1
|
Wang KC, Ojeda NB, Wang H, Chiang HS, Tucci MA, Lee JW, Wei HC, Kaizaki-Mitsumoto A, Tanaka S, Dankhara N, Tien LT, Fan LW. Neonatal brain inflammation enhances methamphetamine-induced reinstated behavioral sensitization in adult rats analyzed with explainable machine learning. Neurochem Int 2024; 176:105743. [PMID: 38641026 PMCID: PMC11102812 DOI: 10.1016/j.neuint.2024.105743] [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: 12/18/2023] [Revised: 03/15/2024] [Accepted: 04/15/2024] [Indexed: 04/21/2024]
Abstract
Neonatal brain inflammation produced by intraperitoneal (i.p.) injection of lipopolysaccharide (LPS) results in long-lasting brain dopaminergic injury and motor disturbances in adult rats. The goal of the present work is to investigate the effect of neonatal systemic LPS exposure (1 or 2 mg/kg, i.p. injection in postnatal day 5, P5, male rats)-induced dopaminergic injury to examine methamphetamine (METH)-induced behavioral sensitization as an indicator of drug addiction. On P70, subjects underwent a treatment schedule of 5 once daily subcutaneous (s.c.) administrations of METH (0.5 mg/kg) (P70-P74) to induce behavioral sensitization. Ninety-six hours following the 5th treatment of METH (P78), the rats received one dose of 0.5 mg/kg METH (s.c.) to reintroduce behavioral sensitization. Hyperlocomotion is a critical index caused by drug abuse, and METH administration has been shown to produce remarkable locomotor-enhancing effects. Therefore, a random forest model was used as the detector to extract the feature interaction patterns among the collected high-dimensional locomotor data. Our approaches identified neonatal systemic LPS exposure dose and METH-treated dates as features significantly associated with METH-induced behavioral sensitization, reinstated behavioral sensitization, and perinatal inflammation in this experimental model of drug addiction. Overall, the analysis suggests that the implementation of machine learning strategies is sensitive enough to detect interaction patterns in locomotor activity. Neonatal LPS exposure also enhanced METH-induced reduction of dopamine transporter expression and [3H]dopamine uptake, reduced mitochondrial complex I activity, and elevated interleukin-1β and cyclooxygenase-2 concentrations in the P78 rat striatum. These results indicate that neonatal systemic LPS exposure produces a persistent dopaminergic lesion leading to a long-lasting change in the brain reward system as indicated by the enhanced METH-induced behavioral sensitization and reinstated behavioral sensitization later in life. These findings indicate that early-life brain inflammation may enhance susceptibility to drug addiction development later in life, which provides new insights for developing potential therapeutic treatments for drug addiction.
Collapse
Affiliation(s)
- Kuo-Ching Wang
- Department of Anesthesiology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei City, Taiwan
| | - Norma B Ojeda
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA; Department of Advanced Biomedical Education, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Haifeng Wang
- Department of Industrial and Systems Engineering, Mississippi State University, Mississippi State, MS, 39762, USA
| | - Han-Sun Chiang
- School of Medicine, Fu Jen Catholic University, Xinzhuang Dist, New Taipei City, 24205, Taiwan
| | - Michelle A Tucci
- Department of Anesthesiology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Jonathan W Lee
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Han-Chi Wei
- School of Medicine, Fu Jen Catholic University, Xinzhuang Dist, New Taipei City, 24205, Taiwan
| | - Asuka Kaizaki-Mitsumoto
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA; Department of Toxicology, Showa University Graduate School of Pharmacy, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Sachiko Tanaka
- Center for Research and Development in Pharmacy Education, School of Pharmacy, Nihon University, Funabashi, Chiba, 274-8555, Japan
| | - Nilesh Dankhara
- 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, Xinzhuang Dist, New Taipei City, 24205, Taiwan.
| | - Lir-Wan Fan
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA.
| |
Collapse
|
2
|
Geng R, Zhao Y, Xu W, Ma X, Jiang Y, Han X, Zhao L, Li Y. SIRPB1 regulates inflammatory factor expression in the glioma microenvironment via SYK: functional and bioinformatics insights. J Transl Med 2024; 22:338. [PMID: 38594692 PMCID: PMC11003053 DOI: 10.1186/s12967-024-05149-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 03/31/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND SIRPB1 expression is upregulated in various tumor types, including gliomas, and is known to contribute to tumor progression; nevertheless, its function in the immune milieu of gliomas is still mainly unknown. METHODS This study, we analyzed 1152 normal samples from the GTEx database and 670 glioma samples from the TCGA database to investigate the relationship between the expression of SIRPB1 and clinicopathological features. Moreover, SIRPB1 gene knockout THP-1 cell lines were constructed using CRISPR/Cas9 and were induced into a co-culture of macrophages and glioma cells in vitro to learn more about the role of SIRPB1 in the glioma immune milieu. Lastly, we established a prognostic model to predict the effect of SIRPB1 on prognosis. RESULTS Significantly higher levels of SIRPB1 expression were found in gliomas, which had an adverse effect on the immune milieu and correlated poorly with patient survival. SIRPB1 activation with certain antibodies results in SYK phosphorylation and the subsequent activation of calcium, MAPK, and NF-κB signaling pathways. This phenomenon is primarily observed in myeloid-derived cells as opposed to glioma cells. In vitro co-culture demonstrated that macrophages with SIRPB1 knockout showed decreased IL1RA, CCL2, and IL-8, which were recovered upon ectopic expression of SIRPB1 but reduced again following treatment with SYK inhibitor GS9973. Critically, a lower overall survival rate was linked to increased SIRPB1 expression. Making use of SIRPB1 expression along with additional clinicopathological variables, we established a nomogram that showed a high degree of prediction accuracy. CONCLUSIONS Our study demonstrates that glioma cells can be activated by macrophages via SIRPB1, subsequently reprogramming the TME, suggesting that SIRPB1 could serve as a promising therapeutic target for gliomas.
Collapse
Affiliation(s)
- Ren Geng
- Department of Neurosurgery, First Hospital of Jilin University, No. 1, Xinmin Street, Chaoyang District, Changchun, China
| | - Yao Zhao
- Department of Neurosurgery, First Hospital of Jilin University, No. 1, Xinmin Street, Chaoyang District, Changchun, China
| | - Wanzhen Xu
- Department of Neurosurgery, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, China
| | - Xiaoshan Ma
- Department of Neurosurgery, First Hospital of Jilin University, No. 1, Xinmin Street, Chaoyang District, Changchun, China
| | - Yining Jiang
- Department of Neurosurgery, First Hospital of Jilin University, No. 1, Xinmin Street, Chaoyang District, Changchun, China
| | - Xuefei Han
- Department of Neurosurgery, First Hospital of Jilin University, No. 1, Xinmin Street, Chaoyang District, Changchun, China
| | - Liyan Zhao
- Department of Blood Transfusion, Second Hospital of Jilin University, No. 4026, Yatai Street, Nanguan District, Changchun, China.
| | - Yunqian Li
- Department of Neurosurgery, First Hospital of Jilin University, No. 1, Xinmin Street, Chaoyang District, Changchun, China.
| |
Collapse
|
3
|
Yang Y, Yao Z, Wang H, Jia S, Wang M, Wang S, Yun D. Severe inflammation in C57/BL6 mice leads to prolonged cognitive impairment by initiating the IL-1β/TRPM2 pathway. Int Immunopharmacol 2024; 128:111380. [PMID: 38176340 DOI: 10.1016/j.intimp.2023.111380] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/26/2023] [Accepted: 12/11/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND Sepsis could lead to chronic cognitive impairment by unclear molecular mechanisms. Transient receptor potential melastatin-2 (TRPM2) is essential against immunity-related activities and inflammation. Our study attempted to decipher the relationship between cognitive impairment caused by severe inflammation and TRPM2 expression levels. METHODS Severe inflammation was induced by intraperitoneally injecting C57/BL6 mice with a high dosage (5 mg kg-1) of Lipopolysaccharide (LPS). Fear conditioning and a Morris water maze test were performed to examine the cognitive abilities of the mice. Moreover, the signaling and expression of pro-inflammatory cytokines and TRPM2 were measured using Western blotting and Reverse transcription-polymerase chain reaction (RT-PCR). Flow cytometry and immunofluorescence staining helped to determine the astrocyte apoptosis rate. RESULTS Severe inflammation can lead to long-term cognitive impairment in C57/BL6 mice. The interleukin-1 beta (IL-1β) levels intra-hippocampus were significantly elevated until P14 post-LPS introduction. At both P7 and P14, there is an up-regulation of TRPM2 expression within hippocampus. Administration of recombinant IL-1β to astrocytes results in a significant up-regulation of TRPM2 expression. IL-1β or TRPM2 level knockdown helped counter the cognitive impairment caused by significant inflammation. CONCLUSIONS A continuous increase in IL-1β levels within the hippocampus can lead to cognitive impairment by enhancing TRPM2 levels caused by severe inflammation.
Collapse
Affiliation(s)
- Yujiao Yang
- Department of Anesthesiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China; Department of Anesthesiology, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Zhihua Yao
- Department of Anesthesiology, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Hushan Wang
- Department of Anesthesiology, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Shuaiying Jia
- Department of Anesthesiology, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Mingfei Wang
- Department of Anesthesiology, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Shan Wang
- Department of Anesthesiology, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Debo Yun
- Department of Neurosurgery, Nanchong Central Hospital, Nanchong, Sichuan, China.
| |
Collapse
|
4
|
Xiang W, Xiang H, Wang J, Jiang Y, Pan C, Ji B, Zhang A. Fecal microbiota transplantation: a novel strategy for treating Alzheimer's disease. Front Microbiol 2023; 14:1281233. [PMID: 38033557 PMCID: PMC10687436 DOI: 10.3389/fmicb.2023.1281233] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
Alzheimer's disease is a common neurological disorder, which has become one of the major factors affecting human health due to its serious impact on individuals, families and society. It has been confirmed that gut microbiota can affect the occurrence and development of Alzheimer's disease. Especially, fecal microbiota transplantation plays a positive role in the treatment of Alzheimer's disease. The mechanisms for improving Alzheimer's disease might include anti-inflammation and regulation of amyloid β-protein, synaptic plasticity, short-chain fatty acids, and histone acetylation. In this mini-review, the relationship between fecal microbiota transplantation and Alzheimer's disease was summarized. It is hoped that fecal microbiota transplantation would play a positive role in the prevention and treatment of Alzheimer's disease in the future.
Collapse
Affiliation(s)
- Wu Xiang
- Department of Rehabilitation, Beibei Traditional Chinese Medical Hospital, Chongqing, China
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Han Xiang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Junyu Wang
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Yiqin Jiang
- Department of Rehabilitation, Beibei Traditional Chinese Medical Hospital, Chongqing, China
| | - Chuanhui Pan
- Department of Rehabilitation, Beibei Traditional Chinese Medical Hospital, Chongqing, China
| | - Bingjin Ji
- Department of Rehabilitation, Beibei Traditional Chinese Medical Hospital, Chongqing, China
| | - Anren Zhang
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Monet MC, Quan N. Complex Neuroimmune Involvement in Neurodevelopment: A Mini-Review. J Inflamm Res 2023; 16:2979-2991. [PMID: 37489149 PMCID: PMC10363380 DOI: 10.2147/jir.s410562] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 07/11/2023] [Indexed: 07/26/2023] Open
Abstract
It is increasingly evident that cells and molecules of the immune system play significant roles in neurodevelopment. As perinatal infection is associated with the development of neurodevelopmental disorders, previous research has focused on demonstrating that the induction of neuroinflammation in the developing brain is capable of causing neuropathology and behavioral changes. Recent studies, however, have revealed that immune cells and molecules in the brain can influence neurodevelopment without the induction of overt inflammation, identifying neuroimmune activities as integral parts of normal neurodevelopment. This mini-review describes the shift in literature that has moved from emphasizing the intrusion of inflammatory events as a main culprit of neurodevelopmental disorders to evaluating the deviation of the normal neuroimmune activities in neurodevelopment as a potential pathogenic mechanism.
Collapse
Affiliation(s)
- Marianne C Monet
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Ning Quan
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Jupiter, FL, USA
| |
Collapse
|
6
|
Pre- and Neonatal Exposure to Lead (Pb) Induces Neuroinflammation in the Forebrain Cortex, Hippocampus and Cerebellum of Rat Pups. Int J Mol Sci 2020; 21:ijms21031083. [PMID: 32041252 PMCID: PMC7037720 DOI: 10.3390/ijms21031083] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/01/2020] [Accepted: 02/03/2020] [Indexed: 12/11/2022] Open
Abstract
Lead (Pb) is a heavy metal with a proven neurotoxic effect. Exposure is particularly dangerous to the developing brain in the pre- and neonatal periods. One postulated mechanism of its neurotoxicity is induction of inflammation. This study analyzed the effect of exposure of rat pups to Pb during periods of brain development on the concentrations of selected cytokines and prostanoids in the forebrain cortex, hippocampus and cerebellum. Methods: Administration of 0.1% lead acetate (PbAc) in drinking water ad libitum, from the first day of gestation to postnatal day 21, resulted in blood Pb in rat pups reaching levels below the threshold considered safe for humans by the Centers for Disease Control and Prevention (10 µg/dL). Enzyme-linked immunosorbent assay (ELISA) method was used to determine the levels of interleukins IL-1β, IL-6, transforming growth factor-β (TGF-β), prostaglandin E2 (PGE2) and thromboxane B2 (TXB2). Western blot and quantitative real-time PCR were used to determine the expression levels of cyclooxygenases COX-1 and COX-2. Finally, Western blot was used to determine the level of nuclear factor kappa B (NF-κB). Results: In all studied brain structures (forebrain cortex, hippocampus and cerebellum), the administration of Pb caused a significant increase in all studied cytokines and prostanoids (IL-1β, IL-6, TGF-β, PGE2 and TXB2). The protein and mRNA expression of COX-1 and COX-2 increased in all studied brain structures, as did NF-κB expression. Conclusions: Chronic pre- and neonatal exposure to Pb induces neuroinflammation in the forebrain cortex, hippocampus and cerebellum of rat pups.
Collapse
|
7
|
Bai N, Zhang Q, Zhang W, Liu B, Yang F, Brann D, Wang R. G-protein-coupled estrogen receptor activation upregulates interleukin-1 receptor antagonist in the hippocampus after global cerebral ischemia: implications for neuronal self-defense. J Neuroinflammation 2020; 17:45. [PMID: 32007102 PMCID: PMC6995076 DOI: 10.1186/s12974-020-1715-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 01/13/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND G-protein-coupled estrogen receptor (GPER/GPR30) is a novel membrane-associated estrogen receptor that can induce rapid kinase signaling in various cells. Activation of GPER can prevent hippocampal neuronal cell death following transient global cerebral ischemia (GCI), although the mechanisms remain unclear. In the current study, we sought to address whether GPER activation exerts potent anti-inflammatory effects in the rat hippocampus after GCI as a potential mechanism to limit neuronal cell death. METHODS GCI was induced by four-vessel occlusion in ovariectomized female SD rats. Specific agonist G1 or antagonist G36 of GPER was administrated using minipump, and antisense oligonucleotide (AS) of interleukin-1β receptor antagonist (IL1RA) was administrated using brain infusion kit. Protein expression of IL1RA, NF-κB-P65, phosphorylation of CREB (p-CREB), Bcl2, cleaved caspase 3, and microglial markers Iba1, CD11b, as well as inflammasome components NLRP3, ASC, cleaved caspase 1, and Cle-IL1β in the hippocampal CA1 region were investigated by immunofluorescent staining and Western blot analysis. The Duolink II in situ proximity ligation assay (PLA) was performed to detect the interaction between NLRP3 and ASC. Immunofluorescent staining for NeuN and TUNEL analysis were used to analyze neuronal survival and apoptosis, respectively. We performed Barnes maze and Novel object tests to compare the cognitive function of the rats. RESULTS The results showed that G1 attenuated GCI-induced elevation of Iba1 and CD11b in the hippocampal CA1 region at 14 days of reperfusion, and this effect was blocked by G36. G1 treatment also markedly decreased expression of the NLRP3-ASC-caspase 1 inflammasome and IL1β activation, as well as downstream NF-κB signaling, the effects reversed by G36 administration. Intriguingly, G1 caused a robust elevation in neurons of a well-known endogenous anti-inflammatory factor IL1RA, which was reversed by G36 treatment. G1 also enhanced p-CREB level in the hippocampus, a transcription factor known to enhance expression of IL1RA. Finally, in vivo IL1RA-AS abolished the anti-inflammatory, neuroprotective, and anti-apoptotic effects of G1 after GCI and reversed the cognitive-enhancing effects of G1 at 14 days after GCI. CONCLUSIONS Taken together, the current results suggest that GPER preserves cognitive function following GCI in part by exerting anti-inflammatory effects and enhancing the defense mechanism of neurons by upregulating IL1RA.
Collapse
Affiliation(s)
- Ning Bai
- Neurobiology Institute of Medical Research Center, North China University of Science and Technology, Tangshan, 063210 Hebei China
| | - Quanguang Zhang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912 USA
| | - Wenli Zhang
- Neurobiology Institute of Medical Research Center, North China University of Science and Technology, Tangshan, 063210 Hebei China
| | - Bin Liu
- First Department of Neurology, Hospital Affiliated to North China University of Science and Technology, Tangshan, 063000 Hebei China
| | - Fang Yang
- Neurobiology Institute of Medical Research Center, North China University of Science and Technology, Tangshan, 063210 Hebei China
| | - Darrell Brann
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912 USA
| | - Ruimin Wang
- Neurobiology Institute of Medical Research Center, North China University of Science and Technology, Tangshan, 063210 Hebei China
- Key Laboratory of Dementia and Cognitive Disorder in Tangshan, North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, 21 Bohai Road, Caofeidian Xincheng, Tangshan, 063210 Hebei China
| |
Collapse
|
8
|
Omer M, Melo AM, Kelly L, Mac Dermott EJ, Leahy TR, Killeen O, Saugstad OD, Savani RC, Molloy EJ. Emerging Role of the NLRP3 Inflammasome and Interleukin-1β in Neonates. Neonatology 2020; 117:545-554. [PMID: 33075792 DOI: 10.1159/000507584] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/28/2020] [Indexed: 11/19/2022]
Abstract
Infection and persistent inflammation have a prominent role in the pathogenesis of brain injury and cerebral palsy, as well as other conditions associated with prematurity such as bronchopulmonary dysplasia. The NLRP3 inflammasome-interleukin (IL)-1β pathway has been extensively studied in adults and pre-clinical models, improving our understanding of innate immunity and offering an attractive therapeutic target that is already contributing to clinical management in many auto-inflammatory disorders. IL-1 blockade has transformed the course and outcome of conditions such as chronic infantile neurological, cutaneous, articular (CINCA/NOMID) syndrome. Inflammasome activation and upregulation has recently been implicated in neonatal brain and lung inflammatory disease and may be a novel therapeutic target.
Collapse
Affiliation(s)
- Murwan Omer
- Discipline of Paediatrics, Trinity College Dublin, The University of Dublin, Dublin, Ireland.,Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland
| | - Ashanty Maggvie Melo
- Discipline of Paediatrics, Trinity College Dublin, The University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute, St. James Hospital, Dublin, Ireland
| | - Lynne Kelly
- Discipline of Paediatrics, Trinity College Dublin, The University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute, St. James Hospital, Dublin, Ireland
| | - Emma Jane Mac Dermott
- Department of Paediatrics, Coombe Women's and Infant's University Hospital, Dublin, Ireland
| | - Timothy Ronan Leahy
- Department of Paediatrics, Coombe Women's and Infant's University Hospital, Dublin, Ireland
| | - Orla Killeen
- Department of Paediatrics, Coombe Women's and Infant's University Hospital, Dublin, Ireland
| | - Ola Didrik Saugstad
- Department of Pediatric Research, University of Oslo, Oslo, Norway.,Ann and Robert H. Lurie Children's Hospital of Chicago, Department of Neonatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Rashmin C Savani
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Eleanor J Molloy
- Discipline of Paediatrics, Trinity College Dublin, The University of Dublin, Dublin, Ireland, .,Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland, .,Trinity Translational Medicine Institute, St. James Hospital, Dublin, Ireland, .,Department of Paediatrics, Coombe Women's and Infant's University Hospital, Dublin, Ireland, .,Department of Immunology, Rheumatology, and Neonatology, CHI at Crumlin, Dublin, Ireland,
| |
Collapse
|
9
|
Peng T, Jiang Y, Farhan M, Lazarovici P, Chen L, Zheng W. Anti-inflammatory Effects of Traditional Chinese Medicines on Preclinical in vivo Models of Brain Ischemia-Reperfusion-Injury: Prospects for Neuroprotective Drug Discovery and Therapy. Front Pharmacol 2019; 10:204. [PMID: 30930774 PMCID: PMC6423897 DOI: 10.3389/fphar.2019.00204] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 02/18/2019] [Indexed: 12/28/2022] Open
Abstract
Acquired brain ischemia-and reperfusion-injury (IRI), including both Ischemic stroke (IS) and Traumatic Brain injury (TBI), is one of the most common causes of disability and death in adults and represents a major burden in both western and developing countries worldwide. China’s clinical neurological therapeutic experience in the use of traditional Chinese medicines (TCMs), including TCM-derived active compounds, Chinese herbs, TCM formulations and decoction, in brain IRI diseases indicated a trend of significant improvement in patients’ neurological deficits, calling for blind, placebo-controlled and randomized clinical trials with careful meta-analysis evaluation. There are many TCMs in use for brain IRI therapy in China with significant therapeutic effects in preclinical studies using different brain IRI-animal. The basic hypothesis in this field claims that in order to avoid the toxicity and side effects of the complex TCM formulas, individual isolated and identified compounds that exhibited neuroprotective properties could be used as lead compounds for the development of novel drugs. China’s efforts in promoting TCMs have contributed to an explosive growth of the preclinical research dedicated to the isolation and identification of TCM-derived neuroprotective lead compounds. Tanshinone, is a typical example of TCM-derived lead compounds conferring neuroprotection toward IRI in animals with brain middle cerebral artery occlusion (MCAO) or TBI models. Recent reports show the significance of the inflammatory response accompanying brain IRI. This response appears to contribute to both primary and secondary ischemic pathology, and therefore anti-inflammatory strategies have become popular by targeting pro-inflammatory and anti-inflammatory cytokines, other inflammatory mediators, reactive oxygen species, nitric oxide, and several transcriptional factors. Here, we review recent selected studies and discuss further considerations for critical reevaluation of the neuroprotection hypothesis of TCMs in IRI therapy. Moreover, we will emphasize several TCM’s mechanisms of action and attempt to address the most promising compounds and the obstacles to be overcome before they will enter the clinic for IRI therapy. We hope that this review will further help in investigations of neuroprotective effects of novel molecular entities isolated from Chinese herbal medicines and will stimulate performance of clinical trials of Chinese herbal medicine-derived drugs in IRI patients.
Collapse
Affiliation(s)
- Tangming Peng
- Center of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Macau, China.,Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China.,Neurosurgical Clinical Research Center of Sichuan Province, Luzhou, China
| | - Yizhou Jiang
- Center of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Mohd Farhan
- Center of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Philip Lazarovici
- Faculty of Medicine, School of Pharmacy, The Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ligang Chen
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China.,Neurosurgical Clinical Research Center of Sichuan Province, Luzhou, China
| | - Wenhua Zheng
- Center of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| |
Collapse
|
10
|
Bonapersona V, Joëls M, Sarabdjitsingh RA. Effects of early life stress on biochemical indicators of the dopaminergic system: A 3 level meta-analysis of rodent studies. Neurosci Biobehav Rev 2018; 95:1-16. [PMID: 30201218 DOI: 10.1016/j.neubiorev.2018.09.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 09/03/2018] [Accepted: 09/05/2018] [Indexed: 12/31/2022]
Abstract
Adverse early life events are a well-established risk factor for the precipitation of behavioral disorders characterized by anomalies in the dopaminergic system, such as schizophrenia and addiction. The correlation between early life conditions and the dopaminergic system has been causally investigated in more than 90 rodent publications. Here, we tested the validity of the hypothesis that early life stress (ELS) alters dopamine signaling by performing an extensive 3-level mixed effect meta-analysis. We included several ELS models and biochemical indicators of the dopaminergic system in a variety of brain areas, for a total of 1009 comparisons. Contrary to our expectations, only a few comparisons displayed a significant effect. Specifically, the striatal area was the most vulnerable, displaying decreased dopamine precursor and increased metabolites after ELS. To make all data openly accessible, we created MaDEapp (https://osf.io/w25m4/), a tool to explore data of the meta-analysis with the intent to guide future (pre)clinical research and allow power calculations. All in all, ELS induces a few yet robust changes on biochemical indicators of the dopaminergic system.
Collapse
Affiliation(s)
- V Bonapersona
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center, Utrecht University, The Netherlands.
| | - M Joëls
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center, Utrecht University, The Netherlands; University Medical Center Groningen, University of Groningen, The Netherlands
| | - R A Sarabdjitsingh
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center, Utrecht University, The Netherlands
| |
Collapse
|
11
|
Chevin M, Guiraut C, Sébire G. Effect of hypothermia on interleukin-1 receptor antagonist pharmacodynamics in inflammatory-sensitized hypoxic-ischemic encephalopathy of term newborns. J Neuroinflammation 2018; 15:214. [PMID: 30060742 PMCID: PMC6066954 DOI: 10.1186/s12974-018-1258-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/17/2018] [Indexed: 12/21/2022] Open
Abstract
Background Hypothermia is increasingly tested in several neurological conditions, such as neonatal encephalopathy, stroke, traumatic brain injury, subarachnoid hemorrhage, spinal cord injury, and neurological outcomes of cardiac arrest. Current studies aim to increase benefits of hypothermia with new add-on therapies including immunomodulatory agents. Hypothermia has been shown to affect the metabolism of commonly used drugs, including those acting on neuroimmune pathways. Objective This study focuses on the effect of hypothermia on interleukin-1 receptor antagonist pharmacodynamics in a model of neonatal encephalopathy. Methods The effect of hypothermia on (i) the tissue concentration of the interleukin-1 receptor antagonist, (ii) the interleukin-1 inflammatory cascade, and (iii) the neuroprotective potential of interleukin-1 receptor antagonist has been assessed on our rat model of neonatal encephalopathy resulting from inflammation induced by bacterial compound plus hypoxia-ischemia. Results Hypothermia reduced the surface of core and penumbra lesions, as well as alleviated the brain weight loss induced by LPS+HI exposure. Hypothermia compared to normothermia significantly increased (range 50–65%) the concentration of the interleukin-1 receptor antagonist within the central nervous system. Despite this increase of intracerebral interleukin-1 receptor antagonist concentration, the intracerebral interleukin-1-induced tumor necrosis factor-alpha cascade was upregulated. In hypothermic condition, the known neuroprotective effect of interleukin-1 receptor antagonist was neutralized (50 mg/kg/12 h for 72 h) or even reversed (200 mg/kg/12 h for 72 h) as compared to normothermic condition. Conclusion Hypothermia interferes with the pharmacodynamic parameters of the interleukin-1 receptor antagonist, through a bioaccumulation of the drug within the central nervous system and a paradoxical upregulation of the interleukin-1 pathway. These effects seem to be at the origin of the loss of efficiency or even toxicity of the interleukin-1 receptor antagonist when combined with hypothermia. Such bioaccumulation could happen similarly with the use of other drugs combined to hypothermia in a clinical context.
Collapse
Affiliation(s)
- Mathilde Chevin
- Department of Pediatrics, McGill University, Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, (Glen site, Block E, M0.3211), Montreal, Quebec, H4A 3J1, Canada
| | - Clémence Guiraut
- Department of Pediatrics, McGill University, Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, (Glen site, Block E, M0.3211), Montreal, Quebec, H4A 3J1, Canada
| | - Guillaume Sébire
- Department of Pediatrics, McGill University, Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, (Glen site, Block E, M0.3211), Montreal, Quebec, H4A 3J1, Canada.
| |
Collapse
|
12
|
Wang J, Zhang MY, Xu SQ, Cheng J, Yu ZJ, Hu XM. Down-regulation of telomerase reverse transcriptase-related anti-apoptotic function in a rat model of acrylamide induced neurobehavioral deficits. Biotech Histochem 2018; 93:512-518. [PMID: 29926741 DOI: 10.1080/10520295.2018.1471523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Although the precise mechanism is unknown, neuron apoptosis is believed to participate in neuropathy caused by acrylamide (ACR). Telomerase reverse transcriptase (TERT) exhibits an anti-apoptotic function, but its contribution to the pathogenesis of ACR neurotoxicity is unclear. We investigated adult male rats that were given 30, 40 and 50 mg/kg ACR three times/week for 4 weeks. We found that ACR treatment caused significant deficits in sensory/motor function as measured by gait score, landing foot spread distance, movement initiation test and tail immersion test. Histological examination showed that the cerebral cortex in all ACR treated animals exhibited fewer neurons and more condensed nuclei than normal cortex. A significant increase in apoptosis was found in the cerebral cortex of rat brains subjected to ACR treatment in a dose-dependent manner. The expression of TERT in the brain was significantly reduced by ACR treatment. The pro-apoptotic cleaved caspase-3 protein level was increased, while the anti-apoptotic Bcl-2 protein level was decreased by 30 - 50 mg/kg ACR. Our findings indicate that TERT and its downstream regulators of neuron apoptosis, including Bcl-2 and cleaved caspase-3, were involved in ACR neurotoxicity.
Collapse
Affiliation(s)
- J Wang
- a Department of Pharmacy , College of Medicine, Wuhan University of Science and Technology , Wuhan.,b Hubei Province Key Laboratory of Occupational Hazard Identification and Control , Wuhan University of Science and Technology , Wuhan
| | - M Y Zhang
- a Department of Pharmacy , College of Medicine, Wuhan University of Science and Technology , Wuhan.,b Hubei Province Key Laboratory of Occupational Hazard Identification and Control , Wuhan University of Science and Technology , Wuhan
| | - S Q Xu
- a Department of Pharmacy , College of Medicine, Wuhan University of Science and Technology , Wuhan.,b Hubei Province Key Laboratory of Occupational Hazard Identification and Control , Wuhan University of Science and Technology , Wuhan
| | - J Cheng
- a Department of Pharmacy , College of Medicine, Wuhan University of Science and Technology , Wuhan.,b Hubei Province Key Laboratory of Occupational Hazard Identification and Control , Wuhan University of Science and Technology , Wuhan
| | - Z J Yu
- a Department of Pharmacy , College of Medicine, Wuhan University of Science and Technology , Wuhan.,b Hubei Province Key Laboratory of Occupational Hazard Identification and Control , Wuhan University of Science and Technology , Wuhan
| | - X M Hu
- a Department of Pharmacy , College of Medicine, Wuhan University of Science and Technology , Wuhan.,b Hubei Province Key Laboratory of Occupational Hazard Identification and Control , Wuhan University of Science and Technology , Wuhan.,c College of Pharmacy , Shanghai University of Medicine & Health Sciences , Shanghai , China
| |
Collapse
|
13
|
Tsimis ME, Lei J, Rosenzweig JM, Arif H, Shabi Y, Alshehri W, Talbot CC, Baig-Ward KM, Segars J, Graham EM, Burd I. P2X7 receptor blockade prevents preterm birth and perinatal brain injury in a mouse model of intrauterine inflammation. Biol Reprod 2018; 97:230-239. [PMID: 29044426 DOI: 10.1093/biolre/iox081] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 07/25/2017] [Indexed: 01/02/2023] Open
Abstract
The P2X7 is an adenosine triphosphate (ATP)-gated ion channel involved in several facets of immune activation and neuronal function through its importance in interleukin (IL)-1β secretion. We hypothesized that blockade of P2X7 would prevent perinatal brain injury associated with exposure to intrauterine (IU) inflammation. Dams received 45 mg/kg of Brilliant Blue G (BBG), a specific P2X7 receptor (P2X7R) antagonist, on gestation day 17 (E17) prior to administration of lipopolysaccharide (LPS) or phosphate-buffered saline (PBS). Furthermore, we utilized embryo transfer experiments to delineate whether the P2X7 was the key mediator of IU inflammation-associated brain injury on maternal or fetal sides. In these experiments, P2X7-/- dams were embryo-transferred wild type embryos and wild type dams were embryo-transferred P2X7-/- embryos. In the mouse model of intrauterine inflammation, pharmacologic blockade of P2X7R reduced preterm birth rate, improved offspring performance on neuromotor tests as well as the dendritic arborization and density of cortical neurons. Embryo transfer experiments demonstrated the importance of maternal P2X7R in IU inflammation-mediated effects on offspring. Both genetic and pharmacologic blockade of IL-1β signaling, by targeting maternal P2X7R, ameliorated perinatal brain injury following exposure to IU inflammation. Specific targeting of maternal P2X7R may provide a clinically useful tool to prevent both preterm birth and prematurity-associated perinatal brain injury, and further studies are urgently needed.
Collapse
Affiliation(s)
- Michael E Tsimis
- Department of Gynecology and Obstetrics, Division of Maternal-Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jun Lei
- Department of Gynecology and Obstetrics, Division of Maternal-Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jason M Rosenzweig
- Department of Gynecology and Obstetrics, Division of Maternal-Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hattan Arif
- Department of Gynecology and Obstetrics, Division of Maternal-Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yahya Shabi
- Department of Gynecology and Obstetrics, Division of Maternal-Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wael Alshehri
- Department of Gynecology and Obstetrics, Division of Maternal-Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Connie C Talbot
- Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - K Maravet Baig-Ward
- Department of Gynecology and Obstetrics, Division of Reproductive Science and Women's Health Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - James Segars
- Department of Gynecology and Obstetrics, Division of Reproductive Science and Women's Health Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ernest M Graham
- Department of Gynecology and Obstetrics, Division of Maternal-Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Neuroscience Intensive Care Nursery Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Irina Burd
- Department of Gynecology and Obstetrics, Division of Maternal-Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Neuroscience Intensive Care Nursery Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
14
|
Schwarz AP, Trofimov AN, Zubareva OE, Lioudyno VI, Kosheverova VV, Ischenko AM, Klimenko VM. Prefrontal mRNA expression of long and short isoforms of D2 dopamine receptor: Possible role in delayed learning deficit caused by early life interleukin-1β treatment. Behav Brain Res 2017; 333:118-122. [PMID: 28673768 DOI: 10.1016/j.bbr.2017.06.052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 06/26/2017] [Accepted: 06/30/2017] [Indexed: 01/11/2023]
Abstract
Long (D2L) and short (D2S) isoform of the D2 dopamine receptor are believed to play different roles in behavioral regulation. However, little is known about differential regulation of these isoforms mRNA expression during the process of learning in physiological and pathological states. In this study, we have investigated the combined effect of training in active avoidance (AA) paradigm and chronic early life treatment with pro-inflammatory cytokine interleukin (IL)-1β (1μg/kg i.p., P15-21) on D2S and D2L dopamine receptor mRNA expression in the medial prefrontal cortex (mPFC) of adult rats. We have shown differential regulation of D2 short and long mRNA isoform expression in the mPFC. There was no effect of AA-training on D2S mRNA expression, while D2L mRNA was downregulated in AA-trained control (intact and saline-treated) animals, and this effect was not observed in rats treated with IL-1β. D2S mRNA expression level negatively correlated with learning ability within control (saline-treated and intact) groups but not in IL-1β-treated animals. Thus, prefrontal expression of distinct D2 dopamine receptor splice variants is supposed to be implicated in cognitive decline caused by early life immune challenge.
Collapse
Affiliation(s)
- Alexander P Schwarz
- Laboratory of Neurobiology of the Brain Integrative Functions, I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 St. Petersburg, Russia.
| | - Alexander N Trofimov
- Laboratory of Neurobiology of the Brain Integrative Functions, I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 St. Petersburg, Russia
| | - Olga E Zubareva
- Laboratory of Neurobiology of the Brain Integrative Functions, I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 St. Petersburg, Russia; Laboratory of Molecular Mechanisms of Neuronal Interactions, I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Thorez Avenue 44, 199223 St. Petersburg, Russia
| | - Victoria I Lioudyno
- Laboratory of Neurobiology of the Brain Integrative Functions, I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 St. Petersburg, Russia
| | - Vera V Kosheverova
- Laboratory of Intracellular Membranes Dynamics, Department of the Intracellular Signalling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Avenue 4, 194064 St. Petersburg, Russia
| | - Alexander M Ischenko
- Laboratory of Protein Biochemistry, Research Institute of Highly Pure Biopreparations, Pudozhskaya 7, 197110 St. Petersburg, Russia
| | - Victor M Klimenko
- Laboratory of Neurobiology of the Brain Integrative Functions, I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 St. Petersburg, Russia
| |
Collapse
|
15
|
Dammann O, Allred EN, Fichorova RN, Kuban K, O'Shea TM, Leviton A. Duration of Systemic Inflammation in the First Postnatal Month Among Infants Born Before the 28th Week of Gestation. Inflammation 2017; 39:672-7. [PMID: 26676589 DOI: 10.1007/s10753-015-0293-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Extremely low gestational age newborns (ELGANs, <28 completed weeks of gestation) that exhibit fetal and neonatal systemic inflammatory responses are at increased risk for developmental adversity, especially if the inflammatory process is sustained. We evaluated pro-inflammatory cytokine patterns in whole blood of 1220 ELGANs on one or more of postnatal days 1, 7, 14, 21, and 28. Protein concentrations were divided into quartiles within gestational week categories. We calculated odds ratios (OR) with 99% confidence intervals (CI) for having a concentration in the top quartile for each protein given that the infant had a protein concentration in the top quartile 1 week or more earlier compared to infants who did not. ELGANs who have elevated systemic levels of IL-6R, TNF-α, or RANTES on their first postnatal day are approximately twice as likely to have elevated levels of these cytokines at the end of each of the first postnatal month. In some, this twofold risk increase persisted for the entire first postnatal month. In extremely preterm newborns, inflammatory processes can be sustained over weeks.
Collapse
Affiliation(s)
- Olaf Dammann
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA, USA. .,Perinatal Neuropidemiology Unit, Hannover Medical School, Hannover, Germany.
| | - Elizabeth N Allred
- Neuroepidemiology Unit, Department of Neurology, Boston Children's Hospital, Harvard University, Boston, MA, USA
| | - Raina N Fichorova
- Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology, and Reproductive Biology, Brigham and Women's Hospital, Boston, MA, USA
| | - Karl Kuban
- Division of Neurology, Department of Pediatrics, Boston Medical Center and Boston University, Boston, MA, USA
| | - T Michael O'Shea
- Department of Pediatrics, Wake Forest University, Winston-Salem, NC, USA
| | - Alan Leviton
- Neuroepidemiology Unit, Department of Neurology, Boston Children's Hospital, Harvard University, Boston, MA, USA
| | | |
Collapse
|
16
|
Pierre WC, Smith PLP, Londono I, Chemtob S, Mallard C, Lodygensky GA. Neonatal microglia: The cornerstone of brain fate. Brain Behav Immun 2017; 59:333-345. [PMID: 27596692 DOI: 10.1016/j.bbi.2016.08.018] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/30/2016] [Accepted: 08/29/2016] [Indexed: 12/16/2022] Open
Abstract
Microglia, mainly known for their role in innate immunity and modulation of neuroinflammation, play an active role in central nervous system development and homeostasis. Depending on the context and environmental stimuli, microglia adopt a broad spectrum of activation status from pro-inflammatory, associated with neurotoxicity, to anti-inflammatory linked to neuroprotection. Pro-inflammatory microglial activation is a key hallmark of white matter injury in preterm infants and is involved in developmental origin of adult neurological diseases. Characterization of neonatal microglia function in brain development and inflammation has allowed the investigation of promising therapeutic targets with potential long-lasting neuroprotective effects. True prevention of neuro-degenerative diseases might eventually occur as early as the perinatal period.
Collapse
Affiliation(s)
- Wyston C Pierre
- Sainte-Justine Hospital and Research Center, Department of Pediatrics, Université de Montréal, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, Québec, Canada
| | - Peter L P Smith
- Perinatal Center, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Irène Londono
- Sainte-Justine Hospital and Research Center, Department of Pediatrics, Université de Montréal, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, Québec, Canada
| | - Sylvain Chemtob
- Sainte-Justine Hospital and Research Center, Department of Pediatrics, Université de Montréal, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, Québec, Canada; Departments of Ophtalmology, Université de Montréal, Montreal, Quebec, Canada; Departments of Pharmacology, Université de Montréal, Montreal, Quebec, Canada
| | - Carina Mallard
- Perinatal Center, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Gregory A Lodygensky
- Sainte-Justine Hospital and Research Center, Department of Pediatrics, Université de Montréal, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, Québec, Canada; Montreal Heart Institute, 5000 Rue Bélanger, Montreal, Quebec, Canada; Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada; Departments of Pharmacology, Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
17
|
Chevin M, Guiraut C, Maurice-Gelinas C, Deslauriers J, Grignon S, Sébire G. Neuroprotective effects of hypothermia in inflammatory-sensitized hypoxic-ischemic encephalopathy. Int J Dev Neurosci 2016; 55:1-8. [PMID: 27616300 DOI: 10.1016/j.ijdevneu.2016.09.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 09/05/2016] [Accepted: 09/06/2016] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Despite the recent introduction of hypothermia as a mandatory standard of care, the incidence of neonatal encephalopathy in full-term newborns and its devastating neuro-behavioral outcomes continues to be a major individual, familial and social issue. Neonatal encephalopathy is mainly due to the compounding and interacting effects of hypoxia-ischemia and inflammation resulting from placental and other perinatal infections. It is unclear why hypothermia is effective in alleviating neonatal encephalopathy in some, but not all, full-term newborns. However, newborns exposed to inflammatory-sensitized hypoxia-ischemia seem to have less therapeutic benefit from hypothermia than those exposed to hypoxia-ischemia alone. OBJECTIVES To clarify this uncertainty, we tested the efficacy of hypothermia in a double-hit model of neonatal encephalopathy induced by inflammatory-sensitized hypoxia-ischemia. METHODS Using a rat preclinical model of endotoxin plus hypoxia-ischemia-induced neonatal encephalopathy of term newborns, we assessed the following in pups exposed (or not) to hypothermia: the extent of brain injuries and the expressions of molecules implicated in neural cell death, namely: pro-inflammatory cytokines, matrix metalloproteinase-9, antioxidant enzymes, as well as receptor-interacting protein-3. RESULTS Hypothermia was neuroprotective on inflammatory-sensitized hypoxia-ischemia-induced penumbra, but not core, brain injuries. This beneficial effect was associated with a hypothermia-induced increase of antioxidant enzymes (superoxide dismutase-1, glutathione peroxidase-1), but was not associated with any variations of the other inflammatory mediators tested, namely: interleukin-1β, interleukin-1 receptor antagonist, tumor necrosis factor-α and matrix metalloproteinase-9. CONCLUSION Hypothermia is neuroprotective against inflammatory-sensitized hypoxia-ischemia possibly through a hypothermia-induced increase of antioxidant enzymes. This neuroprotective effect seems to be independent of the interleukin-1 system.
Collapse
Affiliation(s)
- Mathilde Chevin
- Département de Pédiatrie, Université de Sherbrooke, 3001- 12e avenue Nord, Sherbrooke (Quebec), J1H 5N4, Canada.
| | - Clémence Guiraut
- Department of Pediatrics, McGill University, Research Institute of the McGill University Health Centre, 1001- Decarie boulevard, Montreal (Quebec), H4A 3J1, Canada.
| | - Caroline Maurice-Gelinas
- Département de Pharmacologie-physiologie, Université de Sherbrooke, 3001- 12e avenue Nord, Sherbrooke (Quebec), J1H 5N4, Canada.
| | - Jessica Deslauriers
- Department of Psychiatry, University of California, 9500 Gilman Drive #0804 San Diego, (CA) La Jolla, 92093, United States.
| | - Sylvain Grignon
- Département de Psychiatrie, Université de Sherbrooke, 3001- 12e avenue Nord, Sherbrooke (Quebec), J1H 5N4, Canada.
| | - Guillaume Sébire
- Département de Pédiatrie, Université de Sherbrooke, 3001- 12e avenue Nord, Sherbrooke (Quebec), J1H 5N4, Canada; Department of Pediatrics, McGill University, Research Institute of the McGill University Health Centre, 1001- Decarie boulevard, Montreal (Quebec), H4A 3J1, Canada.
| |
Collapse
|
18
|
Schmidt AF, Kannan PS, Chougnet CA, Danzer SC, Miller LA, Jobe AH, Kallapur SG. Intra-amniotic LPS causes acute neuroinflammation in preterm rhesus macaques. J Neuroinflammation 2016; 13:238. [PMID: 27596440 PMCID: PMC5011884 DOI: 10.1186/s12974-016-0706-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 08/25/2016] [Indexed: 01/06/2023] Open
Abstract
Background Chorioamnionitis is associated with an increased risk of brain injury in preterm neonates. Inflammatory changes in brain could underlie this injury. Here, we evaluated whether neuroinflammation is induced by chorioamnionitis in a clinically relevant model. Methods Rhesus macaque fetuses were exposed to either intra-amniotic (IA) saline, or IA lipopolysaccharide (LPS) (1 mg) 16 or 48 h prior to delivery at 130 days (85 % of gestation) (n = 4–5 animals/group). We measured cytokines in the cerebrospinal fluid (CSF), froze samples from the left brain for molecular analysis, and immersion fixed the right brain hemisphere for immunohistology. We analyzed the messenger RNA (mRNA) levels of the pro-inflammatory cytokines IL-1β, CCL2, TNF-α, IL-6, IL-8, IL-10, and COX-2 in the periventricular white matter (PVWM), cortex, thalamus, hippocampus, and cerebellum by RT-qPCR. Brain injury was assessed by immunohistology for myelin basic protein (MBP), IBA1 (microglial marker), GFAP (astrocyte marker), OLIG2 (oligodendrocyte marker), NeuN (neuronal marker), CD3 (T cells), and CD14 (monocytes). Microglial proliferation was assessed by co-immunostaining for IBA1 and Ki67. Data were analyzed by ANOVA with Tukey’s post-test. Results IA LPS increased mRNA expression of pro-inflammatory cytokines in the PVWM, thalamus, and cerebellum, increased IL-6 concentration in the CSF, and increased apoptosis in the periventricular area after 16 h. Microglial proliferation in the white matter was increased 48 h after IA LPS. Conclusions LPS-induced chorioamnionitis caused neuroinflammation, microglial proliferation, and periventricular apoptosis in a clinically relevant model of chorioamnionitis in fetal rhesus macaques. These findings identify specific responses in the fetal brain and support the hypothesis that neuroinflammatory changes may mediate the adverse neurodevelopmental outcomes associated with chorioamnionitis. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0706-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Augusto F Schmidt
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Paranthaman S Kannan
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Claire A Chougnet
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Steve C Danzer
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Lisa A Miller
- California National Primate Research Center and Department of Pediatrics and Cell Biology and Human Anatomy, University of California, Davis, CA, USA
| | - Alan H Jobe
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Suhas G Kallapur
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA.
| |
Collapse
|
19
|
Lu S, Shaffery JP, Pang Y, Tien LT, Fan LW. Rapid Eye Movement Sleep Homeostatic Response: A Potential Marker for Early Detection of Parkinson's Disease. ACTA ACUST UNITED AC 2016; 6. [PMID: 27713856 DOI: 10.4172/2161-0460.1000255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Silu Lu
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - James P Shaffery
- Department of Psychiatry and Human Behavior, Animal Behavior Core, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Yi Pang
- 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, Xinzhuang Dist, New Taipei City 24205, Taiwan
| | - Lir-Wan Fan
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
20
|
Neonatal proinflammatory challenge in male Wistar rats: Effects on behavior, synaptic plasticity, and adrenocortical stress response. Behav Brain Res 2016; 304:1-10. [DOI: 10.1016/j.bbr.2016.02.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 01/31/2016] [Accepted: 02/01/2016] [Indexed: 12/14/2022]
|
21
|
Lv YL, Wu ZZ, Chen LX, Wu BX, Chen LL, Qin GC, Gui B, Zhou JY. Neuroprotective effects of tetrandrine against vascular dementia. Neural Regen Res 2016; 11:454-9. [PMID: 27127485 PMCID: PMC4829011 DOI: 10.4103/1673-5374.179058] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Tetrandrine is one of the major active ingredients in Menispermaceae Stephania tetrandra S. Moore, and has specific therapeutic effects in ischemic cerebrovascular disease. Its use in vascular dementia has not been studied fully. Here, we investigated whether tetrandrine would improve behavioral and cellular impairments in a two-vessel occlusion rat model of chronic vascular dementia. Eight weeks after model establishment, rats were injected intraperitoneally with 10 or 30 mg/kg tetrandrine every other day for 4 weeks. Behavioral assessment in the Morris water maze showed that model rats had longer escape latencies in training trials, and spent less time swimming in the target quadrant in probe trials, than sham-operated rats. However, rats that had received tetrandrine showed shorter escape latencies and longer target quadrant swimming time than untreated model rats. Hematoxylin-eosin and Nissl staining revealed less neuronal necrosis and pathological damage, and more living cells, in the hippocampus of rats treated with tetrandrine than in untreated model rats. Western blot assay showed that interleukin-1β expression, and phosphorylation of the N-methyl-D-aspartate 2B receptor at tyrosine 1472, were lower in model rats that received tetrandrine than in those that did not. The present findings suggest that tetrandrine may be neuroprotective in chronic vascular dementia by reducing interleukin-1β expression, N-methyl-D-aspartate receptor 2B phosphorylation at tyrosine 1472, and neuronal necrosis.
Collapse
Affiliation(s)
- Yan-Ling Lv
- Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ze-Zhi Wu
- Key Laboratory of Biorheological Science and Technology of the State Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Li-Xue Chen
- Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bai-Xue Wu
- Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lian-Lian Chen
- Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guang-Cheng Qin
- Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bei Gui
- Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ji-Ying Zhou
- Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
22
|
Asakawa T, Fang H, Sugiyama K, Nozaki T, Hong Z, Yang Y, Hua F, Ding G, Chao D, Fenoy AJ, Villarreal SJ, Onoe H, Suzuki K, Mori N, Namba H, Xia Y. Animal behavioral assessments in current research of Parkinson's disease. Neurosci Biobehav Rev 2016; 65:63-94. [PMID: 27026638 DOI: 10.1016/j.neubiorev.2016.03.016] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 03/22/2016] [Accepted: 03/22/2016] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD), a neurodegenerative disorder, is traditionally classified as a movement disorder. Patients typically suffer from many motor dysfunctions. Presently, clinicians and scientists recognize that many non-motor symptoms are associated with PD. There is an increasing interest in both motor and non-motor symptoms in clinical studies on PD patients and laboratory research on animal models that imitate the pathophysiologic features and symptoms of PD patients. Therefore, appropriate behavioral assessments are extremely crucial for correctly understanding the mechanisms of PD and accurately evaluating the efficacy and safety of novel therapies. This article systematically reviews the behavioral assessments, for both motor and non-motor symptoms, in various animal models involved in current PD research. We addressed the strengths and weaknesses of these behavioral tests and their appropriate applications. Moreover, we discussed potential mechanisms behind these behavioral tests and cautioned readers against potential experimental bias. Since most of the behavioral assessments currently used for non-motor symptoms are not particularly designed for animals with PD, it is of the utmost importance to greatly improve experimental design and evaluation in PD research with animal models. Indeed, it is essential to develop specific assessments for non-motor symptoms in PD animals based on their characteristics. We concluded with a prospective view for behavioral assessments with real-time assessment with mobile internet and wearable device in future PD research.
Collapse
Affiliation(s)
- Tetsuya Asakawa
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu-city, Shizuoka, Japan; Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu-city, Shizuoka, Japan.
| | - Huan Fang
- Department of Pharmacy, Jinshan Hospital of Fudan University, Shanghai, China
| | - Kenji Sugiyama
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu-city, Shizuoka, Japan
| | - Takao Nozaki
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu-city, Shizuoka, Japan
| | - Zhen Hong
- Department of Neurology, Huashan Hospital of Fudan University, Shanghai, China
| | - Yilin Yang
- The First People's Hospital of Changzhou, Soochow University School of Medicine, Changzhou, China
| | - Fei Hua
- The First People's Hospital of Changzhou, Soochow University School of Medicine, Changzhou, China
| | - Guanghong Ding
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China
| | - Dongman Chao
- Department of Neurosurgery, The University of Texas McGovern Medical School,Houston, TX, USA
| | - Albert J Fenoy
- Department of Neurosurgery, The University of Texas McGovern Medical School,Houston, TX, USA
| | - Sebastian J Villarreal
- Department of Neurosurgery, The University of Texas McGovern Medical School,Houston, TX, USA
| | - Hirotaka Onoe
- Functional Probe Research Laboratory, RIKEN Center for Life Science Technologies, Kobe, Japan
| | - Katsuaki Suzuki
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu-city, Shizuoka, Japan
| | - Norio Mori
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu-city, Shizuoka, Japan
| | - Hiroki Namba
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu-city, Shizuoka, Japan
| | - Ying Xia
- Department of Neurosurgery, The University of Texas McGovern Medical School,Houston, TX, USA.
| |
Collapse
|
23
|
Patil J, Matte A, Nissbrandt H, Mallard C, Sandberg M. Sustained Effects of Neonatal Systemic Lipopolysaccharide on IL-1β and Nrf2 in Adult Rat Substantia Nigra Are Partly Normalized by a Spirulina-Enriched Diet. Neuroimmunomodulation 2016; 23:250-259. [PMID: 27931028 PMCID: PMC5266664 DOI: 10.1159/000452714] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 10/19/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND/AIM Neonatal infection can sensitize the adult substantia nigra (SN) to secondary insults, causing a decrease in antioxidant capacity which may lead to Parkinson's disease in adults. We studied the prolonged effect of systemic infection by (i.p.) administration of lipopolysaccharide (LPS) on interleukin (IL)-1β, the antioxidant regulator nuclear factor-erythroid 2-related factor 2 (Nrf2), and the peroxisome proliferator-activated receptor γ coactivator (PGC)-1α in rat SN. METHOD AND RESULTS Five-day-old rat pups were treated with LPS (i.p. 2 mg/kg). After 65 days, the mRNA level of IL-1β was significantly increased, in parallel with a decrease in that of the rate-limiting enzyme in glutathione synthesis, the γ-glutamylcysteine ligase catalytic subunit (γGCLc), Nrf2, and brain-derived neurotrophic factor (BDNF). Protein levels of γGCLc and Nrf2 were decreased while IL-1β protein was significantly increased. These LPS-induced long-term changes correlated with a decrease in phosphorylated (active) AKT (pAKT) and phosphorylated (inactive) GSK-3β (pGSK-3β). In another set of experiments, a 0.1% Spirulina-containing diet was given to lactating mothers 24 h before the LPS treatment of the pups. The Spirulina-supplemented diet decreased IL-1β protein expression in SN and elevated the mRNA level of γGCLc, Nrf2 protein, PGC-1α protein, and pAKT. CONCLUSION Early-life infection can negatively affect Nrf2, pAKT, and pGSK-3β for a long time in SN. A diet enriched with antioxidant and anti-inflammatory phytochemicals can partly restore some, but not all, of the effects on the antioxidant defense, possibly via normalizing effects on pAKT.
Collapse
Affiliation(s)
- Jaspal Patil
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Sweden
| | - Ashok Matte
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Sweden
| | - Hans Nissbrandt
- Department of Pharmacology, Institute of Neuroscience and Physiology, University of Gothenburg, Sweden
| | - Carina Mallard
- Department of Physiology, Institute of Neuroscience and Physiology, University of Gothenburg, Sweden
| | - Mats Sandberg
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Sweden
| |
Collapse
|
24
|
Magnesium sulfate prevents maternal inflammation-induced impairment of learning ability and memory in rat offspring. Am J Obstet Gynecol 2015; 213:851.e1-8. [PMID: 26232507 DOI: 10.1016/j.ajog.2015.07.042] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 07/10/2015] [Accepted: 07/22/2015] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Maternal chorioamnionitis is associated with newborn neurologic injury. Recent evidence suggests that maternal administration of magnesium sulphate (MG) may protect fetuses from white matter injury. Previously we demonstrated evidence by magnetic resonance imaging that MG may prevent maternal inflammation-induced gray matter injury of offspring. Thus, we sought to determine the potential of maternal inflammation to induce fetal neurological/behavioral deficits and assess whether maternal MG attenuates these effects. STUDY DESIGN Pregnant rats at day 18 received injections of intraperitoneal lipopolysaccharide (LPS) or saline. Dams were treated with subcutaneous saline/MG (270 mg/kg followed by 27 mg/kg every 20 minutes) for 2 hours before and following LPS/saline injections. Pups were delivered spontaneously. At 1 and 3 months of age, 11-12 offspring of each group (saline, LPS, MG, LPS-MG) underwent a 2-way shuttle box avoidance testing. The shuttle box is divided in half and the animal moves between compartments to avoid an electric shock in response to an auditory stimulus. RESULTS Control offspring demonstrated significantly improved learning and memory abilities from age 1 to 3 months. At 1 month, LPS-treated dams' offspring were similar to controls with no improvement in learning abilities at 3 months. MG treatment of LPS dams significantly improved offspring learning at 3 months, to equal or better than that of controls. CONCLUSION LPS-stimulated inflammation during pregnancy impairs offspring learning ability and memory, which is ameliorated by maternal MG treatment. These results suggest that maternal MG therapy may prevent white and gray matter injuries associated with maternal infection/inflammation.
Collapse
|