markers in synaptojanin-1 knockout mouse brain. Additional experiments showed that the Sac1-like phosphatase domain of synaptojanin-1 is responsible for the observed astrogliogenic effect. Our results strongly indicate that phosphatidylinositol metabolism plays a key role in astrogliogenesis. The relevance of our findings for Down's syndrome pathology is discussed.
Collapse
Affiliation(s)
- Federico Herrera
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California, USA
| | - Qi Chen
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California, USA
| | - Wolfgang H. Fischer
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California, USA
| | - Pamela Maher
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California, USA
| | - David Schubert
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California, USA
| |
Collapse
|
25
|
Tan K, Duquette M, Liu JH, Lawler J, Wang JH. The crystal structure of the heparin-binding reelin-N domain of f-spondin. J Mol Biol 2008; 381:1213-23. [PMID: 18602404 PMCID: PMC2561254 DOI: 10.1016/j.jmb.2008.06.045] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2008] [Revised: 06/10/2008] [Accepted: 06/18/2008] [Indexed: 01/07/2023]
Abstract
The extracellular matrix protein F-spondin mediates axon guidance during neuronal development. Its N-terminal domain, termed the reelin-N domain, is conserved in F-spondins, reelins, and other extracellular matrix proteins. In this study, a recombinant human reelin-N domain has been expressed, purified, and shown to bind heparin. The crystal structure of the reelin-N domain resolved to 2.0 A reveals a variant immunoglobulin-like fold and potential heparin-binding sites. Substantial conformational variations even in secondary structure are observed between the two chemically identical reelin-N domains in one crystallographic asymmetric unit. The variations may result from extensive, highly specific interactions across the interface of the two reelin-N domains. The calculated values of buried surface area and the interface's shape complementarity are consistent with the formation of a weak dimer. The homophilic asymmetric dimer can potentially offer advantages in binding to ligands such as glycosaminoglycans, which may, in turn, bridge the two reelin-N domains and stabilize the dimer.
Collapse
Affiliation(s)
- Kemin Tan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.
| | | | | | | | | |
Collapse
|
26
|
Woo WM, Berry E, Hudson ML, Swale RE, Goncharov A, Chisholm AD. The C. elegans F-spondin family protein SPON-1 maintains cell adhesion in neural and non-neural tissues. Development 2008; 135:2747-2756. [PMID: 18614580 PMCID: PMC2633776 DOI: 10.1242/dev.015289] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The F-spondin family of extracellular matrix proteins has been implicated in axon outgrowth, fasciculation and neuronal cell migration, as well as in the differentiation and proliferation of non-neuronal cells. In screens for mutants defective in C. elegans embryonic morphogenesis, we identified SPON-1, the only C. elegans member of the spondin family. SPON-1 is synthesized in body muscles and localizes to integrin-containing structures on body muscles and to other basement membranes. SPON-1 maintains strong attachments of muscles to epidermis; in the absence of SPON-1, muscles progressively detach from the epidermis, causing defective epidermal elongation. In animals with reduced integrin function, SPON-1 becomes dose dependent, suggesting that SPON-1 and integrins function in concert to promote the attachment of muscles to the basement membrane. Although spon-1 mutants display largely normal neurite outgrowth, spon-1 synergizes with outgrowth defective mutants, revealing a cryptic role for SPON-1 in axon extension. In motoneurons, SPON-1 acts in axon guidance and fasciculation, whereas in interneurons SPON-1 maintains process position. Our results show that a spondin maintains cell-matrix adhesion in multiple tissues.
Collapse
Affiliation(s)
- Wei-Meng Woo
- Department of Molecular, Cell, and Developmental Biology, Sinsheimer Laboratories, University of California, Santa Cruz, CA 95064
| | - Emily Berry
- Department of Molecular, Cell, and Developmental Biology, Sinsheimer Laboratories, University of California, Santa Cruz, CA 95064
- Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093
| | - Martin L Hudson
- Department of Molecular, Cell, and Developmental Biology, Sinsheimer Laboratories, University of California, Santa Cruz, CA 95064
| | - Ryann E Swale
- Department of Molecular, Cell, and Developmental Biology, Sinsheimer Laboratories, University of California, Santa Cruz, CA 95064
| | - Alexandr Goncharov
- Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093
- Howard Hughes Medical Institute
| | - Andrew D Chisholm
- Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093
| |
Collapse
|
27
|
Regulated proteolysis of APP and ApoE receptors. Mol Neurobiol 2008; 37:64-72. [PMID: 18415033 DOI: 10.1007/s12035-008-8017-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2008] [Accepted: 03/24/2008] [Indexed: 10/22/2022]
Abstract
The beta-amyloid precursor protein (APP) shares intracellular and extracellular-binding partners with the family of receptors for apolipoprotein E (apoE). Binding of APP and apoE receptors to specific extracellular matrix proteins (F-spondin and Reelin) promotes their presence on the cell surface and influences whether they will interact with specific cytoplasmic adaptor proteins. Cleavage of APP and apoE receptors at the cell surface occurs by alpha-secretase activities; thus, the processing of these proteins can be regulated by their trafficking either to or from the cell surface. Their cleavages can also be regulated by tissue inhibitor of metalloproteinase-3 (TIMP-3), a metalloprotease inhibitor in the extracellular matrix. ApoE receptors have functions in neuronal migration during development and in proper synaptic function in the adult. Thus, the functions of apoE receptors and by analogy of APP will be modified by the various extracellular and intracellular interactions reviewed in this paper.
Collapse
|
28
|
Svensson P, Williams C, Lundeberg J, Rydén P, Bergqvist I, Edlund H. Gene array identification of Ipf1/Pdx1-/- regulated genes in pancreatic progenitor cells. BMC DEVELOPMENTAL BIOLOGY 2007; 7:129. [PMID: 18036209 PMCID: PMC2212654 DOI: 10.1186/1471-213x-7-129] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2007] [Accepted: 11/23/2007] [Indexed: 01/29/2023]
Abstract
Background The homeodomain transcription factor IPF1/PDX1 exerts a dual role in the pancreas; Ipf1/Pdx1 global null mutants fail to develop a pancreas whereas conditional inactivation of Ipf1/Pdx1 in β-cells leads to impaired β-cell function and diabetes. Although several putative target genes have been linked to the β-cell function of Ipf1/Pdx1, relatively little is known with respect to genes regulated by IPF1/PDX1 in early pancreatic progenitor cells. Results Microarray analyses identified a total of 111 genes that were differentially expressed in e10.5 pancreatic buds of Ipf1/Pdx1-/- embryos. The expression of one of these, Spondin 1, which encodes an extracellular matrix protein, has not previously been described in the pancreas. Quantitative real-time RT-PCR analyses and immunohistochemical analyses also revealed that the expression of FgfR2IIIb, that encodes the receptor for FGF10, was down-regulated in Ipf1/Pdx1-/- pancreatic progenitor cells. Conclusion This microarray analysis has identified a number of candidate genes that are differentially expressed in Ipf1/Pdx1-/- pancreatic buds. Several of the differentially expressed genes were known to be important for pancreatic progenitor cell proliferation and differentiation whereas others have not previously been associated with pancreatic development.
Collapse
Affiliation(s)
- Per Svensson
- Umeå Center for Molecular Medicine, Umeå University, SE-901 87 Umeå, Sweden.
| | | | | | | | | | | |
Collapse
|
29
|
Xiao Y, Fu H, Prasadam I, Yang YC, Hollinger JO. Gene expression profiling of bone marrow stromal cells from juvenile, adult, aged and osteoporotic rats: with an emphasis on osteoporosis. Bone 2007; 40:700-15. [PMID: 17166785 DOI: 10.1016/j.bone.2006.10.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2006] [Revised: 10/24/2006] [Accepted: 10/24/2006] [Indexed: 10/23/2022]
Abstract
PURPOSE Osteoporosis is a multi-factorial, age-related disease with a complex etiology and mode of regulation involving a large numbers of genes. To better understand the possible relationships among genes, we fingerprinted genes in a rat model induced by ovariectomy to determine differences among osteoporotic, non-osteoporotic, aged and juvenile rats. METHODS We applied genome wide cDNA microarray technology to analyze genes expressed in bone marrow mesenchymal stromal cells (BMSC) and compared non-osteoporotic adult vs. osteoporotic, non-osteoporotic adult vs. aged, and non-osteoporotic adult vs. juvenile. Rigorous statistical analysis of functional annotation (EASE program) identified over-represented biological and molecular functions with significant group wide changes (p< or =0.05). Some of the expressed genes were further confirmed by quantitative RT-PCR (reverse transcription-polymerase chain reaction). RESULTS Differences in gene expression were observed by identifying transcripts selected by t-test that were consistently changed by a minimum of two-fold. There were 195 transcripts that showed an increased expression and 109 transcripts that showed decreased expression relative to the osteoporotic condition. Of these, 75% transcripts were unknown gene products or ESTs (expressed sequence tag). A number of genes found in the aged and juvenile groups were not present in the osteoporotic rats. Functional clustering of the genes using the EASE bioinformatics program revealed that transcripts in osteoporosis were associated with signal transduction, lipid metabolism, protein metabolism, ionic and protein transport, neuropeptide and G protein signaling pathways. Although some of the genes have previously been shown to play a key role in osteoporosis, several genes were uniquely identified in this study and likely play a role in developing aged related osteoporosis that could have compelling implications in the development of new diagnostic strategies and therapeutics for osteoporosis. CONCLUSIONS These data suggest that osteoporosis is associated with changes of multiple novel gene expression and that numerous pathways could play important roles in osteoporosis pathogenesis.
Collapse
Affiliation(s)
- Yin Xiao
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.
| | | | | | | | | |
Collapse
|