1
|
Park J, Lee SH, Shin D, Kim Y, Kim YS, Seong MY, Lee JJ, Seo HG, Cho WS, Ro YS, Kim Y, Oh BM. Multiplexed Quantitative Proteomics Reveals Proteomic Alterations in Two Rodent Traumatic Brain Injury Models. J Proteome Res 2024; 23:249-263. [PMID: 38064581 DOI: 10.1021/acs.jproteome.3c00544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2024]
Abstract
In many cases of traumatic brain injury (TBI), conspicuous abnormalities, such as scalp wounds and intracranial hemorrhages, abate over time. However, many unnoticeable symptoms, including cognitive, emotional, and behavioral dysfunction, often last from several weeks to years after trauma, even for mild injuries. Moreover, the cause of such persistence of symptoms has not been examined extensively. Recent studies have implicated the dysregulation of the molecular system in the injured brain, necessitating an in-depth analysis of the proteome and signaling pathways that mediate the consequences of TBI. Thus, in this study, the brain proteomes of two TBI models were examined by quantitative proteomics during the recovery period to determine the molecular mechanisms of TBI. Our results show that the proteomes in both TBI models undergo distinct changes. A bioinformatics analysis demonstrated robust activation and inhibition of signaling pathways and core proteins that mediate biological processes after brain injury. These findings can help determine the molecular mechanisms that underlie the persistent effects of TBI and identify novel targets for drug interventions.
Collapse
Affiliation(s)
- Junho Park
- Department of Pharmacology, School of Medicine, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
- Proteomics Research Team, CHA Future Medicine Research Institute, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
- Research Institute for Basic Medical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| | - Seung Hak Lee
- Department of Rehabilitation Medicine, Asan Medical Center, 88 Olympic-Ro 43-Gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Dongyoon Shin
- Proteomics Research Team, CHA Future Medicine Research Institute, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| | - Yeongshin Kim
- Department of Life Science, School of Medicine, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| | - Young Sik Kim
- Proteomics Research Team, CHA Future Medicine Research Institute, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| | - Min Yong Seong
- Department of Rehabilitation Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Jin Joo Lee
- Department of Rehabilitation Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Han Gil Seo
- Department of Rehabilitation Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Won-Sang Cho
- Department of Neurosurgery, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Young Sun Ro
- Department of Emergency Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Youngsoo Kim
- Proteomics Research Team, CHA Future Medicine Research Institute, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
- Department of Life Science, School of Medicine, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| | - Byung-Mo Oh
- Department of Rehabilitation Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
- Institute of Aging, Seoul National University College of Medicine, 71 Ihwajang-gil, Jongno-gu, Seoul 03080, Republic of Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
- National Traffic Injury Rehabilitation Hospital, 260 Jungang-ro, Yangpyeong-gun 12564, Gyeonggi-do, Republic of Korea
| |
Collapse
|
2
|
Bandyopadhyay S, Garland P, Gaastra B, Zolnourian A, Bulters D, Galea I. The Haptoglobin Response after Aneurysmal Subarachnoid Haemorrhage. Int J Mol Sci 2023; 24:16922. [PMID: 38069244 PMCID: PMC10707007 DOI: 10.3390/ijms242316922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/21/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
Haptoglobin is the body's first line of defence against the toxicity of extracellular haemoglobin released following a subarachnoid haemorrhage (SAH). We investigated the haptoglobin response after SAH in cerebrospinal fluid (CSF) and serum. Paired CSF and serum samples from 19 controls and 92 SAH patients were assayed as follows: ultra-performance liquid chromatography for CSF haemoglobin and haptoglobin, immunoassay for serum haptoglobin and multiplexed CSF cytokines, and colorimetry for albumin. There was marked CSF haptoglobin deficiency: 99% of extracellular haemoglobin was unbound. The quotients for both CSF/serum albumin (qAlb) and haptoglobin (qHp) were used to compute the CSF haptoglobin index (qHp/qAlb). CSF from SAH patients had a significantly lower haptoglobin index compared to controls, especially in Haptoglobin-1 allele carriers. Serum haptoglobin levels increased after SAH and were correlated with CSF cytokine levels. Haptoglobin variables were not associated with long-term clinical outcomes post-SAH. We conclude that: (1) intrathecal haptoglobin consumption occurs after SAH, more so in haptoglobin-1 allele carriers; (2) serum haptoglobin is upregulated after SAH, in keeping with the liver acute phase response to central inflammation; (3) haptoglobin in the CSF is so low that any variation is too small for this to affect long-term outcomes, emphasising the potential for therapeutic haptoglobin supplementation.
Collapse
Affiliation(s)
- Soham Bandyopadhyay
- Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (S.B.); (P.G.); (B.G.)
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK;
| | - Patrick Garland
- Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (S.B.); (P.G.); (B.G.)
| | - Ben Gaastra
- Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (S.B.); (P.G.); (B.G.)
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK;
| | - Ardalan Zolnourian
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK;
| | - Diederik Bulters
- Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (S.B.); (P.G.); (B.G.)
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK;
| | - Ian Galea
- Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (S.B.); (P.G.); (B.G.)
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK;
| |
Collapse
|
3
|
Tsymbalyuk O, Gerzanich V, Simard JM, Rathinam CV. Traumatic brain injury alters dendritic cell differentiation and distribution in lymphoid and non-lymphoid organs. J Neuroinflammation 2022; 19:238. [PMID: 36183126 PMCID: PMC9526328 DOI: 10.1186/s12974-022-02609-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 09/24/2022] [Indexed: 11/10/2022] Open
Abstract
Background Pathophysiological consequences of traumatic brain injury (TBI) mediated secondary injury remain incompletely understood. In particular, the impact of TBI on the differentiation and maintenance of dendritic cells (DCs), which are regarded as the most professional antigen presenting cells of the immune system, remains completely unknown. Here, we report that DC-differentiation, maintenance and functions are altered on day 3 and day 7 after TBI. Methods Long bones, spleen, peripheral lymph nodes (pLNs), mesenteric lymph nodes (mLNs), liver, lungs, skin and blood were collected from mice with either moderate-level cortical impact (CCI) or sham on day 1, day 3 or day 7 after TBI. Bone marrow cells were isolated from the tibias and femurs of hind limb through flushing. Tissues were digested with Collagenase-D and DNase I. Skin biopsies were digested in the presence of liberase + DNase I. Single cell suspensions were made, red blood cells were lysed with Ammonium chloride (Stem Cell Technology) and subsequently filtered using a 70 μM nylon mesh. DC subsets of the tissues and DC progenitors of the BM were identified through 10-color flow cytometry-based immunophenotyping studies. Intracellular reactive oxygen species (ROS) were identified through H2DCFDA staining. Results Our studies identify that; (1) frequencies and absolute numbers of DCs in the spleen and BM are altered on day 3 and day 7 after TBI; (2) surface expression of key molecules involved in antigen presentation of DCs were affected on day 3 and day 7 after TBI; (3) distribution and functions of tissue-specific DC subsets of both circulatory and lymphatic systems were imbalanced following TBI; (4) early differentiation program of DCs, especially the commitment of hematopoietic stem cells to common DC progenitors (CDPs), were deregulated after TBI; and (5) intracellular ROS levels were reduced in DC progenitors and differentiated DCs on day 3 and day 7 after TBI. Conclusions Our data demonstrate, for the first time, that TBI affects the distribution pattern of DCs and induces an imbalance among DC subsets in both lymphoid and non-lymphoid organs. In addition, the current study demonstrates that TBI results in reduced levels of ROS in DCs on day 3 and day 7 after TBI, which may explain altered DC differentiation paradigm following TBI. A deeper understanding on the molecular mechanisms that contribute to DC defects following TBI would be essential and beneficial in treating infections in patients with acute central nervous system (CNS) injuries, such as TBI, stroke and spinal cord injury.
Collapse
Affiliation(s)
- Orest Tsymbalyuk
- Department of Neurosurgery, University of Maryland School of Medicine, MD, Baltimore, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, MD, Baltimore, USA
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, MD, Baltimore, USA.,Research Service, Veterans Affairs Maryland Health Care System, MD, Baltimore, USA.,Department of Pathology, University of Maryland School of Medicine, MD, Baltimore, USA.,Department of Physiology, University of Maryland School of Medicine, MD, Baltimore, USA
| | - Chozha Vendan Rathinam
- Institute of Human Virology, University of Maryland School of Medicine, 725 West Lombard Street, Baltimore, MD, 21201, USA. .,Center for Stem Cell and Regenerative Medicine, University of Maryland School of Medicine, MD, 21201, Baltimore, USA.
| |
Collapse
|
4
|
Tallon C, Picciolini S, Yoo SW, Thomas AG, Pal A, Alt J, Carlomagno C, Gualerzi A, Rais R, Haughey NJ, Bedoni M, Slusher BS. Inhibition of neutral sphingomyelinase 2 reduces extracellular vesicle release from neurons, oligodendrocytes, and activated microglial cells following acute brain injury. Biochem Pharmacol 2021; 194:114796. [PMID: 34678224 DOI: 10.1016/j.bcp.2021.114796] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 12/16/2022]
Abstract
Extracellular Vesicles (EVs) are implicated in the spread of pathogenic proteinsin a growing number of neurological diseases. Given this, there is rising interest in developing inhibitors of Neutral Sphingomyelinase 2 (nSMase2), an enzyme critical in EV biogenesis. Our group recently discovered phenyl(R)-(1-(3-(3,4-dimethoxyphenyl)-2,6-dimethylimidazo[1,2-b]pyridazin-8-yl)pyrrolidin-3-yl)carbamate (PDDC), the first potent, selective, orally-available, and brain-penetrable nSMase2 inhibitor, capable of dose-dependently reducing EVs release in vitro and in vivo. Herein, using multiplexed Surface Plasmon Resonance imaging (SPRi), we evaluated which brain cell-derived EVs were affected by PDDC following acute brain injury. Mice were fed PDDC-containing chow at doses which gave steady PDDC brain exposures exceeding its nSMase2 IC50. Mice were then administered an intra-striatal IL-1β injection and two hours later plasma and brain were collected. IL-1β injection significantly increased striatal nSMase2 activity which was completely normalized by PDDC. Using SPRi, we found that IL-1β-induced injury selectively increased plasma levels of CD171 + and PLP1 + EVs; this EV increase was normalized by PDDC. In contrast, GLAST1 + EVs were unchanged by IL-1β or PDDC. IL-1β injection selectively increased EVs released from activated versus non-activated microglia, indicated by the CD11b+/IB4 + ratio. The increase in EVs from CD11b + microglia was dramatically attenuated with PDDC. Taken together, our data demonstrate that following acute injury, brain nSMase2 activity is elevated. EVs released from neurons, oligodendrocytes, and activated microglial are increased in plasma and inhibition of nSMase2 with PDDC reduced these IL-1β-induced changes implicating nSMase2 inhibition as a therapeutic target for acute brain injury.
Collapse
Affiliation(s)
- Carolyn Tallon
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Silvia Picciolini
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Laboratory of Nanomedicine and Clinical Biophotonics (LABION), Milan, Italy
| | - Seung-Wan Yoo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ajit G Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Arindom Pal
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jesse Alt
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Cristiano Carlomagno
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Laboratory of Nanomedicine and Clinical Biophotonics (LABION), Milan, Italy
| | - Alice Gualerzi
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Laboratory of Nanomedicine and Clinical Biophotonics (LABION), Milan, Italy
| | - Rana Rais
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Norman J Haughey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Marzia Bedoni
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Laboratory of Nanomedicine and Clinical Biophotonics (LABION), Milan, Italy.
| | - Barbara S Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
5
|
Wu YH, Rosset S, Lee TR, Dragunow M, Park T, Shim V. In Vitro Models of Traumatic Brain Injury: A Systematic Review. J Neurotrauma 2021; 38:2336-2372. [PMID: 33563092 DOI: 10.1089/neu.2020.7402] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Traumatic brain injury (TBI) is a major public health challenge that is also the third leading cause of death worldwide. It is also the leading cause of long-term disability in children and young adults worldwide. Despite a large body of research using predominantly in vivo and in vitro rodent models of brain injury, there is no medication that can reduce brain damage or promote brain repair mainly due to our lack of understanding in the mechanisms and pathophysiology of the TBI. The aim of this review is to examine in vitro TBI studies conducted from 2008-2018 to better understand the TBI in vitro model available in the literature. Specifically, our focus was to perform a detailed analysis of the in vitro experimental protocols used and their subsequent biological findings. Our review showed that the uniaxial stretch is the most frequently used way of load application, accounting for more than two-thirds of the studies reviewed. The rate and magnitude of the loading were varied significantly from study to study but can generally be categorized into mild, moderate, and severe injuries. The in vitro studies reviewed here examined key processes in TBI pathophysiology such as membrane disruptions leading to ionic dysregulation, inflammation, and the subsequent damages to the microtubules and axons, as well as cell death. Overall, the studies examined in this review contributed to the betterment of our understanding of TBI as a disease process. Yet, our review also revealed the areas where more work needs to be done such as: 1) diversification of load application methods that will include complex loading that mimics in vivo head impacts; 2) more widespread use of human brain cells, especially patient-matched human cells in the experimental set-up; and 3) need for building a more high-throughput system to be able to discover effective therapeutic targets for TBI.
Collapse
Affiliation(s)
- Yi-Han Wu
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
- Center for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Samuel Rosset
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Tae-Rin Lee
- Advanced Institute of Convergence Technology, Seoul National University, Seoul, Korea
| | - Mike Dragunow
- Center for Brain Research, The University of Auckland, Auckland, New Zealand
- Department of Pharmacology, The University of Auckland, Auckland, New Zealand
| | - Thomas Park
- Center for Brain Research, The University of Auckland, Auckland, New Zealand
- Department of Pharmacology, The University of Auckland, Auckland, New Zealand
| | - Vickie Shim
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
6
|
Postolache TT, Wadhawan A, Can A, Lowry CA, Woodbury M, Makkar H, Hoisington AJ, Scott AJ, Potocki E, Benros ME, Stiller JW. Inflammation in Traumatic Brain Injury. J Alzheimers Dis 2021; 74:1-28. [PMID: 32176646 DOI: 10.3233/jad-191150] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
There is an increasing evidence that inflammation contributes to clinical and functional outcomes in traumatic brain injury (TBI). Many successful target-engaging, lesion-reducing, symptom-alleviating, and function-improving interventions in animal models of TBI have failed to show efficacy in clinical trials. Timing and immunological context are paramount for the direction, quality, and intensity of immune responses to TBI and the resulting neuroanatomical, clinical, and functional course. We present components of the immune system implicated in TBI, potential immune targets, and target-engaging interventions. The main objective of our article is to point toward modifiable molecular and cellular mechanisms that may modify the outcomes in TBI, and contribute to increasing the translational value of interventions that have been identified in animal models of TBI.
Collapse
Affiliation(s)
- Teodor T Postolache
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Aurora, CO, USA.,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO, USA.,Mental Illness Research, Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 5, VA Capitol Health Care Network, Baltimore, MD, USA
| | - Abhishek Wadhawan
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,Saint Elizabeths Hospital, Department of Psychiatry, Washington, DC, USA
| | - Adem Can
- School of Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Christopher A Lowry
- Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Aurora, CO, USA.,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO, USA.,Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA.,Department of Physical Medicine and Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Margaret Woodbury
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,VA Maryland Healthcare System, Baltimore VA Medical Center, Baltimore, MD, USA
| | - Hina Makkar
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Andrew J Hoisington
- Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Aurora, CO, USA.,Systems Engineering and Management, Air Force Institute of Technology, Wright-Patterson AFB, OH, USA
| | - Alison J Scott
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD, USA
| | - Eileen Potocki
- VA Maryland Healthcare System, Baltimore VA Medical Center, Baltimore, MD, USA
| | - Michael E Benros
- Copenhagen Research Center for Mental Health-CORE, Mental Health Centre Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - John W Stiller
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,Maryland State Athletic Commission, Baltimore, MD, USA.,Saint Elizabeths Hospital, Neurology Consultation Services, Washington, DC, USA
| |
Collapse
|
7
|
Yates AG, Jogia T, Gillespie ER, Couch Y, Ruitenberg MJ, Anthony DC. Acute IL-1RA treatment suppresses the peripheral and central inflammatory response to spinal cord injury. J Neuroinflammation 2021; 18:15. [PMID: 33407641 PMCID: PMC7788822 DOI: 10.1186/s12974-020-02050-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/09/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The acute phase response (APR) to CNS insults contributes to the overall magnitude and nature of the systemic inflammatory response. Aspects of this response are thought to drive secondary inflammatory pathology at the lesion site, and suppression of the APR can therefore afford some neuroprotection. In this study, we examined the APR in a mouse model of traumatic spinal cord injury (SCI), along with its relationship to neutrophil recruitment during the immediate aftermath of the insult. We specifically investigated the effect of IL-1 receptor antagonist (IL-1RA) administration on the APR and leukocyte recruitment to the injured spinal cord. METHODS Adult female C57BL/6 mice underwent either a 70kD contusive SCI, or sham surgery, and tissue was collected at 2, 6, 12, and 24 hours post-operation. For IL-1RA experiments, SCI mice received two intraperitoneal injections of human IL-1RA (100mg/kg), or saline as control, immediately following, and 5 hours after impact, and animals were sacrificed 6 hours later. Blood, spleen, liver and spinal cord were collected to study markers of central and peripheral inflammation by flow cytometry, immunohistochemistry and qPCR. Results were analysed by two-way ANOVA or student's t-test, as appropriate. RESULTS SCI induced a robust APR, hallmarked by elevated hepatic expression of pro-inflammatory marker genes and a significantly increased neutrophil presence in the blood, liver and spleen of these animals, as early as 2 hours after injury. This peripheral response preceded significant neutrophil infiltration of the spinal cord, which peaked 24 hours post-SCI. Although expression of IL-1RA was also induced in the liver following SCI, its response was delayed compared to IL-1β. Exogenous administration of IL-1RA during this putative therapeutic window was able to suppress the hepatic APR, as evidenced by a reduction in CXCL1 and SAA-2 expression as well as a significant decrease in neutrophil infiltration in both the liver and the injured spinal cord itself. CONCLUSIONS Our data indicate that peripheral administration of IL-1RA can attenuate the APR which in turn reduces immune cell infiltration at the spinal cord lesion site. We propose IL-1RA treatment as a viable therapeutic strategy to minimise the harmful effects of SCI-induced inflammation.
Collapse
Affiliation(s)
- Abi G Yates
- Department of Pharmacology, The University of Oxford, Mansfield Road, Oxford, UK
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Queensland, Australia
| | - Trisha Jogia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Queensland, Australia
| | - Ellen R Gillespie
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Queensland, Australia
| | - Yvonne Couch
- Acute Stroke Programme, RDM-Investigative Medicine, The University of Oxford, Oxford, UK
| | - Marc J Ruitenberg
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Queensland, Australia
| | - Daniel C Anthony
- Department of Pharmacology, The University of Oxford, Mansfield Road, Oxford, UK.
- Sechenov First Moscow State Medical University, Moscow, Russia.
| |
Collapse
|
8
|
Kim TJ, Park SH, Jeong HB, Ha EJ, Cho WS, Kang HS, Kim JE, Ko SB. Optimizing Nitrogen Balance Is Associated with Better Outcomes in Neurocritically Ill Patients. Nutrients 2020; 12:nu12103137. [PMID: 33066539 PMCID: PMC7602201 DOI: 10.3390/nu12103137] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/07/2020] [Accepted: 10/11/2020] [Indexed: 12/12/2022] Open
Abstract
Marked protein catabolism is common in critically ill patients. We hypothesized that optimal protein supplementation using nitrogen balance might be associated with better outcomes in the neurointensive care unit (NICU) patients. A total of 175 patients admitted to the NICU between July 2017 and December 2018 were included. Nitrogen balance was measured after NICU admission and measurements were repeated in 77 patients. The outcomes were compared according to initial nitrogen balance results and improvement of nitrogen balance on follow-up measurements. A total of 140 (80.0%) patients had a negative nitrogen balance on initial assessments. The negative balance group had more events of in-hospital mortality and poor functional outcome at three months. In follow-up measurement patients, 39 (50.6%) showed an improvement in nitrogen balance. The improvement group had fewer events of in-hospital mortality (p = 0.047) and poor functional outcomes (p = 0.046). Moreover, improvement of nitrogen balance was associated with a lower risk of poor functional outcomes (Odds ratio, 0.247; 95% confidence interval, 0.066–0.925, p = 0.038). This study demonstrated that a significant proportion of patients in the NICU were under protein hypercatabolism. Moreover, an improvement in protein balance was related to improved outcomes in neurocritically ill patients. Further studies are needed to confirm the relationship between protein balance and outcomes.
Collapse
Affiliation(s)
- Tae Jung Kim
- Department of Neurology, Seoul National University Hospital, Seoul 03080, Korea; (T.J.K.); (H.-B.J.)
- Department of Critical Care Medicine, Seoul National University Hospital, Seoul 03080, Korea;
| | - Soo-Hyun Park
- Department of Neurology, Inha University Hospital, Incheon 22332, Korea;
| | - Hae-Bong Jeong
- Department of Neurology, Seoul National University Hospital, Seoul 03080, Korea; (T.J.K.); (H.-B.J.)
| | - Eun Jin Ha
- Department of Critical Care Medicine, Seoul National University Hospital, Seoul 03080, Korea;
- Department of Neurosurgery, Seoul National University Hospital, Seoul 03080, Korea; (W.S.C.); (H.-S.K.); (J.E.K.)
| | - Won Sang Cho
- Department of Neurosurgery, Seoul National University Hospital, Seoul 03080, Korea; (W.S.C.); (H.-S.K.); (J.E.K.)
| | - Hyun-Seung Kang
- Department of Neurosurgery, Seoul National University Hospital, Seoul 03080, Korea; (W.S.C.); (H.-S.K.); (J.E.K.)
| | - Jeong Eun Kim
- Department of Neurosurgery, Seoul National University Hospital, Seoul 03080, Korea; (W.S.C.); (H.-S.K.); (J.E.K.)
| | - Sang-Bae Ko
- Department of Neurology, Seoul National University Hospital, Seoul 03080, Korea; (T.J.K.); (H.-B.J.)
- Department of Critical Care Medicine, Seoul National University Hospital, Seoul 03080, Korea;
- Correspondence: ; Tel.: +82-2-2072-2278
| |
Collapse
|
9
|
Yates AG, Anthony DC, Ruitenberg MJ, Couch Y. Systemic Immune Response to Traumatic CNS Injuries-Are Extracellular Vesicles the Missing Link? Front Immunol 2019; 10:2723. [PMID: 31824504 PMCID: PMC6879545 DOI: 10.3389/fimmu.2019.02723] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/06/2019] [Indexed: 12/16/2022] Open
Abstract
Inflammation following traumatic injury to the central nervous system (CNS) persists long after the primary insult and is known to exacerbate cell death and worsen functional outcomes. Therapeutic interventions targeting this inflammation have been unsuccessful, which has been attributed to poor bioavailability owing to the presence of blood-CNS barrier. Recent studies have shown that the magnitude of the CNS inflammatory response is dependent on systemic inflammatory events. The acute phase response (APR) to CNS injury presents an alternative strategy to modulating the secondary phase of injury. However, the communication pathways between the CNS and the periphery remain poorly understood. Extracellular vesicles (EVs) are membrane bound nanoparticles that are regulators of intercellular communication. They are shed from cells of the CNS including microglia, astrocytes, neurons and endothelial cells, and are able to cross the blood-CNS barrier, thus providing an attractive candidate for initiating the APR after acute CNS injury. The purpose of this review is to summarize the current evidence that EVs play a critical role in the APR following CNS injuries.
Collapse
Affiliation(s)
- Abi G Yates
- Department of Pharmacology, Medical Sciences Division, University of Oxford, Oxford, United Kingdom.,School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Daniel C Anthony
- Department of Pharmacology, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Marc J Ruitenberg
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Yvonne Couch
- Acute Stroke Programme, RDM-Investigative Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
10
|
Rojas C, Sala M, Thomas AG, Datta Chaudhuri A, Yoo SW, Li Z, Dash RP, Rais R, Haughey NJ, Nencka R, Slusher B. A novel and potent brain penetrant inhibitor of extracellular vesicle release. Br J Pharmacol 2019; 176:3857-3870. [PMID: 31273753 DOI: 10.1111/bph.14789] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 06/12/2019] [Accepted: 06/13/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Extracellular vesicles (EVs) are constitutively shed from cells and released by various stimuli. Their protein and RNA cargo are modified by the stimulus, and in disease conditions can carry pathological cargo involved in disease progression. Neutral sphingomyelinase 2 (nSMase2) is a major regulator in at least one of several independent routes of EV biogenesis, and its inhibition is a promising new therapeutic approach for neurological disorders. Unfortunately, known inhibitors exhibit μM potency, poor physicochemical properties, and/or limited brain penetration. Here, we sought to identify a drug-like inhibitor of nSMase2. EXPERIMENTAL APPROACH We conducted a human nSMase2 high throughput screen (>365,000 compounds). Selected hits were optimized focusing on potency, selectivity, metabolic stability, pharmacokinetics, and ability to inhibit EV release in vitro and in vivo. KEY RESULTS We identified phenyl(R)-(1-(3-(3,4-dimethoxyphenyl)-2,6-dimethylimidazo[1,2-b]pyridazin-8-yl)pyrrolidin-3-yl)-carbamate (PDDC), a potent (pIC50 = 6.57) and selective non-competitive inhibitor of nSMase2. PDDC was metabolically stable, with excellent oral bioavailability (%F = 88) and brain penetration (AUCbrain /AUCplasma = 0.60). PDDC dose-dependently (pEC50 = 5.5) inhibited release of astrocyte-derived extracellular vesicles (ADEV). In an in vivo inflammatory brain injury model, PDDC robustly inhibited ADEV release and the associated peripheral immunological response. A closely related inactive PDDC analogue was ineffective. CONCLUSION AND IMPLICATIONS PDDC is a structurally novel, potent, orally available, and brain penetrant inhibitor of nSMase2. PDDC inhibited release of ADEVs in tissue culture and in vivo. PDDC is actively being tested in animal models of neurological disease and, along with closely related analogues, is being considered for clinical translation.
Collapse
Affiliation(s)
- Camilo Rojas
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland.,Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Michal Sala
- Chem Research Group, Institute of Organic Chemistry and Biochemistry, Prague, Czech Republic
| | - Ajit G Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | | | - Seung-Wan Yoo
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Zhigang Li
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Ranjeet P Dash
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Rana Rais
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland.,Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Norman J Haughey
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Radim Nencka
- Chem Research Group, Institute of Organic Chemistry and Biochemistry, Prague, Czech Republic
| | - Barbara Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland.,Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland.,Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland.,Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland.,Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland.,Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
11
|
Wen M, Jin Y, Zhang H, Sun X, Kuai Y, Tan W. Proteomic Analysis of Rat Cerebral Cortex in the Subacute to Long-Term Phases of Focal Cerebral Ischemia-Reperfusion Injury. J Proteome Res 2019; 18:3099-3118. [PMID: 31265301 DOI: 10.1021/acs.jproteome.9b00220] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Stroke is a leading cause of mortality and disability, and ischemic stroke accounts for more than 80% of the disease occurrence. Timely reperfusion is essential in the treatment of ischemic stroke, but it is known to cause ischemia-reperfusion (I/R) injury and the relevant studies have mostly focused on the acute phase. Here we reported on a global proteomic analysis to investigate the development of cerebral I/R injury in the subacute and long-term phases. A rat model was used, with 2 h-middle cerebral artery occlusion (MCAO) followed with 1, 7, and 14 days of reperfusion. The proteins of cerebral cortex were analyzed by SDS-PAGE, whole-gel slicing, and quantitative LC-MS/MS. Totally 5621 proteins were identified, among which 568, 755, and 492 proteins were detected to have significant dys-regulation in the model groups with 1, 7, and 14 days of reperfusion, respectively, when compared with the corresponding sham groups (n = 4, fold change ≥1.5 or ≤0.67 and p ≤ 0.05). Bioinformatic analysis on the functions and reperfusion time-dependent dys-regulation profiles of the proteins exhibited changes of structures and biological processes in cytoskeleton, synaptic plasticity, energy metabolism, inflammation, and lysosome from subacute to long-term phases of cerebral I/R injury. Disruption of cytoskeleton and synaptic structures, impairment of energy metabolism processes, and acute inflammation responses were the most significant features in the subacute phase. With the elongation of reperfusion time to the long-term phase, a tendency of recovery was detected on cytoskeleton, while inflammation pathways different from the subacute phase were activated. Also, lysosomal structures and functions might be restored. This is the first work reporting the proteome changes that occurred at different time points from the subacute to long-term phases of cerebral I/R injury and we expect it would provide useful information to improve the understanding of the mechanisms involved in the development of cerebral I/R injury and suggest candidates for treatment.
Collapse
Affiliation(s)
- Meiling Wen
- School of Biology and Biological Engineering , South China University of Technology , Guangzhou 510006 , P. R. China
| | - Ya Jin
- Institute of Biomedical and Pharmaceutical Sciences , Guangdong University of Technology , Guangzhou 510006 , P. R. China
| | - Hao Zhang
- Institute of Biomedical and Pharmaceutical Sciences , Guangdong University of Technology , Guangzhou 510006 , P. R. China
| | - Xiaoou Sun
- Institute of Biomedical and Pharmaceutical Sciences , Guangdong University of Technology , Guangzhou 510006 , P. R. China
| | - Yihe Kuai
- Institute of Biomedical and Pharmaceutical Sciences , Guangdong University of Technology , Guangzhou 510006 , P. R. China
| | - Wen Tan
- Institute of Biomedical and Pharmaceutical Sciences , Guangdong University of Technology , Guangzhou 510006 , P. R. China
| |
Collapse
|
12
|
Srivastava AK, Prabhakara KS, Kota DJ, Bedi SS, Triolo F, Brown KS, Skiles ML, Brown HL, Cox CS, Olson SD. Human umbilical cord blood cells restore vascular integrity in injured rat brain and modulate inflammation in vitro. Regen Med 2019; 14:295-307. [PMID: 31074319 DOI: 10.2217/rme-2018-0106] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aim: Traumatic brain injury is a complex condition consisting of a mechanical injury with neurovascular disruption and inflammation with limited clinical interventions available. A growing number of studies report systemic delivery of human umbilical cord blood (HUCB) as a therapy for neural injuries. Materials & methods: HUCB cells from five donors were tested to improve blood-brain barrier integrity in a traumatic brain injury rat model at a dose of 2.5 × 107 cells/kg at 24 or 72 h postinjury and for immunomodulatory activity in vitro. Results & Conclusion: We observed that cells delivered 72 h postinjury significantly restored blood-brain barrier integrity. HUCB cells reduced the amount of TNF-α and IFN-γ released by activated primary rat splenocytes, which correlated with the expression of COX2 and IDO1.
Collapse
Affiliation(s)
- Amit K Srivastava
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Sciences Center at Houston, Houston, TX, 77030, USA
| | - Karthik S Prabhakara
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Sciences Center at Houston, Houston, TX, 77030, USA
| | - Daniel J Kota
- Emory Personalized Immunotherapy Core Labs, Emory University, School of Medicine, Atlanta, GA 30322, USA
| | - Supinder S Bedi
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Sciences Center at Houston, Houston, TX, 77030, USA
| | - Fabio Triolo
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Sciences Center at Houston, Houston, TX, 77030, USA
| | | | | | | | - Charles S Cox
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Sciences Center at Houston, Houston, TX, 77030, USA
| | - Scott D Olson
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Sciences Center at Houston, Houston, TX, 77030, USA
| |
Collapse
|
13
|
Xu FF, Zhang ZB, Wang YY, Wang TH. Brain-Derived Glia Maturation Factor β Participates in Lung Injury Induced by Acute Cerebral Ischemia by Increasing ROS in Endothelial Cells. Neurosci Bull 2018; 34:1077-1090. [PMID: 30191459 PMCID: PMC6246848 DOI: 10.1007/s12264-018-0283-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/03/2018] [Indexed: 02/05/2023] Open
Abstract
Brain damage can cause lung injury. To explore the mechanism underlying the lung injury induced by acute cerebral ischemia (ACI), we established a middle cerebral artery occlusion (MCAO) model in male Sprague-Dawley rats. We focused on glia maturation factor β (GMFB) based on quantitative analysis of the global rat serum proteome. Polymerase chain reaction, western blotting, and immunofluorescence revealed that GMFB was over-expressed in astrocytes in the brains of rats subjected to MCAO. We cultured rat primary astrocytes and confirmed that GMFB was also up-regulated in primary astrocytes after oxygen-glucose deprivation (OGD). We subjected the primary astrocytes to Gmfb RNA interference before OGD and collected the conditioned medium (CM) after OGD. We then used the CM to culture pulmonary microvascular endothelial cells (PMVECs) acquired in advance and assessed their status. The viability of the PMVECs improved significantly when Gmfb was blocked. Moreover, ELISA assays revealed an elevation in GMFB concentration in the medium after OGD. Cell cultures containing recombinant GMFB showed increased levels of reactive oxygen species and a deterioration in the state of the cells. In conclusion, GMFB is up-regulated in astrocytes after ACI, and brain-derived GMFB damages PMVECs by increasing reactive oxygen species. GMFB might thus be an initiator of the lung injury induced by ACI.
Collapse
Affiliation(s)
- Fei-Fei Xu
- Institute of Neurological Disease, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zi-Bin Zhang
- Institute of Neurological Disease, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yang-Yang Wang
- Institute of Neurological Disease, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ting-Hua Wang
- Institute of Neurological Disease, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
14
|
Dickens AM, Posti JP, Takala RSK, Ala-Seppälä H, Mattila I, Coles JP, Frantzén J, Hutchinson PJ, Katila AJ, Kyllönen A, Maanpää HR, Newcombe V, Outtrim J, Tallus J, Carpenter KLH, Menon DK, Hyötyläinen T, Tenovuo O, Orešic M. Serum Metabolites Associated with Computed Tomography Findings after Traumatic Brain Injury. J Neurotrauma 2018; 35:2673-2683. [PMID: 29947291 DOI: 10.1089/neu.2017.5272] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
There is a need to rapidly detect patients with traumatic brain injury (TBI) who require head computed tomography (CT). Given the energy crisis in the brain following TBI, we hypothesized that serum metabolomics would be a useful tool for developing a set of biomarkers to determine the need for CT and to distinguish among different types of injuries observed. Logistical regression models using metabolite data from the discovery cohort (n = 144, Turku, Finland) were used to distinguish between patients with traumatic intracranial findings and those with negative findings on head CT. The resultant models were then tested in the validation cohort (n = 66, Cambridge, United Kingdom). The levels of glial fibrillary acidic protein and ubiquitin C-terminal hydrolase-L1 were also quantified in the serum from the same patients. Despite there being significant differences in the protein biomarkers in patients with TBI, the model that determined the need for a CT scan validated poorly (area under the curve [AUC] = 0.64: Cambridge patients). However, using a combination of six metabolites (two amino acids, three sugar derivatives, and one ketoacid) it was possible to discriminate patients with intracranial abnormalities on CT and patients with a normal CT (AUC = 0.77 in Turku patients and AUC = 0.73 in Cambridge patients). Further, a combination of three metabolites could distinguish between diffuse brain injuries and mass lesions (AUC = 0.87 in Turku patients and AUC = 0.68 in Cambridge patients). This study identifies a set of validated serum polar metabolites, which associate with the need for a CT scan. Additionally, serum metabolites can also predict the nature of the brain injury. These metabolite markers may prevent unnecessary CT scans, thus reducing the cost of diagnostics and radiation load.
Collapse
Affiliation(s)
- Alex M Dickens
- 1 Turku Centre for Biotechnology, University of Turku , Turku, Finland
| | - Jussi P Posti
- 2 Turku Brain Injury Centre, Turku University Hospital , Turku, Finland .,3 Department of Neurology, University of Turku , Turku, Finland .,4 Division of Clinical Neurosciences, Department of Neurosurgery, Turku University Hospital , Turku, Finland
| | - Riikka S K Takala
- 5 Perioperative Services, Intensive Care Medicine and Pain Management, Turku University Hospital and University of Turku , Turku, Finland
| | | | - Ismo Mattila
- 6 Steno Diabetes Center Copenhagen , Gentofte, Denmark
| | - Jonathan P Coles
- 7 Division of Anaesthesia, Department of Medicine, University of Cambridge , Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Janek Frantzén
- 2 Turku Brain Injury Centre, Turku University Hospital , Turku, Finland .,3 Department of Neurology, University of Turku , Turku, Finland .,4 Division of Clinical Neurosciences, Department of Neurosurgery, Turku University Hospital , Turku, Finland
| | - Peter J Hutchinson
- 8 Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge , Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Ari J Katila
- 5 Perioperative Services, Intensive Care Medicine and Pain Management, Turku University Hospital and University of Turku , Turku, Finland
| | - Anna Kyllönen
- 3 Department of Neurology, University of Turku , Turku, Finland
| | | | - Virginia Newcombe
- 7 Division of Anaesthesia, Department of Medicine, University of Cambridge , Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Joanne Outtrim
- 7 Division of Anaesthesia, Department of Medicine, University of Cambridge , Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Jussi Tallus
- 3 Department of Neurology, University of Turku , Turku, Finland
| | - Keri L H Carpenter
- 8 Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge , Addenbrooke's Hospital, Cambridge, United Kingdom
| | - David K Menon
- 7 Division of Anaesthesia, Department of Medicine, University of Cambridge , Addenbrooke's Hospital, Cambridge, United Kingdom
| | | | - Olli Tenovuo
- 2 Turku Brain Injury Centre, Turku University Hospital , Turku, Finland .,3 Department of Neurology, University of Turku , Turku, Finland
| | - Matej Orešic
- 1 Turku Centre for Biotechnology, University of Turku , Turku, Finland .,10 Schools of Medical Science, Örebro University , Örebro, Sweden
| |
Collapse
|
15
|
Validation of sodium/glucose cotransporter proteins in human brain as a potential marker for temporal narrowing of the trauma formation. Int J Legal Med 2018; 133:1107-1114. [PMID: 30073510 DOI: 10.1007/s00414-018-1893-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 07/17/2018] [Indexed: 02/07/2023]
Abstract
In many forensic cases, the existence of a traumatic brain injury (TBI) is an essential factor, and the determination of the survival time is nearly as important as the determination of whether or not a trauma exists. Since it is known that glucose uptake increases in injured brain cells in order to perpetuate the neuronal integrity, this study focuses on the pathomechanism of brain glucose supply via sodium/glucose cotransporters 1 and 2 (SGLT1, SGLT2) following traumatization. Human cerebrum tissue of male and female individuals who died following TBI was taken from the contusional and contralateral regions, as well as from individuals deceased due to cardiac arrest (control group). Total SGLT1 and SGLT2 protein expression was analyzed by immunoblotting comparing injured and non-injured tissue. The immunoreactivity in contusional cerebral cortex region began to increase 3 to 7 h following traumatization. We found that both SGLT1 and SGLT2 protein expression increased significantly 37 h post-injury compared to the control group. SGLT1 rose significantly at 52 h post-injury and peaked significantly at 72 h, while SGLT2 rose significantly at 52 and 72 h after injury. By compiling these data, we predict a standard operator via SGLT expression as a comparative expression assertion to determine post-injury survival time for unknown cases. Our result suggests that SGLT1 and SGLT2 protein expression may be useful in forensic practice as an effective target to analyze the existence of a TBI and to determine the time of the traumatization.
Collapse
|
16
|
Sá-Pereira I, Roodselaar J, Couch Y, Consentino Kronka Sosthenes M, Evans MC, Anthony DC, Stolp HB. Hepatic acute phase response protects the brain from focal inflammation during postnatal window of susceptibility. Brain Behav Immun 2018; 69:486-498. [PMID: 29355821 PMCID: PMC5871396 DOI: 10.1016/j.bbi.2018.01.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 01/15/2018] [Accepted: 01/16/2018] [Indexed: 12/11/2022] Open
Abstract
Perinatal inflammation is known to contribute to neurodevelopmental diseases. Animal models of perinatal inflammation have revealed that the inflammatory response within the brain is age dependent, but the regulators of this variation remain unclear. In the adult, the peripheral acute phase response (APR) is known to be pivotal in the downstream recruitment of leukocytes to the injured brain. The relationship between perinatal brain injury and the APR has not been established. Here, we generated focal inflammation in the brain using interleukin (IL)-1β at postnatal day (P)7, P14, P21 and P56 and studied both the central nervous system (CNS) and hepatic inflammatory responses at 4 h. We found that there is a significant window of susceptibility in mice at P14, when compared to mice at P7, P21 and P56. This was reflected in increased neutrophil recruitment to the CNS, as well as an increase in blood-brain barrier permeability. To investigate phenomena underlying this window of susceptibility, we performed a dose response of IL-1β. Whilst induction of endogenous IL-1β or intercellular adhesion molecule (ICAM)-1 in the brain and induction of a hepatic APR were dose dependent, the recruitment of neutrophils and associated blood-brain barrier breakdown was inversely proportional. Furthermore, in contrast to adult animals, an additional peripheral challenge (intravenous IL-1β) reduced the degree of CNS inflammation, rather than exacerbating it. Together these results suggest a unique window of susceptibility to CNS injury, meaning that suppressing systemic inflammation after brain injury may exacerbate the damage caused, in an age-dependent manner.
Collapse
Affiliation(s)
- Inês Sá-Pereira
- Department of Pharmacology, University of Oxford, United Kingdom
| | - Jay Roodselaar
- Department of Pharmacology, University of Oxford, United Kingdom
| | - Yvonne Couch
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, United Kingdom
| | - Marcia Consentino Kronka Sosthenes
- Department of Pharmacology, University of Oxford, United Kingdom,Universidade Federal do Pará, Laboratório de Investigações em Neurodegeneração e Infecção, ICB/HUJBB, Belém, Brazil
| | - Matthew C. Evans
- Department of Pharmacology, University of Oxford, United Kingdom
| | - Daniel C. Anthony
- Department of Pharmacology, University of Oxford, United Kingdom,Corresponding author at: Department of Pharmacology, University of Oxford, Oxford OX1 3QT, United Kingdom.Department of PharmacologyUniversity of OxfordOxfordOX1 3QTUnited Kingdom
| | - Helen B. Stolp
- Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, St Thomas’ Hospital, King’s College London, United Kingdom,Royal Veterinary College, London, United Kingdom
| |
Collapse
|
17
|
Griffin ÉW, Yssel JD, O’Neill E, Ryan KJ, Boyle N, Harper P, Harkin A, Connor T. The β2-adrenoceptor agonist clenbuterol reduces the neuroinflammatory response, neutrophil infiltration and apoptosis following intra-striatal IL-1β administration to rats. Immunopharmacol Immunotoxicol 2018; 40:99-106. [DOI: 10.1080/08923973.2017.1418882] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Éadaoin W. Griffin
- Neuroimmunology Research Group, Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
- Department of Physiology, School of Medicine, Trinity College, Dublin, Ireland
| | - Justin D. Yssel
- Department of Physiology, School of Medicine, Trinity College, Dublin, Ireland
- Neuropsychopharmacology Research Group, Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Eoin O’Neill
- Neuropsychopharmacology Research Group, Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
- School of Pharmacy and Pharmaceutical Sciences, Trinity College, Dublin, Ireland
| | - Katie J. Ryan
- Neuroimmunology Research Group, Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
- Department of Physiology, School of Medicine, Trinity College, Dublin, Ireland
| | - Noreen Boyle
- Neuroimmunology Research Group, Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
- Department of Physiology, School of Medicine, Trinity College, Dublin, Ireland
| | - Peter Harper
- Neuroimmunology Research Group, Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Andrew Harkin
- Neuropsychopharmacology Research Group, Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
- School of Pharmacy and Pharmaceutical Sciences, Trinity College, Dublin, Ireland
| | - Thomas Connor
- Neuroimmunology Research Group, Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
- Department of Physiology, School of Medicine, Trinity College, Dublin, Ireland
| |
Collapse
|
18
|
Neuroimmunology of Traumatic Brain Injury: Time for a Paradigm Shift. Neuron 2017; 95:1246-1265. [PMID: 28910616 DOI: 10.1016/j.neuron.2017.07.010] [Citation(s) in RCA: 467] [Impact Index Per Article: 66.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 07/07/2017] [Accepted: 07/10/2017] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) is a leading cause of morbidity and disability, with a considerable socioeconomic burden. Heterogeneity of pathoanatomical subtypes and diversity in the pathogenesis and extent of injury contribute to differences in the course and outcome of TBI. Following the primary injury, extensive and lasting damage is sustained through a complex cascade of events referred to as "secondary injury." Neuroinflammation is proposed as an important manipulable aspect of secondary injury in animal and human studies. Because neuroinflammation can be detrimental or beneficial, before developing immunomodulatory therapies, it is necessary to better understand the timing and complexity of the immune responses that follow TBI. With a rapidly increasing body of literature, there is a need for a clear summary of TBI neuroimmunology. This review presents our current understanding of the immune response to TBI in a chronological and compartment-based manner, highlighting early changes in gene expression and initial signaling pathways that lead to activation of innate and adaptive immunity. Based on recent advances in our understanding of innate immune cell activation, we propose a new paradigm to study innate immune cells following TBI that moves away from the existing M1/M2 classification of activation states toward a stimulus- and disease-specific understanding of polarization state based on transcriptomic and proteomic profiling.
Collapse
|
19
|
Couch Y, Akbar N, Roodselaar J, Evans MC, Gardiner C, Sargent I, Romero IA, Bristow A, Buchan AM, Haughey N, Anthony DC. Circulating endothelial cell-derived extracellular vesicles mediate the acute phase response and sickness behaviour associated with CNS inflammation. Sci Rep 2017; 7:9574. [PMID: 28851955 PMCID: PMC5575066 DOI: 10.1038/s41598-017-09710-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 07/27/2017] [Indexed: 12/19/2022] Open
Abstract
Brain injury elicits a systemic acute-phase response (APR), which is responsible for co-ordinating the peripheral immunological response to injury. To date, the mechanisms responsible for signalling the presence of injury or disease to selectively activate responses in distant organs were unclear. Circulating endogenous extracellular vesicles (EVs) are increased after brain injury and have the potential to carry targeted injury signals around the body. Here, we examined the potential of EVs, isolated from rats after focal inflammatory brain lesions using IL-1β, to activate a systemic APR in recipient naïve rats, as well as the behavioural consequences of EV transfer. Focal brain lesions increased EV release, and, following isolation and transfer, the EVs were sequestered by the liver where they initiated an APR. Transfer of blood-borne EVs from brain-injured animals was also enough to suppress exploratory behaviours in recipient naïve animals. EVs derived from brain endothelial cell cultures treated with IL-1β also activated an APR and altered behaviour in recipient animals. These experiments reveal that inflammation-induced circulating EVs derived from endothelial cells are able to initiate the APR to brain injury and are sufficient to generate the associated sickness behaviours, and are the first demonstration that EVs are capable of modifying behavioural responses.
Collapse
Affiliation(s)
- Yvonne Couch
- Acute Stroke Programme, RDM-Investigative Medicine, University of Oxford, Oxford, UK.
| | - Naveed Akbar
- Division of Cardiovascular Medicine, RDM, University of Oxford, Oxford, UK
| | - Jay Roodselaar
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Matthew C Evans
- Department of Pharmacology, University of Oxford, Oxford, UK
| | | | - Ian Sargent
- Department of Obstetrics and Gynaecology, University of Oxford, Oxford, UK
| | - Ignacio A Romero
- Department of Life, Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | | | - Alastair M Buchan
- Acute Stroke Programme, RDM-Investigative Medicine, University of Oxford, Oxford, UK
| | - Norman Haughey
- Department of Neurology and Psychiatry, Johns Hopkins University, Baltimore, Maryland, USA
| | | |
Collapse
|
20
|
Dickens AM, Tovar-Y-Romo LB, Yoo SW, Trout AL, Bae M, Kanmogne M, Megra B, Williams DW, Witwer KW, Gacias M, Tabatadze N, Cole RN, Casaccia P, Berman JW, Anthony DC, Haughey NJ. Astrocyte-shed extracellular vesicles regulate the peripheral leukocyte response to inflammatory brain lesions. Sci Signal 2017; 10:10/473/eaai7696. [PMID: 28377412 PMCID: PMC5590230 DOI: 10.1126/scisignal.aai7696] [Citation(s) in RCA: 199] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Brain injury induces a peripheral acute cytokine response that directs the transmigration of leukocytes into the brain. Because this brain-to-peripheral immune communication affects patient recovery, understanding its regulation is important. Using a mouse model of inflammatory brain injury, we set out to find a soluble mediator for this phenomenon. We found that extracellular vesicles (EVs) shed from astrocytes in response to intracerebral injection of interleukin-1β (IL-1β) rapidly entered into peripheral circulation and promoted the transmigration of leukocytes through modulation of the peripheral acute cytokine response. Bioinformatic analysis of the protein and microRNA cargo of EVs identified peroxisome proliferator-activated receptor α (PPARα) as a primary molecular target of astrocyte-shed EVs. We confirmed in mice that astrocytic EVs promoted the transmigration of leukocytes into the brain by inhibiting PPARα, resulting in the increase of nuclear factor κB (NF-κB) activity that triggered the production of cytokines in liver. These findings expand our understanding of the mechanisms regulating communication between the brain and peripheral immune system and identify astrocytic EVs as a molecular regulator of the immunological response to inflammatory brain damage.
Collapse
Affiliation(s)
- Alex M Dickens
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Luis B Tovar-Y-Romo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Seung-Wan Yoo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Amanda L Trout
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mihyun Bae
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Marlene Kanmogne
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Bezawit Megra
- Departments of Pathology, Microbiology, and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Dionna W Williams
- Departments of Pathology, Microbiology, and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Kennith W Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mar Gacias
- Department of Neuroscience, Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nino Tabatadze
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Robert N Cole
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Patrizia Casaccia
- Department of Neuroscience, Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joan W Berman
- Departments of Pathology, Microbiology, and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Daniel C Anthony
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Norman J Haughey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA. .,Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
21
|
Hu G, Witwer KW, Bond VC, Haughey N, Kashanchi F, Pulliam L, Buch S. Proceedings of the ISEV symposium on "HIV, NeuroAIDS, drug abuse & EVs". J Extracell Vesicles 2017; 6:1294360. [PMID: 28800366 PMCID: PMC5373676 DOI: 10.1080/20013078.2017.1294360] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 02/06/2017] [Indexed: 12/17/2022] Open
Abstract
Extracellular vesicles (EVs) are globular, membrane bound nanovesicles (30-100 nm range) that are shed both during normal cellular functioning and under pathological conditions by most cell types. In recent years, there has been significant interest in the study of these vesicles as conduits for the delivery of information between cells from both analogous and disparate tissues. Their ability to carry specialised cargo including signalling mediators, proteins, messenger RNA and miRNAs characterises these vesicles as primary facilitators of cell-to-cell communication and regulation. EVs have also been demonstrated to play important roles in the field of cancer biology and metastasis. However, significant knowledge gaps exist in the role these vesicles play in the context of HIV infection and drug abuse. To foster discussion in this area a satellite symposium on "HIV, NeuroAIDS, Drug Abuse & EVs", was held in conjunction with the annual meeting of the International Society for Extracellular Vesicles (ISEV) in Bethesda, in April 2015. Experts in HIV and drug abuse fields were invited to share their findings on the role of EVs in HIV-1 infection and drug addiction. Additional discussion included current areas of research in EV biology in HIV infection and drug abuse.
Collapse
Affiliation(s)
- Guoku Hu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vincent C Bond
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Norman Haughey
- Department of Neurology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Psychiatry, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fatah Kashanchi
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Lynn Pulliam
- Departments of Laboratory Medicine and Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
22
|
Targeting the NF-E2-Related Factor 2 Pathway: a Novel Strategy for Traumatic Brain Injury. Mol Neurobiol 2017; 55:1773-1785. [PMID: 28224478 DOI: 10.1007/s12035-017-0456-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 02/13/2017] [Indexed: 12/30/2022]
Abstract
As an essential component of cellular defense against a variety of endogenous and exogenous stresses, nuclear factor erythroid 2-related factor 2 (Nrf2) has received increased attention in the past decades. Multiple studies indicate that Nrf2 acts not only as an important protective factor in injury models but also as a downstream target of therapeutic agents. Activation of Nrf2 has increasingly been linked to many human diseases, especially in central nervous system (CNS) injury such as traumatic brain injury (TBI). Several researches have deciphered that activation of Nrf2 exerts antioxidative stress, antiapoptosis, and antiinflammation influence in TBI via different molecules and pathways including heme oxygenase-1 (HO-1), NADPH:quinine oxidoreductase-1 (NQO-1), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), and nicotinamide adenine dinucleotide phosphate oxidase 2 (NOX2). Hence, Nrf2 shows great promise as a molecular target in TBI. In the present article, we provide an updated review of the current state of our knowledge about relationship between Nrf2 and TBI, highlighting the specific roles of Nrf2 in TBI.
Collapse
|
23
|
Nizamutdinov D, DeMorrow S, McMillin M, Kain J, Mukherjee S, Zeitouni S, Frampton G, Bricker PCS, Hurst J, Shapiro LA. Hepatic alterations are accompanied by changes to bile acid transporter-expressing neurons in the hypothalamus after traumatic brain injury. Sci Rep 2017; 7:40112. [PMID: 28106051 PMCID: PMC5247752 DOI: 10.1038/srep40112] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 12/02/2016] [Indexed: 12/29/2022] Open
Abstract
Annually, there are over 2 million incidents of traumatic brain injury (TBI) and treatment options are non-existent. While many TBI studies have focused on the brain, peripheral contributions involving the digestive and immune systems are emerging as factors involved in the various symptomology associated with TBI. We hypothesized that TBI would alter hepatic function, including bile acid system machinery in the liver and brain. The results show activation of the hepatic acute phase response by 2 hours after TBI, hepatic inflammation by 6 hours after TBI and a decrease in hepatic transcription factors, Gli 1, Gli 2, Gli 3 at 2 and 24 hrs after TBI. Bile acid receptors and transporters were decreased as early as 2 hrs after TBI until at least 24 hrs after TBI. Quantification of bile acid transporter, ASBT-expressing neurons in the hypothalamus, revealed a significant decrease following TBI. These results are the first to show such changes following a TBI, and are compatible with previous studies of the bile acid system in stroke models. The data support the emerging idea of a systemic influence to neurological disorders and point to the need for future studies to better define specific mechanisms of action.
Collapse
Affiliation(s)
- Damir Nizamutdinov
- Department of Surgery, Texas A&M University Health Science Center, College of Medicine, Temple, Texas, 76504, USA.,Department of Neurosurgery, Neuroscience Research Institute, Baylor Scott &White Health, Temple, Texas, 76504, USA
| | - Sharon DeMorrow
- Departent of Internal Medicine, Texas A&M University Health Science Center, College of Medicine, Temple, Texas, 76504, USA.,Central Texas Veterans Health Care System, Temple, Texas, 76504, USA
| | - Matthew McMillin
- Departent of Internal Medicine, Texas A&M University Health Science Center, College of Medicine, Temple, Texas, 76504, USA.,Central Texas Veterans Health Care System, Temple, Texas, 76504, USA
| | - Jessica Kain
- Department of Surgery, Texas A&M University Health Science Center, College of Medicine, Temple, Texas, 76504, USA
| | - Sanjib Mukherjee
- Department of Surgery, Texas A&M University Health Science Center, College of Medicine, Temple, Texas, 76504, USA
| | - Suzanne Zeitouni
- Department of Surgery, Texas A&M University Health Science Center, College of Medicine, Temple, Texas, 76504, USA
| | - Gabriel Frampton
- Departent of Internal Medicine, Texas A&M University Health Science Center, College of Medicine, Temple, Texas, 76504, USA.,Central Texas Veterans Health Care System, Temple, Texas, 76504, USA
| | - Paul Clint S Bricker
- Department of Surgery, Texas A&M University Health Science Center, College of Medicine, Temple, Texas, 76504, USA
| | - Jacob Hurst
- Department of Surgery, Texas A&M University Health Science Center, College of Medicine, Temple, Texas, 76504, USA
| | - Lee A Shapiro
- Department of Surgery, Texas A&M University Health Science Center, College of Medicine, Temple, Texas, 76504, USA.,Department of Neurosurgery, Neuroscience Research Institute, Baylor Scott &White Health, Temple, Texas, 76504, USA
| |
Collapse
|
24
|
Brady RD, Shultz SR, Sun M, Romano T, van der Poel C, Wright DK, Wark JD, O'Brien TJ, Grills BL, McDonald SJ. Experimental Traumatic Brain Injury Induces Bone Loss in Rats. J Neurotrauma 2016; 33:2154-2160. [PMID: 25686841 DOI: 10.1089/neu.2014.3836] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Few studies have investigated the influence of traumatic brain injury (TBI) on bone homeostasis; however, pathophysiological mechanisms involved in TBI have potential to be detrimental to bone. The current study assessed the effect of experimental TBI in rats on the quantity and quality of two different weight-bearing bones, the femur and humerus. Rats were randomly assigned into either sham or lateral fluid percussion injury (FPI) groups. Open-field testing to assess locomotion was conducted at 1, 4, and 12 weeks post-injury, with the rats killed at 1 and 12 weeks post-injury. Bones were analyzed using peripheral quantitative computed tomography (pQCT), histomorphometric analysis, and three-point bending. pQCT analysis revealed that at 1 and 12 weeks post-injury, the distal metaphyseal region of femora from FPI rats had reduced cortical content (10% decrease at 1 week, 8% decrease at 12 weeks; p < 0.01) and cortical thickness (10% decrease at 1 week, 11% decrease at 12 weeks p < 0.001). There was also a 23% reduction in trabecular bone volume ratio at 1 week post-injury and a 27% reduction at 12 weeks post-injury in FPI rats compared to sham (p < 0.001). There were no differences in bone quantity and mechanical properties of the femoral midshaft between sham and TBI animals. There were no differences in locomotor outcomes, which suggested that post-TBI changes in bone were not attributed to immobility. Taken together, these findings indicate that this rat model of TBI was detrimental to bone and suggests a link between TBI and altered bone remodeling.
Collapse
Affiliation(s)
- Rhys D Brady
- 1 Department of Physiology, Anatomy and Microbiology, La Trobe University , Bundoora, VIC, Australia
| | - Sandy R Shultz
- 2 Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne , Parkville, VIC, Australia
| | - Mujun Sun
- 2 Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne , Parkville, VIC, Australia
| | - Tania Romano
- 1 Department of Physiology, Anatomy and Microbiology, La Trobe University , Bundoora, VIC, Australia
| | - Chris van der Poel
- 1 Department of Physiology, Anatomy and Microbiology, La Trobe University , Bundoora, VIC, Australia
| | - David K Wright
- 3 Anatomy and Neuroscience, The University of Melbourne , Parkville, VIC, Australia .,4 The Florey Institute of Neuroscience and Mental Health, The University of Melbourne , Parkville, VIC, Australia
| | - John D Wark
- 2 Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne , Parkville, VIC, Australia
| | - Terence J O'Brien
- 2 Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne , Parkville, VIC, Australia
| | - Brian L Grills
- 1 Department of Physiology, Anatomy and Microbiology, La Trobe University , Bundoora, VIC, Australia
| | - Stuart J McDonald
- 1 Department of Physiology, Anatomy and Microbiology, La Trobe University , Bundoora, VIC, Australia
| |
Collapse
|
25
|
Powner DJ, Miller ER, Levine RL. CVP and PAoP Measurements Are Discordant During Fluid Therapy After Traumatic Brain Injury. J Intensive Care Med 2016; 20:28-33. [PMID: 15665257 DOI: 10.1177/0885066604271750] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The objective of the study was to compare measurements of central venous pressure (CVP) and pulmonary artery occlusion pressures (PAoP) as estimates of intravascular volume during the first 96 hours of fluid therapy after traumatic brain injury (TBI). One thousand five hundred ten simultaneous CVP and PAoP measurements from 31 patients entered into the National Acute Brain Injury Study: Hypothermia (NABISH:H) protocol were retrospectively compared. The effect of fluid administration and body temperature upon the paired measurements was statistically assessed. Agreement between CVP and PAoP values was poor. The CVP and PAoP were equal in only 11% of paired values. The CVP was always higher than PAoP in 1 patient, whereas PAoP always exceeded the CVP in 5 others. In 74% of the pairs, the PAoP was higher than the CVP, whereas in 15%, CVP was greater than PAoP. For any CVP measurement, the PAoP was either 3 mm Hg above or below the CVP in 67% of the pairs and at least 5 mm Hg above or below the CVP in 21% of the pairs. In 21 (68%) patients, PAoP was= 5 mm Hg above CVP in more than 4 readings, a clinically important difference. Discordance was not attributed to the fluid administered or to the temperature protocol. Neurological outcome appears affected by the volume of fluid administration. However, during initial therapy, estimates of intravascular volume provided by the CVP and PAoP are discordant. Although documented in other clinical conditions, the disparity noted here after TBI has not been previously reported. Assessment of intravascular volume to avoid hypovolemia should utilize other measurement techniques.
Collapse
Affiliation(s)
- David J Powner
- Department of Neurosurgery, University of Texas Health Science Center, Houston, TX 77030, USA.
| | | | | |
Collapse
|
26
|
Glushakov AV, Arias RA, Tolosano E, Doré S. Age-Dependent Effects of Haptoglobin Deletion in Neurobehavioral and Anatomical Outcomes Following Traumatic Brain Injury. Front Mol Biosci 2016; 3:34. [PMID: 27486583 PMCID: PMC4949397 DOI: 10.3389/fmolb.2016.00034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 07/05/2016] [Indexed: 12/11/2022] Open
Abstract
Cerebral hemorrhages are common features of traumatic brain injury (TBI) and their presence is associated with chronic disabilities. Recent clinical and experimental evidence suggests that haptoglobin (Hp), an endogenous hemoglobin-binding protein most abundant in blood plasma, is involved in the intrinsic molecular defensive mechanism, though its role in TBI is poorly understood. The aim of this study was to investigate the effects of Hp deletion on the anatomical and behavioral outcomes in the controlled cortical impact model using wildtype (WT) C57BL/6 mice and genetically modified mice lacking the Hp gene (Hp(-∕-)) in two age cohorts [2-4 mo-old (young adult) and 7-8 mo-old (older adult)]. The data obtained suggest age-dependent significant effects on behavioral and anatomical TBI outcomes and recovery from injury. Moreover, in the adult cohort, neurological deficits in Hp(-∕-) mice at 24 h were significantly improved compared to WT, whereas there were no significant differences in brain pathology between these genotypes. In contrast, in the older adult cohort, Hp(-∕-) mice had significantly larger lesion volumes compared to WT, but neurological deficits were not significantly different. Immunohistochemistry for ionized calcium-binding adapter molecule 1 (Iba1) and glial fibrillary acidic protein (GFAP) revealed significant differences in microglial and astrocytic reactivity between Hp(-∕-) and WT in selected brain regions of the adult but not the older adult-aged cohort. In conclusion, the data obtained in the study provide clarification on the age-dependent aspects of the intrinsic defensive mechanisms involving Hp that might be involved in complex pathways differentially affecting acute brain trauma outcomes.
Collapse
Affiliation(s)
- Alexander V Glushakov
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine Gainesville, FL, USA
| | - Rodrigo A Arias
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine Gainesville, FL, USA
| | - Emanuela Tolosano
- Departments of Molecular Biotechnology and Health Sciences, University of Torino Torino, Italy
| | - Sylvain Doré
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, University of Florida College of MedicineGainesville, FL, USA; Departments of Anesthesiology, Neurology, Psychiatry, Psychology, Pharmaceutics and Neuroscience, University of Florida College of MedicineGainesville, FL, USA
| |
Collapse
|
27
|
Di Battista AP, Rhind SG, Hutchison MG, Hassan S, Shiu MY, Inaba K, Topolovec-Vranic J, Neto AC, Rizoli SB, Baker AJ. Inflammatory cytokine and chemokine profiles are associated with patient outcome and the hyperadrenergic state following acute brain injury. J Neuroinflammation 2016; 13:40. [PMID: 26883121 PMCID: PMC4754875 DOI: 10.1186/s12974-016-0500-3] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Accepted: 02/01/2016] [Indexed: 01/25/2023] Open
Abstract
Background Traumatic brain injury (TBI) elicits intense sympathetic nervous system (SNS) activation with profuse catecholamine secretion. The resultant hyperadrenergic state is linked to immunomodulation both within the brain and systemically. Dysregulated inflammation post-TBI exacerbates secondary brain injury and contributes to unfavorable patient outcomes including death. The aim of this study was to characterize the early dynamic profile of circulating inflammatory cytokines/chemokines in patients admitted for moderate-to-severe TBI, to examine interrelationships between these mediators and catecholamines, as well as clinical indices of injury severity and neurological outcome. Methods Blood was sampled from 166 isolated TBI patients (aged 45 ± 20.3 years; 74.7 % male) on admission, 6-, 12-, and 24-h post-injury and from healthy controls (N = 21). Plasma cytokine [interleukin (IL)-1β, -2, -4, -5, -10, -12p70, -13, tumor necrosis factor (TNF)-α, interferon (IFN)-γ] and chemokine [IL-8, eotaxin, eotaxin-3, IFN-γ-induced protein (IP)-10, monocyte chemoattractant protein (MCP)-1, -4, macrophage-derived chemokine (MDC), macrophage inflammatory protein (MIP)-1β, thymus activation regulated chemokine (TARC)] concentrations were analyzed using high-sensitivity electrochemiluminescence multiplex immunoassays. Plasma catecholamines [epinephrine (Epi), norepinephrine (NE)] were measured by immunoassay. Neurological outcome at 6 months was assessed using the extended Glasgow outcome scale (GOSE) dichotomized as good (>4) or poor (≤4) outcomes. Results Patients showed altered levels of IL-10 and all chemokines assayed relative to controls. Significant differences in a number of markers were evident between moderate and severe TBI cohorts. Elevated IL-8, IL-10, and TNF-α, as well as alterations in 8 of 9 chemokines, were associated with poor outcome at 6 months. Notably, a positive association was found between Epi and IL-1β, IL-10, Eotaxin, IL-8, and MCP-1. NE was positively associated with IL-1β, IL-10, TNF-α, eotaxin, IL-8, IP-10, and MCP-1. Conclusions Our results provide further evidence that exaggerated SNS activation acutely after isolated TBI in humans may contribute to harmful peripheral inflammatory cytokine/chemokine dysregulation. These findings are consistent with a potentially beneficial role for therapies aimed at modulating the inflammatory response and hyperadrenergic state acutely post-injury. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0500-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alex P Di Battista
- Defence Research & Development Canada, Toronto Research Centre, Toronto, ON, Canada. .,Institute of Medical Science, University of Toronto, Toronto, ON, Canada.
| | - Shawn G Rhind
- Defence Research & Development Canada, Toronto Research Centre, Toronto, ON, Canada. .,Faculty of Kinesiology & Physical Education, University of Toronto, Toronto, ON, Canada.
| | - Michael G Hutchison
- Faculty of Kinesiology & Physical Education, University of Toronto, Toronto, ON, Canada. .,Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hopsital, Toronto, ON, Canada.
| | - Syed Hassan
- Defence Research & Development Canada, Toronto Research Centre, Toronto, ON, Canada. .,Institute of Medical Science, University of Toronto, Toronto, ON, Canada.
| | - Maria Y Shiu
- Defence Research & Development Canada, Toronto Research Centre, Toronto, ON, Canada. .,Faculty of Kinesiology & Physical Education, University of Toronto, Toronto, ON, Canada.
| | - Kenji Inaba
- Division of Trauma & Critical Care, University of Southern California, Los Angeles, CA, USA. .,LA County+ USC Medical Center, Los Angeles, CA, USA.
| | - Jane Topolovec-Vranic
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hopsital, Toronto, ON, Canada.
| | | | - Sandro B Rizoli
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hopsital, Toronto, ON, Canada. .,Department of Critical Care, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada. .,Department of Anesthesia, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada. .,Department of Surgery, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada.
| | - Andrew J Baker
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada. .,Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hopsital, Toronto, ON, Canada. .,Department of Critical Care, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada. .,Department of Anesthesia, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada. .,Department of Surgery, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
28
|
Wang L, Liu L, Pan Z, Zeng Y. Serum leptin, bone mineral density and the healing of long bone fractures in men with spinal cord injury. Bosn J Basic Med Sci 2015; 15:69-74. [PMID: 26614856 DOI: 10.17305/bjbms.2015.693] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 10/16/2015] [Accepted: 10/17/2015] [Indexed: 11/16/2022] Open
Abstract
Previously reported fracture rates in patients with spinal cord injury range from 1% to 20%. However, the exact role of spinal cord injury in bone metabolism has not yet been clarified. In order to investigate the effects of serum leptin and bone mineral density on the healing of long bone fractures in men with spinal cord injury, 15 male SCI patients and 15 matched controls were involved in our study. The outcome indicated that at 4 and 8 weeks after bone fracture, callus production in patients with spinal cord injury was lower than that in controls. Besides, bone mineral density was significantly reduced at 2, 4 and 8 weeks. In addition, it was found that at each time point, patients with spinal cord injury had significantly higher serum leptin levels than controls and no association was found between serum leptin level and bone mineral density of lumbar vertebrae. Moreover, bone mineral density was positively correlated with bone formation in both of the groups. These findings suggest that in early phases i.e. week 4 and 8, fracture healing was impaired in patients with spinal cord injury and that various factors participated in the complicated healing process, such as hormonal and mechanical factors.
Collapse
Affiliation(s)
- Lei Wang
- Department of Orthopedics, the Affiliated People's Hospital of Jiangsu University, China.
| | | | | | | |
Collapse
|
29
|
Weaver LC, Bao F, Dekaban GA, Hryciw T, Shultz SR, Cain DP, Brown A. CD11d integrin blockade reduces the systemic inflammatory response syndrome after traumatic brain injury in rats. Exp Neurol 2015; 271:409-22. [PMID: 26169930 DOI: 10.1016/j.expneurol.2015.07.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 07/04/2015] [Indexed: 12/15/2022]
Abstract
Traumatic CNS injury triggers a systemic inflammatory response syndrome (SIRS), in which circulating inflammatory cells invade body organs causing local inflammation and tissue damage. We have shown that the SIRS caused by spinal cord injury is greatly reduced by acute intravenous treatment with an antibody against the CD11d subunit of the CD11d/CD18 integrin expressed by neutrophils and monocyte/macrophages, a treatment that reduces their efflux from the circulation. Traumatic brain injury (TBI) is a frequently occurring injury after motor vehicle accidents, sporting and military injuries, and falls. Our studies have shown that the anti-CD11d treatment diminishes brain inflammation and oxidative injury after moderate or mild TBI, improving neurological outcomes. Accordingly, we examined the impact of this treatment on the SIRS triggered by TBI. The anti-CD11d treatment was given at 2h after a single moderate (2.5-3.0 atm) or 2 and 24h after each of three consecutive mild (1.0-1.5 atm) fluid percussion TBIs. Sham-injured, saline-treated rats served as controls. At 24h, 72 h, and 4 or 8 weeks after the single TBI and after the third of three TBIs, lungs of rats were examined histochemically, immunocytochemically and biochemically for downstream effects of SIRS including inflammation, tissue damage and expression of oxidative enzymes. Lung sections revealed that both the single moderate and repeated mild TBI caused alveolar disruption, thickening of inter-alveolar tissue, hemorrhage into the parenchyma and increased density of intra-and peri-alveolar macrophages. The anti-CD11d treatment decreased the intrapulmonary influx of neutrophils and the density of activated macrophages and the activity of myeloperoxidase after these TBIs. Moreover, Western blotting studies showed that the treatment decreased lung protein levels of oxidative enzymes gp91(phox), inducible nitric oxide synthase and cyclooxygenase-2, as well as the apoptotic pathway enzyme caspase-3 and levels of 4-hydroxynonenal-bound proteins (an indicator of lipid peroxidation). Decreased expression of the cytoprotective transcription factor Nrf2 reflected decreased lung oxidative stress. Anti-CD11d treatment also diminished the lung concentration of free radicals and tissue aldehydes. In conclusion, the substantial lung component of the SIRS after single or repeated TBIs is significantly decreased by a simple, minimally invasive and short-lasting anti-inflammatory treatment.
Collapse
Affiliation(s)
- Lynne C Weaver
- Spinal Cord Injury Team, Molecular Medicine, Robarts Research Institute, The University of Western Ontario, London, Ontario N6A 5B7, Canada; Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario N6A 5B7, Canada.
| | - Feng Bao
- Spinal Cord Injury Team, Molecular Medicine, Robarts Research Institute, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Gregory A Dekaban
- Spinal Cord Injury Team, Molecular Medicine, Robarts Research Institute, The University of Western Ontario, London, Ontario N6A 5B7, Canada; Department of Microbiology and Immunology, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Todd Hryciw
- Spinal Cord Injury Team, Molecular Medicine, Robarts Research Institute, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Sandy R Shultz
- Program in Neuroscience, The University of Western Ontario, London, Ontario N6A 5B7, Canada; Department of Psychology, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Donald P Cain
- Program in Neuroscience, The University of Western Ontario, London, Ontario N6A 5B7, Canada; Department of Psychology, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Arthur Brown
- Spinal Cord Injury Team, Molecular Medicine, Robarts Research Institute, The University of Western Ontario, London, Ontario N6A 5B7, Canada; Program in Neuroscience, The University of Western Ontario, London, Ontario N6A 5B7, Canada; Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| |
Collapse
|
30
|
Ansari MA. Temporal profile of M1 and M2 responses in the hippocampus following early 24h of neurotrauma. J Neurol Sci 2015; 357:41-9. [PMID: 26148932 DOI: 10.1016/j.jns.2015.06.062] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 06/10/2015] [Accepted: 06/26/2015] [Indexed: 01/16/2023]
Abstract
Traumatic brain injury (TBI) elicits complex inflammatory assets (M1 and M2 responses) in the brain that include the expression of various cytokines/chemokines and the recruitment of blood cells, contributing secondary injury cascades (SIC), and also recovery processes. The modulation of such inflammatory assets might be a therapeutic option following TBI. The present study assesses a temporal profile of various molecular markers of M1 and M2 response in the hippocampus after TBI. Following a unilateral controlled cortical impact (CCI) on young rats, hippocampal tissues of each brain were harvested at 2, 4, 6, 10, and 24h post trauma. Including shams (craniotomy only), half of the rats were assessed for gene expression and half for the protein of various markers for M1 [interferon-gamma (IFNγ), tumor necrosis factor-α (TNFα), interleukin (IL)-1-β (IL-1β), and IL-6] and M2 [IL-4, IL-10, IL-13, arginase 1 (Arg1), YM1, FIZZ1, and mannose receptor C-1 (MRC1)] responses. Analysis revealed that molecular markers of M1 and M2 responses have heterogeneous injury effects in the hippocampus and that "time-post-injury" is an important factor in determining inflammation status. With the heterogeneous gene expression of pro-inflammatory cytokines, M1 response was significantly elevated at 2h and declined at 24h after TBI, however, their levels remained higher than the sham rats. Except IFNγ, proteins of M1 cytokines were significantly elevated in the first 24h, and peaked between 2-6h [TNFα (2h), IL-1β (6h), and IL-6 (4-6h)]. With the heterogeneous relative gene expression of Arg1, YM1, FIZZ1, and MRC1, levels of M2 cytokines were peaked at 24h post TBI. IL-10 and IL-13 expression appeared biphasic in the first 24h. Protein values of IL-4 and IL-13 peaked at 24h and IL-10 at 6h post injury. Results suggest that the M1 response rises rapidly after injury and overpowers the initial, comparatively smaller, or transient M2 response. A treatment that can modulate inflammation, reduce SIC, and improve recovery should be initiated early (within 10h) after TBI.
Collapse
Affiliation(s)
- Mubeen A Ansari
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA; Spinal Cord Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
31
|
Losey P, Ladds E, Laprais M, Guevel B, Burns L, Bordet R, Anthony DC. The role of PPAR activation during the systemic response to brain injury. J Neuroinflammation 2015; 12:99. [PMID: 25994490 PMCID: PMC4450490 DOI: 10.1186/s12974-015-0295-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 04/02/2015] [Indexed: 01/22/2023] Open
Abstract
Background Fenofibrate, a PPAR-α activator, has shown promising results as a neuroprotective therapy, with proposed anti-inflammatory and anti-oxidant effects. However, it displays poor blood-brain barrier permeability leading to some ambiguity over its mechanism of action. Experimentally induced brain injury has been shown to elicit a hepatic acute phase response that modulates leukocyte recruitment to the injured brain. Here, we sought to discover whether one effect of fenofibrate might include the suppression of the acute phase response (APR) following brain injury. Methods A 1-h intraluminal thread middle cerebral artery occlusion (MCAO) model followed by a 6-h reperfusion was performed in C57/BL6 mice. Quantitative reverse transcriptase-polymerase chain reaction was then used to measure hepatic expression of chemokine (C-X-C motif) ligand 1 (CXCL1), chemokine ligand 10 (CXCL10) and serum amyloid A-1 (SAA-1), and immunohistochemical analysis was used to quantify brain and hepatic neutrophil infiltration following stroke. Results The MCAO and sham surgery induced the expression of all three acute phase reactants. A 14-day fenofibrate pre-treatment decreased reactant production, infarct volume, and neutrophil recruitment to the brain and liver, which is a hallmark of the APR. Conclusions The data highlight a novel mechanism of action for fenofibrate and lend further evidence towards the promotion of its use as a prophylactic therapy in patients at risk of cerebral ischaemia. Further research is required to elucidate the mechanistic explanation underlying its actions.
Collapse
Affiliation(s)
- Patrick Losey
- Department of Pharmacology, Experimental Neuropathology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK.,EA 1046, Pharmacology, Faculty of Medicine, Research Branch, IMPRT, University of Lille North of France, Place de Verdun, Lille, Cedex, 59045, France
| | - Emma Ladds
- North Bristol NHS Trust, Southmead Road, Bristol, BS10 5NB, UK
| | - Maud Laprais
- EA 1046, Pharmacology, Faculty of Medicine, Research Branch, IMPRT, University of Lille North of France, Place de Verdun, Lille, Cedex, 59045, France
| | - Borna Guevel
- Department of Pharmacology, Experimental Neuropathology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Laura Burns
- Department of Pharmacology, Experimental Neuropathology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Regis Bordet
- EA 1046, Pharmacology, Faculty of Medicine, Research Branch, IMPRT, University of Lille North of France, Place de Verdun, Lille, Cedex, 59045, France.
| | - Daniel C Anthony
- Department of Pharmacology, Experimental Neuropathology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK. .,EA 1046, Pharmacology, Faculty of Medicine, Research Branch, IMPRT, University of Lille North of France, Place de Verdun, Lille, Cedex, 59045, France.
| |
Collapse
|
32
|
Sauerbeck AD, Laws JL, Bandaru VVR, Popovich PG, Haughey NJ, McTigue DM. Spinal cord injury causes chronic liver pathology in rats. J Neurotrauma 2014; 32:159-69. [PMID: 25036371 DOI: 10.1089/neu.2014.3497] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Traumatic spinal cord injury (SCI) causes major disruption to peripheral organ innervation and regulation. Relatively little work has investigated these post-SCI systemic changes, however, despite considerable evidence that multiple organ system dysfunction contributes to chronic impairments in health. Because metabolic dysfunction is common after SCI and the liver is a pivotal site for metabolic homeostasis, we sought to determine if liver pathology occurs as a result of SCI in a rat spinal contusion model. Histologic evidence showed excess lipid accumulation in the liver for at least 21 days post-injury after cervical or midthoracic SCI. Lipidomic analysis revealed an acute increase in hepatic ceramides as well as chronically elevated lactosylceramide. Post-SCI hepatic changes also included increased proinflammatory gene expression, including interleukin (IL)-1α, IL-1β, chemokine ligand-2, and tumor necrosis factor-α mRNA. These were coincident with increased CD68+ macrophages in the liver through 21 days post-injury. Serum alanine transaminase, used clinically to detect liver damage, was significantly increased at 21 days post-injury, suggesting that early metabolic and inflammatory damage preceded overt liver pathology. Surprisingly, liver inflammation was even detected after lumbar SCI. Collectively, these results suggest that SCI produces chronic liver injury with symptoms strikingly similar to those of nonalcoholic steatohepatitis (fatty liver disease). These clinically significant hepatic changes after SCI are known to contribute to systemic inflammation, cardiovascular disease, and metabolic syndrome, all of which are more prevalent in persons with SCI. Targeting acute and prolonged hepatic pathology may improve recovery and reduce long-term complications after SCI.
Collapse
Affiliation(s)
- Andrew D Sauerbeck
- 1 Department of Neuroscience, The Ohio State University , Columbus, Ohio
| | | | | | | | | | | |
Collapse
|
33
|
Couch Y, Davis AE, Sá-Pereira I, Campbell SJ, Anthony DC. Viral pre-challenge increases central nervous system inflammation after intracranial interleukin-1β injection. J Neuroinflammation 2014; 11:178. [PMID: 25323767 PMCID: PMC4201684 DOI: 10.1186/s12974-014-0178-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 10/01/2014] [Indexed: 12/29/2022] Open
Abstract
Introduction Systemic inflammation has been shown to significantly worsen the outcome of neurological disease. However, after acute injuries to the brain both pre- and post-conditioning with bacterial endotoxin has been shown to reduce leukocyte recruitment to the CNS. Here, we sought to determine whether viral pre-challenge would have an effect on the outcome of acute CNS inflammation that was distinct from endotoxin. Methods Animals received a single intracranial microinjection of IL-1β in the presence or absence of a viral pre-challenge 24 hours prior to surgery. Liver and brain tissue were analysed for chemokine expression by qRT-PCR and leukocyte and monocyte infiltration 12 hours, 3 days and 7 days after the IL-1β injection. Results Here, a single injection of adenovirus prior to IL-1β injection resulted in adhesion molecule expression, chemokine expression and the recruitment of neutrophils to the injured CNS in significantly higher numbers than in IL-1β injected animals. The distribution and persistence of leukocytes within the CNS was also greater after pre-challenge, with neutrophils being found in both the ipsilateral and contralateral hemispheres. Thus, despite the absence of virus within the CNS, the presence of virus within the periphery was sufficient to exacerbate CNS disease. Conclusions These data suggest that the effect of a peripheral inflammatory challenge on the outcome of CNS injury or disease is not generic and will be highly dependent on the nature of the pathogen. Electronic supplementary material The online version of this article (doi:10.1186/s12974-014-0178-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | - Daniel C Anthony
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK.
| |
Collapse
|
34
|
Wang L, Yao X, Xiao L, Tang X, Ding H, Zhang H, Yuan J. The effects of spinal cord injury on bone healing in patients with femoral fractures. J Spinal Cord Med 2014; 37:414-9. [PMID: 24621032 PMCID: PMC4116725 DOI: 10.1179/2045772313y.0000000155] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
BACKGROUND Orthopedic literature states that fractures of long bones, when associated with traumatic brain injuries, frequently heal with excessive callus and faster than normal. Few studies, however, have reported these phenomena being induced by spinal cord injury (SCI). Our objective is to compare the extent of callus and the rate of healing of long-bone fractures in patients with or without SCI. Subgroup comparisons were performed among the patients with SCI in terms of different levels of SCI. METHODS The final mean volume of callus formation and the rate of union of nailed fractures of the femur were determined radiologically in 22 femoral fracture patients with SCI (seven cervical, six thoracic, and nine lumbar spine injury) and compared with those in a group of 22 patients with similar types of fractures but without SCI. RESULTS The final mean callus volume in the fracture/SCI group was significantly higher than the fracture-only group (P < 0.001). The fractures in the fracture/SCI group united in an average time of 22.86 weeks compared with 25.04 weeks in the fracture-only group (P < 0.05). We observed 84.6% (11 of 13) of patients with cervical and thoracic SCI patients with accelerated fracture healing (cervical 6 of 7, thoracic 5 of 6), but only 44.4% (4 of 9) of patients with lumbar SCI appeared to show this phenomenon (P < 0.05). CONCLUSIONS These results confirm that SCI may be associated with accelerated fracture healing and enhanced callus formation. Furthermore, our study revealed a trend toward enhanced osteogenesis in cervical or thoracic SCI compared with lumbar SCI.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jishan Yuan
- Correspondence to: Yuan Jishan, The Affiliated People's Hospital with Jiangsu University, Zhenjiang, Jiangsu Province, China.
| |
Collapse
|
35
|
Meling S, Kvalheim OM, Arneberg R, Bårdsen K, Hjelle A, Ulvund MJ. Investigation of serum protein profiles in scrapie infected sheep by means of SELDI-TOF-MS and multivariate data analysis. BMC Res Notes 2013; 6:466. [PMID: 24229425 PMCID: PMC3843553 DOI: 10.1186/1756-0500-6-466] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 11/04/2013] [Indexed: 01/18/2023] Open
Abstract
Background Classical scrapie in sheep is a fatal neurodegenerative disease associated with the conversion PrPC to PrPSc. Much is known about genetic susceptibility, uptake and dissemination of PrPSc in the body, but many aspects of prion diseases are still unknown. Different proteomic techniques have been used during the last decade to investigate differences in protein profiles between affected animals and healthy controls. We have investigated the protein profiles in serum of sheep with scrapie and healthy controls by SELDI-TOF-MS and LC-MS/MS. Latent Variable methods such as Principal Component Analysis, Partial Least Squares-Discriminant Analysis and Target Projection methods were used to describe the MS data. Results The serum proteomic profiles showed variable differences between the groups both throughout the incubation period and at the clinical end stage of scrapie. At the end stage, the target projection model separated the two groups with a sensitivity of 97.8%, and serum amyloid A was identified as one of the protein peaks that differed significantly between the groups. Conclusions At the clinical end stage of classical scrapie, ten SELDI peaks significantly discriminated the scrapie group from the healthy controls. During the non-clinical incubation period, individual SELDI peaks were differently expressed between the groups at different time points. Investigations of differences in -omic profiles can contribute to new insights into the underlying disease processes and pathways, and advance our understanding of prion diseases, but comparison and validation across laboratories is difficult and challenging.
Collapse
Affiliation(s)
- Siv Meling
- Department of Production Animal Clinical Sciences, Section for Small Ruminant Research, Norwegian School of Veterinary Science, Kyrkjevegen 332-334, N-4325, Sandnes, Norway.
| | | | | | | | | | | |
Collapse
|
36
|
Dąbrowski R, Kostro K, Szczubiał M. Concentrations of C-reactive protein, serum amyloid A, and haptoglobin in uterine arterial and peripheral blood in bitches with pyometra. Theriogenology 2013; 80:494-7. [DOI: 10.1016/j.theriogenology.2013.05.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 05/16/2013] [Accepted: 05/16/2013] [Indexed: 10/26/2022]
|
37
|
Liao Y, Liu P, Guo F, Zhang ZY, Zhang Z. Oxidative burst of circulating neutrophils following traumatic brain injury in human. PLoS One 2013; 8:e68963. [PMID: 23894384 PMCID: PMC3722225 DOI: 10.1371/journal.pone.0068963] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 06/04/2013] [Indexed: 12/16/2022] Open
Abstract
Besides secondary injury at the lesional site, Traumatic brain injury (TBI) can cause a systemic inflammatory response, which may cause damage to initially unaffected organs and potentially further exacerbate the original injury. Here we investigated plasma levels of important inflammatory mediators, oxidative activity of circulating leukocytes, particularly focusing on neutrophils, from TBI subjects and control subjects with general trauma from 6 hours to 2 weeks following injury, comparing with values from uninjured subjects. We observed increased plasma level of inflammatory cytokines/molecules TNF-α, IL-6 and CRP, dramatically increased circulating leukocyte counts and elevated expression of TNF-α and iNOS in circulating leukocytes from TBI patients, which suggests a systemic inflammatory response following TBI. Our data further showed increased free radical production in leukocyte homogenates and elevated expression of key oxidative enzymes iNOS, COX-2 and NADPH oxidase (gp91phox) in circulating leukocytes, indicating an intense induction of oxidative burst following TBI, which is significantly greater than that in control subjects with general trauma. Furthermore, flow cytometry assay proved neutrophils as the largest population in circulation after TBI and showed significantly up-regulated oxidative activity and suppressed phagocytosis rate for circulating neutrophils following brain trauma. It suggests that the highly activated neutrophils might play an important role in the secondary damage, even outside the injured brain. Taken together, the potent systemic inflammatory response induced by TBI, especially the intensively increase oxidative activity of circulating leukocytes, mainly neutrophils, may lead to a systemic damage, dysfunction/damage of bystander tissues/organs and even further exacerbate secondary local damage. Controlling these pathophysiological processes may be a promising therapeutic strategy and will protect unaffected organs and the injured brain from the secondary damage.
Collapse
Affiliation(s)
- Yiliu Liao
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Peng Liu
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Fangyuan Guo
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Zhi-Yuan Zhang
- Department of Neuropathology, University of Tuebingen, Tuebingen, Germany
- * E-mail: (ZYZ); (ZZ)
| | - Zhiren Zhang
- Institute of Immunology, Third Military Medical University of PLA, Chongqing, People’s Republic of China
- * E-mail: (ZYZ); (ZZ)
| |
Collapse
|
38
|
Bonestroo HJC, Nijboer CHA, van Velthoven CTJ, Kavelaars A, Hack CE, van Bel F, Heijnen CJ. Cerebral and hepatic inflammatory response after neonatal hypoxia-ischemia in newborn rats. Dev Neurosci 2013; 35:197-211. [PMID: 23689428 DOI: 10.1159/000346685] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 12/19/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Neonatal encephalopathy induced by perinatal asphyxia is a serious condition associated with high mortality and morbidity. Inflammation after the insult is thought to contribute to brain injury. This inflammatory response to hypoxia-ischemia (HI) may not only occur in the brain but also in peripheral organs. The aim of the present study was to investigate the effect of neonatal HI on the inflammatory response in the liver in comparison to inflammation in the brain. METHODS HI was induced in P7 Wistar rats by unilateral carotid artery occlusion and hypoxia. Cytokine and chemokine mRNA levels were determined in the brain and liver by quantitative PCR. Polarization of brain macrophages to the M1/M2-like phenotype and infiltration of neutrophils were characterized by immunohistochemistry. RESULTS 3 h after HI, an upregulation of the proinflammatory cytokines TNF-α and IL-1β and anti-inflammatory IL-10 was observed in the ipsilateral hemisphere of the brain compared to mRNA levels in sham-operated animals. Additionally, cerebral CINC-1 and MCP-1 mRNA expressions were increased. We also observed increased numbers of macrophages/microglia of the M1-like phenotype as well as a small increase in granulocyte influx in the ipsilateral hemisphere. Conversely, in the liver 3 h after HI, a downregulation of TNF-α, IL-1β, and MCP-1 and a trend towards an upregulation of IL-10 were observed compared to mRNA levels of sham-operated animals. However, hepatic CINC-1 expression was increased compared to levels in sham-operated animals. Following systemic hypoxia only, no significant changes in the expression of TNF-α, CINC-1 or MCP-1 were observed in the liver compared to sham-operated littermates, except for an upregulation in hepatic IL-1β expression 3 h after hypoxia. Twenty-four hours after insult, cerebral ipsilateral TNF-α, MCP-1 and CINC-1 mRNA expression was still increased, together with an increase in TGF-β expression. Moreover, an increase in macrophages/microglia of the M1-like phenotype was observed together with the appearance of macrophages/microglia of the M2-like phenotype around the cerebral lesion as well as an increase in granulocyte influx in comparison to 3 h after HI. In the liver, 24 h after HI, cytokine and chemokine responses were similar to mRNA levels in sham-operated animals except for a decrease in IL-10 and MCP-1. CONCLUSION We describe for the first time that brain damage following neonatal HI induces an early downregulation of the proinflammatory response in the liver. HI induces an early proinflammatory response in the brain with a concomitant increase in influx of neutrophils and polarization of macrophages/microglia to the M1-like phenotype starting at 3 h and increasing up to 24 h after HI. The inflammatory state of the brain changes after 24 h, with an increase in the anti-inflammatory cytokine TGF-β together with the appearance of macrophages/microglia of the M2-like phenotype. The downregulation of proinflammatory cytokines in the liver is not due to systemic hypoxia only, but is induced by the cerebral damage.
Collapse
Affiliation(s)
- H J C Bonestroo
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
39
|
Abrams MB, Nilsson I, Lewandowski SA, Kjell J, Codeluppi S, Olson L, Eriksson U. Imatinib enhances functional outcome after spinal cord injury. PLoS One 2012; 7:e38760. [PMID: 22723886 PMCID: PMC3378614 DOI: 10.1371/journal.pone.0038760] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 05/12/2012] [Indexed: 01/24/2023] Open
Abstract
We investigated whether imatinib (Gleevec®, Novartis), a tyrosine kinase inhibitor, could improve functional outcome in experimental spinal cord injury. Rats subjected to contusion spinal cord injury were treated orally with imatinib for 5 days beginning 30 minutes after injury. We found that imatinib significantly enhanced blood-spinal cord-barrier integrity, hindlimb locomotor function, sensorimotor integration, and bladder function, as well as attenuated astrogliosis and deposition of chondroitin sulfate proteoglycans, and increased tissue preservation. These improvements were associated with enhanced vascular integrity and reduced inflammation. Our results show that imatinib improves recovery in spinal cord injury by preserving axons and other spinal cord tissue components. The rapid time course of these beneficial effects suggests that the effects of imatinib are neuroprotective rather than neurorestorative. The positive effects on experimental spinal cord injury, obtained by oral delivery of a clinically used drug, makes imatinib an interesting candidate drug for clinical trials in spinal cord injury.
Collapse
Affiliation(s)
- Mathew B Abrams
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | | | | | |
Collapse
|
40
|
Anthony DC, Couch Y, Losey P, Evans MC. The systemic response to brain injury and disease. Brain Behav Immun 2012; 26:534-40. [PMID: 22085588 DOI: 10.1016/j.bbi.2011.10.011] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2011] [Revised: 10/27/2011] [Accepted: 10/30/2011] [Indexed: 10/15/2022] Open
Abstract
The idea that the brain is immunologically privileged and displays an atypical leukocyte recruitment profile following injury has influenced our ideas about how signals might be carried between brain and the periphery. For many, this has encouraged a cerebrocentric view of immunological responses to CNS injury, with little reference to the potential contribution from other organs. However, it is clear that bidirectional pathways between the brain and the peripheral immune system are important in the pathogenesis of CNS disease. In recent years, we have begun to understand the signals that are carried to the periphery and discovered new functions for known chemokines, made by the liver in response to brain injury, as important regulators of the CNS inflammatory response.
Collapse
Affiliation(s)
- Daniel C Anthony
- Experimental Neuropathology, Department of Pharmacology, Mansfield Road, University of Oxford, Oxford OX1 3QT, UK.
| | | | | | | |
Collapse
|
41
|
Bao F, Omana V, Brown A, Weaver LC. The systemic inflammatory response after spinal cord injury in the rat is decreased by α4β1 integrin blockade. J Neurotrauma 2012; 29:1626-37. [PMID: 22150233 DOI: 10.1089/neu.2011.2190] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Abstract
Abstract The systemic inflammatory response syndrome (SIRS) follows spinal cord injury (SCI) and causes damage to the lungs, kidney, and liver due to an influx of inflammatory cells from the circulation. After SCI in rats, the SIRS develops within 12 h and is sustained for at least 3 days. We have previously shown that blockade of CD11d/CD18 integrin reduces inflammation-driven secondary damage to the spinal cord. This treatment reduces the SIRS after SCI. In another study we found that blockade of α4β1 integrin limited secondary cord damage more effectively than blockade of CD11d/CD18. Therefore we considered it important to assess the effects of anti-α4β1 treatment on the SIRS in the lung, kidney, and liver after SCI. An anti-α4 antibody was given IV at 2 h after SCI at the fourth thoracic segment and the effects on the organs were evaluated at 24 h post-injury. The migration of neutrophils into the lungs and liver was markedly reduced and all three organs contained fewer macrophages. In the lungs and liver, the activation of the oxidative enzymes myeloperoxidase (MPO), inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), and gp91(phox), the production of free radicals, lipid peroxidation, and cell death were substantially and similarly reduced. Treatment effects were less robust in the kidney. Overall, the efficacy of the anti-α4β1 treatment did not differ greatly from that of the anti-CD11d antibody, although details of the results differed. The SIRS after SCI impedes recovery, and attenuation of the SIRS with an anti-integrin treatment is an important, clinically-relevant finding.
Collapse
Affiliation(s)
- Feng Bao
- Spinal Cord Injury Team, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | | | | | | |
Collapse
|
42
|
Perioperative glucose control in neurosurgical patients. Anesthesiol Res Pract 2012; 2012:690362. [PMID: 22400022 PMCID: PMC3286889 DOI: 10.1155/2012/690362] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2011] [Revised: 09/29/2011] [Accepted: 10/21/2011] [Indexed: 02/06/2023] Open
Abstract
Many neurosurgery patients may have unrecognized diabetes or may develop stress-related hyperglycemia in the perioperative period. Diabetes patients have a higher perioperative risk of complications and have longer hospital stays than individuals without diabetes. Maintenance of euglycemia using intensive insulin therapy (IIT) continues to be investigated as a therapeutic tool to decrease morbidity and mortality associated with derangements in glucose metabolism due to surgery. Suboptimal perioperative glucose control may contribute to increased morbidity, mortality, and aggravate concomitant illnesses. The challenge is to minimize the effects of metabolic derangements on surgical outcomes, reduce blood glucose excursions, and prevent hypoglycemia. Differences in cerebral versus systemic glucose metabolism, time course of cerebral response to injury, and heterogeneity of pathophysiology in the neurosurgical patient populations are important to consider in evaluating the risks and benefits of IIT. While extremes of glucose levels are to be avoided, there are little data to support an optimal blood glucose level or recommend a specific use of IIT for euglycemia maintenance in the perioperative management of neurosurgical patients. Individualized treatment should be based on the local level of blood glucose control, outpatient treatment regimen, presence of complications, nature of the surgical procedure, and type of anesthesia administered.
Collapse
|
43
|
Moreno B, Jukes JP, Vergara-Irigaray N, Errea O, Villoslada P, Perry VH, Newman TA. Systemic inflammation induces axon injury during brain inflammation. Ann Neurol 2012; 70:932-42. [PMID: 22190366 DOI: 10.1002/ana.22550] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Axon injury is a key contributor to the progression of disability in multiple sclerosis (MS). Systemic infections, which frequently precede relapses in MS, have been linked to clinical progression in Alzheimer's disease. There is evidence of a role for the innate immune system in MS lesions, as axonal injury is associated with macrophage activation. We hypothesize that systemic inflammation leads to enhanced axonal damage in MS as a consequence of innate immune system activation. METHODS Monophasic experimental allergic encephalomyelitis (EAE) was induced in a cohort of Lewis rats. The animals received a systemic challenge with either an inflammagen (lipopolysaccharide [LPS]) or saline as a control, at 1, 3, or 6 weeks into the remission phase of the disease. The clinical outcome, cellular recruitment to lesions, degree of tissue damage, and cytokine profiles were measured. RESULTS We found that systemic inflammation activates the central nervous system (CNS) innate immune response and results in a switch in the macrophage/microglia phenotype. This switch was accompanied by inducible nitric oxide synthase (iNOS) and interleukin-1β (IL-1β) expression and increased axon injury. This increased injury occurred independently of the re-emergence of overt clinical signs. INTERPRETATION Our evidence indicates that microglia/macrophages, associated with lesions, respond to circulating cytokines, produced in response to an inflammatory event outside the CNS, by producing immune mediators that lead to tissue damage. This has implications for people with MS, in which prevention and stringent management of systemic infectious diseases may slow disease progression.
Collapse
Affiliation(s)
- Beatriz Moreno
- CNS Inflammation Group, School of Biological Sciences, University of Southampton, Southampton, UK
| | | | | | | | | | | | | |
Collapse
|
44
|
Wang L, Tang X, Zhang H, Yuan J, Ding H, Wei Y. Elevated leptin expression in rat model of traumatic spinal cord injury and femoral fracture. J Spinal Cord Med 2011; 34:501-9. [PMID: 22118258 PMCID: PMC3184488 DOI: 10.1179/2045772311y.0000000034] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
BACKGROUND Few studies have reported a relationship between leptin induced by spinal cord injury (SCI) and healing bone tissue. OBJECTIVE To observe serum and callus leptin expression within the setting of fracture and traumatic SCI. METHODS Seventy-two male Sprague Dawley rats were randomized equally into four groups: control, SCI group, fracture group, and fracture/SCI group. Rats were sacrificed at 7, 14, 21, and 28 days post-fracture/SCI. Serum leptin was detected using radioimmunoassay at 1, 7, 14, 21, and 28 days, and callus formation was measured radiologically at 14, 21, and 28 days. Callus leptin was analyzed by means of immunohistochemistry. RESULTS Serum leptin in the fracture group, SCI group, and combined fracture/SCI group were all significantly increased compared to control group at the 1, 7, 14, and 2-day time points (P < 0.05). Serum leptin in the combined fracture/SCI group was significantly higher than in the fracture group at 7, 14, and 21 days (P < 0.05), and higher than in SCI groups at 14 and 21days after operation (P < 0.05). The percentage of leptin-positive cells in the fracture/SCI callus, and callus volume was significantly higher than in the fracture-only group (P < 0.001). CONCLUSIONS Overall, elevated leptin expression was demonstrated within healing bone especially in the 21 days of a rat model combining fracture and SCI. A close association exists between leptin levels and the degree of callus formation in fractures.
Collapse
Affiliation(s)
- Lei Wang
- Department of Orthopedics, The Affiliated People's Hospital with Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Xingguo Tang
- Department of Orthopedics, The Affiliated People's Hospital with Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Hongxi Zhang
- Department of Orthopedics, The Affiliated People's Hospital with Jiangsu University, Zhenjiang, Jiangsu Province, China,Correspondence to: Hongxi Zhang, Dianli Road 8, Zhenjiang 212001, Jiangsu Province, China.
| | - Jishan Yuan
- Department of Orthopedics, The Affiliated People's Hospital with Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Hua Ding
- Department of Orthopedics, The Affiliated People's Hospital with Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Yongzhong Wei
- Department of Orthopedics, The First Affiliated Hospital with Nanjing University, Jiangsu Province, China
| |
Collapse
|
45
|
CD11d integrin blockade reduces the systemic inflammatory response syndrome after spinal cord injury. Exp Neurol 2011; 231:272-83. [PMID: 21784069 DOI: 10.1016/j.expneurol.2011.07.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Revised: 05/26/2011] [Accepted: 07/07/2011] [Indexed: 11/20/2022]
Abstract
Traumatic injury to the spinal cord triggers a systemic inflammatory response syndrome (SIRS), in which inflammatory cells from the circulation invade organs such as the liver, lung and kidney, leading to damage of these organs. Our previous study (Gris, et al, Exp. Neurol, 2008) demonstrated that spinal cord injury (SCI) activates circulating neutrophils that then invade the lung and kidney from 2 to 24 h after injury, increasing myeloperoxidase activity, cyclooxygenase-2 and matrix metalloproteinase-9 expression and lipid peroxidation in these organs. The present study was designed to ascertain whether a treatment that limits the influx of leukocytes into the injured spinal cord would also be effective in reducing the SIRS after SCI. This treatment is intravenous delivery of a monoclonal antibody (mAb) against the CD11d subunit of the CD11d/CD18 integrin expressed by neutrophils and monocytes. We delivered the anti-CD11d mAb at 2 h post moderate clip compression SCI at the 4th or 12th thoracic segments and assessed inflammation, oxidative activity and cellular damage within the lung, kidney and liver at 12 h post-injury. In some analyses we compared high and low thoracic injuries to evaluate the importance of injury level on the intensity of the SIRS. After T4 injury, treatment with the anti-integrin mAb reduced the presence of neutrophils and macrophages in the lung, with associated decreases in expression of NF-κB and oxidative enzymes and in the concentration of free radicals in this organ. The treatment also reduced lipid peroxidation, protein nitration and cell death in the lung. The anti-CD11d treatment also reduced the inflammatory cells within the kidney after T4 injury, as well as the free radical concentration and amount of lipid peroxidation. In the liver, the mAb treatment reduced the influx of neutrophils but most of the other measures examined were unaffected by SCI. The inflammatory responses within the lung and kidney were often greater after T4 than T12 injury. Clinical studies show that SIRS, with its associated organ failure, contributes significantly to the morbidity and mortality of SCI patients. This anti-integrin treatment may block the onset of SIRS after SCI.
Collapse
|
46
|
Ferrari CC, Tarelli R. Parkinson's disease and systemic inflammation. PARKINSONS DISEASE 2011; 2011:436813. [PMID: 21403862 PMCID: PMC3049348 DOI: 10.4061/2011/436813] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 01/07/2011] [Indexed: 12/20/2022]
Abstract
Peripheral inflammation triggers exacerbation in the central brain's ongoing damage in several neurodegenerative diseases. Systemic inflammatory stimulus induce a general response known as sickness behaviour, indicating that a peripheral stimulus can induce the synthesis of cytokines in the brain. In Parkinson's disease (PD), inflammation was mainly associated with microglia activation that can underlie the neurodegeneration of neurons in the substantia nigra (SN). Peripheral inflammation can transform the “primed” microglia into an “active” state, which can trigger stronger responses dealing with neurodegenerative processes. Numerous evidences show that systemic inflammatory processes exacerbate ongoing neurodegeneration in PD patient and animal models. Anti-inflammatory treatment in PD patients exerts a neuroprotective effect. In the present paper, we analyse the effect of peripheral infections in the etiology and progression in PD patients and animal models, suggesting that these peripheral immune challenges can exacerbate the symptoms in the disease.
Collapse
Affiliation(s)
- Carina C Ferrari
- Laboratorio de Terapias Regenerativas y Protectoras del Sistema Nervioso, Fundación Instituto Leloir, Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
| | | |
Collapse
|
47
|
Wang HJ, Zakhari S, Jung MK. Alcohol, inflammation, and gut-liver-brain interactions in tissue damage and disease development. World J Gastroenterol 2010; 16:1304-13. [PMID: 20238396 PMCID: PMC2842521 DOI: 10.3748/wjg.v16.i11.1304] [Citation(s) in RCA: 179] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Chronic inflammation is often associated with alcohol-related medical conditions. The key inducer of such inflammation, and also the best understood, is gut microflora-derived lipopolysaccharide (LPS). Alcohol can significantly increase the translocation of LPS from the gut. In healthy individuals, the adverse effects of LPS are kept in check by the actions and interactions of multiple organs. The liver plays a central role in detoxifying LPS and producing a balanced cytokine milieu. The central nervous system contributes to anti-inflammatory regulation through neuroimmunoendocrine actions. Chronic alcohol use impairs not only gut and liver functions, but also multi-organ interactions, leading to persistent systemic inflammation and ultimately, to organ damage. The study of these interactions may provide potential new targets for therapeutic intervention.
Collapse
|
48
|
Wei Y, Wang L, Clark JCM, Dass CR, Choong PFM. Elevated leptin expression in a rat model of fracture and traumatic brain injury. J Pharm Pharmacol 2010. [DOI: 10.1211/jpp.60.12.0013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Abstract
A few studies have reported a relationship between leptin induced by brain injury and healing of bone tissue. Our objective was to measure serum and callus leptin expression within the setting of fracture and traumatic brain injury (TBI). Sixty-four male Sprague-Dawley rats were randomised equally into four groups: control, TBI group, fracture group and fracture/TBI group. Rats were sacrificed at 2, 4, 8 and 12 weeks after fracture/TBI. Serum leptin was detected using radio-immunoassay, and callus formation was measured radiologically. Callus leptin was analysed with immunohistochemistry. Serum leptin was significantly increased in the fracture, TBI and combined fracture/TBI groups compared with the control group at 2 weeks (P < 0.05). Serum leptin was significantly higher in the combined fracture/TBI group than in the fracture and TBI groups at 4 and 8 weeks (P < 0.05). The percentage of leptin-positive cells in the callus and callus volume were significantly higher in the fracture/TBI group than in the fracture-only group (P < 0.001). Thus, we demonstrated elevated leptin expression within healing bone, particularly in the first 8 weeks of a rat model combining fracture and TBI. A close association exists between leptin levels and the degree of callus formation in fractures.
Collapse
Affiliation(s)
- Yongzhong Wei
- Department of Orthopedics, The First Affiliated Hospital with Nanjing Medical University, China
| | - Lei Wang
- Department of Orthopedics, The First Affiliated Hospital with Nanjing Medical University, China
| | - Jonathan C M Clark
- Department of Orthopedics, St Vincent's Hospital Melbourne and Melbourne University, Australia
| | - Crispin R Dass
- Department of Orthopedics, St Vincent's Hospital Melbourne and Melbourne University, Australia
| | - Peter F M Choong
- Department of Orthopedics, St Vincent's Hospital Melbourne and Melbourne University, Australia
| |
Collapse
|
49
|
Cunningham O, Campion S, Perry VH, Murray C, Sidenius N, Docagne F, Cunningham C. Microglia and the urokinase plasminogen activator receptor/uPA system in innate brain inflammation. Glia 2010; 57:1802-14. [PMID: 19459212 PMCID: PMC2816357 DOI: 10.1002/glia.20892] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The urokinase plasminogen activator (uPA) receptor (uPAR) is a GPI-linked cell surface protein that facilitates focused plasmin proteolytic activity at the cell surface. uPAR has been detected in macrophages infiltrating the central nervous system (CNS) and soluble uPAR has been detected in the cerebrospinal fluid during a number of CNS pathologies. However, its expression by resident microglial cells in vivo remains uncertain. In this work, we aimed to elucidate the murine CNS expression of uPAR and uPA as well as that of tissue plasminogen activator and plasminogen activator inhibitor 1 (PAI-1) during insults generating distinct and well-characterized inflammatory responses; acute intracerebral lipopolysaccharide (LPS), acute kainate-induced neurodegeneration, and chronic neurodegeneration induced by prion disease inoculation. All three insults induced marked expression of uPAR at both mRNA and protein level compared to controls (naïve, saline, or control inoculum-injected). uPAR expression was microglial in all cases. Conversely, uPA transcription and activity was only markedly increased during chronic neurodegeneration. Dissociation of uPA and uPAR levels in acute challenges is suggestive of additional proteolysis-independent roles for uPAR. PAI-1 was most highly expressed upon LPS challenge, whereas tissue plasminogen activator mRNA was constitutively present and less responsive to all insults studied. These data are novel and suggest much wider involvement of the uPAR/uPA system in CNS function and pathology than previously supposed.
Collapse
|
50
|
Bao F, Bailey CS, Gurr KR, Bailey SI, Rosas-Arellano MP, Dekaban GA, Weaver LC. Increased oxidative activity in human blood neutrophils and monocytes after spinal cord injury. Exp Neurol 2008; 215:308-16. [PMID: 19056384 DOI: 10.1016/j.expneurol.2008.10.022] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2008] [Revised: 10/03/2008] [Accepted: 10/26/2008] [Indexed: 10/21/2022]
Abstract
Traumatic injury can cause a systemic inflammatory response, increasing oxidative activity of circulating leukocytes and potentially exacerbating the original injury, as well as causing damage to initially unaffected organs. Although the importance of intraspinal inflammation after human spinal cord injury is appreciated, the role of the systemic inflammatory response to this injury is not widely recognised. We investigated oxidative activity of blood leukocytes from nine cord-injured subjects and six trauma controls (bone fractures without CNS injury) at 6 h-2 weeks after injury, comparing values to those of ten uninjured subjects. Neutrophil and monocyte free radical production, evaluated by flow cytometry, increased significantly more in cord injury subjects than in trauma controls (6-fold vs 50% increases). In leukocyte homogenates, the concentration of free radicals increased significantly more in cord injury subjects (2-fold) than in the trauma controls (1.6-fold) as did activity of myeloperoxidase (2.3-fold vs. 1.7-fold). Moreover, in homogenates and blood smears, expression of the NADPH oxidase subunit gp91(phox) and of the oxidative enzyme, inducible nitric oxide synthetase was 20-25% greater in cord injury subjects than in trauma controls. Expression of the pro-inflammatory transcription factor NF-kappaB and of cyclooxygenase-2 increased similarly after both injuries. Finally, aldehyde products of tissue-damaging lipid peroxidation also increased significantly more in the plasma of spinal cord injury subjects than in trauma controls (2.6 fold vs. 1.9-fold). Spinal cord injury causes a particularly intense systemic inflammatory response. Limiting this response briefly after cord injury should protect the spinal cord and tissues/organs outside the CNS from secondary damage.
Collapse
Affiliation(s)
- Feng Bao
- Spinal Cord Injury Team, BioTherapeutics Research Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, 100 Perth Drive, London, ON, Canada N6A 5K8
| | | | | | | | | | | | | |
Collapse
|