1
|
Yang W, Liu X, Fan D. Low CD3 level is a risk factor for amyotrophic lateral sclerosis: a Mendelian randomization study. Amyotroph Lateral Scler Frontotemporal Degener 2025; 26:64-72. [PMID: 39316061 DOI: 10.1080/21678421.2024.2407408] [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: 08/09/2024] [Revised: 09/06/2024] [Accepted: 09/16/2024] [Indexed: 09/25/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive and fatal disease characterized by neuronal degeneration of the spinal cord and brain and believed to be related to the immune system. In this study, our aim is to use Mendelian randomization (MR) to search for immune markers related to ALS. A total of 731 immune cell traits were included in this study. MR analysis was used to identify the causality between 731 immune cell traits (with 3,757 Europeans) and ALS (with 138,086 Europeans). Colocalization analysis was used to verify the found causality, protein-protein interaction prediction was used to look for the interacting proteins that are known to be involved in ALS. We found low expression levels of CD3 on central memory CD8+ T cell is risk factor for ALS (OR = 0.90, 95% CI: 0.86-0.95, P = 0.0000303). CD3 can interact with three ALS-related proteins: VCP, HLA-DRA and HLA-DRB5, which are associated with adaptive immune response. Our study reported for the first time that low-level CD3 is a risk factor for ALS and the possible mechanism, which could provide a potential strategy for ALS diagnosis and therapy.
Collapse
Affiliation(s)
- Wenzhi Yang
- Department of Neurology, Peking University Third Hospital Beijing, China and
- Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Xiangyi Liu
- Department of Neurology, Peking University Third Hospital Beijing, China and
- Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital Beijing, China and
- Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| |
Collapse
|
2
|
Zhou L, Liu X, Wu T, Liu Q, Jing M, Li H, Xu N, Tang H. Identification of survival related key genes and long-term survival specific differentially expressed genes related key miRNA network of primary glioblastoma. Heliyon 2024; 10:e28439. [PMID: 38601561 PMCID: PMC11004527 DOI: 10.1016/j.heliyon.2024.e28439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 04/12/2024] Open
Abstract
Primary glioblastoma(pGBM) is the most malignant tumor of the central nervous system. Radiotherapy, chemotherapy and surgical treatment have little effect on the survival of pGBM patients. The prognosis is often poorly once the tumor recurs. It is urgent to develop new therapies for patients. In recent years, studies have been clarified that miRNA have a powerful regulating effect on the genes. However, the main group of miRNAs in regulating long-term survival specific related genes of pGBM is still unclear. Given that the survival period of most glioma patients is relatively short, studying long-term survival patients with pGBM is of great value for this disease. Our study aim to identify key miRNAs with long-term survival related genes present in pGBM and uncover their potential mechanisms. The gene expression profiles of GSE53733, GSE15824, GSE30563, GSE50161 were obtained from the Gene Expression Omnibus database. Firstly, samples were divided into 3 groups according to its survival time and each group compare to the normal control group. Then we obtained differential expression genes (DEGs) with a long-term survival specific (LTSDEGs) and a short-term survival specific DEGs (STSDEGs). Next, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were conducted with LTSDEGs and STSDEGs together. Moreover, we used the UALCAN database to verify LTSDEGs and STSDEGs, and obtained long-term verified survival specific DEGs(LTVSDEGs) and short-term verified survival specific DEGs(STVSDEGs). Finally, we established the predicted key miRNAs-LTVSDEGs interaction network. The protein expressions of the top 4 LTVSDEGs were verified in the HPA database with immunohistochemical staining. In total, we found 260 genes changed in LTSDEGs and 822 genes changed in STSDEGs. GO and KEGG results shown that the major changes are focused on tumor metabolism. 9 LTVSDEGs and 18 STVSDEGs were verified in UALCAN database. As for protein expression verification in top 4 LTVSDEGs, ZNF630, BLVRB and RPA3 were verified, while TPBG was not detected. We obtained 59 key miRNA from the predicted key miRNAs-LTVSDEGs interaction network. 25 key miRNAs were verified using GSE90603. Finally, we constructed the key miRNAs-LTVSDEGs network using a Sankey diagram, including 25 miRNAs and 7 LTVSDEGs. In conclusion, our study shows that there is a close relationship between metabolic changes and survival in pGBM. Besides, we established a key miRNAs-LTVSDEGs network for pGBM, which could be the key path in prolonging the life of pGBM patients.
Collapse
Affiliation(s)
- Lingqi Zhou
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou, 510623, China
- Guangzhou Key Laboratory of Child Neurodevelopment, Guangzhou, 510623, China
- Institute Pasteur of Shanghai, Chinese Academy of Science, Shanghai, 200031, China
| | - Xuemei Liu
- Department of Gynecology, Shunde Hospital,Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, 528308, China
| | - Tong Wu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080,China
| | - Qundi Liu
- Guangdong Jiangmen Chinese Medicine College, Jiangmen, 529000, China
| | - Meilian Jing
- Guangdong Jiangmen Chinese Medicine College, Jiangmen, 529000, China
| | - Huahan Li
- Guangdong Jiangmen Chinese Medicine College, Jiangmen, 529000, China
| | - Ning Xu
- Department of Clinical Laboratory, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518111, China
| | - Hai Tang
- Guangdong Jiangmen Chinese Medicine College, Jiangmen, 529000, China
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080,China
| |
Collapse
|
3
|
Mimic S, Aru B, Pehlivanoğlu C, Sleiman H, Andjus PR, Yanıkkaya Demirel G. Immunology of amyotrophic lateral sclerosis - role of the innate and adaptive immunity. Front Neurosci 2023; 17:1277399. [PMID: 38105925 PMCID: PMC10723830 DOI: 10.3389/fnins.2023.1277399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/07/2023] [Indexed: 12/19/2023] Open
Abstract
This review aims to summarize the latest evidence about the role of innate and adaptive immunity in Amyotrophic Lateral Sclerosis (ALS). ALS is a devastating neurodegenerative disease affecting upper and lower motor neurons, which involves essential cells of the immune system that play a basic role in innate or adaptive immunity, that can be neurotoxic or neuroprotective for neurons. However, distinguishing between the sole neurotoxic or neuroprotective function of certain cells such as astrocytes can be challenging due to intricate nature of these cells, the complexity of the microenvironment and the contextual factors. In this review, in regard to innate immunity we focus on the involvement of monocytes/macrophages, microglia, the complement, NK cells, neutrophils, mast cells, and astrocytes, while regarding adaptive immunity, in addition to humoral immunity the most important features and roles of T and B cells are highlighted, specifically different subsets of CD4+ as well as CD8+ T cells. The role of autoantibodies and cytokines is also discussed in distinct sections of this review.
Collapse
Affiliation(s)
- Stefan Mimic
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Jean Giaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Başak Aru
- Immunology Department, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Cemil Pehlivanoğlu
- Immunology Department, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Hadi Sleiman
- Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Pavle R. Andjus
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Jean Giaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | | |
Collapse
|
4
|
Moțățăianu A, Șerban G, Andone S. The Role of Short-Chain Fatty Acids in Microbiota-Gut-Brain Cross-Talk with a Focus on Amyotrophic Lateral Sclerosis: A Systematic Review. Int J Mol Sci 2023; 24:15094. [PMID: 37894774 PMCID: PMC10606032 DOI: 10.3390/ijms242015094] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 09/27/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Amyotrophic lateral sclerosis is a devastating neurodegenerative disease characterized by the gradual loss of motor neurons in the brain and spinal cord, leading to progressive motor function decline. Unfortunately, there is no effective treatment, and its increasing prevalence is linked to an aging population, improved diagnostics, heightened awareness, and changing lifestyles. In the gastrointestinal system, the gut microbiota plays a vital role in producing metabolites, neurotransmitters, and immune molecules. Short-chain fatty acids, of interest for their potential health benefits, are influenced by a fiber- and plant-based diet, promoting a diverse and balanced gut microbiome. These fatty acids impact the body by binding to receptors on enteroendocrine cells, influencing hormones like glucagon-like peptide-1 and peptide YY, which regulate appetite and insulin sensitivity. Furthermore, these fatty acids impact the blood-brain barrier, neurotransmitter levels, and neurotrophic factors, and directly stimulate vagal afferent nerves, affecting gut-brain communication. The vagus nerve is a crucial link between the gut and the brain, transmitting signals related to appetite, inflammation, and various processes. Dysregulation of this pathway can contribute to conditions like obesity and irritable bowel syndrome. Emerging evidence suggests the complex interplay among these fatty acids, the gut microbiota, and environmental factors influences neurodegenerative processes via interconnected pathways, including immune function, anti-inflammation, gut barrier, and energy metabolism. Embracing a balanced, fiber-rich diet may foster a diverse gut microbiome, potentially impacting neurodegenerative disease risk. Comprehensive understanding requires further research into interventions targeting the gut microbiome and fatty acid production and their potential therapeutic role in neurodegeneration.
Collapse
Affiliation(s)
- Anca Moțățăianu
- 1st Neurology Clinic, Mures County Clinical Emergency Hospital, 540136 Târgu Mures, Romania
- Department of Neurology, University of Medicine, Pharmacy, Science and Technology of Târgu Mures ‘George Emil Palade’, 540142 Târgu Mures, Romania
| | - Georgiana Șerban
- Doctoral School, University of Medicine, Pharmacy, Science and Technology of Târgu Mures ‘George Emil Palade’, 540142 Târgu Mures, Romania
| | - Sebastian Andone
- 1st Neurology Clinic, Mures County Clinical Emergency Hospital, 540136 Târgu Mures, Romania
- Department of Neurology, University of Medicine, Pharmacy, Science and Technology of Târgu Mures ‘George Emil Palade’, 540142 Târgu Mures, Romania
| |
Collapse
|
5
|
Han Y, Wang B, Gao H, He C, Hua R, Liang C, Zhang S, Wang Y, Xin S, Xu J. Vagus Nerve and Underlying Impact on the Gut Microbiota-Brain Axis in Behavior and Neurodegenerative Diseases. J Inflamm Res 2022; 15:6213-6230. [PMID: 36386584 PMCID: PMC9656367 DOI: 10.2147/jir.s384949] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/20/2022] [Indexed: 11/10/2022] Open
Abstract
The gut microbiota is the most abundant and diverse microbiota in the human body and the vagus nerve is the most widely distributed and complex nerve in the body, both of them are essential in maintaining homeostasis. The most important phenomenon is how they coordinate to regulate functions, which has attracted the great attention of scientists. The academic literature on the correlation with a host of intestinal diseases and even systemic diseases has revealed the bidirectional communication between the gut microbiota and the brain, which can be carried out via multiple patterns. In the review, firstly, we have a general overview of the gut microbiota and the gut microbiota-brain axis. Secondly, according to the distribution characteristics of the vagus nerve, we analyzed and summarized its function in the intestinal tract. At the same time, we have summarized the underlying mechanism of some behavior changes such as depressive and anxiety-like behaviors and related neurodegenerative diseases caused by the vagus nerve and intestinal microecological environment disorders, and then we also analyzed inconsistency of the experimental evidence in order to propose novel strategies for the clinical practice.
Collapse
Affiliation(s)
- Yimin Han
- Department of Oral Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Boya Wang
- Undergraduate Student of 2018 Eight Program of Clinical Medicine, Peking University People’s Hospital, Beijing, 100083, People’s Republic of China
| | - Han Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Chengwei He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Rongxuan Hua
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Chen Liang
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Sitian Zhang
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Ying Wang
- Department of Dermatology, Beijing Tong Ren Hospital, Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Shuzi Xin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Jingdong Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People’s Republic of China
- Correspondence: Jingdong Xu, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, No. 10, Xitoutiao, Youanmenwai, Fengtai District, Beijing, 100069, People’s Republic of China, Tel/Fax +86 10-8391-1469, Email
| |
Collapse
|
6
|
Ashraf H, Solla P, Sechi LA. Current Advancement of Immunomodulatory Drugs as Potential Pharmacotherapies for Autoimmunity Based Neurological Diseases. Pharmaceuticals (Basel) 2022; 15:ph15091077. [PMID: 36145298 PMCID: PMC9504155 DOI: 10.3390/ph15091077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Dramatic advancement has been made in recent decades to understand the basis of autoimmunity-mediated neurological diseases. These diseases create a strong influence on the central nervous system (CNS) and the peripheral nervous system (PNS), leading to various clinical manifestations and numerous symptoms. Multiple sclerosis (MS) is the most prevalent autoimmune neurological disease while NMO spectrum disorder (NMOSD) is less common. Furthermore, evidence supports the presence of autoimmune mechanisms contributing to the pathogenesis of amyotrophic lateral sclerosis (ALS), which is a neurodegenerative disorder characterized by the progressive death of motor neurons. Additionally, autoimmunity is believed to be involved in the basis of Alzheimer’s and Parkinson’s diseases. In recent years, the prevalence of autoimmune-based neurological disorders has been elevated and current findings strongly suggest the role of pharmacotherapies in controlling the progression of autoimmune diseases. Therefore, this review focused on the current advancement of immunomodulatory drugs as novel approaches in the management of autoimmune neurological diseases and their future outlook.
Collapse
Affiliation(s)
- Hajra Ashraf
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Paolo Solla
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Leonardo Atonio Sechi
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
- Complex Structure of Microbology and Virology, AOU Sassari, 07100 Sassari, Italy
- Correspondence:
| |
Collapse
|
7
|
Mirian A, Moszczynski A, Soleimani S, Aubert I, Zinman L, Abrahao A. Breached Barriers: A Scoping Review of Blood-Central Nervous System Barrier Pathology in Amyotrophic Lateral Sclerosis. Front Cell Neurosci 2022; 16:851563. [PMID: 35431812 PMCID: PMC9009245 DOI: 10.3389/fncel.2022.851563] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Recent studies have implicated changes in the blood-central nervous system barriers (BCNSB) in amyotrophic lateral sclerosis (ALS). The objective of this scoping review is to synthesize the current evidence for BCNSB structure and functional abnormalities in ALS studies and propose how BCNSB pathology may impact therapeutic development. Methods A literature search was conducted using Ovid Medline, EMBASE, and Web of Science, from inception to November 2021 and limited to entries in English language. Simplified search strategy included the terms ALS/motor neuron disease and [BCNSB or blood-brain barrier (BBB) or blood-spinal cord barrier (BSCB)]. Henceforth, BCNSB is used as a term that is inclusive of the BBB and BSCB. Four independent reviewers conducted a title and abstract screening, hand-searched the reference lists of review papers, and performed a full text review of eligible studies. Included studies were original peer-reviewed full text publications, evaluating the structure and function of the BCNSB in preclinical models of ALS, clinical ALS, or postmortem human ALS tissue. There was no restriction on study design. The four reviewers independently extracted the data. Results The search retrieved 2,221 non-duplicated articles and 48 original studies were included in the synthesis. There was evidence that the integrity of the BCNSB is disrupted throughout the course of the disease in rodent models, beginning prior to symptom onset and detectable neurodegeneration. Increased permeability, pharmacoresistance with upregulated efflux transporters, and morphological changes in the supporting cells of the BCNSB, including pericytes, astrocytes, and endothelial cells were observed in animal models. BCNSB abnormalities were also demonstrated in postmortem studies of ALS patients. Therapeutic interventions targeting BCNSB dysfunction were associated with improved motor neuron survival in animal models of ALS. Conclusion BCNSB structural and functional abnormalities are likely implicated in ALS pathophysiology and may occur upstream to neurodegeneration. Promising therapeutic strategies targeting BCNSB dysfunction have been tested in animals and can be translated into ALS clinical trials.
Collapse
Affiliation(s)
- Ario Mirian
- Clinical Neurological Sciences, Western University, London Health Sciences, London, ON, Canada
| | | | - Serena Soleimani
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI, United States
| | - Isabelle Aubert
- Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Lorne Zinman
- Division of Neurology, Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Division of Neurology, Department of Medicine, Sunnybrook Health Science Centre, Toronto, ON, Canada
- Evaluative Clinical Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Agessandro Abrahao
- Division of Neurology, Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Division of Neurology, Department of Medicine, Sunnybrook Health Science Centre, Toronto, ON, Canada
- Evaluative Clinical Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Toronto, ON, Canada
| |
Collapse
|
8
|
Polgár TF, Meszlényi V, Nógrádi B, Körmöczy L, Spisák K, Tripolszki K, Széll M, Obál I, Engelhardt JI, Siklós L, Patai R. Passive Transfer of Blood Sera from ALS Patients with Identified Mutations Results in Elevated Motoneuronal Calcium Level and Loss of Motor Neurons in the Spinal Cord of Mice. Int J Mol Sci 2021; 22:ijms22189994. [PMID: 34576165 PMCID: PMC8470779 DOI: 10.3390/ijms22189994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/09/2021] [Accepted: 09/13/2021] [Indexed: 02/08/2023] Open
Abstract
Introduction: Previously, we demonstrated the degeneration of axon terminals in mice after repeated injections of blood sera from amyotrophic lateral sclerosis (ALS) patients with identified mutations. However, whether a similar treatment affects the cell body of motor neurons (MNs) remained unresolved. Methods: Sera from healthy individuals or ALS patients with a mutation in different ALS-related genes were intraperitoneally injected into ten-week-old male Balb/c mice (n = 3/serum) for two days. Afterward, the perikaryal calcium level was measured using electron microscopy. Furthermore, the optical disector method was used to evaluate the number of lumbar MNs. Results: The cytoplasmic calcium level of the lumbar MNs of the ALS-serum-treated mice, compared to untreated and healthy-serum-treated controls, was significantly elevated. While injections of the healthy serum did not reduce the number of MNs compared to the untreated control group, ALS sera induced a remarkable loss of MNs. Discussion: Similarly to the distant motor axon terminals, the injection of blood sera of ALS patients has a rapid degenerative effect on MNs. Analogously, the magnitude of the evoked changes was specific to the type of mutation; furthermore, the degeneration was most pronounced in the group treated with sera from ALS patients with a mutation in the chromosome 9 open reading frame 72 gene.
Collapse
Affiliation(s)
- Tamás F. Polgár
- Biological Research Centre, Institute of Biophysics, 62 Temesvári krt., 6726 Szeged, Hungary; (T.F.P.); (V.M.); (B.N.); (L.K.); (K.S.)
- Theoretical Medicine Doctoral School, University of Szeged, 97 Tisza Lajos krt., 6722 Szeged, Hungary
| | - Valéria Meszlényi
- Biological Research Centre, Institute of Biophysics, 62 Temesvári krt., 6726 Szeged, Hungary; (T.F.P.); (V.M.); (B.N.); (L.K.); (K.S.)
- Albert Szent-Györgyi Health Centre, Department of Neurology, University of Szeged, 6 Semmelweis u., 6725 Szeged, Hungary; (I.O.); (J.I.E.)
| | - Bernát Nógrádi
- Biological Research Centre, Institute of Biophysics, 62 Temesvári krt., 6726 Szeged, Hungary; (T.F.P.); (V.M.); (B.N.); (L.K.); (K.S.)
- Albert Szent-Györgyi Health Centre, Department of Neurology, University of Szeged, 6 Semmelweis u., 6725 Szeged, Hungary; (I.O.); (J.I.E.)
| | - Laura Körmöczy
- Biological Research Centre, Institute of Biophysics, 62 Temesvári krt., 6726 Szeged, Hungary; (T.F.P.); (V.M.); (B.N.); (L.K.); (K.S.)
| | - Krisztina Spisák
- Biological Research Centre, Institute of Biophysics, 62 Temesvári krt., 6726 Szeged, Hungary; (T.F.P.); (V.M.); (B.N.); (L.K.); (K.S.)
| | - Kornélia Tripolszki
- Department of Medical Genetics, University of Szeged, 4/B Szőkefalvi-Nagy Béla u., 6720 Szeged, Hungary; (K.T.); (M.S.)
| | - Márta Széll
- Department of Medical Genetics, University of Szeged, 4/B Szőkefalvi-Nagy Béla u., 6720 Szeged, Hungary; (K.T.); (M.S.)
- Dermatological Research Group, Hungarian Academy of Sciences, University of Szeged, 4/B Szőkefalvi-Nagy Béla u., 6720 Szeged, Hungary
| | - Izabella Obál
- Albert Szent-Györgyi Health Centre, Department of Neurology, University of Szeged, 6 Semmelweis u., 6725 Szeged, Hungary; (I.O.); (J.I.E.)
- Department of Neurology, Aalborg University Hospital, 15 Skovvej Sdr., 9000 Aalborg, Denmark
| | - József I. Engelhardt
- Albert Szent-Györgyi Health Centre, Department of Neurology, University of Szeged, 6 Semmelweis u., 6725 Szeged, Hungary; (I.O.); (J.I.E.)
| | - László Siklós
- Biological Research Centre, Institute of Biophysics, 62 Temesvári krt., 6726 Szeged, Hungary; (T.F.P.); (V.M.); (B.N.); (L.K.); (K.S.)
- Correspondence: (L.S.); (R.P.); Tel.: +36-62-599-611 (L.S.); +36-62-599-600/431 (R.P.)
| | - Roland Patai
- Biological Research Centre, Institute of Biophysics, 62 Temesvári krt., 6726 Szeged, Hungary; (T.F.P.); (V.M.); (B.N.); (L.K.); (K.S.)
- Correspondence: (L.S.); (R.P.); Tel.: +36-62-599-611 (L.S.); +36-62-599-600/431 (R.P.)
| |
Collapse
|
9
|
Beneficial Effects of Transplanted Human Bone Marrow Endothelial Progenitors on Functional and Cellular Components of Blood-Spinal Cord Barrier in ALS Mice. eNeuro 2021; 8:ENEURO.0314-21.2021. [PMID: 34479980 PMCID: PMC8451202 DOI: 10.1523/eneuro.0314-21.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 08/26/2021] [Accepted: 08/30/2021] [Indexed: 12/18/2022] Open
Abstract
Convincing evidence of blood-spinal cord barrier (BSCB) alterations has been demonstrated in amyotrophic lateral sclerosis (ALS) and barrier repair is imperative to prevent motor neuron dysfunction. We showed benefits of human bone marrow-derived CD34+ cells (hBM34+) and endothelial progenitor cells (hBM-EPCs) intravenous transplantation into symptomatic G93A SOD1 mutant mice on barrier reparative processes. These gains likely occurred by replacement of damaged endothelial cells, prolonging motor neuron survival. However, additional investigations are needed to confirm the effects of administered cells on integrity of the microvascular endothelium. The aim of this study was to determine tight junction protein levels, capillary pericyte coverage, microvascular basement membrane, and endothelial filamentous actin (F-actin) status in spinal cord capillaries of G93A SOD1 mutant mice treated with human bone marrow-derived stem cells. Tight junction proteins were detected in the spinal cords of cell-treated versus non-treated mice via Western blotting at four weeks after transplant. Capillary pericyte, basement membrane laminin, and endothelial F-actin magnitudes were determined in cervical/lumbar spinal cord tissues in ALS mice, including controls, by immunohistochemistry and fluorescent staining. Results showed that cell-treated versus media-treated ALS mice substantially increased tight junction protein levels, capillary pericyte coverage, basement membrane laminin immunoexpressions, and endothelial cytoskeletal F-actin fluorescent expressions. The greatest benefits were detected in mice receiving hBM-EPCs versus hBM34+ cells. These study results support treatment with a specific cell type derived from human bone marrow toward BSCB repair in ALS. Thus, hBM-EPCs may be advanced for clinical applications as a cell-specific approach for ALS therapy through restored barrier integrity.
Collapse
|
10
|
De Marchi F, Munitic I, Amedei A, Berry JD, Feldman EL, Aronica E, Nardo G, Van Weehaeghe D, Niccolai E, Prtenjaca N, Sakowski SA, Bendotti C, Mazzini L. Interplay between immunity and amyotrophic lateral sclerosis: Clinical impact. Neurosci Biobehav Rev 2021; 127:958-978. [PMID: 34153344 PMCID: PMC8428677 DOI: 10.1016/j.neubiorev.2021.06.027] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 05/07/2021] [Accepted: 06/17/2021] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a debilitating and rapidly fatal neurodegenerative disease. Despite decades of research and many new insights into disease biology over the 150 years since the disease was first described, causative pathogenic mechanisms in ALS remain poorly understood, especially in sporadic cases. Our understanding of the role of the immune system in ALS pathophysiology, however, is rapidly expanding. The aim of this manuscript is to summarize the recent advances regarding the immune system involvement in ALS, with particular attention to clinical translation. We focus on the potential pathophysiologic mechanism of the immune system in ALS, discussing local and systemic factors (blood, cerebrospinal fluid, and microbiota) that influence ALS onset and progression in animal models and people. We also explore the potential of Positron Emission Tomography to detect neuroinflammation in vivo, and discuss ongoing clinical trials of therapies targeting the immune system. With validation in human patients, new evidence in this emerging field will serve to identify novel therapeutic targets and provide realistic hope for personalized treatment strategies.
Collapse
Affiliation(s)
- Fabiola De Marchi
- Department of Neurology and ALS Centre, University of Piemonte Orientale, Maggiore Della Carità Hospital, Corso Mazzini 18, Novara, 28100, Italy
| | - Ivana Munitic
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, R. Matejcic 2, 51000, Rijeka, Croatia
| | - Amedeo Amedei
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - James D Berry
- Sean M. Healey & AMG Center for ALS, Department of Neurology, Massachusetts General Hospital, 165 Cambridge Street, Suite 600, Boston, MA, 02114, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Giovanni Nardo
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milanm, 20156, Italy
| | - Donatienne Van Weehaeghe
- Division of Nuclear Medicine, Department of Imaging and Pathology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Elena Niccolai
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Nikolina Prtenjaca
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, R. Matejcic 2, 51000, Rijeka, Croatia
| | - Stacey A Sakowski
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Caterina Bendotti
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milanm, 20156, Italy
| | - Letizia Mazzini
- Department of Neurology and ALS Centre, University of Piemonte Orientale, Maggiore Della Carità Hospital, Corso Mazzini 18, Novara, 28100, Italy.
| |
Collapse
|
11
|
Ca 2+ homeostasis in brain microvascular endothelial cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 362:55-110. [PMID: 34253298 DOI: 10.1016/bs.ircmb.2021.01.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Blood brain barrier (BBB) is formed by the brain microvascular endothelial cells (BMVECs) lining the wall of brain capillaries. Its integrity is regulated by multiple mechanisms, including up/downregulation of tight junction proteins or adhesion molecules, altered Ca2+ homeostasis, remodeling of cytoskeleton, that are confined at the level of BMVECs. Beside the contribution of BMVECs to BBB permeability changes, other cells, such as pericytes, astrocytes, microglia, leukocytes or neurons, etc. are also exerting direct or indirect modulatory effects on BBB. Alterations in BBB integrity play a key role in multiple brain pathologies, including neurological (e.g. epilepsy) and neurodegenerative disorders (e.g. Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis etc.). In this review, the principal Ca2+ signaling pathways in brain microvascular endothelial cells are discussed and their contribution to BBB integrity is emphasized. Improving the knowledge of Ca2+ homeostasis alterations in BMVECa is fundamental to identify new possible drug targets that diminish/prevent BBB permeabilization in neurological and neurodegenerative disorders.
Collapse
|
12
|
Ouali Alami N, Tang L, Wiesner D, Commisso B, Bayer D, Weishaupt J, Dupuis L, Wong P, Baumann B, Wirth T, Boeckers TM, Yilmazer-Hanke D, Ludolph A, Roselli F. Multiplexed chemogenetics in astrocytes and motoneurons restore blood-spinal cord barrier in ALS. Life Sci Alliance 2020; 3:3/11/e201900571. [PMID: 32900826 PMCID: PMC7479971 DOI: 10.26508/lsa.201900571] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 08/24/2020] [Accepted: 08/31/2020] [Indexed: 12/20/2022] Open
Abstract
Chemogenetic motoneuron excitation and astrocyte GPCR-Gi signaling restore blood–spinal cord barrier, disrupted in four ALS mouse models, revealing its role in disease progression but not initiation. Blood–spinal cord barrier (BSCB) disruption is thought to contribute to motoneuron (MN) loss in amyotrophic lateral sclerosis (ALS). It is currently unclear whether impairment of the BSCB is the cause or consequence of MN dysfunction and whether its restoration may be directly beneficial. We revealed that SOD1G93A, FUSΔNLS, TDP43G298S, and Tbk1+/− ALS mouse models commonly shared alterations in the BSCB, unrelated to motoneuron loss. We exploit PSAM/PSEM chemogenetics in SOD1G93A mice to demonstrate that the BSCB is rescued by increased MN firing, whereas inactivation worsens it. Moreover, we use DREADD chemogenetics, alone or in multiplexed form, to show that activation of Gi signaling in astrocytes restores BSCB integrity, independently of MN firing, with no effect on MN disease markers and dissociating them from BSCB disruption. We show that astrocytic levels of the BSCB stabilizers Wnt7a and Wnt5a are decreased in SOD1G93A mice and strongly enhanced by Gi signaling, although further decreased by MN inactivation. Thus, we demonstrate that BSCB impairment follows MN dysfunction in ALS pathogenesis but can be reversed by Gi-induced expression of astrocytic Wnt5a/7a.
Collapse
Affiliation(s)
- Najwa Ouali Alami
- Department of Neurology, Ulm University, Ulm, Germany.,International Graduate School in Molecular Medicine Ulm, Ulm, Germany.,Department of Neurology, Clinical Neuroanatomy, Ulm University, Ulm, Germany
| | - Linyun Tang
- Department of Neurology, Ulm University, Ulm, Germany
| | - Diana Wiesner
- Department of Neurology, Ulm University, Ulm, Germany.,German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | | | - David Bayer
- Department of Neurology, Ulm University, Ulm, Germany.,CEMMA Graduate School, Ulm University, Ulm, Germany
| | | | - Luc Dupuis
- Inserm U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence; Université de Strasbourg, Faculté de Médecine, Strasbourg, France
| | - Phillip Wong
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bernd Baumann
- Institute of Physiological Chemistry, Ulm University, Ulm, Germany
| | - Thomas Wirth
- Institute of Physiological Chemistry, Ulm University, Ulm, Germany
| | - Tobias M Boeckers
- German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany.,Department of Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | | | - Albert Ludolph
- Department of Neurology, Ulm University, Ulm, Germany.,German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Francesco Roselli
- Department of Neurology, Ulm University, Ulm, Germany .,German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| |
Collapse
|
13
|
Béland LC, Markovinovic A, Jakovac H, De Marchi F, Bilic E, Mazzini L, Kriz J, Munitic I. Immunity in amyotrophic lateral sclerosis: blurred lines between excessive inflammation and inefficient immune responses. Brain Commun 2020; 2:fcaa124. [PMID: 33134918 PMCID: PMC7585698 DOI: 10.1093/braincomms/fcaa124] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/07/2020] [Accepted: 07/13/2020] [Indexed: 12/11/2022] Open
Abstract
Despite wide genetic, environmental and clinical heterogeneity in amyotrophic lateral sclerosis, a rapidly fatal neurodegenerative disease targeting motoneurons, neuroinflammation is a common finding. It is marked by local glial activation, T cell infiltration and systemic immune system activation. The immune system has a prominent role in the pathogenesis of various chronic diseases, hence some of them, including some types of cancer, are successfully targeted by immunotherapeutic approaches. However, various anti-inflammatory or immunosuppressive therapies in amyotrophic lateral sclerosis have failed. This prompted increased scrutiny over the immune-mediated processes underlying amyotrophic lateral sclerosis. Perhaps the biggest conundrum is that amyotrophic lateral sclerosis pathogenesis exhibits features of three otherwise distinct immune dysfunctions-excessive inflammation, autoimmunity and inefficient immune responses. Epidemiological and genome-wide association studies show only minimal overlap between amyotrophic lateral sclerosis and autoimmune diseases, so excessive inflammation is usually thought to be secondary to protein aggregation, mitochondrial damage or other stresses. In contrast, several recently characterized amyotrophic lateral sclerosis-linked mutations, including those in TBK1, OPTN, CYLD and C9orf72, could lead to inefficient immune responses and/or damage pile-up, suggesting that an innate immunodeficiency may also be a trigger and/or modifier of this disease. In such cases, non-selective immunosuppression would further restrict neuroprotective immune responses. Here we discuss multiple layers of immune-mediated neuroprotection and neurotoxicity in amyotrophic lateral sclerosis. Particular focus is placed on individual patient mutations that directly or indirectly affect the immune system, and the mechanisms by which these mutations influence disease progression. The topic of immunity in amyotrophic lateral sclerosis is timely and relevant, because it is one of the few common and potentially malleable denominators in this heterogenous disease. Importantly, amyotrophic lateral sclerosis progression has recently been intricately linked to patient T cell and monocyte profiles, as well as polymorphisms in cytokine and chemokine receptors. For this reason, precise patient stratification based on immunophenotyping will be crucial for efficient therapies.
Collapse
Affiliation(s)
| | - Andrea Markovinovic
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
- ENCALS Center Zagreb, 10000 Zagreb, Croatia
| | - Hrvoje Jakovac
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, 51000 Rijeka, Croatia
| | - Fabiola De Marchi
- Department of Neurology, ALS Centre, University of Piemonte Orientale, “Maggiore della Carità” Hospital, 28100 Novara, Italy
| | - Ervina Bilic
- Department of Neurology, Clinical Hospital Centre Zagreb, 10000 Zagreb, Croatia
- ENCALS Center Zagreb, 10000 Zagreb, Croatia
| | - Letizia Mazzini
- Department of Neurology, ALS Centre, University of Piemonte Orientale, “Maggiore della Carità” Hospital, 28100 Novara, Italy
| | - Jasna Kriz
- CERVO Research Centre, Laval University, Quebec City, Quebec G1J 2G3, Canada
| | - Ivana Munitic
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
14
|
Gómez-Budia M, Konttinen H, Saveleva L, Korhonen P, Jalava PI, Kanninen KM, Malm T. Glial smog: Interplay between air pollution and astrocyte-microglia interactions. Neurochem Int 2020; 136:104715. [DOI: 10.1016/j.neuint.2020.104715] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 12/15/2022]
|
15
|
Bukhari SI, Alfawaz H, Al-Dbass A, Bhat RS, Moubayed NMS, Bukhari W, Hassan SA, Merghani N, Elsamaligy S, El-Ansary A. Efficacy of Novavit in ameliorating the neurotoxicity of propionic acid. Transl Neurosci 2020; 11:134-146. [PMID: 33312719 PMCID: PMC7705989 DOI: 10.1515/tnsci-2020-0103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 03/08/2020] [Accepted: 03/24/2020] [Indexed: 01/01/2023] Open
Abstract
Oxidative stress, abnormal fatty acid metabolism, and impaired gut microbiota play a serious role in the pathology of autism. The use of dietary supplements to improve the core symptoms of autism is a common therapeutic strategy. The present study analyzed the effects of oral supplementation with Novavit, a multi-ingredient supplement, on ameliorating oxidative stress and impaired lipid metabolism in a propionic acid (PPA)-induced rodent model of autism. Male western albino rats were divided into three groups. The first group is the control, the second group was given an oral neurotoxic dose of PPA (250 mg/kg body weight/day) for 3 days and then received buffered saline until the end of the experiment. The third group received Novavit (70 mg/kg body weight/day for 30 days after the 3-day PPA treatment). Markers of oxidative stress and impaired fatty acid metabolism were measured in brain homogenates obtained from each group. Novavit modulation of the gut microbiota was also evaluated. While PPA induced significant increases in lipid peroxides and 5-lipoxygenase, together with significantly decreased glutathione, and cyclooxygenase 2, oral supplementation with Novavit ameliorated PPA-induced oxidative stress and impaired fatty acid metabolism. Our results showed that the presence of multivitamins, coenzyme Q10, minerals, and colostrum, the major components of Novavit, protects against PPA-induced neurotoxicity.
Collapse
Affiliation(s)
- Sarah I. Bukhari
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hanan Alfawaz
- Department of Food Science and Nutrition, College of Food Science and Agriculture, King Saud University, Riyadh, Saudi Arabia
| | - Abeer Al-Dbass
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ramesa Shafi Bhat
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Nadine MS Moubayed
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Wadha Bukhari
- Central Laboratory, Female Center for Medical Studies and Scientific Section, King Saud University, P O Box 22452, Riyadh, Saudi Arabia
| | | | - Nada Merghani
- Central Laboratory, Female Center for Medical Studies and Scientific Section, King Saud University, P O Box 22452, Riyadh, Saudi Arabia
| | - Samar Elsamaligy
- Department of Pharamaceutics and Industrial Pharmacy, Helwan University, Ain Helwan, Cairo, Egypt
| | - Afaf El-Ansary
- Central Laboratory, Female Center for Medical Studies and Scientific Section, King Saud University, P O Box 22452, Riyadh, Saudi Arabia
- Therapeutic Department, National Research Centre, Dokki, Egypt
| |
Collapse
|
16
|
Abstract
The microbiome is the ecological community of commensal, symbiotic, and pathogenic microorganisms that share our body space (Medical and Health Genomics, 2016, page 15-28). The human gut is the location where the maximum number of microorganisms can be found. Among the different microorganisms they can be broadly classified into two groups: the beneficial and harmful. In the human gut there is always a balance between the beneficial and the opportunistic microorganism which maintains human health. However, if the balance is not maintained and homeostasis is disturbed, with an increase in opportunistic microorganisms, it may result in various diseases like inflammatory bowel disease, irritable bowel disease, ulcerative colitis, Crohn's disease, colorectal cancer, metabolic disorders and neurodegenerative diseases including motor neuron diseases. In the present chapter we discuss the role of gut bacteria in motor neuron diseases like multiple sclerosis, Parkinson's disease and amyotrophic lateral sclerosis.
Collapse
|
17
|
Phenotypic characteristics of human bone marrow-derived endothelial progenitor cells in vitro support cell effectiveness for repair of the blood-spinal cord barrier in ALS. Brain Res 2019; 1724:146428. [PMID: 31493389 DOI: 10.1016/j.brainres.2019.146428] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/18/2019] [Accepted: 08/28/2019] [Indexed: 01/01/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) was recently recognized as a neurovascular disease. Accumulating evidence demonstrated blood-spinal-cord barrier (BSCB) impairment mainly via endothelial cell (EC) degeneration in ALS patients and animal models. BSCB repair may be a therapeutic approach for ALS. We showed benefits of human bone marrow endothelial progenitor cell (hBMEPC) transplantation into symptomatic ALS mice on barrier restoration; however, cellular mechanisms remain unclear. The study aimed to characterize hBMEPCs in vitro under normogenic conditions. hBMEPCs were cultured at different time points. Enzyme-linked immunosorbent assay (ELISA) was used to detect concentrations of angiogenic factors (VEGF-A, angiogenin-1, and endoglin) and angiogenic inhibitor endostatin in conditioned media. Double immunocytochemical staining for CD105, ZO-1, and occludin with F-actin was performed. Results showed predominantly gradual significant post-culture increases of VEGF-A and angiogenin-1 levels. Cultured cells displayed distinct rounded or elongated cellular morphologies and positively immunoexpressed for CD105, indicating EC phenotype. Cytoskeletal F-actin filaments were re-arranged according to cell morphologies. Immunopositive expressions for ZO-1 were detected near inner cell membrane and for occludin on cell membrane surface of adjacent hBMEPCs. Together, secretion of angiogenic factors by cultured cells provides evidence for a potential mechanism underlying endogenous EC repair in ALS through hBMEPC transplantation, leading to restored barrier integrity. Also, ZO-1 and occludin immunoexpressions, confirming hBMEPC interactions in vitro, may reflect post-transplant cell actions in vivo.
Collapse
|
18
|
Human Bone Marrow Endothelial Progenitor Cell Transplantation into Symptomatic ALS Mice Delays Disease Progression and Increases Motor Neuron Survival by Repairing Blood-Spinal Cord Barrier. Sci Rep 2019; 9:5280. [PMID: 30918315 PMCID: PMC6437219 DOI: 10.1038/s41598-019-41747-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 03/14/2019] [Indexed: 12/11/2022] Open
Abstract
Convincing evidence demonstrated impairment of the blood-spinal cord barrier (BSCB) in Amyotrophic Lateral Sclerosis (ALS), mainly by endothelial cell (EC) alterations. Replacing damaged ECs by cell transplantation is a potential barrier repair strategy. Recently, we showed that intravenous (iv) administration of human bone marrow CD34+ (hBM34+) cells into symptomatic ALS mice benefits BSCB restoration and postpones disease progression. However, delayed effect on motor function and some severely damaged capillaries were noted. We hypothesized that hematopoietic cells from a restricted lineage would be more effective. This study aimed to establish the effects of human bone marrow-derived endothelial progenitor cells (hBMEPCs) systemically transplanted into G93A mice at symptomatic disease stage. Results showed that transplanted hBMEPCs significantly improved behavioral disease outcomes, engrafted widely into capillaries of the gray/white matter spinal cord and brain motor cortex/brainstem, substantially restored capillary ultrastructure, significantly decreased EB extravasation into spinal cord parenchyma, meaningfully re-established perivascular astrocyte end-feet, and enhanced spinal cord motor neuron survival. These results provide novel evidence that transplantation of hBMEPCs effectively repairs the BSCB, potentially preventing entry of detrimental peripheral factors, including immune/inflammatory cells, which contribute to motor neuron dysfunction. Transplanting EC progenitor cells may be a promising strategy for barrier repair therapy in this disease.
Collapse
|
19
|
Young TL, Zychowski KE, Denson JL, Campen MJ. Blood-brain barrier at the interface of air pollution-associated neurotoxicity and neuroinflammation. ROLE OF INFLAMMATION IN ENVIRONMENTAL NEUROTOXICITY 2019. [DOI: 10.1016/bs.ant.2018.10.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
20
|
Page S, Patel R, Raut S, Al-Ahmad A. Neurological diseases at the blood-brain barrier: Stemming new scientific paradigms using patient-derived induced pluripotent cells. Biochim Biophys Acta Mol Basis Dis 2018; 1866:165358. [PMID: 30593893 DOI: 10.1016/j.bbadis.2018.12.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/15/2018] [Accepted: 12/05/2018] [Indexed: 02/08/2023]
Abstract
The blood-brain barrier (BBB) is a component of the neurovascular unit formed by specialized brain microvascular endothelial cells (BMECs) surrounded by a specific basement membrane interacting with astrocytes, neurons, and pericytes. The BBB plays an essential function in the maintenance of brain homeostasis, by providing a physical and chemical barrier against pathogens and xenobiotics. Although the disruption of the BBB occurs with several neurological disorders, the scarcity of patient material source and lack of reliability of current in vitro models hindered our ability to model the BBB during such neurological conditions. The development of novel in vitro models based on patient-derived stem cells opened new venues in modeling the human BBB in vitro, by being more accurate than existing in vitro models, but also bringing such models closer to the in vivo setting. In addition, patient-derived models of the BBB opens the avenue to address the contribution of genetic factors commonly associated with certain neurological diseases on the BBB pathophysiology. This review provides a comprehensive understanding of the BBB, the current development of stem cell-based models in the field, the current challenges and limitations of such models.
Collapse
Affiliation(s)
- Shyanne Page
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, TX, United States of America
| | - Ronak Patel
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, TX, United States of America
| | - Snehal Raut
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, TX, United States of America
| | - Abraham Al-Ahmad
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, TX, United States of America.
| |
Collapse
|
21
|
Shigemoto-Mogami Y, Hoshikawa K, Sato K. Activated Microglia Disrupt the Blood-Brain Barrier and Induce Chemokines and Cytokines in a Rat in vitro Model. Front Cell Neurosci 2018; 12:494. [PMID: 30618641 PMCID: PMC6300509 DOI: 10.3389/fncel.2018.00494] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 11/30/2018] [Indexed: 12/25/2022] Open
Abstract
Severe neuroinflammation is associated with blood brain barrier (BBB) disruption in CNS diseases. Although microglial activation and the subsequent changes in cytokine/chemokine (C/C) concentrations are thought to be key steps in the development of neuroinflammation, little data are available concerning the interaction of microglia with BBB cells. In this study, we investigated this interaction by adding LPS-activated microglia (LPS-MG) to the abluminal side of a BBB model composed of endothelial cells (EC), pericytes (Peri) and astrocytes (Ast). We then examined the abluminal concentrations of 27 C/Cs and the interactions between the LPS-MG and BBB cells. LPS-MG caused collapse of the BBB, revealed by decreases in the trans-endothelial electrical resistance (TEER) and by changes in the expression levels of tight junction (TJ) proteins. Under these conditions, 19 C/Cs were markedly increased on the abluminal side. Unexpectedly, although LPS-MG alone released 10 of the 19 C/Cs, their concentrations were much lower than those detected on the abluminal side of the BBB model supplemented with LPS-MG. Co-culture of LPS-MG with Ast caused marked increases in 12 of the 19 C/Cs, while co-culture of LPS-MG with EC and Peri resulted in a significant increase in only 1 of the 19 C/Cs (fractalkine). These results suggest that C/C dynamics in this system are not only caused by activated microglia but also are due to the interaction between activated microglia and astrocytes.
Collapse
Affiliation(s)
- Yukari Shigemoto-Mogami
- Laboratory of Neuropharmacology, Division of Pharmacology, National Institute of Health Sciences, Kawasaki-ku, Japan.,Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kazue Hoshikawa
- Laboratory of Neuropharmacology, Division of Pharmacology, National Institute of Health Sciences, Kawasaki-ku, Japan
| | - Kaoru Sato
- Laboratory of Neuropharmacology, Division of Pharmacology, National Institute of Health Sciences, Kawasaki-ku, Japan
| |
Collapse
|
22
|
Transplantation of human bone marrow stem cells into symptomatic ALS mice enhances structural and functional blood-spinal cord barrier repair. Exp Neurol 2018; 310:33-47. [PMID: 30172620 DOI: 10.1016/j.expneurol.2018.08.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 08/27/2018] [Accepted: 08/29/2018] [Indexed: 12/11/2022]
Abstract
Accumulating evidence shows alterations in the blood-brain barrier (BBB) and blood-spinal cord barrier (BSCB) in ALS patients and in animal models of disease, mainly by endothelial cell (EC) damage. Repair of the altered barrier in the CNS by replacement of ECs via cell transplantation may be a new therapeutic approach for ALS. Recently, we demonstrated positive effects towards BSCB repair by intravenous administration of unmodified human bone marrow CD34+ (hBM34+) cells at different doses into symptomatic ALS mice. However, particular benefits of these transplanted cells on microvascular integrity in symptomatic ALS mice are still unclear. The aim of the present study was to determine the structural and functional spinal cord capillary integrity in symptomatic ALS mice after intravenous administration of hBM34+ cells. The G93A mice at 13 weeks of age intravenously received one of three different cell doses (5 × 104, 5 × 105, or 1 × 106) and were euthanized at 17 weeks of age (4 weeks post-transplant). Control groups were media-treated and non-carrier mutant SOD1 gene mice. Capillary ultrastructural (electron microscopy), immunohistochemical (laminin and HuNu), and histological (myelin and capillary density) analyses were performed in the cervical and lumbar spinal cords. Capillary permeability in the spinal cords was determined by Evans Blue (EB) injection. Results showed significant restoration of ultrastructural capillary morphology, improvement of basement membrane integrity, enhancement of axonal myelin coherence, and stabilization of capillary density in the spinal cords primarily of ALS mice receiving the high dose of 1 × 106 cells. Moreover, substantial reduction of parenchymal EB levels was determined in these mice, confirming our previous results on capillary permeability. Additionally, transplanted cells were detected in blood smears of sacrificed late symptomatic mice by HuNu marker. Altogether, these results provide novel evidence that unmodified bone marrow hematopoietic stem cell treatment at optimal dose might be beneficial for structural and functional repair of the damaged BSCB in advanced stage of ALS, potentially resulting in delayed disease progression by increased motor neuron survival.
Collapse
|
23
|
Spielman LJ, Gibson DL, Klegeris A. Unhealthy gut, unhealthy brain: The role of the intestinal microbiota in neurodegenerative diseases. Neurochem Int 2018; 120:149-163. [PMID: 30114473 DOI: 10.1016/j.neuint.2018.08.005] [Citation(s) in RCA: 186] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 08/09/2018] [Accepted: 08/13/2018] [Indexed: 02/08/2023]
Abstract
The number of bacterial cells living within the human body is approximately equal to, or greater than, the total number of human cells. This dynamic population of microorganisms, termed the human microbiota, resides mainly within the gastrointestinal tract. It is widely accepted that highly diverse and stable microbiota promote overall human health. Colonization of the gut with maladaptive and pathogenic microbiota, a state also known as dysbiosis, is associated with a variety of peripheral diseases ranging from type 2 diabetes mellitus to cardiovascular and inflammatory bowel disease. More recently, microbial dysbiosis has been associated with a number of brain pathologies, including autism spectrum disorder, Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS), suggesting a direct or indirect communication between intestinal bacteria and the central nervous system (CNS). In this review, we illustrate two pathways implicated in the crosstalk between gut microbiota and CNS involving 1) the vagus nerve and 2) transmission of signaling molecules through the circulatory system and across the blood-brain barrier (BBB). We summarize the available evidence of the specific changes in the intestinal microbiota, as well as microorganism-induced modifications to intestinal and BBB permeability, which have been linked to several neurodegenerative disorders including ALS, AD, and PD. Even though each of these diseases arises from unique pathogenetic mechanisms, all are characterized, at least in part, by chronic neuroinflammation. We provide an interpretation for the substantial evidence that healthy intestinal microbiota have the ability to positively regulate the neuroimmune responses in the CNS. Even though the evidence is mainly associative, it has been suggested that bacterial dysbiosis could contribute to an adverse neuroinflammatory state leading to increased risk of neurodegenerative diseases. Thus, developing strategies for regulating and maintaining healthy intestinal microbiota could be a valid approach for lowering individual risk and prevalence of neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Deanna Lynn Gibson
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, Canada
| | - Andis Klegeris
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, Canada
| |
Collapse
|
24
|
Milošević M, Milićević K, Božić I, Lavrnja I, Stevanović I, Bijelić D, Dubaić M, Živković I, Stević Z, Giniatullin R, Andjus P. Immunoglobulins G from Sera of Amyotrophic Lateral Sclerosis Patients Induce Oxidative Stress and Upregulation of Antioxidative System in BV-2 Microglial Cell Line. Front Immunol 2017; 8:1619. [PMID: 29218049 PMCID: PMC5703705 DOI: 10.3389/fimmu.2017.01619] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 11/08/2017] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder with a very fast progression, no diagnostic tool for the presymptomatic phase, and still no effective treatment of the disease. Although ALS affects motor neurons, the overall pathophysiological condition points out to the non-cell autonomous mechanisms, where astrocytes and microglia play crucial roles in the disease progression. We have already shown that IgG from sera of ALS patients (ALS IgG) induce calcium transients and an increase in the mobility of acidic vesicles in cultured rat astrocytes. Having in mind the role of microglia in neurodegeneration, and a well-documented fact that oxidative stress is one of the many components contributing to the disease, we decided to examine the effect of ALS IgG on activation, oxidative stress and antioxidative system of BV-2 microglia, and to evaluate their acute effect on cytosolic peroxide, pH, and on reactive oxygen species (ROS) generation. All tested ALS IgGs (compared to control IgG) induced oxidative stress (rise in nitric oxide and the index of lipid peroxidation) followed by release of TNF-α and higher antioxidative defense (elevation of Mn- and CuZn-superoxide dismutase, catalase, and glutathione reductase with a decrease of glutathione peroxidase and glutathione) after 24 h treatment. Both ALS IgG and control IgG showed same localization on the membrane of BV-2 cells following 24 h treatment. Cytosolic peroxide and pH alteration were evaluated with fluorescent probes HyPer and SypHer, respectively, having in mind that HyPer also reacts to pH changes. Out of 11 tested IgGs from ALS patients, 4 induced slow exponential rise of HyPer signal, with maximal normalized fluorescence in the range 0.2–0.5, also inducing similar increase of SypHer intensity, but of a lower amplitude. None of the control IgGs induced changes with neither of the indicators. Acute ROS generation was detected in one out of three tested ALS samples with carboxy-H2DCFDA. The observed phenomena demonstrate the potential role of inflammatory humoral factors, IgGs, as potential triggers of the activation in microglia, known to occur in later stages of ALS. Therefore, revealing the ALS IgG signaling cascade in microglial cells could offer a valuable molecular biomarker and/or a potential therapeutic target.
Collapse
Affiliation(s)
- Milena Milošević
- Center for Laser Microscopy, Department for Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Katarina Milićević
- Center for Laser Microscopy, Department for Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Iva Božić
- Institute for Biological Research "Siniša Stanković", University of Belgrade, Belgrade, Serbia
| | - Irena Lavrnja
- Institute for Biological Research "Siniša Stanković", University of Belgrade, Belgrade, Serbia
| | - Ivana Stevanović
- Institute for Medical Research, Military Medical Academy, Belgrade, Serbia
| | - Dunja Bijelić
- Center for Laser Microscopy, Department for Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Marija Dubaić
- Center for Laser Microscopy, Department for Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Irena Živković
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia
| | - Zorica Stević
- Clinic of Neurology, Clinical Center of Serbia, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Rashid Giniatullin
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.,Laboratory in Neurobiology, Kazan Federal University, Kazan, Russia
| | - Pavle Andjus
- Center for Laser Microscopy, Department for Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
25
|
Deng S, Liu H, Qiu K, You H, Lei Q, Lu W. Role of the Golgi Apparatus in the Blood-Brain Barrier: Golgi Protection May Be a Targeted Therapy for Neurological Diseases. Mol Neurobiol 2017; 55:4788-4801. [PMID: 28730529 DOI: 10.1007/s12035-017-0691-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 07/13/2017] [Indexed: 12/17/2022]
Abstract
The blood-brain barrier (BBB) protects the brain from toxic material in the blood, provides nutrients for brain tissues, and screens harmful substances from the brain. The specific brain microvascular endothelial cells (BMVECs), tight junction between endothelial cells, and astrocytes ensure proper function of the central nervous system (CNS). Pathological factors disrupt the integrity of the BBB by destroying the normal function of endothelial cells and decreasing the production of tight junction proteins or the expression of proteins specifically localized on astrocytes. Interestingly, fragmentation of the Golgi apparatus is observed in neurological diseases and is involved in the destruction of the BBB function. The Golgi acts as a processing center in which proteins are transported after being processed in the endoplasmic reticulum. Besides reprocessing, classifying, and packaging proteins, the Golgi apparatus (GA) also acts as a signaling platform and calcium pool. In this review, we summarized the current literature on the potential relationship between the Golgi and endothelial cells, tight junction, and astrocytes. The normal function of the BBB is maintained as long as the normal function and morphology of the GA are not disturbed. Furthermore, we speculate that protecting the Golgi may be a novel therapeutic approach to protect the BBB and treat neurological diseases due to BBB dysfunction.
Collapse
Affiliation(s)
- Shuwen Deng
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China
| | - Hui Liu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China
| | - Ke Qiu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China
| | - Hong You
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China
| | - Qiang Lei
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China
| | - Wei Lu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China.
| |
Collapse
|
26
|
Vatine GD, Al-Ahmad A, Barriga BK, Svendsen S, Salim A, Garcia L, Garcia VJ, Ho R, Yucer N, Qian T, Lim RG, Wu J, Thompson LM, Spivia WR, Chen Z, Van Eyk J, Palecek SP, Refetoff S, Shusta EV, Svendsen CN. Modeling Psychomotor Retardation using iPSCs from MCT8-Deficient Patients Indicates a Prominent Role for the Blood-Brain Barrier. Cell Stem Cell 2017; 20:831-843.e5. [PMID: 28526555 PMCID: PMC6659720 DOI: 10.1016/j.stem.2017.04.002] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/29/2016] [Accepted: 04/07/2017] [Indexed: 12/27/2022]
Abstract
Inactivating mutations in the thyroid hormone (TH) transporter Monocarboxylate transporter 8 (MCT8) cause severe psychomotor retardation in children. Animal models do not reflect the biology of the human disease. Using patient-specific induced pluripotent stem cells (iPSCs), we generated MCT8-deficient neural cells that showed normal TH-dependent neuronal properties and maturation. However, the blood-brain barrier (BBB) controls TH entry into the brain, and reduced TH availability to neural cells could instead underlie the diseased phenotype. To test potential BBB involvement, we generated an iPSC-based BBB model of MCT8 deficiency, and we found that MCT8 was necessary for polarized influx of the active form of TH across the BBB. We also found that a candidate drug did not appreciably cross the mutant BBB. Our results therefore clarify the underlying physiological basis of this disorder, and they suggest that circumventing the diseased BBB to deliver active TH to the brain could be a viable therapeutic strategy.
Collapse
Affiliation(s)
- Gad D Vatine
- The Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Abraham Al-Ahmad
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Bianca K Barriga
- The Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Soshana Svendsen
- The Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ariel Salim
- The Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Leslie Garcia
- The Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Veronica J Garcia
- The Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ritchie Ho
- The Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Nur Yucer
- The Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Tongcheng Qian
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Ryan G Lim
- Department of Biological Chemistry, University of California, Irvine (UCI), Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders, University of California, Irvine (UCI), Irvine, CA 92697, USA
| | - Jie Wu
- Department of Biological Chemistry, University of California, Irvine (UCI), Irvine, CA 92697, USA
| | - Leslie M Thompson
- Department of Biological Chemistry, University of California, Irvine (UCI), Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders, University of California, Irvine (UCI), Irvine, CA 92697, USA; Department of Neurobiology and Behavior, University of California, Irvine (UCI), Irvine, CA 92697, USA; Department of Psychiatry and Human Behavior, University of California, Irvine (UCI), Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Center, University of California, Irvine (UCI), Irvine, CA 92697, USA
| | - Weston R Spivia
- Advanced Clinical Biosystems Research Institute, Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Zhaohui Chen
- Advanced Clinical Biosystems Research Institute, Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jennifer Van Eyk
- Advanced Clinical Biosystems Research Institute, Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Samuel Refetoff
- Department of Medicine, Pediatrics and Committee on Genetics, The University of Chicago, Chicago, IL 60637, USA
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA.
| | - Clive N Svendsen
- The Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
27
|
Choi JY, Yoon SS, Kim SE, Ahn Jo S. KDM4B histone demethylase and G9a regulate expression of vascular adhesion proteins in cerebral microvessels. Sci Rep 2017; 7:45005. [PMID: 28327608 PMCID: PMC5361151 DOI: 10.1038/srep45005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 02/17/2017] [Indexed: 12/16/2022] Open
Abstract
Intercellular adhesion molecule 1 (ICAM1) mediates the adhesion and transmigration of leukocytes across the endothelium, promoting inflammation. We investigated the epigenetic mechanism regulating ICAM1 expression. The pro-inflammatory cytokine TNF-α dramatically increased ICAM1 mRNA and protein levels in human brain microvascular endothelial cells and mouse brain microvessels. Chromatin immunoprecipitation revealed that TNF-α reduced methylation of histone H3 at lysines 9 and 27 (H3K9 and H3K27), well-known residues involved in gene suppression. Inhibition of G9a and EZH2, histone methyltransferases responsible for methylation at H3K9 and H3K27, respectively as well as G9a overexpression demonstrated the involvement of G9a in TNF-α-induced ICAM1 expression and leukocyte adhesion and transmigration. A specific role for KDM4B, a histone demethylase targeting H3K9me2, in TNF-α-induced ICAM1 upregulation was validated with siRNA. Moreover, treating mice with a KDM4 inhibitor ML324 blocked TNF-α-mediated neutrophil adhesion. Similarly, TNF-α-induced VCAM1 expression was suppressed by G9a overexpression and KDM4B knockdown. Collectively, we demonstrated that modification of H3K9me2 by G9a and KDM4B regulates expression of vascular adhesion molecules, and that depletion of these proteins or KDM4B reduces inflammation-induced leukocyte extravasation. Thus, blocking ICAM1 or KDM4B could offer a novel therapeutic opportunity treating brain diseases.
Collapse
Affiliation(s)
- Ji-Young Choi
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 330-714, South Korea
| | - Sang-Sun Yoon
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 330-714, South Korea
| | - Sang-Eun Kim
- Department of Pharmacology, College of Pharmacy, Dankook University, Cheonan 330-714, South Korea
| | - Sangmee Ahn Jo
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 330-714, South Korea
- Department of Pharmacology, College of Pharmacy, Dankook University, Cheonan 330-714, South Korea
| |
Collapse
|
28
|
Wu B, De SK, Kulinich A, Salem AF, Koeppen J, Wang R, Barile E, Wang S, Zhang D, Ethell I, Pellecchia M. Potent and Selective EphA4 Agonists for the Treatment of ALS. Cell Chem Biol 2017; 24:293-305. [PMID: 28196613 DOI: 10.1016/j.chembiol.2017.01.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 09/16/2016] [Accepted: 01/19/2017] [Indexed: 12/16/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive degenerative disease that affects motor neurons. Recent studies identified the receptor tyrosine kinase EphA4 as a disease-modifying gene that is critical for the progression of motor neuron degeneration. We report on the design and characterization of a family of EphA4 targeting agents that bind to its ligand binding domain with nanomolar affinity. The molecules exhibit excellent selectivity and display efficacy in a SOD1 mutant mouse model of ALS. Interestingly, the molecules appear to act as agonists for the receptor in certain surrogate cellular assays. While the exact mechanisms responsible for the therapeutic effect of the new agonists remain to be elucidated, we believe that the described agent represents both an invaluable pharmacological tool to further decipher the role of the EphA4 in ALS and potentially other human diseases, and a significant stepping stone for the development of novel treatments.
Collapse
Affiliation(s)
- Bainan Wu
- Sanford-Burnham-Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Surya K De
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, 900 University Avenue, Riverside, CA 92521, USA
| | - Anna Kulinich
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, 900 University Avenue, Riverside, CA 92521, USA
| | - Ahmed F Salem
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, 900 University Avenue, Riverside, CA 92521, USA
| | - Jordan Koeppen
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, 900 University Avenue, Riverside, CA 92521, USA
| | - Rengang Wang
- Sanford-Burnham-Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA; Brain Development, University of California San Diego, San Diego, CA 92090, USA
| | - Elisa Barile
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, 900 University Avenue, Riverside, CA 92521, USA
| | - Si Wang
- Sanford-Burnham-Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Dongxiang Zhang
- Sanford-Burnham-Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Iryna Ethell
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, 900 University Avenue, Riverside, CA 92521, USA
| | - Maurizio Pellecchia
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, 900 University Avenue, Riverside, CA 92521, USA.
| |
Collapse
|