1
|
Ma YZ, Cao JX, Zhang YS, Su XM, Jing YH, Gao LP. T Cells Trafficking into the Brain in Aging and Alzheimer's Disease. J Neuroimmune Pharmacol 2024; 19:47. [PMID: 39180590 DOI: 10.1007/s11481-024-10147-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 08/05/2024] [Indexed: 08/26/2024]
Abstract
The meninges, choroid plexus (CP) and blood-brain barrier (BBB) are recognized as important gateways for peripheral immune cell trafficking into the central nervous system (CNS). Accumulation of peripheral immune cells in brain parenchyma can be observed during aging and Alzheimer's disease (AD). However, the mechanisms by which peripheral immune cells enter the CNS through these three pathways and how they interact with resident cells within the CNS to cause brain injury are not fully understood. In this paper, we review recent research on T cells recruitment in the brain during aging and AD. This review focuses on the possible pathways through which T cells infiltrate the brain, the evidence that T cells are recruited to the brain, and how infiltrating T cells interact with the resident cells in the CNS during aging and AD. Unraveling these issues will contribute to a better understanding of the mechanisms of aging and AD from the perspective of immunity, and hopefully develop new therapeutic strategies for brain aging and AD.
Collapse
Affiliation(s)
- Yue-Zhang Ma
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Jia-Xin Cao
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yi-Shu Zhang
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiao-Mei Su
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yu-Hong Jing
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
| | - Li-Ping Gao
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
| |
Collapse
|
2
|
Sacharczuk M, Mickael ME, Kubick N, Kamińska A, Horbańczuk JO, Atanasov AG, Religa P, Ławiński M. The Current Landscape of Hypotheses Describing the Contribution of CD4+ Heterogeneous Populations to ALS. Curr Issues Mol Biol 2024; 46:7846-7861. [PMID: 39194682 DOI: 10.3390/cimb46080465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/11/2024] [Accepted: 07/22/2024] [Indexed: 08/29/2024] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a poorly understood and fatal disease. It has a low prevalence and a 2-4 year survival period. Various theories and hypotheses relating to its development process have been proposed, albeit with no breakthrough in its treatment. Recently, the role of the adaptive immune system in ALS, particularly CD4+ T cells, has begun to be investigated. CD4+ T cells are a heterogeneous group of immune cells. They include highly pro-inflammatory types such as Th1 and Th17, as well as highly anti-inflammatory cells such as Tregs. However, the landscape of the role of CD4+ T cells in ALS is still not clearly understood. This review covers current hypotheses that elucidate how various CD4+ T cells can contribute to ALS development. These hypotheses include the SWITCH model, which suggests that, in the early stages of the disease, Tregs are highly capable of regulating the immune response. However, in the later stages of the disease, it seems that pro-inflammatory cells such as Th1 and Th17 are capable of overwhelming Treg function. The reason why this occurs is not known. Several research groups have proposed that CD4+ T cells as a whole might experience aging. Others have proposed that gamma delta T cells might directly target Tregs. Additionally, other research groups have argued that less well-known CD4+ T cells, such as Emoes+ CD4+ T cells, may be directly responsible for neuron death by producing granzyme B. We propose that the ALS landscape is highly complicated and that there is more than one feasible hypothesis. However, it is critical to take into consideration the differences in the ability of different populations of CD4+ T cells to infiltrate the blood-brain barrier, taking into account the brain region and the time of infiltration. Shedding more light on these still obscure factors can help to create a personalized therapy capable of regaining the balance of power in the battle between the anti-inflammatory and pro-inflammatory cells in the central nervous system of ALS patients.
Collapse
Affiliation(s)
- Mariusz Sacharczuk
- Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Jastrzębiec, Poland
- Department of Pharmacodynamics, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1B, 02-091 Warsaw, Poland
| | - Michel-Edwar Mickael
- Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Jastrzębiec, Poland
| | - Norwin Kubick
- Department of Biology, Institute of Plant Science and Microbiology, University of Hamburg, Ohnhorststr. 18, 22609 Hamburg, Germany
| | - Agnieszka Kamińska
- Faculty of Medicine, Collegium Medicum Cardinal Stefan Wyszyński University in Warsaw, 01-938 Warsaw, Poland
| | - Jarosław Olav Horbańczuk
- Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Jastrzębiec, Poland
| | - Atanas G Atanasov
- Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Jastrzębiec, Poland
- Ludwig Boltzmann Institute Digital Health and Patient Safety, Medical University of Vienna, 1090 Vienna, Austria
| | - Piotr Religa
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institute, SE-141 86 Stockholm, Sweden
| | - Michał Ławiński
- Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Jastrzębiec, Poland
- Department of General Surgery, Gastroenterology and Oncology, Medical University of Warsaw, 02-091 Warsaw, Poland
| |
Collapse
|
3
|
Shang Y, Zheng L, Du Y, Shang T, Liu X, Zou W. Role of Regulatory T Cells in Intracerebral Hemorrhage. Mol Neurobiol 2024:10.1007/s12035-024-04281-7. [PMID: 38877366 DOI: 10.1007/s12035-024-04281-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/03/2024] [Indexed: 06/16/2024]
Abstract
Intracerebral hemorrhage (ICH) is a common cerebrovascular disease that can lead to severe neurological dysfunction in surviving patients, resulting in a heavy burden on patients and their families. When ICH occurs, the blood‒brain barrier is disrupted, thereby promoting immune cell migration into damaged brain tissue. As important immunosuppressive T cells, regulatory T (Treg) cells are involved in the maintenance of immune homeostasis and the suppression of immune responses after ICH. Treg cells mitigate brain tissue damage after ICH in a variety of ways, such as inhibiting the neuroinflammatory response, protecting against blood‒brain barrier damage, reducing oxidative stress damage and promoting nerve repair. In this review, we discuss the changes in Treg cells in ICH clinical patients and experimental animals, the mechanisms by which Treg cells regulate ICH and treatments targeting Treg cells in ICH, aiming to support new therapeutic strategies for clinical treatment.
Collapse
Affiliation(s)
- Yaxin Shang
- The Graduate School, Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Lei Zheng
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, People's Republic of China
- Molecular Biology Laboratory of Clinical Integrated of Traditional Chinese and Western Medicine of Heilong Jiang Province, Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Yunpeng Du
- The Graduate School, Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Tong Shang
- The Graduate School, Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Xueting Liu
- The Graduate School, Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Wei Zou
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, People's Republic of China.
- Molecular Biology Laboratory of Clinical Integrated of Traditional Chinese and Western Medicine of Heilong Jiang Province, Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, People's Republic of China.
| |
Collapse
|
4
|
Milne SM, Lahiri A, Sanchez CL, Marshall MJ, Jahan I, Meares GP. Myelin oligodendrocyte glycoprotein reactive Th17 cells drive Janus Kinase 1 dependent transcriptional reprogramming in astrocytes and alter cell surface cytokine receptor profiles during experimental autoimmune encephalomyelitis. Sci Rep 2024; 14:13146. [PMID: 38849434 PMCID: PMC11161502 DOI: 10.1038/s41598-024-63877-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/03/2024] [Indexed: 06/09/2024] Open
Abstract
Multiple sclerosis (MS) is an autoimmune demyelinating disease affecting the central nervous system (CNS). T helper (Th) 17 cells are involved in the pathogenesis of MS and its animal model of experimental autoimmune encephalomyelitis (EAE) by infiltrating the CNS and producing effector molecules that engage resident glial cells. Among these glial cells, astrocytes have a central role in coordinating inflammatory processes by responding to cytokines and chemokines released by Th17 cells. In this study, we examined the impact of pathogenic Th17 cells on astrocytes in vitro and in vivo. We identified that Th17 cells reprogram astrocytes by driving transcriptomic changes partly through a Janus Kinase (JAK)1-dependent mechanism, which included increased chemokines, interferon-inducible genes, and cytokine receptors. In vivo, we observed a region-specific heterogeneity in the expression of cell surface cytokine receptors on astrocytes, including those for IFN-γ, IL-1, TNF-α, IL-17, TGFβ, and IL-10. Additionally, these receptors were dynamically regulated during EAE induced by adoptive transfer of myelin-reactive Th17 cells. This study overall provides evidence of Th17 cell reprogramming of astrocytes, which may drive changes in the astrocytic responsiveness to cytokines during autoimmune neuroinflammation.
Collapse
MESH Headings
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Animals
- Astrocytes/metabolism
- Th17 Cells/immunology
- Th17 Cells/metabolism
- Mice
- Myelin-Oligodendrocyte Glycoprotein
- Receptors, Cytokine/metabolism
- Receptors, Cytokine/genetics
- Janus Kinase 1/metabolism
- Mice, Inbred C57BL
- Cytokines/metabolism
- Cellular Reprogramming
- Female
- Cells, Cultured
Collapse
Affiliation(s)
- Sarah M Milne
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, 26506, USA
| | - Anirudhya Lahiri
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, 26506, USA
| | - Cristina L Sanchez
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, 26506, USA
| | - Micah J Marshall
- Department of Neurology, The Ohio State University College of Medicine, IBMR 415D, 460 Medical Center Drive, Columbus, OH, 43210, USA
| | - Ishrat Jahan
- Department of Neurology, The Ohio State University College of Medicine, IBMR 415D, 460 Medical Center Drive, Columbus, OH, 43210, USA
| | - Gordon P Meares
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, 26506, USA.
- Department of Neurology, The Ohio State University College of Medicine, IBMR 415D, 460 Medical Center Drive, Columbus, OH, 43210, USA.
- Department of Neuroscience, West Virginia University, Morgantown, WV, 26506, USA.
- Rockefeller Neuroscience Institute, Morgantown, WV, 26506, USA.
| |
Collapse
|
5
|
Lee SY, Chung WS. Astrocytic crosstalk with brain and immune cells in healthy and diseased conditions. Curr Opin Neurobiol 2024; 84:102840. [PMID: 38290370 DOI: 10.1016/j.conb.2024.102840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/04/2023] [Accepted: 01/05/2024] [Indexed: 02/01/2024]
Abstract
Astrocytes interact with various cell types, including neurons, vascular cells, microglia, and peripheral immune cells. These interactions are crucial for regulating normal brain functions as well as modulating neuroinflammation in pathological conditions. Recent transcriptomic and proteomic studies have identified critical molecules involved in astrocytic crosstalk with other cells, shedding light on their roles in maintaining brain homeostasis in both healthy and diseased conditions. Astrocytes perform these various roles through either direct or indirect physical associations with neuronal synapses and vasculature. Furthermore, astrocytes can communicate with other immune cells, such as microglia, T cells, and natural killer cells, through secreted molecules during neuroinflammation. In this review, we discuss the critical molecular basis of this astrocytic crosstalk and the underlying mechanisms of astrocyte communication with other cells. We propose that astrocytes function as a central hub in inter-connecting neurons, vasculatures, and immune cells in healthy and diseased brains.
Collapse
Affiliation(s)
- Se Young Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea. https://twitter.com/SYLee_neuro
| | - Won-Suk Chung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea.
| |
Collapse
|
6
|
Zolfaghari Baghbadorani P, Rayati Damavandi A, Moradi S, Ahmadi M, Bemani P, Aria H, Mottedayyen H, Rayati Damavandi A, Eskandari N, Fathi F. Current advances in stem cell therapy in the treatment of multiple sclerosis. Rev Neurosci 2023; 34:613-633. [PMID: 36496351 DOI: 10.1515/revneuro-2022-0102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/18/2022] [Indexed: 08/04/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory disease related to the central nervous system (CNS) with a significant global burden. In this illness, the immune system plays an essential role in its pathophysiology and progression. The currently available treatments are not recognized as curable options and, at best, might slow the progression of MS injuries to the CNS. However, stem cell treatment has provided a new avenue for treating MS. Stem cells may enhance CNS healing and regulate immunological responses. Likewise, stem cells can come from various sources, including adipose, neuronal, bone marrow, and embryonic tissues. Choosing the optimal cell source for stem cell therapy is still a difficult verdict. A type of stem cell known as mesenchymal stem cells (MSCs) is obtainable from different sources and has a strong immunomodulatory impact on the immune system. According to mounting data, the umbilical cord and adipose tissue may serve as appropriate sources for the isolation of MSCs. Human amniotic epithelial cells (hAECs), as novel stem cell sources with immune-regulatory effects, regenerative properties, and decreased antigenicity, can also be thought of as a new upcoming contender for MS treatment. Overall, the administration of stem cells in different sets of animal and clinical trials has shown immunomodulatory and neuroprotective results. Therefore, this review aims to discuss the different types of stem cells by focusing on MSCs and their mechanisms, which can be used to treat and improve the outcomes of MS disease.
Collapse
Affiliation(s)
| | - Amirmasoud Rayati Damavandi
- Students' Scientific Research Center, Exceptional Talents Development Center, Tehran University of Medical Sciences, Keshavarz Blvrd, Vesal Shirazi St., Tehran 1417613151, Iran
| | - Samira Moradi
- School of Medicine, Hormozgan University of Medical Sciences Chamran Blvrd., Hormozgan 7919693116, Bandar Abbass, Iran
| | - Meysam Ahmadi
- School of Medicine, Shiraz University of Medical Sciences, Fars, Zand St., Shiraz 7134814336, Iran
| | - Peyman Bemani
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Hezar Jerib St., Isfahan 8174673461, Iran
| | - Hamid Aria
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Hezar Jerib St., Isfahan 8174673461, Iran
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fars, Ibn Sina Sq., Fasa 7461686688, Iran
| | - Hossein Mottedayyen
- Department of Immunology, School of Medicine, Kashan University of Medical Sciences, Ravandi Blvrd, Isfahan, Kashan 8715988141, Iran
| | - Amirhossein Rayati Damavandi
- Student's Research Committee, Pharmaceutical Sciences Branch, Islamic Azad University, Yakhchal St., Tehran 193951498, Iran
| | - Nahid Eskandari
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Hezar Jerib St., Isfahan 8174673461, Iran
| | - Farshid Fathi
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Hezar Jerib St., Isfahan 8174673461, Iran
| |
Collapse
|
7
|
Manjili MH. The adaptation model of immunity: A new insight into aetiology and treatment of multiple sclerosis. Scand J Immunol 2023; 97:e13255. [PMID: 36680379 DOI: 10.1111/sji.13255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 12/04/2022] [Accepted: 01/18/2023] [Indexed: 01/22/2023]
Abstract
Current research and drug development for multiple sclerosis (MS) is fully influenced by the self-nonself (SNS) model of immunity, suggesting that breakage of immunological tolerance towards self-antigens expressed in the central nervous system (CNS) is responsible for pathogenesis of MS; thus, immune suppressive drugs are recommended for the management of the disease. However, this model provides incomplete understanding of the causes and pathways involved in the onset and progression of MS and limits our ability to effectively treat this neurological disease. Some pre-clinical and clinical reports have been misunderstood; some others have been underappreciated because of the lack of a theoretical model that can explain them. Also, current immunotherapies are guided according to the models that are not designed to explain the functional outcomes of an immune response. The adaptation model of immunity is proposed to offer a new understanding of the existing data and galvanize a new direction for the treatment of MS. According to this model, the immune system continuously communicates with the CNS through the adaptation receptors (AdRs) and adaptation ligands (AdLs) or co-receptors, signal IV, to support cell growth and neuroplasticity. Alterations in the expression of the neuronal AdRs results in MS by shifting the cerebral inflammatory immune responses from remyelination to demyelination. Therefore, novel therapeutics for MS should be focused on the discovery and targeting of the AdR/AdL axis in the CNS rather than carrying on with immune suppressive interventions.
Collapse
Affiliation(s)
- Masoud H Manjili
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, Massey Cancer Center, Richmond, Virginia, USA
| |
Collapse
|
8
|
Feng W, Zhang Y, Sun P, Xiao M. Acquired immunity and Alzheimer's disease. J Biomed Res 2023; 37:15-29. [PMID: 36165328 PMCID: PMC9898041 DOI: 10.7555/jbr.36.20220083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by progressive cognitive defects. The role of the central immune system dominated by microglia in the progression of AD has been extensively investigated. However, little is known about the peripheral immune system in AD pathogenesis. Recently, with the discovery of the meningeal lymphatic vessels and glymphatic system, the roles of the acquired immunity in the maintenance of central homeostasis and neurodegenerative diseases have attracted an increasing attention. The T cells not only regulate the function of neurons, astrocytes, microglia, oligodendrocytes and brain microvascular endothelial cells, but also participate in the clearance of β-amyloid (Aβ) plaques. Apart from producing antibodies to bind Aβ peptides, the B cells affect Aβ-related cascades via a variety of antibody-independent mechanisms. This review systemically summarizes the recent progress in understanding pathophysiological roles of the T cells and B cells in AD.
Collapse
Affiliation(s)
- Weixi Feng
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu 211166, China,Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Chinese Academy of Sciences, Shanghai 200031, China,Weixi Feng, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, Jiangsu 211166, China. Tel: +86-25-86869338; E-mail:
| | - Yanli Zhang
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu 211166, China,Brain Institute, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Peng Sun
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu 211166, China,Brain Institute, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Ming Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu 211166, China,Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Chinese Academy of Sciences, Shanghai 200031, China,Brain Institute, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
9
|
Broekaart DWM, Zimmer TS, Cohen ST, Tessers R, Anink JJ, de Vries HE, Gorter JA, Prades R, Aronica E, van Vliet EA. The Gelatinase Inhibitor ACT-03 Reduces Gliosis in the Rapid Kindling Rat Model of Epilepsy, and Attenuates Inflammation and Loss of Barrier Integrity In Vitro. Biomedicines 2022; 10:biomedicines10092117. [PMID: 36140216 PMCID: PMC9495904 DOI: 10.3390/biomedicines10092117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/12/2022] [Accepted: 08/20/2022] [Indexed: 11/25/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are endopeptidases responsible for the cleavage of intra- and extracellular proteins. Several brain MMPs have been implicated in neurological disorders including epilepsy. We recently showed that the novel gelatinase inhibitor ACT-03 has disease-modifying effects in models of epilepsy. Here, we studied its effects on neuroinflammation and blood–brain barrier (BBB) integrity. Using the rapid kindling rat model of epilepsy, we examined whether ACT-03 affected astro- and microgliosis in the brain using immunohistochemistry. Cellular and molecular alterations were further studied in vitro using human fetal astrocyte and brain endothelial cell (hCMEC/D3) cultures, with a focus on neuroinflammatory markers as well as on barrier permeability using an endothelial and astrocyte co-culture model. We observed less astro- and microgliosis in the brains of kindled animals treated with ACT-03 compared to control vehicle-treated animals. In vitro, ACT-03 treatment attenuated stimulation-induced mRNA expression of several pro-inflammatory factors in human fetal astrocytes and brain endothelial cells, as well as a loss of barrier integrity in endothelial and astrocyte co-cultures. Since ACT-03 has disease-modifying effects in epilepsy models, possibly via limiting gliosis, inflammation, and barrier integrity loss, it is of interest to further evaluate its effects in a clinical trial.
Collapse
Affiliation(s)
- Diede W. M. Broekaart
- Amsterdam UMC, Location University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Till S. Zimmer
- Amsterdam UMC, Location University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Sophie T. Cohen
- Amsterdam UMC, Location University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Rianne Tessers
- Amsterdam UMC, Location University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Jasper J. Anink
- Amsterdam UMC, Location University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Helga E. de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, 1081 HV Amsterdam, The Netherlands
| | - Jan A. Gorter
- Swammerdam Institute for Life Sciences Center for Neuroscience, University of Amsterdam, 1098 XH Amsterdam, The Netherlands
| | - Roger Prades
- Accure Therapeutics S.L., 08028 Barcelona, Spain
| | - Eleonora Aronica
- Amsterdam UMC, Location University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), 2103 SW Heemstede, The Netherlands
- Correspondence: (E.A.); (E.A.v.V.)
| | - Erwin A. van Vliet
- Amsterdam UMC, Location University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Swammerdam Institute for Life Sciences Center for Neuroscience, University of Amsterdam, 1098 XH Amsterdam, The Netherlands
- Correspondence: (E.A.); (E.A.v.V.)
| |
Collapse
|
10
|
Cystatin C Plays a Sex-Dependent Detrimental Role in Experimental Autoimmune Encephalomyelitis. Cell Rep 2021; 33:108236. [PMID: 33027652 PMCID: PMC8603395 DOI: 10.1016/j.celrep.2020.108236] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/17/2020] [Accepted: 09/15/2020] [Indexed: 12/31/2022] Open
Abstract
The cysteine protease inhibitor Cystatin C (CST3) is highly expressed in the brains of multiple sclerosis (MS) patients and C57BL/6J mice with experimental autoimmune encephalomyelitis (EAE; a model of MS), but its roles in the diseases are unknown. Here, we show that CST3 plays a detrimental function in myelin oligodendrocyte glycoprotein 35–55 (MOG35–55)-induced EAE but only in female animals. Female Cst3 null mice display significantly lower clinical signs of disease compared to wild-type (WT) littermates. This difference is associated with reduced interleukin-6 production and lower expression of key proteins (CD80, CD86, major histocompatibility complex [MHC] II, LC3A/B) involved in antigen processing, presentation, and co-stimulation in antigen-presenting cells (APCs). In contrast, male WT and Cst3−/− mice and cells show no differences in EAE signs or APC function. Further, the sex-dependent effect of CST3 in EAE is sensitive to gonadal hormones. Altogether, we have shown that CST3 has a sex-dependent role in MOG35–55-induced EAE. Cystatin C (CST3) is increased in the brains of multiple sclerosis patients, but its role is unknown. In a mouse model of the disease, Hoghooghi et al. find that CST3 has a detrimental function but only in female animals. The effect is related to activation of antigen-presenting cells of the immune system.
Collapse
|
11
|
Qiu YM, Zhang CL, Chen AQ, Wang HL, Zhou YF, Li YN, Hu B. Immune Cells in the BBB Disruption After Acute Ischemic Stroke: Targets for Immune Therapy? Front Immunol 2021; 12:678744. [PMID: 34248961 PMCID: PMC8260997 DOI: 10.3389/fimmu.2021.678744] [Citation(s) in RCA: 167] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/31/2021] [Indexed: 12/15/2022] Open
Abstract
Blood-Brain Barrier (BBB) disruption is an important pathophysiological process of acute ischemic stroke (AIS), resulting in devastating malignant brain edema and hemorrhagic transformation. The rapid activation of immune cells plays a critical role in BBB disruption after ischemic stroke. Infiltrating blood-borne immune cells (neutrophils, monocytes, and T lymphocytes) increase BBB permeability, as they cause microvascular disorder and secrete inflammation-associated molecules. In contrast, they promote BBB repair and angiogenesis in the latter phase of ischemic stroke. The profound immunological effects of cerebral immune cells (microglia, astrocytes, and pericytes) on BBB disruption have been underestimated in ischemic stroke. Post-stroke microglia and astrocytes can adopt both an M1/A1 or M2/A2 phenotype, which influence BBB integrity differently. However, whether pericytes acquire microglia phenotype and exert immunological effects on the BBB remains controversial. Thus, better understanding the inflammatory mechanism underlying BBB disruption can lead to the identification of more promising biological targets to develop treatments that minimize the onset of life-threatening complications and to improve existing treatments in patients. However, early attempts to inhibit the infiltration of circulating immune cells into the brain by blocking adhesion molecules, that were successful in experimental stroke failed in clinical trials. Therefore, new immunoregulatory therapeutic strategies for acute ischemic stroke are desperately warranted. Herein, we highlight the role of circulating and cerebral immune cells in BBB disruption and the crosstalk between them following acute ischemic stroke. Using a robust theoretical background, we discuss potential and effective immunotherapeutic targets to regulate BBB permeability after acute ischemic stroke.
Collapse
Affiliation(s)
| | | | | | | | | | - Ya-nan Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
Astrocytes in Multiple Sclerosis-Essential Constituents with Diverse Multifaceted Functions. Int J Mol Sci 2021; 22:ijms22115904. [PMID: 34072790 PMCID: PMC8198285 DOI: 10.3390/ijms22115904] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 11/19/2022] Open
Abstract
In multiple sclerosis (MS), astrocytes respond to the inflammatory stimulation with an early robust process of morphological, transcriptional, biochemical, and functional remodeling. Recent studies utilizing novel technologies in samples from MS patients, and in an animal model of MS, experimental autoimmune encephalomyelitis (EAE), exposed the detrimental and the beneficial, in part contradictory, functions of this heterogeneous cell population. In this review, we summarize the various roles of astrocytes in recruiting immune cells to lesion sites, engendering the inflammatory loop, and inflicting tissue damage. The roles of astrocytes in suppressing excessive inflammation and promoting neuroprotection and repair processes is also discussed. The pivotal roles played by astrocytes make them an attractive therapeutic target. Improved understanding of astrocyte function and diversity, and the mechanisms by which they are regulated may lead to the development of novel approaches to selectively block astrocytic detrimental responses and/or enhance their protective properties.
Collapse
|
13
|
Kaddatz H, Joost S, Nedelcu J, Chrzanowski U, Schmitz C, Gingele S, Gudi V, Stangel M, Zhan J, Santrau E, Greiner T, Frenz J, Müller-Hilke B, Müller M, Amor S, van der Valk P, Kipp M. Cuprizone-induced demyelination triggers a CD8-pronounced T cell recruitment. Glia 2020; 69:925-942. [PMID: 33245604 DOI: 10.1002/glia.23937] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 01/25/2023]
Abstract
The loss of myelinating oligodendrocytes is a key characteristic of many neurological diseases, including Multiple Sclerosis (MS). In progressive MS, where effective treatment options are limited, peripheral immune cells can be found at the site of demyelination and are suggested to play a functional role during disease progression. In this study, we hypothesize that metabolic oligodendrocyte injury, caused by feeding the copper chelator cuprizone, is a potent trigger for peripheral immune cell recruitment into the central nervous system (CNS). We used immunohistochemistry and flow cytometry to evaluate the composition, density, and activation status of infiltrating T lymphocytes in cuprizone-intoxicated mice and post-mortem progressive MS tissues. Our results demonstrate a predominance of CD8+ T cells along with high proliferation rates and cytotoxic granule expression, indicating an antigenic and pro-inflammatory milieu in the CNS of cuprizone-intoxicated mice. Numbers of recruited T cells and the composition of lymphocytic infiltrates in cuprizone-intoxicated mice were found to be comparable to those found in progressive MS lesions. Finally, amelioration of the cuprizone-induced pathology by treating mice with laquinimod significantly reduces the number of recruited T cells. Overall, this study provides strong evidence that toxic demyelination is a sufficient trigger for T cells to infiltrate the demyelinated CNS. Further investigation of the mode of action and functional consequence of T cell recruitment might offer promising new therapeutic approaches for progressive MS.
Collapse
Affiliation(s)
- Hannes Kaddatz
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
| | - Sarah Joost
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
| | - Julia Nedelcu
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany.,Institute of Anatomy II, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Uta Chrzanowski
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany.,Institute of Anatomy II, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Christoph Schmitz
- Institute of Anatomy II, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Stefan Gingele
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Viktoria Gudi
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Martin Stangel
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Jiangshan Zhan
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
| | - Emily Santrau
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
| | - Theresa Greiner
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
| | - Julia Frenz
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
| | - Brigitte Müller-Hilke
- Core Facility for Cell Sorting and Analysing, Rostock University Medical Center, Rostock, Germany
| | - Michael Müller
- Core Facility for Cell Sorting and Analysing, Rostock University Medical Center, Rostock, Germany
| | - Sandra Amor
- Department of Pathology, Amsterdam UMC, Amsterdam, The Netherlands.,Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | - Markus Kipp
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
14
|
Drugs Modulating CD4+ T Cells Blood-Brain Barrier Interaction in Alzheimer's Disease. Pharmaceutics 2020; 12:pharmaceutics12090880. [PMID: 32948022 PMCID: PMC7558445 DOI: 10.3390/pharmaceutics12090880] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/07/2020] [Accepted: 09/14/2020] [Indexed: 12/21/2022] Open
Abstract
The effect of Alzheimer’s disease (AD) medications on CD4+ T cells homing has not been thoroughly investigated. CD4+ T cells could both exacerbate and reduce AD symptoms based on their infiltrating subpopulations. Proinflammatory subpopulations such as Th1 and Th17 constitute a major source of proinflammatory cytokines that reduce endothelial integrity and stimulate astrocytes, resulting in the production of amyloid β. Anti-inflammatory subpopulations such as Th2 and Tregs reduce inflammation and regulate the function of Th1 and Th17. Recently, pathogenic Th17 has been shown to have a superior infiltrating capacity compared to other major CD4+ T cell subpopulations. Alzheimer’s drugs such as donepezil (Aricept), rivastigmine (Exelon), galantamine (Razadyne), and memantine (Namenda) are known to play an important part in regulating the mechanisms of the neurotransmitters. However, little is known about the effect of these drugs on CD4+ T cell subpopulations’ infiltration of the brain during AD. In this review, we focus on understanding the influence of AD drugs on CD4+ T cell subpopulation interactions with the BBB in AD. While current AD therapies improve endothelial integrity and reduce astrocytes activations, they vary according to their influence on various CD4+ T cell subpopulations. Donepezil reduces the numbers of Th1 but not Th2, Rivastigmine inhibits Th1 and Th17 but not Th2, and memantine reduces Th1 but not Treg. However, none of the current AD drugs is specifically designed to target the dysregulated balance in the Th17/Treg axis. Future drug design approaches should specifically consider inhibiting CD4+ Th17 to improve AD prognosis.
Collapse
|
15
|
da Silva LC, Lima IVDA, da Silva MCM, Corrêa TA, de Souza VP, de Almeida MV, de Oliveira ACP, Ferreira AP. A new lipophilic amino alcohol, chemically similar to compound FTY720, attenuates the pathogenesis of experimental autoimmune encephalomyelitis by PI3K/Akt pathway inhibition. Int Immunopharmacol 2020; 88:106919. [PMID: 32871475 DOI: 10.1016/j.intimp.2020.106919] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/16/2020] [Accepted: 08/17/2020] [Indexed: 01/11/2023]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is one of the main animal models used for the study of Multiple Sclerosis (MS). Long-chain lipophilic amino alcohols with immunoregulatory activities have already been studied in some models of inflammatory diseases, but the action of these compounds in EAE and MS is still unknown. In this study, we investigated whether the lipophilic amino alcohol 4b would act to improve the clinical signs of EAE and reduce the demyelination process and the neuroinflammatory parameters in the spinal cord, as well as the inflammatory process in the inguinal lymph nodes, of C57Bl/6 mice induced with EAE after stimulation with MOG35-55 and pertussis toxin. The 4b treatment (1.0 mg/kg/day) was orally administered, starting on the day of onset of clinical signs of the disease (10th) and ending on the 20th day after immunization. This treatment was able to reduce the cell count on the inguinal lymph nodes, the migration of inflammatory cells into the central nervous system (CNS), as well as the processes of microgliosis, astrogliosis, and the production of chemokines and pro-inflammatory cytokines, thus increasing the IL-10 anti-inflammatory cytokine levels in EAE mice. The inhibition of Akt phosphorylation in the CNS of EAE mice after treatment with 4b indicates that the immunoregulatory action of 4b is related to the PI3K/Akt signaling pathway. Our results indicate the immunoregulatory efficacy of the new compound 4b in the control of some inflammatory parameters and in the glial proliferation. In addition, 4b was able to reduce the demyelination of neurons and the worsening of clinical signs of EAE as effectively as the compound FTY720, the first oral drug approved by the FDA for the treatment of MS.
Collapse
Affiliation(s)
- Luan Cristian da Silva
- Department of Parasitology, Microbiology and Immunology, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil.
| | - Isabel Vieira de Assis Lima
- Department of Parasitology, Microbiology and Immunology, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | | | - Taís Arthur Corrêa
- Department of Exact and Earth Sciences, State University of Minas Gerais, Frutal 38200-000, Brazil
| | - Viviane Passos de Souza
- Department of Parasitology, Microbiology and Immunology, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | | | | | - Ana Paula Ferreira
- Department of Parasitology, Microbiology and Immunology, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| |
Collapse
|
16
|
Spampinato SF, Merlo S, Fagone E, Fruciano M, Sano Y, Kanda T, Sortino MA. Reciprocal Interplay Between Astrocytes and CD4+ Cells Affects Blood-Brain Barrier and Neuronal Function in Response to β Amyloid. Front Mol Neurosci 2020; 13:120. [PMID: 32719583 PMCID: PMC7347984 DOI: 10.3389/fnmol.2020.00120] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022] Open
Abstract
Background: In Alzheimer’s disease (AD) neuronal degeneration is associated with gliosis and infiltration of peripheral blood mononuclear cells (PBMCs), which participate in neuroinflammation. Defects at the blood-brain barrier (BBB) facilitate PBMCs migration towards the central nervous system (CNS) and in particular CD4+ T cells have been found in areas severely affected in AD. However, the role of T cells, once they migrate into the CNS, is not well defined. CD4+ cells interact with astrocytes able to release several factors and cytokines that can modulate T cell polarization; similarly, astrocytic properties are modulated after interaction with T cells. Methods: In in vitro models, astrocytes were primed with β-amyloid (Aβ; 2.5 μM, 5 h) and then co-cultured with magnetically isolated CD4+ cells. Cytokines expression was evaluated both in co-cultured CD4+ cells and astrocytes. The effects of this crosstalk were further evaluated by co-culturing CD4+ cells with the neuronal-like SH-SY5Y cell line and astrocytes with endothelial cells. Results: The pattern of cytokines and trophic factors expressed by CD4+ cells were strongly modulated in the presence of Aβ-primed astrocytes. Specifically, the percentage of IL-4+ and IFNγ+ CD4+ cells was significantly increased and reduced, respectively. Further, increased BDNF mRNA levels were observed in CD4+ cells. When SH-SY5Y cells were co-cultured with astrocyte-conditioned CD4+ cells and exposed to Aβ, the reduction of the presynaptic protein synaptophysin was prevented with a BDNF-dependent mechanism. In astrocytes co-cultured with CD4+ cells, reduced mRNA levels of inflammatory cytokines and VEGF were observed. This was paralleled by the prevention of the reduction of claudin-5 when astrocytes were co-cultured with endothelial cells. Conclusion: Following Aβ exposure, there exists reciprocal crosstalk between infiltrating peripheral cells and astrocytes that in turn affects not only endothelial function and thus BBB properties, but also neuronal behavior. Since astrocytes are the first cells that lymphocytes interact with and are among the principal players in neuroinflammation occurring in AD, understanding this crosstalk may disclose new potential targets of intervention in the treatment of neurodegeneration.
Collapse
Affiliation(s)
- Simona Federica Spampinato
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Sara Merlo
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Evelina Fagone
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Mary Fruciano
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Yasuteru Sano
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Takashi Kanda
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Maria Angela Sortino
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| |
Collapse
|
17
|
Beneficial and Detrimental Effects of Regulatory T Cells in Neurotropic Virus Infections. Int J Mol Sci 2020; 21:ijms21051705. [PMID: 32131483 PMCID: PMC7084400 DOI: 10.3390/ijms21051705] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 02/07/2023] Open
Abstract
Neurotropic viruses infect the central nervous system (CNS) and cause acute or chronic neurologic disabilities. Regulatory T cells (Treg) play a critical role for immune homeostasis, but may inhibit pathogen-specific immunity in infectious disorders. The present review summarizes the current knowledge about Treg in human CNS infections and their animal models. Besides dampening pathogen-induced immunopathology, Treg have the ability to facilitate protective responses by supporting effector T cell trafficking to the infection site and the development of resident memory T cells. Moreover, Treg can reduce virus replication by inducing apoptosis of infected macrophages and attenuate neurotoxic astrogliosis and pro-inflammatory microglial responses. By contrast, detrimental effects of Treg are caused by suppression of antiviral immunity, allowing for virus persistence and latency. Opposing disease outcomes following Treg manipulation in different models might be attributed to differences in technique and timing of intervention, infection route, genetic background, and the host’s age. In addition, mouse models of virus-induced demyelination revealed that Treg are able to reduce autoimmunity and immune-mediated CNS damage in a disease phase-dependent manner. Understanding the unique properties of Treg and their complex interplay with effector cells represents a prerequisite for the development of new therapeutic approaches in neurotropic virus infections.
Collapse
|
18
|
Wang Y, Zhang JH, Sheng J, Shao A. Immunoreactive Cells After Cerebral Ischemia. Front Immunol 2019; 10:2781. [PMID: 31849964 PMCID: PMC6902047 DOI: 10.3389/fimmu.2019.02781] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/13/2019] [Indexed: 12/20/2022] Open
Abstract
The immune system is rapidly activated after ischemic stroke. As immune cells migrate and infiltrate across the blood-brain barrier into the ischemic region, a cascade of cellular and molecular biological reactions occur, involving migrated immune cells, resident glial cells, and the vascular endothelium. These events regulate infarction evolution and thus influence the outcome of ischemic stroke. Most immune cells exert dual effects on cerebral ischemia, and some crucial cells may become central targets in ischemic stroke treatment and rehabilitation.
Collapse
Affiliation(s)
- Yijie Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Jifang Sheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
19
|
Lin W, Chen W, Liu W, Xu Z, Zhang L. Sirtuin4 suppresses the anti-neuroinflammatory activity of infiltrating regulatory T cells in the traumatically injured spinal cord. Immunology 2019; 158:362-374. [PMID: 31559637 DOI: 10.1111/imm.13123] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 08/25/2019] [Accepted: 09/05/2019] [Indexed: 12/24/2022] Open
Abstract
The neuroinflammation following traumatic spinal cord injury (SCI) is a critical process that impacts both the injury and the recovery of spinal cord parenchyma. Infiltrating regulatory T (Treg) cells are potent anti-inflammatory cells that restrain post-SCI neuroinflammation. To understand the molecular mechanisms underlying the activity of infiltrating Treg cells, we used a mouse spinal cord compression injury model to analyze the role of Sirtuins (SIRTs) in the modulation of infiltrating Treg cell functions. We found that the expressions of SIRT4 and SIRT6 were up-regulated in infiltrating Treg cells. Using lentivirus-mediated gene expression or RNA interference, we revealed that SIRT4 substantially inhibited the expression of Foxp3, interleukin-10, and transforming growth factor-β in Treg cells, whereas SIRT6 had little effect on Treg cells. Consistently, SIRT4 overexpression weakened the suppressive effect of Treg cells on lipopolysaccharide-stimulated spinal cord CD11b+ myeloid cells. Knock-down of SIRT4 enhanced the anti-inflammatory activity of infiltrating Treg cells in the parenchyma of injured spinal cords. Additionally, SIRT4 overexpression blocked in vitro Treg cell generation from conventional T cells. Furthermore, SIRT4 down-regulated 5' AMP-activated protein kinase (AMPK) signaling in Treg cells, whereas the AMPK agonist AICAR restored the expression of Foxp3 and interleukin-10 in SIRT4-overexpressing Treg cells. In conclusion, our research unveils a new mechanism by which the post-SCI neuroinflammation is regulated.
Collapse
Affiliation(s)
- Wenping Lin
- Department of Spine Surgery, Shenzhen Pingle Orthopedic Hospital, Shenzhen, Guangdong, China.,Department of Orthopedic Surgery, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Wenkai Chen
- Department of Orthopedic Surgery, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Weifeng Liu
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Zhengquan Xu
- Department of Spine Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Liqun Zhang
- Department of Spine Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
20
|
Astrocytes in multiple sclerosis and experimental autoimmune encephalomyelitis: Star-shaped cells illuminating the darkness of CNS autoimmunity. Brain Behav Immun 2019; 80:10-24. [PMID: 31125711 DOI: 10.1016/j.bbi.2019.05.029] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/16/2019] [Accepted: 05/20/2019] [Indexed: 12/18/2022] Open
Abstract
Neuropathology in the human autoimmune disease multiple sclerosis (MS) is considered to be mediated by autoreactive leukocytes, such as T cells, B cells, and macrophages. However, the inflammation and tissue damage in MS and its animal model experimental autoimmune encephalomyelitis (EAE) is also critically regulated by astrocytes, the most abundant cell population in the central nervous system (CNS). Under physiological conditions, astrocytes are integral to the development and function of the CNS, whereas in CNS autoimmunity, astrocytes influence the pathogenesis, progression, and recovery of the diseases. In this review, we summarize recent advances in astrocytic functions in the context of MS and EAE, which are categorized into two opposite aspects, one being detrimental and the other beneficial. Inhibition of the detrimental functions and/or enhancement of the beneficial functions of astrocytes might be favorable for the treatment of MS.
Collapse
|
21
|
Noor S, Sun MS, Vanderwall AG, Havard MA, Sanchez JE, Harris NW, Nysus MV, Norenberg JP, West HT, Wagner CR, Jantzie LL, Mellios N, Milligan ED. LFA-1 antagonist (BIRT377) similarly reverses peripheral neuropathic pain in male and female mice with underlying sex divergent peripheral immune proinflammatory phenotypes. ACTA ACUST UNITED AC 2019; 6. [PMID: 31763376 PMCID: PMC6873931 DOI: 10.20517/2347-8659.2019.18] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Aim: The majority of preclinical studies investigating aberrant glial-neuroimmune actions underlying neuropathic pain have focused on male rodent models. Recently, studies have shown peripheral immune cells play a more prominent role than glial cells in mediating pathological pain in females. Here, we compared the onset and duration of allodynia in males and females, and the anti-allodynic action of a potentially novel therapeutic drug (BIRT377) that not only antagonizes the action of lymphocyte function-associated antigen-1 (LFA-1) to reduce cell migration in the periphery, but may also directly alter the cellular inflammatory bias. Methods: Male and female mice were subjected to peripheral nerve injury chronic constriction injury (CCI) applying two methods, using either 4–0 or 5–0 chromic gut suture material, to examine potential sex differences in the onset, magnitude and duration of allodynia. Hindpaw sensitivity before and after CCI and application of intravenous BIRT377 was assessed. Peripheral and spinal tissues were analyzed for protein (multiplex electrochemiluminescence technology) and mRNA expression (quantitative real-time PCR). The phenotype of peripheral T cells was determined using flow cytometry. Results: Sex differences in proinflammatory CCL2 and IL-1β and the anti-inflammatory IL-10 were observed from a set of cytokines analyzed. A profound proinflammatory T cell (Th17) response in the periphery and spinal cord was also observed in neuropathic females. BIRT377 reversed pain, reduced IL-1β and TNF, and increased IL-10 and transforming growth factor (TGF)-β1, also an anti-inflammatory cytokine, in both sexes. However, female-derived T cell cytokines are transcriptionally regulated by BIRT377, as demonstrated by reducing proinflammatory IL-17A production with concurrent increases in IL-10, TGF-β1 and the anti-inflammatory regulatory T cell-related factor, FOXP3. Conclusion: This study supports that divergent peripheral immune and neuroimmune responses during neuropathy exists between males and females. Moreover, the modulatory actions of BIRT377 on T cells during neuropathy are predominantly specific to females. These data highlight the necessity of including both sexes for studying drug efficacy and mechanisms of action in preclinical studies and clinical trials.
Collapse
Affiliation(s)
- Shahani Noor
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| | - Melody S Sun
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| | - Arden G Vanderwall
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA.,Department of Anesthesiology and Critical Care, University of New Mexico, Albuquerque, NM 87131, USA
| | - Mara A Havard
- Department of Anesthesiology and Critical Care, University of New Mexico, Albuquerque, NM 87131, USA
| | - Jacob E Sanchez
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| | - Nathan W Harris
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| | - Monique V Nysus
- Department of Radiopharmaceutical Sciences, College of Pharmacy, New Mexico Center for Isotopes in Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| | - Jeffrey P Norenberg
- Department of Radiopharmaceutical Sciences, College of Pharmacy, New Mexico Center for Isotopes in Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| | - Harrison T West
- Department of Medicinal Chemistry, University of Minnesota, College of Pharmacy, Minneapolis, MN 55455, USA
| | - Carsten R Wagner
- Department of Medicinal Chemistry, University of Minnesota, College of Pharmacy, Minneapolis, MN 55455, USA
| | - Lauren L Jantzie
- Department of Pediatrics and Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205-2196, USA
| | - Nikolaos Mellios
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| | - Erin D Milligan
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| |
Collapse
|
22
|
Ulivieri C, De Tommaso D, Finetti F, Ortensi B, Pelicci G, D'Elios MM, Ballerini C, Baldari CT. A T Cell Suppressive Circuitry Mediated by CD39 and Regulated by ShcC/Rai Is Induced in Astrocytes by Encephalitogenic T Cells. Front Immunol 2019; 10:1041. [PMID: 31134091 PMCID: PMC6524536 DOI: 10.3389/fimmu.2019.01041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/23/2019] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis is an autoimmune disease caused by autoreactive immune cell infiltration into the central nervous system leading to inflammation, demyelination, and neuronal loss. While myelin-reactive Th1 and Th17 are centrally implicated in multiple sclerosis pathogenesis, the local CNS microenvironment, which is shaped by both infiltrated immune cells and central nervous system resident cells, has emerged a key player in disease onset and progression. We have recently demonstrated that ShcC/Rai is as a novel astrocytic adaptor whose loss in mice protects from experimental autoimmune encephalomyelitis. Here, we have explored the mechanisms that underlie the ability of Rai-/- astrocytes to antagonize T cell-dependent neuroinflammation. We show that Rai deficiency enhances the ability of astrocytes to upregulate the expression and activity of the ectonucleotidase CD39, which catalyzes the conversion of extracellular ATP to the immunosuppressive metabolite adenosine, through both contact-dependent and-independent mechanisms. As a result, Rai-deficient astrocytes acquire an enhanced ability to suppress T-cell proliferation, which involves suppression of T cell receptor signaling and upregulation of the inhibitory receptor CTLA-4. Additionally, Rai-deficient astrocytes preferentially polarize to the neuroprotective A2 phenotype. These results identify a new mechanism, to which Rai contributes to a major extent, by which astrocytes modulate the pathogenic potential of autoreactive T cells.
Collapse
Affiliation(s)
| | | | | | - Barbara Ortensi
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy.,Department of Translational Medicine, Piemonte Orientale University "Amedeo Avogadro", Novara, Italy
| | - Giuliana Pelicci
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy.,Department of Translational Medicine, Piemonte Orientale University "Amedeo Avogadro", Novara, Italy
| | - Mario Milco D'Elios
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Clara Ballerini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | |
Collapse
|
23
|
Lee GA, Lin TN, Chen CY, Mau SY, Huang WZ, Kao YC, Ma RY, Liao NS. Interleukin 15 blockade protects the brain from cerebral ischemia-reperfusion injury. Brain Behav Immun 2018; 73:562-570. [PMID: 29959050 DOI: 10.1016/j.bbi.2018.06.021] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 06/02/2018] [Accepted: 06/22/2018] [Indexed: 01/08/2023] Open
Abstract
Acute ischemic stroke is followed by a complex interplay between the brain and the immune system in which ischemia-reperfusion leads to a detrimental inflammatory response that causes brain injury. In the brain, IL-15 is expressed by astrocytes, neurons and microglia. Previous study showed that ischemia-reperfusion induces expression of IL-15 by astrocytes. Transgenic over-expression of IL-15 in astrocytes aggravates ischemia-reperfusion brain damage by increasing the levels and promoting the effector functions of CD8+ T and NK cells. Treatment of neonatal rats with IL-15 neutralizing antibody before hypoxia-ischemia induction reduces the infarct volume. However, as stroke-induced inflammatory responses differ between neonate and adult brain, the effects of IL-15 blockade on the injury and immune response arising from stroke in adult animals has remained unclear. In this study, we examined the effect of post-ischemia/reperfusion IL-15 blockade on the pathophysiology of cerebral ischemia-reperfusion in adult mice. Using a cerebral ischemia-reperfusion model, we compared infarct size and the infiltrating immune cells in the brain of wild type (WT) mice and Il15-/- mice lacking NK and memory CD8+ T cells. We also evaluated the effects of IL-15 neutralizing antibody treatment on brain infarct volume, motor function, and the status of brain-infiltrating immune cells in WT mice. Il15-/- mice show a smaller infarct volume and lower numbers of activated brain-infiltrating NK, CD8+ T, and CD4+ T cells compared to WT mice after cerebral ischemia-reperfusion. Post-ischemia/reperfusion IL-15 blockade reduces infarct size and improves motor and locomotor activity. Furthermore, IL-15 blockade reduces the effector function of NK, CD8+ T, and CD4+ T cells in the ischemia-reperfusion brain of WT mice. Ablation of IL-15 responses after cerebral ischemia-reperfusion ameliorates brain injury in adult mice. Therefore, targeting IL-15 is a potential effective therapy for ischemic stroke.
Collapse
Affiliation(s)
- Gilbert Aaron Lee
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan.
| | - Teng-Nan Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Cheng-Yu Chen
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
| | - Shin-Yi Mau
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
| | - Wan-Zhen Huang
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yu-Chieh Kao
- Translational Imaging Research Center, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ruo-Yu Ma
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Nan-Shih Liao
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
24
|
Cerebral Ischemic Postconditioning Plays a Neuroprotective Role through Regulation of Central and Peripheral Glutamate. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6316059. [PMID: 30112410 PMCID: PMC6077516 DOI: 10.1155/2018/6316059] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/12/2018] [Indexed: 01/30/2023]
Abstract
Following cerebral ischemia/reperfusion (I/R) injury, a series of pathophysiological processes are stimulated in both the central nervous system (CNS) and the periphery, including, but not limited to, the peripheral immune and endocrine systems and underregulation of the neuroendocrine-immune network. Glutamate (Glu) is an important excitatory neurotransmitter in the CNS; its excitotoxicity following cerebral ischemia has been a focus of study for several decades. In addition, as a novel immunoregulator, Glu also regulates immune activity in both the CNS and periphery and may connect the CNS and periphery through regulation of the neuroendocrine-immune network. Ischemic postconditioning (IPostC) is powerful and activates various endogenous neuroprotective mechanisms following cerebral I/R, but only a few studies have focused on the mechanisms associated with Glu to date. Given that Glu plays an important and complex pathophysiological role, the understanding of Glu-related mechanisms of IPostC is an interesting area of research, which we review here.
Collapse
|
25
|
Orihara K, Odemuyiwa SO, Stefura WP, Ilarraza R, HayGlass KT, Moqbel R. Neurotransmitter signalling via NMDA receptors leads to decreased T helper type 1-like and enhanced T helper type 2-like immune balance in humans. Immunology 2017; 153:368-379. [PMID: 28940416 DOI: 10.1111/imm.12846] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 08/17/2017] [Accepted: 09/17/2017] [Indexed: 12/20/2022] Open
Abstract
Given the pivotal roles that CD4+ T cell imbalance plays in human immune disorders, much interest centres on better understanding influences that regulate human helper T-cell subset dominance in vivo. Here, using primary CD4+ T cells and short-term T helper type 1 (Th1) and Th2-like lines, we investigated roles and mechanisms by which neurotransmitter receptors may influence human type 1 versus type 2 immunity. We hypothesized that N-methyl-d-aspartate receptors (NMDA-R), which play key roles in memory and learning, can also regulate human CD4+ T cell function through induction of excitotoxicity. Fresh primary CD4+ T cells from healthy donors express functional NMDA-R that are strongly up-regulated upon T cell receptor (TCR) mediated activation. Synthetic and physiological NMDA-R agonists elicited Ca2+ flux and led to marked inhibition of type 1 but not type 2 or interleukin-10 cytokine responses. Among CD4+ lines, NMDA and quinolinic acid preferentially reduced cytokine production, Ca2+ flux, proliferation and survival of Th1-like cells through increased induction of cell death whereas Th2-like cells were largely spared. Collectively, the findings demonstrate that (i) NMDA-R is rapidly up-regulated upon CD4+ T cell activation in humans and (ii) Th1 versus Th2 cell functions such as proliferation, cytokine production and cell survival are differentially affected by NMDA-R agonists. Differential cytokine production and proliferative capacity of Th1 versus Th2 cells is attributable in part to increased physiological cell death among fully committed Th1 versus Th2 cells, leading to increased Th2-like dominance. Hence, excitotoxicity, beyond its roles in neuronal plasticity, may contribute to ongoing modulation of human T cell responses.
Collapse
Affiliation(s)
- Kanami Orihara
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Solomon O Odemuyiwa
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - William P Stefura
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Ramses Ilarraza
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada.,Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Kent T HayGlass
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Redwan Moqbel
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada.,Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
26
|
Di Benedetto S, Müller L, Wenger E, Düzel S, Pawelec G. Contribution of neuroinflammation and immunity to brain aging and the mitigating effects of physical and cognitive interventions. Neurosci Biobehav Rev 2017; 75:114-128. [PMID: 28161508 DOI: 10.1016/j.neubiorev.2017.01.044] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/24/2017] [Accepted: 01/30/2017] [Indexed: 01/08/2023]
Abstract
It is widely accepted that the brain and the immune system continuously interact during normal as well as pathological functioning. Human aging is commonly accompanied by low-grade inflammation in both the immune and central nervous systems, thought to contribute to many age-related diseases. This review of the current literature focuses first on the normal neuroimmune interactions occurring in the brain, which promote learning, memory and neuroplasticity. Further, we discuss the protective and dynamic role of barriers to neuroimmune interactions, which have become clearer with the recent discovery of the meningeal lymphatic system. Next, we consider age-related changes of the immune system and possible deleterious influences of immunosenescence and low-grade inflammation (inflammaging) on neurodegenerative processes in the normally aging brain. We survey the major immunomodulators and neuroregulators in the aging brain and their highly tuned dynamic and reciprocal interactions. Finally, we consider our current understanding of how physical activity, as well as a combination of physical and cognitive interventions, may mediate anti-inflammatory effects and thus positively impact brain aging.
Collapse
Affiliation(s)
- Svetlana Di Benedetto
- Max Planck Institute for Human Development, Center for Lifespan Psychology, Lentzeallee 94, 14195, Berlin, Germany; Center for Medical Research, Department of Internal Medicine II, University of Tübingen, Waldhörnlestr. 22, 72072 Tübingen, Germany
| | - Ludmila Müller
- Max Planck Institute for Human Development, Center for Lifespan Psychology, Lentzeallee 94, 14195, Berlin, Germany.
| | - Elisabeth Wenger
- Max Planck Institute for Human Development, Center for Lifespan Psychology, Lentzeallee 94, 14195, Berlin, Germany
| | - Sandra Düzel
- Max Planck Institute for Human Development, Center for Lifespan Psychology, Lentzeallee 94, 14195, Berlin, Germany
| | - Graham Pawelec
- Center for Medical Research, Department of Internal Medicine II, University of Tübingen, Waldhörnlestr. 22, 72072 Tübingen, Germany
| |
Collapse
|
27
|
Filipello F, Pozzi D, Proietti M, Romagnani A, Mazzitelli S, Matteoli M, Verderio C, Grassi F. Ectonucleotidase activity and immunosuppression in astrocyte-CD4 T cell bidirectional signaling. Oncotarget 2017; 7:5143-56. [PMID: 26784253 PMCID: PMC4868677 DOI: 10.18632/oncotarget.6914] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 01/01/2016] [Indexed: 12/04/2022] Open
Abstract
Astrocytes play a crucial role in neuroinflammation as part of the glia limitans, which regulates infiltration of the brain parenchyma by leukocytes. The signaling pathways and molecular events, which result from the interaction of activated T cells with astrocytes are poorly defined. Here we show that astrocytes promote the expression and enzymatic activity of CD39 and CD73 ectonucleotidases in recently activated CD4 cells by a contact dependent mechanism that is independent of T cell receptor interaction with class II major histocompatibility complex (MHC). Transforming growth factor-β (TGF-β) is robustly upregulated and sufficient to promote ectonucleotidases expression. T cell adhesion to astrocyte results in differentiation to an immunosuppressive phenotype defined by expression of the transcription factor Rorγt, which characterizes the CD4 T helper 17 subset. CD39 activity in T cells in turn inhibits spontaneous calcium oscillations in astrocytes that correlated with enhanced and reduced transcription of CCL2 chemokine and Sonic hedgehog (Shh), respectively. We hypothesize this TCR-independent interaction promote an immunosuppressive program in T cells to control possible brain injury by deregulated T cell activation during neuroinflammation. On the other hand, the increased secretion of CCL2 with concomitant reduction of Shh might promote leukocytes extravasation into the brain parenchyma.
Collapse
Affiliation(s)
- Fabia Filipello
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, Rozzano, Italy
| | - Davide Pozzi
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, Rozzano, Italy
| | - Michele Proietti
- Institute for Research in Biomedicine, Bellinzona, Switzerland.,Center of Chronic Immunodeficiency, University Medical Center, Freiburg, Germany
| | - Andrea Romagnani
- Institute for Research in Biomedicine, Bellinzona, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Sonia Mazzitelli
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, Rozzano, Italy.,Hertie Institute for Clinical Brain Research, University of Tubingen, Department of Cellular Neurology, Tubingen, Germany
| | - Michela Matteoli
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, Rozzano, Italy.,CNR Institute of Neuroscience, Milano, Italy
| | - Claudia Verderio
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, Rozzano, Italy.,CNR Institute of Neuroscience, Milano, Italy
| | - Fabio Grassi
- Institute for Research in Biomedicine, Bellinzona, Switzerland.,Department of Medical Biotechnology and Translational Medicine, University of Milan, Istituto Nazionale di Genetica Molecolare, Milan, Italy
| |
Collapse
|
28
|
Slack M, Wang T, Wang R. T cell metabolic reprogramming and plasticity. Mol Immunol 2015; 68:507-12. [PMID: 26277274 DOI: 10.1016/j.molimm.2015.07.036] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 07/26/2015] [Indexed: 12/21/2022]
Abstract
Upon antigen stimulation, small and quiescent naïve T cells undergo an approximately 24h growth phase followed by rapid proliferation. Depending on the nature of the antigen and cytokine milieu, these proliferating T cells differentiate into distinctive functional subgroups that are essential for appropriate immune defense and regulation. T cells undergo a characteristic metabolic rewiring that fulfills the dramatically increased bioenergetic and biosynthetic demands during the transition between resting, activation and differentiation. Beyond this, T cells are distributed throughout the body and are able to function in a wide range of physio-pathological environments, including some with a dramatic metabolic derangement. As such, T cells must quickly respond to and adapt to fluctuations in environmental nutrient levels. We consider such responsiveness and adaptation in terms of metabolic plasticity, that is, an evolutionarilly selected process which allows T cells to illicit robust immune functions in response to either a continuous or disrupted nutrient supply. In this review, we illustrate the relevant metabolic pathways in T cells and discuss the ability of T cells to change their metabolic substrates in response to changes in the environment.
Collapse
Affiliation(s)
- Maria Slack
- Department of Pediatrics, The Ohio State University School of Medicine, Columbus, OH, USA; Division of Allergy and Immunology Nationwide Children's Hospital, Columbus, OH, USA; Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, OH, USA
| | - Tingting Wang
- Center for Childhood Cancer and Blood Disease, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Ruoning Wang
- Center for Childhood Cancer and Blood Disease, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Hematology/Oncology & BMT, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University School of Medicine, Columbus, OH, USA.
| |
Collapse
|
29
|
Dumont CM, Park J, Shea LD. Controlled release strategies for modulating immune responses to promote tissue regeneration. J Control Release 2015; 219:155-166. [PMID: 26264833 DOI: 10.1016/j.jconrel.2015.08.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 08/04/2015] [Accepted: 08/05/2015] [Indexed: 01/06/2023]
Abstract
Advances in the field of tissue engineering have enhanced the potential of regenerative medicine, yet the efficacy of these strategies remains incomplete, and is limited by the innate and adaptive immune responses. The immune response associated with injury or disease combined with that mounted to biomaterials, transplanted cells, proteins, and gene therapies vectors can contribute to the inability to fully restore tissue function. Blocking immune responses such as with anti-inflammatory or immunosuppressive agents are either ineffective, as the immune response contributes significantly to regeneration, or have significant side effects. This review describes targeted strategies to modulate the immune response in order to limit tissue damage following injury, promote an anti-inflammatory environment that leads to regeneration, and induce antigen (Ag)-specific tolerance that can target degenerative diseases that destroy tissues and promote engraftment of transplanted cells. Focusing on targeted immuno-modulation, we describe local delivery techniques to sites of inflammation as well as systemic approaches that preferentially target subsets of immune populations.
Collapse
Affiliation(s)
- Courtney M Dumont
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| | - Jonghyuck Park
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48105, USA.
| |
Collapse
|
30
|
Lokensgard JR, Schachtele SJ, Mutnal MB, Sheng WS, Prasad S, Hu S. Chronic reactive gliosis following regulatory T cell depletion during acute MCMV encephalitis. Glia 2015; 63:1982-1996. [PMID: 26041050 DOI: 10.1002/glia.22868] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 05/12/2015] [Indexed: 12/21/2022]
Abstract
Long-term, persistent central nervous system inflammation is commonly seen following brain infection. Using a murine model of viral encephalitis (murine cytomegalovirus, MCMV) we have previously shown that post-encephalitic brains are maintained in an inflammatory state consisting of glial cell reactivity, retention of brain-infiltrating tissue-resident memory CD8+ T-cells, and long-term persistence of antibody-producing cells of the B-lineage. Here, we report that this neuroinflammation occurs concomitantly with accumulation and retention of immunosuppressive regulatory T-cells (Tregs), and is exacerbated following their ablation. However, the extent to which these Tregs function to control neuroimmune activation following MCMV encephalitis is unknown. In this study, we used Foxp3-diphtheria toxin receptor-GFP (Foxp3-DTR-GFP) transgenic mice, which upon administration of low-dose diphtheria toxin (DTx) results in the specific depletion of Tregs, to investigate their function. We found treatment with DTx during the acute phase of viral brain infection (0-4 dpi) resulted in depletion of Tregs from the brain, exacerbation of encephalitis (i.e., increased presence of CD4+ and CD8+ T-cells), and chronic reactive phenotypes of resident glial cells (i.e., elevated MHC Class II as well as PD-L1 levels, sustained microgliosis, and increased glial fibrillary acidic protein (GFAP) expression on astrocytes) versus untreated, infected animals. This chronic proinflammatory environment was associated with reduced cognitive performance in spatial learning and memory tasks (Barnes Maze) by convalescent animals. These data demonstrate that chronic glial cell activation, unremitting post-encephalitic neuroinflammation, and its associated long-term neurological sequelae in response to viral brain infection are modulated by the immunoregulatory properties of Tregs. GLIA 2015;63:1982-1996.
Collapse
Affiliation(s)
- James R Lokensgard
- Center for Infectious Diseases and Microbiology Translational Research, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Scott J Schachtele
- Center for Infectious Diseases and Microbiology Translational Research, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Manohar B Mutnal
- Center for Infectious Diseases and Microbiology Translational Research, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Wen S Sheng
- Center for Infectious Diseases and Microbiology Translational Research, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Sujata Prasad
- Center for Infectious Diseases and Microbiology Translational Research, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Shuxian Hu
- Center for Infectious Diseases and Microbiology Translational Research, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
31
|
Regulation of the Neurodegenerative Process Associated to Parkinson's Disease by CD4+ T-cells. J Neuroimmune Pharmacol 2015; 10:561-75. [PMID: 26018603 DOI: 10.1007/s11481-015-9618-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 05/19/2015] [Indexed: 01/09/2023]
Abstract
Neuroinflammation constitutes a fundamental process involved in the physiopathology of Parkinson's disease (PD). Microglial cells play a central role in the outcome of neuroinflammation and consequent neurodegeneration of dopaminergic neurons in the substantia nigra. Current evidence indicates that CD4+ T-cells infiltrate the central nervous system (CNS) in PD, where they play a critical role determining the functional phenotype of microglia, thus regulating the progression of the neurodegenerative process. Here, we first analysed the pathogenic role of inflammatory phenotypes and the beneficial role of anti-inflammatory phenotypes of encephalitogenic CD4+ T-cells involved in the physiopathology of PD. Next, we discussed how alterations of neurotransmitter levels observed in the basal ganglia throughout the time course of PD progression could be strongly affecting the behaviour of encephalitogenic CD4+ T-cells and thereby the outcome of the neuroinflammatory process and the consequent neurodegeneration of dopaminergic neurons. Afterward, we integrated the evidence indicating the involvement of an antigen-specific immune response mediated by T-cells and B-cells against CNS-derived self-constituents in PD. Consistent with the involvement of a relevant autoimmune component in PD, we also reviewed the polymorphisms of both, class I and class II major histocompatibility complexes, associated to the risk of PD. Overall, this study gives an overview of how an autoimmune component involved in PD plays a fundamental role in the progression of the neurodegenerative process.
Collapse
|
32
|
Interaction of astrocytes and T cells in physiological and pathological conditions. Brain Res 2015; 1623:63-73. [PMID: 25813828 DOI: 10.1016/j.brainres.2015.03.026] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 03/10/2015] [Accepted: 03/12/2015] [Indexed: 12/24/2022]
Abstract
The central nervous system (CNS) has long been recognized as a site of 'immune privilege' because of the existence of the blood brain barrier (BBB) which presumably isolates CNS from the peripheral immunosurveillance. Different from the peripheral organs, CNS is unique in response to all forms of CNS injury and disease which is mainly mediated by resident microglia and astrocyte. There is increasing evidence that immune cells are not only involved in neuroinflammation process but also the maintenance of CNS homeostasis. T cells, an important immune cell population, are involved in the pathogenesis of some neurological diseases by inducing either innate or adaptive immune responses. Astrocytes, which are the most abundant cell type in the CNS, maintain the integrity of BBB and actively participate in the initiation and progression of neurological diseases. Surprisingly, how astrocytes and T cells interact and the consequences of their interaction are not clear. In this review we briefly summarized T cells diversity and astrocyte function. Then, we examined the evidence for the astrocytes and T cells interaction under physiological and pathological conditions including ischemic stroke, multiple sclerosis, viral infection, and Alzheimer's disease. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
|