1
|
Wellman SM, Forrest AM, Douglas MM, Subbaraman A, Zhang G, Kozai TDY. Dynamic changes in the structure and function of brain mural cells around chronically implanted microelectrodes. Biomaterials 2025; 315:122963. [PMID: 39547137 DOI: 10.1016/j.biomaterials.2024.122963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/25/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
Integration of neural interfaces with minimal tissue disruption in the brain is ideal to develop robust tools that can address essential neuroscience questions and combat neurological disorders. However, implantation of intracortical devices provokes severe tissue inflammation within the brain, which requires a high metabolic demand to support a complex series of cellular events mediating tissue degeneration and wound healing. Pericytes, peri-vascular cells involved in blood-brain barrier maintenance, vascular permeability, waste clearance, and angiogenesis, have recently been implicated as potential perpetuators of neurodegeneration in brain injury and disease. While the intimate relationship between pericytes and the cortical microvasculature have been explored in other disease states, their behavior following microelectrode implantation, which is responsible for direct blood vessel disruption and dysfunction, is currently unknown. Using two-photon microscopy we observed dynamic changes in the structure and function of pericytes during implantation of a microelectrode array over a 4-week implantation period. Pericytes respond to electrode insertion through transient increases in intracellular calcium and underlying constriction of capillary vessels. Within days following the initial insertion, we observed an influx of new, proliferating pericytes which contribute to new blood vessel formation. Additionally, we discovered a potentially novel population of reactive immune cells in close proximity to the electrode-tissue interface actively engaging in encapsulation of the microelectrode array. Finally, we determined that intracellular pericyte calcium can be modulated by intracortical microstimulation in an amplitude- and frequency-dependent manner. This study provides a new perspective on the complex biological sequelae occurring at the electrode-tissue interface and will foster new avenues of potential research consideration and lead to development of more advanced therapeutic interventions towards improving the biocompatibility of neural electrode technology.
Collapse
Affiliation(s)
- Steven M Wellman
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Adam M Forrest
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Madeline M Douglas
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ashwat Subbaraman
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Guangfeng Zhang
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, USA.
| |
Collapse
|
2
|
Chen X, Habib S, Alexandru M, Chauhan J, Evan T, Troka JM, Rahimi A, Esapa B, Tull TJ, Ng WZ, Fitzpatrick A, Wu Y, Geh JLC, Lloyd-Hughes H, Palhares LCGF, Adams R, Bax HJ, Whittaker S, Jacków-Malinowska J, Karagiannis SN. Chondroitin Sulfate Proteoglycan 4 (CSPG4) as an Emerging Target for Immunotherapy to Treat Melanoma. Cancers (Basel) 2024; 16:3260. [PMID: 39409881 PMCID: PMC11476251 DOI: 10.3390/cancers16193260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 10/20/2024] Open
Abstract
Immunotherapies, including checkpoint inhibitor antibodies, have precipitated significant improvements in clinical outcomes for melanoma. However, approximately half of patients do not benefit from approved treatments. Additionally, apart from Tebentafusp, which is approved for the treatment of uveal melanoma, there is a lack of immunotherapies directly focused on melanoma cells. This is partly due to few available targets, especially those expressed on the cancer cell surface. Chondroitin sulfate proteoglycan 4 (CSPG4) is a cell surface molecule overexpressed in human melanoma, with restricted distribution and low expression in non-malignant tissues and involved in several cancer-promoting and dissemination pathways. Here, we summarize the current understanding of the expression and functional significance of CSPG4 in health and melanoma, and we outline immunotherapeutic strategies. These include monoclonal antibodies, antibody-drug conjugates (ADCs), chimeric-antigen receptor (CAR) T cells, and other strategies such as anti-idiotypic and mimotope vaccines to raise immune responses against CSPG4-expressing melanomas. Several showed promising functions in preclinical models of melanoma, yet few have reached clinical testing, and none are approved for therapeutic use. Obstacles preventing that progress include limited knowledge of CSPG4 function in human cancer and a lack of in vivo models that adequately represent patient immune responses and human melanoma biology. Despite several challenges, immunotherapy directed to CSPG4-expressing melanoma harbors significant potential to transform the treatment landscape.
Collapse
Affiliation(s)
- Xinyi Chen
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Shabana Habib
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Madalina Alexandru
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Jitesh Chauhan
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Theodore Evan
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Joanna M. Troka
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Avigail Rahimi
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Benjamina Esapa
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Thomas J. Tull
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Wen Zhe Ng
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Amanda Fitzpatrick
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
- Oncology Department, Guy’s and St Thomas’ Hospitals, London SE1 9RT, UK
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King’s College London, Innovation Hub, Guy’s Hospital, London SE1 9RT, UK
| | - Yin Wu
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King’s College London, Innovation Hub, Guy’s Hospital, London SE1 9RT, UK
- Peter Gorer Department of Immunobiology, Centre for Inflammation Biology and Cancer Immunology, School of Immunology and Microbial Sciences, King’s College London, London SE1 9RT, UK
| | - Jenny L. C. Geh
- St John’s Institute of Dermatology, Guy’s, King’s and St. Thomas’ Hospitals NHS Foundation Trust, London SE1 9RT, UK
- Department of Plastic Surgery, Guy’s, King’s and St. Thomas’ Hospitals, London SE1 9RT, UK
| | - Hawys Lloyd-Hughes
- Department of Plastic Surgery, Guy’s, King’s and St. Thomas’ Hospitals, London SE1 9RT, UK
| | - Lais C. G. F. Palhares
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Rebecca Adams
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Heather J. Bax
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Sean Whittaker
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Joanna Jacków-Malinowska
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Sophia N. Karagiannis
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King’s College London, Innovation Hub, Guy’s Hospital, London SE1 9RT, UK
| |
Collapse
|
3
|
Bottero M, Pessina G, Bason C, Vigo T, Uccelli A, Ferrara G. Nerve-Glial antigen 2: unmasking the enigmatic cellular identity in the central nervous system. Front Immunol 2024; 15:1393842. [PMID: 39136008 PMCID: PMC11317297 DOI: 10.3389/fimmu.2024.1393842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 07/05/2024] [Indexed: 08/15/2024] Open
Abstract
Chondroitin sulfate proteoglycans (CSPGs) are fundamental components of the extracellular matrix in the central nervous system (CNS). Among these, the Nerve-Glial antigen 2 (NG2) stands out as a transmembrane CSPG exclusively expressed in a different population of cells collectively termed NG2-expressing cells. These enigmatic cells, found throughout the developing and adult CNS, have been indicated with various names, including NG2 progenitor cells, polydendrocytes, synantocytes, NG2 cells, and NG2-Glia, but are more commonly referred to as oligodendrocyte progenitor cells. Characterized by high proliferation rates and unique morphology, NG2-expressing cells stand apart from neurons, astrocytes, and oligodendrocytes. Intriguingly, some NG2-expressing cells form functional glutamatergic synapses with neurons, challenging the long-held belief that only neurons possess the intricate machinery required for neurotransmission. In the CNS, the complexity surrounding NG2-expressing cells extends to their classification. Additionally, NG2 expression has been documented in pericytes and immune cells, suggesting a role in regulating brain innate immunity and neuro-immune crosstalk in homeostasis. Ongoing debates revolve around their heterogeneity, potential as progenitors for various cell types, responses to neuroinflammation, and the role of NG2. Therefore, this review aims to shed light on the enigma of NG2-expressing cells by delving into their structure, functions, and signaling pathways. We will critically evaluate the literature on NG2 expression across the CNS, and address the contentious issues surrounding their classification and roles in neuroinflammation and neurodegeneration. By unraveling the intricacies of NG2-expressing cells, we hope to pave the way for a more comprehensive understanding of their contributions to CNS health and during neurological disorders.
Collapse
Affiliation(s)
- Marta Bottero
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Giada Pessina
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | | | - Tiziana Vigo
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Antonio Uccelli
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Neurology, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | | |
Collapse
|
4
|
KUROKAWA T, IMAI K. Chondroitin sulfate proteoglycan 4: An attractive target for antibody-based immunotherapy. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2024; 100:293-308. [PMID: 38735753 PMCID: PMC11260911 DOI: 10.2183/pjab.100.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/11/2024] [Indexed: 05/14/2024]
Abstract
Multifunctional molecules involved in tumor progression and metastasis have been identified as valuable targets for immunotherapy. Among these, chondroitin sulfate proteoglycan 4 (CSPG4), a significant tumor cell membrane-bound proteoglycan, has emerged as a promising target, especially in light of advances in chimeric antigen receptor (CAR) T-cell therapy. The profound bioactivity of CSPG4 and its role in pivotal processes such as tumor proliferation, migration, and neoangiogenesis underline its therapeutic potential. We reviewed the molecular intricacies of CSPG4, its functional attributes within tumor cells, and the latest clinical-translational advances targeting it. Strategies such as blocking monoclonal antibodies, conjugate therapies, bispecific antibodies, small-molecule inhibitors, CAR T-cell therapies, trispecific killer engagers, and ribonucleic acid vaccines against CSPG4 were assessed. CSPG4 overexpression in diverse tumors and its correlation with adverse prognostic outcomes emphasize its significance in cancer biology. These findings suggest that targeting CSPG4 offers a promising avenue for future cancer therapy, with potential synergistic effects when combined with existing treatments.
Collapse
Affiliation(s)
- Tomohiro KUROKAWA
- Department of Medical Epigenomics Research, Fukushima Medical University, Fukushima, Japan
- Department of Surgery, Jyoban Hospital of Tokiwa Foundation, Fukushima, Japan
| | - Kohzoh IMAI
- Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
5
|
Larabee JL, Doyle DA, Ahmed UKB, Shadid TM, Sharp RR, Jones KL, Kim YM, Li S, Ballard JD. Discovery of Hippo signaling as a regulator of CSPG4 expression and as a therapeutic target for Clostridioides difficile disease. PLoS Pathog 2023; 19:e1011272. [PMID: 36972308 PMCID: PMC10079225 DOI: 10.1371/journal.ppat.1011272] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 04/06/2023] [Accepted: 03/08/2023] [Indexed: 03/29/2023] Open
Abstract
The signaling pathways and networks regulating expression of chondroitin sulfate proteoglycan 4 (CSPG4), a cancer-related protein that serves as a receptor for Clostridiodes difficile TcdB, are poorly defined. In this study, TcdB-resistant/CSPG4-negative HeLa cells were generated by exposure to increasing concentrations of the toxin. The cells that emerged (HeLa R5) lost expression of CSPG4 mRNA and were resistant to binding by TcdB. mRNA expression profiles paired with integrated pathway analysis correlated changes in the Hippo and estrogen signaling pathways with a CSPG4 decrease in HeLa R5 cells. Both signaling pathways altered CSPG4 expression when modulated chemically or through CRISPR-mediated deletion of key transcriptional regulators in the Hippo pathway. Based on the in vitro findings, we predicted and experimentally confirmed that a Hippo pathway inactivating drug (XMU-MP-1) provides protection from C. difficile disease in a mouse model. These results provide insights into key regulators of CSPG4 expression and identify a therapeutic for C. difficile disease.
Collapse
Affiliation(s)
- Jason L. Larabee
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - D. Annie Doyle
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Ummey Khalecha Bintha Ahmed
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Tyler M. Shadid
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Rachel R. Sharp
- Laboratory for Molecular Biology and Cytometry Research, Harold Hamm Diabetes Center, Oklahoma City, Oklahoma, United States of America
| | - Kenneth L. Jones
- Laboratory for Molecular Biology and Cytometry Research, Harold Hamm Diabetes Center, Oklahoma City, Oklahoma, United States of America
- Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Young Mi Kim
- Department of Pediatrics, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Shibo Li
- Department of Pediatrics, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Jimmy D. Ballard
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| |
Collapse
|
6
|
Bassotti G, Fruganti A, Stracci F, Marconi P, Fettucciari K. Cytotoxic synergism of Clostridioides difficile toxin B with proinflammatory cytokines in subjects with inflammatory bowel diseases. World J Gastroenterol 2023; 29:582-596. [PMID: 36742168 PMCID: PMC9896618 DOI: 10.3748/wjg.v29.i4.582] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/03/2022] [Accepted: 12/27/2022] [Indexed: 01/20/2023] Open
Abstract
Clostridioides difficile (C. difficile) is progressively colonizing humans and animals living with humans. During this process, hypervirulent strains and mutated toxin A and B of C. difficile (TcdA and TcdB) are originating and developing. While in healthy subjects colonization by C. difficile becomes a risk after the use of antibiotics that alter the microbiome, other categories of people are more susceptible to infection and at risk of relapse, such as those with inflammatory bowel disease (IBD). Recent in vitro studies suggest that this increased susceptibility could be due to the strong cytotoxic synergism between TcdB and proinflammatory cytokines the tumor necrosis factor-alpha and interferon-gamma (CKs). Therefore, in subjects with IBD the presence of an inflammatory state in the colon could be the driver that increases the susceptibility to C. difficile infection and its progression and relapses. TcdB is internalized in the cell via three receptors: chondroitin sulphate proteoglycan 4; poliovirus receptor-like 3; and Wnt receptor frizzled family. Chondroitin sulphate proteoglycan 4 and Wnt receptor frizzled family are involved in cell death by apoptosis or necrosis depending on the concentration of TcdB and cell types, while poliovirus receptor-like 3 induces only necrosis. It is possible that cytokines could also induce a greater expression of receptors for TcdB that are more involved in necrosis than in apoptosis. Therefore, in subjects with IBD there are the conditions: (1) For greater susceptibility to C. difficile infection, such as the inflammatory state, and abnormalities of the microbiome and of the immune system; (2) for the enhancement of the cytotoxic activity of TcdB +Cks; and (3) for a greater expression of TcdB receptors stimulated by cytokines that induce cell death by necrosis rather than apoptosis. The only therapeutic approach currently possible in IBD patients is monitoring of C. difficile colonization for interventions aimed at reducing tumor necrosis factor-alpha and interferon-gamma levels when the infection begins. The future perspective is to generate bacteriophages against C. difficile for targeted therapy.
Collapse
Affiliation(s)
- Gabrio Bassotti
- Department of Medicine and Surgery, Gastroenterology, Hepatology & Digestive Endoscopy Section University of Perugia Medical School, Piazza Lucio Severi, Perugia 06132, Italy, and Santa Maria della Misericordia Hospital, Gastroenterology & Hepatology Unit Perugia 06156, Italy
| | - Alessandro Fruganti
- School of Biosciences and Veterinary Medicine, University of Camerino, Matelica 62024, Italy
| | - Fabrizio Stracci
- Medicine and Surgery, Hygiene and Public Health Section, University of Perugia, Perugia 06123, Italy
| | - Pierfrancesco Marconi
- Medicine and Surgery, Biosciences & Medical Embryology Section, University of Perugia, Perugia 06132, Italy
| | - Katia Fettucciari
- Medicine and Surgery, Biosciences & Medical Embryology Section, University of Perugia, Perugia 06132, Italy
| |
Collapse
|
7
|
Scheller A, Meyer E. Pathology-induced NG2 proteoglycan expression in microglia. Neural Regen Res 2023; 18:801-802. [DOI: 10.4103/1673-5374.353488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
8
|
Xu L, He D, Zhang C, Bai Y, Zhang C. The regulate function of polysaccharides and oligosaccharides that with sulfate group on immune-related disease. J Funct Foods 2022. [DOI: 10.1016/j.jff.2021.104870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
9
|
Du X, Zhang Z, Zhou H, Zhou J. Differential Modulators of NG2-Glia Differentiation into Neurons and Glia and Their Crosstalk. Cell Mol Neurobiol 2021; 41:1-15. [PMID: 32285247 PMCID: PMC11448640 DOI: 10.1007/s10571-020-00843-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/06/2020] [Indexed: 02/08/2023]
Abstract
As the fifth main cell population in the brain, NG2-glia are also known as oligodendrocyte precursor cells. NG2-glia express receptors and ion channels for fast modulation of neuronal activities and signaling with neuronal synapses, which are of functional significance in both physiological and pathological states. NG2-glia also participate in fast signaling with peripheral neurons via direct synaptic contacts in the brain. These distinctive glia have the unique capability of proliferating and differentiating into oligodendrocytes, which are critical for axonal myelination in the early developing brain. In neurodegenerative diseases, NG2-glia play an important role and undergo morphological modification, adapt the expression of their membrane receptors and ion channels, and display gene-modulated cell reprogramming and excitotoxicity-caused cell death. These modifications directly and indirectly influence populations of neurons and other glial cells. NG2-glia regulate their action and dynamics in response to neuronal behavior and disease, indicating a critical function to preserve and remodel myelin in physiological states and to repair it in pathological states. Here, we review in detail the differential modulators of NG2-glia into neurons and astrocytes, as well as interactions of NG2-glia with neurons, astrocytes, and microglia. We will also summarize a future potential exploitation of NG2-glia.
Collapse
Affiliation(s)
- Xiaohuang Du
- Department of Scientific Research, Army Medical University, Chongqing, 400037, China
| | - Zuo Zhang
- National Drug Clinical Trial Institution, Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China
| | - Hongli Zhou
- National Drug Clinical Trial Institution, Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China
| | - Jiyin Zhou
- National Drug Clinical Trial Institution, Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China.
| |
Collapse
|
10
|
Ito N, Sasaki K, Takemoto H, Kobayashi Y, Isoda H, Odaguchi H. Emotional Impairments and Neuroinflammation are Induced in Male Mice Invulnerable to Repeated Social Defeat Stress. Neuroscience 2020; 443:148-163. [PMID: 32707290 DOI: 10.1016/j.neuroscience.2020.07.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/10/2020] [Accepted: 07/14/2020] [Indexed: 11/26/2022]
Abstract
Prolonged stress triggers neuroinflammation, which plays a significant role in the development of depression; however, stressed people do not always suffer from depression because of individual differences in stress vulnerability. Negative cognitive bias (NCB) toward pessimistic judgment often underlies depressive episodes. However, a relationship between stress vulnerability, neuroinflammation, and NCB remains elusive. In addition, an animal model with all the traits would be a powerful tool for studying the etiology of depression and its therapeutic approaches. Accordingly, this study evaluated the effect of stress vulnerability on neuroinflammation and depression-related behaviors, including NCB in males, using a modified version of repeated social defeat stress (mRSDS) paradigm, a validated animal model of psychosocial stress. Exposure to mRSDS, consisting of 5 min of social defeat by unfamiliar CD-1 aggressor mice for five consecutive days, caused NCB, which co-occurred with depressive- and anxiety-like behaviors, and neuroinflammation in male BALB/c mice. Treatment with minocycline, an antibiotic with anti-inflammatory property, blocked mRSDS-induced depressive-like behaviors and neuroinflammation, but not NCB, indicating the limited effect of an anti-inflammatory intervention. In addition, marked differences were found in neuroinflammatory profiles and hippocampal gene expression patterns between resilient and unstressed mice, as well as between susceptible and resilient mice. Therefore, mice resilient to mRSDS are indeed not intact. Our findings provide insights into the unique features of the mRSDS model in male BALB/c mice, which could be used to investigate the etiological mechanisms underlying depression as well as bridge the gap in the relationship between stress vulnerability, neuroinflammation, and NCB in males.
Collapse
Affiliation(s)
- Naoki Ito
- Department of Clinical Research, Oriental Medicine Research Center, Kitasato University, Minato-ku, Tokyo 108-8642, Japan.
| | - Kazunori Sasaki
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba-shi, Ibaraki 305-8572, Japan; Interdisciplinary Research Center for Catalytic Chemistry, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba-shi, Ibaraki 305-8565, Japan; Faculty of Pure and Applied Sciences, University of Tsukuba, Tsukuba-shi, Ibaraki 305-8571, Japan
| | - Hiroaki Takemoto
- School of Pharmacy, Kitasato University, Minato-ku, Tokyo 108-8642, Japan
| | - Yoshinori Kobayashi
- Department of Clinical Research, Oriental Medicine Research Center, Kitasato University, Minato-ku, Tokyo 108-8642, Japan; School of Pharmacy, Kitasato University, Minato-ku, Tokyo 108-8642, Japan
| | - Hiroko Isoda
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba-shi, Ibaraki 305-8572, Japan; Interdisciplinary Research Center for Catalytic Chemistry, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba-shi, Ibaraki 305-8565, Japan; Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba-shi, Ibaraki 305-8572, Japan
| | - Hiroshi Odaguchi
- Department of Clinical Research, Oriental Medicine Research Center, Kitasato University, Minato-ku, Tokyo 108-8642, Japan
| |
Collapse
|
11
|
Hein ZM, Kraiwattanapirom N, Mukda S, Chetsawang B. The induction of Neuron-Glial2 (NG2) expressing cells in methamphetamine toxicity-induced neuroinflammation in rat brain are averted by melatonin. J Neuroimmunol 2020; 344:577232. [PMID: 32311585 DOI: 10.1016/j.jneuroim.2020.577232] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 03/31/2020] [Accepted: 03/31/2020] [Indexed: 01/26/2023]
Abstract
Neuron-Glial2 (NG2) expressing cells are described as the oligodendrocyte precursor cells in the brain. This study aimed to investigate the possible involvement of NG2 cells under the methamphetamine (METH)-induced neurotoxicity and neuroprotective capacity of melatonin. The results showed that the levels of NG2 in rat brain gradually increase from postnatal day 0 to postnatal day 8 and then the lower levels of NG2 are shown in adults. In adult rats, the levels of NG2 and COX-2 in the brain were significantly increased in lipopolysaccharide treatment. Pretreatment of 10 mg/kg melatonin prior to treating with METH was able to reduce an increase in the levels of NG2 and activation in astrocyte and microglia. These findings would extend the contribution of NG2 expressing cells in the adult brain during pathological conditions such as neuroinflammation.
Collapse
Affiliation(s)
- Zaw Myo Hein
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | - Natcharee Kraiwattanapirom
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | - Sujira Mukda
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | - Banthit Chetsawang
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand.
| |
Collapse
|
12
|
Jin X, Riew TR, Kim S, Kim HL, Lee MY. Spatiotemporal Profile and Morphological Changes of NG2 Glia in the CA1 Region of the Rat Hippocampus after Transient Forebrain Ischemia. Exp Neurobiol 2020; 29:50-69. [PMID: 32122108 PMCID: PMC7075659 DOI: 10.5607/en.2020.29.1.50] [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: 12/03/2019] [Revised: 01/17/2020] [Accepted: 01/18/2020] [Indexed: 02/06/2023] Open
Abstract
Neuron-glial antigen-2 (NG2) glia undergo proliferation and morphological changes following brain insults. Here, we show that NG2 glia is activated in a characteristic time- and layer-specific manner in the ischemia-vulnerable CA1 region of the rat hippocampus. Resting NG2 glia of the pyramidal cell layer (somatic region) shared morphological features with those of the neighboring dendritic stratum radiatum. During the postischemic period, reactive NG2 glia of the pyramidal cell layer exhibited shortened, scarcely branched processes, while those of the stratum radiatum had multiple branching processes with their arborization being almost indiscernible 7~14 days after reperfusion. Immunoelectron microscopy demonstrated that NG2 immunoreactivity was specifically associated with the plasma membrane and the adjacent extracellular matrix of NG2 glia in the stratum radiatum at 14 days. NG2 glia also exhibited differences in their numbers and proliferation profiles in the two examined hippocampal strata after ischemia. In addition, induced NG2 expression in activated microglia/macrophages exhibited a characteristic strata-dependent pattern in the ischemic CA1 hippocampus. NG2 induction was prominent in macrophage-like phenotypes which were predominantly localized in the pyramidal cell layer, compared with activated stellate microglial cells in the stratum radiatum. Thus, our data demonstrate that activation of NG2 glia and the induction of NG2 expression in activated microglia/macrophages occur in a distinct time- and layer-specific manner in the ischemic CA1 hippocampus. These characteristic profiles of reactive NG2 glia could be secondary to the degeneration processes occurring in the cell bodies or dendritic domains of hippocampal CA1 pyramidal neurons after ischemic insults.
Collapse
Affiliation(s)
- Xuyan Jin
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea.,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea
| | - Tae-Ryong Riew
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea
| | - Soojin Kim
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea
| | - Hong Lim Kim
- Integrative Research Support Center, Laboratory of Electron Microscope, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Mun-Yong Lee
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea.,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea
| |
Collapse
|
13
|
Finocchiaro G, Pellegatta S. NG2/CSPG4 in glioblastoma: about flexibility. Neuro Oncol 2020; 21:697-698. [PMID: 30918960 DOI: 10.1093/neuonc/noz055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Affiliation(s)
- Gaetano Finocchiaro
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Serena Pellegatta
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
14
|
Chondroitin Sulphate Proteoglycans in the Tumour Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1272:73-92. [PMID: 32845503 DOI: 10.1007/978-3-030-48457-6_5] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Proteoglycans are macromolecules that are essential for the development of cells, human diseases and malignancies. In particular, chondroitin sulphate proteoglycans (CSPGs) accumulate in tumour stroma and play a key role in tumour growth and invasion by driving multiple oncogenic pathways in tumour cells and promoting crucial interactions in the tumour microenvironment (TME). These pathways involve receptor tyrosine kinase (RTK) signalling via the mitogen-activated protein kinase (MAPK) cascade and integrin signalling via the activation of focal adhesion kinase (FAK), which sustains the activation of extracellular signal-regulated kinases 1/2 (ERK1/2).Human CSPG4 is a type I transmembrane protein that is associated with the growth and progression of human brain tumours. It regulates cell signalling and migration by interacting with components of the extracellular matrix, extracellular ligands, growth factor receptors, intracellular enzymes and structural proteins. Its overexpression by tumour cells, perivascular cells and precursor/progenitor cells in gliomas suggests that it plays a role in their origin, progression and neo-angiogenesis and its aberrant expression in tumour cells may be a promising biomarker to monitor malignant progression and patient survival.The aim of this chapter is to review and discuss the role of CSPG4 in the TME of human gliomas, including its potential as a druggable therapeutic target.
Collapse
|
15
|
Glycans and glycosaminoglycans in neurobiology: key regulators of neuronal cell function and fate. Biochem J 2018; 475:2511-2545. [PMID: 30115748 DOI: 10.1042/bcj20180283] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 07/14/2018] [Accepted: 07/18/2018] [Indexed: 12/16/2022]
Abstract
The aim of the present study was to examine the roles of l-fucose and the glycosaminoglycans (GAGs) keratan sulfate (KS) and chondroitin sulfate/dermatan sulfate (CS/DS) with selected functional molecules in neural tissues. Cell surface glycans and GAGs have evolved over millions of years to become cellular mediators which regulate fundamental aspects of cellular survival. The glycocalyx, which surrounds all cells, actuates responses to growth factors, cytokines and morphogens at the cellular boundary, silencing or activating downstream signaling pathways and gene expression. In this review, we have focused on interactions mediated by l-fucose, KS and CS/DS in the central and peripheral nervous systems. Fucose makes critical contributions in the area of molecular recognition and information transfer in the blood group substances, cytotoxic immunoglobulins, cell fate-mediated Notch-1 interactions, regulation of selectin-mediated neutrophil extravasation in innate immunity and CD-34-mediated new blood vessel development, and the targeting of neuroprogenitor cells to damaged neural tissue. Fucosylated glycoproteins regulate delivery of synaptic neurotransmitters and neural function. Neural KS proteoglycans (PGs) were examined in terms of cellular regulation and their interactive properties with neuroregulatory molecules. The paradoxical properties of CS/DS isomers decorating matrix and transmembrane PGs and the positive and negative regulatory cues they provide to neurons are also discussed.
Collapse
|
16
|
Ilieva KM, Cheung A, Mele S, Chiaruttini G, Crescioli S, Griffin M, Nakamura M, Spicer JF, Tsoka S, Lacy KE, Tutt ANJ, Karagiannis SN. Chondroitin Sulfate Proteoglycan 4 and Its Potential As an Antibody Immunotherapy Target across Different Tumor Types. Front Immunol 2018; 8:1911. [PMID: 29375561 PMCID: PMC5767725 DOI: 10.3389/fimmu.2017.01911] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 12/14/2017] [Indexed: 12/18/2022] Open
Abstract
Overexpression of the chondroitin sulfate proteoglycan 4 (CSPG4) has been associated with the pathology of multiple types of such as melanoma, breast cancer, squamous cell carcinoma, mesothelioma, neuroblastoma, adult and pediatric sarcomas, and some hematological cancers. CSPG4 has been reported to exhibit a role in the growth and survival as well as in the spreading and metastasis of tumor cells. CSPG4 is overexpressed in several malignant diseases, while it is thought to have restricted and low expression in normal tissues. Thus, CSPG4 has become the target of numerous anticancer treatment approaches, including monoclonal antibody-based therapies. This study reviews key potential anti-CSPG4 antibody and immune-based therapies and examines their direct antiproliferative/metastatic and immune activating mechanisms of action.
Collapse
Affiliation(s)
- Kristina M Ilieva
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom.,Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, United Kingdom
| | - Anthony Cheung
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom.,Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, United Kingdom
| | - Silvia Mele
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom
| | - Giulia Chiaruttini
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom
| | - Silvia Crescioli
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom
| | - Merope Griffin
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom
| | - Mano Nakamura
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom.,Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, London, United Kingdom
| | - James F Spicer
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, United Kingdom
| | - Sophia Tsoka
- Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, London, United Kingdom
| | - Katie E Lacy
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom
| | - Andrew N J Tutt
- Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, United Kingdom.,Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, United Kingdom
| | - Sophia N Karagiannis
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom.,Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, United Kingdom
| |
Collapse
|
17
|
Distinct NG2 proteoglycan-dependent roles of resident microglia and bone marrow-derived macrophages during myelin damage and repair. PLoS One 2017; 12:e0187530. [PMID: 29095924 PMCID: PMC5667885 DOI: 10.1371/journal.pone.0187530] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 10/20/2017] [Indexed: 12/21/2022] Open
Abstract
We used a bone marrow transplantation approach to distinguish the activities of bone marrow-derived macrophages from the activities of central nervous system-resident microglia in phenomena associated with axon demyelination and remyelination. We transplanted wild type or germline NG2 null beta-actin-EGFP expressing bone marrow into irradiated wild type or NG2 null recipient mice, followed by analysis of lysolecithin-induced spinal cord demyelination and remyelination and quantification of Iba-1+/ F4/80+/ EGFP+ macrophages and Iba-1+/ F4/80+/ EGFP- microglia. One week after microinjection of 1% lysolecithin into the spinal cord, wild type recipients receiving NG2 null bone marrow exhibit greatly reduced infiltration of macrophages into lesions, compared to wild type recipients receiving wild type bone marrow. Wild type bone marrow recipients also exhibit larger numbers of demyelinated axons than NG2 null recipients, indicative of macrophage participation in the initial myelin damage. However, wild type bone marrow recipients also exhibit superior myelin repair at 6 weeks post-injury, compared to NG2 null bone marrow recipients, demonstrating the additional importance of macrophages in remyelination. Incompletely repaired lesions in NG2 null bone marrow recipients at 6 weeks post-injury retain elevated numbers of macrophages, in contrast to lower numbers of macrophages in more completely repaired lesions in wild type bone marrow recipients. This suggests that NG2 expression renders macrophages more effective in myelin repair and less likely to promote chronic inflammation. Effective macrophage involvement in myelin repair is due in part to effects on the proliferation and/or recruitment of oligodendrocyte progenitor cells. Reduced numbers of oligodendrocyte progenitors are seen in lesions in NG2 null bone marrow recipients, likely due to deficits in macrophage production of oligodendrocyte progenitor-relevant mitogens and in phagocytosis of inhibitory myelin debris. Microglia also appear to be important for clearance of myelin debris, as indicated by reduced phagocytosis in NG2 null recipients receiving wild type bone marrow.
Collapse
|
18
|
Nastase MV, Janicova A, Wygrecka M, Schaefer L. Signaling at the Crossroads: Matrix-Derived Proteoglycan and Reactive Oxygen Species Signaling. Antioxid Redox Signal 2017; 27:855-873. [PMID: 28510506 DOI: 10.1089/ars.2017.7165] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Proteoglycans (PGs), besides their structural contribution, have emerged as dynamic components that mediate a multitude of cellular events. The various roles of PGs are attributed to their structure, spatial localization, and ability to act as ligands and receptors. Reactive oxygen species (ROS) are small mediators that are generated in physiological and pathological conditions. Besides their reactivity and ability to induce oxidative stress, a growing body of data suggests that ROS signaling is more relevant than direct radical damage in development of human pathologies. Recent Advances: Cell surface transmembrane PGs (syndecans, cluster of differentiation 44) represent receptors in diverse and complex transduction networks, which involve redox signaling with implications in cancer, fibrosis, renal dysfunction, or Alzheimer's disease. Through NADPH oxidase (NOX)-dependent ROS, the extracellular PG, hyaluronan is involved in osteoclastogenesis and cancer. The ROS sources, NOX1 and NOX4, increase biglycan-induced inflammation, while NOX2 is a negative regulator. CRITICAL ISSUES The complexity of the mechanisms that bring ROS into the light of PG biology might be the foundation of a new research area with significant promise for understanding health and disease. Important aspects need to be investigated in PG/ROS signaling: the discovery of specific targets of ROS, the precise ROS-induced chemical modifications of these targets, and the study of their pathological relevance. FUTURE DIRECTIONS As we become more and more aware of the interactions between PG and ROS signaling underlying intracellular communication and cell fate decisions, it is quite conceivable that this field will allow to identify new therapeutic targets.-Antioxid. Redox Signal. 27, 855-873.
Collapse
Affiliation(s)
- Madalina-Viviana Nastase
- 1 Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe Universität , Frankfurt am Main, Germany .,2 National Institute for Chemical-Pharmaceutical Research and Development , Bucharest, Romania
| | - Andrea Janicova
- 1 Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe Universität , Frankfurt am Main, Germany
| | - Malgorzata Wygrecka
- 3 Department of Biochemistry, Faculty of Medicine, Justus Liebig University , Giessen, Germany
| | - Liliana Schaefer
- 1 Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe Universität , Frankfurt am Main, Germany
| |
Collapse
|
19
|
Schmitt BM, Laschke MW, Rössler OG, Huang W, Scheller A, Menger MD, Ampofo E. Nerve/glial antigen (NG) 2 is a crucial regulator of intercellular adhesion molecule (ICAM)-1 expression. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1865:57-66. [PMID: 28964848 DOI: 10.1016/j.bbamcr.2017.09.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 09/01/2017] [Accepted: 09/26/2017] [Indexed: 12/13/2022]
Abstract
The proteoglycan nerve/glial antigen (NG) 2 is expressed on multiple cell types and mediates cell proliferation and migration. However, little is known about its function in gene regulation. In this study, we demonstrate that in pericytes and glioblastoma cells intercellular adhesion molecule (ICAM)-1, an essential protein for leukocyte adhesion and transmigration, underlies a NG2-dependent expression. As shown by flow cytometry, Western blot analysis and quantitative real-time polymerase chain reaction (qRT-PCR), silencing of NG2 in human placenta-derived pericytes increased the expression of ICAM-1. Pathway analyses revealed that this is mediated by extracellular-regulated-kinases (ERK) 1/2 signaling. Moreover, leukocyte adhesion to NG2 siRNA-treated pericytes was significantly enhanced when compared to scrambled (scr) siRNA-treated control cells. In vivo, we detected increased ICAM-1 protein levels in the retina of mice lacking NG2 expression. To exclude that this novel mechanism is pericyte-specific, we additionally analyzed the expression of ICAM-1 in dependency of NG2 in two glioblastoma cell lines. We found that A1207 and M059K cells exhibit an inverse expression pattern of NG2 and ICAM-1. Finally, downregulation of NG2 in A1207 cells significantly increased ICAM-1 expression. Taken together, these findings indicate that NG2 may represent a promising target for the modulation of ICAM-1-mediated immune responses.
Collapse
Affiliation(s)
- Beate M Schmitt
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Oliver G Rössler
- Medical Biochemistry and Molecular Biology, Saarland University, 66421 Homburg/Saar, Germany
| | - Wenhui Huang
- Department of Molecular Physiology, CIPMM (Center for Integrative Physiology and Molecular Medicine), Saarland University, 66421 Homburg/Saar, Germany
| | - Anja Scheller
- Department of Molecular Physiology, CIPMM (Center for Integrative Physiology and Molecular Medicine), Saarland University, 66421 Homburg/Saar, Germany
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Emmanuel Ampofo
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany.
| |
Collapse
|
20
|
Song FE, Huang JL, Lin SH, Wang S, Ma GF, Tong XP. Roles of NG2-glia in ischemic stroke. CNS Neurosci Ther 2017; 23:547-553. [PMID: 28317272 DOI: 10.1111/cns.12690] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 02/23/2017] [Accepted: 02/24/2017] [Indexed: 12/20/2022] Open
Abstract
Recent studies have shown that a widely distributed class of glial cells, termed NG2-glia, engages in rapid signaling with surrounding neurons through direct synaptic contacts in the developing and mature central nervous system (CNS). This unique glial cell group has a typical function of proliferating and differentiating into oligodendrocytes during early development of the brain, which is crucial to axon myelin formation. Therefore, NG2-glia are also called oligodendrocyte precursor cells (OPCs). In vitro and in vivo studies reveal that NG2-glia expressing receptors and ion channels demonstrate functional significance for rapid signaling with neuronal synapses and modulation of neuronal activities in both physiological and pathological conditions. Although it is well known that NG2-glia play an important role in demyelinating diseases such as multiple sclerosis, little is known about how NG2-glia or OPCs impact neurons and brain function following ischemic injury. This review summarizes recent progress on the roles of NG2-glia in ischemic stroke and illustrates new approaches for targeting NG2-glia in the brain to treat this disease.
Collapse
Affiliation(s)
- Fei-Er Song
- Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia-Lv Huang
- Department of Clinical Medicine, Research-Based Learning training program (RBL2015-29), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Si-Han Lin
- Department of Clinical Medicine, Research-Based Learning training program (RBL2015-29), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuo Wang
- Department of Clinical Medicine, Research-Based Learning training program (RBL2015-29), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guo-Fen Ma
- Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Ping Tong
- Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
21
|
Ampofo E, Schmitt BM, Menger MD, Laschke MW. The regulatory mechanisms of NG2/CSPG4 expression. Cell Mol Biol Lett 2017; 22:4. [PMID: 28536635 PMCID: PMC5415841 DOI: 10.1186/s11658-017-0035-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 02/22/2017] [Indexed: 12/24/2022] Open
Abstract
Neuron-glial antigen 2 (NG2), also known as chondroitin sulphate proteoglycan 4 (CSPG4), is a surface type I transmembrane core proteoglycan that is crucially involved in cell survival, migration and angiogenesis. NG2 is frequently used as a marker for the identification and characterization of certain cell types, but little is known about the mechanisms regulating its expression. In this review, we provide evidence that the regulation of NG2 expression underlies inflammation and hypoxia and is mediated by methyltransferases, transcription factors, including Sp1, paired box (Pax) 3 and Egr-1, and the microRNA miR129-2. These regulatory factors crucially determine NG2-mediated cellular processes such as glial scar formation in the central nervous system (CNS) or tumor growth and metastasis. Therefore, they are potential targets for the establishment of novel NG2-based therapeutic strategies in the treatment of CNS injuries, cancer and other conditions of these types.
Collapse
Affiliation(s)
- Emmanuel Ampofo
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Germany
| | - Beate M Schmitt
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Germany
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Germany
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Germany
| |
Collapse
|
22
|
Li P, Li HX, Jiang HY, Zhu L, Wu HY, Li JT, Lai JH. Expression of NG2 and platelet-derived growth factor receptor alpha in the developing neonatal rat brain. Neural Regen Res 2017; 12:1843-1852. [PMID: 29239330 PMCID: PMC5745838 DOI: 10.4103/1673-5374.219045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Platelet-derived growth factor receptor alpha (PDGFRα) is a marker of oligodendrocyte precursor cells in the central nervous system. NG2 is also considered a marker of oligodendrocyte precursor cells. However, whether there are differences in the distribution and morphology of oligodendrocyte precursor cells labeled by NG2 or PDGFRα in the developing neonatal rat brain remains unclear. In this study, by immunohistochemical staining, NG2 positive (NG2+) cells were ubiquitous in the molecular layer, external pyramidal layer, internal pyramidal layer, and polymorphic layer of the cerebral cortex, and corpus callosum, external capsule, piriform cortex, and medial septal nucleus. NG2+ cells were stellate or fusiform in shape with long processes that were progressively decreased and shortened over the course of brain development. The distribution and morphology of PDGFRα positive (PDGFRα+) cells were coincident with NG2+ cells. The colocalization of NG2 and PDGFRα in the cell bodies and processes of some cells was confirmed by double immunofluorescence labeling. Moreover, cells double-labeled for NG2 and PDGFRα were predominantly in the early postnatal stage of development. The numbers of NG2+/PDGFRα+ cells and PDGFRα+ cells decreased, but the number of NG2+ cells increased from postnatal days 3 to 14 in the developing brain. In addition, amoeboid microglial cells of the corpus callosum, newborn brain macrophages in the normal developing brain, did not express NG2 or PDGFRα, but NG2 expression was detected in amoeboid microglia after hypoxia. The present results suggest that NG2 and PDGFRα are specific markers of oligodendrocyte precursor cells at different stages during early development. Additionally, the NG2 protein is involved in inflammatory and pathological processes of amoeboid microglial cells.
Collapse
Affiliation(s)
- Ping Li
- College of Forensic Science, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi Province; Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan Province, China
| | - Heng-Xi Li
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan Province, China
| | - Hong-Yan Jiang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan Province, China
| | - Lie Zhu
- Department of Plastic Surgery, Changzheng Hospital, Shanghai, China
| | - Hai-Ying Wu
- Department of Emergency and Intensive Care Unit, First Affiliated Hospital, Kunming Medical University, Kunming, Yunnan Province, China
| | - Jin-Tao Li
- Neuroscience Institute, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan Province, China
| | - Jiang-Hua Lai
- College of Forensic Science, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| |
Collapse
|
23
|
Ferrara G, Errede M, Girolamo F, Morando S, Ivaldi F, Panini N, Bendotti C, Perris R, Furlan R, Virgintino D, Kerlero de Rosbo N, Uccelli A. NG2, a common denominator for neuroinflammation, blood-brain barrier alteration, and oligodendrocyte precursor response in EAE, plays a role in dendritic cell activation. Acta Neuropathol 2016; 132:23-42. [PMID: 27026411 PMCID: PMC4911384 DOI: 10.1007/s00401-016-1563-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 03/15/2016] [Accepted: 03/15/2016] [Indexed: 02/07/2023]
Abstract
In adult CNS, nerve/glial-antigen 2 (NG2) is expressed by oligodendrocyte progenitor cells (OPCs) and is an early marker of pericyte activation in pathological conditions. NG2 could, therefore, play a role in experimental autoimmune encephalomyelitis (EAE), a disease associated with increased blood–brain barrier (BBB) permeability, inflammatory infiltrates, and CNS damage. We induced EAE in NG2 knock-out (NG2KO) mice and used laser confocal microscopy immunofluorescence and morphometry to dissect the effect of NG2 KO on CNS pathology. NG2KO mice developed milder EAE than their wild-type (WT) counterparts, with less intense neuropathology associated with a significant improvement in BBB stability. In contrast to WT mice, OPC numbers did not change in NG2KO mice during EAE. Through FACS and confocal microscopy, we found that NG2 was also expressed by immune cells, including T cells, macrophages, and dendritic cells (DCs). Assessment of recall T cell responses to the encephalitogen by proliferation assays and ELISA showed that, while WT and NG2KO T cells proliferated equally to the encephalitogenic peptide MOG35-55, NG2KO T cells were skewed towards a Th2-type response. Because DCs could be responsible for this effect, we assessed their expression of IL-12 by PCR and intracellular FACS. IL-12-expressing CD11c+ cells were significantly decreased in MOG35-55-primed NG2KO lymph node cells. Importantly, in WT mice, the proportion of IL-12-expressing cells was significantly lower in CD11c+ NG2- cells than in CD11c+ NG2+ cells. To assess the relevance of NG2 at immune system and CNS levels, we induced EAE in bone-marrow chimeric mice, generated with WT recipients of NG2KO bone-marrow cells and vice versa. Regardless of their original phenotype, mice receiving NG2KO bone marrow developed milder EAE than those receiving WT bone marrow. Our data suggest that NG2 plays a role in EAE not only at CNS/BBB level, but also at immune response level, impacting on DC activation and thereby their stimulation of reactive T cells, through controlling IL-12 expression.
Collapse
|
24
|
NG2 expression in microglial cells affects the expression of neurotrophic and proinflammatory factors by regulating FAK phosphorylation. Sci Rep 2016; 6:27983. [PMID: 27306838 PMCID: PMC4910048 DOI: 10.1038/srep27983] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/27/2016] [Indexed: 12/01/2022] Open
Abstract
Neural/glial antigen 2 (NG2), a chondroitin sulfate proteoglycan, is significantly upregulated in a subset of glial cells in the facial motor nucleus (FMN) following CNS injury. NG2 is reported to promote the resulting inflammatory reaction, however, the mechanism by which NG2 mediates these effects is yet to be determined. In this study, we examined the changes in NG2 expressing microglial cells in the FMN in response to facial nerve axotomy (FNA) in mice. Our findings indicated that NG2 expression was progressively induced and upregulated specifically in the ipsilateral facial nucleus following FNA. To further investigate the effects of NG2 expression, in vivo studies in NG2-knockout mice and in vitro studies in rat microglial cells transfected with NG2 shRNAs were performed. Abolition of NG2 expression both in vitro and in vivo resulted in increased expression of neurotrophic factors (nerve growth factor and glial derived neurotrophic factor), decreased expression of inflammatory mediators (tumor necrosis factor-α and interleukin-1β) and decreased apoptosis in the ipsilateral facial nucleus in response to FNA. Furthermore, we demonstrated the role of FAK in these NG2-induced effects. Taken together, our findings suggest that NG2 expression mediates inflammatory reactions and neurodegeneration in microglial cells in response to CNS injury, potentially by regulating FAK phosphorylation.
Collapse
|
25
|
Haas SJP, Zhou X, Machado V, Wree A, Krieglstein K, Spittau B. Expression of Tgfβ1 and Inflammatory Markers in the 6-hydroxydopamine Mouse Model of Parkinson's Disease. Front Mol Neurosci 2016; 9:7. [PMID: 26869879 PMCID: PMC4737885 DOI: 10.3389/fnmol.2016.00007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 01/14/2016] [Indexed: 11/13/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder that is characterized by loss of midbrain dopaminergic (mDA) neurons in the substantia nigra (SN). Microglia-mediated neuroinflammation has been described as a common hallmark of PD and is believed to further trigger the progression of neurodegenerative events. Injections of 6-hydroxydopamine (6-OHDA) are widely used to induce degeneration of mDA neurons in rodents as an attempt to mimic PD and to study neurodegeneration, neuroinflammation as well as potential therapeutic approaches. In the present study, we addressed microglia and astroglia reactivity in the SN and the caudatoputamen (CPu) after 6-OHDA injections into the medial forebrain bundle (MFB), and further analyzed the temporal and spatial expression patterns of pro-inflammatory and anti-inflammatory markers in this mouse model of PD. We provide evidence that activated microglia as well as neurons in the lesioned SN and CPu express Transforming growth factor β1 (Tgfβ1), which overlaps with the downregulation of pro-inflammatory markers Tnfα, and iNos, and upregulation of anti-inflammatory markers Ym1 and Arg1. Taken together, the data presented in this study suggest an important role for Tgfβ1 as a lesion-associated factor that might be involved in regulating microglia activation states in the 6-OHDA mouse model of PD in order to prevent degeneration of uninjured neurons by microglia-mediated release of neurotoxic factors such as Tnfα and nitric oxide (NO).
Collapse
Affiliation(s)
| | - Xiaolai Zhou
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Albert-Ludwigs-UniversityFreiburg, Germany; Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell UniversityIthaca, NY, USA
| | - Venissa Machado
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Albert-Ludwigs-UniversityFreiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-UniversityFreiburg, Germany; Faculty of Biology, Albert-Ludwigs-UniversityFreiburg, Germany
| | - Andreas Wree
- Institute of Anatomy, Rostock University Medical Center Rostock, Germany
| | - Kerstin Krieglstein
- Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-University Freiburg, Germany
| | - Björn Spittau
- Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-University Freiburg, Germany
| |
Collapse
|
26
|
Xiang P, Zhu L, Jiang H, He BP. The activation of NG2 expressing cells is downstream to microglial reaction and mediated by the transforming growth factor beta 1. J Neuroimmunol 2015; 279:50-63. [PMID: 25670001 DOI: 10.1016/j.jneuroim.2015.01.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 12/25/2014] [Accepted: 01/14/2015] [Indexed: 11/30/2022]
Abstract
In the present study, we investigated the mechanism of activation of NG2 expressing cells. Application of microglial inhibitors not only attenuated morphological changes but also significantly retarded increase in the number of NG2 expressing cells. Intracerebral injection of TGF-β1 led to a profound activation of NG2 glia as well as an earlier accumulation of NG2(+)-microglia, whilst inhibition of TGF-β1 Smad2/3 signalling pathway eventually attenuated their active responses. We conclude that the activation of NG2 expressing cells is an event downstream to microglial reaction and TGF-β1 secreted from microglia might play an important role in modulation of the function of NG2 expressing cells.
Collapse
Affiliation(s)
- Ping Xiang
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Lie Zhu
- Department of Plastic Surgery, Chang Zheng Hospital, Shanghai, China
| | - Hua Jiang
- Department of Plastic Surgery, Chang Zheng Hospital, Shanghai, China
| | - Bei Ping He
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
27
|
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
|
28
|
Wennström M, Janelidze S, Bay-Richter C, Minthon L, Brundin L. Pro-inflammatory cytokines reduce the proliferation of NG2 cells and increase shedding of NG2 in vivo and in vitro. PLoS One 2014; 9:e109387. [PMID: 25285951 PMCID: PMC4186831 DOI: 10.1371/journal.pone.0109387] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 09/09/2014] [Indexed: 11/18/2022] Open
Abstract
Neuron glial 2 (NG2) cells become strongly activated in injured brain areas. The activation is characterized by increased proliferation as well as increased expression and shedding of the proteoglycan NG2 expressed on their cell surface. It is currently not known how these cells respond to low-grade neuroinflammation provoked by systemic inflammation. To investigate this, we analyzed NG2 cell proliferation as well as soluble NG2 (sNG2) in cerebrospinal fluid (CSF) from rats treated with an acute intraperitoneal (i.p) injection of lipopolysaccharides (LPS) or saline and sacrificed after 2 or 24 hours. The systemically induced neuroinflammation was confirmed as elevated levels of cytokines, including interleukin (IL)-6 and IL-1β, and MHCII expressing microglia were found 24 h after LPS treatment. At this time point NG2 cell proliferation was significantly decreased in both amygdala and hippocampus and sNG2 levels in CSF were increased twofold. We also exposed human NG2 cells in culture to IL-6 and IL-1β for 24 h and found, in line with our in vivo study, a direct impact of these cytokines reducing cell proliferation and increasing shedding of NG2. We conclude that LPS induced systemic inflammation significantly affects NG2 cell proliferation and shedding and that these two events at least in in part are mediated by IL-6 and IL-1β.
Collapse
Affiliation(s)
- Malin Wennström
- Lund University, Department of Clinical Sciences, Clinical Memory Research Unit, Wallenberg Laboratory, Malmö, Sweden
- * E-mail:
| | - Shorena Janelidze
- Lund University, Department of Clinical Sciences, Clinical Memory Research Unit, Wallenberg Laboratory, Malmö, Sweden
| | - Cecilie Bay-Richter
- Aarhus University, Department of Clinical Medicine, Translational Neuropsychiatry Unit, Risskov, Denmark
| | - Lennart Minthon
- Lund University, Department of Clinical Sciences, Clinical Memory Research Unit, Wallenberg Laboratory, Malmö, Sweden
| | - Lena Brundin
- Michigan State University, College of Human Medicine, Department of Psychiatry and Behavioral Medicine, Grand Rapids, Michigan, United States of America
- Van Andel Research Institute, Laboratory of Behavioral Medicine, Grand Rapids, Michigan, United States of America
| |
Collapse
|
29
|
Chen SH, Oyarzabal EA, Sung YF, Chu CH, Wang Q, Chen SL, Lu RB, Hong JS. Microglial regulation of immunological and neuroprotective functions of astroglia. Glia 2014; 63:118-31. [PMID: 25130274 DOI: 10.1002/glia.22738] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 07/23/2014] [Indexed: 01/06/2023]
Abstract
Microglia and astroglia play critical roles in the development, function, and survival of neurons in the CNS. However, under inflammatory conditions the role of astrogliosis in the inflammatory process and its effects on neurons remains unclear. Here, we used several types of cell cultures treated with the bacterial inflammogen LPS to address these questions. We found that the presence of astroglia reduced inflammation-driven neurotoxicity, suggesting that astrogliosis is principally neuroprotective. Neutralization of supernatant glial cell line-derived neurotrophic factor (GDNF) released from astroglia significantly reduced this neuroprotective effect during inflammation. To determine the immunological role of astroglia, we optimized a highly-enriched astroglial culture protocol and demonstrated that LPS failed to induce the synthesis and release of TNF-α and iNOS/NO. Instead we found significant enhancement of TNF-α and iNOS expression in highly-enriched astroglial cultures required the presence of 0.5-1% microglia, respectively. Thus suggesting that microglial-astroglial interactions are required for LPS to induce the expression of pro-inflammatory factors and GDNF from astroglia. Specifically, we found that microglia-derived TNF-α plays a pivotal role as a paracrine signal to regulate the neuroprotective functions of astrogliosis. Taken together, these findings suggest that astroglia may not possess the ability to directly recognize the innate immune stimuli LPS, but rather depend on crosstalk with microglia to elicit release of neurotrophic factors as a counterbalance to support neuronal survival from the collateral damage generated by activated microglia during neuroinflammation.
Collapse
Affiliation(s)
- Shih-Heng Chen
- Laboratory of Toxicology and Pharmacology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Zhang H, Zhang Y, Xu H, Wang L, Adilijiang A, Wang J, Hartle K, Zhang Z, Zhang D, Tan Q, Kong J, Huang Q, Li XM. Olanzapine ameliorates neuropathological changes and increases IGF-1 expression in frontal cortex of C57BL/6 mice exposed to cuprizone. Psychiatry Res 2014; 216:438-45. [PMID: 24613202 DOI: 10.1016/j.psychres.2014.02.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 11/06/2013] [Accepted: 02/10/2014] [Indexed: 02/05/2023]
Abstract
Cuprizone (CPZ) induced demyelinating mouse has been used as an animal model to examine the assumed roles of altered oligodendrocytes in the pathophysiology and treatment of schizophrenia. The objectives of this study were to examine the effect of olanzapine, an atypical antipsychotic, on cuprizone-induced neuropathological changes in the frontal cortex of C57BL/6 mice, and to explore the underlying mechanism for the possible protective effects. The effects of six-week olanzapine (10 mg/kg/day) treatments on neuropathological changes were examined by immunohistochemistry and Western-blot analyses. Olanzapine treatment for six weeks effectively decreased the breakdown of myelin and oligodendrocytes loss of cuprizone-fed mice. Reactive cellular changes, including astrocyte gliosis, microglia accumulation and increased activation of oligodendrocyte progenitor cells, were also attenuated by olanzapine. However, the cortical expression level of insulin-like growth factor 1 (IGF-1) was significantly increased by olanzapine treatment in cuprizone-fed mice as measured by the quantitative real-time polymerase chain reaction (PCR) method. Olanzapine treatment in control mice consuming normal food had no effect on all above measures. These results provide the first in vivo evidence for the protective effects of olanzapine on cuprizone-induced neuropathological changes and suggest that up-regulated insulin-like growth factor 1 may contribute to the protective effects of this antipsychotic.
Collapse
Affiliation(s)
- Handi Zhang
- Mental Health Center, Shantou University Medical College, Shantou 515065, Guangdong, China
| | - Yanbo Zhang
- Department of Psychiatry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Haiyun Xu
- Mental Health Center, Shantou University Medical College, Shantou 515065, Guangdong, China
| | - Lingyan Wang
- Department of Cardiology, China-Japan Friendship Hospital of Jilin University, Jilin University, Chuangchun, Jilin, China
| | | | - Junhui Wang
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| | - Kelly Hartle
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| | - Zhijun Zhang
- Department of Neuropsychiatry, Affiliated ZhongDa Hospital Southeast University, Nanjing, Jiangsu, China
| | - Dai Zhang
- Institute of Mental Health, Peking University, Beijing, China
| | - Qingrong Tan
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi׳an, Shannxi, China
| | - Jiming Kong
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Qingjun Huang
- Mental Health Center, Shantou University Medical College, Shantou 515065, Guangdong, China.
| | - Xin-Min Li
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
31
|
Sugimoto K, Nishioka R, Ikeda A, Mise A, Takahashi H, Yano H, Kumon Y, Ohnishi T, Tanaka J. Activated microglia in a rat stroke model express NG2 proteoglycan in peri-infarct tissue through the involvement of TGF-β1. Glia 2013; 62:185-98. [PMID: 24311432 DOI: 10.1002/glia.22598] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 10/20/2013] [Accepted: 10/22/2013] [Indexed: 12/12/2022]
Abstract
We investigated activated microglia in ischemic brain lesions from rats that had been subjected to transient middle cerebral artery occlusion. Activated microglia expressing NG2 chondroitin sulfate proteoglycan (NG2) were found only in the narrow zone (demarcation zone) that demarcated the peri-infarct tissue and ischemic core. NG2(-) activated microglia were abundantly distributed in the peri-infarct tissue outside the demarcation zone. NG2(+) microglia but not NG2(-) microglia expressed both CD68 and a triggering receptor expressed on myeloid cells 2 (TREM-2), suggesting that NG2(+) microglia eliminated apoptotic neurons. In fact, NG2(+) microglia often attached to degenerating neurons and sometimes internalized NeuN(+) or neurofilament protein(+) material. Kinetic studies using quantitative real-time RT-PCR revealed that expression of transforming growth factor-β1 (TGF-β1) was most evident in the ischemic core; with this marker produced mainly by macrophages located in this region. TGF-β receptor mRNA expression peaked at 3 days post reperfusion (dpr) in the peri-infarct tissue, including the demarcation zone. Primary cultured rat microglia also expressed the receptor mRNA. In response to TGF-β1, primary microglia enhanced the expression of NG2 protein and TREM-2 mRNA as well as migratory activity. A TGF-β1 inhibitor, SB525334, abolished these effects. The present results suggest that TGF-β1 produced in the ischemic core diffused toward the peri-infarct tissue, driving activated microglial cells to eliminate degenerating neurons. Appropriate control of NG2(+) microglia in the demarcation zone might be a novel target for the suppression of secondary neurodegeneration in the peri-infarct tissue.
Collapse
Affiliation(s)
- Kana Sugimoto
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Toon, Ehime, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Nielsen HM, Ek D, Avdic U, Orbjörn C, Hansson O, Veerhuis R, Rozemuller AJM, Brun A, Minthon L, Wennström M. NG2 cells, a new trail for Alzheimer's disease mechanisms? Acta Neuropathol Commun 2013; 1:7. [PMID: 24252600 PMCID: PMC4046664 DOI: 10.1186/2051-5960-1-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 02/28/2013] [Indexed: 02/07/2023] Open
Abstract
Background Neuron Glial 2 (NG2) cells are glial cells known to serve as oligodendrocyte progenitors as well as modulators of the neuronal network. Altered NG2 cell morphology and up-regulation as well as increased shedding of the proteoglycan NG2 expressed on the cell surface have been described in rodent models of brain injury. Here we describe alterations in the human NG2 cell population in response to pathological changes characteristic of Alzheimer’s disease (AD). Results Immunohistological stainings of postmortem brain specimens from clinically diagnosed and postmortem verified AD patients and non-demented controls revealed reduced NG2 immunoreactivity as well as large numbers of NG2 positive astrocytes in individuals with high amyloid beta plaque load. Since fibrillar amyloid beta (Aβ)1-42 is the major component of AD-related senile plaques, we exposed human NG2 cells to oligomer- and fibril enriched preparations of Aβ1-42. We found that both oligomeric and fibrillar Aβ1-42 induced changes in NG2 cell morphology. Further, in vitro exposure to fibrillar Aβ1-42 decreased the NG2 concentrations in both cell lysates and supernatants. Interestingly, we also found significantly decreased levels of soluble NG2 in the cerebrospinal fluid (CSF) from clinically diagnosed AD patients compared to non-demented individuals. Additionally, the CSF NG2 levels were found to significantly correlate with the core AD biomarkers Aß1-42, T-tau and P-tau. Conclusion Our results demonstrate major alterations in the NG2 cell population in relation to AD pathology which highlights the NG2 cell population as a new attractive research target in the search for cellular mechanisms associated with AD pathogenesis. Electronic supplementary material The online version of this article (doi:10.1186/2051-5960-1-7) contains supplementary material, which is available to authorized users.
Collapse
|
33
|
Chew LJ, Fusar-Poli P, Schmitz T. Oligodendroglial alterations and the role of microglia in white matter injury: relevance to schizophrenia. Dev Neurosci 2013; 35:102-29. [PMID: 23446060 PMCID: PMC4531048 DOI: 10.1159/000346157] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 11/07/2012] [Indexed: 12/12/2022] Open
Abstract
Schizophrenia is a chronic and debilitating mental illness characterized by a broad range of abnormal behaviors, including delusions and hallucinations, impaired cognitive function, as well as mood disturbances and social withdrawal. Due to the heterogeneous nature of the disease, the causes of schizophrenia are very complex; its etiology is believed to involve multiple brain regions and the connections between them, and includes alterations in both gray and white matter regions. The onset of symptoms varies with age and severity, and there is some debate over a degenerative or developmental etiology. Longitudinal magnetic resonance imaging studies have detected progressive gray matter loss in the first years of disease, suggesting neurodegeneration; but there is also increasing recognition of a temporal association between clinical complications at birth and disease onset that supports a neurodevelopmental origin. Presently, neuronal abnormalities in schizophrenia are better understood than alterations in myelin-producing cells of the brain, the oligodendrocytes, which are the predominant constituents of white matter structures. Proper white matter development and its structural integrity critically impacts brain connectivity, which affects sensorimotor coordination and cognitive ability. Evidence of defective white matter growth and compromised white matter integrity has been found in individuals at high risk of psychosis, and decreased numbers of mature oligodendrocytes are detected in schizophrenia patients. Inflammatory markers, including proinflammatory cytokines and chemokines, are also associated with psychosis. A relationship between risk of psychosis, white matter defects and prenatal inflammation is being established. Animal models of perinatal brain injury are successful in producing white matter damage in the brain, typified by hypomyelination and/or dysmyelination, impaired motor coordination and prepulse inhibition of the acoustic startle reflex, recapitulating structural and functional characteristics observed in schizophrenia. In addition, elevated expression of inflammation-related genes in brain tissue and increased production of cytokines by blood cells from patients with schizophrenia indicate immunological dysfunction and abnormal inflammatory responses, which are also important underlying features in experimental models. Microglia, resident immune defenders of the central nervous system, play important roles in the development and protection of neural cells, but can contribute to injury under pathological conditions. This article discusses oligodendroglial changes in schizophrenia and focuses on microglial activity in the context of the disease, in neonatal brain injury and in various experimental models of white matter damage. These include disorders associated with premature birth, and animal models of perinatal bacterial and viral infection, oxygen deprivation (hypoxia) and excess (hyperoxia), and elevated systemic proinflammatory cytokine levels. We briefly review the effects of treatment with antipsychotic and anti-inflammatory agents in models of perinatal brain injury, and comment on the therapeutic potential of these strategies. By understanding the neurobiological basis of oligodendroglial abnormalities in schizophrenia, it is hoped that patients will benefit from the availability of targeted and more efficacious treatment options.
Collapse
Affiliation(s)
- Li-Jin Chew
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA.
| | | | | |
Collapse
|
34
|
Xu L, Yao Z, Wu H, Wang F, Zhang S. The immune regulation of κ-carrageenan oligosaccharide and its desulfated derivatives on LPS-activated microglial cells. Neurochem Int 2012; 61:689-96. [DOI: 10.1016/j.neuint.2012.06.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 05/25/2012] [Accepted: 06/23/2012] [Indexed: 10/28/2022]
|
35
|
Honsa P, Pivonkova H, Dzamba D, Filipova M, Anderova M. Polydendrocytes display large lineage plasticity following focal cerebral ischemia. PLoS One 2012; 7:e36816. [PMID: 22590616 PMCID: PMC3349640 DOI: 10.1371/journal.pone.0036816] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 04/07/2012] [Indexed: 11/18/2022] Open
Abstract
Polydendrocytes (also known as NG2 glial cells) constitute a fourth major glial cell type in the adult mammalian central nervous system (CNS) that is distinct from other cell types. Although much evidence suggests that these cells are multipotent in vitro, their differentiation potential in vivo under physiological or pathophysiological conditions is still controversial. To follow the fate of polydendrocytes after CNS pathology, permanent middle cerebral artery occlusion (MCAo), a commonly used model of focal cerebral ischemia, was carried out on adult NG2creBAC:ZEG double transgenic mice, in which enhanced green fluorescent protein (EGFP) is expressed in polydendrocytes and their progeny. The phenotype of the EGFP+ cells was analyzed using immunohistochemistry and the patch-clamp technique 3, 7 and 14 days after MCAo. In sham-operated mice (control), EGFP+ cells in the cortex expressed protein markers and displayed electrophysiological properties of polydendrocytes and oligodendrocytes. We did not detect any co-labeling of EGFP with neuronal, microglial or astroglial markers in this region, thus proving polydendrocyte unipotent differentiation potential under physiological conditions. Three days after MCAo the number of EGFP+ cells in the gliotic tissue dramatically increased when compared to control animals, and these cells displayed properties of proliferating cells. However, in later phases after MCAo a large subpopulation of EGFP+ cells expressed protein markers and electrophysiological properties of astrocytes that contribute to the formation of glial scar. Importantly, some EGFP+ cells displayed membrane properties typical for neural precursor cells, and moreover these cells expressed doublecortin (DCX) – a marker of newly-derived neuronal cells. Taken together, our data indicate that polydendrocytes in the dorsal cortex display multipotent differentiation potential after focal ischemia.
Collapse
Affiliation(s)
- Pavel Honsa
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic
- 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Helena Pivonkova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic
- 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - David Dzamba
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic
- 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marcela Filipova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Miroslava Anderova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic
- * E-mail:
| |
Collapse
|
36
|
Kan EM, Ling EA, Lu J. Microenvironment changes in mild traumatic brain injury. Brain Res Bull 2012; 87:359-72. [PMID: 22289840 DOI: 10.1016/j.brainresbull.2012.01.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 01/10/2012] [Accepted: 01/12/2012] [Indexed: 02/08/2023]
Abstract
Traumatic brain injury (TBI) is a major public-health problem for which mild TBI (MTBI) makes up majority of the cases. MTBI is a poorly-understood health problem and can persist for years manifesting into neurological and non-neurological problems that can affect functional outcome. Presently, diagnosis of MTBI is based on symptoms reporting with poor understanding of ongoing pathophysiology, hence precluding prognosis and intervention. Other than rehabilitation, there is still no pharmacological treatment for the treatment of secondary injury and prevention of the development of cognitive and behavioural problems. The lack of external injuries and absence of detectable brain abnormalities lend support to MTBI developing at the cellular and biochemical level. However, the paucity of suitable and validated non-invasive methods for accurate diagnosis of MTBI poses as a substantial challenge. Hence, it is crucial that a clinically useful evaluation and management procedure be instituted for MTBI that encompasses both molecular pathophysiology and functional outcome. The acute microenvironment changes post-MTBI presents an attractive target for modulation of MTBI symptoms and the development of cognitive changes later in life.
Collapse
Affiliation(s)
- Enci Mary Kan
- Combat Care Laboratory, Defence Medical and Environmental Research Institute, DSO National Laboratories, 27 Medical Drive, Singapore 117510, Singapore
| | | | | |
Collapse
|
37
|
Kucharova K, Chang Y, Boor A, Yong VW, Stallcup WB. Reduced inflammation accompanies diminished myelin damage and repair in the NG2 null mouse spinal cord. J Neuroinflammation 2011; 8:158. [PMID: 22078261 PMCID: PMC3229456 DOI: 10.1186/1742-2094-8-158] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Accepted: 11/13/2011] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a demyelinating disease in which blood-derived immune cells and activated microglia damage myelin in the central nervous system. While oligodendrocyte progenitor cells (OPCs) are essential for generating oligodendrocytes for myelin repair, other cell types also participate in the damage and repair processes. The NG2 proteoglycan is expressed by OPCs, pericytes, and macrophages/microglia. In this report we investigate the effects of NG2 on these cell types during spinal cord demyelination/remyelination. METHODS Demyelinated lesions were created by microinjecting 1% lysolecithin into the lumbar spinal cord. Following demyelination, NG2 expression patterns in wild type mice were studied via immunostaining. Immunolabeling was also used in wild type and NG2 null mice to compare the extent of myelin damage, the kinetics of myelin repair, and the respective responses of OPCs, pericytes, and macrophages/microglia. Cell proliferation was quantified by studies of BrdU incorporation, and cytokine expression levels were evaluated using qRT-PCR. RESULTS The initial volume of spinal cord demyelination in wild type mice is twice as large as in NG2 null mice. However, over the ensuing 5 weeks there is a 6-fold improvement in myelination in wild type mice, versus only a 2-fold improvement in NG2 null mice. NG2 ablation also results in reduced numbers of each of the three affected cell types. BrdU incorporation studies reveal that reduced cell proliferation is an important factor underlying NG2-dependent decreases in each of the three key cell populations. In addition, NG2 ablation reduces macrophage/microglial cell migration and shifts cytokine expression from a pro-inflammatory to anti-inflammatory phenotype. CONCLUSIONS Loss of NG2 expression leads to decreased proliferation of OPCs, pericytes, and macrophages/microglia, reducing the abundance of all three cell types in demyelinated spinal cord lesions. As a result of these NG2-dependent changes, the course of demyelination and remyelination in NG2 null mice differs from that seen in wild type mice, with both myelin damage and repair being reduced in the NG2 null mouse. These studies identify NG2 as an important factor in regulating myelin processing, suggesting that therapeutic targeting of the proteoglycan might offer a means of manipulating cell behavior in demyelinating diseases.
Collapse
|
38
|
Sharifi K, Morihiro Y, Maekawa M, Yasumoto Y, Hoshi H, Adachi Y, Sawada T, Tokuda N, Kondo H, Yoshikawa T, Suzuki M, Owada Y. FABP7 expression in normal and stab-injured brain cortex and its role in astrocyte proliferation. Histochem Cell Biol 2011; 136:501-13. [PMID: 21938553 PMCID: PMC3192944 DOI: 10.1007/s00418-011-0865-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2011] [Indexed: 02/07/2023]
Abstract
Reactive gliosis, in which astrocytes as well as other types of glial cells undergo massive proliferation, is a common hallmark of all brain pathologies. Brain-type fatty acid-binding protein (FABP7) is abundantly expressed in neural stem cells and astrocytes of developing brain, suggesting its role in differentiation and/or proliferation of glial cells through regulation of lipid metabolism and/or signaling. However, the role of FABP7 in proliferation of glial cells during reactive gliosis is unknown. In this study, we examined the expression of FABP7 in mouse cortical stab injury model and also the phenotype of FABP7-KO mice in glial cell proliferation. Western blotting showed that FABP7 expression was increased significantly in the injured cortex compared with the contralateral side. By immunohistochemistry, FABP7 was localized to GFAP+ astrocytes (21% of FABP7+ cells) and NG2+ oligodendrocyte progenitor cells (62%) in the normal cortex. In the injured cortex there was no change in the population of FABP7+/NG2+ cells, while there was a significant increase in FABP7+/GFAP+ cells. In the stab-injured cortex of FABP7-KO mice there was decrease in the total number of reactive astrocytes and in the number of BrdU+ astrocytes compared with wild-type mice. Primary cultured astrocytes from FABP7-KO mice also showed a significant decrease in proliferation and omega-3 fatty acid incorporation compared with wild-type astrocytes. Overall, these data suggest that FABP7 is involved in the proliferation of astrocytes by controlling cellular fatty acid homeostasis.
Collapse
Affiliation(s)
- Kazem Sharifi
- Department of Organ Anatomy, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Wohl SG, Schmeer CW, Friese T, Witte OW, Isenmann S. In situ dividing and phagocytosing retinal microglia express nestin, vimentin, and NG2 in vivo. PLoS One 2011; 6:e22408. [PMID: 21850226 PMCID: PMC3151247 DOI: 10.1371/journal.pone.0022408] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 06/21/2011] [Indexed: 11/18/2022] Open
Abstract
Background Following injury, microglia become activated with subsets expressing nestin as well as other neural markers. Moreover, cerebral microglia can give rise to neurons in vitro. In a previous study, we analysed the proliferation potential and nestin re-expression of retinal macroglial cells such as astrocytes and Müller cells after optic nerve (ON) lesion. However, we were unable to identify the majority of proliferative nestin+ cells. Thus, the present study evaluates expression of nestin and other neural markers in quiescent and proliferating microglia in naïve retina and following ON transection in adult rats in vivo. Methodology/Principal Findings For analysis of cell proliferation and cells fates, rats received BrdU injections. Microglia in retinal sections or isolated cells were characterized using immunofluorescence labeling with markers for microglia (e.g., Iba1, CD11b), cell proliferation, and neural cells (e.g., nestin, vimentin, NG2, GFAP, Doublecortin etc.). Cellular analyses were performed using confocal laser scanning microscopy. In the naïve adult rat retina, about 60% of resting ramified microglia expressed nestin. After ON transection, numbers of nestin+ microglia peaked to a maximum at 7 days, primarily due to in situ cell proliferation of exclusively nestin+ microglia. After 8 weeks, microglia numbers re-attained control levels, but 20% were still BrdU+ and nestin+, although no further local cell proliferation occurred. In addition, nestin+ microglia co-expressed vimentin and NG2, but not GFAP or neuronal markers. Fourteen days after injury and following retrograde labeling of retinal ganglion cells (RGCs) with Fluorogold (FG), nestin+NG2+ microglia were positive for the dye indicating an active involvement of a proliferating cell population in phagocytosing apoptotic retinal neurons. Conclusions/Significance The current study provides evidence that in adult rat retina, a specific resident population of microglia expresses proteins of immature neural cells that are involved in injury-induced cell proliferation and phagocytosis while transdifferentiation was not observed.
Collapse
Affiliation(s)
- Stefanie G Wohl
- Hans Berger Clinic of Neurology, Jena University Hospital, Jena, Germany.
| | | | | | | | | |
Collapse
|
40
|
Moransard M, Dann A, Staszewski O, Fontana A, Prinz M, Suter T. NG2 expressed by macrophages and oligodendrocyte precursor cells is dispensable in experimental autoimmune encephalomyelitis. ACTA ACUST UNITED AC 2011; 134:1315-30. [PMID: 21596769 DOI: 10.1093/brain/awr070] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Increased expression of the chondroitin proteoglycan NG2 is a prominent feature in central nervous system injury with unknown cellular source and biological relevance. Here, we describe the first detailed analysis of experimental autoimmune encephalomyelitis in NG2 knockout mice and NG2 knockout bone marrow chimeras. We show that both macrophages and oligodendrocyte progenitor cells express and secrete NG2 in response to transforming growth factor-β. A subpopulation of macrophages expresses NG2 within leucocyte infiltrates in the central nervous system, but only oligodendrocyte progenitor cells contribute to NG2 accumulation. Notably, NG2 plays no role in experimental autoimmune encephalomyelitis initiation, progression or recuperation. In concurrence, the immune response is unaltered in NG2-deficient mice as are the extent of central nervous system damage and degree of remyelination.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Antigens/genetics
- Antigens/metabolism
- Antigens, CD/metabolism
- Cell Proliferation
- Cells, Cultured
- Disease Models, Animal
- Disease Progression
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Flow Cytometry
- Glial Fibrillary Acidic Protein/metabolism
- Macrophages/metabolism
- Macrophages/ultrastructure
- Mice
- Mice, Inbred C57BL
- Microscopy, Electron, Transmission
- Neurons/metabolism
- Neurons/pathology
- Oligodendroglia/metabolism
- Oligodendroglia/ultrastructure
- Proteoglycans/genetics
- Proteoglycans/metabolism
- RNA, Messenger/metabolism
- Spinal Cord/pathology
- Stem Cells/metabolism
- Transforming Growth Factor beta/metabolism
- Transforming Growth Factor beta/pharmacology
- Up-Regulation/drug effects
- Up-Regulation/physiology
Collapse
Affiliation(s)
- Martijn Moransard
- Clinical Immunology, University Hospital Zurich, Häldeliweg 4, CH-8044 Zürich, Switzerland.
| | | | | | | | | | | |
Collapse
|
41
|
Zhu L, Lu J, Tay SSW, Jiang H, He BP. Induced NG2 expressing microglia in the facial motor nucleus after facial nerve axotomy. Neuroscience 2010; 166:842-51. [PMID: 20060036 DOI: 10.1016/j.neuroscience.2009.12.057] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2009] [Revised: 12/23/2009] [Accepted: 12/23/2009] [Indexed: 11/26/2022]
Abstract
Chondroitin sulfate proteoglycan (NG2) expressing cells, ubiquitously distributed in the CNS respond to injured or diseased neurons; however, their behaviors toward injured neurons have remained to be fully explored. In the present study, along with astrocytic and microglial responses, NG2 expressing cells reacted swiftly and robustly in the facial motor nucleus (FMN) subjected to axotomy. With time, hypertrophic NG2 expressing cells gradually adhered to and enwrapped the axotomized motoneurons. Tight encapsulations around axotomized motoneurons were eventually formed at 7, 14, and 28 days after axotomy. NG2 positive processes appeared to interpose between synapsin-1 immunoreactive nerve terminals and surfaces of axotomized motoneurons. Double labeling results showed that NG2 expressing cells encapsulating axotomized facial motoneurons were mainly microglia marked by OX42 and lectin; only a few of them were positive to platelet-derived growth factor-alpha receptor and none of them positive to ED-1. No Rhodamine particle was detected in the FMN ipsilateral to axotomy after venous injection of the particles. The results suggest that activated microglia in lesioned FMN were induced to express NG2 molecules. It is concluded that axotomized FMN showed two types of NG2 expressing cells namely constitutive NG2 cells and induced-NG2 expressing microglia.
Collapse
Affiliation(s)
- L Zhu
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, 117597 Singapore; Department of Plastic Surgery, Chang Zheng Hospital, Shanghai 200003, PR China
| | | | | | | | | |
Collapse
|