51
|
Wang D, Liu K, Wake H, Teshigawara K, Mori S, Nishibori M. Anti-high mobility group box-1 (HMGB1) antibody inhibits hemorrhage-induced brain injury and improved neurological deficits in rats. Sci Rep 2017; 7:46243. [PMID: 28393932 PMCID: PMC5385548 DOI: 10.1038/srep46243] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 03/09/2017] [Indexed: 02/06/2023] Open
Abstract
As one of the most lethal stroke subtypes, intracerebral hemorrhage (ICH) is acknowledged as a serious clinical problem lacking effective treatment. Available evidence from preclinical and clinical studies suggests that inflammatory mechanisms are involved in the progression of ICH-induced secondary brain injury. High mobility group box-1 (HMGB1) is a ubiquitous and abundant nonhistone DNA-binding protein, and is also an important proinflammatory molecule once released into the extracellular space from the nuclei. Here, we show that treatment with neutralizing anti-HMGB1 mAb (1 mg/kg, i.v. twice) remarkably ameliorated ICH-injury induced by local injection of collagenase IV in the striatum of rats. Administration of anti-HMGB1 mAb inhibited the release of HMGB1 into the extracellular space in the peri-hematomal region, reduced serum HMGB1 levels and decreased brain edema by protecting blood-brain barrier integrity, in association with decreased activated microglia and the expression of inflammation-related factors at 24 h after ICH. Consequently, anti-HMGB1 mAb reduced the oxidative stress and improved the behavioral performance of rats. These results strongly indicate that HMGB1 plays a critical role in the development of ICH-induced secondary injury through the amplification of plural inflammatory responses. Intravenous injection of neutralizing anti-HMGB1 mAb has potential as a novel therapeutic strategy for ICH.
Collapse
Affiliation(s)
- Dengli Wang
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Keyue Liu
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hidenori Wake
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kiyoshi Teshigawara
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shuji Mori
- School of Pharmacy, Shujitsu University, Okayama, Japan
| | - Masahiro Nishibori
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
52
|
Deng Z, Wang Y, Zhou L, Shan Y, Tan S, Cai W, Liao S, Peng L, Lu Z. High salt-induced activation and expression of inflammatory cytokines in cultured astrocytes. Cell Cycle 2017; 16:785-794. [PMID: 28296539 DOI: 10.1080/15384101.2017.1301330] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Salt (sodium chloride, NaCl) accumulation in the brain is associated with various diseases of central nervous system (CNS). Activation of astrocytes is an important manifestation of pathophysiological processes in the CNS. However, the direct impact of high salt (HS) environment on astrocytes is unclear. In the current study, we found that high salt treatment can induce activation of astrocytes both in vivo and in vitro, manifested as morphological alteration coupled with increased expression of glial fibrillary acidic protein (GFAP). Additionally, HS upregulated the expression of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), interleukin-1β (IL-1β) and vascular endothelial growth factor (VEGF); however, its effects on transforming growth factor-β (TGF-β) expression were not evident. Furthermore, HS treatment induced increased phosphorylation of signal transducer and activator transcription 3 (STAT 3). Inhibition of Janus kinase 2 (JAK 2) by specific pharmacological antagonists, AG490, attenuated the activation of JAK2/STAT3 pathway and induction of GFAP and other pro-inflammatory factors, respectively. The results suggest that the aforementioned multiple inflammatory cytokines and mediators that may be linked to the HS induced pathogenesis of CNS via the JAK2/STAT3 signaling pathways.
Collapse
Affiliation(s)
- Zhezhi Deng
- a Department of Neurology , Third Affiliated Hospital of Sun Yat-sen University , Guangzhou , China
| | - Yuge Wang
- a Department of Neurology , Third Affiliated Hospital of Sun Yat-sen University , Guangzhou , China
| | - Li Zhou
- a Department of Neurology , Third Affiliated Hospital of Sun Yat-sen University , Guangzhou , China
| | - Yilong Shan
- a Department of Neurology , Third Affiliated Hospital of Sun Yat-sen University , Guangzhou , China
| | - Sha Tan
- a Department of Neurology , Third Affiliated Hospital of Sun Yat-sen University , Guangzhou , China
| | - Wei Cai
- a Department of Neurology , Third Affiliated Hospital of Sun Yat-sen University , Guangzhou , China
| | - Siyuan Liao
- a Department of Neurology , Third Affiliated Hospital of Sun Yat-sen University , Guangzhou , China
| | - Lisheng Peng
- a Department of Neurology , Third Affiliated Hospital of Sun Yat-sen University , Guangzhou , China
| | - Zhengqi Lu
- a Department of Neurology , Third Affiliated Hospital of Sun Yat-sen University , Guangzhou , China
| |
Collapse
|
53
|
Daniels BP, Jujjavarapu H, Durrant DM, Williams JL, Green RR, White JP, Lazear HM, Gale M, Diamond MS, Klein RS. Regional astrocyte IFN signaling restricts pathogenesis during neurotropic viral infection. J Clin Invest 2017; 127:843-856. [PMID: 28134626 PMCID: PMC5330728 DOI: 10.1172/jci88720] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 12/06/2016] [Indexed: 01/09/2023] Open
Abstract
Type I IFNs promote cellular responses to viruses, and IFN receptor (IFNAR) signaling regulates the responses of endothelial cells of the blood-brain barrier (BBB) during neurotropic viral infection. However, the role of astrocytes in innate immune responses of the BBB during viral infection of the CNS remains to be fully elucidated. Here, we have demonstrated that type I IFNAR signaling in astrocytes regulates BBB permeability and protects the cerebellum from infection and immunopathology. Mice with astrocyte-specific loss of IFNAR signaling showed decreased survival after West Nile virus infection. Accelerated mortality was not due to expanded viral tropism or increased replication. Rather, viral entry increased specifically in the hindbrain of IFNAR-deficient mice, suggesting that IFNAR signaling critically regulates BBB permeability in this brain region. Pattern recognition receptors and IFN-stimulated genes had higher basal and IFN-induced expression in human and mouse cerebellar astrocytes than did cerebral cortical astrocytes, suggesting that IFNAR signaling has brain region-specific roles in CNS immune responses. Taken together, our data identify cerebellar astrocytes as key responders to viral infection and highlight the existence of distinct innate immune programs in astrocytes from evolutionarily disparate regions of the CNS.
Collapse
Affiliation(s)
- Brian P. Daniels
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Immunology, University of Washington, Seattle, Washington, USA
| | - Harsha Jujjavarapu
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Douglas M. Durrant
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Biological Sciences, California State Polytechnic University, Pomona, California, USA
| | - Jessica L. Williams
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Richard R. Green
- Department of Immunology, University of Washington, Seattle, Washington, USA
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, Washington, USA
| | - James P. White
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Helen M. Lazear
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Michael Gale
- Department of Immunology, University of Washington, Seattle, Washington, USA
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, Washington, USA
| | - Michael S. Diamond
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Robyn S. Klein
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
54
|
Webster KM, Sun M, Crack P, O'Brien TJ, Shultz SR, Semple BD. Inflammation in epileptogenesis after traumatic brain injury. J Neuroinflammation 2017; 14:10. [PMID: 28086980 PMCID: PMC5237206 DOI: 10.1186/s12974-016-0786-1] [Citation(s) in RCA: 192] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 12/28/2016] [Indexed: 01/02/2023] Open
Abstract
Background Epilepsy is a common and debilitating consequence of traumatic brain injury (TBI). Seizures contribute to progressive neurodegeneration and poor functional and psychosocial outcomes for TBI survivors, and epilepsy after TBI is often resistant to existing anti-epileptic drugs. The development of post-traumatic epilepsy (PTE) occurs in a complex neurobiological environment characterized by ongoing TBI-induced secondary injury processes. Neuroinflammation is an important secondary injury process, though how it contributes to epileptogenesis, and the development of chronic, spontaneous seizure activity, remains poorly understood. A mechanistic understanding of how inflammation contributes to the development of epilepsy (epileptogenesis) after TBI is important to facilitate the identification of novel therapeutic strategies to reduce or prevent seizures. Body We reviewed previous clinical and pre-clinical data to evaluate the hypothesis that inflammation contributes to seizures and epilepsy after TBI. Increasing evidence indicates that neuroinflammation is a common consequence of epileptic seizure activity, and also contributes to epileptogenesis as well as seizure initiation (ictogenesis) and perpetuation. Three key signaling factors implicated in both seizure activity and TBI-induced secondary pathogenesis are highlighted in this review: high-mobility group box protein-1 interacting with toll-like receptors, interleukin-1β interacting with its receptors, and transforming growth factor-β signaling from extravascular albumin. Lastly, we consider age-dependent differences in seizure susceptibility and neuroinflammation as mechanisms which may contribute to a heightened vulnerability to epileptogenesis in young brain-injured patients. Conclusion Several inflammatory mediators exhibit epileptogenic and ictogenic properties, acting on glia and neurons both directly and indirectly influence neuronal excitability. Further research is required to establish causality between inflammatory signaling cascades and the development of epilepsy post-TBI, and to evaluate the therapeutic potential of pharmaceuticals targeting inflammatory pathways to prevent or mitigate the development of PTE.
Collapse
Affiliation(s)
- Kyria M Webster
- Department of Medicine (The Royal Melbourne Hospital), The University of Melbourne, Kenneth Myer Building, Melbourne Brain Centre, Royal Parade, Parkville, VIC, 3050, Australia
| | - Mujun Sun
- Department of Medicine (The Royal Melbourne Hospital), The University of Melbourne, Kenneth Myer Building, Melbourne Brain Centre, Royal Parade, Parkville, VIC, 3050, Australia
| | - Peter Crack
- Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, VIC, 3050, Australia
| | - Terence J O'Brien
- Department of Medicine (The Royal Melbourne Hospital), The University of Melbourne, Kenneth Myer Building, Melbourne Brain Centre, Royal Parade, Parkville, VIC, 3050, Australia
| | - Sandy R Shultz
- Department of Medicine (The Royal Melbourne Hospital), The University of Melbourne, Kenneth Myer Building, Melbourne Brain Centre, Royal Parade, Parkville, VIC, 3050, Australia
| | - Bridgette D Semple
- Department of Medicine (The Royal Melbourne Hospital), The University of Melbourne, Kenneth Myer Building, Melbourne Brain Centre, Royal Parade, Parkville, VIC, 3050, Australia.
| |
Collapse
|
55
|
Walker SJ, Beavers DP, Fortunato J, Krigsman A. A Putative Blood-Based Biomarker for Autism Spectrum Disorder-Associated Ileocolitis. Sci Rep 2016; 6:35820. [PMID: 27767057 PMCID: PMC5073317 DOI: 10.1038/srep35820] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 10/06/2016] [Indexed: 12/11/2022] Open
Abstract
Gastrointestinal symptoms are common in children with autism spectrum disorder (ASD). A significant proportion of children with ASD and gastrointestinal symptoms have histologic evidence of ileocolitis (inflammation of the terminal ileum and/or colon). We previously reported the molecular characterization of gastrointestinal biopsy tissue from ASD children with ileocolitis (ASDIC+) compared to anatomically similar inflamed tissue from typically developing children with inflammatory bowel disease (IBD; i.e. Crohn’s disease or ulcerative colitis) and typically developing children with gastrointestinal symptoms but no evidence of gastrointestinal mucosal inflammation (TDIC−). ASDIC+ children had a gene expression profile that, while primarily overlapping with known IBD, had distinctive differences. The present study confirms these findings and replicates this molecular characterization in a second cohort of cases (ASDIC+) and controls (TDIC−). In these two separate case/control mucosal-based cohorts, we have demonstrated overlap of 59 differentially expressed transcripts (DETs) unique to inflamed ileocolonic tissue from symptomatic ASDIC+ children. We now report that 9 of these 59 transcripts are also differentially expressed in the peripheral blood of the second cohort of ASDIC+ children. This set of transcripts represents a putative blood-based biomarker for ASD-associated ileocolonic inflammation.
Collapse
Affiliation(s)
- Stephen J Walker
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston Salem, NC, USA
| | - Daniel P Beavers
- Department of Biostatistical Sciences, Public Health Sciences, Wake Forest University Health Sciences, Winston Salem, NC, USA
| | - John Fortunato
- Pediatric Gastroenterology, Hepatology, and Nutrition, Ann &Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Arthur Krigsman
- Pediatric Gastroenterology Resources, 148 Beach 9th Street, Suite 2B, Far Rockaway, NY, USA
| |
Collapse
|
56
|
Kolomeets NS, Vostrikov VM, Uranova NA. [The effects of blood serum from schizophrenia patients under olanzapine monotherapy on the ultrastructure of astrocytes in human fetal brain organotypic culture]. Zh Nevrol Psikhiatr Im S S Korsakova 2016; 116:65-70. [PMID: 27240183 DOI: 10.17116/jnevro20161165165-70] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To study effects of blood serum (BS) from schizophrenia patients under olanzapine monotherapy on astrocytes in the human fetal brain organotypic culture. MATERIAL AND METHODS Authors studied the human fetal brain organotypic culture after the application of BS from 20 normals and 33 patients (ICD-10 schizophrenia, paranoid type, F20.02; F20.22) taken before and after 8 and 28 weeks of olanzapine treatment. A qualitative electron microscopic study of glial cells, neurons and neuropil as well as morphometric study of the ultrastructure of astrocytes were performed. RESULTS Authors found no effects of BS from the patients with schizophrenia on neurons and synaptic contacts. The qualitative and morphometric studies revealed different effects of BS from the patients on the astrocyte ultrastructure before and after olanzapine treatment. The application of BS from untreated schizophrenia patients induced dystrophic alterations of astrocytes, BS from patients who received olanzapine during 8 weeks did not influence the astrocyte ultrastructure. After 28 weeks of olanzapine treatment,a hypertrophy of astrocytes (an increase (р≤0.05) of the area of cells and the number of mitochondria (p=0,015) and unaltered volume density of mitochondria) was found as compared to normal control cultures. CONCLUSION BS from patients with schizophrenia before and after olanzapine treatment induced opposite types of ultrastructural changes of astrocytes in the human fetal brain organotypic culture. The differences might be due to the previously reported changes of the level of circulating immune complexes and interleukins in blood serum of schizophrenia patients and due to the effects of olanzapine on these parameters.
Collapse
Affiliation(s)
| | | | - N A Uranova
- Mental Health Research Center, Moscow, Russia
| |
Collapse
|
57
|
Pan Y, Shen B, Gao Q, Zhu J, Dong J, Zhang L, Zhang Y. Caspase-1 inhibition attenuates activation of BV2 microglia induced by LPS-treated RAW264.7 macrophages. J Biomed Res 2016; 30:225-33. [PMID: 27533933 PMCID: PMC4885171 DOI: 10.7555/jbr.30.20150141] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Revised: 11/23/2015] [Accepted: 03/13/2016] [Indexed: 11/29/2022] Open
Abstract
Neuroinflammation has been recognized as a factor in the pathogenesis of neurodegenerative diseases. Emerging evidence suggests that peripheral inflammation, besides neuroinflammation, functions as a modulator of disease progression and neuropathology in several neurodegenerative diseases. However, detailed correlations among peripheral inflammation, neuroinflammation and neurodegeneration remain unknown. In the present study, we prepared a peripheral inflammation model with lipopolysaccharides (LPS)-stimulated RAW264.7 macrophages to explore its activation on BV2 microglia. We found that LPS induced the production of IL-1β, IL-6 and TNF-α in the culture medium of RAW264.7 macrophages. We further showed that LPS plus ATP activated inflammasome, evidenced by the upregulation of caspase-1 and IL-1β, which was suppressed by ZYVAD, a caspase-1 inhibitor. Furthermore, the conditioned medium obtained from LPS-treated RAW264.7 macrophages activated BV2 microglia, stimulating the release of IL-1β, IL-6 and TNF-α from BV2 cells. ZYVAD pretreatment markedly suppressed BV2 microglia activation induced by RAW264.7 cells conditioned medium. Taken together, our study indicates that macrophage-mediated peripheral inflammation subsequently evokes neuroinflammation and may aggravate neural damage. Inflammasome and caspase-1 may be potential targets for modulating systemic inflammatory responses in neurodegenerative diseases.
Collapse
Affiliation(s)
- Yang Pan
- Department of Neurology, Nanjing First Hospital affiliated to Nanjing Medical University, Nanjing, Jiangsu 210006, China.,Department of Geriatrics, Nanjing Brain Hospital affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Bo Shen
- Department of Geriatrics, Nanjing Brain Hospital affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Qin Gao
- Department of Neurology, Nanjing First Hospital affiliated to Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Jun Zhu
- Department of Geriatrics, Nanjing Brain Hospital affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jingde Dong
- Department of Geriatrics, Nanjing Brain Hospital affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Li Zhang
- Department of Geriatrics, Nanjing Brain Hospital affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, China.
| | - Yingdong Zhang
- Department of Neurology, Nanjing First Hospital affiliated to Nanjing Medical University, Nanjing, Jiangsu 210006, China.
| |
Collapse
|
58
|
Haile M, Boutajangout A, Chung K, Chan J, Stolper T, Vincent N, Batchan M, D’Urso J, Lin Y, Kline R, Yaghmoor F, Jahfal S, Kamal R, Aljohani W, Blanck T, Bekker A, Wisniewski T. The Cox-2 Inhibitor Meloxicam Ameliorates Neuroinflammation and Depressive Behavior in Adult Mice after Splenectomy. JOURNAL OF NEUROPHYSIOLOGY AND NEUROLOGICAL DISORDERS 2016; 3:101. [PMID: 28393111 PMCID: PMC5380921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
BACKGROUND Peripheral surgical trauma may incite neuroinflammation that leads to neuronal dysfunction associated with both depression and cognitive deficits. In a previous study, we found that adult mice developed neuroinflammation and short-term working memory dysfunction in a delayed, transient manner after splenectomy that was ameliorated by the cyclooxygenase-2 inhibitor meloxicam. We tested the hypothesis that splenectomy in mice would also cause anhedonia, the diminished response to pleasure or rewarding stimuli that is a hallmark of depression, and that treatment with meloxicam would be ameliorative. METHODS After Institutional Animal Care and Use Committee approval, Swiss-Webster mice underwent sucrose preference training before being randomized into groups on day 0, when they had either splenectomy and anesthesia or anesthesia alone. Within each group, half were randomized to receive intraperitoneal saline at 24 hours, while the other half received intraperitoneal meloxicam at 24 hours. Sucrose preference ratios were determined on days 1, 5, 9, and 14. Additional mice were randomized into groups for brain histochemistry. Specimens were stained for glial fibrillary acidic protein (GFAP), a marker of astrocytes, and CD45, a protein tyrosine phosphatase that identifies microglial activation. RESULTS On day 5, mice receiving splenectomy and saline demonstrated diminished sucrose preference, which was not seen in mice receiving splenectomy and meloxicam. Semiquantitative analysis of histological slides taken from splenectomized mice treated with meloxicam revealed reduced microglial-based neuroinflammation and reactive astrocytosis compared to mice receiving saline. CONCLUSION Splenectomy in mice is associated with neuroinflammation and anhedonia, as evidenced by reactive microgliosis, astrocytosis, and behavioral changes. Postsurgical treatment with meloxicam attenuates both neuroinflammation and anhedonia. These findings suggest that cyclooxygenase-2-dependent mechanisms may play a role in the development of postoperative mood disorders, possibly via modulation of peripheral effects on neuroinflammation.
Collapse
Affiliation(s)
- Michael Haile
- Department of Anesthesiology, New York University School of Medicine,Corresponding authors: Michael Haile, NYU Langone Medical Center, Department of Anesthesiology, 550 First Avenue, Tisch 530, New York, NY 10016, Tel: (212) 263-5072; Fax: (212) 263-0664; , Thomas Wisniewski, NYU Langone Medical Center, Departments of Neurology, Pathology and Psychiatry, 450 East 29th Street, Alexandria, New York, 10016. NY, Tel: (212)263-7993; Fax: (212) 263-7528;
| | - Allal Boutajangout
- Department of Neurology, New York University School of Medicine,Department of Psychiatry, New York University School of Medicine,Department of Physiology, Neuroscience, New York, New York,King Abdulaziz University, Jeddah, Saudi Arabia
| | - Kevin Chung
- Department of Anesthesiology, New York University School of Medicine
| | - Jeffrey Chan
- Department of Anesthesiology, New York University School of Medicine
| | - Tanya Stolper
- Department of Anesthesiology, New York University School of Medicine
| | - Nemahun Vincent
- Department of Anesthesiology, New York University School of Medicine
| | - Marc Batchan
- Department of Anesthesiology, New York University School of Medicine
| | - John D’Urso
- Department of Anesthesiology, New York University School of Medicine
| | - Yan Lin
- Department of Psychiatry, New York University School of Medicine
| | - Richard Kline
- Department of Anesthesiology, New York University School of Medicine
| | - Faris Yaghmoor
- Department of Neurology, New York University School of Medicine
| | - Saad Jahfal
- Department of Neurology, New York University School of Medicine
| | - Robel Kamal
- Department of Neurology, New York University School of Medicine
| | - Waleed Aljohani
- Department of Neurology, New York University School of Medicine
| | - Thomas Blanck
- Department of Anesthesiology, New York University School of Medicine
| | - Alex Bekker
- Rutgers New Jersey Medical School, New York University School of Medicine, New Jersey
| | - Thomas Wisniewski
- Department of Neurology, New York University School of Medicine,Department of Psychiatry, New York University School of Medicine,Department of Pathology, New York University School of Medicine,Corresponding authors: Michael Haile, NYU Langone Medical Center, Department of Anesthesiology, 550 First Avenue, Tisch 530, New York, NY 10016, Tel: (212) 263-5072; Fax: (212) 263-0664; , Thomas Wisniewski, NYU Langone Medical Center, Departments of Neurology, Pathology and Psychiatry, 450 East 29th Street, Alexandria, New York, 10016. NY, Tel: (212)263-7993; Fax: (212) 263-7528;
| |
Collapse
|
59
|
Mediouni S, Jablonski J, Paris JJ, Clementz MA, Thenin-Houssier S, McLaughlin JP, Valente ST. Didehydro-cortistatin A inhibits HIV-1 Tat mediated neuroinflammation and prevents potentiation of cocaine reward in Tat transgenic mice. Curr HIV Res 2015; 13:64-79. [PMID: 25613133 DOI: 10.2174/1570162x13666150121111548] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 11/26/2014] [Accepted: 01/14/2015] [Indexed: 11/22/2022]
Abstract
HIV-1 Tat protein has been shown to have a crucial role in HIV-1-associated neurocognitive disorders (HAND), which includes a group of syndromes ranging from undetectable neurocognitive impairment to dementia. The abuse of psychostimulants, such as cocaine, by HIV infected individuals, may accelerate and intensify neurological damage. On the other hand, exposure to Tat potentiates cocaine-mediated reward mechanisms, which further promotes HAND. Here, we show that didehydro-Cortistatin A (dCA), an analog of a natural steroidal alkaloid, crosses the blood-brain barrier, cross-neutralizes Tat activity from several HIV-1 clades and decreases Tat uptake by glial cell lines. In addition, dCA potently inhibits Tat mediated dysregulation of IL-1β, TNF-α and MCP-1, key neuroinflammatory signaling proteins. Importantly, using a mouse model where doxycycline induces Tat expression, we demonstrate that dCA reverses the potentiation of cocaine-mediated reward. Our results suggest that adding a Tat inhibitor, such as dCA, to current antiretroviral therapy may reduce HIV-1-related neuropathogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Susana T Valente
- Department of Infectious diseases, The Scripps Research Institute, 130 Scripps Way, 3C1, Jupiter, FL 33458, USA.
| |
Collapse
|
60
|
Borgmann K, Ghorpade A. HIV-1, methamphetamine and astrocytes at neuroinflammatory Crossroads. Front Microbiol 2015; 6:1143. [PMID: 26579077 PMCID: PMC4621459 DOI: 10.3389/fmicb.2015.01143] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/05/2015] [Indexed: 12/30/2022] Open
Abstract
As a popular psychostimulant, methamphetamine (METH) use leads to long-lasting, strong euphoric effects. While METH abuse is common in the general population, between 10 and 15% of human immunodeficiency virus-1 (HIV-1) patients report having abused METH. METH exacerbates the severity and onset of HIV-1-associated neurocognitive disorders (HAND) through direct and indirect mechanisms. Repetitive METH use impedes adherence to antiretroviral drug regimens, increasing the likelihood of HIV-1 disease progression toward AIDS. METH exposure also directly affects both innate and adaptive immunity, altering lymphocyte numbers and activity, cytokine signaling, phagocytic function and infiltration through the blood brain barrier. Further, METH triggers the dopamine reward pathway and leads to impaired neuronal activity and direct toxicity. Concurrently, METH and HIV-1 alter the neuroimmune balance and induce neuroinflammation, which modulates a wide range of brain functions including neuronal signaling and activity, glial activation, viral infection, oxidative stress, and excitotoxicity. Pathologically, reactive gliosis is a hallmark of both HIV-1- and METH-associated neuroinflammation. Significant commonality exists in the neurotoxic mechanisms for both METH and HAND; however, the pathways dysregulated in astroglia during METH exposure are less clear. Thus, this review highlights alterations in astrocyte intracellular signaling pathways, gene expression and function during METH and HIV-1 comorbidity, with special emphasis on HAND-associated neuroinflammation. Importantly, this review carefully evaluates interventions targeting astrocytes in HAND and METH as potential novel therapeutic approaches. This comprehensive overview indicates, without a doubt, that during HIV-1 infection and METH abuse, a complex dialog between all neural cells is orchestrated through astrocyte regulated neuroinflammation.
Collapse
Affiliation(s)
- Kathleen Borgmann
- Department of Cell Biology and Immunology, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Anuja Ghorpade
- Department of Cell Biology and Immunology, University of North Texas Health Science Center Fort Worth, TX, USA
| |
Collapse
|
61
|
Mediouni S, Marcondes MCG, Miller C, McLaughlin JP, Valente ST. The cross-talk of HIV-1 Tat and methamphetamine in HIV-associated neurocognitive disorders. Front Microbiol 2015; 6:1164. [PMID: 26557111 PMCID: PMC4615951 DOI: 10.3389/fmicb.2015.01164] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/07/2015] [Indexed: 12/15/2022] Open
Abstract
Antiretroviral therapy has dramatically improved the lives of human immunodeficiency virus 1 (HIV-1) infected individuals. Nonetheless, HIV-associated neurocognitive disorders (HAND), which range from undetectable neurocognitive impairments to severe dementia, still affect approximately 50% of the infected population, hampering their quality of life. The persistence of HAND is promoted by several factors, including longer life expectancies, the residual levels of virus in the central nervous system (CNS) and the continued presence of HIV-1 regulatory proteins such as the transactivator of transcription (Tat) in the brain. Tat is a secreted viral protein that crosses the blood–brain barrier into the CNS, where it has the ability to directly act on neurons and non-neuronal cells alike. These actions result in the release of soluble factors involved in inflammation, oxidative stress and excitotoxicity, ultimately resulting in neuronal damage. The percentage of methamphetamine (MA) abusers is high among the HIV-1-positive population compared to the general population. On the other hand, MA abuse is correlated with increased viral replication, enhanced Tat-mediated neurotoxicity and neurocognitive impairments. Although several strategies have been investigated to reduce HAND and MA use, no clinically approved treatment is currently available. Here, we review the latest findings of the effects of Tat and MA in HAND and discuss a few promising potential therapeutic developments.
Collapse
Affiliation(s)
- Sonia Mediouni
- Department of Infectious Diseases, The Scripps Research Institute , Jupiter, FL, USA
| | | | - Courtney Miller
- Department of Metabolism and Aging, The Scripps Research Institute , Jupiter, FL, USA ; Department of Neuroscience, The Scripps Research Institute , Jupiter, FL, USA
| | - Jay P McLaughlin
- Department of Pharmacodynamics, University of Florida , Gainesville, FL, USA
| | - Susana T Valente
- Department of Infectious Diseases, The Scripps Research Institute , Jupiter, FL, USA
| |
Collapse
|
62
|
Prathab Balaji S, Vijay Chand C, Justin A, Ramanathan M. Telmisartan mediates anti-inflammatory and not cognitive function through PPAR-γ agonism via SARM and MyD88 signaling. Pharmacol Biochem Behav 2015; 137:60-8. [DOI: 10.1016/j.pbb.2015.08.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 08/03/2015] [Accepted: 08/05/2015] [Indexed: 10/23/2022]
|
63
|
Steardo L, Bronzuoli MR, Iacomino A, Esposito G, Steardo L, Scuderi C. Does neuroinflammation turn on the flame in Alzheimer's disease? Focus on astrocytes. Front Neurosci 2015; 9:259. [PMID: 26283900 PMCID: PMC4518161 DOI: 10.3389/fnins.2015.00259] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 07/10/2015] [Indexed: 12/18/2022] Open
Abstract
Data from animal models and Alzheimer's disease (AD) subjects provide clear evidence for an activation of inflammatory pathways during the pathogenetic course of such illness. Biochemical and neuropathological studies highlighted an important cause/effect relationship between inflammation and AD progression, revealing a wide range of genetic, cellular, and molecular changes associated with the pathology. In this context, glial cells have been proved to exert a crucial role. These cells, in fact, undergo important morphological and functional changes and are now considered to be involved in the onset and progression of AD. In particular, astrocytes respond quickly to pathology with changes that have been increasingly recognized as a continuum, with potentially beneficial and/or negative consequences. Although it is now clear that activated astrocytes trigger the neuroinflammatory process, however, the precise mechanisms have not been completely elucidated. Neuroinflammation is certainly a multi-faceted and complex phenomenon and, especially in the early stages, exerts a reparative intent. However, for reasons not yet all well known, this process goes beyond the physiologic control and contributes to the exacerbation of the damage. Here we scrutinize some evidence supporting the role of astrocytes in the neuroinflammatory process and the possibility that these cells could be considered a promising target for future AD therapies.
Collapse
Affiliation(s)
- Luca Steardo
- Department of Psychiatry, University of Naples SUNNaples, Italy
| | - Maria R. Bronzuoli
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of RomeRome, Italy
| | - Aniello Iacomino
- Faculty of Psychology, University of Rome “G. Marconi”Rome, Italy
| | - Giuseppe Esposito
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of RomeRome, Italy
| | - Luca Steardo
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of RomeRome, Italy
| | - Caterina Scuderi
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of RomeRome, Italy
| |
Collapse
|
64
|
Gustin A, Kirchmeyer M, Koncina E, Felten P, Losciuto S, Heurtaux T, Tardivel A, Heuschling P, Dostert C. NLRP3 Inflammasome Is Expressed and Functional in Mouse Brain Microglia but Not in Astrocytes. PLoS One 2015; 10:e0130624. [PMID: 26091541 PMCID: PMC4474809 DOI: 10.1371/journal.pone.0130624] [Citation(s) in RCA: 287] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 05/21/2015] [Indexed: 11/19/2022] Open
Abstract
Neuroinflammation is the local reaction of the brain to infection, trauma, toxic molecules or protein aggregates. The brain resident macrophages, microglia, are able to trigger an appropriate response involving secretion of cytokines and chemokines, resulting in the activation of astrocytes and recruitment of peripheral immune cells. IL-1β plays an important role in this response; yet its production and mode of action in the brain are not fully understood and its precise implication in neurodegenerative diseases needs further characterization. Our results indicate that the capacity to form a functional NLRP3 inflammasome and secretion of IL-1β is limited to the microglial compartment in the mouse brain. We were not able to observe IL-1β secretion from astrocytes, nor do they express all NLRP3 inflammasome components. Microglia were able to produce IL-1β in response to different classical inflammasome activators, such as ATP, Nigericin or Alum. Similarly, microglia secreted IL-18 and IL-1α, two other inflammasome-linked pro-inflammatory factors. Cell stimulation with α-synuclein, a neurodegenerative disease-related peptide, did not result in the release of active IL-1β by microglia, despite a weak pro-inflammatory effect. Amyloid-β peptides were able to activate the NLRP3 inflammasome in microglia and IL-1β secretion occurred in a P2X7 receptor-independent manner. Thus microglia-dependent inflammasome activation can play an important role in the brain and especially in neuroinflammatory conditions.
Collapse
Affiliation(s)
- Audrey Gustin
- Life Sciences Research Unit, Faculty of Science, Technology and Communication, University of Luxembourg, Luxembourg, Luxembourg
| | - Mélanie Kirchmeyer
- Life Sciences Research Unit, Faculty of Science, Technology and Communication, University of Luxembourg, Luxembourg, Luxembourg
| | - Eric Koncina
- Life Sciences Research Unit, Faculty of Science, Technology and Communication, University of Luxembourg, Luxembourg, Luxembourg
| | - Paul Felten
- Life Sciences Research Unit, Faculty of Science, Technology and Communication, University of Luxembourg, Luxembourg, Luxembourg
| | - Sophie Losciuto
- Life Sciences Research Unit, Faculty of Science, Technology and Communication, University of Luxembourg, Luxembourg, Luxembourg
| | - Tony Heurtaux
- Life Sciences Research Unit, Faculty of Science, Technology and Communication, University of Luxembourg, Luxembourg, Luxembourg
| | - Aubry Tardivel
- Biochemistry Institute, University of Lausanne, Epalinges, Switzerland
| | - Paul Heuschling
- Life Sciences Research Unit, Faculty of Science, Technology and Communication, University of Luxembourg, Luxembourg, Luxembourg
| | - Catherine Dostert
- Life Sciences Research Unit, Faculty of Science, Technology and Communication, University of Luxembourg, Luxembourg, Luxembourg
- * E-mail:
| |
Collapse
|
65
|
The opioid antagonist, β-funaltrexamine, inhibits NF-κB signaling and chemokine expression in human astrocytes and in mice. Eur J Pharmacol 2015; 762:193-201. [PMID: 26007645 DOI: 10.1016/j.ejphar.2015.05.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 05/19/2015] [Accepted: 05/20/2015] [Indexed: 01/12/2023]
Abstract
Opioid-immune crosstalk occurs when opioid drugs alter the activity of the immune system. In this study, the opioid antagonist β-funaltrexamine (β-FNA) decreases the expression and release of an inflammatory chemokine, interferon-γ inducible protein-10 (CXCL10) from normal human astrocytes stimulated by interleukin 1β (IL-1β). β-FNA decreased CXCL10 by an unknown action that did not involve the mu opioid receptor (MOR). As IL-1β acts through its receptor to activate NF-κB/MAPK signaling pathways which leads to CXCL10 expression and release, key steps in the IL-1β signaling pathways were examined following β-FNA treatment. IL-1β-induced activation of p38 mitogen-activated protein kinases (p38 MAPK) was inhibited by β-FNA as shown by decreased p38 MAPK phosphorylation in treated cells. β-FNA also decreased the levels of activated subunits of NF-κB (p50 and p65) in treated astrocytes. The impact of β-FNA was also observed in proteins that act to negatively regulate NF-κB signaling. IL-1β upregulated the expression of A20, a ubiquitin (Ub)-editing enzyme that dampens NF-κB signaling by altering ubiquination patterns on IL-1 receptor second messengers, and the increase in A20 was significantly inhibited by β-FNA treatment. Inhibition of the Ub-activating enzyme E1 by the inhibitor PYR41 also decreased CXCL10 release, like β-FNA, and concurrent treatment with both PYR41 and β-FNA inhibited CXCL10 more than did either agent alone. In mice, lipopolysaccharide-induced CXCL10 expression in the brain was inhibited by treatment with β-FNA. These findings suggest that β-FNA exerts an anti-inflammatory action in vitro and in vivo that is MOR-independent and possibly due to the alkylating ability of β-FNA.
Collapse
|
66
|
Lee KM, MacLean AG. New advances on glial activation in health and disease. World J Virol 2015; 4:42-55. [PMID: 25964871 PMCID: PMC4419121 DOI: 10.5501/wjv.v4.i2.42] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 01/23/2015] [Accepted: 02/11/2015] [Indexed: 02/05/2023] Open
Abstract
In addition to being the support cells of the central nervous system (CNS), astrocytes are now recognized as active players in the regulation of synaptic function, neural repair, and CNS immunity. Astrocytes are among the most structurally complex cells in the brain, and activation of these cells has been shown in a wide spectrum of CNS injuries and diseases. Over the past decade, research has begun to elucidate the role of astrocyte activation and changes in astrocyte morphology in the progression of neural pathologies, which has led to glial-specific interventions for drug development. Future therapies for CNS infection, injury, and neurodegenerative disease are now aimed at targeting astrocyte responses to such insults including astrocyte activation, astrogliosis and other morphological changes, and innate and adaptive immune responses.
Collapse
|
67
|
Interactions of early adversity with stress-related gene polymorphisms impact regional brain structure in females. Brain Struct Funct 2015; 221:1667-79. [PMID: 25630611 DOI: 10.1007/s00429-015-0996-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 01/21/2015] [Indexed: 12/17/2022]
Abstract
Early adverse life events (EALs) have been associated with regional thinning of the subgenual cingulate cortex (sgACC), a brain region implicated in the development of disorders of mood and affect, and often comorbid functional pain disorders, such as irritable bowel syndrome (IBS). Regional neuroinflammation related to chronic stress system activation has been suggested as a possible mechanism underlying these neuroplastic changes. However, the interaction of genetic and environmental factors in these changes is poorly understood. The current study aimed to evaluate the interactions of EALs and candidate gene polymorphisms in influencing thickness of the sgACC. 210 female subjects (137 healthy controls; 73 IBS) were genotyped for stress and inflammation-related gene polymorphisms. Genetic variation with EALs, and diagnosis on sgACC thickness was examined, while controlling for race, age, and total brain volume. Compared to HCs, IBS had significantly reduced sgACC thickness (p = 0.03). Regardless of disease group (IBS vs. HC), thinning of the left sgACC was associated with a significant gene-gene environment interaction between the IL-1β genotype, the NR3C1 haplotype, and a history of EALs (p = 0.05). Reduced sgACC thickness in women with the minor IL-1β allele, was associated with EAL total scores regardless of NR3C1 haplotype status (p = 0.02). In subjects homozygous for the major IL-1β allele, reduced sgACC with increasing levels of EALs was seen only with the less common NR3C1 haplotype (p = 0.02). These findings support an interaction between polymorphisms related to stress and inflammation and early adverse life events in modulating a key region of the emotion arousal circuit.
Collapse
|
68
|
Caiazzo M, Giannelli S, Valente P, Lignani G, Carissimo A, Sessa A, Colasante G, Bartolomeo R, Massimino L, Ferroni S, Settembre C, Benfenati F, Broccoli V. Direct conversion of fibroblasts into functional astrocytes by defined transcription factors. Stem Cell Reports 2014; 4:25-36. [PMID: 25556566 PMCID: PMC4297873 DOI: 10.1016/j.stemcr.2014.12.002] [Citation(s) in RCA: 168] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 11/28/2014] [Accepted: 12/01/2014] [Indexed: 01/04/2023] Open
Abstract
Direct cell reprogramming enables direct conversion of fibroblasts into functional neurons and oligodendrocytes using a minimal set of cell-lineage-specific transcription factors. This approach is rapid and simple, generating the cell types of interest in one step. However, it remains unknown whether this technology can be applied to convert fibroblasts into astrocytes, the third neural lineage. Astrocytes play crucial roles in neuronal homeostasis, and their dysfunctions contribute to the origin and progression of multiple human diseases. Herein, we carried out a screening using several transcription factors involved in defining the astroglial cell fate and identified NFIA, NFIB, and SOX9 to be sufficient to convert with high efficiency embryonic and postnatal mouse fibroblasts into astrocytes (iAstrocytes). We proved both by gene-expression profiling and functional tests that iAstrocytes are comparable to native brain astrocytes. This protocol can be then employed to generate functional iAstrocytes for a wide range of experimental applications. NFIA, NFIB, and SOX9 reprogram fibroblasts into induced astrocytes (iAstrocytes) iAstrocytes reprogramming induces a global change in gene-expression profiling iAstrocytes are functionally comparable to native astrocytes NFIA, NFIB, and SOX9 induce an astrocytic phenotype in human fibroblasts
Collapse
Affiliation(s)
- Massimiliano Caiazzo
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy.
| | - Serena Giannelli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Pierluigi Valente
- Section of Physiology, Department of Experimental Medicine, University of Genoa and National Institute of Neuroscience, 16132 Genoa, Italy
| | - Gabriele Lignani
- Department of Neuroscience and Brain Technologies, Italian Institute of Technology, 16132 Genoa, Italy
| | | | - Alessandro Sessa
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Gaia Colasante
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Rosa Bartolomeo
- Telethon Institute of Genetics and Medicine, Naples 80131, Italy; Dulbecco Telethon Institute
| | - Luca Massimino
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Stefano Ferroni
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Carmine Settembre
- Telethon Institute of Genetics and Medicine, Naples 80131, Italy; Dulbecco Telethon Institute; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Medical Genetics, Department of Medical and Translational Science Unit, Federico II University, Via Pansini 5, 80131 Naples, Italy
| | - Fabio Benfenati
- Section of Physiology, Department of Experimental Medicine, University of Genoa and National Institute of Neuroscience, 16132 Genoa, Italy; Department of Neuroscience and Brain Technologies, Italian Institute of Technology, 16132 Genoa, Italy
| | - Vania Broccoli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy.
| |
Collapse
|
69
|
Donzis EJ, Tronson NC. Modulation of learning and memory by cytokines: signaling mechanisms and long term consequences. Neurobiol Learn Mem 2014; 115:68-77. [PMID: 25151944 PMCID: PMC4250287 DOI: 10.1016/j.nlm.2014.08.008] [Citation(s) in RCA: 193] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 08/12/2014] [Accepted: 08/13/2014] [Indexed: 11/16/2022]
Abstract
This review describes the role of cytokines and their downstream signaling cascades on the modulation of learning and memory. Immune proteins are required for many key neural processes and dysregulation of these functions by systemic inflammation can result in impairments of memory that persist long after the resolution of inflammation. Recent research has demonstrated that manipulations of individual cytokines can modulate learning, memory, and synaptic plasticity. The many conflicting findings, however, have prevented a clear understanding of the precise role of cytokines in memory. Given the complexity of inflammatory signaling, understanding its modulatory role requires a shift in focus from single cytokines to a network of cytokine interactions and elucidation of the cytokine-dependent intracellular signaling cascades. Finally, we propose that whereas signal transduction and transcription may mediate short-term modulation of memory, long-lasting cellular and molecular mechanisms such as epigenetic modifications and altered neurogenesis may be required for the long lasting impact of inflammation on memory and cognition.
Collapse
Affiliation(s)
- Elissa J Donzis
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Natalie C Tronson
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
70
|
Azizi G, Khannazer N, Mirshafiey A. The Potential Role of Chemokines in Alzheimer's Disease Pathogenesis. Am J Alzheimers Dis Other Demen 2014; 29:415-25. [PMID: 24408754 PMCID: PMC10852600 DOI: 10.1177/1533317513518651] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder and leading cause of dementia, which begins with impaired memory. The neuropathological hallmarks of AD include destructive alterations of neurons by neurofibrillary tangles, neuritic amyloid plaques, and neuroinflammatory process in the brain. Chemokines have a major role in inflammatory cell attraction and glial cell activation and/or modulation in the central nervous system. Moreover, the clinical and immunopathological evidence could show dual key role of chemokines in their pro- and anti-inflammatory properties in AD. However, their effects in neurodegeneration and/or neuroprotection remain an area of investigation. This review article provides an overview of characteristic, cellular source and activity of chemokines, and their roles in neuronal glial cell interaction in AD.
Collapse
Affiliation(s)
- Gholamreza Azizi
- Imam Hassan Mojtaba Hospital, Alborz University of Medical Sciences, Karaj, Iran
| | - Nikoo Khannazer
- Department of Molecular and Cellular Biology, College of Science, University of Tehran, Tehran, Iran
| | - Abbas Mirshafiey
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
71
|
Vartak-Sharma N, Gelman BB, Joshi C, Borgamann K, Ghorpade A. Astrocyte elevated gene-1 is a novel modulator of HIV-1-associated neuroinflammation via regulation of nuclear factor-κB signaling and excitatory amino acid transporter-2 repression. J Biol Chem 2014; 289:19599-612. [PMID: 24855648 DOI: 10.1074/jbc.m114.567644] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Astrocyte elevated gene-1 (AEG-1), a novel human immunodeficiency virus (HIV)-1 and tumor necrosis factor (TNF)-α-inducible oncogene, has generated significant interest in the field of cancer research as a therapeutic target for many metastatic aggressive tumors. However, little is known about its role in astrocyte responses during HIV-1 central nervous system (CNS) infection and whether it contributes toward the development of HIV-associated neurocognitive disorders (HAND). Therefore, in this study, we investigated changes in AEG-1 CNS expression in HIV-1-infected brain tissues and elucidated a potential mechanism of AEG-1-mediated regulation of HAND. Immunoblotting and immunohistochemical analyses of HIV-1 seropositive and HIV-1 encephalitic human brain tissues revealed significantly elevated levels of AEG-1 protein. Immunohistochemical analyses of HIV-1 Tat transgenic mouse brain tissues also showed a marked increase in AEG-1 staining. Similar to in vivo observations, cultured astrocytes expressing HIV-1 Tat also revealed AEG-1 and cytokine up-regulation. Astrocytes treated with HAND-relevant stimuli, TNF-α, interleukin (IL)-1β, and HIV-1, also significantly induced AEG-1 expression and nuclear translocation via activation of the nuclear factor (NF)-κB pathway. Co-immunoprecipitation studies demonstrated IL-1β- or TNF-α-induced AEG-1 interaction with NF-κB p65 subunit. AEG-1 knockdown decreased NF-κB activation, nuclear translocation, and transcriptional output in TNF-α-treated astrocytes. Moreover, IL-1β treatment of AEG-1-overexpressing astrocytes significantly lowered expression of excitatory amino acid transporter 2, increased expression of excitatory amino acid transporter 2 repressor ying yang 1, and reduced glutamate clearance, a major transducer of excitotoxic neuronal damage. Findings from this study identify a novel transcriptional co-factor function of AEG-1 and further implicate AEG-1 in HAND-associated neuroinflammation.
Collapse
Affiliation(s)
- Neha Vartak-Sharma
- From the Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas 76107 and
| | - Benjamin B Gelman
- the Departments of Pathology and Neuroscience & Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Chaitanya Joshi
- From the Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas 76107 and
| | - Kathleen Borgamann
- From the Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas 76107 and
| | - Anuja Ghorpade
- From the Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas 76107 and
| |
Collapse
|
72
|
Waknine-Grinberg JH, Even-Chen S, Avichzer J, Turjeman K, Bentura-Marciano A, Haynes RK, Weiss L, Allon N, Ovadia H, Golenser J, Barenholz Y. Glucocorticosteroids in nano-sterically stabilized liposomes are efficacious for elimination of the acute symptoms of experimental cerebral malaria. PLoS One 2013; 8:e72722. [PMID: 23991146 PMCID: PMC3753236 DOI: 10.1371/journal.pone.0072722] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 07/12/2013] [Indexed: 01/07/2023] Open
Abstract
Cerebral malaria is the most severe complication of Plasmodium falciparum infection, and a leading cause of death in children under the age of five in malaria-endemic areas. We report high therapeutic efficacy of a novel formulation of liposome-encapsulated water-soluble glucocorticoid prodrugs, and in particular β-methasone hemisuccinate (BMS), for treatment of experimental cerebral malaria (ECM), using the murine P. berghei ANKA model. BMS is a novel derivative of the potent steroid β-methasone, and was specially synthesized to enable remote loading into nano-sterically stabilized liposomes (nSSL), to form nSSL-BMS. The novel nano-drug, composed of nSSL remote loaded with BMS, dramatically improves drug efficacy and abolishes the high toxicity seen upon administration of free BMS. nSSL-BMS reduces ECM rates in a dose-dependent manner and creates a survival time-window, enabling administration of an antiplasmodial drug, such as artemisone. Administration of artemisone after treatment with the nSSL-BMS results in complete cure. Treatment with BMS leads to lower levels of cerebral inflammation, demonstrated by changes in cytokines, chemokines, and cell markers, as well as diminished hemorrhage and edema, correlating with reduced clinical score. Administration of the liposomal formulation results in accumulation of BMS in the brains of sick mice but not of healthy mice. This steroidal nano-drug effectively eliminates the adverse effects of the cerebral syndrome even when the treatment is started at late stages of disease, in which disruption of the blood-brain barrier has occurred and mice show clear signs of neurological impairment. Overall, sequential treatment with nSSL-BMS and artemisone may be an efficacious and well-tolerated therapy for prevention of CM, elimination of parasites, and prevention of long-term cognitive damage.
Collapse
Affiliation(s)
- Judith H. Waknine-Grinberg
- Laboratory of Membrane and Liposome Research, Department of Biochemistry, Institute for Medical Research – Israel-Canada (IMRIC), The Hebrew University - Hadassah Medical School, Jerusalem, Israel
- Department of Microbiology and Molecular Genetics, The Kuvin Center for the Study of Infectious and Tropical Diseases, The Hebrew University - Hadassah Medical School, Jerusalem, Israel
| | - Simcha Even-Chen
- Laboratory of Membrane and Liposome Research, Department of Biochemistry, Institute for Medical Research – Israel-Canada (IMRIC), The Hebrew University - Hadassah Medical School, Jerusalem, Israel
| | - Jasmine Avichzer
- Agnes Ginges Center for Human Neurogenetics, Department of Neurology, Hadassah University Hospital, Jerusalem, Israel
| | - Keren Turjeman
- Laboratory of Membrane and Liposome Research, Department of Biochemistry, Institute for Medical Research – Israel-Canada (IMRIC), The Hebrew University - Hadassah Medical School, Jerusalem, Israel
| | - Annael Bentura-Marciano
- Department of Microbiology and Molecular Genetics, The Kuvin Center for the Study of Infectious and Tropical Diseases, The Hebrew University - Hadassah Medical School, Jerusalem, Israel
| | - Richard K. Haynes
- Department of Chemistry, Institute of Molecular Technology for Drug Discovery and Synthesis, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Lola Weiss
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Hadassah University Hospital, Jerusalem, Israel
| | - Nahum Allon
- Laboratory of Membrane and Liposome Research, Department of Biochemistry, Institute for Medical Research – Israel-Canada (IMRIC), The Hebrew University - Hadassah Medical School, Jerusalem, Israel
| | - Haim Ovadia
- Agnes Ginges Center for Human Neurogenetics, Department of Neurology, Hadassah University Hospital, Jerusalem, Israel
| | - Jacob Golenser
- Department of Microbiology and Molecular Genetics, The Kuvin Center for the Study of Infectious and Tropical Diseases, The Hebrew University - Hadassah Medical School, Jerusalem, Israel
| | - Yechezkel Barenholz
- Laboratory of Membrane and Liposome Research, Department of Biochemistry, Institute for Medical Research – Israel-Canada (IMRIC), The Hebrew University - Hadassah Medical School, Jerusalem, Israel
- * E-mail: (YB), (JG)
| |
Collapse
|
73
|
Meraz-Ríos MA, Toral-Rios D, Franco-Bocanegra D, Villeda-Hernández J, Campos-Peña V. Inflammatory process in Alzheimer's Disease. Front Integr Neurosci 2013; 7:59. [PMID: 23964211 PMCID: PMC3741576 DOI: 10.3389/fnint.2013.00059] [Citation(s) in RCA: 278] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Accepted: 07/25/2013] [Indexed: 12/26/2022] Open
Abstract
Alzheimer Disease (AD) is a neurodegenerative disorder and the most common form of dementia. Histopathologically is characterized by the presence of two major hallmarks, the intracellular neurofibrillary tangles (NFTs) and extracellular neuritic plaques (NPs) surrounded by activated astrocytes and microglia. NFTs consist of paired helical filaments of truncated tau protein that is abnormally hyperphosphorylated. The main component in the NP is the amyloid-β peptide (Aβ), a small fragment of 40–42 amino acids with a molecular weight of 4 kD. It has been proposed that the amyloid aggregates and microglia activation are able to favor the neurodegenerative process observed in AD patients. However, the role of inflammation in AD is controversial, because in early stages the inflammation could have a beneficial role in the pathology, since it has been thought that the microglia and astrocytes activated could be involved in Aβ clearance. Nevertheless the chronic activation of the microglia has been related with an increase of Aβ and possibly with tau phosphorylation. Studies in AD brains have shown an upregulation of complement molecules, pro-inflammatory cytokines, acute phase reactants and other inflammatory mediators that could contribute with the neurodegenerative process. Clinical trials and animal models with non-steroidal anti-inflammatory drugs (NSAIDs) indicate that these drugs may decrease the risk of developing AD and apparently reduce Aβ deposition. Finally, further studies are needed to determine whether treatment with anti-inflammatory strategies, may decrease the neurodegenerative process that affects these patients.
Collapse
Affiliation(s)
- Marco A Meraz-Ríos
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados Mexico City, Mexico
| | | | | | | | | |
Collapse
|
74
|
Liu MH, Lin YS, Sheu SY, Sun JS. Anti-inflammatory effects of daidzein on primary astroglial cell culture. Nutr Neurosci 2013; 12:123-34. [DOI: 10.1179/147683009x423274] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
75
|
Wyss-Coray T, Rogers J. Inflammation in Alzheimer disease-a brief review of the basic science and clinical literature. Cold Spring Harb Perspect Med 2013; 2:a006346. [PMID: 22315714 DOI: 10.1101/cshperspect.a006346] [Citation(s) in RCA: 695] [Impact Index Per Article: 57.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Biochemical and neuropathological studies of brains from individuals with Alzheimer disease (AD) provide clear evidence for an activation of inflammatory pathways, and long-term use of anti-inflammatory drugs is linked with reduced risk to develop the disease. As cause and effect relationships between inflammation and AD are being worked out, there is a realization that some components of this complex molecular and cellular machinery are most likely promoting pathological processes leading to AD, whereas other components serve to do the opposite. The challenge will be to find ways of fine tuning inflammation to delay, prevent, or treat AD.
Collapse
Affiliation(s)
- Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305-5235, USA; Geriatric Research Education and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304, USA
| | | |
Collapse
|
76
|
Ljubimova JY, Kleinman MT, Karabalin NM, Inoue S, Konda B, Gangalum P, Markman JL, Ljubimov AV, Black KL. Gene expression changes in rat brain after short and long exposures to particulate matter in Los Angeles basin air: Comparison with human brain tumors. ACTA ACUST UNITED AC 2013; 65:1063-71. [PMID: 23688656 DOI: 10.1016/j.etp.2013.04.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 04/13/2013] [Accepted: 04/19/2013] [Indexed: 12/19/2022]
Abstract
Air pollution negatively impacts pulmonary, cardiovascular, and central nervous systems. Although its influence on brain cancer is unclear, toxic pollutants can cause blood-brain barrier disruption, enabling them to reach the brain and cause alterations leading to tumor development. By gene microarray analysis validated by quantitative RT-PCR and immunostaining we examined whether rat (n=104) inhalation exposure to air pollution particulate matter (PM) resulted in brain molecular changes similar to those associated with human brain tumors. Global brain gene expression was analyzed after exposure to PM (coarse, 2.5-10μm; fine, <2.5μm; or ultrafine, <0.15μm) and purified air for different times, short (0.5, 1, and 3 months) and chronic (10 months), for 5h per day, four days per week. Expression of select gene products was also studied in human brain (n=7) and in tumors (n=83). Arc/Arg3.1 and Rac1 genes, and their protein products were selected for further examination. Arc was elevated upon two-week to three-month exposure to coarse PM and declined after 10-month exposure. Rac1 was significantly elevated upon 10-month coarse PM exposure. On human brain tumor sections, Arc was expressed in benign meningiomas and low-grade gliomas but was much lower in high-grade tumors. Conversely, Rac1 was elevated in high-grade vs. low-grade gliomas. Arc is thus associated with early brain changes and low-grade tumors, whereas Rac1 is associated with long-term PM exposure and highly aggressive tumors. In summary, exposure to air PM leads to distinct changes in rodent brain gene expression similar to those observed in human brain tumors.
Collapse
Affiliation(s)
- Julia Y Ljubimova
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States.
| | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Guha D, Nagilla P, Redinger C, Srinivasan A, Schatten GP, Ayyavoo V. Neuronal apoptosis by HIV-1 Vpr: contribution of proinflammatory molecular networks from infected target cells. J Neuroinflammation 2012; 9:138. [PMID: 22727020 PMCID: PMC3425332 DOI: 10.1186/1742-2094-9-138] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 05/19/2012] [Indexed: 01/13/2023] Open
Abstract
Background Human immunodeficiency virus type 1 (HIV-1) induces neuronal dysfunction through host cellular factors and viral proteins including viral protein R (Vpr) released from infected macrophages/microglia. Vpr is important for infection of terminally differentiated cells such as macrophages. The objective of this study was to assess the effect of Vpr in the context of infectious virus particles on neuronal death through proinflammatory cytokines released from macrophages. Methods Monocyte-derived macrophages (MDM) were infected with either HIV-1 wild type (HIV-1wt), Vpr deleted mutant (HIV-1∆Vpr) or mock. Cell lysates and culture supernatants from MDMs were analyzed for the expression and release of proinflammatory cytokines by quantitative reverse transcription-PCR and enzyme-linked immunosorbent assay respectively. Mitogen-activated protein kinases (MAPK) were analyzed in activated MDMs by western blots. Further, the effect of Vpr on neuronal apoptosis was examined using primary neurons exposed to culture supernatants from HIV-1wt, HIV-1∆Vpr or mock-infected MDMs by Annexin-V staining, MTT and Caspase - Glo® 3/7 assays. The role of interleukin (IL)-1β, IL-8 and tumor necrosis factor (TNF)-α on neuronal apoptosis was also evaluated in the presence or absence of neutralizing antibodies against these cytokines. Results HIV-1∆Vpr-infected MDMs exhibited reduced infection over time and specifically a significant downregulation of IL-1β, IL-8 and TNF-α at the transcriptional and/or protein levels compared to HIV-1wt-infected cultures. This downregulation was due to impaired activation of p38 and stress-activated protein kinase (SAPK)/c-Jun N-terminal kinase (JNK) in HIV-1∆Vpr-infected MDMs. The association of SAPK/JNK and p38 to IL-1β and IL-8 production was confirmed by blocking MAPKs that prevented the elevation of IL-1β and IL-8 in HIV-1wt more than in HIV-1∆Vpr-infected cultures. Supernatants from HIV-1∆Vpr-infected MDMs containing lower concentrations of IL-1β, IL-8 and TNF-α as well as viral proteins showed a reduced neurotoxicity compared to HIV-1wt-infected MDM supernatants. Reduction of neuronal death in the presence of anti-IL-1β and anti-IL-8 antibodies only in HIV-1wt-infected culture implies that the effect of Vpr on neuronal death is in part mediated through released proinflammatory factors. Conclusion Collectively, these results demonstrate the ability of HIV-1∆Vpr to restrict neuronal apoptosis through dysregulation of multiple proinflammatory cytokines in the infected target cells either directly or indirectly by suppressing viral replication.
Collapse
Affiliation(s)
- Debjani Guha
- Department of Infectious Diseases & Microbiology, Graduate School of Public Health, University of Pittsburgh, 130 DeSoto Street, Pittsburgh, PA 15261, USA
| | | | | | | | | | | |
Collapse
|
78
|
Schäfer S, Calas AG, Vergouts M, Hermans E. Immunomodulatory influence of bone marrow-derived mesenchymal stem cells on neuroinflammation in astrocyte cultures. J Neuroimmunol 2012; 249:40-8. [PMID: 22633273 DOI: 10.1016/j.jneuroim.2012.04.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 04/23/2012] [Accepted: 04/26/2012] [Indexed: 11/28/2022]
Abstract
The therapeutic benefits associated with mesenchymal stem cells (MSCs) largely result from their immunomodulatory and neurotrophic properties. In this study, we evaluated the effects of MSCs on astrocyte cultures exposed to lipopolysaccharide. In response to this inflammatory trigger, astrocytes showed an increased expression of pro-inflammatory genes (IL-1β, TNFα, IL-6), which was attenuated by pre-exposure to MSC conditioned medium. Furthermore, mediators released by MSCs increased cell proliferation and altered the regulation of intermediate filaments (GFAP, vimentin), pro-inflammatory enzymes (iNOS, COX-2) and receptors (TLR4, CD14, mGluR3, mGluR5). These data demonstrate that MSCs influence diverse cell types participating in the response to neuroinflammation.
Collapse
Affiliation(s)
- Sabrina Schäfer
- Institute of Neuroscience (IoNS), Group of Neuropharmacology, Université catholique de Louvain, Av. Hippocrate 54, Brussels, Belgium
| | | | | | | |
Collapse
|
79
|
Wang J, Li G, Wang Z, Zhang X, Yao L, Wang F, Liu S, Yin J, Ling EA, Wang L, Hao A. High glucose-induced expression of inflammatory cytokines and reactive oxygen species in cultured astrocytes. Neuroscience 2011; 202:58-68. [PMID: 22178606 DOI: 10.1016/j.neuroscience.2011.11.062] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 11/25/2011] [Accepted: 11/28/2011] [Indexed: 11/16/2022]
Abstract
Astrocyte activation plays important roles both in physiological and pathological process in the CNS. In the latter, the process is further aggravated by hyperglycemia, leading to diabetes complications of CNS. We report here that high glucose (HG) treatment stimulated astrocytic morphological alteration coupled with changes in glial fibrillary acidic protein (GFAP) and vimentin expression. Additionally, HG upregulated the expression of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), interleukin-1β (IL-1β), interleukin-4 (IL-4), and vascular endothelial growth factor (VEGF); however, its effects on transforming growth factor-β (TGF-β) expression were not evident. HG treatment induced increased production of reactive oxygen species (ROS) as well as activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and signal transducer and activator transcription 3 (STAT 3). HG-induced expression of TNF-α, IL-6, IL-1β, IL-4, and VEGF was blocked by ROS scavenger and inhibitors specific for NF-κB and STAT 3, respectively. The results suggest that the aforementioned multiple inflammatory cytokines and mediators that may be linked to the pathogenesis of the diabetes complications of CNS are induced by HG via the key signaling pathways.
Collapse
Affiliation(s)
- J Wang
- Department of Neurosurgery, Qi Lu Hospital, Shandong University, No. 107, Wenhua Xi Road, Jinan, Shandong, 250012, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Miljković D, Timotijević G, Stojković MM. Astrocytes in the tempest of multiple sclerosis. FEBS Lett 2011; 585:3781-8. [DOI: 10.1016/j.febslet.2011.03.047] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2011] [Revised: 03/21/2011] [Accepted: 03/23/2011] [Indexed: 12/11/2022]
|
81
|
Freeman LR, Small BJ, Bickford PC, Umphlet C, Granholm AC. A high-fat/high-cholesterol diet inhibits growth of fetal hippocampal transplants via increased inflammation. Cell Transplant 2011; 20:1499-514. [PMID: 21396159 PMCID: PMC4830280 DOI: 10.3727/096368910x557281] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
A diet containing high levels of saturated fat and cholesterol is detrimental to many aspects of health and is known to lead to obesity, metabolic syndrome, heart disease, diabetes, and cancer. However, the effects of a diet rich in saturated fat and cholesterol on the brain are not currently well understood. In order to determine direct effects of a high saturated fat and cholesterol diet upon fetal hippocampal tissue, we transplanted hippocampal grafts from embryonic day 18 rats to the anterior eye chamber of 16-month-old host animals that were fed either a normal rat chow diet or a 10% hydrogenated coconut oil + 2% cholesterol diet (HFHC diet) for 8 weeks. One eye per rat received topical application of an IL-1 receptor antagonist (IL-1Ra, Kineret®) and the other served as a saline control. Results revealed that the HFHC diet led to a marked reduction in hippocampal transplant growth, and detrimental effects of the diet were alleviated by the IL-1 receptor antagonist IL-1Ra. Graft morphology demonstrated that the HFHC diet reduced organotypical development of the hippocampal neuronal cell layers, which was also alleviated by IL-1Ra. Finally, grafts were evaluated with markers for glucose transporter expression, astrocytes, and activated microglia. Our results demonstrate significant effects of the HFHC diet on hippocampal morphology, including elevated microglial activation and reduced neuronal development. IL-1Ra largely blocked the detrimental effects of this diet, suggesting a potential use for this agent in neurological disorders involving neuroinflammation.
Collapse
Affiliation(s)
- L R Freeman
- Department of Neurosciences and the Center on Aging, Medical University of South Carolina, Charleston, SC, USA.
| | | | | | | | | |
Collapse
|
82
|
Yilmaz G, Granger DN. Leukocyte recruitment and ischemic brain injury. Neuromolecular Med 2010; 12:193-204. [PMID: 19579016 PMCID: PMC2878882 DOI: 10.1007/s12017-009-8074-1] [Citation(s) in RCA: 223] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2009] [Accepted: 06/16/2009] [Indexed: 01/23/2023]
Abstract
Leukocytes are recruited into the cerebral microcirculation following an ischemic insult. The leukocyte-endothelial cell adhesion manifested within a few hours after ischemia (followed by reperfusion, I/R) largely reflects an infiltration of neutrophils, while other leukocyte populations appear to dominate the adhesive interactions with the vessel wall at 24 h of reperfusion. The influx of rolling and adherent leukocytes is accompanied by the recruitment of adherent platelets, which likely enhances the cytotoxic potential of the leukocytes to which they are attached. The recruitment of leukocytes and platelets in the postischemic brain is mediated by specific adhesion glycoproteins expressed by the activated blood cells and on cerebral microvascular endothelial cells. This process is also modulated by different signaling pathways (e.g., CD40/CD40L, Notch) and cytokines (e.g., RANTES) that are activated/released following I/R. Some of the known risk factors for cardiovascular disease, including hypercholesterolemia and obesity appear to exacerbate the leukocyte and platelet recruitment elicited by brain I/R. Although lymphocyte-endothelial cell and -platelet interactions in the postischemic cerebral microcirculation have not been evaluated to date, recent evidence in experimental animals implicate both CD4+ and CD8+ T-lymphocytes in the cerebral microvascular dysfunction, inflammation, and tissue injury associated with brain I/R. Evidence implicating regulatory T-cells as cerebroprotective modulators of the inflammatory and tissue injury responses to brain I/R support a continued focus on leukocytes as a target for therapeutic intervention in ischemic stroke.
Collapse
Affiliation(s)
- Gokhan Yilmaz
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA 71130-3932, USA
| | - D. Neil Granger
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA 71130-3932, USA
| |
Collapse
|
83
|
Rajaraman P, Brenner AV, Neta G, Pfeiffer R, Wang SS, Yeager M, Thomas G, Fine HA, Linet MS, Rothman N, Chanock SJ, Inskip PD. Risk of meningioma and common variation in genes related to innate immunity. Cancer Epidemiol Biomarkers Prev 2010; 19:1356-61. [PMID: 20406964 PMCID: PMC3169167 DOI: 10.1158/1055-9965.epi-09-1151] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND The etiology of meningioma, the second most common type of adult brain tumor in the United States, is largely unknown. Prior studies indicate that history of immune-related conditions may affect the risk of meningioma. METHODS To identify genetic markers for meningioma in genes involved with innate immunity, we conducted an exploratory association study of 101 meningioma cases and 330 frequency-matched controls of European ancestry using subjects from a hospital-based study conducted by the National Cancer Institute. We genotyped 1,407 "tag" single nucleotide polymorphisms (SNP) in 148 genetic regions chosen on the basis of an r2>0.8 and minor allele frequency of >5% in Caucasians in HapMap1. Risk of meningioma was estimated by odds ratios and 95% confidence intervals. RESULTS Seventeen SNPs distributed across 12 genetic regions (NFKB1 (3), FCER1G (3), CCR6 (2), VCAM1, CD14, TNFRSF18, RAC2, XDH, C1D, TLR1/TLR10/TLR6, NOS1, and DEFA5) were associated with the risk of meningioma with P<0.01. Although individual SNP tests were not significant after controlling for multiple comparisons, gene region-based tests were statistically significant (P<0.05) for TNFRSF18, NFKB1, FCER1G, CD14, C1D, CCR6, and VCAM1. CONCLUSIONS AND IMPACT Our results indicate that common genetic polymorphisms in innate immunity genes may be associated with risk of meningioma. Given the small sample size, replication of these results in a larger study of meningioma is needed.
Collapse
Affiliation(s)
- Preetha Rajaraman
- Division of Cancer Epidemiology and Genetics, Neuro-oncology Branch, National Cancer Institute, NIH, Department of Health and Human Services, 6120 Executive Boulevard, EPS Room 7058, Bethesda, MD 20892-7238, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Yang Y, Wu J, Lu Y. Mechanism of HIV-1-TAT induction of interleukin-1beta from human monocytes: Involvement of the phospholipase C/protein kinase C signaling cascade. J Med Virol 2010; 82:735-46. [PMID: 20336759 PMCID: PMC2849807 DOI: 10.1002/jmv.21720] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Human immunodeficiency virus TAT plays an important role in the disregulation of cytokine production associated with the neurological disorders that follow HIV infection. IL-1beta is one of the important inflammatory cytokines secreted by immune-activated monocytes/macrophages. Previous reports have shown that extracellular TAT stimulates IL-1beta expression in monocytes/macrophages. However, little is known about the mechanisms and possible TAT-responsive elements within the IL-1beta promoter. The present study shows that TAT increases the production of IL-1beta in human monocytes; PLC-PKC pathway-dependent phosphorylation of p44/42 and JNK MAP kinases participates partially in IL-1beta induction by TAT; specific C/EBP and NF-kappaB transcription factor binding elements within the IL-1beta promoter are involved in TAT regulation of IL-1beta production. This study identifies a signaling mechanism for HIV-1-induced IL-1beta production in human monocytes that may be involved in the neuropathogenesis of HIV-associated dementia.
Collapse
Affiliation(s)
- Yongbo Yang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
- Department of Public Health Science, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, 96822, U.S.A
| | - Jianguo Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Yuanan Lu
- Department of Public Health Science, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, 96822, U.S.A
| |
Collapse
|
85
|
Janowitz T, Menon DK. Exploring new routes for neuroprotective drug development in traumatic brain injury. Sci Transl Med 2010; 2:27rv1. [PMID: 20393189 DOI: 10.1126/scitranslmed.3000330] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Worldwide, traumatic brain injury (TBI) is a major cause of mortality and morbidity with a substantial predicted increase in incidence. Despite an obvious need, there are no pharmacological treatment options for TBI because translation of neuroprotection from preclinical studies to clinical practice has so far failed. Here, we identify potential causes for this failure. We suggest that the monitoring and investigation tools that are commonly used in patients with TBI may provide an experimental medicine route to facilitate a more rational approach to translational research. This suggestion is underpinned by existing research data on disease biology, drug delivery, and treatment response obtained with these methods.
Collapse
Affiliation(s)
- T Janowitz
- Department of Medicine, Translational Medicine and Therapeutics, Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 2QQ, UK
| | | |
Collapse
|
86
|
Dutta K, Ghosh D, Nazmi A, Kumawat KL, Basu A. A common carcinogen benzo[a]pyrene causes neuronal death in mouse via microglial activation. PLoS One 2010; 5:e9984. [PMID: 20376308 PMCID: PMC2848611 DOI: 10.1371/journal.pone.0009984] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Accepted: 03/15/2010] [Indexed: 12/04/2022] Open
Abstract
Background Benzo[a]pyrene (B[a]P) belongs to a class of polycyclic aromatic hydrocarbons that serve as micropollutants in the environment. B[a]P has been reported as a probable carcinogen in humans. Exposure to B[a]P can take place by ingestion of contaminated (especially grilled, roasted or smoked) food or water, or inhalation of polluted air. There are reports available that also suggests neurotoxicity as a result of B[a]P exposure, but the exact mechanism of action is unknown. Methodology/Principal Findings Using neuroblastoma cell line and primary cortical neuron culture, we demonstrated that B[a]P has no direct neurotoxic effect. We utilized both in vivo and in vitro systems to demonstrate that B[a]P causes microglial activation. Using microglial cell line and primary microglial culture, we showed for the first time that B[a]P administration results in elevation of reactive oxygen species within the microglia thereby causing depression of antioxidant protein levels; enhanced expression of inducible nitric oxide synthase, that results in increased production of NO from the cells. Synthesis and secretion of proinflammatory cytokines were also elevated within the microglia, possibly via the p38MAP kinase pathway. All these factors contributed to bystander death of neurons, in vitro. When administered to animals, B[a]P was found to cause microglial activation and astrogliosis in the brain with subsequent increase in proinflammatory cytokine levels. Conclusions/Significance Contrary to earlier published reports we found that B[a]P has no direct neurotoxic activity. However, it kills neurons in a bystander mechanism by activating the immune cells of the brain viz the microglia. For the first time, we have provided conclusive evidence regarding the mechanism by which the micropollutant B[a]P may actually cause damage to the central nervous system. In today's perspective, where rising pollution levels globally are a matter of grave concern, our study throws light on other health hazards that such pollutants may exert.
Collapse
Affiliation(s)
- Kallol Dutta
- National Brain Research Centre, Manesar, Haryana, India
| | | | - Arshed Nazmi
- National Brain Research Centre, Manesar, Haryana, India
| | | | - Anirban Basu
- National Brain Research Centre, Manesar, Haryana, India
- * E-mail:
| |
Collapse
|
87
|
Identification of distinct cellular pools of interleukin-1β during the evolution of the neuroinflammatory response induced by transient middle cerebral artery occlusion in the brain of rat. Brain Res 2010; 1313:259-69. [DOI: 10.1016/j.brainres.2009.12.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Revised: 12/01/2009] [Accepted: 12/04/2009] [Indexed: 11/23/2022]
|
88
|
Laird MD, Sukumari-Ramesh S, Swift AEB, Meiler SE, Vender JR, Dhandapani KM. Curcumin attenuates cerebral edema following traumatic brain injury in mice: a possible role for aquaporin-4? J Neurochem 2010; 113:637-48. [PMID: 20132469 DOI: 10.1111/j.1471-4159.2010.06630.x] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Traumatic brain injury is a devastating neurological injury associated with significant morbidity and mortality. Medical therapies to limit cerebral edema, a cause of increased intracranial hypertension and poor clinical outcome, are largely ineffective, emphasizing the need for novel therapeutic approaches. In the present study, pre-treatment with curcumin (75, 150 mg/kg) or 30 min post-treatment with 300 mg/kg significantly reduced brain water content and improved neurological outcome following a moderate controlled cortical impact in mice. The protective effect of curcumin was associated with a significant attenuation in the acute pericontusional expression of interleukin-1beta, a pro-inflammatory cytokine, after injury. Curcumin also reversed the induction of aquaporin-4, an astrocytic water channel implicated in the development of cellular edema following head trauma. Notably, curcumin blocked IL-1beta-induced aquaporin-4 expression in cultured astrocytes, an effect mediated, at least in part, by reduced activation of the p50 and p65 subunits of nuclear factor kappaB. Consistent with this notion, curcumin preferentially attenuated phosphorylated p65 immunoreactivity in pericontusional astrocytes and decreased the expression of glial fibrillary acidic protein, a reactive astrocyte marker. As a whole, these data suggest clinically achievable concentrations of curcumin reduce glial activation and cerebral edema following neurotrauma, a finding which warrants further investigation.
Collapse
Affiliation(s)
- Melissa D Laird
- Department of Neurosurgery, Medical College of Georgia, Augusta, Georgia, USA
| | | | | | | | | | | |
Collapse
|
89
|
Morga E, Mouad-Amazzal L, Felten P, Heurtaux T, Moro M, Michelucci A, Gabel S, Grandbarbe L, Heuschling P. Jagged1 regulates the activation of astrocytes via modulation of NFkappaB and JAK/STAT/SOCS pathways. Glia 2010; 57:1741-53. [PMID: 19455581 DOI: 10.1002/glia.20887] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The Notch pathway is implicated in many aspects of the central nervous system (CNS) development and functions. Recently, we and others identified the Notch pathway to be involved in inflammatory events of the CNS. To understand the implication of this pathway on astrocytes, we have studied the Jagged-Notch-Hes pathway under inflammatory conditions. LPS exposure induced an upregulation of Jagged1 expression on cultured astrocytes. To address the role of Jagged1 in the modulation of inflammation, we used a siRNA mediated silencing of Jagged1 (siRNA J1). Jagged1 inhibition induced important variations on the Notch pathway components like Hes1, Hes5, Notch3, and RBP-Jkappa. siRNA J1 repressed the mRNA expression of genes known as hallmarks of the gliosis like GFAP and endothelin(B) receptor. On activated astrocytes, the inhibition of Jagged1 had antiinflammatory effects and resulted in a decrease of LPS-induced proinflammatory cytokines (IL1beta, IL1alpha, and TNFalpha) as well as the iNOS expression. The inhibition of Jagged1 induced a modulation of the JAK/STAT/SOCS signaling pathway. Most interestingly, the siRNA J1 decreased the LPS-induced translocation of NFkappaB p65 and this could be correlated to the phosphorylation of IkappaBalpha. These results suggest that during inflammatory and gliotic events of the CNS, Jagged1/Notch signaling sustains the inflammation mainly through NFkappaB and in part through JAK/STAT/SOCS signaling pathways.
Collapse
Affiliation(s)
- Eleonora Morga
- Laboratoire de Neurobiologie, Life Sciences, Université du Luxembourg, Luxembourg, 1511 Luxembourg.
| | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Munoz L, Ammit AJ. Targeting p38 MAPK pathway for the treatment of Alzheimer's disease. Neuropharmacology 2009; 58:561-8. [PMID: 19951717 DOI: 10.1016/j.neuropharm.2009.11.010] [Citation(s) in RCA: 295] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2009] [Revised: 10/30/2009] [Accepted: 11/24/2009] [Indexed: 10/20/2022]
Abstract
Accumulating evidence indicates that p38 mitogen-activated protein kinase (MAPK) could play more than one role in Alzheimer's disease (AD) pathophysiology and that patients suffering from AD dementia could benefit from p38 MAPK inhibitors. The p38 MAPK signalling has been widely accepted as a cascade contributing to neuroinflammation. However, deepening insight into the underlying biology of Alzheimer's disease reveals that p38 MAPK operates in other events related to AD, such as excitotoxicity, synaptic plasticity and tau phosphorylation. Although quantification of behavioural improvements upon p38 MAPK inhibition and in vivo evaluation of p38 MAPK significance to various aspects of AD pathology is still missing, the p38 MAPK is emerging as a new Alzheimer's disease treatment strategy. Thus, we present here an update on the role of p38 MAPK in neurodegeneration, with a focus on Alzheimer's disease, by summarizing recent literature and several key papers from earlier years.
Collapse
Affiliation(s)
- Lenka Munoz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The University of Sydney, NSW 2006, Australia.
| | | |
Collapse
|
91
|
Xia Y, Zhai Q. IL-1β enhances the antibacterial activity of astrocytes by activation of NF-κB. Glia 2009; 58:244-52. [DOI: 10.1002/glia.20921] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
92
|
Ghosh D, Mishra MK, Das S, Kaushik DK, Basu A. Tobacco carcinogen induces microglial activation and subsequent neuronal damage. J Neurochem 2009; 110:1070-81. [PMID: 19500213 DOI: 10.1111/j.1471-4159.2009.06203.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
4-Methylnitrosamino-1-(3-pyridyl)-1-butanone (NNK) is a tobacco-specific procarcinogen. We have investigated whether NNK causes inflammatory upheaval in the brain by activation of resident microglia and astrocyte and result in bystander neuronal damage. We have carried out the work in both in vitro and in vivo models. We have found that treatment with NNK causes significant activation of mouse microglial (BV2) cell line as evident by increase in reactive oxygen species and nitric oxide level. Western blot analysis has showed increase in proinflammatory signaling proteins, proinflammatory effector proteins, and other stress-related proteins. Interestingly, increased levels of proinflammatory cytokines like interleukin (IL)-6, tumor necrosis factor-alpha, monocyte chemoattractant protein 1 (MCP1), and IL-12p70 are also detected. Work from our in vivo studies has demonstrated similar increase in proinflammatory signaling and effector molecules along with the proinflammatory cytokine levels, following NNK treatment. Immunohistochemical staining of the brain sections of NNK-treated mice reveals massive microglial and astrocyte activation along with distinct foci of neuronal damage. Both in vitro and in vivo results provide strong indication that NNK causes significant upheaval of the inflammatory condition of brain and inflicts subsequent neuronal damage.
Collapse
|
93
|
The multifaceted profile of activated microglia. Mol Neurobiol 2009; 40:139-56. [PMID: 19629762 DOI: 10.1007/s12035-009-8077-9] [Citation(s) in RCA: 250] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2009] [Accepted: 06/17/2009] [Indexed: 12/17/2022]
Abstract
Although relatively neglected previously, research efforts in the past decade or so have identified a pivotal role for glial cells in regulating neuronal function. Particular emphasis has been placed on increasing our understanding of the function of microglia because a change from the ramified "resting" state of these cells has been associated with the pathogenesis of several neurodegenerative diseases, notably Alzheimer's disease. However, it is not clear whether activation of microglia and the associated inflammatory changes play a part in triggering disease processes or whether cell activation is a response to the early changes associated with the disease. In either case, the possibility exists that modulation of microglial activation may be beneficial in some circumstances, underlying the need to pursue research in this area. The original morphological categorization of microglia by Del Rio Hortega into ameboid, ramified, and intermediate forms, must now be elaborated to encompass a functional description. The evidence which has been generated recently suggests that microglia are probably never in a "resting" state and that several intermediate transitional states, based on function and morphology, probably exist. A more complete understanding of these states and the triggers which lead to a change from one to another state, and the factors which modulate the molecular switch that determines the persistence of the "activated" state remain to be identified.
Collapse
|
94
|
Decker Y, McBean G, Godson C. Lipoxin A4 inhibits IL-1beta-induced IL-8 and ICAM-1 expression in 1321N1 human astrocytoma cells. Am J Physiol Cell Physiol 2009; 296:C1420-7. [PMID: 19357230 DOI: 10.1152/ajpcell.00380.2008] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
There is a growing appreciation that endogenously produced mediators may actively promote the resolution of inflammation. Lipoxins (LX) are a group of recently discovered lipid mediators that have been shown to exert anti-inflammatory and proresolution effects on cells of myeloid and nonmyeloid origin. LXs mediate a number of processes, including regression of pro-inflammatory cytokine production, inhibition of cell proliferation, and stimulation of phagocytosis of apoptotic leukocytes by macrophages. Lipoxin A(4) (LXA(4)) is one of the principal LXs formed by mammalian cells. Recently, a G protein-coupled receptor that binds LXA(4,) the lipoxin A(4) receptor, was identified in astrocytes and microglia, suggesting that these cells may be a target for LX action in the brain. In this study, we have investigated the potential of LXA(4) to modify inflammatory responses of astrocytes, using the 1321N1 human astrocytoma cell line as a model system. As shown by quantitative RT-PCR, LXA(4) (10 nM) significantly inhibited (P < 0.05) the IL-1beta-induced stimulation of IL-8 and ICAM-1 expression in these cells. Furthermore, LXA(4) (10 nM) decreased the expression of IL-1beta-induced IL-8 protein levels (P < 0.05). LXA(4) (10 nM) was found to inhibit IL-1beta-induced degradation of IkappaBalpha (P < 0.05), and the activation of an NFkappaB regulated reporter gene construct (P < 0.05). Overall, these data suggest that LXA(4) exerts anti-inflammatory effects in 1321N1 astrocytoma cells at least in part via an NFkappaB-dependent mechanism. It is concluded that LXA(4) may represent a potentially novel therapeutic approach to acute or chronic inflammation in the brain.
Collapse
Affiliation(s)
- Yann Decker
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | |
Collapse
|
95
|
Chen Q, Zhang H, Li Q, An Y, Herkenham M, Lai W, Popovich P, Agarwal S, Quan N. Three Promoters Regulate Tissue- and Cell Type-specific Expression of Murine Interleukin-1 Receptor Type I. J Biol Chem 2009; 284:8703-13. [PMID: 19196714 DOI: 10.1074/jbc.m808261200] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The type 1 interleukin-1 receptor (IL-1R1) mediates diverse functions of interleukin-1 (IL-1) in the nervous, immune, and neuroendocrine systems. It has been suggested previously that the versatile functions of IL-1 may in part be conferred by the multiple promoters of IL-1R1 that have been identified for the human IL-1R1 gene. Promoters for murine IL-1R1 (mIL-1R1) gene have not been studied in detail. We performed 5'-rapid amplification of cDNA ends to determine the transcription start sites (TSS) in mIL-1R1, using mRNAs derived from 24 different tissues. The results revealed three putative TSSs of mIL-1R1. Three full-length cDNAs containing these distinct TSSs were recovered in screens of cloned cDNA libraries. Translation of these cDNAs produced IL-1R1 proteins that were verified by Western blot analysis. IL-1 stimulation of the individual IL-1R1 proteins resulted in the activation of NF-kappaB. Promoter-reporter assay for genomic DNA sequences immediately upstream of the three TSSs validated that the sequences possess promoter activity in a cell type-specific manner. These promoters are termed P1, P2, and P3 of the mIL-1R1, in 5' to 3' order. Quantitative PCR analysis of P1-, P2-, and P3-specific mIL-1R1 mRNAs showed that there is tissue-specific distribution of these mRNAs in vivo, and there are distinct patterns of P1, P2, and P3 mRNA expression in different cell lines. In the brain, P3 mRNA is expressed preferentially in the dentate gyrus. Further, glucocorticoids differentially regulate these promoters in a cell type-specific manner. Together, these results suggest that the different IL-1R1 promoters contribute to the discrete and diverse actions of IL-1.
Collapse
Affiliation(s)
- Qun Chen
- Department of Oral Biology, Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Sama MA, Mathis DM, Furman JL, Abdul HM, Artiushin IA, Kraner SD, Norris CM. Interleukin-1beta-dependent signaling between astrocytes and neurons depends critically on astrocytic calcineurin/NFAT activity. J Biol Chem 2008; 283:21953-64. [PMID: 18541537 PMCID: PMC2494911 DOI: 10.1074/jbc.m800148200] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Revised: 06/04/2008] [Indexed: 01/13/2023] Open
Abstract
Interleukin-1beta (IL-1beta) and the Ca(2+)/calmodulin-dependent protein phosphatase, calcineurin, have each been shown to play an important role in neuroinflammation. However, whether these signaling molecules interact to coordinate immune/inflammatory processes and neurodegeneration has not been investigated. Here, we show that exogenous application of IL-1beta (10 ng/ml) recruited calcineurin/NFAT (nuclear factor of activated T cells) activation in primary astrocyte-enriched cultures within minutes, through a pathway involving IL-1 receptors and L-type Ca(2+) channels. Adenovirus-mediated delivery of the NFAT inhibitor, VIVIT, suppressed the IL-1beta-dependent induction of several inflammatory mediators and/or markers of astrocyte activation, including tumor necrosis factor alpha, granulocyte/macrophage colony-stimulating factor, and vimentin. Expression of an activated form of calcineurin in one set of astrocyte cultures also triggered the release of factors that, in turn, stimulated NFAT activity in a second set of "naive" astrocytes. This effect was prevented when calcineurin-expressing cultures co-expressed VIVIT, suggesting that the calcineurin/NFAT pathway coordinates positive feedback signaling between astrocytes. In the presence of astrocytes and neurons, 48-h delivery of IL-1beta was associated with several excitotoxic effects, including NMDA receptor-dependent neuronal death, elevated extracellular glutamate, and hyperexcitable synaptic activity. Each of these effects were reversed or ameliorated by targeted delivery of VIVIT to astrocytes. IL-1beta also caused an NFAT-dependent reduction in excitatory amino acid transporter levels, indicating a possible mechanism for IL-1beta-mediated excitotoxicity. Taken together, the results have potentially important implications for the propagation and maintenance of neuroinflammatory signaling processes associated with many neurodegenerative conditions and diseases.
Collapse
Affiliation(s)
- Michelle A Sama
- Department of Molecular and Biomedical Pharmacology, Graduate Center for Gerontology, University of Kentucky, Lexington, Kentucky 40536, USA
| | | | | | | | | | | | | |
Collapse
|
97
|
Ben Menachem-Zidon O, Goshen I, Kreisel T, Ben Menahem Y, Reinhartz E, Ben Hur T, Yirmiya R. Intrahippocampal transplantation of transgenic neural precursor cells overexpressing interleukin-1 receptor antagonist blocks chronic isolation-induced impairment in memory and neurogenesis. Neuropsychopharmacology 2008; 33:2251-62. [PMID: 17987063 DOI: 10.1038/sj.npp.1301606] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The proinflammatory cytokine interleukin-1 (IL-1) within the brain is critically involved in mediating the memory impairment induced by acute inflammatory challenges and psychological stress. However, the role of IL-1 in memory impairment and suppressed neurogenesis induced by chronic stress exposure has not been investigated before now. We report here that mice that were isolated for 4 weeks displayed a significant elevation in hippocampal IL-1beta levels concomitantly with body weight loss, specific impairment in hippocampal-dependent memory, and decreased hippocampal neurogenesis. To examine the causal role of IL-1 in these effects, we developed a novel approach for long-term delivery of IL-1 receptor antagonist (IL-1ra) into the brain, using transplantation of neural precursor cells (NPCs), obtained from neonatal mice with transgenic overexpression of IL-1ra (IL-1raTG) under the glial fibrillary acidic protein promoter. Four weeks following intrahippocampal transplantation of IL-1raTG NPCs labeled with PKH-26, the transplanted cells were incorporated within the dentate gyrus and expressed mainly astrocytic markers. IL-1ra levels were markedly elevated in the hippocampus, but not in other brain regions, by 10 days and for at least 4 weeks post-transplantation. Transplantation of IL-1raTG NPCs completely rescued the chronic isolation-induced body weight loss, memory impairment, and suppressed hippocampal neurogenesis, compared with isolated mice transplanted with WT cells or sham operated. The transplantation had no effect in group-housed mice. These findings elucidate the role of IL-1 in the pathophysiology of chronic isolation and suggest that transplantation of IL-1raTG NPCs may provide a useful therapeutic procedure for IL-1-mediated memory disturbances in chronic inflammatory and neurological conditions.
Collapse
Affiliation(s)
- Ofra Ben Menachem-Zidon
- Department of Psychology, The Hebrew University of Jerusalem, Department of Neurology, Hadassah-Hebrew University Hospital, Jerusalem, Israel
| | | | | | | | | | | | | |
Collapse
|
98
|
Lin ST, Wang Y, Xue Y, Feng DC, Xu Y, Xu LY. Tetrandrine suppresses LPS-induced astrocyte activation via modulating IKKs-IκBα-NF-κB signaling pathway. Mol Cell Biochem 2008; 315:41-9. [DOI: 10.1007/s11010-008-9787-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Accepted: 05/05/2008] [Indexed: 11/24/2022]
|
99
|
Calderón-Garcidueñas L, Solt AC, Henríquez-Roldán C, Torres-Jardón R, Nuse B, Herritt L, Villarreal-Calderón R, Osnaya N, Stone I, García R, Brooks DM, González-Maciel A, Reynoso-Robles R, Delgado-Chávez R, Reed W. Long-term air pollution exposure is associated with neuroinflammation, an altered innate immune response, disruption of the blood-brain barrier, ultrafine particulate deposition, and accumulation of amyloid beta-42 and alpha-synuclein in children and young adults. Toxicol Pathol 2008; 36:289-310. [PMID: 18349428 DOI: 10.1177/0192623307313011] [Citation(s) in RCA: 651] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Air pollution is a serious environmental problem. We investigated whether residency in cities with high air pollution is associated with neuroinflammation/neurodegeneration in healthy children and young adults who died suddenly. We measured mRNA cyclooxygenase-2, interleukin-1beta, and CD14 in target brain regions from low (n = 12) or highly exposed residents (n = 35) aged 25.1 +/- 1.5 years. Upregulation of cyclooxygenase-2, interleukin-1beta, and CD14 in olfactory bulb, frontal cortex, substantia nigrae and vagus nerves; disruption of the blood-brain barrier; endothelial activation, oxidative stress, and inflammatory cell trafficking were seen in highly exposed subjects. Amyloid beta42 (Abeta42) immunoreactivity was observed in 58.8% of apolipoprotein E (APOE) 3/3 < 25 y, and 100% of the APOE 4 subjects, whereas alpha-synuclein was seen in 23.5% of < 25 y subjects. Particulate material (PM) was seen in olfactory bulb neurons, and PM < 100 nm were observed in intraluminal erythrocytes from lung, frontal, and trigeminal ganglia capillaries. Exposure to air pollution causes neuroinflammation, an altered brain innate immune response, and accumulation of Abeta42 and alpha-synuclein starting in childhood. Exposure to air pollution should be considered a risk factor for Alzheimer's and Parkinson's diseases, and carriers of the APOE 4 allele could have a higher risk of developing Alzheimer's disease if they reside in a polluted environment.
Collapse
|
100
|
Mustafina OE, Bakhtijarova KZ, Mikhailova AM, Tuktarova IA, Khusainova AN, Nasibullin TR, Magjanov RV. Analysis of the association of allelic variants of apolypoprotein E and interleukin 1 beta genes with multiple sclerosis in ethnic Tatars. RUSS J GENET+ 2008. [DOI: 10.1134/s1022795408030174] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|