1
|
Yamazaki R, Azuma M, Osanai Y, Kouki T, Inagaki T, Kakita A, Takao M, Ohno N. Type I collagen secreted in white matter lesions inhibits remyelination and functional recovery. Cell Death Dis 2025; 16:285. [PMID: 40221393 PMCID: PMC11993711 DOI: 10.1038/s41419-025-07633-w] [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: 09/30/2024] [Revised: 03/31/2025] [Accepted: 04/04/2025] [Indexed: 04/14/2025]
Abstract
White matter injury is caused by cerebral blood flow disturbances associated with stroke and demyelinating diseases such as multiple sclerosis. Remyelination is induced spontaneously after white matter injury, but progressive multiple sclerosis and white matter stroke are usually characterised by remyelination failure. However, the mechanisms underlying impaired remyelination in lesions caused by demyelination and stroke remain unclear. In the current study, we demonstrated that collagen fibres accumulated in the demyelinated lesions of multiple sclerosis patients (age range 23-80 years) and white matter lesions of stroke patients (age range 80-87 years), suggesting that the accumulation of collagen fibres correlates with remyelination failure in these lesions. To investigate the function of collagen fibres in the white matter lesions, we generated two types of white matter injury in mice. We induced focal demyelination by lysolecithin (LPC) injection and ischemic stroke by endothelin 1 (ET1) injection into the internal capsule. We found that type I collagen fibres were secreted in ET1-induced lesions with impaired white matter regeneration in the chronic phase of disease. We also showed that monocyte-derived macrophages that infiltrated into lesions from the peripheral blood produced type I collagen after white matter injury, and that type I collagen also exacerbated microglial activation, astrogliosis, and axonal injury. Finally, we demonstrated that oligodendrocyte differentiation and remyelination were inhibited in the presence of type I collagen after LPC-induced demyelination. These results suggest that type I collagen secreted by monocyte-derived macrophages inhibited white matter regeneration, and therefore, the modulation of type I collagen metabolism might be a novel therapeutic target for white matter injury.
Collapse
Affiliation(s)
- Reiji Yamazaki
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Tochigi, Japan.
| | - Morio Azuma
- Department of Pharmacology, Division of Molecular Pharmacology, School of Medicine, Jichi Medical University, Tochigi, Japan
| | - Yasuyuki Osanai
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Tochigi, Japan
| | - Tom Kouki
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Tochigi, Japan
| | - Takeshi Inagaki
- Department of Anatomy, Division of Forensic Medicine, School of Medicine, Jichi Medical University, Tochigi, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masaki Takao
- Department of Clinical Laboratory, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Nobuhiko Ohno
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Tochigi, Japan
- Division of Ultrastructural Research, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| |
Collapse
|
2
|
Abiramalatha T, Amboiram P, Ponnala AK, Murkunde YV, Punnoose AM, Vivekanandhan A, Tamrakar S, Cruze LD, Mahadevan A. Development of a New Animal Model for Non-Cystic Periventricular Leukomalacia. Neurol India 2025; 73:320-324. [PMID: 40176223 DOI: 10.4103/neurol-india.neurol-india-d-23-00580] [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: 07/06/2024] [Accepted: 07/23/2024] [Indexed: 04/04/2025]
Abstract
Periventricular leukomalacia (PVL) is a major cause of cerebral palsy in preterm neonates. Though many interventions are being explored as therapeutic options in PVL, all these therapies are still in preclinical phase. This emphasizes the need for the development of robust animal models for PVL. We aimed to develop a new animal (rat) model for PVL using natural pattern of perinatal insults as in humans. The animal model was developed using a combination of three insults: prenatal inflammation, prenatal near total ischemia and postnatal recurrent hypoxia. Pathological and immunohistochemical analysis of pup brains was performed on day 28. We had 33 pups in experimental (inflammation, ischemia and hypoxia, LIH) group and 34 normal pups as controls. Histopathological analysis and immunohistochemistry with anti myelin basic protein (MBP) antibody showed myelin loss in corpus callosum or periventricular white matter in all pups in LIH group. Nissl's stain showed focal to generalized cortical disorganization. On functional assessment, LIH pups had prolonged escape latency in morris water maze test suggestive of cognitive/memory impairment. To conclude, we have developed a new animal model for PVL in rat, using a similar pattern of perinatal insults as in human preterm neonates. The model resembles non-cystic PVL in both pathological and functional analysis.
Collapse
Affiliation(s)
- Thangaraj Abiramalatha
- Department of Neonatology, Sri Ramachandra Institute of Higher Education and Research (SRIHER), Chennai, Tamil Nadu, India
| | - Prakash Amboiram
- Department of Neonatology, Sri Ramachandra Institute of Higher Education and Research (SRIHER), Chennai, Tamil Nadu, India
| | - Ananda Kumar Ponnala
- Centre for Toxicology and Developmental Research (CEFTE), Sri Ramachandra Institute of Higher Education and Research (SRIHER), Chennai, Tamil Nadu, India
| | - Yogeshkumar V Murkunde
- Centre for Toxicology and Developmental Research (CEFTE), Sri Ramachandra Institute of Higher Education and Research (SRIHER), Chennai, Tamil Nadu, India
| | - Alan Mathew Punnoose
- Department of Regenerative Medicine and Stem Cell Research, Sri Ramachandra Institute of Higher Education and Research (SRIHER), Chennai, Tamil Nadu, India
| | | | - Shonam Tamrakar
- Centre for Toxicology and Developmental Research (CEFTE), Sri Ramachandra Institute of Higher Education and Research (SRIHER), Chennai, Tamil Nadu, India
| | - Lawrence D Cruze
- Department of Pathology, Sri Ramachandra Institute of Higher Education and Research (SRIHER), Chennai, Tamil Nadu, India
| | - Anita Mahadevan
- Department of Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, Karnataka, India
| |
Collapse
|
3
|
Jiang YQ, Chen QZ, Yang Y, Zang CX, Ma JW, Wang JR, Dong YR, Zhou N, Yang X, Li FF, Bao XQ, Zhang D. White matter lesions contribute to motor and non-motor disorders in Parkinson's disease: a critical review. GeroScience 2025; 47:591-609. [PMID: 39576561 PMCID: PMC11872850 DOI: 10.1007/s11357-024-01428-1] [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: 07/09/2024] [Accepted: 11/08/2024] [Indexed: 03/04/2025] Open
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disease, characterized by movement disorders and non-motor symptoms like cognitive impairment and depression. Degeneration of dopaminergic neurons in the substantia nigra and Lewy bodies have long been considered as main neuropathological changes. However, recent magnetic resonance imaging (MRI) studies have shown that white matter lesions (WMLs) were present in PD patients. WMLs are characterized by loss or impairment of myelin sheath in central nerve fibers, which are closely correlated with motor and cognitive dysfunction in PD. WMLs alterations precede nigrostriatal neuronal losses and can independently affect the clinical severity or characteristics of motor coordination in PD patients. Currently, the exact mechanism of WMLs involvement in the occurrence and development of PD remains unclear. It is speculated that WMLs may participate in the pathogenesis of PD by disrupting important connections in brain or promoting axonal degeneration. In this review, we will discuss the pathological changes and mechanisms of WMLs, elaborate the impact of WMLs on the progression of PD, clarify the importance of WMLs in PD pathogenesis, and thus provide novel targets for PD treatments.
Collapse
Affiliation(s)
- Yue-Qi Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Qiu-Zhu Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Yang Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Cai-Xia Zang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Jing-Wei Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Jin-Rong Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Yi-Rong Dong
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Ning Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Xing Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Fang-Fang Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Xiu-Qi Bao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China.
| | - Dan Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China.
| |
Collapse
|
4
|
Blair HJ, Morales L, Cryan JF, Aburto MR. Neuroglia and the microbiota-gut-brain axis. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:171-196. [PMID: 40122624 DOI: 10.1016/b978-0-443-19104-6.00001-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Glial cells are key players in the regulation of nervous system functioning in both the central and enteric nervous systems. Glial cells are dynamic and respond to environmental cues to modulate their activity. Increasing evidence suggests that these signals include those originating from the gut microbiota, the community of microorganisms, including bacteria, viruses, archaea, and protozoa, that inhabit the gut. The gut microbiota and the brain communicate in a bidirectional manner across multiple signaling pathways and interfaces that together comprise the microbiota-gut-brain axis. Here, we detail the role of glial cells, including astrocytes, microglia, and oligodendrocytes in the central nervous system, and glial cells in the enteric nervous system along this gut-brain axis. We review what is known regarding the modulation of glia by microbial signals, in particular by microbial metabolites which signal to the brain through systemic circulation and via the vagus nerve. In addition, we highlight what is yet to be discovered regarding the role of other gut microbiota signaling pathways in glial cell modulation and the challenges of research in this area.
Collapse
Affiliation(s)
- Hugo J Blair
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Lorena Morales
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland.
| | - María R Aburto
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland.
| |
Collapse
|
5
|
Prasad J, Van Steenwinckel J, Gunn AJ, Bennet L, Korzeniewski SJ, Gressens P, Dean JM. Chronic Inflammation Offers Hints About Viable Therapeutic Targets for Preeclampsia and Potentially Related Offspring Sequelae. Int J Mol Sci 2024; 25:12999. [PMID: 39684715 PMCID: PMC11640791 DOI: 10.3390/ijms252312999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
The combination of hypertension with systemic inflammation during pregnancy is a hallmark of preeclampsia, but both processes also convey dynamic information about its antecedents and correlates (e.g., fetal growth restriction) and potentially related offspring sequelae. Causal inferences are further complicated by the increasingly frequent overlap of preeclampsia, fetal growth restriction, and multiple indicators of acute and chronic inflammation, with decreased gestational length and its correlates (e.g., social vulnerability). This complexity prompted our group to summarize information from mechanistic studies, integrated with key clinical evidence, to discuss the possibility that sustained or intermittent systemic inflammation-related phenomena offer hints about viable therapeutic targets, not only for the prevention of preeclampsia, but also the neurobehavioral and other developmental deficits that appear to be overrepresented in surviving offspring. Importantly, we feel that carefully designed hypothesis-driven observational studies are necessary if we are to translate the mechanistic evidence into child health benefits, namely because multiple pregnancy disorders might contribute to heightened risks of neuroinflammation, arrested brain development, or dysconnectivity in survivors who exhibit developmental problems later in life.
Collapse
Affiliation(s)
- Jaya Prasad
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand; (J.P.); (A.J.G.); (L.B.); (J.M.D.)
| | | | - Alistair J. Gunn
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand; (J.P.); (A.J.G.); (L.B.); (J.M.D.)
| | - Laura Bennet
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand; (J.P.); (A.J.G.); (L.B.); (J.M.D.)
| | - Steven J. Korzeniewski
- C.S. Mott Center for Human Growth and Development, Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Pierre Gressens
- Inserm, Neurodiderot, Université de Paris, 75019 Paris, France;
- Centre for the Developing Brain, Division of Imaging Sciences and Department of Biomedical Engineering, King’s College London, King’s Health Partners, St. Thomas’ Hospital, London SE1 7EH, UK
| | - Justin M. Dean
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand; (J.P.); (A.J.G.); (L.B.); (J.M.D.)
| |
Collapse
|
6
|
Tepebasi MY, Selcuk E, Taner R, Tasan S, Asci H, Gunes AB, Sarisahin B, Aydın B. Potential ameliorative effect of Dapagliflozin on systemic inflammation-induced cardiovascular injury via endoplasmic reticulum stress and autophagy pathway. Mol Biol Rep 2024; 51:1080. [PMID: 39432138 DOI: 10.1007/s11033-024-09990-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 10/06/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND Dapagliflozin (DPG) is a sodium-glucose cotransporter-2 inhibitor and is used in the treatment of diabetes. In this study, we aimed to investigate the effect of DPG on cardiotoxicity caused by systemic inflammation via endoplasmic reticulum (ER) stress and autophagy. METHODS AND RESULTS Four groups of thirty-two Wistar Albino rats were created: Control (1 ml oral physiological saline for five days and intraperitoneal saline on the 5th day), LPS (1 ml oral physiological saline for five days and intraperitoneal 5 mg/kg of LPS on the 5th day), LPS + DPG (10 mg/kg of DPG orally for five days and 5 mg/kg of LPS intraperitoneally on the 5th day), and DPG (10 mg/kg of DPG orally for five days and 5 mg/kg of SF intraperitoneally on the 5th day). Histopathological and immunohistochemical analyses were performed on heart and aorta tissues. ER stress and autophagy gene markers in heart tissues were evaluated by RT-qPCR. Oxidative stress in heart tissues and serum cardiac enzymes were analyzed by spectrophotometric method. The heart and aortic tissues of the LPS group showed increased expressions of Tumor Necrosis Factor-α (TNF-α) and Caspase-3 (Cas-3), along with mild hyperemia, slight inflammatory cell infiltrations, and myocardial cell damage. The heart tissues also showed genetically increased expressions of include binding immunoglobulin protein (BiP/ GRP78), protein kinase RNA-like ER Kinase (PERK), inositol-requiring enzyme 1 (IRE-1), activating transcription factors 4 (ATF-4), activating transcription factors 4 (ATF6), C/EBP homologous protein (CHOP), and BECLIN 1. Furthermore, Creatine kinase-MB (CK-MB) and Lactate dehydrogenase (LDH) levels in blood tissue significantly increased, according to biochemical analysis. With DPG therapy, all of these findings were reversed. CONCLUSION In conclusion, DPG protects against the cardiotoxic effect of systemic inflammation with its antioxidant and anti-inflammatory properties by regulating ER stress and autophagy pathways.
Collapse
Affiliation(s)
| | - Esma Selcuk
- Department of Medical Biology, Faculty of Medicine, University of Suleyman Demirel, Isparta, Turkey
| | - Rumeysa Taner
- Department of Bioengineering, Institute of Science, Suleyman Demirel University, Isparta, Turkey
| | - Serife Tasan
- Department of Pathology, Faculty of Veterinary Medicine, University of Mehmet Akif Ersoy, Burdur, Turkey
| | - Halil Asci
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
- Department of Bioengineering, Institute of Science, Suleyman Demirel University, Isparta, Turkey
| | - Ali Baran Gunes
- Undergraduate student, Faculty of Medicine, University of Suleyman Demirel, Isparta, Turkey
| | - Berkehan Sarisahin
- Undergraduate student, Faculty of Medicine, University of Suleyman Demirel, Isparta, Turkey
| | - Bunyamin Aydın
- Kutahya Evliya Celebi Training and Research Hospital, Division of Endocrinology and Metabolism, Kutahya Health Sciences University, Kütahya, Turkey
| |
Collapse
|
7
|
D'Egidio F, Castelli V, d'Angelo M, Ammannito F, Quintiliani M, Cimini A. Brain incoming call from glia during neuroinflammation: Roles of extracellular vesicles. Neurobiol Dis 2024; 201:106663. [PMID: 39251030 DOI: 10.1016/j.nbd.2024.106663] [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: 06/03/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024] Open
Abstract
The functionality of the central nervous system (CNS) relies on the connection, integration, and the exchange of information among neural cells. The crosstalk among glial cells and neurons is pivotal for a series of neural functions, such as development of the nervous system, electric conduction, synaptic transmission, neural circuit establishment, and brain homeostasis. Glial cells are crucial players in the maintenance of brain functionality in physiological and disease conditions. Neuroinflammation is a common pathological process in various brain disorders, such as neurodegenerative diseases, and infections. Glial cells, including astrocytes, microglia, and oligodendrocytes, are the main mediators of neuroinflammation, as they can sense and respond to brain insults by releasing pro-inflammatory or anti-inflammatory factors. Recent evidence indicates that extracellular vesicles (EVs) are pivotal players in the intercellular communication that underlies physiological and pathological processes. In particular, glia-derived EVs play relevant roles in modulating neuroinflammation, either by promoting or inhibiting the activation of glial cells and neurons, or by facilitating the clearance or propagation of pathogenic proteins. The involvement of EVs in neurodegenerative diseases such as Alzheimer's Disease (AD), Parkinson's Disease (PD), Huntington's Disease (HD), and Multiple Sclerosis (MS)- which share hallmarks such as neuroinflammation and oxidative stress to DNA damage, alterations in neurotrophin levels, mitochondrial impairment, and altered protein dynamics- will be dissected, showing how EVs act as pivotal cell-cell mediators of toxic stimuli, thereby propagating degeneration and cell death signaling. Thus, this review focuses on the EVs secreted by microglia, astrocytes, oligodendrocytes and in neuroinflammatory conditions, emphasizing on their effects on neurons and on central nervous system functions, considering both their beneficial and detrimental effects.
Collapse
Affiliation(s)
- Francesco D'Egidio
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy 67100, Via Vetoio - Coppito1, Building "Renato Ricamo"
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy 67100, Via Vetoio - Coppito1, Building "Renato Ricamo"
| | - Michele d'Angelo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy 67100, Via Vetoio - Coppito1, Building "Renato Ricamo".
| | - Fabrizio Ammannito
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy 67100, Via Vetoio - Coppito1, Building "Renato Ricamo"
| | - Massimiliano Quintiliani
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy 67100, Via Vetoio - Coppito1, Building "Renato Ricamo"
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy 67100, Via Vetoio - Coppito1, Building "Renato Ricamo"
| |
Collapse
|
8
|
Waddell J, Lin S, Carter K, Truong T, Hebert M, Ojeda N, Fan LW, Bhatt A, Pang Y. Early Postnatal Neuroinflammation Produces Key Features of Diffuse Brain White Matter Injury in Rats. Brain Sci 2024; 14:976. [PMID: 39451991 PMCID: PMC11505921 DOI: 10.3390/brainsci14100976] [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: 08/08/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Perinatal infection is a major risk factor for diffuse white matter injury (dWMI), which remains the most common form of neurological disability among very preterm infants. The disease primarily targets oligodendrocytes (OL) lineage cells in the white matter but also involves injury and/or dysmaturation of neurons of the gray matter. This study aimed to investigate whether neuroinflammation preferentially affects the cellular compositions of the white matter or gray matter. METHOD Neuroinflammation was initiated by intracerebral administration of lipopolysaccharide (LPS) to rat pups at postnatal (P) day 5, and neurobiological and behavioral outcomes were assessed between P6 and P21. RESULTS LPS challenge rapidly activates microglia and astrocytes, which is associated with the inhibition of OL and neuron differentiation leading to myelination deficits. Specifically, neuroinflammation reduces the immature OLs but not progenitors and causes acute axonal injury (β-amyloid precursor protein immunopositivity) and impaired dendritic maturation (reduced MAP2+ neural fiber density) in the cortical area at P7. Neuroinflammation also reduces the expression of doublecortin in the hippocampus, suggesting compromise in neurogenesis. Utilizing a battery of behavioral assessments, we found that LPS-exposed animals exhibited deficits in sensorimotor, neuromuscular, and cognitive domains. CONCLUSION Our overall results indicate that neuroinflammation alone in the early postnatal period can produce cardinal neuropathological features of dWMI.
Collapse
Affiliation(s)
- John Waddell
- Division of Neonatology, Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.W.); (K.C.); (N.O.); (L.-W.F.); (A.B.)
| | - Shuying Lin
- Department of Physical Therapy, School of Health-Related Professionals, University of Mississippi Medical Center, Jackson, MS 39216, USA;
| | - Kathleen Carter
- Division of Neonatology, Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.W.); (K.C.); (N.O.); (L.-W.F.); (A.B.)
| | - Tina Truong
- Undergraduate Summer Research Program, University of Mississippi Medical Center, Jackson, MS 39216, USA; (T.T.)
| | - May Hebert
- Undergraduate Summer Research Program, University of Mississippi Medical Center, Jackson, MS 39216, USA; (T.T.)
| | - Norma Ojeda
- Division of Neonatology, Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.W.); (K.C.); (N.O.); (L.-W.F.); (A.B.)
| | - Lir-Wan Fan
- Division of Neonatology, Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.W.); (K.C.); (N.O.); (L.-W.F.); (A.B.)
| | - Abhay Bhatt
- Division of Neonatology, Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.W.); (K.C.); (N.O.); (L.-W.F.); (A.B.)
| | - Yi Pang
- Division of Neonatology, Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.W.); (K.C.); (N.O.); (L.-W.F.); (A.B.)
| |
Collapse
|
9
|
Zota I, Chanoumidou K, Gravanis A, Charalampopoulos I. Stimulating myelin restoration with BDNF: a promising therapeutic approach for Alzheimer's disease. Front Cell Neurosci 2024; 18:1422130. [PMID: 39285941 PMCID: PMC11402763 DOI: 10.3389/fncel.2024.1422130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 08/12/2024] [Indexed: 09/19/2024] Open
Abstract
Alzheimer's Disease (AD) is a chronic neurodegenerative disorder constituting the most common form of dementia (60%-70% of cases). Although AD presents majorly a neurodegenerative pathology, recent clinical evidence highlights myelin impairment as a key factor in disease pathogenesis. The lack of preventive or restorative treatment is emphasizing the need to develop novel therapeutic approaches targeting to the causes of the disease. Recent studies in animals and patients have highlighted the loss of myelination of the neuronal axons as an extremely aggravating factor in AD, in addition to the formation of amyloid plaques and neurofibrillary tangles that are to date the main pathological hallmarks of the disease. Myelin breakdown represents an early stage event in AD. However, it is still unclear whether myelin loss is attributed only to exogenous factors like inflammatory processes of the tissue or to impaired oligodendrogenesis as well. Neurotrophic factors are well established protective molecules under many pathological conditions of the neural tissue, contributing also to proper myelination. Due to their inability to be used as drugs, many research efforts are focused on substituting neurotrophic activity with small molecules. Our research team has recently developed novel micromolecular synthetic neurotrophin mimetics (MNTs), selectively acting on neurotrophin receptors, and thus offering a unique opportunity for innovative therapies against neurodegenerative diseases. These small sized, lipophilic molecules address the underlying biological effect of these diseases (neuroprotective action), but also they exert significant neurogenic actions inducing neuronal replacement of the disease areas. One of the significant neurotrophin molecules in the Central Nervous System is Brain-Derived-Neurotrophin-Factor (BDNF). BDNF is a neurotrophin that not only supports neuroprotection and adult neurogenesis, but also mediates pro-myelinating effects in the CNS. BDNF binds with high-affinity on the TrkB neurotrophin receptor and enhances myelination by increasing the density of oligodendrocyte progenitor cells (OPCs) and playing an important role in CNS myelination. Conclusively, in the present review, we discuss the myelin pathophysiology in Alzheimer's Diseases, as well as the role of neurotrophins, and specifically BDNF, in myelin maintenance and restoration, revealing its valuable therapeutic potential against AD.
Collapse
Affiliation(s)
- Ioanna Zota
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas (IMBB-FORTH), Heraklion, Greece
| | - Konstantina Chanoumidou
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas (IMBB-FORTH), Heraklion, Greece
| | - Achille Gravanis
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas (IMBB-FORTH), Heraklion, Greece
| | - Ioannis Charalampopoulos
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas (IMBB-FORTH), Heraklion, Greece
| |
Collapse
|
10
|
Zota I, Chanoumidou K, Charalampopoulos I, Gravanis A. Dynamics of myelin deficits in the 5xFAD mouse model for Alzheimer's disease and the protective role of BDNF. Glia 2024; 72:809-827. [PMID: 38205694 DOI: 10.1002/glia.24505] [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: 06/11/2023] [Revised: 12/08/2023] [Accepted: 12/14/2023] [Indexed: 01/12/2024]
Abstract
Recent findings highlight myelin breakdown as a decisive early event in Alzheimer's Disease (AD) acting as aggravating factor of its progression. However, it is still unclear whether myelin loss is attributed to increased oligodendrocyte vulnerability, reduced repairing capacity or toxic stimuli. In the present study, we sought to clarify the starting point of myelin disruption accompanied with Oligodendrocyte Progenitor Cell (OPC) elimination in the brain of the 5xFAD mouse model of AD at 6 months of age in Dentate Gyrus of the hippocampus in relation to neurotrophin system. Prominent inflammation presence was detected since the age of 6 months playing a key role in myelin disturbance and AD progression. Expression analysis of neurotrophin receptors in OPCs was performed to identify new targets that could increase myelination in health and disease. OPCs in both control and 5xFAD mice express TrkB, TrkC and p75 receptors but not TrkA. Brain-derived neurotrophic factor (BDNF) that binds to TrkB receptor is well-known about its pro-myelination effect, promoting oligodendrocytes proliferation and differentiation, so we focused our investigation on its effects in OPCs under neurodegenerative conditions. Our in vitro results showed that BDNF rescues OPCs from death and promotes their proliferation and differentiation in presence of the toxic Amyloid-β 1-42. Collectively, our results indicate that BDNF possess an additional neuroprotective role through its actions on oligodendrocytic component and its use could be proposed as a drug-based myelin-enhancing strategy, complementary to amyloid and tau centered therapies in AD.
Collapse
Affiliation(s)
- Ioanna Zota
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (IMBB-FORTH), Heraklion, Crete, Greece
| | - Konstantina Chanoumidou
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (IMBB-FORTH), Heraklion, Crete, Greece
| | - Ioannis Charalampopoulos
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (IMBB-FORTH), Heraklion, Crete, Greece
| | - Achille Gravanis
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (IMBB-FORTH), Heraklion, Crete, Greece
| |
Collapse
|
11
|
Van Steenwinckel J, Bokobza C, Laforge M, Shearer IK, Miron VE, Rua R, Matta SM, Hill‐Yardin EL, Fleiss B, Gressens P. Key roles of glial cells in the encephalopathy of prematurity. Glia 2024; 72:475-503. [PMID: 37909340 PMCID: PMC10952406 DOI: 10.1002/glia.24474] [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: 07/19/2023] [Revised: 09/17/2023] [Accepted: 09/19/2023] [Indexed: 11/03/2023]
Abstract
Across the globe, approximately one in 10 babies are born preterm, that is, before 37 weeks of a typical 40 weeks of gestation. Up to 50% of preterm born infants develop brain injury, encephalopathy of prematurity (EoP), that substantially increases their risk for developing lifelong defects in motor skills and domains of learning, memory, emotional regulation, and cognition. We are still severely limited in our abilities to prevent or predict preterm birth. No longer just the "support cells," we now clearly understand that during development glia are key for building a healthy brain. Glial dysfunction is a hallmark of EoP, notably, microgliosis, astrogliosis, and oligodendrocyte injury. Our knowledge of glial biology during development is exponentially expanding but hasn't developed sufficiently for development of effective neuroregenerative therapies. This review summarizes the current state of knowledge for the roles of glia in infants with EoP and its animal models, and a description of known glial-cell interactions in the context of EoP, such as the roles for border-associated macrophages. The field of perinatal medicine is relatively small but has worked passionately to improve our understanding of the etiology of EoP coupled with detailed mechanistic studies of pre-clinical and human cohorts. A primary finding from this review is that expanding our collaborations with computational biologists, working together to understand the complexity of glial subtypes, glial maturation, and the impacts of EoP in the short and long term will be key to the design of therapies that improve outcomes.
Collapse
Affiliation(s)
| | - Cindy Bokobza
- NeuroDiderot, INSERMUniversité Paris CitéParisFrance
| | | | - Isabelle K. Shearer
- School of Health and Biomedical SciencesSTEM College, RMIT UniversityBundooraVictoriaAustralia
| | - Veronique E. Miron
- Barlo Multiple Sclerosis CentreSt. Michael's HospitalTorontoOntarioCanada
- Department of ImmunologyUniversity of TorontoTorontoOntarioCanada
- College of Medicine and Veterinary MedicineThe Dementia Research Institute at The University of EdinburghEdinburghUK
| | - Rejane Rua
- CNRS, INSERM, Centre d'Immunologie de Marseille‐Luminy (CIML), Turing Centre for Living SystemsAix‐Marseille UniversityMarseilleFrance
| | - Samantha M. Matta
- School of Health and Biomedical SciencesSTEM College, RMIT UniversityBundooraVictoriaAustralia
| | - Elisa L. Hill‐Yardin
- School of Health and Biomedical SciencesSTEM College, RMIT UniversityBundooraVictoriaAustralia
| | - Bobbi Fleiss
- NeuroDiderot, INSERMUniversité Paris CitéParisFrance
- School of Health and Biomedical SciencesSTEM College, RMIT UniversityBundooraVictoriaAustralia
| | | |
Collapse
|
12
|
Sun J, Wang W, Ma Q, Pan X, Zhai H, Wang J, Han Y, Li Y, Wang Y. Necrostatin-1s Suppresses RIPK1-driven Necroptosis and Inflammation in Periventricular Leukomalacia Neonatal Mice. Neurochem Res 2024; 49:129-141. [PMID: 37642893 DOI: 10.1007/s11064-023-04013-8] [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: 06/06/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/31/2023]
Abstract
Periventricular leukomalacia (PVL), a predominant form of brain injury in preterm survivors, is characterized by hypomyelination and microgliosis and is also the major cause of long-term neurobehavioral abnormalities in premature infants. Receptor-interacting protein kinase 1 (RIPK1) plays a pivotal role in mediating cell death and inflammatory signaling cascade. However, very little is known about the potential effect of RIPK1 in PVL and the underlying mechanism. Herein, we found that the expression level of RIPK1 was drastically increased in the brain of PVL neonatal mice models, and treatment of PVL neonatal mice with Necrostatin-1s (Nec-1s), an inhibitor of RIPK1, greatly ameliorated cerebral ischemic injury, exhibiting an increase of body weights, reduction of cerebral infarct size, neuronal loss, and occurrence of necrosis-like cells, and significantly improved the long-term abnormal neurobehaviors of PVL. In addition, Nec-1s significantly suppressed hypomyelination and promoted the differentiation of oligodendrocyte precursor cells (OPCs), as demonstrated by the increased expression levels of MBP and Olig2, the decreased expression level of GPR17, a significant increase in the number of CC-1-positive cells, and suppression of myelin ultrastructure impairment. Moreover, the mechanism of neuroprotective effects of Nec-1s against PVL is closely associated with its suppression of the RIPK1-mediated necrosis signaling molecules, RIPK1, PIPK3, and MLKL. More importantly, inhibition of RIPK1 could reduce microglial inflammatory injury by triggering the M1 to M2 microglial phenotype, appreciably decreasing the levels of M1 marker CD86 and increasing the levels of M2 markers Arg1 or CD206 in PVL mice. Taken together, inhibition of RIPK1 markedly ameliorates the brain injury and long-term neurobehavioral abnormalities of PVL mice through the reduction of neural cell necroptosis and reversing neuroinflammation.
Collapse
Affiliation(s)
- Jinping Sun
- School of Basic Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, P.R. China
- Department of Pathology, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, P.R. China
| | - Wei Wang
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, P.R. China
| | - Quanrui Ma
- School of Basic Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, P.R. China
| | - Xiaoli Pan
- Department of Pathology, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, P.R. China
| | - Hualiang Zhai
- Department of Pathology, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, P.R. China
| | - Junyan Wang
- School of Basic Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, P.R. China
| | - Yong Han
- School of Basic Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, P.R. China
| | - Yunhong Li
- School of Basic Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, P.R. China.
| | - Yin Wang
- School of Basic Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, P.R. China.
| |
Collapse
|
13
|
Sullivan-Toole H, Jobson KR, Hoffman LJ, Stewart LC, Olson IR, Olino TM. Adolescents at risk for depression show increased white matter microstructure with age across diffuse areas of the brain. Dev Cogn Neurosci 2023; 64:101307. [PMID: 37813039 PMCID: PMC10570597 DOI: 10.1016/j.dcn.2023.101307] [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: 06/03/2023] [Revised: 08/22/2023] [Accepted: 09/23/2023] [Indexed: 10/11/2023] Open
Abstract
Maternal history of depression is a strong predictor of depression in offspring and linked to structural and functional alterations in the developing brain. However, very little work has examined differences in white matter in adolescents at familial risk for depression. In a sample aged 9-14 (n = 117), we used tract-based spatial statistics (TBSS) to examine differences in white matter microstructure between adolescents with (n = 42) and without (n = 75) maternal history of depression. Microstructure was indexed using fractional anisotropy (FA). Threshold-free cluster enhancement was applied and cluster maps were thresholded at whole-brain family-wise error < .05. There was no significant main effect of risk status on FA. However, there was a significant interaction between risk status and age, such that large and diffuse portions of the white matter skeleton showed relatively increased FA with age for youth with a maternal history of depression compared to those without. Most tracts identified by the interaction were robust to controlling for sex, youth internalizing, in-scanner motion, neighborhood SES, and intra-cranial volume, evidence that maternal depression is a unique predictor of white matter alterations in youth. Widespread increases in FA with age may correspond to a global pattern of accelerated brain maturation in youth at risk for depression.
Collapse
Affiliation(s)
| | - Katie R Jobson
- Department of Psychology and Neuroscience, Temple University, USA
| | - Linda J Hoffman
- Department of Psychology and Neuroscience, Temple University, USA
| | | | - Ingrid R Olson
- Department of Psychology and Neuroscience, Temple University, USA
| | - Thomas M Olino
- Department of Psychology and Neuroscience, Temple University, USA
| |
Collapse
|
14
|
Sawada K, Kamiya S, Kobayashi T. Neonatal Exposure to Lipopolysaccharide Promotes Neurogenesis of Subventricular Zone Progenitors in the Developing Neocortex of Ferrets. Int J Mol Sci 2023; 24:14962. [PMID: 37834410 PMCID: PMC10573966 DOI: 10.3390/ijms241914962] [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: 09/06/2023] [Revised: 09/29/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023] Open
Abstract
Lipopolysaccharide (LPS) is a natural agonist of toll-like receptor 4 that serves a role in innate immunity. The current study evaluated the LPS-mediated regulation of neurogenesis in the subventricular zone (SVZ) progenitors, that is, the basal radial glia and intermediate progenitors (IPs), in ferrets. Ferret pups were subcutaneously injected with LPS (500 μg/g of body weight) on postnatal days (PDs) 6 and 7. Furthermore, 5-ethynyl-2'-deoxyuridine (EdU) and 5-bromo-2'-deoxyuridine (BrdU) were administered on PDs 5 and 7, respectively, to label the post-proliferative and proliferating cells in the inner SVZ (iSVZ) and outer SVZ (oSVZ). A significantly higher density of BrdU single-labeled proliferating cells was observed in the iSVZ of LPS-exposed ferrets than in controls but not in post-proliferative EdU single-labeled and EdU/BrdU double-labeled self-renewing cells. BrdU single-labeled cells exhibited a lower proportion of Tbr2 immunostaining in LPS-exposed ferrets (22.2%) than in controls (42.6%) and a higher proportion of Ctip2 immunostaining in LPS-exposed ferrets (22.2%) than in controls (8.6%). The present findings revealed that LPS modified the neurogenesis of SVZ progenitors. Neonatal LPS exposure facilitates the proliferation of SVZ progenitors, followed by the differentiation of Tbr2-expressing IPs into Ctip2-expressing immature neurons.
Collapse
Affiliation(s)
- Kazuhiko Sawada
- Department of Nutrition, Faculty of Medical and Health Sciences, Tsukuba International University, Tsuchiura 300-0051, Japan
| | - Shiori Kamiya
- Department of Regulation Biology, Faculty of Science, Saitama University, Saitama 338-8570, Japan; (S.K.); (T.K.)
| | - Tetsuya Kobayashi
- Department of Regulation Biology, Faculty of Science, Saitama University, Saitama 338-8570, Japan; (S.K.); (T.K.)
| |
Collapse
|
15
|
Millán Solano MV, Salinas Lara C, Sánchez-Garibay C, Soto-Rojas LO, Escobedo-Ávila I, Tena-Suck ML, Ortíz-Butrón R, Choreño-Parra JA, Romero-López JP, Meléndez Camargo ME. Effect of Systemic Inflammation in the CNS: A Silent History of Neuronal Damage. Int J Mol Sci 2023; 24:11902. [PMID: 37569277 PMCID: PMC10419139 DOI: 10.3390/ijms241511902] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/21/2023] [Accepted: 06/24/2023] [Indexed: 08/13/2023] Open
Abstract
Central nervous system (CNS) infections including meningitis and encephalitis, resulting from the blood-borne spread of specific microorganisms, provoke nervous tissue damage due to the inflammatory process. Moreover, different pathologies such as sepsis can generate systemic inflammation. Bacterial lipopolysaccharide (LPS) induces the release of inflammatory mediators and damage molecules, which are then released into the bloodstream and can interact with structures such as the CNS, thus modifying the blood-brain barrier's (BBB´s) and blood-cerebrospinal fluid barrier´s (BCSFB´s) function and inducing aseptic neuroinflammation. During neuroinflammation, the participation of glial cells (astrocytes, microglia, and oligodendrocytes) plays an important role. They release cytokines, chemokines, reactive oxygen species, nitrogen species, peptides, and even excitatory amino acids that lead to neuronal damage. The neurons undergo morphological and functional changes that could initiate functional alterations to neurodegenerative processes. The present work aims to explain these processes and the pathophysiological interactions involved in CNS damage in the absence of microbes or inflammatory cells.
Collapse
Affiliation(s)
- Mara Verónica Millán Solano
- Red MEDICI, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.V.M.S.); (C.S.-G.); (L.O.S.-R.); (I.E.-Á.); (J.P.R.-L.)
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cos’ıo Villegas, Mexico City 14080, Mexico;
| | - Citlaltepetl Salinas Lara
- Red MEDICI, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.V.M.S.); (C.S.-G.); (L.O.S.-R.); (I.E.-Á.); (J.P.R.-L.)
- Departamento de Neuropatología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, Mexico City 14269, Mexico;
| | - Carlos Sánchez-Garibay
- Red MEDICI, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.V.M.S.); (C.S.-G.); (L.O.S.-R.); (I.E.-Á.); (J.P.R.-L.)
- Departamento de Neuropatología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, Mexico City 14269, Mexico;
| | - Luis O. Soto-Rojas
- Red MEDICI, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.V.M.S.); (C.S.-G.); (L.O.S.-R.); (I.E.-Á.); (J.P.R.-L.)
- Laboratorio de Patogénesis Molecular, Laboratorio 4, Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
| | - Itzel Escobedo-Ávila
- Red MEDICI, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.V.M.S.); (C.S.-G.); (L.O.S.-R.); (I.E.-Á.); (J.P.R.-L.)
- Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autonoma de Mexico, Mexico City 04510, Mexico
| | - Martha Lilia Tena-Suck
- Departamento de Neuropatología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, Mexico City 14269, Mexico;
| | - Rocío Ortíz-Butrón
- Laboratorio de Neurobiología, Departamento de Fisiología de ENCB, Instituto Politécnico Nacional, Mexico City 07738, Mexico;
| | - José Alberto Choreño-Parra
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cos’ıo Villegas, Mexico City 14080, Mexico;
| | - José Pablo Romero-López
- Red MEDICI, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.V.M.S.); (C.S.-G.); (L.O.S.-R.); (I.E.-Á.); (J.P.R.-L.)
- Laboratorio de Patogénesis Molecular, Laboratorio 4, Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
| | - María Estela Meléndez Camargo
- Laboratorio de Farmacología, Departamento de Farmacia, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Av. Wilfrido Massieu Esq. Manuel Luis Stampa S/N, U.P. Adolfo López Mateos, Mexico City 07738, Mexico;
| |
Collapse
|
16
|
Murray CJ, Vecchiarelli HA, Tremblay MÈ. Enhancing axonal myelination in seniors: A review exploring the potential impact cannabis has on myelination in the aged brain. Front Aging Neurosci 2023; 15:1119552. [PMID: 37032821 PMCID: PMC10073480 DOI: 10.3389/fnagi.2023.1119552] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/22/2023] [Indexed: 04/11/2023] Open
Abstract
Consumption of cannabis is on the rise as public opinion trends toward acceptance and its consequent legalization. Specifically, the senior population is one of the demographics increasing their use of cannabis the fastest, but research aimed at understanding cannabis' impact on the aged brain is still scarce. Aging is characterized by many brain changes that slowly alter cognitive ability. One process that is greatly impacted during aging is axonal myelination. The slow degradation and loss of myelin (i.e., demyelination) in the brain with age has been shown to associate with cognitive decline and, furthermore, is a common characteristic of numerous neurological diseases experienced in aging. It is currently not known what causes this age-dependent degradation, but it is likely due to numerous confounding factors (i.e., heightened inflammation, reduced blood flow, cellular senescence) that impact the many cells responsible for maintaining overall homeostasis and myelin integrity. Importantly, animal studies using non-human primates and rodents have also revealed demyelination with age, providing a reliable model for researchers to try and understand the cellular mechanisms at play. In rodents, cannabis was recently shown to modulate the myelination process. Furthermore, studies looking at the direct modulatory impact cannabis has on microglia, astrocytes and oligodendrocyte lineage cells hint at potential mechanisms to prevent some of the more damaging activities performed by these cells that contribute to demyelination in aging. However, research focusing on how cannabis impacts myelination in the aged brain is lacking. Therefore, this review will explore the evidence thus far accumulated to show how cannabis impacts myelination and will extrapolate what this knowledge may mean for the aged brain.
Collapse
Affiliation(s)
- Colin J. Murray
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- *Correspondence: Colin J. Murray,
| | | | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Départment de Médicine Moléculaire, Université Laval, Québec City, QC, Canada
- Axe Neurosciences, Center de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Neurology and Neurosurgery Department, McGill University, Montréal, QC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
- Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada
- Marie-Ève Tremblay,
| |
Collapse
|
17
|
Pierre WC, Zhang E, Londono I, De Leener B, Lesage F, Lodygensky GA. Non-invasive in vivo MRI detects long-term microstructural brain alterations related to learning and memory impairments in a model of inflammation-induced white matter injury. Behav Brain Res 2022; 428:113884. [DOI: 10.1016/j.bbr.2022.113884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 03/18/2022] [Accepted: 04/03/2022] [Indexed: 11/28/2022]
|
18
|
Girolamo F, Errede M, Bizzoca A, Virgintino D, Ribatti D. Central Nervous System Pericytes Contribute to Health and Disease. Cells 2022; 11:1707. [PMID: 35626743 PMCID: PMC9139243 DOI: 10.3390/cells11101707] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 12/11/2022] Open
Abstract
Successful neuroprotection is only possible with contemporary microvascular protection. The prevention of disease-induced vascular modifications that accelerate brain damage remains largely elusive. An improved understanding of pericyte (PC) signalling could provide important insight into the function of the neurovascular unit (NVU), and into the injury-provoked responses that modify cell-cell interactions and crosstalk. Due to sharing the same basement membrane with endothelial cells, PCs have a crucial role in the control of endothelial, astrocyte, and oligodendrocyte precursor functions and hence blood-brain barrier stability. Both cerebrovascular and neurodegenerative diseases impair oxygen delivery and functionally impair the NVU. In this review, the role of PCs in central nervous system health and disease is discussed, considering their origin, multipotency, functions and also dysfunction, focusing on new possible avenues to modulate neuroprotection. Dysfunctional PC signalling could also be considered as a potential biomarker of NVU pathology, allowing us to individualize therapeutic interventions, monitor responses, or predict outcomes.
Collapse
Affiliation(s)
- Francesco Girolamo
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (M.E.); (D.V.); (D.R.)
| | - Mariella Errede
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (M.E.); (D.V.); (D.R.)
| | - Antonella Bizzoca
- Physiology Unit, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy;
| | - Daniela Virgintino
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (M.E.); (D.V.); (D.R.)
| | - Domenico Ribatti
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (M.E.); (D.V.); (D.R.)
| |
Collapse
|
19
|
Pierre WC, Londono I, Quiniou C, Chemtob S, Lodygensky GA. Modulatory effect of IL‐1 inhibition following lipopolysaccharide‐induced neuroinflammation in neonatal microglia and astrocytes. Int J Dev Neurosci 2022; 82:243-260. [DOI: 10.1002/jdn.10179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 02/23/2022] [Accepted: 03/17/2022] [Indexed: 11/09/2022] Open
Affiliation(s)
- Wyston C. Pierre
- Sainte‐Justine Hospital and Research Center, Department of Pediatrics Université de Montréal Montréal, Québec Canada
- Department of Pharmacology and Physiology Université de Montréal Montréal Canada
| | - Irène Londono
- Sainte‐Justine Hospital and Research Center, Department of Pediatrics Université de Montréal Montréal, Québec Canada
| | - Christiane Quiniou
- Sainte‐Justine Hospital and Research Center, Department of Pediatrics Université de Montréal Montréal, Québec Canada
| | - Sylvain Chemtob
- Sainte‐Justine Hospital and Research Center, Department of Pediatrics Université de Montréal Montréal, Québec Canada
- Department of Pharmacology and Physiology Université de Montréal Montréal Canada
- Department of Pharmacology and Therapeutics McGill University Montréal Canada
| | - Gregory A. Lodygensky
- Sainte‐Justine Hospital and Research Center, Department of Pediatrics Université de Montréal Montréal, Québec Canada
- Department of Pharmacology and Physiology Université de Montréal Montréal Canada
| |
Collapse
|
20
|
Spiers JG, Vassileff N, Hill AF. Neuroinflammatory Modulation of Extracellular Vesicle Biogenesis and Cargo Loading. Neuromolecular Med 2022; 24:385-391. [PMID: 35181852 DOI: 10.1007/s12017-022-08704-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 02/04/2022] [Indexed: 12/12/2022]
Abstract
Increasing evidence suggests neuroinflammation is a highly coordinated response involving multiple cell types and utilising several different forms of cellular communication. In addition to the well documented cytokine and chemokine messengers, extracellular vesicles (EVs) have emerged as key regulators of the inflammatory response. EVs act as vectors of intercellular communication, capable of travelling between different cells and tissues to deliver selectively packaged protein, miRNA, and lipids from the parent cell. During neuroinflammation, EVs transmit specific inflammatory mediators, particularly from microglia, to promote inflammatory resolution. This mini-review will highlight the novel neuroinflammatory mechanisms contributing to the biogenesis and selective packaging of EVs.
Collapse
Affiliation(s)
- Jereme G Spiers
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3083, Australia
| | - Natasha Vassileff
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3083, Australia
| | - Andrew F Hill
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3083, Australia.
| |
Collapse
|
21
|
Rodríguez AM, Rodríguez J, Giambartolomei GH. Microglia at the Crossroads of Pathogen-Induced Neuroinflammation. ASN Neuro 2022; 14:17590914221104566. [PMID: 35635133 PMCID: PMC9158411 DOI: 10.1177/17590914221104566] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Microglia are the resident tissue macrophages of the central nervous system (CNS). Recent findings point out that in the steady state the major role of microglia, is to instruct and regulate the correct function of the neuronal networks and different components of the neurovascular unit in the adult CNS, while providing immune surveillance. Paradoxically, during CNS infection immune activation of microglia generates an inflammatory milieu that contributes to the clearance of the pathogen but can, in the process, harm nearby cells of CNS. Most of the knowledge about the harmful effects of activated microglia on CNS has arisen from studies on neurodegenerative diseases. In this review we will focus on the beneficial role and detrimental functions of microglial cells on the neighboring cells of the CNS upon infection.
Collapse
Affiliation(s)
- Ana María Rodríguez
- Instituto de Inmunología, Genética y Metabolismo (INIGEM). CONICET. Facultad de Farmacia y Bioquímica, 28196Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Julia Rodríguez
- Instituto de Inmunología, Genética y Metabolismo (INIGEM). CONICET. Facultad de Farmacia y Bioquímica, 28196Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Guillermo Hernán Giambartolomei
- Instituto de Inmunología, Genética y Metabolismo (INIGEM). CONICET. Facultad de Farmacia y Bioquímica, 28196Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
22
|
Zheng Z, Chen J, Chopp M. Mechanisms of Plasticity Remodeling and Recovery. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00011-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
23
|
Ahn JH, Lee HJ, Lee K, Lim J, Hwang JK, Kim CR, Kim HA, Kim HS, Park HK. Effects of Lipopolysaccharide on Oligodendrocyte Differentiation at Different Developmental Stages: an In Vitro Study. J Korean Med Sci 2021; 36:e332. [PMID: 34931496 PMCID: PMC8688345 DOI: 10.3346/jkms.2021.36.e332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 10/25/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Lipopolysaccharide (LPS) exerts cytotoxic effects on brain cells, especially on those belonging to the oligodendrocyte lineage, in preterm infants. The susceptibility of oligodendrocyte lineage cells to LPS-induced inflammation is dependent on the developmental stage. This study aimed to investigate the effect of LPS on oligodendrocyte lineage cells at different developmental stages in a microglial cell and oligodendrocyte co-culture model. METHODS The primary cultures of oligodendrocytes and microglia cells were prepared from the forebrains of 2-day-old Sprague-Dawley rats. The oligodendrocyte progenitor cells (OPCs) co-cultured with microglial cells were treated with 0 (control), 0.01, 0.1, and 1 µg/mL LPS at the D3 stage to determine the dose of LPS that impairs oligodendrocyte differentiation. The co-culture was treated with 0.01 µg/mL LPS, which was the lowest dose that did not impair oligodendrocyte differentiation, at the developmental stages D1 (early LPS group), D3 (late LPS group), or D1 and D3 (double LPS group). On day 7 of differentiation, oligodendrocytes were subjected to neural glial antigen 2 (NG2) and myelin basic protein (MBP) immunostaining to examine the number of OPCs and mature oligodendrocytes, respectively. RESULTS LPS dose-dependently decreased the proportion of mature oligodendrocytes (MBP+ cells) relative to the total number of cells. The number of MBP+ cells in the early LPS group was significantly lower than that in the late LPS group. Compared with those in the control group, the MBP+ cell numbers were significantly lower and the NG2+ cell numbers were significantly higher in the double LPS group, which exhibited impaired oligodendrocyte lineage cell development, on day 7 of differentiation. CONCLUSION Repetitive LPS stimulation during development significantly inhibited brain cell development by impairing oligodendrocyte differentiation. In contrast, brain cell development was not affected in the late LPS group. These findings suggest that inflammation at the early developmental stage of oligodendrocytes increases the susceptibility of the preterm brain to inflammation-induced injury.
Collapse
Affiliation(s)
- Ja-Hye Ahn
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Korea
| | - Hyun Ju Lee
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Korea
| | - Kyeongmi Lee
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Korea
| | - Jean Lim
- Kangwon National University College of Veterinary Medicine, Chuncheon, Korea
| | - Jae Kyoon Hwang
- Department of Pediatrics, Hanyang University Guri Hospital, Guri, Korea
| | - Chang-Ryul Kim
- Department of Pediatrics, Hanyang University Guri Hospital, Guri, Korea
| | - Hyun A Kim
- Department of Child Psychotherapy, Hanyang University Graduate School of Medicine, Seoul, Korea
| | - Han-Suk Kim
- Department of Pediatrics, Seoul University College of Medicine, Seoul, Korea
| | - Hyun-Kyung Park
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Korea.
| |
Collapse
|
24
|
Ramarao S, Pang Y, Carter K, Bhatt A. Azithromycin Protects Oligodendrocyte Progenitor Cells against Lipopolysaccharide-activated Microglia-induced damage. Dev Neurosci 2021; 44:1-12. [PMID: 34571509 DOI: 10.1159/000519874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 09/21/2021] [Indexed: 11/19/2022] Open
Affiliation(s)
- Sumana Ramarao
- Division of Newborn Medicine, Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Yi Pang
- Division of Newborn Medicine, Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Kathleen Carter
- Division of Newborn Medicine, Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Abhay Bhatt
- Division of Newborn Medicine, Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
25
|
Sams E. Oligodendrocytes in the aging brain. Neuronal Signal 2021; 5:NS20210008. [PMID: 34290887 PMCID: PMC8264650 DOI: 10.1042/ns20210008] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/22/2022] Open
Abstract
More than half of the human brain volume is made up of white matter: regions where axons are coated in myelin, which primarily functions to increase the conduction speed of axon potentials. White matter volume significantly decreases with age, correlating with cognitive decline. Much research in the field of non-pathological brain aging mechanisms has taken a neuron-centric approach, with relatively little attention paid to other neural cells. This review discusses white matter changes, with focus on oligodendrocyte lineage cells and their ability to produce and maintain myelin to support normal brain homoeostasis. Improved understanding of intrinsic cellular changes, general senescence mechanisms, intercellular interactions and alterations in extracellular environment which occur with aging and impact oligodendrocyte cells is paramount. This may lead to strategies to support oligodendrocytes in aging, for example by supporting myelin synthesis, protecting against oxidative stress and promoting the rejuvenation of the intrinsic regenerative potential of progenitor cells. Ultimately, this will enable the protection of white matter integrity thus protecting cognitive function into the later years of life.
Collapse
Affiliation(s)
- Eleanor Catherine Sams
- Blizard Institute, Barts and The London School of Medicine and Dentistry Centre for Neuroscience, Surgery and Trauma, Blizard Institute, 4 Newark Street, Whitechapel E1 2AT, London
| |
Collapse
|
26
|
Kelly SB, Stojanovska V, Zahra VA, Moxham A, Miller SL, Moss TJM, Hooper SB, Nold MF, Nold-Petry CA, Dean JM, Bennet L, Polglase GR, Gunn AJ, Galinsky R. Interleukin-1 blockade attenuates white matter inflammation and oligodendrocyte loss after progressive systemic lipopolysaccharide exposure in near-term fetal sheep. J Neuroinflammation 2021; 18:189. [PMID: 34465372 PMCID: PMC8408978 DOI: 10.1186/s12974-021-02238-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 08/11/2021] [Indexed: 12/14/2022] Open
Abstract
Background Increased systemic and tissue levels of interleukin (IL)-1β are associated with greater risk of impaired neurodevelopment after birth. In this study, we tested the hypothesis that systemic IL-1 receptor antagonist (Ra) administration would attenuate brain inflammation and injury in near-term fetal sheep exposed to lipopolysaccharide (LPS). Methods Chronically instrumented near-term fetal sheep at 0.85 of gestation were randomly assigned to saline infusion (control, n = 9), repeated LPS infusions (0 h = 300 ng, 24 h = 600 ng, 48 h = 1200 ng, n = 8) or repeated LPS plus IL-1Ra infusions (13 mg/kg infused over 4 h) started 1 h after each LPS infusion (n = 9). Sheep were euthanized 4 days after starting infusions for histology. Results LPS infusions increased circulating cytokines and were associated with electroencephalogram (EEG) suppression with transiently reduced mean arterial blood pressure, and increased carotid artery perfusion and fetal heart rate (P < 0.05 vs. control for all). In the periventricular and intragyral white matter, LPS-exposure increased IL-1β immunoreactivity, numbers of caspase 3+ cells and microglia, reduced astrocyte and olig-2+ oligodendrocyte survival but did not change numbers of mature CC1+ oligodendrocytes, myelin expression or numbers of neurons in the cortex and subcortical regions. IL-1Ra infusions reduced circulating cytokines and improved recovery of EEG activity and carotid artery perfusion. Histologically, IL-1Ra reduced microgliosis, IL-1β expression and caspase-3+ cells, and improved olig-2+ oligodendrocyte survival. Conclusion IL-1Ra improved EEG activity and markedly attenuated systemic inflammation, microgliosis and oligodendrocyte loss following LPS exposure in near-term fetal sheep. Further studies examining the long-term effects on brain maturation are now needed. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02238-4.
Collapse
Affiliation(s)
- Sharmony B Kelly
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright street, Melbourne, Victoria, 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia
| | - Vanesa Stojanovska
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright street, Melbourne, Victoria, 3168, Australia
| | - Valerie A Zahra
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright street, Melbourne, Victoria, 3168, Australia
| | - Alison Moxham
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright street, Melbourne, Victoria, 3168, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright street, Melbourne, Victoria, 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia
| | - Timothy J M Moss
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright street, Melbourne, Victoria, 3168, Australia
| | - Stuart B Hooper
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright street, Melbourne, Victoria, 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia
| | - Marcel F Nold
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright street, Melbourne, Victoria, 3168, Australia.,Department of Paediatrics, Monash University, Melbourne, Victoria, Australia.,Monash Newborn, Monash Children's Hospital, Melbourne, Australia
| | - Claudia A Nold-Petry
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright street, Melbourne, Victoria, 3168, Australia.,Department of Paediatrics, Monash University, Melbourne, Victoria, Australia
| | - Justin M Dean
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Graeme R Polglase
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright street, Melbourne, Victoria, 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia
| | - Alistair J Gunn
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Robert Galinsky
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright street, Melbourne, Victoria, 3168, Australia. .,Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
27
|
Delahaye-Duriez A, Dufour A, Bokobza C, Gressens P, Van Steenwinckel J. Targeting Microglial Disturbances to Protect the Brain From Neurodevelopmental Disorders Associated With Prematurity. J Neuropathol Exp Neurol 2021; 80:634-648. [PMID: 34363661 DOI: 10.1093/jnen/nlab049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Microglial activation during critical phases of brain development can result in short- and long-term consequences for neurological and psychiatric health. Several studies in humans and rodents have shown that microglial activation, leading to a transition from the homeostatic state toward a proinflammatory phenotype, has adverse effects on the developing brain and neurodevelopmental disorders. Targeting proinflammatory microglia may be an effective strategy for protecting the brain and attenuating neurodevelopmental disorders induced by inflammation. In this review we focus on the role of inflammation and the activation of immature microglia (pre-microglia) soon after birth in prematurity-associated neurodevelopmental disorders, and the specific features of pre-microglia during development. We also highlight the relevance of immunomodulatory strategies for regulating activated microglia in a rodent model of perinatal brain injury. An original neuroprotective approach involving a nanoparticle-based therapy and targeting microglia, with the aim of improving myelination and protecting the developing brain, is also addressed.
Collapse
Affiliation(s)
- Andrée Delahaye-Duriez
- From the NeuroDiderot, UMR 1141, Inserm, Université de Paris, Paris, France.,UFR SMBH, Université Sorbonne Paris Nord, Bobigny, France.,Assistance Publique des Hôpitaux de Paris, Hôpital Jean Verdier, Service d'Histologie-Embryologie-Cytogénétique, Bondy, France
| | - Adrien Dufour
- From the NeuroDiderot, UMR 1141, Inserm, Université de Paris, Paris, France
| | - Cindy Bokobza
- From the NeuroDiderot, UMR 1141, Inserm, Université de Paris, Paris, France
| | - Pierre Gressens
- From the NeuroDiderot, UMR 1141, Inserm, Université de Paris, Paris, France
| | | |
Collapse
|
28
|
Yeh JH, Wang KC, Kaizaki A, Lee JW, Wei HC, Tucci MA, Ojeda NB, Fan LW, Tien LT. Pioglitazone Ameliorates Lipopolysaccharide-Induced Behavioral Impairment, Brain Inflammation, White Matter Injury and Mitochondrial Dysfunction in Neonatal Rats. Int J Mol Sci 2021; 22:6306. [PMID: 34208374 PMCID: PMC8231261 DOI: 10.3390/ijms22126306] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/29/2021] [Accepted: 06/05/2021] [Indexed: 01/04/2023] Open
Abstract
Previous studies have demonstrated that pioglitazone, a peroxisome proliferator-activated receptor gamma (PPARγ) agonist, inhibits ischemia-induced brain injury. The present study was conducted to examine whether pioglitazone can reduce impairment of behavioral deficits mediated by inflammatory-induced brain white matter injury in neonatal rats. Intraperitoneal (i.p.) injection of lipopolysaccharide (LPS, 2 mg/kg) was administered to Sprague-Dawley rat pups on postnatal day 5 (P5), and i.p. administration of pioglitazone (20 mg/kg) or vehicle was performed 5 min after LPS injection. Sensorimotor behavioral tests were performed 24 h after LPS exposure, and changes in biochemistry of the brain was examined after these tests. The results show that systemic LPS exposure resulted in impaired sensorimotor behavioral performance, reduction of oligodendrocytes and mitochondrial activity, and increases in lipid peroxidation and brain inflammation, as indicated by the increment of interleukin-1β (IL-1β) levels and number of activated microglia in the neonatal rat brain. Pioglitazone treatment significantly improved LPS-induced neurobehavioral and physiological disturbances including the loss of body weight, hypothermia, righting reflex, wire-hanging maneuver, negative geotaxis, and hind-limb suspension in neonatal rats. The neuroprotective effect of pioglitazone against the loss of oligodendrocytes and mitochondrial activity was associated with attenuation of LPS-induced increment of thiobarbituric acid reactive substances (TBARS) content, IL-1β levels and number of activated microglia in neonatal rats. Our results show that pioglitazone prevents neurobehavioral disturbances induced by systemic LPS exposure in neonatal rats, and its neuroprotective effects are associated with its impact on microglial activation, IL-1β induction, lipid peroxidation, oligodendrocyte production and mitochondrial activity.
Collapse
Affiliation(s)
- Jiann-Horng Yeh
- Department of Neurobiology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 111, Taiwan;
- School of Medicine, Fu Jen Catholic University, Xinzhuang Dist., New Taipei City 24205, Taiwan; (K.-C.W.); (H.-C.W.)
| | - Kuo-Ching Wang
- School of Medicine, Fu Jen Catholic University, Xinzhuang Dist., New Taipei City 24205, Taiwan; (K.-C.W.); (H.-C.W.)
- Department of Anesthesiology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 111, Taiwan
| | - Asuka Kaizaki
- Department of Pharmacology, Toxicology and Therapeutics, Division of Toxicology, School of Pharmacy, Showa University, Shingawa-ku, Tokyo 142-8555, Japan;
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.W.L.); (N.B.O.); (L.-W.F.)
| | - Jonathan W. Lee
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.W.L.); (N.B.O.); (L.-W.F.)
| | - Han-Chi Wei
- School of Medicine, Fu Jen Catholic University, Xinzhuang Dist., New Taipei City 24205, Taiwan; (K.-C.W.); (H.-C.W.)
| | - Michelle A. Tucci
- Department of Anesthesiology, University of Mississippi Medical Center, Jackson, MS 39216, USA;
| | - Norma B. Ojeda
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.W.L.); (N.B.O.); (L.-W.F.)
| | - Lir-Wan Fan
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.W.L.); (N.B.O.); (L.-W.F.)
| | - Lu-Tai Tien
- School of Medicine, Fu Jen Catholic University, Xinzhuang Dist., New Taipei City 24205, Taiwan; (K.-C.W.); (H.-C.W.)
| |
Collapse
|
29
|
Kaul D, Schwab SG, Mechawar N, Matosin N. How stress physically re-shapes the brain: Impact on brain cell shapes, numbers and connections in psychiatric disorders. Neurosci Biobehav Rev 2021; 124:193-215. [PMID: 33556389 DOI: 10.1016/j.neubiorev.2021.01.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/20/2021] [Accepted: 01/31/2021] [Indexed: 12/16/2022]
Abstract
Severe stress is among the most robust risk factors for the development of psychiatric disorders. Imaging studies indicate that life stress is integral to shaping the human brain, especially regions involved in processing the stress response. Although this is likely underpinned by changes to the cytoarchitecture of cellular networks in the brain, we are yet to clearly understand how these define a role for stress in human psychopathology. In this review, we consolidate evidence of macro-structural morphometric changes and the cellular mechanisms that likely underlie them. Focusing on stress-sensitive regions of the brain, we illustrate how stress throughout life may lead to persistent remodelling of the both neurons and glia in cellular networks and how these may lead to psychopathology. We support that greater translation of cellular alterations to human cohorts will support parsing the psychological sequalae of severe stress and improve our understanding of how stress shapes the human brain. This will remain a critical step for improving treatment interventions and prevention outcomes.
Collapse
Affiliation(s)
- Dominic Kaul
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong 2522, Australia; Molecular Horizons, School of Chemistry and Molecular Biosciences, University of Wollongong, Northfields Ave, Wollongong 2522, Australia
| | - Sibylle G Schwab
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong 2522, Australia; Molecular Horizons, School of Chemistry and Molecular Biosciences, University of Wollongong, Northfields Ave, Wollongong 2522, Australia
| | - Naguib Mechawar
- Douglas Mental Health University Institute, 6875 LaSalle blvd, Verdun, Qc, H4H 1R3, Canada
| | - Natalie Matosin
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong 2522, Australia; Molecular Horizons, School of Chemistry and Molecular Biosciences, University of Wollongong, Northfields Ave, Wollongong 2522, Australia; Max Planck Institute of Psychiatry, Kraepelinstrasse 2-10, 80804 Munich, Germany.
| |
Collapse
|
30
|
Dhanalakshmi C, Janakiraman U, Moutal A, Fukunaga K, Khanna R, Nelson MA. Evaluation of the effects of the T-type calcium channel enhancer SAK3 in a rat model of TAF1 deficiency. Neurobiol Dis 2021; 149:105224. [PMID: 33359140 PMCID: PMC8230513 DOI: 10.1016/j.nbd.2020.105224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 12/03/2020] [Accepted: 12/16/2020] [Indexed: 11/18/2022] Open
Abstract
The TATA-box binding protein associated factor 1 (TAF1) is part of the TFIID complex that plays a key role during the initiation of transcription. Variants of TAF1 are associated with neurodevelopmental disorders. Previously, we found that CRISPR/Cas9 based editing of the TAF1 gene disrupts the morphology of the cerebral cortex and blunts the expression as well as the function of the CaV3.1 (T-type) voltage gated calcium channel. Here, we tested the efficacy of SAK3 (ethyl 8'-methyl-2', 4-dioxo-2-(piperidin-1-yl)-2'H-spiro [cyclopentane-1, 3'-imidazo [1, 2-a] pyridine]-2-ene-3-carboxylate), a T-type calcium channel enhancer, in an animal model of TAF1 intellectual disability (ID) syndrome. At post-natal day 3, rat pups were subjected to intracerebroventricular (ICV) injection of either gRNA-control or gRNA-TAF1 CRISPR/Cas9 viruses. At post-natal day 21, the rat pups were given SAK3 (0.25 mg/kg, p.o.) or vehicle for 14 days (i.e. till post-natal day 35) and then subjected to behavioral, morphological, and molecular studies. Oral administration of SAK3 (0.25 mg/kg, p.o.) significantly rescued locomotion abnormalities associated with TAF1 gene editing. SAK3 treatment prevented the loss of cortical neurons and GFAP-positive astrocytes observed after TAF1 gene editing. In addition, SAK3 protected cells from apoptosis. SAK3 also restored the Brain-derived neurotrophic factor/protein kinase B/Glycogen Synthase Kinase 3 Beta (BDNF/AKT/GSK3β) signaling axis in TAF1 edited animals. Finally, SAK3 normalized the levels of three GSK3β substrates - CaV3.1, FOXP2, and CRMP2. We conclude that the T-type calcium channel enhancer SAK3 is beneficial against the deleterious effects of TAF1 gene-editing, in part, by stimulating the BDNF/AKT/GSK3β signaling pathway.
Collapse
Affiliation(s)
- Chinnasamy Dhanalakshmi
- Department of Pathology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA
| | - Udaiyappan Janakiraman
- Department of Pathology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA
| | - Aubin Moutal
- Department of Pharmacology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA; The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, AZ, United States; The BIO5 Institute, University of Arizona, United States
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Rajesh Khanna
- Department of Pharmacology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA; The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, AZ, United States; The BIO5 Institute, University of Arizona, United States
| | - Mark A Nelson
- Department of Pathology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA.
| |
Collapse
|
31
|
Abe Y, Ochiai D, Sato Y, Otani T, Fukutake M, Ikenoue S, Kasuga Y, Tanaka M. Amniotic fluid stem cells as a novel strategy for the treatment of fetal and neonatal neurological diseases. Placenta 2021; 104:247-252. [PMID: 33461069 DOI: 10.1016/j.placenta.2021.01.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/26/2020] [Accepted: 01/08/2021] [Indexed: 01/26/2023]
Abstract
Even in the context of modern medicine, infants with fetal and neonatal neurological diseases such as cerebral palsy and myelomeningocele suffer serious long-lasting impairment due to the irreversible neuronal damage. The promotion of neurologically intact survival in patients with perinatal intractable neurological diseases requires the development of novel strategies. One promising strategy involves the use of human amniotic fluid stem cells (hAFSCs), which have attracted much attention in recent years and are known to exert anti-inflammatory and neuroprotective effects. In recent years, the therapeutic effects of hAFSCs on fetal-neonatal neurological diseases have become evident as per intense research efforts by our group and others. Specifically, hAFSCs administered into the nasal cavity migrated to the brain and controlled local inflammation in a rodent model of neonatal hypoxic-ischemic encephalopathy. In contrast, hAFSCs administered intraperitoneally did not migrate to the brain; they rather formed spheroids in the abdominal cavity, resulting in the suppression of systemic inflammation (including in the brain) via the secretion of anti-inflammatory cytokines in concert with peritoneal macrophages in a rodent model of periventricular leukomalacia. Moreover, studies in a rat model of myelomeningocele suggested that hAFSCs administered in utero secreted hepatocyte growth factor and protected the exposed spinal cord during pregnancy. Importantly, autologous hAFSCs, whose use for fetal-neonatal treatment does not raise ethical issues, can be collected during pregnancy and prepared in sufficient numbers for therapeutic use. This article outlines the results of preclinical research on fetal stem cell therapy, mainly involving hAFSCs, in the context of perinatal neurological diseases.
Collapse
Affiliation(s)
- Yushi Abe
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Daigo Ochiai
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan.
| | - Yu Sato
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Toshimitsu Otani
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Marie Fukutake
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Satoru Ikenoue
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Yoshifumi Kasuga
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Mamoru Tanaka
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
32
|
Holloway RK, Ireland G, Sullivan G, Becher JC, Smith C, Boardman JP, Gressens P, Miron VE. Microglial inflammasome activation drives developmental white matter injury. Glia 2021; 69:1268-1280. [PMID: 33417729 PMCID: PMC8607465 DOI: 10.1002/glia.23963] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 12/15/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022]
Abstract
Injury to the developing brain during the perinatal period often causes hypomyelination, leading to clinical deficits for which there is an unmet therapeutic need. Dysregulation of inflammation and microglia have been implicated, yet the molecular mechanisms linking these to hypomyelination are unclear. Using human infant cerebrospinal fluid (CSF) and postmortem tissue, we found that microglial activation of the pro-inflammatory molecular complex the NLRP3 inflammasome is associated with pathology. By developing a novel mouse brain explant model of microglial inflammasome activation, we demonstrate that blocking the inflammasome rescues myelination. In human and mouse, we discovered a link between the inflammasome product IL1β and increased levels of follistatin, an endogenous inhibitor of activin-A. Follistatin treatment was sufficient to reduce myelination, whereas myelination was rescued in injured explants upon follistatin neutralization or supplementation with exogenous activin-A. Our data reveal that inflammasome activation in microglia drives hypomyelination and identifies novel therapeutic strategies to reinstate myelination following developmental injury.
Collapse
Affiliation(s)
- Rebecca K Holloway
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Graeme Ireland
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Gemma Sullivan
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Julie-Clare Becher
- Simpson Centre for Reproductive Health, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Colin Smith
- Centre for Clinical Brain Sciences, Centre for Comparative Pathology, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - James P Boardman
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Pierre Gressens
- Department of Perinatal Imaging and Health, Rayne's Institute, King's College London, London, UK.,PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Veronique E Miron
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
33
|
From "Leaky Gut" to Impaired Glia-Neuron Communication in Depression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1305:129-155. [PMID: 33834399 DOI: 10.1007/978-981-33-6044-0_9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In the last three decades, the robust scientific data emerged, demonstrating that the immune-inflammatory response is a fundamental component of the pathophysiology of major depressive disorder (MDD). Psychological stress and various inflammatory comorbidities contribute to such immune activation. Still, this is not uncommon that patients with depression do not have defined inflammatory comorbidities, and alternative mechanisms of immune activation need to take place. The gastrointestinal (GI) tract, along with gut-associated lymphoid tissue (GALT), constitutes the largest lymphatic organ in the human body and forms the biggest surface of contact with the external environment. It is also the most significant source of bacterial and food-derived antigenic material. There is a broad range of reciprocal interactions between the GI tract, intestinal microbiota, increased intestinal permeability, activation of immune-inflammatory response, and the CNS that has crucial implications in brain function and mental health. This intercommunication takes place within the microbiota-gut-immune-glia (MGIG) axis, and glial cells are the main orchestrator of this communication. A broad range of factors, including psychological stress, inflammation, dysbiosis, may compromise the permeability of this barrier. This leads to excessive bacterial translocation and the excessive influx of food-derived antigenic material that contributes to activation of the immune-inflammatory response and depressive psychopathology. This chapter summarizes the role of increased intestinal permeability in MDD and mechanisms of how the "leaky gut" may contribute to immune-inflammatory response in this disorder.
Collapse
|
34
|
O’Donovan A, Bahorik A, Sidney S, Launer LJ, Yaffe K. Relationships of inflammation trajectories with white matter volume and integrity in midlife. Brain Behav Immun 2021; 91:81-88. [PMID: 32966872 PMCID: PMC7749816 DOI: 10.1016/j.bbi.2020.09.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/01/2020] [Accepted: 09/04/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Elevated inflammation is associated with worse late-life cognitive functioning and brain health. Our goal was to examine the relationship between inflammation trajectories and white matter integrity in midlife. METHODS Participants were 508 adults from the Coronary Artery Risk Development in Young Adults Study (CARDIA; 51% female). Latent class analysis was used to identify inflammation trajectories based on repeated measures of the inflammatory marker C-reactive protein (CRP) over the 18 years before brain magnetic resonance imaging (MRI). Outcomes were brain MRI measures of total and region-specific white matter volume and integrity at a mean age of 50.6 ± 3.4 years. Linear regression was used to examine if inflammation trajectories were associated with brain MRI outcomes, adjusting for potential confounds in all models and for disease and health behaviors in follow-up models. RESULTS Lower-stable (38%), moderate-increasing (7%), and consistently-higher (54%), trajectories emerged. Compared to the lower-stable group, the moderate-increasing group showed lower white matter volume (β = -0.18, 95% CI -0.29, -0.06) and worse white matter integrity as indexed by lower fractional anisotropy (FA; β = -0.37, 95% CI -0.70, -0.04) and higher mean diffusivity (β = 0.44, 95% CI 0.11, 0.78) in the whole brain. The consistently-higher group showed lower whole-brain FA (β = -0.20, -0.38, -0.03). In exploratory analyses, the moderate-increasing group showed lower white matter volume, lower FA and higher MD in the frontal, temporal, and parietal lobes compared to the lower-stable group. The consistently-higher group showed lower white matter volume in the parietal lobe and lower FA in the frontal, temporal, and parietal lobes, but similar MD, compared to the lower-stable group. Findings for the moderate-increasing, but not the consistently-higher, group were robust to adjustment for disease and lifestyle factors. CONCLUSION Increasing or high inflammation trajectories from early to mid adulthood are associated with worse brain health, as indexed by lower white matter volume and/or worse white matter integrity.
Collapse
Affiliation(s)
- Aoife O’Donovan
- University of California, San Francisco, CA,Corresponding Author: Aoife O’Donovan, PhD, Department of Psychiatry, University of California, San Francisco, 4150 Clement Street, San Francisco, CA 94121, Phone: +01 (415) 221-4810 X24959,
| | | | | | | | | |
Collapse
|
35
|
Goulding DS, Vogel RC, Gensel JC, Morganti JM, Stromberg AJ, Miller BA. Acute brain inflammation, white matter oxidative stress, and myelin deficiency in a model of neonatal intraventricular hemorrhage. J Neurosurg Pediatr 2020; 26:613-623. [PMID: 32858507 PMCID: PMC10193502 DOI: 10.3171/2020.5.peds20124] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/18/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Neonatal intraventricular hemorrhage (IVH) leads to posthemorrhagic hydrocephalus (PHH), brain injury, and long-term disability. Current therapy for IVH is based on treating PHH but does not address the underlying brain injury. In order to develop pharmacological treatment for IVH, there must be a better understanding of the underlying pathology of this disease. This study was designed to determine the time course of the acute inflammation and oxidative stress that may underlie the progressive pathology of IVH. The authors sought to understand the temporal relationships among inflammation, oxidative stress, and white matter pathology in a rat model of IVH. METHODS A rat model of IVH consisting of hemoglobin injection into the lateral ventricle was used. Tissue was analyzed via biochemical and histological methods to map the spatiotemporal distribution of innate immune activation and oxidative stress. White matter was quantified using both immunohistochemistry and Western blot for myelin basic protein (MBP) in the corpus callosum. RESULTS IVH led to acute induction of inflammatory cytokines, followed by oxidative stress. Oxidative stress was concentrated in white matter, adjacent to the lateral ventricles. Animals with IVH initially gained weight at a lower rate than control animals and had larger ventricles and less MBP than control animals. CONCLUSIONS Experimental IVH induces global inflammation throughout the brain and oxidative stress concentrated in the white matter. Both of these phenomena occur early after IVH. This has implications for human neonates with immature white matter that is exquisitely sensitive to inflammation and oxidative stress. Antiinflammatory or antioxidant therapy for IVH may need to be initiated early in order to protect developing white matter.
Collapse
Affiliation(s)
- Danielle S. Goulding
- Departments of Neurosurgery
- Spinal Cord and Brain Injury Research Center, University of Kentucky; and
| | - R. Caleb Vogel
- Departments of Neurosurgery
- Spinal Cord and Brain Injury Research Center, University of Kentucky; and
| | - John C. Gensel
- Physiology
- Spinal Cord and Brain Injury Research Center, University of Kentucky; and
| | - Josh M. Morganti
- Neuroscience, and
- Spinal Cord and Brain Injury Research Center, University of Kentucky; and
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky
| | | | - Brandon A. Miller
- Departments of Neurosurgery
- Neuroscience, and
- Spinal Cord and Brain Injury Research Center, University of Kentucky; and
| |
Collapse
|
36
|
Janakiraman U, Dhanalakshmi C, Yu J, Moutal A, Boinon L, Fukunaga K, Khanna R, Nelson MA. The investigation of the T-type calcium channel enhancer SAK3 in an animal model of TAF1 intellectual disability syndrome. Neurobiol Dis 2020; 143:105006. [PMID: 32622085 PMCID: PMC7422587 DOI: 10.1016/j.nbd.2020.105006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/17/2020] [Accepted: 06/27/2020] [Indexed: 01/21/2023] Open
Abstract
T-type calcium channels, in the central nervous system, are involved in the pathogenesis of many neurodegenerative diseases, including TAF1 intellectual disability syndrome (TAF1 ID syndrome). Here, we evaluated the efficacy of a novel T-type Ca2+ channel enhancer, SAK3 (ethyl 8'-methyl-2', 4-dioxo-2-(piperidin-1-yl)-2'H-spiro [cyclopentane-1, 3'-imidazo [1, 2-a] pyridine]-2-ene-3-carboxylate) in an animal model of TAF1 ID syndrome. At post-natal day 3, rat pups were subjected to intracerebroventricular (ICV) injection of either gRNA-control or gRNA-TAF1 CRISPR/Cas9 viruses. At post-natal day 21 animals were given SAK3 (0.25 mg/kg, p.o.) or vehicle up to post-natal day 35 (i.e. 14 days). Rats were subjected to behavioral, morphological, electrophysiological, and molecular studies. Oral administration of SAK3 (0.25 mg/kg, p.o.) significantly rescued the behavior abnormalities in beam walking test and open field test caused by TAF1 gene editing. We observed an increase in calbindin-positive Purkinje cells and GFAP-positive astrocytes as well as a decrease in IBA1-positive microglia cells in SAK3-treated animals. In addition, SAK3 protected the Purkinje and granule cells from apoptosis induced by TAF-1 gene editing. SAK3 also restored the excitatory post synaptic current (sEPSCs) in TAF1 edited Purkinje cells. Finally, SAK3 normalized the BDNF/AKT signaling axis in TAF1 edited animals. Altogether, these observations suggest that SAK3 could be a novel therapeutic agent for TAF1 ID syndrome.
Collapse
Affiliation(s)
- Udaiyappan Janakiraman
- Department of Pathology, University of Arizona College of Medicine, College of Pharmacy, Tucson, AZ, USA
| | - Chinnasamy Dhanalakshmi
- Department of Pathology, University of Arizona College of Medicine, College of Pharmacy, Tucson, AZ, USA
| | - Jie Yu
- Department of Pharmacology, University of Arizona College of Medicine, College of Pharmacy, Tucson, AZ, USA; College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310058, China
| | - Aubin Moutal
- Department of Pharmacology, University of Arizona College of Medicine, College of Pharmacy, Tucson, AZ, USA
| | - Lisa Boinon
- Department of Pharmacology, University of Arizona College of Medicine, College of Pharmacy, Tucson, AZ, USA
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Rajesh Khanna
- Department of Pathology, University of Arizona College of Medicine, College of Pharmacy, Tucson, AZ, USA; Department of Pharmacology, University of Arizona College of Medicine, College of Pharmacy, Tucson, AZ, USA; The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, AZ, USA; The BIO5 Institute, University of Arizona, USA
| | - Mark A Nelson
- Department of Pathology, University of Arizona College of Medicine, College of Pharmacy, Tucson, AZ, USA.
| |
Collapse
|
37
|
Rudzki L, Maes M. The Microbiota-Gut-Immune-Glia (MGIG) Axis in Major Depression. Mol Neurobiol 2020; 57:4269-4295. [DOI: 10.1007/s12035-020-01961-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/28/2020] [Indexed: 02/08/2023]
|
38
|
Dietz AG, Goldman SA, Nedergaard M. Glial cells in schizophrenia: a unified hypothesis. Lancet Psychiatry 2020; 7:272-281. [PMID: 31704113 PMCID: PMC7267935 DOI: 10.1016/s2215-0366(19)30302-5] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 07/23/2019] [Accepted: 07/24/2019] [Indexed: 12/11/2022]
Abstract
The cellular neurobiology of schizophrenia remains poorly understood. We discuss neuroimaging studies, pathological findings, and experimental work supporting the idea that glial cells might contribute to the development of schizophrenia. Experimental studies suggest that abnormalities in the differentiation competence of glial progenitor cells lead to failure in the morphological and functional maturation of oligodendrocytes and astrocytes. We propose that immune activation of microglial cells during development, superimposed upon genetic risk factors, could contribute to defective differentiation competence of glial progenitor cells. The resulting hypomyelination and disrupted white matter integrity might contribute to transmission desynchronisation and dysconnectivity, whereas the failure of astrocytic differentiation results in abnormal glial coverage and support of synapses. The delayed and deficient maturation of astrocytes might, in parallel, lead to disruption of glutamatergic, potassium, and neuromodulatory homoeostasis, resulting in dysregulated synaptic transmission. By highlighting a role for glial cells in schizophrenia, these studies potentially point to new mechanisms for disease modification.
Collapse
Affiliation(s)
- Andrea G Dietz
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Steven A Goldman
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA.
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
39
|
Hsieh CT, Lee YJ, Lee JW, Lu S, Tucci MA, Dai X, Ojeda NB, Lee HJ, Fan LW, Tien LT. Interleukin-1 receptor antagonist ameliorates the pain hypersensitivity, spinal inflammation and oxidative stress induced by systemic lipopolysaccharide in neonatal rats. Neurochem Int 2020; 135:104686. [PMID: 31987865 DOI: 10.1016/j.neuint.2020.104686] [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: 09/15/2019] [Revised: 01/05/2020] [Accepted: 01/13/2020] [Indexed: 02/07/2023]
Abstract
Perinatal inflammation-induced reduction in pain threshold may alter pain sensitivity to hyperalgesia or allodynia which may persist into adulthood. In this study, we investigated the anti-inflammatory protective effect of interleukin-1 receptor antagonist (IL-1ra), an anti-inflammatory cytokine, on systemic lipopolysaccharide (LPS)-induced spinal cord inflammation and oxidative stress, thermal hyperalgesia, and mechanical allodynia in neonatal rats. Intraperitoneal (i.p.) injection of LPS (2 mg/kg) or sterile saline was performed in postnatal day 5 (P5) rat pups, and IL-1ra (100 mg/kg) or saline was administered (i.p.) 5 min after LPS injection. Pain reflex behavior, spinal cord inflammation and oxidative stress were examined 24 h after LPS administration. Systemic LPS exposure led to a reduction of tactile threshold in the von Frey filament tests (mechanical allodynia) and pain response latency in the tail-flick test (thermal hyperalgesia) of P6 neonatal rats. Spinal cord inflammation was indicated by the increased numbers of activated glial cells including microglia (Iba1+) and astrocytes (GFAP+), and elevated levels of pro-inflammatory cytokine interleukin-1β (IL-1β), cyclooxygenase-2 (COX-2), and prostaglandin E2 (PGE2) 24 h after LPS treatment. LPS treatment induced spinal oxidative stress as evidenced by the increase in thiobarbituric acid reactive substances (TBARS) content in the spinal cord. LPS exposure also led to a significant increase in oligodendrocyte lineage population (Olig2+) and mature oligodendrocyte cells (APC+) in the neonatal rat spinal cord. IL-1ra treatment significantly reduced LPS-induced effects including hyperalgesia, allodynia, the increased number of activated microglia, astrocytes and oligodendrocytes, and elevated levels of IL-1β, COX-2, PGE2, and lipid peroxidation (TBARS) in the neonatal rat spinal cord. These data suggest that IL-1ra provides a protective effect against the development of pain hypersensitivity, spinal cord inflammation and oxidative stress in the neonatal rats following LPS exposure, which may be associated with the blockade of LPS-induced pro-inflammatory cytokine IL-1β.
Collapse
Affiliation(s)
- Cheng-Ta Hsieh
- School of Medicine, Fu Jen Catholic University, New Taipei City, 24205, Taiwan; Division of Neurosurgery, Department of Surgery, Cathay General Hospital, Taipei, 10630, Taiwan; Department of Chemistry, Fu Jen Catholic University, New Taipei City, 24205, Taiwan; Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, 24205, Taiwan
| | - Yih-Jing Lee
- School of Medicine, Fu Jen Catholic University, New Taipei City, 24205, Taiwan
| | - Jonathan W Lee
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Silu Lu
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA; Department of Neurology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Michelle A Tucci
- Department of Anesthesiology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Xiaoli Dai
- Department of Anesthesiology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Norma Beatriz Ojeda
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Hyun Joon Lee
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS, 39216, USA; Research Services, G.V. (Sonny) Montgomery Veterans Administration Medical Center, Jackson, MS, 39216, USA
| | - Lir-Wan Fan
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Lu-Tai Tien
- School of Medicine, Fu Jen Catholic University, New Taipei City, 24205, Taiwan.
| |
Collapse
|
40
|
Yang L, Su Y, Guo F, Zhang H, Zhao Y, Huang Q, Xu H. Deep rTMS Mitigates Behavioral and Neuropathologic Anomalies in Cuprizone-Exposed Mice Through Reducing Microglial Proinflammatory Cytokines. Front Integr Neurosci 2020; 14:556839. [PMID: 33250722 PMCID: PMC7674917 DOI: 10.3389/fnint.2020.556839] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 10/05/2020] [Indexed: 02/05/2023] Open
Abstract
In comparison to conventional repetitive transcranial magnetic stimulation (rTMS), theta burst stimulation is stronger and more effective as a brain stimulation approach within short periods. Although this deep rTMS technique is being applied in treating neuropsychiatric disorders, few animal studies have attempted to clarify the neurobiological mechanisms underlying its beneficial effects. This animal study examined the effects of deep rTMS on the cuprizone-induced neuropathologic and behavioral anomalies and explored the underlying mechanism. Adolescent male C57BL/6 mice were fed a rodent chow without or with cuprizone (CPZ; 0.2% w/w) for 5 weeks. Another two groups of mice were subjected to deep rTMS or sham rTMS once a day during weeks 2-5 of the CPZ-feeding period. The behaviors of all mice were assessed after the withdrawal of CPZ before neuropathological and immunological analyses. Compared to the CNT group, mice in CPZ and CPZ + Sham groups showed deficits in social recognition and spatial working memory as well as anxiety-like behavior, in addition to myelin breakdown and OL loss in the corpus callosum (CC), caudate putamen, cerebral cortex, and hippocampus of the brain. Deep rTMS effectively reduced behavioral anomalies and blocked myelin breakdown and OL loss in CPZ-fed mice. Besides, it also dampened microglia activation at lesion sites and rectified cytokines levels (IL-1β, IL-6, and IL-10) in CPZ-affected regions. The most significant effect was seen in the cerebral cortex where alleviated neuropathology co-existed with less microglia activation and higher IL-10 level. These data provided experimental evidence for the beneficial effects of deep rTMS in CPZ-fed mice and revealed a neurobiological mechanism of the modality.
Collapse
Affiliation(s)
- Liu Yang
- The Mental Health Center, Shantou University Medical College, Shantou, China
| | - Yawen Su
- The Mental Health Center, Shantou University Medical College, Shantou, China
| | - Fannv Guo
- The Mental Health Center, Shantou University Medical College, Shantou, China
| | - Handi Zhang
- The Mental Health Center, Shantou University Medical College, Shantou, China
| | - Yinglin Zhao
- The Mental Health Center, Shantou University Medical College, Shantou, China
| | - Qinjun Huang
- The Mental Health Center, Shantou University Medical College, Shantou, China
- *Correspondence: Qinjun Huang Haiyun Xu
| | - Haiyun Xu
- The Mental Health Center, Shantou University Medical College, Shantou, China
- The School of Psychiatry, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Qinjun Huang Haiyun Xu
| |
Collapse
|
41
|
Pierre WC, Legault L, Londono I, McGraw S, Lodygensky GA. Alteration of the brain methylation landscape following postnatal inflammatory injury in rat pups. FASEB J 2020; 34:432-445. [PMID: 31914673 PMCID: PMC6972494 DOI: 10.1096/fj.201901461r] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/26/2019] [Accepted: 10/08/2019] [Indexed: 12/18/2022]
Abstract
Preterm infants are vulnerable to inflammation-induced white matter injury (WMI), which is associated with neurocognitive impairment and increased risk of neuropsychiatric diseases in adulthood. Epigenetic mechanisms, particularly DNA methylation, play a role in normal development and modulate the response to pathological challenges. Our aims were to determine how WMI triggered DNA methylation alterations in brains of neonatal rats and if such changes persisted over time. We used a robust model of WMI by injecting lipopolysaccharide (LPS) or sterile saline in the corpus callosum of 3-day-old (P3) rat pups. Brains were collected 24 hours (P4) and 21 days post-injection (P24). We extracted genomic DNA from the brain to establish genome-wide quantitative DNA methylation profiles using reduced representation bisulfite sequencing. Neonatal LPS exposure induced a persistent increased methylation of genes related to nervous system development and a reduced methylation of genes associated with inflammatory pathways. These findings suggest that early-life neuroinflammatory exposure impacts the cerebral methylation landscape with determining widespread epigenetic modifications especially in genes related to neurodevelopment.
Collapse
Affiliation(s)
- Wyston C. Pierre
- Department of PediatricsUniversité de MontréalResearch Center of the CHU Sainte‐JustineMontréalQCCanada
| | - Lisa‐Marie Legault
- Department of Biochemistry and Molecular MedicineUniversité de MontréalResearch Center of the CHU Sainte‐JustineMontréalQCCanada
| | - Irene Londono
- Department of PediatricsUniversité de MontréalResearch Center of the CHU Sainte‐JustineMontréalQCCanada
| | - Serge McGraw
- Department of Biochemistry and Molecular MedicineUniversité de MontréalResearch Center of the CHU Sainte‐JustineMontréalQCCanada
- Department of Obstetrics & GynecologyUniversité de MontréalResearch Center of the CHU Sainte‐JustineMontréalQCCanada
| | - Gregory A. Lodygensky
- Department of PediatricsUniversité de MontréalResearch Center of the CHU Sainte‐JustineMontréalQCCanada
- Department of Pharmacology and PhysiologyUniversité de MontréalMontréalQCCanada
| |
Collapse
|
42
|
Janakiraman U, Yu J, Moutal A, Chinnasamy D, Boinon L, Batchelor SN, Anandhan A, Khanna R, Nelson MA. TAF1-gene editing alters the morphology and function of the cerebellum and cerebral cortex. Neurobiol Dis 2019; 132:104539. [PMID: 31344492 PMCID: PMC7197880 DOI: 10.1016/j.nbd.2019.104539] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/20/2019] [Accepted: 07/19/2019] [Indexed: 10/26/2022] Open
Abstract
TAF1/MRSX33 intellectual disability syndrome is an X-linked disorder caused by loss-of-function mutations in the TAF1 gene. How these mutations cause dysmorphology, hypotonia, intellectual and motor defects is unknown. Mouse models which have embryonically targeted TAF1 have failed, possibly due to TAF1 being essential for viability, preferentially expressed in early brain development, and intolerant of mutation. Novel animal models are valuable tools for understanding neuronal pathology. Here, we report the development and characterization of a novel animal model for TAF1 ID syndrome in which the TAF1 gene is deleted in embryonic rats using clustered regularly interspaced short palindromic repeats (CRISPR) associated protein 9 (Cas9) technology and somatic brain transgenesis mediated by lentiviral transduction. Rat pups, post-natal day 3, were subjected to intracerebroventricular (ICV) injection of either gRNA-control or gRNA-TAF1 vectors. Rats were subjected to a battery of behavioral tests followed by histopathological analyses of brains at post-natal day 14 and day 35. TAF1-edited rats exhibited behavioral deficits at both the neonatal and juvenile stages of development. Deletion of TAF1 lead to a hypoplasia and loss of the Purkinje cells. We also observed a decreased in GFAP positive astrocytes and an increase in Iba1 positive microglia within the granular layer of the cerebellum in TAF1-edited animals. Immunostaining revealed a reduction in the expression of the CaV3.1 T-type calcium channel. Abnormal motor symptoms in TAF1-edited rats were associated with irregular cerebellar output caused by changes in the intrinsic activity of the Purkinje cells due to loss of pre-synaptic CaV3.1. This animal model provides a powerful new tool for studies of neuronal dysfunction in conditions associated with TAF1 abnormalities and should prove useful for developing therapeutic strategies to treat TAF1 ID syndrome.
Collapse
Affiliation(s)
- Udaiyappan Janakiraman
- Department of Pathology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA
| | - Jie Yu
- Department of Pharmacology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA; College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310058, China
| | - Aubin Moutal
- Department of Pharmacology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA
| | - Dhanalakshmi Chinnasamy
- Department of Pathology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA
| | - Lisa Boinon
- Department of Pharmacology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA
| | - Shelby N Batchelor
- Department of Pathology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA
| | - Annaduri Anandhan
- Department of Pharmacology and Toxicology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA
| | - Rajesh Khanna
- Department of Pathology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA; Department of Pharmacology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA; The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, AZ, United States of America; The BIO5 Institute, University of Arizona, United States of America
| | - Mark A Nelson
- Department of Pathology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA.
| |
Collapse
|
43
|
Chen Z, Chopp M, Zacharek A, Li W, Venkat P, Wang F, Landschoot-Ward J, Chen J. Brain-Derived Microparticles (BDMPs) Contribute to Neuroinflammation and Lactadherin Reduces BDMP Induced Neuroinflammation and Improves Outcome After Stroke. Front Immunol 2019; 10:2747. [PMID: 31993045 PMCID: PMC6968774 DOI: 10.3389/fimmu.2019.02747] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 11/08/2019] [Indexed: 12/28/2022] Open
Abstract
Microparticles (MPs, ~size between 0.1 and 1 mm) are lipid encased containers derived from intact cells which contain antigen from the parent cells. MPs are involved in intercellular communication and regulate inflammation. Stroke increases secretion of brain derived MP (BDMP) which activate macrophages/microglia and induce neuroinflammation. Lactadherin (Milk fat globule–EGF factor-8) binds to anionic phospholipids and extracellular matrices, promotes apoptotic cell clearance and limits pathogenic antigen cross presentation. In this study, we investigate whether BDMP affects stroke-induced neuroinflammation and whether Lactadherin treatment reduces stroke initiated BDMP-induced neuroinflammation, thereby improving functional outcome after stroke. Middle aged (8–9 months old) male C57BL/6J mice were subjected to distal middle cerebral artery occlusion (dMCAo) stroke, and BDMPs were extracted from ischemic brain 24 h after dMCAo by ultracentrifugation. Adult male C57BL/6J mice were subjected to dMCAo and treated via tail vein injection at 3 h after stroke with: (A) +PBS (n = 5/group); (B) +BDMPs (1.5 × 108, n = 6/group); (C) +Lactadherin (400 μg/kg, n = 5/group); (D) +BDMP+Lactadherin (n = 6/group). A battery of neurological function tests were performed and mice sacrificed for immunostaining at 14 days after stroke. Blood plasma was used for Western blot assay. Our data indicate: (1) treatment of Stroke with BDMP significantly increases lesion volume, neurological deficits, blood brain barrier (BBB) leakage, microglial activation, inflammatory cell infiltration (CD45, microglia/macrophages, and neutrophils) into brain, inflammatory factor (TNFα, IL6, and IL1β) expression in brain, increases axon/white matter (WM) damage identified by decreased axon and myelin density, and increases inflammatory factor expression in the plasma when compared to PBS treated stroke mice; (2) when compared to PBS and BDMP treated stroke mice, Lactadherin and BDMP+Lactadherin treatment significantly improves neurological outcome, and decreases lesion volume, BBB leakage, axon/WM injury, inflammatory cell infiltration and inflammatory factor expression in the ischemic brain, respectively. Lactadherin treatment significantly increases anti-inflammatory factor (IL10) expression in ischemic brain and decreases IL1β expression in plasma compared to PBS and BDMP treated stroke mice, respectively. BDMP increases neuroinflammation and aggravates ischemic brain damage after stroke. Thus, Lactadherin exerts anti-inflammatory effects and improves the clearance of MPs to reduce stroke and BDMP induced neurological deficits.
Collapse
Affiliation(s)
- Zhili Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States.,Department of Physics, Oakland University, Rochester, MI, United States
| | - Alex Zacharek
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Wei Li
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Poornima Venkat
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Fenjie Wang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | | | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| |
Collapse
|
44
|
McNamara NB, Miron VE. Microglia in developing white matter and perinatal brain injury. Neurosci Lett 2019; 714:134539. [PMID: 31614181 DOI: 10.1016/j.neulet.2019.134539] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/04/2019] [Accepted: 10/07/2019] [Indexed: 12/13/2022]
Abstract
Perinatal brain injury (PBI) to the developing white matter results in hypomyelination of axons and can cause long-term motor and cognitive deficits e.g. cerebral palsy. There are currently no approved therapies aimed at repairing the white matter following insult, underscoring the need to investigate the mechanisms underlying the pathogenesis of PBI. Microglia have been strongly implicated, but their function and heterogeneity in this context remain poorly understood, posing a barrier to the development of microglia-targeted therapies for white matter repair following PBI. In this review, we discuss the roles of microglia in normal white matter development and in PBI, and potential drug strategies to influence microglial responses in this setting.
Collapse
Affiliation(s)
- Niamh B McNamara
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Veronique E Miron
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
45
|
Pierre WC, Akakpo L, Londono I, Pouliot P, Chemtob S, Lesage F, Lodygensky GA. Assessing therapeutic response non-invasively in a neonatal rat model of acute inflammatory white matter injury using high-field MRI. Brain Behav Immun 2019; 81:348-360. [PMID: 31247289 DOI: 10.1016/j.bbi.2019.06.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 05/20/2019] [Accepted: 06/22/2019] [Indexed: 12/19/2022] Open
Abstract
Perinatal infection and inflammatory episodes in preterm infants are associated with diffuse white matter injury (WMI) and adverse neurological outcomes. Inflammation-induced WMI was previously shown to be linked with later hippocampal atrophy as well as learning and memory impairments in preterm infants. Early evaluation of injury load and therapeutic response with non-invasive tools such as multimodal magnetic resonance imaging (MRI) would greatly improve the search of new therapeutic approaches in preterm infants. Our aim was to evaluate the potential of multimodal MRI to detect the response of interleukin-1 receptor antagonist (IL-1Ra) treatment, known for its neuroprotective properties, during the acute phase of injury on a model of neonatal WMI. Rat pups at postnatal day 3 (P3) received intracerebral injection of lipopolysaccharide with systemic IL-1Ra therapy. 24 h later (P4), rats were imaged with multimodal MRI to assess microstructure by diffusion tensor imaging (DTI) and neurochemical profile of the hippocampus with 1H-magnetic resonance spectroscopy. Astrocyte and microglial activation, apoptosis and the mRNA expression of pro-inflammatory and necroptotic markers were assessed. During the acute phase of injury, neonatal LPS exposure altered the concentration of hippocampus metabolites related to neuronal integrity, neurotransmission and membrane integrity and induced diffusivity restriction. Just 24 h after initiation of therapy, early indication of IL-1Ra neuroprotective effect could be detected in vivo by non-invasive spectroscopy and DTI, and confirmed with immunohistochemical evaluation and mRNA expression of inflammatory markers and cell death. In conclusion, multimodal MRI, particularly DTI, can detect not only injury but also the acute therapeutic effect of IL-1Ra suggesting that MRI could be a useful non-invasive tool to follow, at early time points, the therapeutic response in preterm infants.
Collapse
Affiliation(s)
- Wyston C Pierre
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada; Department of Pharmacology, Université de Montréal, Montréal, Canada
| | - Luis Akakpo
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada; École Polytechnique de Montréal, Montreal, QC, Canada
| | - Irène Londono
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
| | - Philippe Pouliot
- École Polytechnique de Montréal, Montreal, QC, Canada; Montreal Heart Institute, Montreal, QC, Canada
| | - Sylvain Chemtob
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada; Department of Pharmacology, Université de Montréal, Montréal, Canada; Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada
| | - Frédéric Lesage
- École Polytechnique de Montréal, Montreal, QC, Canada; Montreal Heart Institute, Montreal, QC, Canada
| | - Gregory A Lodygensky
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada; Department of Pharmacology, Université de Montréal, Montréal, Canada; Montreal Heart Institute, Montreal, QC, Canada.
| |
Collapse
|
46
|
Quinlan S, Merino-Serrais P, Di Grande A, Dussmann H, Prehn JHM, Ní Chonghaile T, Henshall DC, Jimenez-Mateos EM. The Anti-inflammatory Compound Candesartan Cilexetil Improves Neurological Outcomes in a Mouse Model of Neonatal Hypoxia. Front Immunol 2019; 10:1752. [PMID: 31396238 PMCID: PMC6667988 DOI: 10.3389/fimmu.2019.01752] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 07/10/2019] [Indexed: 11/15/2022] Open
Abstract
Recent studies suggest that mild hypoxia-induced neonatal seizures can trigger an acute neuroinflammatory response leading to long-lasting changes in brain excitability along with associated cognitive and behavioral deficits. The cellular elements and signaling pathways underlying neuroinflammation in this setting remain incompletely understood but could yield novel therapeutic targets. Here we show that brief global hypoxia-induced neonatal seizures in mice result in transient cytokine production, a selective expansion of microglia and long-lasting changes to the neuronal structure of pyramidal neurons in the hippocampus. Treatment of neonatal mice after hypoxia-seizures with the novel anti-inflammatory compound candesartan cilexetil suppressed acute seizure-damage and mitigated later-life aggravated seizure responses and hippocampus-dependent learning deficits. Together, these findings improve our understanding of the effects of neonatal seizures and identify potentially novel treatments to protect against short and long-lasting harmful effects.
Collapse
Affiliation(s)
- Sean Quinlan
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Paula Merino-Serrais
- Division for Neurogeriatrics, Department of Neurobiology Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.,Departamento de Neurobiologia Funcional y de Sistemas, Instituto Cajal, Consejo Superior de Investigaciones Cientificas, Madrid, Spain
| | - Alessandra Di Grande
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Heiko Dussmann
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jochen H M Prehn
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.,FutureNeuro Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Tríona Ní Chonghaile
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - David C Henshall
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.,FutureNeuro Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland.,INFANT Research Centre, UCC, Cork, Ireland
| | - Eva M Jimenez-Mateos
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.,Department of Physiology, School of Medicine, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| |
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW With the incidence of neurodevelopmental disorders on the rise, it is imperative to identify and understand the mechanisms by which environmental contaminants can impact the developing brain and heighten risk. Here, we report on recent findings regarding novel mechanisms of developmental neurotoxicity and highlight chemicals of concern, beyond traditionally defined neurotoxicants. RECENT FINDINGS The perinatal window represents a critical and extremely vulnerable period of time during which chemical insult can alter the morphological and functional trajectory of the developing brain. Numerous chemical classes have been associated with alterations in neurodevelopment including metals, solvents, pesticides, and, more recently, endocrine-disrupting compounds. Although mechanisms of neurotoxicity have traditionally been identified as pathways leading to neuronal cell death, neuropathology, or severe neural injury, recent research highlights alternative mechanisms that result in more subtle but consequential changes in the brain and behavior. These emerging areas of interest include neuroendocrine and immune disruption, as well as indirect toxicity via actions on other organs such as the gut and placenta. Understanding of the myriad ways in which the developing brain is vulnerable to chemical exposures has grown tremendously over the past decade. Further progress and implementation in risk assessment is critical to reducing risk of neurodevelopmental disorders.
Collapse
|
48
|
Improvement in dysmyelination by the inhibition of microglial activation in a mouse model of Sandhoff disease. Neuroreport 2019; 29:962-967. [PMID: 29847465 DOI: 10.1097/wnr.0000000000001060] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Sandhoff disease (SD) is a genetic disorder caused by a mutation of the β-subunit gene β-hexosaminidase B (HexB) in humans, which results in the massive accumulation of the ganglioside GM2 and related glycosphingolipids in the nervous system. SD causes progressive neurodegeneration and changes in white matter in human infants. An animal model of SD has been established, Hexb-deficient (Hexb) mice, which shows abnormalities resembling the severe phenotype found in human infants. Previously, we reported that the activation state of microglia caused astrogliosis in the early stage of Hexb mouse development. To study how the symptoms of SD develop, we explored the difference in gene expression between 4-week-old Hexb and Hexb mouse cerebral cortices by microarray analysis. The data indicated not only the upregulation of immune system-related genes but also the downregulation of myelin-related genes in the 4-week-old Hexb mouse cerebral cortices. To test the correlation between inflammation and dysmyelination, we generated double-knockout mice of Hexb and the Fc receptor γ gene (Fcrγ), which is a regulator of autoimmune responses. Dysmyelination recovered in these double-knockout mice. The number of oligodendrocyte progenitors, which expressed platelet-derived growth factor receptor-α, did not change in the 2-week-old mouse brain. These results indicate that microglial activation plays an important role in the myelination process, without influencing the number of oligodendrocyte progenitors, in the development of Hexb mice.
Collapse
|
49
|
Kentner AC, Bilbo SD, Brown AS, Hsiao EY, McAllister AK, Meyer U, Pearce BD, Pletnikov MV, Yolken RH, Bauman MD. Maternal immune activation: reporting guidelines to improve the rigor, reproducibility, and transparency of the model. Neuropsychopharmacology 2019; 44:245-258. [PMID: 30188509 PMCID: PMC6300528 DOI: 10.1038/s41386-018-0185-7] [Citation(s) in RCA: 180] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 07/04/2018] [Accepted: 08/02/2018] [Indexed: 01/16/2023]
Abstract
The 2017 American College of Neuropychopharmacology (ACNP) conference hosted a Study Group on 4 December 2017, Establishing best practice guidelines to improve the rigor, reproducibility, and transparency of the maternal immune activation (MIA) animal model of neurodevelopmental abnormalities. The goals of this session were to (a) evaluate the current literature and establish a consensus on best practices to be implemented in MIA studies, (b) identify remaining research gaps warranting additional data collection and lend to the development of evidence-based best practice design, and (c) inform the MIA research community of these findings. During this session, there was a detailed discussion on the importance of validating immunogen doses and standardizing the general design (e.g., species, immunogenic compound used, housing) of our MIA models both within and across laboratories. The consensus of the study group was that data does not currently exist to support specific evidence-based model selection or methodological recommendations due to lack of consistency in reporting, and that this issue extends to other inflammatory models of neurodevelopmental abnormalities. This launched a call to establish a reporting checklist focusing on validation, implementation, and transparency modeled on the ARRIVE Guidelines and CONSORT (scientific reporting guidelines for animal and clinical research, respectively). Here we provide a summary of the discussions in addition to a suggested checklist of reporting guidelines needed to improve the rigor and reproducibility of this valuable translational model, which can be adapted and applied to other animal models as well.
Collapse
Affiliation(s)
- Amanda C. Kentner
- 0000 0001 0021 3995grid.416498.6School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA USA
| | - Staci D. Bilbo
- 000000041936754Xgrid.38142.3cDepartment of Pediatrics, Harvard Medical School, Boston, MA USA ,0000 0004 0386 9924grid.32224.35Lurie Center for Autism, Massachusetts General Hospital for Children, Boston, MA USA
| | - Alan S. Brown
- 0000000419368729grid.21729.3fDepartment of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY USA ,0000 0000 8499 1112grid.413734.6New York State Psychiatric Institute, New York, NY USA
| | - Elaine Y. Hsiao
- 0000 0000 9632 6718grid.19006.3eDepartment of Integrative Biology and Physiology, University of California, Los Angeles, USA
| | - A. Kimberley McAllister
- 0000 0004 1936 9684grid.27860.3bCenter for Neuroscience, University of California Davis, Davis, CA USA
| | - Urs Meyer
- 0000 0004 1937 0650grid.7400.3Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse, Zurich, Switzerland ,0000 0004 1937 0650grid.7400.3Neuroscience Centre Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Brad D. Pearce
- 0000 0001 0941 6502grid.189967.8Department of Epidemiology, Rollins School of Public Health, and Graduate Division of Biological and Biomedical Sciences, Neuroscience Program, Emory University, Atlanta, GA USA
| | - Mikhail V. Pletnikov
- 0000 0001 2171 9311grid.21107.35Department of Psychiatry and Behavioral Sciences, Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Robert H. Yolken
- 0000 0001 2171 9311grid.21107.35Department of Pediatrics, Stanley Division of Developmental Neurovirology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Melissa D. Bauman
- 0000 0004 1936 9684grid.27860.3bThe UC Davis MIND Institute, Department of Psychiatry and Behavioral Sciences, California National Primate Research Center, University of California, Davis, USA
| |
Collapse
|
50
|
Promising neuroprotective effects of β-caryophyllene against LPS-induced oligodendrocyte toxicity: A mechanistic study. Biochem Pharmacol 2018; 159:154-171. [PMID: 30529211 DOI: 10.1016/j.bcp.2018.12.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 12/04/2018] [Indexed: 01/09/2023]
Abstract
Myelin loss subsequent to oligodendrocyte death has been reported in a variety of myelin-associated disorders such as multiple sclerosis (MS). Lipopolysaccharide (LPS) has been shown to elicit cellular responses in the central nervous system (CNS) and trigger immune infiltrates and glial cells to release a variety of inflammatory cytokines and mediators. LPS-induced oligodendrocytes toxicity may be chosen as an efficient model to evaluate the role of oligodendrocytes in neuroprotective activities of compounds. β-Caryophyllene (BCP) is a selective type 2 cannabinoid (CB2) receptor agonist. However, the mechanisms underlying the anti-inflammatory effects of BCP are not completely understood. On this basis, we aimed to investigate the protective effects of a wide range of BCP concentrations against LPS-induced toxicity in a proliferative oligodendrocyte cell line (OLN-93) and evaluate the possible correlation between BCP concentration and selective modulation of CB2, Nrf2, sphingomyelinase (SMase) and peroxisome proliferator-activated receptors (PPAR)-γ signaling pathways. We found that LPS significantly increases the levels of reactive oxygen species (ROS), nitric oxide (NO) metabolite and tumor necrosis factor (TNF)-α production while decreases the level of GSH. BCP could prevent LPS-induced cytotoxicity and excessive production of NO, ROS, and TNF-α. Also, we demonstrated that BCP's protective effects against LPS-induced oligodendrocytes toxicity were mediated via the CB2 receptor through different pathways including Nrf2/HO-1/anti-oxidant axis, and PPAR-γ, at low (0.2 and 1 µM), and high (10-50 µM) concentrations, respectively. Additionally, we observed that the addition of SMase inhibitors imipramine (IMP) and fluoxetine (FLX) synergistically increased the protective effects of BCP. Finally, BCP at low concentrations exerted promising protective effects that could be considered for the treatment of neurodegenerative disorders such as MS. However, more studies using other models of neurodegenerative diseases should be undertaken to assess different parameters such as the activity or expression of SMase.
Collapse
|