1
|
Catenacci RB, Galleguillos D, Rhodes A, Phillips S, Calabresi PA. MHC class I and II expression and induction in oligodendrocytes varies with age. Sci Rep 2025; 15:18309. [PMID: 40419671 PMCID: PMC12106765 DOI: 10.1038/s41598-025-03089-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 05/19/2025] [Indexed: 05/28/2025] Open
Abstract
Oligodendroglia expressing major histocompatibility complex (MHC) molecules have become of central interest in multiple sclerosis (MS) research, but their role in aging is still being elucidated. Using MHC class I and II reporter mice, we compared oligodendroglial MHC expression in neonatal, young adult (8-16 weeks), and aging (52 weeks) mice at baseline and after stereotactic delivery of AAV-GFAP-IFNγ. We found that MHC class I-expressing oligodendroglia were present in both the naïve brain and spinal cord, while MHC class II-expressing oligodendroglia were seen only in the spinal cord of aging mice at baseline. After AAV injection, MHC expression in oligodendroglia increased, recapitulating pathology seen in MS mouse models and in MS post-mortem tissue. After IFNγ-AAV injection, the abundance of MHC class I-expressing oligodendroglia did not vary with age, whereas MHC class II-expressing oligodendroglia were more abundant in aging mice than neonatal and young adult mice. Our results suggest that MHC class II-expressing oligodendroglia are more prevalent with aging, which may contribute to the mechanisms underlying progressive MS and other age-related neurodegenerative diseases with central nervous system inflammation.
Collapse
Affiliation(s)
- Riley B Catenacci
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Danny Galleguillos
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Adriana Rhodes
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Sloan Phillips
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Peter A Calabresi
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| |
Collapse
|
2
|
Zheng Z, Chen X, Zheng R, Yan Z, Li L, Chen R, Li L, Liu Y, Guan Y, Zhu H. Progressive Adaptation of Subtype H6N1 Avian Influenza Virus in Taiwan Enhances Mammalian Infectivity, Pathogenicity, and Transmissibility. Viruses 2025; 17:733. [PMID: 40431744 PMCID: PMC12115762 DOI: 10.3390/v17050733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2025] [Revised: 05/15/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
The interspecies transmission of avian influenza viruses remains a significant public health concern. H6 viruses have gained attention following the first human infection by a chicken-origin H6N1 virus (A/Taiwan/02/2013, Hu/13), highlighting their zoonotic potential. To understand the evolutionary trajectory and mammalian adaptation of this Taiwan lineage, we compared two avian isolates (A/Chicken/Taiwan/CF19/2009, Ck/09; A/Chicken/Taiwan/2267/2012, Ck/12) and Hu/13 in vitro and in vivo. Hu/13 exhibited enhanced replication in MDCK cells, producing larger plaques and higher viral titers than Ck/09 and Ck/12. In BALB/c mice, Hu/13 demonstrated the highest pathogenicity and mortality, followed by Ck/12, while Ck/09 induced minimal morbidity. Hu/13 and Ck/12 replicated efficiently in respiratory tissues, eliciting robust cytokine responses and severe pulmonary lesions. In ferrets, Hu/13 showed relatively efficient transmission, infecting all direct physical-contact and two out of three airborne-contact ferrets, whereas Ck/09 failed to transmit. Histopathology confirmed escalating lung pathology from Ck/09 to Ck/12 and Hu/13. Whole-genome sequencing identified adaptive mutations in Hu/13 during ferret replication, though no canonical mammalian-adaptive changes (e.g., PB2-E627K or HA-Q226L) were detected. These findings demonstrate progressive mammalian adaptation, replication efficiency, and transmissibility within the Taiwan H6N1 lineage. Enhanced surveillance is crucial to monitor mammalian-adaptive mutations, informing pandemic preparedness and public health strategies.
Collapse
Affiliation(s)
- Zuoyi Zheng
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases, Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases (Key Laboratory of Ministry of Education), Joint Institute of Virology (Shantou University-The University of Hong Kong), Shantou University Medical College, Shantou University, Shantou 515063, China
- State Key Laboratory of Emerging Infectious Diseases (SKLEID), School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Xifeng Chen
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases, Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases (Key Laboratory of Ministry of Education), Joint Institute of Virology (Shantou University-The University of Hong Kong), Shantou University Medical College, Shantou University, Shantou 515063, China
| | - Rutian Zheng
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases, Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases (Key Laboratory of Ministry of Education), Joint Institute of Virology (Shantou University-The University of Hong Kong), Shantou University Medical College, Shantou University, Shantou 515063, China
| | - Zhigang Yan
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases, Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases (Key Laboratory of Ministry of Education), Joint Institute of Virology (Shantou University-The University of Hong Kong), Shantou University Medical College, Shantou University, Shantou 515063, China
| | - Long Li
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases, Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases (Key Laboratory of Ministry of Education), Joint Institute of Virology (Shantou University-The University of Hong Kong), Shantou University Medical College, Shantou University, Shantou 515063, China
| | - Rirong Chen
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases, Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases (Key Laboratory of Ministry of Education), Joint Institute of Virology (Shantou University-The University of Hong Kong), Shantou University Medical College, Shantou University, Shantou 515063, China
- State Key Laboratory of Emerging Infectious Diseases (SKLEID), School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Lifeng Li
- State Key Laboratory of Emerging Infectious Diseases (SKLEID), School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yongmei Liu
- State Key Laboratory of Emerging Infectious Diseases (SKLEID), School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yi Guan
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases, Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases (Key Laboratory of Ministry of Education), Joint Institute of Virology (Shantou University-The University of Hong Kong), Shantou University Medical College, Shantou University, Shantou 515063, China
- State Key Laboratory of Emerging Infectious Diseases (SKLEID), School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Huachen Zhu
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases, Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases (Key Laboratory of Ministry of Education), Joint Institute of Virology (Shantou University-The University of Hong Kong), Shantou University Medical College, Shantou University, Shantou 515063, China
- State Key Laboratory of Emerging Infectious Diseases (SKLEID), School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
3
|
Vos S, Portillo JAC, Hubal A, Bapputty R, Pfaff A, Aaron R, Weng M, Sun D, Lu ZR, Yu JS, Subauste CS. CD40 Induces Unfolded Protein Response, Upregulation of VEGF, and Vascular Leakage in Diabetic Retinopathy. Diabetes 2025; 74:798-811. [PMID: 39976665 PMCID: PMC12012590 DOI: 10.2337/db23-0538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 02/16/2025] [Indexed: 02/26/2025]
Abstract
The unfolded protein response (UPR) drives events that promote diabetic retinopathy, including vascular endothelial growth factor (VEGF) upregulation in Müller cells. How UPR is activated in vivo in the diabetic retina is not well understood. CD40 is required for development of diabetic retinopathy, but whether CD40 mediates activation of UPR sensors is unknown. CD40 ligation in Müller cells caused phospholipase Cγ1 (PLCγ1)-dependent activation of UPR sensors (PERK, IRE1α, and ATF6α) and VEGF production dependent on PLCγ1 and UPR sensors. Diabetic Cd40-/- mice did not exhibit UPR activation or VEGF upregulation in the retina. These responses were restored in diabetic Cd40-/- mice rescued to express wild-type CD40 in Müller cells but not in mice rescued to express a CD40 mutation unable to recruit TRAF2/3. Intravitreal administration of a cell-permeable CD40-TRAF2/3-disrupting peptide reduced UPR activation, VEGF upregulation, and vascular leakage in diabetic mice. CD40 and TRAF2 in Müller cells from patients with diabetic retinopathy colocalized with activated UPR sensors and VEGF. Our study indicates that CD40 (via TRAF2/3 signaling) is an inducer of UPR activation that triggers VEGF production in Müller cells. This work uncovered inhibition of CD40-TRAF2/3 signaling as a potential approach to impair UPR activation, VEGF upregulation, and vascular leakage in diabetic retinopathy. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Sarah Vos
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH
| | - Jose-Andres C. Portillo
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH
| | - Alyssa Hubal
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH
- Department of Pathology, Case Western Reserve University, Cleveland, OH
| | - Reena Bapputty
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH
| | - Amelia Pfaff
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH
| | - Rachel Aaron
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH
| | - Matthew Weng
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH
| | - Da Sun
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH
| | - Zheng-Rong Lu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH
| | - Jin-Sang Yu
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH
| | - Carlos S. Subauste
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH
- Department of Pathology, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
4
|
Pereyra G, Mateo MI, Miaja P, Martin-Bermejo MJ, Martinez-Baños M, Klaassen R, Gruart A, Rueda-Carrasco J, Fernández-Rodrigo A, López-Merino E, Esteve P, Esteban JA, Smit AB, Delgado-García JM, Bovolenta P. SFRP1 upregulation causes hippocampal synaptic dysfunction and memory impairment. Cell Rep 2025; 44:115535. [PMID: 40198223 DOI: 10.1016/j.celrep.2025.115535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/30/2025] [Accepted: 03/17/2025] [Indexed: 04/10/2025] Open
Abstract
Impaired neuronal and synaptic function are hallmarks of early Alzheimer's disease (AD), preceding other neuropathological traits and cognitive decline. We previously showed that SFRP1, a glial-derived protein elevated in AD brains from preclinical stages, contributes to disease progression, implicating glial factors in early pathogenesis. Here, we generate and analyze transgenic mice overexpressing astrocytic SFRP1. SFRP1 accumulation causes early dendritic and synaptic defects in adult mice, followed by impaired synaptic long-term potentiation and cognitive decline, evident only when the animals age, thereby mimicking AD's structural-functional temporal distinction. This phenotype correlates with proteomic changes, including increased structural synaptic proteins like neurexin, which localizes in close proximity with SFRP1 in cultured hippocampal neurons. We conclude that excessive SFRP1 hinders synaptic protein turnover, reducing synaptic plasticity-a mechanism that may underlie the synaptopathy observed in the brains of prodromal AD patients.
Collapse
Affiliation(s)
- Guadalupe Pereyra
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| | - María Inés Mateo
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| | - Pablo Miaja
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| | - María Jesús Martin-Bermejo
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| | - Marcos Martinez-Baños
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| | - Remco Klaassen
- Center for Neurogenomics and Cognitive Research, VU University Amsterdam, 1081 Amsterdam, the Netherlands
| | - Agnès Gruart
- División de Neurociencias, Universidad Pablo de Olavide, 41013 Seville, Spain
| | - Javier Rueda-Carrasco
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| | - Alba Fernández-Rodrigo
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Esperanza López-Merino
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Pilar Esteve
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| | - José A Esteban
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - August B Smit
- Center for Neurogenomics and Cognitive Research, VU University Amsterdam, 1081 Amsterdam, the Netherlands
| | | | - Paola Bovolenta
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), 28029 Madrid, Spain.
| |
Collapse
|
5
|
Gruol DL. The Neuroimmune System and the Cerebellum. CEREBELLUM (LONDON, ENGLAND) 2024; 23:2511-2537. [PMID: 37950146 PMCID: PMC11585519 DOI: 10.1007/s12311-023-01624-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 10/20/2023] [Indexed: 11/12/2023]
Abstract
The recognition that there is an innate immune system of the brain, referred to as the neuroimmune system, that preforms many functions comparable to that of the peripheral immune system is a relatively new concept and much is yet to be learned. The main cellular components of the neuroimmune system are the glial cells of the brain, primarily microglia and astrocytes. These cell types preform many functions through secretion of signaling factors initially known as immune factors but referred to as neuroimmune factors when produced by cells of the brain. The immune functions of glial cells play critical roles in the healthy brain to maintain homeostasis that is essential for normal brain function, to establish cytoarchitecture of the brain during development, and, in pathological conditions, to minimize the detrimental effects of disease and injury and promote repair of brain structure and function. However, dysregulation of this system can occur resulting in actions that exacerbate or perpetuate the detrimental effects of disease or injury. The neuroimmune system extends throughout all brain regions, but attention to the cerebellar system has lagged that of other brain regions and information is limited on this topic. This article is meant to provide a brief introduction to the cellular and molecular components of the brain immune system, its functions, and what is known about its role in the cerebellum. The majority of this information comes from studies of animal models and pathological conditions, where upregulation of the system facilitates investigation of its actions.
Collapse
Affiliation(s)
- Donna L Gruol
- Neuroscience Department, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
6
|
Richard SA. The pivotal role of autophagy in the pathogenesis and therapy of medulloblastoma. Future Oncol 2024; 20:3313-3324. [PMID: 39513232 PMCID: PMC11633412 DOI: 10.1080/14796694.2024.2420629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 10/21/2024] [Indexed: 11/15/2024] Open
Abstract
Medulloblastoma (MB) is the most frequent malignant brain tumor in children. MB originates from neural precursor cells in distinctive regions of the rhombic lip and their maturation occurs in the cerebellum or the brain stem during embryonal development. Autophagy is also referred to as self-eating' which is a catabolic process that often triggers cellular homeostasis through the salvaging of degenerated proteins as well as organelles. Autophagy influence cell survival via aberrant proteins that could accumulate within the cell and influence potential signaling and transport mechanisms. The role of autophagy in MB aggressiveness as well as tumorigenesis is a very complex process. This review targets specifically data reporting the key roles of autophagy in the pathogenesis and therapy of MB.
Collapse
Affiliation(s)
- Seidu A. Richard
- Department of Biochemistry and Forensic Sciences, School of Chemistry and Biochemical Science, C. K. Tedam University of Technology and Applied Sciences, P. O. Box 24, Navrongo, Ghana
- Institute of Neuroscience, Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052,China
| |
Collapse
|
7
|
Ren P, Yu J, Wang D, Zeng L, Zhang X, Liu X, Cao Y, Hu Z, Zhao X, Yang K. Newcastle disease virus promotes pyroptosis in medulloblastoma cells by regulating interferon-gamma-mediated guanylate-binding protein 1 expression and activating caspase-4. Cytojournal 2024; 21:39. [PMID: 39563668 PMCID: PMC11574683 DOI: 10.25259/cytojournal_39_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/23/2024] [Indexed: 11/21/2024] Open
Abstract
Objective The literature has reported that Newcastle disease virus (NDV) can have inhibitory effects on various tumors. This study aims to investigate the mechanism by which NDV induces pyroptosis in medulloblastoma (MB) cells. Material and Methods We treated MB cell lines Daoy and D283 with NDV or recombinant interferon-gamma (IFN-g) proteins. Guanylate-binding proteins (GBPs) were measured using real-time quantitative polymerase chain reaction. Small interfering RNA-specific targeting GBP1 was transfected into MB cells. Apoptosis was assessed using Terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nucleoside nick end labeling and flow cytometry assays. Pyroptosis-related proteins, including caspase-4, caspase-1, and gasdermin D (GSDMD), were detected using Western blotting. Results Bioinformatics analysis revealed that GBP family genes and interferon-related genes might be responsive to NDV stimulation in MB cells. Treatment with NDV resulted in increased IFN-g levels and upregulated GBP expression, particularly GBP1. In addition, IFN-g treatment induced GBP1 expression and enhanced cell apoptosis. GBP1 knockdown attenuated the decreased cell proliferation and increased cell apoptosis induced by NDV in MB cells. GBP1 overexpression upregulated the expression of pyroptosis-related proteins, including caspase-4, caspase-1, and GSDMD, subsequently leading to inhibition of cell proliferation and an increase in cell apoptosis levels. The silencing of caspase-4 confirmed the regulatory role of GBP1 in MB cell pyroptosis. Conclusion Our findings suggest that NDV elevates IFN-g and GBP1 expression in MB cells, potentially contributing to caspase-4-mediated pyroptosis activation.
Collapse
Affiliation(s)
- Pengwu Ren
- Department of Neurosurgery, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong
| | - Jiayan Yu
- Department of Neurosurgery, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong
| | - Dongxiang Wang
- Department of Dermatology, The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lijuan Zeng
- Department of Neurosurgery, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong
| | - Xianqiang Zhang
- Department of Neurosurgery, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong
| | - Xiaohe Liu
- Department of Neurosurgery, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong
| | - Yongfu Cao
- Department of Neurosurgery, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong
| | - Zijian Hu
- Department of Neurosurgery, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong
| | - Xiaoyong Zhao
- Department of Neurosurgery, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong
| | - Kongbin Yang
- Department of Neurosurgery, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong
| |
Collapse
|
8
|
Packer D, Fresenko EE, Harrington EP. Remyelination in animal models of multiple sclerosis: finding the elusive grail of regeneration. Front Mol Neurosci 2023; 16:1207007. [PMID: 37448959 PMCID: PMC10338073 DOI: 10.3389/fnmol.2023.1207007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 06/09/2023] [Indexed: 07/18/2023] Open
Abstract
Remyelination biology and the therapeutic potential of restoring myelin sheaths to prevent neurodegeneration and disability in multiple sclerosis (MS) has made considerable gains over the past decade with many regeneration strategies undergoing tested in MS clinical trials. Animal models used to investigate oligodendroglial responses and regeneration of myelin vary considerably in the mechanism of demyelination, involvement of inflammatory cells, neurodegeneration and capacity for remyelination. The investigation of remyelination in the context of aging and an inflammatory environment are of considerable interest for the potential translation to progressive multiple sclerosis. Here we review how remyelination is assessed in mouse models of demyelination, differences and advantages of these models, therapeutic strategies that have emerged and current pro-remyelination clinical trials.
Collapse
|
9
|
Yao B, Delaidelli A, Vogel H, Sorensen PH. Pediatric Brain Tumours: Lessons from the Immune Microenvironment. Curr Oncol 2023; 30:5024-5046. [PMID: 37232837 PMCID: PMC10217418 DOI: 10.3390/curroncol30050379] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/01/2023] [Accepted: 05/10/2023] [Indexed: 05/27/2023] Open
Abstract
In spite of recent advances in tumour molecular subtyping, pediatric brain tumours (PBTs) remain the leading cause of cancer-related deaths in children. While some PBTs are treatable with favourable outcomes, recurrent and metastatic disease for certain types of PBTs remains challenging and is often fatal. Tumour immunotherapy has emerged as a hopeful avenue for the treatment of childhood tumours, and recent immunotherapy efforts have been directed towards PBTs. This strategy has the potential to combat otherwise incurable PBTs, while minimizing off-target effects and long-term sequelae. As the infiltration and activation states of immune cells, including tumour-infiltrating lymphocytes and tumour-associated macrophages, are key to shaping responses towards immunotherapy, this review explores the immune landscape of the developing brain and discusses the tumour immune microenvironments of common PBTs, with hopes of conferring insights that may inform future treatment design.
Collapse
Affiliation(s)
- Betty Yao
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada; (B.Y.)
| | - Alberto Delaidelli
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada; (B.Y.)
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Hannes Vogel
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Poul H. Sorensen
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada; (B.Y.)
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
10
|
Holloway RK, Zhang L, Molina-Gonzalez I, Ton K, Nicoll JAR, Boardman JP, Liang Y, Williams A, Miron VE. Localized microglia dysregulation impairs central nervous system myelination in development. Acta Neuropathol Commun 2023; 11:49. [PMID: 36949514 PMCID: PMC10035254 DOI: 10.1186/s40478-023-01543-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/03/2023] [Indexed: 03/24/2023] Open
Abstract
Myelination of neuronal axons is a critical aspect of central nervous system development and function. However, the fundamental cellular and molecular mechanisms influencing human developmental myelination and its failure are not fully understood. Here, we used digital spatial transcriptomics of a rare bank of human developing white matter to uncover that a localized dysregulated innate immune response is associated with impeded myelination. We identified that poorly myelinating areas have a distinct signature of Type II interferon signalling in microglia/macrophages, relative to adjacent myelinating areas. This is associated with a surprising increase in mature oligodendrocytes, which fail to form myelin processes appropriately. We functionally link these findings by showing that conditioned media from interferon-stimulated microglia is sufficient to dysregulate myelin process formation by oligodendrocytes in culture. We identify the Type II interferon inducer, Osteopontin (SPP1), as being upregulated in poorly myelinating brains, indicating a potential biomarker. Our results reveal the importance of microglia-mature oligodendrocyte interaction and interferon signaling in regulating myelination of the developing human brain.
Collapse
Affiliation(s)
- Rebecca K Holloway
- Keenan Research Centre for Biomedial Science at St. Michael's Hospital, 209 Victoria Street, Toronto, ON, M5B 1T8, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, Scotland, UK
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, Scotland, UK
| | - Liang Zhang
- Nanostring Technologies, Inc., Seattle, WA, USA
| | - Irene Molina-Gonzalez
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, Scotland, UK
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, Scotland, UK
| | - Kathy Ton
- Nanostring Technologies, Inc., Seattle, WA, USA
| | - James A R Nicoll
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Department of Cellular Pathology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - James P Boardman
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, Scotland, UK
| | - Yan Liang
- Nanostring Technologies, Inc., Seattle, WA, USA
| | - Anna Williams
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, UK
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, Scotland, UK
| | - Veronique E Miron
- Keenan Research Centre for Biomedial Science at St. Michael's Hospital, 209 Victoria Street, Toronto, ON, M5B 1T8, Canada.
- Department of Immunology, University of Toronto, Toronto, ON, Canada.
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, UK.
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, Scotland, UK.
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, Scotland, UK.
| |
Collapse
|
11
|
Portillo JAC, Yu JS, Vos S, Bapputty R, Lopez Corcino Y, Hubal A, Daw J, Arora S, Sun W, Lu ZR, Subauste CS. Disruption of retinal inflammation and the development of diabetic retinopathy in mice by a CD40-derived peptide or mutation of CD40 in Müller cells. Diabetologia 2022; 65:2157-2171. [PMID: 35920844 PMCID: PMC9630214 DOI: 10.1007/s00125-022-05775-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 06/09/2022] [Indexed: 01/11/2023]
Abstract
AIMS/HYPOTHESIS CD40 expressed in Müller cells is a central driver of diabetic retinopathy. CD40 causes phospholipase Cγ1 (PLCγ1)-dependent ATP release in Müller cells followed by purinergic receptor (P2X7)-dependent production of proinflammatory cytokines in myeloid cells. In the diabetic retina, CD40 and P2X7 upregulate a broad range of inflammatory molecules that promote development of diabetic retinopathy. The molecular event downstream of CD40 that activates the PLCγ1-ATP-P2X7-proinflammatory cytokine cascade and promotes development of diabetic retinopathy is unknown. We hypothesise that disruption of the CD40-driven molecular events that trigger this cascade prevents/treats diabetic retinopathy in mice. METHODS B6 and transgenic mice with Müller cell-restricted expression of wild-type (WT) CD40 or CD40 with mutations in TNF receptor-associated factor (TRAF) binding sites were made diabetic using streptozotocin. Leucostasis was assessed using FITC-conjugated concanavalin A. Histopathology was examined in the retinal vasculature. Expression of inflammatory molecules and phospho-Tyr783 PLCγ1 (p-PLCγ1) were assessed using real-time PCR, immunoblot and/or immunohistochemistry. Release of ATP and cytokines were measured by ATP bioluminescence and ELISA, respectively. RESULTS Human Müller cells with CD40 ΔT2,3 (lacks TRAF2,3 binding sites) were unable to phosphorylate PLCγ1 and release ATP in response to CD40 ligation, and could not induce TNF-α/IL-1β secretion in bystander myeloid cells. CD40-TRAF signalling acted via Src to induce PLCγ1 phosphorylation. Diabetic mice in which WT CD40 in Müller cells was replaced by CD40 ΔT2,3 failed to exhibit phosphorylation of PLCγ1 in these cells and upregulate P2X7 and TNF-α in microglia/macrophages. P2x7 (also known as P2rx7), Tnf-α (also known as Tnf), Il-1β (also known as Il1b), Nos2, Icam-1 (also known as Icam1) and Ccl2 mRNA were not increased in these mice and the mice did not develop retinal leucostasis and capillary degeneration. Diabetic B6 mice treated intravitreally with a cell-permeable peptide that disrupts CD40-TRAF2,3 signalling did not exhibit either upregulation of P2X7 and inflammatory molecules in the retina or leucostasis. CONCLUSIONS/INTERPRETATION CD40-TRAF2,3 signalling activated the CD40-PLCγ1-ATP-P2X7-proinflammatory cytokine pathway. Src functioned as a link between CD40-TRAF2,3 and PLCγ1. Replacing WT CD40 with CD40 ΔT2,3 impaired activation of PLCγ1 in Müller cells, upregulation of P2X7 in microglia/macrophages, upregulation of a broad range of inflammatory molecules in the diabetic retina and the development of diabetic retinopathy. Administration of a peptide that disrupts CD40-TRAF2,3 signalling reduced retinal expression of inflammatory molecules and reduced leucostasis in diabetic mice, supporting the therapeutic potential of pharmacological inhibition of CD40-TRAF2,3 in diabetic retinopathy.
Collapse
Affiliation(s)
- Jose-Andres C Portillo
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Jin-Sang Yu
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Sarah Vos
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Reena Bapputty
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Yalitza Lopez Corcino
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Alyssa Hubal
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Jad Daw
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Sahil Arora
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Wenyu Sun
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Zheng-Rong Lu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Carlos S Subauste
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA.
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
12
|
Gharagozloo M, Mace JW, Calabresi PA. Animal models to investigate the effects of inflammation on remyelination in multiple sclerosis. Front Mol Neurosci 2022; 15:995477. [PMID: 36407761 PMCID: PMC9669474 DOI: 10.3389/fnmol.2022.995477] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/22/2022] [Indexed: 09/19/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory, demyelinating, and neurodegenerative disease of the central nervous system (CNS). In people with MS, impaired remyelination and axonal loss lead to debilitating long-term neurologic deficits. Current MS disease-modifying drugs mainly target peripheral immune cells and have demonstrated little efficacy for neuroprotection or promoting repair. To elucidate the pathological mechanisms and test therapeutic interventions, multiple animal models have been developed to recapitulate specific aspects of MS pathology, particularly the acute inflammatory stage. However, there are few animal models that facilitate the study of remyelination in the presence of inflammation, and none fully replicate the biology of chronic demyelination in MS. In this review, we describe the animal models that have provided insight into the mechanisms underlying demyelination, myelin repair, and potential therapeutic targets for remyelination. We highlight the limitations of studying remyelination in toxin-based demyelination models and discuss the combinatorial models that recapitulate the inflammatory microenvironment, which is now recognized to be a major inhibitor of remyelination mechanisms. These models may be useful in identifying novel therapeutics that promote CNS remyelination in inflammatory diseases such as MS.
Collapse
Affiliation(s)
- Marjan Gharagozloo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jackson W. Mace
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Peter A. Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
13
|
Clark DN, Begg LR, Filiano AJ. Unique aspects of IFN-γ/STAT1 signaling in neurons. Immunol Rev 2022; 311:187-204. [PMID: 35656941 PMCID: PMC10120860 DOI: 10.1111/imr.13092] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/01/2022] [Accepted: 05/12/2022] [Indexed: 01/05/2023]
Abstract
The IFN-γ/STAT1 immune signaling pathway impacts many homeostatic and pathological aspects of neurons, beyond its canonical role in controlling intracellular pathogens. Well known for its potent pro-inflammatory and anti-viral functions in the periphery, the IFN-γ/STAT1 pathway is rapidly activated then deactivated to prevent excessive inflammation; however, neurons utilize unique IFN-γ/STAT1 activation patterns, which may contribute to the non-canonical neuron-specific downstream effects. Though it is now well-established that the immune system interacts and supports the CNS in health and disease, many aspects regarding IFN-γ production in the CNS and how neurons respond to IFN-γ are unclear. Additionally, it is not well understood how the diversity of the IFN-γ/STAT1 pathway is regulated in neurons to control homeostatic functions, support immune surveillance, and prevent pathologies. In this review, we discuss the neuron-specific mechanisms and kinetics of IFN-γ/STAT1 activation, the potential sources and entry sites of IFN-γ in the CNS, and the diverse set of homeostatic and pathological effects IFN-γ/STAT1 signaling in neurons has on CNS health and disease. We will also highlight the different contexts and conditions under which IFN-γ-induced STAT1 activation has been studied in neurons, and how various factors might contribute to the vast array of downstream effects observed.
Collapse
Affiliation(s)
- Danielle N. Clark
- Department of Immunology, Duke University, Durham, North Carolina, USA
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA
| | - Lauren R. Begg
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Anthony J. Filiano
- Department of Immunology, Duke University, Durham, North Carolina, USA
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA
- Department of Pathology, Duke University, Durham, North Carolina, USA
- Department of Neurosurgery, Duke University, Durham, North Carolina, USA
| |
Collapse
|
14
|
Seguella L, Gulbransen BD. Enteric glial biology, intercellular signalling and roles in gastrointestinal disease. Nat Rev Gastroenterol Hepatol 2021; 18:571-587. [PMID: 33731961 PMCID: PMC8324524 DOI: 10.1038/s41575-021-00423-7] [Citation(s) in RCA: 179] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/28/2021] [Indexed: 02/07/2023]
Abstract
One of the most transformative developments in neurogastroenterology is the realization that many functions normally attributed to enteric neurons involve interactions with enteric glial cells: a large population of peripheral neuroglia associated with enteric neurons throughout the gastrointestinal tract. The notion that glial cells function solely as passive support cells has been refuted by compelling evidence that demonstrates that enteric glia are important homeostatic cells of the intestine. Active signalling mechanisms between enteric glia and neurons modulate gastrointestinal reflexes and, in certain circumstances, function to drive neuroinflammatory processes that lead to long-term dysfunction. Bidirectional communication between enteric glia and immune cells contributes to gastrointestinal immune homeostasis, and crosstalk between enteric glia and cancer stem cells regulates tumorigenesis. These neuromodulatory and immunomodulatory roles place enteric glia in a unique position to regulate diverse gastrointestinal disease processes. In this Review, we discuss current concepts regarding enteric glial development, heterogeneity and functional roles in gastrointestinal pathophysiology and pathophysiology, with a focus on interactions with neurons and immune cells. We also present a working model to differentiate glial states based on normal function and disease-induced dysfunctions.
Collapse
Affiliation(s)
- Luisa Seguella
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
- Department of Physiology, Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Brian D Gulbransen
- Department of Physiology, Neuroscience Program, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
15
|
Chen Y, Kunjamma RB, Weiner M, Chan JR, Popko B. Prolonging the integrated stress response enhances CNS remyelination in an inflammatory environment. eLife 2021; 10:e65469. [PMID: 33752802 PMCID: PMC7987340 DOI: 10.7554/elife.65469] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/08/2021] [Indexed: 12/13/2022] Open
Abstract
The inflammatory environment of demyelinated lesions in multiple sclerosis (MS) patients contributes to remyelination failure. Inflammation activates a cytoprotective pathway, the integrated stress response (ISR), but it remains unclear whether enhancing the ISR can improve remyelination in an inflammatory environment. To examine this possibility, the remyelination stage of experimental autoimmune encephalomyelitis (EAE), as well as a mouse model that incorporates cuprizone-induced demyelination along with CNS delivery of the proinflammatory cytokine IFN-γ were used here. We demonstrate that either genetic or pharmacological ISR enhancement significantly increased the number of remyelinating oligodendrocytes and remyelinated axons in the inflammatory lesions. Moreover, the combined treatment of the ISR modulator Sephin1 with the oligodendrocyte differentiation enhancing reagent bazedoxifene increased myelin thickness of remyelinated axons to pre-lesion levels. Taken together, our findings indicate that prolonging the ISR protects remyelinating oligodendrocytes and promotes remyelination in the presence of inflammation, suggesting that ISR enhancement may provide reparative benefit to MS patients.
Collapse
Affiliation(s)
- Yanan Chen
- Department of Neurology, Division of Multiple Sclerosis and Neuroimmunology, Northwestern University Feinberg School of MedicineChicagoUnited States
| | - Rejani B Kunjamma
- Department of Neurology, Division of Multiple Sclerosis and Neuroimmunology, Northwestern University Feinberg School of MedicineChicagoUnited States
| | - Molly Weiner
- Department of Neurology, Division of Multiple Sclerosis and Neuroimmunology, Northwestern University Feinberg School of MedicineChicagoUnited States
| | - Jonah R Chan
- Weill Institute for Neuroscience, Department of Neurology, University of California, San FranciscoSan FranciscoUnited States
| | - Brian Popko
- Department of Neurology, Division of Multiple Sclerosis and Neuroimmunology, Northwestern University Feinberg School of MedicineChicagoUnited States
| |
Collapse
|
16
|
Yu X, Nagai J, Khakh BS. Improved tools to study astrocytes. Nat Rev Neurosci 2020; 21:121-138. [DOI: 10.1038/s41583-020-0264-8] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2020] [Indexed: 12/21/2022]
|
17
|
Abstract
Activation of the unfolded protein response in response to endoplasmic reticulum stress preserves cell viability and function under stressful conditions. Nevertheless, persistent, unresolvable activation of the unfolded protein response can trigger apoptosis to eliminate stressed cells. Recent studies show that the unfolded protein response plays an important role in the pathogenesis of various disorders of myelin, including multiples sclerosis, Charcot-Marie-Tooth disease, Pelizaeus-Merzbacher disease, vanishing white matter disease, spinal cord injury, tuberous sclerosis complex, and hypoxia-induced perinatal white matter injury. In this review we summarize the current literature on the unfolded protein response and the evidence for its role in the pathogenesis of myelin disorders.
Collapse
Affiliation(s)
- Wensheng Lin
- Department of Neuroscience; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Sarrabeth Stone
- Department of Neuroscience; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
18
|
Dufor T, Grehl S, Tang AD, Doulazmi M, Traoré M, Debray N, Dubacq C, Deng ZD, Mariani J, Lohof AM, Sherrard RM. Neural circuit repair by low-intensity magnetic stimulation requires cellular magnetoreceptors and specific stimulation patterns. SCIENCE ADVANCES 2019; 5:eaav9847. [PMID: 31692960 PMCID: PMC6821463 DOI: 10.1126/sciadv.aav9847] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 09/16/2019] [Indexed: 05/10/2023]
Abstract
Although electromagnetic brain stimulation is a promising treatment in neurology and psychiatry, clinical outcomes are variable, and underlying mechanisms are ill-defined, which impedes the development of new effective stimulation protocols. Here, we show, in vivo and ex vivo, that repetitive transcranial magnetic stimulation at low-intensity (LI-rTMS) induces axon outgrowth and synaptogenesis to repair a neural circuit. This repair depends on stimulation pattern, with complex biomimetic patterns being particularly effective, and the presence of cryptochrome, a putative magnetoreceptor. Only repair-promoting LI-rTMS patterns up-regulated genes involved in neuronal repair; almost 40% of were cryptochrome targets. Our data open a new framework to understand the mechanisms underlying structural neuroplasticity induced by electromagnetic stimulation. Rather than neuronal activation by induced electric currents, we propose that weak magnetic fields act through cryptochrome to activate cellular signaling cascades. This information opens new routes to optimize electromagnetic stimulation and develop effective treatments for different neurological diseases.
Collapse
Affiliation(s)
- T. Dufor
- Sorbonne Université and CNRS, IBPS, UMR 8256 Biological Adaptation and Ageing, Paris, France
| | - S. Grehl
- Sorbonne Université and CNRS, IBPS, UMR 8256 Biological Adaptation and Ageing, Paris, France
- Experimental and Regenerative Neuroscience, School of Animal Biology, University of Western Australia, Perth, Australia
| | - A. D. Tang
- Experimental and Regenerative Neuroscience, School of Animal Biology, University of Western Australia, Perth, Australia
| | - M. Doulazmi
- Sorbonne Université and CNRS, IBPS, UMR 8256 Biological Adaptation and Ageing, Paris, France
| | | | - N. Debray
- Sorbonne Université and CNRS, IBPS, UMR 8256 Biological Adaptation and Ageing, Paris, France
| | - C. Dubacq
- Sorbonne Université, IBPS, CNRS UMR 8246 and INSERM U1130 Neuroscience Paris Seine, Paris, France
| | - Z.-D. Deng
- Noninvasive Neuromodulation Unit, Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - J. Mariani
- Sorbonne Université and CNRS, IBPS, UMR 8256 Biological Adaptation and Ageing, Paris, France
- Sorbonne Université and Assistance Publique Hôpitaux de Paris, Institut de la Longévité, Charles Foix Hospital, Ivry-sur-Seine, France
| | - A. M. Lohof
- Sorbonne Université and CNRS, IBPS, UMR 8256 Biological Adaptation and Ageing, Paris, France
| | - R. M. Sherrard
- Sorbonne Université and CNRS, IBPS, UMR 8256 Biological Adaptation and Ageing, Paris, France
- Sorbonne Université and Assistance Publique Hôpitaux de Paris, Institut de la Longévité, Charles Foix Hospital, Ivry-sur-Seine, France
- Corresponding author.
| |
Collapse
|
19
|
Kirby L, Jin J, Cardona JG, Smith MD, Martin KA, Wang J, Strasburger H, Herbst L, Alexis M, Karnell J, Davidson T, Dutta R, Goverman J, Bergles D, Calabresi PA. Oligodendrocyte precursor cells present antigen and are cytotoxic targets in inflammatory demyelination. Nat Commun 2019; 10:3887. [PMID: 31467299 PMCID: PMC6715717 DOI: 10.1038/s41467-019-11638-3] [Citation(s) in RCA: 268] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 07/24/2019] [Indexed: 01/09/2023] Open
Abstract
Oligodendrocyte precursor cells (OPCs) are abundant in the adult central nervous system, and have the capacity to regenerate oligodendrocytes and myelin. However, in inflammatory diseases such as multiple sclerosis (MS) remyelination is often incomplete. To investigate how neuroinflammation influences OPCs, we perform in vivo fate-tracing in an inflammatory demyelinating mouse model. Here we report that OPC differentiation is inhibited by both effector T cells and IFNγ overexpression by astrocytes. IFNγ also reduces the absolute number of OPCs and alters remaining OPCs by inducing the immunoproteasome and MHC class I. In vitro, OPCs exposed to IFNγ cross-present antigen to cytotoxic CD8 T cells, resulting in OPC death. In human demyelinated MS brain lesions, but not normal appearing white matter, oligodendroglia exhibit enhanced expression of the immunoproteasome subunit PSMB8. Therefore, OPCs may be co-opted by the immune system in MS to perpetuate the autoimmune response, suggesting that inhibiting immune activation of OPCs may facilitate remyelination.
Collapse
Affiliation(s)
- Leslie Kirby
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jing Jin
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | - Matthew D Smith
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Kyle A Martin
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | - Hayley Strasburger
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Leyla Herbst
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Maya Alexis
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | | | - Ranjan Dutta
- Department of Neuroscience, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Joan Goverman
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Dwight Bergles
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
20
|
Purvis IJ, Avilala J, Guda MR, Venkataraman S, Vibhakar R, Tsung AJ, Velpula KK, Asuthkar S. Role of MYC-miR-29-B7-H3 in Medulloblastoma Growth and Angiogenesis. J Clin Med 2019; 8:jcm8081158. [PMID: 31382461 PMCID: PMC6723910 DOI: 10.3390/jcm8081158] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 07/23/2019] [Accepted: 07/30/2019] [Indexed: 12/14/2022] Open
Abstract
Medulloblastoma (MB) is the most common embryonal neuroepithelial tumor, with poor patient outcomes and secondary complications. In this study, we investigated the role of the B7 family of immune checkpoint homolog 3 (B7-H3) expression in MB angiogenesis. B7-H3, a co-inhibitory immune checkpoint, is highly expressed and is associated with lower overall survival in MYC+ MB's. Evidence for a direct transcriptional role of MYC on the B7-H3 gene promoter was confirmed by MYC inhibition and anti-MYC antibody ChIP analysis. Interestingly, MYC inhibition not only downregulated the B7-H3 protein expression, but also rescued miR-29 expression, thus indicating a triangular regulatory relationship between MYC, miR-29, and B7-H3 in Group 3 MB cells. From RNA seq and IPAD assay, we observed a negative feedback loop between miR-29 and MYC that may control B7-H3 expression levels in MB cells. Our studies show that B7-H3 expression levels play a crucial role in promoting MB angiogenesis which can be inhibited by miR-29 overexpression via miR-29-mediated B7-H3 downregulation. The tumor suppressor role of miR-29 is mediated by the activation of JAK/STAT1 signaling that further plays a role in MYC-B7-H3 downregulation in MB. This study highlights B7-H3 as a viable target in MB angiogenesis, and that the expression of miR-29 can inhibit B7-H3 and sensitize MB cells to treatment with MYC-inhibiting drugs.
Collapse
Affiliation(s)
- Ian J Purvis
- Departments of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
| | - Janardhan Avilala
- Departments of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
| | - Maheedhara R Guda
- Departments of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
| | - Sujatha Venkataraman
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Rajeev Vibhakar
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Andrew J Tsung
- Departments of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- Departments of Neurosurgery, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
| | - Kiran K Velpula
- Departments of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- Departments of Neurosurgery, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- Departments of Pediatrics, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
| | - Swapna Asuthkar
- Departments of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA.
| |
Collapse
|
21
|
GSK3β overexpression driven by GFAP promoter improves rotarod performance. Brain Res 2019; 1712:47-54. [DOI: 10.1016/j.brainres.2019.01.040] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 11/20/2022]
|
22
|
Zhang H, Kuick R, Park SS, Peabody C, Yoon J, Fernández EC, Wang J, Thomas D, Viollet B, Inoki K, Camelo-Piragua S, Rual JF. Loss of AMPKα2 Impairs Hedgehog-Driven Medulloblastoma Tumorigenesis. Int J Mol Sci 2018; 19:ijms19113287. [PMID: 30360486 PMCID: PMC6274763 DOI: 10.3390/ijms19113287] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 10/11/2018] [Accepted: 10/19/2018] [Indexed: 12/20/2022] Open
Abstract
The AMP-activated protein kinase (AMPK) is a sensor of cellular energy status that has a dual role in cancer, i.e., pro- or anti-tumorigenic, depending on the context. In medulloblastoma, the most frequent malignant pediatric brain tumor, several in vitro studies previously showed that AMPK suppresses tumor cell growth. The role of AMPK in this disease context remains to be tested in vivo. Here, we investigate loss of AMPKα2 in a genetically engineered mouse model of sonic hedgehog (SHH)-medulloblastoma. In contrast to previous reports, our study reveals that AMPKα2 KO impairs SHH medulloblastoma tumorigenesis. Moreover, we performed complementary molecular and genomic analyses that support the hypothesis of a pro-tumorigenic SHH/AMPK/CNBP axis in medulloblastoma. In conclusion, our observations further underline the context-dependent role of AMPK in cancer, and caution is warranted for the previously proposed hypothesis that AMPK agonists may have therapeutic benefits in medulloblastoma patients. Note: an abstract describing the project was previously submitted to the American Society for Investigative Pathology PISA 2018 conference and appears in The American Journal of Pathology (Volume 188, Issue 10, October 2018, Page 2433).
Collapse
Affiliation(s)
- Honglai Zhang
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Rork Kuick
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Sung-Soo Park
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Claire Peabody
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Justin Yoon
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Ester Calvo Fernández
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Junying Wang
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Dafydd Thomas
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Benoit Viollet
- Inserm, U1016, Institut Cochin, 75014 Paris, France.
- CNRS, UMR8104, 75014 Paris, France.
- Université Paris Descartes, Sorbonne Paris cité, 75014 Paris, France.
| | - Ken Inoki
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Sandra Camelo-Piragua
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Jean-François Rual
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
23
|
Stone S, Wu S, Jamison S, Durose W, Pallais JP, Lin W. Activating transcription factor 6α deficiency exacerbates oligodendrocyte death and myelin damage in immune-mediated demyelinating diseases. Glia 2018; 66:1331-1345. [PMID: 29436030 DOI: 10.1002/glia.23307] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 01/16/2018] [Accepted: 01/29/2018] [Indexed: 01/06/2023]
Abstract
Endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) play a critical role in immune-mediated demyelinating diseases, including multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE), by regulating the viability of oligodendrocytes. Our previous studies show that activation of the PERK branch of the UPR protects myelinating oligodendrocytes against ER stress in young, developing mice that express IFN-γ, a key pro-inflammatory cytokine in MS and EAE, in the CNS. Several studies also demonstrate that PERK activation preserves oligodendrocyte viability and function, protecting mice against EAE. While evidence suggests activation of the ATF6α branch of the UPR in oligodendrocytes under normal and disease conditions, the effects of ATF6α activation on oligodendrocytes in immune-mediated demyelinating diseases remain unknown. Herein, we showed that ATF6α deficiency had no effect on oligodendrocytes under normal conditions. Interestingly, we showed that ATF6α deficiency exacerbated ER stressed-induced myelinating oligodendrocyte death and subsequent myelin loss in the developing CNS of IFN-γ-expressing mice. Moreover, we found that ATF6α deficiency increased EAE severity and aggravated EAE-induced oligodendrocyte loss and demyelination, without affecting inflammation. Thus, these data suggest the protective effects of ATF6α activation on oligodendrocytes in immune-mediated demyelinating diseases.
Collapse
Affiliation(s)
- Sarrabeth Stone
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota.,Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Shuangchan Wu
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota.,Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Stephanie Jamison
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota.,Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Wilaiwan Durose
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota.,Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Jean Pierre Pallais
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota.,Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Wensheng Lin
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota.,Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
24
|
IDO1 involvement in mTOR pathway: a molecular mechanism of resistance to mTOR targeting in medulloblastoma. Oncotarget 2018; 7:52900-52911. [PMID: 27174915 PMCID: PMC5288157 DOI: 10.18632/oncotarget.9284] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 04/11/2016] [Indexed: 12/14/2022] Open
Abstract
Medulloblastoma (MB) is the most common malignant brain tumor in children. Despite therapeutic advancements, high-risk groups still present significant mortality. A deeper knowledge of the signaling pathways contributing to MB formation and aggressiveness would help develop new successful therapies. The target of rapamycin, mTOR signaling, is known to be involved in MB and is already targetable in the clinical setting. Furthermore, mTOR is a master metabolic regulator able to control cell growth versus autophagy decisions in conditions of amino-acid deprivation that can be due to IDO1 enzymatic activity. IDO1 has been also implicated in the regulation of inflammation, as well as of T cell-mediated immune responses, in a variety of pathological conditions, including brain tumors. In particular, IDO1 induces expansion of regulatory T-cells (Treg), preventing immune response against tumor cells. Analysis of 27 MB tissue specimens for the expression of both mTOR and IDO1 showed their widespread expression in all samples. Testing their cooperation in vitro, a significant involvement of IDO1 in mTOR immunogenic pathway was found, able to counteract the aim of rapamycin treatment. In MB cell lines, inhibition of mTOR strongly induced IDO1 expression and activity, corroborating its ability to recruit Treg cells in the tumor microenvironment. The mTOR/IDO1 cross talk was found to be strictly specific of MB cells. We demonstrated that mTOR pathway cross talks with IDO1 pathway to promote MB immune escape, possibly contributing to failure of mTOR- targeted therapy.
Collapse
|
25
|
Neumann JE, Swartling FJ, Schüller U. Medulloblastoma: experimental models and reality. Acta Neuropathol 2017; 134:679-689. [PMID: 28725965 DOI: 10.1007/s00401-017-1753-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 06/27/2017] [Accepted: 07/16/2017] [Indexed: 12/11/2022]
Abstract
Medulloblastoma is the most frequent malignant brain tumor in childhood, but it may also affect infants, adolescents, and young adults. Recent advances in the understanding of the disease have shed light on molecular and clinical heterogeneity, which is now reflected in the updated WHO classification of brain tumors. At the same time, it is well accepted that preclinical research and clinical trials have to be subgroup-specific. Hence, valid models have to be generated specifically for every medulloblastoma subgroup to properly mimic molecular fingerprints, clinical features, and responsiveness to targeted therapies. This review summarizes the availability of experimental medulloblastoma models with a particular focus on how well these models reflect the actual disease subgroup. We further describe technical advantages and disadvantages of the models and finally point out how some models have successfully been used to introduce new drugs and why some medulloblastoma subgroups are extraordinary difficult to model.
Collapse
Affiliation(s)
- Julia E Neumann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fredrik J Swartling
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Ulrich Schüller
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Research Institute Children's Cancer Center, Martinistrasse 52, 20251, Hamburg, Germany.
| |
Collapse
|
26
|
Forsberg D, Ringstedt T, Herlenius E. Astrocytes release prostaglandin E2 to modify respiratory network activity. eLife 2017; 6:29566. [PMID: 28976306 PMCID: PMC5648524 DOI: 10.7554/elife.29566] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 10/03/2017] [Indexed: 12/31/2022] Open
Abstract
Previously (Forsberg et al., 2016), we revealed that prostaglandin E2 (PGE2), released during hypercapnic challenge, increases calcium oscillations in the chemosensitive parafacial respiratory group (pFRG/RTN). Here, we demonstrate that pFRG/RTN astrocytes are the PGE2 source. Two distinct astrocyte subtypes were found using transgenic mice expressing GFP and MrgA1 receptors in astrocytes. Although most astrocytes appeared dormant during time-lapse calcium imaging, a subgroup displayed persistent, rhythmic oscillating calcium activity. These active astrocytes formed a subnetwork within the respiratory network distinct from the neuronal network. Activation of exogenous MrgA1Rs expressed in astrocytes tripled astrocytic calcium oscillation frequency in both the preBötzinger complex and pFRG/RTN. However, neurons in the preBötC were unaffected, whereas neuronal calcium oscillatory frequency in pFRG/RTN doubled. Notably, astrocyte activation in pFRG/RTN triggered local PGE2 release and blunted the hypercapnic response. Thus, astrocytes play an active role in respiratory rhythm modulation, modifying respiratory-related behavior through PGE2 release in the pFRG/RTN.
Collapse
Affiliation(s)
- David Forsberg
- Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Thomas Ringstedt
- Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Eric Herlenius
- Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
27
|
NF-κB Activation Protects Oligodendrocytes against Inflammation. J Neurosci 2017; 37:9332-9344. [PMID: 28842413 PMCID: PMC5607472 DOI: 10.1523/jneurosci.1608-17.2017] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/07/2017] [Accepted: 08/16/2017] [Indexed: 01/13/2023] Open
Abstract
NF-κB is a key player in inflammatory diseases, including multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). However, the effects of NF-κB activation on oligodendrocytes in MS and EAE remain unknown. We generated a mouse model that expresses IκBαΔN, a super-suppressor of NF-κB, specifically in oligodendrocytes and demonstrated that IκBαΔN expression had no effect on oligodendrocytes under normal conditions (both sexes). Interestingly, we showed that oligodendrocyte-specific expression of IκBαΔN blocked NF-κB activation in oligodendrocytes and resulted in exacerbated oligodendrocyte death and hypomyelination in young, developing mice that express IFN-γ ectopically in the CNS (both sexes). We also showed that NF-κB inactivation in oligodendrocytes aggravated IFN-γ-induced remyelinating oligodendrocyte death and remyelination failure in the cuprizone model (male mice). Moreover, we found that NF-κB inactivation in oligodendrocytes increased the susceptibility of mice to EAE (female mice). These findings imply the cytoprotective effects of NF-κB activation on oligodendrocytes in MS and EAE.SIGNIFICANCE STATEMENT Multiple sclerosis (MS) is an inflammatory demyelinating disease of the CNS. NF-κB is a major player in inflammatory diseases that acts by regulating inflammation and cell viability. Data indicate that NF-κB activation in inflammatory cells facilitates the development of MS. However, to date, attempts to understand the role of NF-κB activation in oligodendrocytes in MS have been unsuccessful. Herein, we generated a mouse model that allows for inactivation of NF-κB specifically in oligodendrocytes and then used this model to determine the precise role of NF-κB activation in oligodendrocytes in models of MS. The results presented in this study represent the first demonstration that NF-κB activation acts cell autonomously to protect oligodendrocytes against inflammation in animal models of MS.
Collapse
|
28
|
Xu T, Zhang H, Park SS, Venneti S, Kuick R, Ha K, Michael LE, Santi M, Uchida C, Uchida T, Srinivasan A, Olson JM, Dlugosz AA, Camelo-Piragua S, Rual JF. Loss of Pin1 Suppresses Hedgehog-Driven Medulloblastoma Tumorigenesis. Neoplasia 2017; 19:216-225. [PMID: 28167297 PMCID: PMC5293723 DOI: 10.1016/j.neo.2017.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 01/09/2017] [Indexed: 01/30/2023] Open
Abstract
Medulloblastoma is the most common malignant brain tumor in children. Therapeutic approaches to medulloblastoma (combination of surgery, radiotherapy, and chemotherapy) have led to significant improvements, but these are achieved at a high cost to quality of life. Alternative therapeutic approaches are needed. Genetic mutations leading to the activation of the Hedgehog pathway drive tumorigenesis in ~30% of medulloblastoma. In a yeast two-hybrid proteomic screen, we discovered a novel interaction between GLI1, a key transcription factor for the mediation of Hedgehog signals, and PIN1, a peptidylprolyl cis/trans isomerase that regulates the postphosphorylation fate of its targets. The GLI1/PIN1 interaction was validated by reciprocal pulldowns using epitope-tagged proteins in HEK293T cells as well as by co-immunoprecipiations of the endogenous proteins in a medulloblastoma cell line. Our results support a molecular model in which PIN1 promotes GLI1 protein abundance, thus contributing to the positive regulation of Hedgehog signals. Most importantly, in vivo functional analyses of Pin1 in the GFAP-tTA;TRE-SmoA1 mouse model of Hedgehog-driven medulloblastoma demonstrate that the loss of Pin1 impairs tumor development and dramatically increases survival. In summary, the discovery of the GLI1/PIN1 interaction uncovers PIN1 as a novel therapeutic target in Hedgehog-driven medulloblastoma tumorigenesis.
Collapse
Affiliation(s)
- Tao Xu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Honglai Zhang
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Sung-Soo Park
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Sriram Venneti
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Rork Kuick
- Center for Cancer Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kimberly Ha
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Lowell Evan Michael
- Departments of Dermatology and Cell & Developmental Biology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Mariarita Santi
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Chiyoko Uchida
- Department of Human Development and Culture, Fukushima University, Fukushima, 960-1296, Japan
| | - Takafumi Uchida
- Department of Molecular Cell Science, Tohoku University, Sendai 981-8555, Japan
| | - Ashok Srinivasan
- Department of Radiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - James M Olson
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Andrzej A Dlugosz
- Departments of Dermatology and Cell & Developmental Biology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Sandra Camelo-Piragua
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Jean-François Rual
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
29
|
Portillo JAC, Lopez Corcino Y, Miao Y, Tang J, Sheibani N, Kern TS, Dubyak GR, Subauste CS. CD40 in Retinal Müller Cells Induces P2X7-Dependent Cytokine Expression in Macrophages/Microglia in Diabetic Mice and Development of Early Experimental Diabetic Retinopathy. Diabetes 2017; 66:483-493. [PMID: 27474370 PMCID: PMC5248988 DOI: 10.2337/db16-0051] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 07/07/2016] [Indexed: 12/11/2022]
Abstract
Müller cells and macrophages/microglia are likely important for the development of diabetic retinopathy; however, the interplay between these cells in this disease is not well understood. An inflammatory process is linked to the onset of experimental diabetic retinopathy. CD40 deficiency impairs this process and prevents diabetic retinopathy. Using mice with CD40 expression restricted to Müller cells, we identified a mechanism by which Müller cells trigger proinflammatory cytokine expression in myeloid cells. During diabetes, mice with CD40 expressed in Müller cells upregulated retinal tumor necrosis factor-α (TNF-α), interleukin 1β (IL-1β), intracellular adhesion molecule 1 (ICAM-1), and nitric oxide synthase (NOS2), developed leukostasis and capillary degeneration. However, CD40 did not cause TNF-α or IL-1β secretion in Müller cells. TNF-α was not detected in Müller cells from diabetic mice with CD40+ Müller cells. Rather, TNF-α was upregulated in macrophages/microglia. CD40 ligation in Müller cells triggered phospholipase C-dependent ATP release that caused P2X7-dependent production of TNF-α and IL-1β by macrophages. P2X7-/- mice and mice treated with a P2X7 inhibitor were protected from diabetes-induced TNF-α, IL-1β, ICAM-1, and NOS2 upregulation. Our studies indicate that CD40 in Müller cells is sufficient to upregulate retinal inflammatory markers and appears to promote experimental diabetic retinopathy and that Müller cells orchestrate inflammatory responses in myeloid cells through a CD40-ATP-P2X7 pathway.
Collapse
Affiliation(s)
- Jose-Andres C Portillo
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH
| | - Yalitza Lopez Corcino
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH
| | - Yanling Miao
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH
| | - Jie Tang
- Division of Molecular Endocrinology, Department of Medicine, Case Western Reserve University, Cleveland, OH
| | - Nader Sheibani
- Department of Ophthalmology, University of Wisconsin-Madison, Madison, WI
| | - Timothy S Kern
- Division of Molecular Endocrinology, Department of Medicine, Case Western Reserve University, Cleveland, OH
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH
- Louis Stokes Cleveland Veterans Administration Medical Center, Research Service 151, Cleveland, OH
| | - George R Dubyak
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH
| | - Carlos S Subauste
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH
- Department of Pathology, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
30
|
Supasai S, Aimo L, Adamo AM, Mackenzie GG, Oteiza PI. Zinc deficiency affects the STAT1/3 signaling pathways in part through redox-mediated mechanisms. Redox Biol 2017; 11:469-481. [PMID: 28086195 PMCID: PMC5228099 DOI: 10.1016/j.redox.2016.12.027] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 12/03/2016] [Accepted: 12/26/2016] [Indexed: 01/03/2023] Open
Abstract
Zinc deficiency affects the development of the central nervous system (CNS) through mechanisms only partially understood. We previously showed that zinc deficiency causes CNS oxidative stress, damaging microtubules and impairing protein nuclear shuttling. STAT1 and STAT3 transcription factors, which require nuclear import for their functions, play major roles in CNS development. Thus, we investigated whether zinc deficiency disrupts STAT1 and STAT3 signaling pathways in the developing fetal CNS, characterizing the involvement of oxidative stress and the cytoskeleton in the adverse effects. Maternal (gestation day 0–19) marginal zinc deficiency (MZD) reduced STAT1 and STAT3 tyrosine phosphorylation and their nuclear translocation in the embryonic day 19 (E19) rat brain. Similar effects were observed in zinc depleted IMR-32 neuroblastoma cells, with an associated decrease in STAT1- and STAT3-dependent gene transactivation. Zinc deficiency caused oxidative stress (increased 4-hydroxynonenal-protein adducts) in E19 brain and IMR-32 cells, which was prevented in cells by supplementation with 0.5 mM α-lipoic acid (LA). In zinc depleted IMR-32 cells, the low tyrosine phosphorylation of STAT1, but not that of STAT3, recovered upon incubation with LA. STAT1 and STAT3 nuclear transports were also restored by LA. Accordingly, chemical disruption of the cytoskeleton partially reduced STAT1 and STAT3 nuclear levels. In summary, the redox-dependent tyrosine phosphorylation, and oxidant-mediated disruption of the cytoskeleton are involved in the deleterious effects of zinc deficit on STAT1 and STAT3 activation and nuclear translocation. Therefore, disruption of the STAT1 and STAT3 signaling pathways may in part explain the deleterious effects of maternal MZD on fetal brain development. Zinc deficits impair STAT1/STAT3 signaling in fetal brain and neuroblastoma cells. Zinc deficiency inhibits STAT1 and STAT3 tyrosine phosphorylation and nuclear translocation. Zinc deficiency causes oxidative stress (high HNE-protein adducts) in fetal brain and cells. Lipoic acid reverts zinc deficiency-associated decreased STAT1/STAT3 nuclear shuttling. Zinc deficiency-associated oxidative stress impairs STAT1/STAT3 modulation.
Collapse
Affiliation(s)
- S Supasai
- Department of Nutrition, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA; Department of Environmental Toxicology, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| | - L Aimo
- Department of Nutrition, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA; Department of Environmental Toxicology, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| | - A M Adamo
- Department of Biological Chemistry and IQUIFIB (UBA-CONICET), School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| | - G G Mackenzie
- Department of Nutrition, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| | - P I Oteiza
- Department of Nutrition, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA; Department of Environmental Toxicology, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA.
| |
Collapse
|
31
|
Stone S, Ho Y, Li X, Jamison S, Harding HP, Ron D, Lin W. Dual role of the integrated stress response in medulloblastoma tumorigenesis. Oncotarget 2016; 7:64124-64135. [PMID: 27802424 PMCID: PMC5325430 DOI: 10.18632/oncotarget.11873] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 09/01/2016] [Indexed: 02/02/2023] Open
Abstract
In response to endoplasmic reticulum (ER) stress, activation of pancreatic ER kinase (PERK) coordinates an adaptive program known as the integrated stress response (ISR) by phosphorylating translation initiation factor 2α (eIF2α). Phosphorylated eIF2α is quickly dephosphorylated by the protein phosphatase 1 and growth arrest and DNA damage 34 (GADD34) complex. Data indicate that the ISR can either promote or suppress tumor development. Our previous studies showed that the ISR is activated in medulloblastoma in both human patients and animal models, and that the decreased ISR via PERK heterozygous deficiency attenuates medulloblastoma formation in Patched1 heterozygous deficient (Ptch1+/-) mice by enhancing apoptosis of pre-malignant granule cell precursors (GCPs) during cell transformation. We showed here that GADD34 heterozygous mutation moderately enhanced the ISR and noticeably increased the incidence of medulloblastoma in adult Ptch1+/- mice. Surprisingly, GADD34 homozygous mutation strongly enhanced the ISR, but significantly decreased the incidence of medulloblastoma in adult Ptch1+/- mice. Intriguingly, GADD34 homozygous mutation significantly enhanced pre-malignant GCP apoptosis in cerebellar hyperplastic lesions and reduced the lesion numbers in young Ptch1+/- mice. Nevertheless, neither GADD34 heterozygous mutation nor GADD34 homozygous mutation had a significant effect on medulloblastoma cells in adult Ptch1+/- mice. Collectively, these data imply the dual role of the ISR, promoting and inhibiting, in medulloblastoma tumorigenesis by regulating apoptosis of pre-malignant GCPs during the course of malignant transformation.
Collapse
Affiliation(s)
- Sarrabeth Stone
- 1 Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States,2 Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States,3 Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States
| | - Yeung Ho
- 1 Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States,2 Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States,3 Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States
| | - Xiting Li
- 1 Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States,2 Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States,3 Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States,4 Department of Periodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Stephanie Jamison
- 1 Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States,2 Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States,3 Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States
| | - Heather P. Harding
- 5 Cambridge Institute of Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - David Ron
- 5 Cambridge Institute of Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Wensheng Lin
- 1 Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States,2 Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States,3 Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States
| |
Collapse
|
32
|
Forsberg D, Horn Z, Tserga E, Smedler E, Silberberg G, Shvarev Y, Kaila K, Uhlén P, Herlenius E. CO2-evoked release of PGE2 modulates sighs and inspiration as demonstrated in brainstem organotypic culture. eLife 2016; 5. [PMID: 27377173 PMCID: PMC4974055 DOI: 10.7554/elife.14170] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Accepted: 06/21/2016] [Indexed: 12/20/2022] Open
Abstract
Inflammation-induced release of prostaglandin E2 (PGE2) changes breathing patterns and the response to CO2 levels. This may have fatal consequences in newborn babies and result in sudden infant death. To elucidate the underlying mechanisms, we present a novel breathing brainstem organotypic culture that generates rhythmic neural network and motor activity for 3 weeks. We show that increased CO2 elicits a gap junction-dependent release of PGE2. This alters neural network activity in the preBötzinger rhythm-generating complex and in the chemosensitive brainstem respiratory regions, thereby increasing sigh frequency and the depth of inspiration. We used mice lacking eicosanoid prostanoid 3 receptors (EP3R), breathing brainstem organotypic slices and optogenetic inhibition of EP3R+/+ cells to demonstrate that the EP3R is important for the ventilatory response to hypercapnia. Our study identifies a novel pathway linking the inflammatory and respiratory systems, with implications for inspiration and sighs throughout life, and the ability to autoresuscitate when breathing fails. DOI:http://dx.doi.org/10.7554/eLife.14170.001 Humans and other mammals breathe air to absorb oxygen into the body and to remove carbon dioxide. We know that in a part of the brain called the brainstem, several regions work together to create breaths, but it is not clear precisely how this works. These regions adjust our breathing to the demands placed on the body by different activities, such as sleeping or exercising. Sometimes, especially in newborn babies, the brainstem’s monitoring of oxygen and carbon dioxide does not work properly, which can lead to abnormal breathing and possibly death. In the brain, cells called neurons form networks that can rapidly transfer information via electrical signals. Here, Forsberg et al. investigated the neural networks in the brainstem that generate and control breathing in mice. They used slices of mouse brainstem that had been kept alive in a dish in the laboratory. The slice contained an arrangement of neurons and supporting cells that allowed it to continue to produce patterns of electrical activity that are associated with breathing. Over a three-week period, Forsberg et al. monitored the activity of the cells and calculated how they were connected to each other. The experiments show that the neurons responsible for breathing were organized in a “small-world” network, in which the neurons are connected to each other directly or via small numbers of other neurons. Further experiments tested how various factors affect the behavior of the network. For example, carbon dioxide triggered the release of a small molecule called prostaglandin E2 from cells. This molecule is known to play a role in inflammation and fever. However, in the carbon dioxide sensing region of the brainstem it acted as a signaling molecule that increased activity. Therefore, inflammation could interfere with the body’s normal response to carbon dioxide and lead to potentially life-threatening breathing problems. Furthermore, prostaglandin E2 induced deeper breaths known as sighs, which may be vital for newborn babies to be able to take their first deep breaths of life. Future challenges include understanding how the brainstem neural networks generate breathing and translate this knowledge to improve the treatment of breathing difficulties in babies. DOI:http://dx.doi.org/10.7554/eLife.14170.002
Collapse
Affiliation(s)
- David Forsberg
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Karolinska University Hospital, Stockholm, Sweden
| | - Zachi Horn
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Karolinska University Hospital, Stockholm, Sweden
| | - Evangelia Tserga
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Karolinska University Hospital, Stockholm, Sweden
| | - Erik Smedler
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Gilad Silberberg
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Yuri Shvarev
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Karolinska University Hospital, Stockholm, Sweden
| | - Kai Kaila
- Department of Biosciences and Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Per Uhlén
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Eric Herlenius
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
33
|
Ho Y, Li X, Jamison S, Harding HP, McKinnon PJ, Ron D, Lin W. PERK Activation Promotes Medulloblastoma Tumorigenesis by Attenuating Premalignant Granule Cell Precursor Apoptosis. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1939-1951. [PMID: 27181404 PMCID: PMC4929388 DOI: 10.1016/j.ajpath.2016.03.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 02/18/2016] [Accepted: 03/04/2016] [Indexed: 12/26/2022]
Abstract
Evidence suggests that activation of pancreatic endoplasmic reticulum kinase (PERK) signaling in response to endoplasmic reticulum stress negatively or positively influences cell transformation by regulating apoptosis. Patched1 heterozygous deficient (Ptch1(+/-)) mice reproduce human Gorlin's syndrome and are regarded as the best animal model to study tumorigenesis of the sonic hedgehog subgroup of medulloblastomas. It is believed that medulloblastomas in Ptch1(+/-) mice results from the transformation of granule cell precursors (GCPs) in the developing cerebellum. Here, we determined the role of PERK signaling on medulloblastoma tumorigenesis by assessing its effects on premalignant GCPs and tumor cells. We found that PERK signaling was activated in both premalignant GCPs in young Ptch1(+/-) mice and medulloblastoma cells in adult mice. We demonstrated that PERK haploinsufficiency reduced the incidence of medulloblastomas in Ptch1(+/-) mice. Interestingly, PERK haploinsufficiency enhanced apoptosis of premalignant GCPs in young Ptch1(+/-) mice but had no significant effect on medulloblastoma cells in adult mice. Moreover, we showed that the PERK pathway was activated in medulloblastomas in humans. These results suggest that PERK signaling promotes medulloblastoma tumorigenesis by attenuating apoptosis of premalignant GCPs during the course of malignant transformation.
Collapse
Affiliation(s)
- Yeung Ho
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota; Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Xiting Li
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota; Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota; Department of Periodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Stephanie Jamison
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota; Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Heather P Harding
- Cambridge Institute of Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Peter J McKinnon
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - David Ron
- Cambridge Institute of Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Wensheng Lin
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota; Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
34
|
Jurado-Arjona J, Llorens-Martín M, Ávila J, Hernández F. GSK3β Overexpression in Dentate Gyrus Neural Precursor Cells Expands the Progenitor Pool and Enhances Memory Skills. J Biol Chem 2016; 291:8199-213. [PMID: 26887949 DOI: 10.1074/jbc.m115.674531] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Indexed: 11/06/2022] Open
Abstract
In restricted areas of the adult brain, like the subgranular zone of the dentate gyrus (DG), there is continuous production of new neurons. This process, named adult neurogenesis, is involved in important cognitive functions such as memory and learning. It requires the presence of newborn neurons that arise from neuronal stem cells, which divide and differentiate through successive stages in adulthood. In this work, we demonstrate that overexpression of glycogen synthase kinase (GSK) 3β in neural precursor cells (NPCs) using the glial fibrillary acidic protein promoter during DG development produces an increase in the neurogenic process, increasing NPCs numbers. Moreover, the transgenic mice show higher DG volume and increased number of mature granule neurons. In an attempt to compensate for these alterations, glial fibrillary acidic protein/GSK3β-overexpressing mice show increased levels of Dkk1 and sFRP3, two inhibitors of the Wnt-frizzled complex. We have also found behavioral differences between wild type and transgenic mice, indicating a higher rating in memory tasks for GSK3β-overexpressing mice compared with wild type mice. These data indicate that GSK3β is a crucial kinase in NPC physiology and suggest that this molecule plays a key role in the correct development of DG and adult neurogenesis in this region.
Collapse
Affiliation(s)
- Jerónimo Jurado-Arjona
- From the Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid (CSIC/UAM), Cantoblanco, 28049 Madrid, Spain and the Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - María Llorens-Martín
- From the Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid (CSIC/UAM), Cantoblanco, 28049 Madrid, Spain and the Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Jesús Ávila
- From the Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid (CSIC/UAM), Cantoblanco, 28049 Madrid, Spain and the Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Félix Hernández
- From the Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid (CSIC/UAM), Cantoblanco, 28049 Madrid, Spain and the Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| |
Collapse
|
35
|
Pardo L, Schlüter A, Valor LM, Barco A, Giralt M, Golbano A, Hidalgo J, Jia P, Zhao Z, Jové M, Portero-Otin M, Ruiz M, Giménez-Llort L, Masgrau R, Pujol A, Galea E. Targeted activation of CREB in reactive astrocytes is neuroprotective in focal acute cortical injury. Glia 2016; 64:853-74. [PMID: 26880229 DOI: 10.1002/glia.22969] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 01/07/2016] [Accepted: 01/08/2016] [Indexed: 01/07/2023]
Abstract
The clinical challenge in acute injury as in traumatic brain injury (TBI) is to halt the delayed neuronal loss that occurs hours and days after the insult. Here we report that the activation of CREB-dependent transcription in reactive astrocytes prevents secondary injury in cerebral cortex after experimental TBI. The study was performed in a novel bitransgenic mouse in which a constitutively active CREB, VP16-CREB, was targeted to astrocytes with the Tet-Off system. Using histochemistry, qPCR, and gene profiling we found less neuronal death and damage, reduced macrophage infiltration, preserved mitochondria, and rescued expression of genes related to mitochondrial metabolism in bitransgenic mice as compared to wild type littermates. Finally, with meta-analyses using publicly available databases we identified a core set of VP16-CREB candidate target genes that may account for the neuroprotective effect. Enhancing CREB activity in astrocytes thus emerges as a novel avenue in acute brain post-injury therapeutics.
Collapse
Affiliation(s)
- Luis Pardo
- Institut De Neurociències and Unitat De Bioquímica, Facultat De Medicina, Universitat Autònoma De Barcelona, Bellaterra, Barcelona, 08193, Spain
| | - Agatha Schlüter
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, L'hospitalet De Llobregat, Barcelona, 08907, Spain
| | - Luis M Valor
- Instituto De Neurociencias De Alicante, Universidad Miguel Hernández/Consejo Superior De Investigaciones Científicas, Sant Joan D'alacant, Alicante, 03550, Spain
| | - Angel Barco
- Instituto De Neurociencias De Alicante, Universidad Miguel Hernández/Consejo Superior De Investigaciones Científicas, Sant Joan D'alacant, Alicante, 03550, Spain
| | - Mercedes Giralt
- Institut De Neurociències and Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences, Universitat Autònoma, Barcelona, 08193, Spain
| | - Arantxa Golbano
- Institut De Neurociències and Unitat De Bioquímica, Facultat De Medicina, Universitat Autònoma De Barcelona, Bellaterra, Barcelona, 08193, Spain
| | - Juan Hidalgo
- Institut De Neurociències and Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences, Universitat Autònoma, Barcelona, 08193, Spain
| | - Peilin Jia
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee.,Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Zhongming Zhao
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee.,Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Mariona Jové
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida, Lleida, 25198, Spain
| | - Manuel Portero-Otin
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida, Lleida, 25198, Spain
| | - Montserrat Ruiz
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, L'hospitalet De Llobregat, Barcelona, 08907, Spain
| | - Lydia Giménez-Llort
- Institut De Neurociènces and Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma De Barcelona, Bellaterra, 08193, Spain
| | - Roser Masgrau
- Institut De Neurociències and Unitat De Bioquímica, Facultat De Medicina, Universitat Autònoma De Barcelona, Bellaterra, Barcelona, 08193, Spain
| | - Aurora Pujol
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, L'hospitalet De Llobregat, Barcelona, 08907, Spain.,Institució Catalana De Recerca I Estudis Avançats (ICREA), Barcelona, Spain
| | - Elena Galea
- Institut De Neurociències and Unitat De Bioquímica, Facultat De Medicina, Universitat Autònoma De Barcelona, Bellaterra, Barcelona, 08193, Spain.,Institució Catalana De Recerca I Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
36
|
Way SW, Popko B. Harnessing the integrated stress response for the treatment of multiple sclerosis. Lancet Neurol 2016; 15:434-43. [PMID: 26873788 PMCID: PMC4792730 DOI: 10.1016/s1474-4422(15)00381-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 11/09/2015] [Accepted: 12/02/2015] [Indexed: 12/13/2022]
Abstract
Multiple sclerosis (MS) is a chronic demyelinating autoimmune disease of the central nervous system (CNS) with no known cure. Though a dozen immunomodulatory therapies exist, their impact on progression of disease appears limited. The field has hence focused on alternate strategies for treatment such as enhancing remyelination and CNS repair. Recent progress has been made on a third complimentary treatment approach that involves protecting oligodendrocytes, and the myelin they generate and maintain, from inflammatory-mediated death via enhancement of the integrated stress response (ISR). Studies in cells and mouse models of MS have demonstrated that the ISR, an innate protective pathway that maintains proteostasis, may be effectively harnessed to aid in the protection of oligodendrocytes and myelin during inflammation. With one ISR-modifying drug already in clinical trial and a number of potential future therapies under investigation, this approach may offer an important component in halting the progression of multiple sclerosis.
Collapse
Affiliation(s)
- Sharon W Way
- Department of Neurology, The University of Chicago Center for Peripheral Neuropathy, The University of Chicago, Chicago, IL, USA
| | - Brian Popko
- Department of Neurology, The University of Chicago Center for Peripheral Neuropathy, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
37
|
McClain JL, Fried DE, Gulbransen BD. Agonist-evoked Ca 2+ signaling in enteric glia drives neural programs that regulate intestinal motility in mice. Cell Mol Gastroenterol Hepatol 2015; 1:631-645. [PMID: 26693173 PMCID: PMC4673674 DOI: 10.1016/j.jcmgh.2015.08.004] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 08/14/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Gastrointestinal motility is regulated by enteric neural circuitry that includes enteric neurons and glia. Enteric glia monitor synaptic activity and exhibit responses to neurotransmitters that are encoded by intracellular calcium (Ca2+) signaling. What role evoked glial responses play in the neural regulation of gut motility is unknown. We tested how evoking Ca2+ signaling in enteric glia affects the neural control of intestinal motility. METHODS We used a novel chemogenetic mouse model that expresses the designer receptor hM3Dq under the transcriptional control of the glial fibrillary acidic protein (GFAP) promoter (GFAP::hM3Dq mice) to selectively trigger glial Ca2+ signaling. We used in situ Ca2+ imaging and immunohistochemistry to validate this model and assessed gut motility by measuring pellet output and composition, colonic bead expulsion time, small intestinal transit time, total gut transit time, colonic migrating motor complex (CMMC) recordings and muscle tension recordings. RESULTS hM3Dq receptor expression is confined to GFAP-positive enteric glia in the intestines of GFAP::hM3Dq mice. In these mice, application of the hM3Dq agonist clozapine-N-oxide (CNO) selectively triggers intracellular Ca2+ responses in enteric glia. Glial activation drove neurogenic contractions in the ileum and colon but had no effect on neurogenic relaxations. CNO enhanced the amplitude and frequency of CMMCs in ex vivo preparations of the colon and CNO increased colonic motility in vivo. CNO had no effect on the composition of fecal matter, small intestinal transit or whole gut transit. CONCLUSIONS Glial excitability encoded by intracellular Ca2+ signaling functions to modulate excitatory enteric circuits. Selectively triggering glial Ca2+ signaling might be a novel strategy to improve gut function in motility disorders.
Collapse
Affiliation(s)
| | - David E. Fried
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Brian D. Gulbransen
- Department of Physiology, Michigan State University, East Lansing, Michigan
- Neuroscience Program, Michigan State University, East Lansing, Michigan
| |
Collapse
|
38
|
Stone S, Lin W. The unfolded protein response in multiple sclerosis. Front Neurosci 2015; 9:264. [PMID: 26283904 PMCID: PMC4518158 DOI: 10.3389/fnins.2015.00264] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 07/14/2015] [Indexed: 01/08/2023] Open
Abstract
The unfolded protein response (UPR) occurs in response to endoplasmic reticulum (ER) stress caused by the accumulation of unfolded or misfolded proteins in the ER. The UPR is comprised of three signaling pathways that promote cytoprotective functions to correct ER stress; however, if ER stress cannot be resolved the UPR results in apoptosis of affected cells. The UPR is an important feature of various human diseases, including multiple sclerosis (MS). Recent studies have shown several components of the UPR are upregulated in the multiple cell types in MS lesions, including oligodendrocytes, T cells, microglia/macrophages, and astrocytes. Data from animal model studies, particularly studies of experimental autoimmune encephalomyelitis (EAE) and the cuprizone model, imply an important role of the UPR activation in oligodendrocytes in the development of MS. In this review we will cover current literature on the UPR and the evidence for its role in the development of MS.
Collapse
Affiliation(s)
- Sarrabeth Stone
- Department of Neuroscience, University of Minnesota Minneapolis, MN, USA ; Institute for Translational Neuroscience, University of Minnesota Minneapolis, MN, USA
| | - Wensheng Lin
- Department of Neuroscience, University of Minnesota Minneapolis, MN, USA ; Institute for Translational Neuroscience, University of Minnesota Minneapolis, MN, USA
| |
Collapse
|
39
|
Klingler S, Guo B, Yao J, Yan H, Zhang L, Vaseva AV, Chen S, Canoll P, Horner JW, Wang YA, Paik JH, Ying H, Zheng H. Development of Resistance to EGFR-Targeted Therapy in Malignant Glioma Can Occur through EGFR-Dependent and -Independent Mechanisms. Cancer Res 2015; 75:2109-19. [PMID: 25808866 DOI: 10.1158/0008-5472.can-14-3122] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 02/24/2015] [Indexed: 01/20/2023]
Abstract
Epidermal growth factor receptor (EGFR) is highly amplified, mutated, and overexpressed in human malignant gliomas. Despite its prevalence and growth-promoting functions, therapeutic strategies to inhibit EGFR kinase activity have not been translated into profound beneficial effects in glioma clinical trials. To determine the roles of oncogenic EGFR signaling in gliomagenesis and tumor maintenance, we generated a novel glioma mouse model driven by inducible expression of a mutant EGFR (EGFR*). Using combined genetic and pharmacologic interventions, we revealed that EGFR*-driven gliomas were insensitive to EGFR tyrosine kinase inhibitors, although they could efficiently inhibit EGFR* autophosphorylation in vitro and in vivo. This is in contrast with the genetic suppression of EGFR* induction that led to significant tumor regression and prolonged animal survival. However, despite their initial response to genetic EGFR* extinction, all tumors would relapse and propagate independent of EGFR*. We further showed that EGFR*-independent tumor cells existed prior to treatment and were responsible for relapse following genetic EGFR* suppression. And, the addition of a PI3K/mTOR inhibitor could significantly delay relapse and prolong animal survival. Our findings shed mechanistic insight into EGFR drug resistance in glioma and provide a platform to test therapies targeting aberrant EGFR signaling in this setting.
Collapse
Affiliation(s)
| | - Baofeng Guo
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Jun Yao
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Haiyan Yan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ling Zhang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | | | - Sida Chen
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University, New York, New York
| | - James W Horner
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Y Alan Wang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ji-Hye Paik
- Department of Pathology, Weill-Cornell Medical College, New York, New York
| | - Haoqiang Ying
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hongwu Zheng
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York.
| |
Collapse
|
40
|
Orr AG, Hsiao EC, Wang MM, Ho K, Kim DH, Wang X, Guo W, Kang J, Yu GQ, Adame A, Devidze N, Dubal DB, Masliah E, Conklin BR, Mucke L. Astrocytic adenosine receptor A2A and Gs-coupled signaling regulate memory. Nat Neurosci 2015; 18:423-34. [PMID: 25622143 PMCID: PMC4340760 DOI: 10.1038/nn.3930] [Citation(s) in RCA: 206] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 12/15/2014] [Indexed: 01/18/2023]
Abstract
Astrocytes express a variety of G protein-coupled receptors and might influence cognitive functions, such as learning and memory. However, the roles of astrocytic Gs-coupled receptors in cognitive function are not known. We found that humans with Alzheimer's disease (AD) had increased levels of the Gs-coupled adenosine receptor A2A in astrocytes. Conditional genetic removal of these receptors enhanced long-term memory in young and aging mice and increased the levels of Arc (also known as Arg3.1), an immediate-early gene that is required for long-term memory. Chemogenetic activation of astrocytic Gs-coupled signaling reduced long-term memory in mice without affecting learning. Like humans with AD, aging mice expressing human amyloid precursor protein (hAPP) showed increased levels of astrocytic A2A receptors. Conditional genetic removal of these receptors enhanced memory in aging hAPP mice. Together, these findings establish a regulatory role for astrocytic Gs-coupled receptors in memory and suggest that AD-linked increases in astrocytic A2A receptor levels contribute to memory loss.
Collapse
MESH Headings
- Alzheimer Disease/pathology
- Animals
- Animals, Newborn
- Astrocytes/metabolism
- Cytoskeletal Proteins/genetics
- Cytoskeletal Proteins/metabolism
- Exploratory Behavior/drug effects
- Exploratory Behavior/physiology
- Gene Expression Regulation/physiology
- Glial Fibrillary Acidic Protein/genetics
- Glial Fibrillary Acidic Protein/metabolism
- Humans
- Indoles/pharmacology
- Maze Learning/physiology
- Memory, Long-Term/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Receptor, Adenosine A2A/genetics
- Receptor, Adenosine A2A/metabolism
- Receptors, Serotonin, 5-HT4/genetics
- Receptors, Serotonin, 5-HT4/metabolism
- Recognition, Psychology/drug effects
- Recognition, Psychology/physiology
- Serotonin Antagonists/pharmacology
- Signal Transduction/physiology
- Sulfonamides/pharmacology
Collapse
Affiliation(s)
- Anna G. Orr
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
- Department of Neurology, University of California, San Francisco, CA 94158
| | - Edward C. Hsiao
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158
- Division of Endocrinology and Metabolism, Department of Medicine, and the Institute for Human Genetics, University of California, San Francisco, CA 94158
| | - Max M. Wang
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Kaitlyn Ho
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Daniel H. Kim
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Xin Wang
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Weikun Guo
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Jing Kang
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Gui-Qiu Yu
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Anthony Adame
- Departments of Neuroscience and Pathology, University of California, San Diego, La Jolla, CA
| | - Nino Devidze
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Dena B. Dubal
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
- Department of Neurology, University of California, San Francisco, CA 94158
| | - Eliezer Masliah
- Departments of Neuroscience and Pathology, University of California, San Diego, La Jolla, CA
| | - Bruce R. Conklin
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158
| | - Lennart Mucke
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
- Department of Neurology, University of California, San Francisco, CA 94158
| |
Collapse
|
41
|
The role of inflammatory cytokines as key modulators of neurogenesis. Trends Neurosci 2015; 38:145-57. [PMID: 25579391 DOI: 10.1016/j.tins.2014.12.006] [Citation(s) in RCA: 264] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 10/20/2014] [Accepted: 12/08/2014] [Indexed: 12/20/2022]
Abstract
Neurogenesis is an important process in the regulation of brain function and behaviour, highly active in early development and continuing throughout life. Recent studies have shown that neurogenesis is modulated by inflammatory cytokines in response to an activated immune system. To disentangle the effects of the different cytokines on neurogenesis, here we summarise and discuss in vitro studies on individual cytokines. We show that inflammatory cytokines have both a positive and negative role on proliferation and neuronal differentiation. Hence, this strengthens the notion that inflammation is involved in molecular and cellular mechanisms associated with complex cognitive processes and, therefore, that alterations in brain-immune communication are relevant to the development of neuropsychiatric disorders.
Collapse
|
42
|
Chew LJ, DeBoy CA, Senatorov VV. Finding degrees of separation: experimental approaches for astroglial and oligodendroglial cell isolation and genetic targeting. J Neurosci Methods 2014; 236:125-47. [PMID: 25169049 PMCID: PMC4171043 DOI: 10.1016/j.jneumeth.2014.08.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 08/15/2014] [Accepted: 08/18/2014] [Indexed: 12/20/2022]
Abstract
The study of CNS glial cell function requires experimental methods to detect, purify, and manipulate each cell population with fidelity and specificity. With the identification and cloning of cell- and stage-specific markers, glial cell analysis techniques have grown beyond physical methods of tissue dissociation and cell culture, and become highly specific with immunoselection of cell cultures in vitro and genetic targeting in vivo. The unique plasticity of glial cells offers the potential for cell replacement therapies in neurological disease that utilize neural cells derived from transplanted neural stem and progenitor cells. In this mini-review, we outline general physical and genetic approaches for macroglial cell generation. We summarize cell culture methods to obtain astrocytes and oligodendrocytes and their precursors, from developing and adult tissue, as well as approaches to obtain human neural progenitor cells through the establishment of stem cells. We discuss popular targeting rodent strains designed for cell-specific detection, selection and manipulation of neuroglial cell progenitors and their committed progeny. Based on shared markers between astrocytes and stem cells, we discuss genetically modified mouse strains with overlapping expression, and highlight SOX-expressing strains available for targeting of stem and progenitor cell populations. We also include recently established mouse strains for detection, and tag-assisted RNA and miRNA analysis. This discussion aims to provide a brief overview of the rapidly expanding collection of experimental approaches and genetic resources for the isolation and targeting of macroglial cells, their sources, progeny and gene products to facilitate our understanding of their properties and potential application in pathology.
Collapse
Affiliation(s)
- Li-Jin Chew
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC, United States.
| | - Cynthia A DeBoy
- Biology Department, Trinity Washington University, Washington, DC, United States
| | - Vladimir V Senatorov
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, United States
| |
Collapse
|
43
|
Tian F, Yourek G, Shi X, Yang Y. The development of Wilms tumor: from WT1 and microRNA to animal models. Biochim Biophys Acta Rev Cancer 2014; 1846:180-7. [PMID: 25018051 DOI: 10.1016/j.bbcan.2014.07.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 07/04/2014] [Accepted: 07/07/2014] [Indexed: 01/01/2023]
Abstract
Wilms tumor recapitulates the development of the kidney and represents a unique opportunity to understand the relationship between normal and tumor development. This has been illustrated by the findings that mutations of Wnt/β-catenin pathway-related WT1, β-catenin, and WTX together account for about one-third of Wilms tumor cases. While intense efforts are being made to explore the genetic basis of the other two-thirds of tumor cases, it is worth noting that, epigenetic changes, particularly the loss of imprinting of the DNA region encoding the major fetal growth factor IGF2, which results in its biallelic over-expression, are closely associated with the development of many Wilms tumors. Recent investigations also revealed that mutations of Drosha and Dicer, the RNases required for miRNA generation, and Dis3L2, the 3'-5' exonuclease that normally degrades miRNAs and mRNAs, could cause predisposition to Wilms tumors, demonstrating that miRNA can play a pivotal role in Wilms tumor development. Interestingly, Lin28, a direct target of miRNA let-7 and potent regulator of stem cell self-renewal and differentiation, is significantly elevated in some Wilms tumors, and enforced expression of Lin28 during kidney development could induce Wilms tumor. With the success in establishing mice nephroblastoma models through over-expressing IGF2 and deleting WT1, and advances in understanding the ENU-induced rat model, we are now able to explore the molecular and cellular mechanisms induced by these genetic, epigenetic, and miRNA alterations in animal models to understand the development of Wilms tumor. These animal models may also serve as valuable systems to assess new treatment targets and strategies for Wilms tumor.
Collapse
Affiliation(s)
- Fang Tian
- Department of Pathophysiology, School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, PR China
| | | | - Xiaolei Shi
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, PR China
| | - Yili Yang
- Center for Translational Medicine, Changzheng Hospital, Second Military Medical University, Shanghai, PR China.
| |
Collapse
|
44
|
Cekanaviciute E, Dietrich HK, Axtell RC, Williams AM, Egusquiza R, Wai KM, Koshy AA, Buckwalter MS. Astrocytic TGF-β signaling limits inflammation and reduces neuronal damage during central nervous system Toxoplasma infection. THE JOURNAL OF IMMUNOLOGY 2014; 193:139-49. [PMID: 24860191 DOI: 10.4049/jimmunol.1303284] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The balance between controlling infection and limiting inflammation is particularly precarious in the brain because of its unique vulnerability to the toxic effects of inflammation. Astrocytes have been implicated as key regulators of neuroinflammation in CNS infections, including infection with Toxoplasma gondii, a protozoan parasite that naturally establishes a chronic CNS infection in mice and humans. In CNS toxoplasmosis, astrocytes are critical to controlling parasite growth. They secrete proinflammatory cytokines and physically encircle parasites. However, the molecular mechanisms used by astrocytes to limit neuroinflammation during toxoplasmic encephalitis have not yet been identified. TGF-β signaling in astrocytes is of particular interest because TGF-β is universally upregulated during CNS infection and serves master regulatory and primarily anti-inflammatory functions. We report in this study that TGF-β signaling is activated in astrocytes during toxoplasmic encephalitis and that inhibition of astrocytic TGF-β signaling increases immune cell infiltration, uncouples proinflammatory cytokine and chemokine production from CNS parasite burden, and increases neuronal injury. Remarkably, we show that the effects of inhibiting astrocytic TGF-β signaling are independent of parasite burden and the ability of GFAP(+) astrocytes to physically encircle parasites.
Collapse
Affiliation(s)
- Egle Cekanaviciute
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305; Neurosciences Graduate Program, Stanford Neurosciences Institute, Stanford University, Stanford, CA 94305
| | - Hans K Dietrich
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, CA 94305; BIO5 Institute, University of Arizona, Tucson, AZ 85721
| | - Robert C Axtell
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305
| | - Aaron M Williams
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305
| | - Riann Egusquiza
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305
| | - Karen M Wai
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305
| | - Anita A Koshy
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305; Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, CA 94305; BIO5 Institute, University of Arizona, Tucson, AZ 85721; Department of Neurology, University of Arizona, Tucson, AZ 85721; Department of Immunobiology, University of Arizona, Tucson, AZ 85721; and
| | - Marion S Buckwalter
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305; Department of Neurosurgery, Stanford University, Stanford, CA 94305
| |
Collapse
|
45
|
Yang D, Baumann JM, Sun YY, Tang M, Dunn RS, Akeson AL, Kernie SG, Kallapur S, Lindquist DM, Huang EJ, Potter SS, Liang HC, Kuan CY. Overexpression of vascular endothelial growth factor in the germinal matrix induces neurovascular proteases and intraventricular hemorrhage. Sci Transl Med 2014; 5:193ra90. [PMID: 23843451 DOI: 10.1126/scitranslmed.3005794] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Intracranial hemorrhage in preterm neonates may result in neonatal mortality and functional disabilities, but its pathogenic mechanisms are poorly defined and better therapies are needed. We used a tetracycline-regulated transgenic system to test whether the induction of vascular endothelial growth factor (VEGF) in the germinal matrix leads to intracranial hemorrhage. This genetic strategy initially induced a dense network of loosely adjoined endothelial cells and pericytes near lateral ventricles, similar to the immature vascular rete in human fetal brains. Yet, this rich vascular network transformed into low-vasculature patches correlated with hemorrhage and caspase-3 activation near birth. Gene expression and biochemical analyses suggested that downstream mediators of VEGF in this network include transcriptional factors ETS1 and HIF2α (hypoxia-inducible factor 2α), components of the PDGFβ (platelet-derived growth factor β) and TGFβ (transforming growth factor-β) receptor signaling pathways, matrix metalloproteinase-9 (MMP-9), and cathepsins. Prenatal administration of glucocorticoids markedly reduced mortality and cerebral hemorrhage in mutant animals, as in human neonates. This protective effect was not due to blocking vasculogenesis, but was instead associated with inhibition of neurovascular proteases, notably MMP-9, cathepsin B, and caspase-3. Collectively, these results support a causative role of VEGF in perinatal cerebral hemorrhage and implicate its downstream proteases as potential therapeutic targets.
Collapse
Affiliation(s)
- Dianer Yang
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Effector molecules released by Th1 but not Th17 cells drive an M1 response in microglia. Brain Behav Immun 2014; 37:248-59. [PMID: 24412213 DOI: 10.1016/j.bbi.2014.01.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Revised: 12/23/2013] [Accepted: 01/02/2014] [Indexed: 12/15/2022] Open
Abstract
Microglia act as sensors of inflammation in the central nervous system (CNS) and respond to many stimuli. Other key players in neuroinflammatory diseases are CD4+ T helper cell (Th) subsets that characteristically secrete IFN-γ (Th1) or IL-17 (Th17). However, the potential of a distinct cytokine milieu generated by these effector T cell subsets to modulate microglial phenotype and function is poorly understood. We therefore investigated the ability of factors secreted by Th1 and Th17 cells to induce microglial activation. In vitro experiments wherein microglia were cultured in the presence of supernatants derived from polarized Th1 or Th17 cultures, revealed that Th1-associated factors could directly activate and trigger a proinflammatory M1-type gene expression profile in microglia that was cell-cell contact independent, whereas Th17 cells or its associated factors did not have any direct influence on microglia. To assess the effects of the key Th17 effector cytokine IL-17A in vivo we used transgenic mice in which IL-17A is specifically expressed in astrocytes. Flow cytometric and histological analysis revealed only subtle changes in the phenotype of microglia suggesting only minimal effects of constitutively produced IL-17A on microglia in vivo. Neither IL-23 signaling nor addition of GM-CSF, a recently described effector molecule of Th17 cells, changed the incapacity of Th17 cells to activate microglia. These findings demonstrate a potent effect of Th1 cells on microglia, however, the mechanism of how Th17 cells achieve their effect in CNS inflammation remains unclear.
Collapse
|
47
|
Lin Y, Huang G, Jamison S, Li J, Harding HP, Ron D, Lin W. PERK activation preserves the viability and function of remyelinating oligodendrocytes in immune-mediated demyelinating diseases. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 184:507-19. [PMID: 24269558 DOI: 10.1016/j.ajpath.2013.10.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 09/24/2013] [Accepted: 10/23/2013] [Indexed: 10/26/2022]
Abstract
Remyelination occurs in multiple sclerosis (MS) lesions but is generally considered to be insufficient. One of the major challenges in MS research is to understand the causes of remyelination failure and to identify therapeutic targets that promote remyelination. Activation of pancreatic endoplasmic reticulum kinase (PERK) signaling in response to endoplasmic reticulum stress modulates cell viability and function under stressful conditions. There is evidence that PERK is activated in remyelinating oligodendrocytes in demyelinated lesions in both MS and its animal model, experimental autoimmune encephalomyelitis (EAE). In this study, we sought to determine the role of PERK signaling in remyelinating oligodendrocytes in MS and EAE using transgenic mice that allow temporally controlled activation of PERK signaling specifically in oligodendrocytes. We demonstrated that persistent PERK activation was not deleterious to myelinating oligodendrocytes in young, developing mice or to remyelinating oligodendrocytes in cuprizone-induced demyelinated lesions. We found that enhancing PERK activation, specifically in (re)myelinating oligodendrocytes, protected the cells and myelin against the detrimental effects of interferon-γ, a key proinflammatory cytokine in MS and EAE. More important, we showed that enhancing PERK activation in remyelinating oligodendrocytes at the recovery stage of EAE promoted cell survival and remyelination in EAE demyelinated lesions. Thus, our data provide direct evidence that PERK activation cell-autonomously enhances the survival and preserves function of remyelinating oligodendrocytes in immune-mediated demyelinating diseases.
Collapse
Affiliation(s)
- Yifeng Lin
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Guangcun Huang
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Stephanie Jamison
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Jin Li
- Department of Ophthalmology, 9th Hospital, Shanghai Jiaotong University School of Medical Science, Shanghai, China
| | - Heather P Harding
- University of Cambridge Metabolic Research Laboratories, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, United Kingdom
| | - David Ron
- University of Cambridge Metabolic Research Laboratories, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, United Kingdom
| | - Wensheng Lin
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
48
|
Liu W, Zhang L, Wu R. Differential expression of STAT1 and IFN-γ in primary and invasive or metastatic wilms tumors. J Surg Oncol 2013; 108:152-6. [PMID: 23794088 DOI: 10.1002/jso.23364] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2013] [Accepted: 05/28/2013] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND OBJECTIVES IFN/STAT1 signaling has been found to be not only associated with an aggressive tumor phenotype but also activated and functional during metanephric development. This study was undertaken to evaluate STAT1 and IFN-γ expression and its relation to histopathological features of primary and invasive/metastatic Wilms tumors. METHODS Immunohistochemistry was used to determine the expression and cellular distribution of STAT1 and IFN-γ in 18 pairs of primary and corresponding invasive/metastatic Wilms tumors and 40 primary tumors without invasion or metastasis. RESULTS Positive rate of STAT1/IFN-γ expression was 66.7%/61.1% and 72.2%/77.8% in 18 pairs of primary and associated invasive/metastatic Wilms tumor tissues, while 35.0%/27.5% in 40 primary tumors without invasion or metastasis. The expression of STAT1 and IFN-γ was significantly associated with invasion/metastasis (P = 0.025; P = 0.015). There was a positive correlation between STAT1 and IFN-γ expression in all Wilms tumor tissues (χ(2) = 23.408, P = 0.05, r = 0.555). The expression of STAT1 and IFN-γ between primary and matched invasive/metastatic tissues was concordance, respectively (P = 0.710 and P = 0.375). CONCLUSIONS These results suggest that IFN-γ/STAT1 signaling might have clinical potential as a promising predictor to identify individuals with poor prognostic potential and as a possible novel target molecule of therapy for Wilms tumor.
Collapse
Affiliation(s)
- Wei Liu
- Department of Pediatric Surgery, Provincial Hospital Affiliated to Shandong University, Jinan, China
| | | | | |
Collapse
|
49
|
Heng MY, Lin ST, Verret L, Huang Y, Kamiya S, Padiath QS, Tong Y, Palop JJ, Huang EJ, Ptácχek LJ, Fu YH. Lamin B1 mediates cell-autonomous neuropathology in a leukodystrophy mouse model. J Clin Invest 2013; 123:2719-29. [PMID: 23676464 PMCID: PMC3668844 DOI: 10.1172/jci66737] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 03/19/2013] [Indexed: 01/20/2023] Open
Abstract
Adult-onset autosomal-dominant leukodystrophy (ADLD) is a progressive and fatal neurological disorder characterized by early autonomic dysfunction, cognitive impairment, pyramidal tract and cerebellar dysfunction, and white matter loss in the central nervous system. ADLD is caused by duplication of the LMNB1 gene, which results in increased lamin B1 transcripts and protein expression. How duplication of LMNB1 leads to myelin defects is unknown. To address this question, we developed a mouse model of ADLD that overexpresses lamin B1. These mice exhibited cognitive impairment and epilepsy, followed by age-dependent motor deficits. Selective overexpression of lamin B1 in oligodendrocytes also resulted in marked motor deficits and myelin defects, suggesting these deficits are cell autonomous. Proteomic and genome-wide transcriptome studies indicated that lamin B1 overexpression is associated with downregulation of proteolipid protein, a highly abundant myelin sheath component that was previously linked to another myelin-related disorder, Pelizaeus-Merzbacher disease. Furthermore, we found that lamin B1 overexpression leads to reduced occupancy of Yin Yang 1 transcription factor at the promoter region of proteolipid protein. These studies identify a mechanism by which lamin B1 overexpression mediates oligodendrocyte cell-autonomous neuropathology in ADLD and implicate lamin B1 as an important regulator of myelin formation and maintenance during aging.
Collapse
Affiliation(s)
- Mary Y. Heng
- Department of Neurology, UCSF, San Francisco, California, USA.
Gladstone Institute of Neurological Disease, San Francisco,
California, USA. Howard Hughes Medical Institute, San Francisco,
California, USA. Department of Pathology, UCSF, San Francisco,
California, USA. Veterans Affairs Medical Center, San Francisco,
California, USA
| | - Shu-Ting Lin
- Department of Neurology, UCSF, San Francisco, California, USA.
Gladstone Institute of Neurological Disease, San Francisco,
California, USA. Howard Hughes Medical Institute, San Francisco,
California, USA. Department of Pathology, UCSF, San Francisco,
California, USA. Veterans Affairs Medical Center, San Francisco,
California, USA
| | - Laure Verret
- Department of Neurology, UCSF, San Francisco, California, USA.
Gladstone Institute of Neurological Disease, San Francisco,
California, USA. Howard Hughes Medical Institute, San Francisco,
California, USA. Department of Pathology, UCSF, San Francisco,
California, USA. Veterans Affairs Medical Center, San Francisco,
California, USA
| | - Yong Huang
- Department of Neurology, UCSF, San Francisco, California, USA.
Gladstone Institute of Neurological Disease, San Francisco,
California, USA. Howard Hughes Medical Institute, San Francisco,
California, USA. Department of Pathology, UCSF, San Francisco,
California, USA. Veterans Affairs Medical Center, San Francisco,
California, USA
| | - Sherry Kamiya
- Department of Neurology, UCSF, San Francisco, California, USA.
Gladstone Institute of Neurological Disease, San Francisco,
California, USA. Howard Hughes Medical Institute, San Francisco,
California, USA. Department of Pathology, UCSF, San Francisco,
California, USA. Veterans Affairs Medical Center, San Francisco,
California, USA
| | - Quasar S. Padiath
- Department of Neurology, UCSF, San Francisco, California, USA.
Gladstone Institute of Neurological Disease, San Francisco,
California, USA. Howard Hughes Medical Institute, San Francisco,
California, USA. Department of Pathology, UCSF, San Francisco,
California, USA. Veterans Affairs Medical Center, San Francisco,
California, USA
| | - Ying Tong
- Department of Neurology, UCSF, San Francisco, California, USA.
Gladstone Institute of Neurological Disease, San Francisco,
California, USA. Howard Hughes Medical Institute, San Francisco,
California, USA. Department of Pathology, UCSF, San Francisco,
California, USA. Veterans Affairs Medical Center, San Francisco,
California, USA
| | - Jorge J. Palop
- Department of Neurology, UCSF, San Francisco, California, USA.
Gladstone Institute of Neurological Disease, San Francisco,
California, USA. Howard Hughes Medical Institute, San Francisco,
California, USA. Department of Pathology, UCSF, San Francisco,
California, USA. Veterans Affairs Medical Center, San Francisco,
California, USA
| | - Eric J. Huang
- Department of Neurology, UCSF, San Francisco, California, USA.
Gladstone Institute of Neurological Disease, San Francisco,
California, USA. Howard Hughes Medical Institute, San Francisco,
California, USA. Department of Pathology, UCSF, San Francisco,
California, USA. Veterans Affairs Medical Center, San Francisco,
California, USA
| | - Louis J. Ptácχek
- Department of Neurology, UCSF, San Francisco, California, USA.
Gladstone Institute of Neurological Disease, San Francisco,
California, USA. Howard Hughes Medical Institute, San Francisco,
California, USA. Department of Pathology, UCSF, San Francisco,
California, USA. Veterans Affairs Medical Center, San Francisco,
California, USA
| | - Ying-Hui Fu
- Department of Neurology, UCSF, San Francisco, California, USA.
Gladstone Institute of Neurological Disease, San Francisco,
California, USA. Howard Hughes Medical Institute, San Francisco,
California, USA. Department of Pathology, UCSF, San Francisco,
California, USA. Veterans Affairs Medical Center, San Francisco,
California, USA
| |
Collapse
|
50
|
Hofer MJ, Campbell IL. Type I interferon in neurological disease-the devil from within. Cytokine Growth Factor Rev 2013; 24:257-67. [PMID: 23548179 DOI: 10.1016/j.cytogfr.2013.03.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 03/05/2013] [Indexed: 12/31/2022]
Abstract
The members of the type I interferon (IFN-I) family of cytokines are pleiotropic factors that have seminal roles in host defence, acting as antimicrobial and antitumor mediators as well as potent immunomodulatory factors that bridge the innate and adaptive immune responses. Despite these beneficial actions there is mounting evidence that link inappropriate or chronic production of IFN-I in the CNS to the development of a number of severe neuroinflammatory disorders. The most persuasive example is the genetically determined inflammatory encephalopathy, Aicardi-Goutières syndrome (AGS) in which patients have chronically elevated IFN-α production in the CNS. The presentation of AGS can often mimic congenital viral infection, however, molecular genetic studies have identified mutations in six genes that can cause AGS, most likely via dysregulated nucleic acid metabolism and activation of the innate immune response leading to increased intrathecal production of IFN-α. The role of IFN-α as a pathogenic factor in AGS and other neurological disorders has gained considerable support from experimental studies. In particular, a transgenic mouse model with CNS-restricted production of IFN-α replicates many of the cardinal neuropathologic features of AGS and reveal IFN-I to be the "devil from within", mediating molecular and cellular damage within the CNS. Thus, targeting IFN-I may be an effective strategy for the treatment of AGS as well as some other autoimmune and infectious neurological "interferonopathies".
Collapse
Affiliation(s)
- Markus J Hofer
- School of Molecular Bioscience and the Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia.
| | | |
Collapse
|