1
|
Seplovich G, Bouchi Y, de Rivero Vaccari JP, Pareja JCM, Reisner A, Blackwell L, Mechref Y, Wang KK, Tyndall JA, Tharakan B, Kobeissy F. Inflammasome links traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease. Neural Regen Res 2025; 20:1644-1664. [PMID: 39104096 PMCID: PMC11688549 DOI: 10.4103/nrr.nrr-d-24-00107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/20/2024] [Accepted: 06/03/2024] [Indexed: 08/07/2024] Open
Abstract
Traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease are three distinct neurological disorders that share common pathophysiological mechanisms involving neuroinflammation. One sequela of neuroinflammation includes the pathologic hyperphosphorylation of tau protein, an endogenous microtubule-associated protein that protects the integrity of neuronal cytoskeletons. Tau hyperphosphorylation results in protein misfolding and subsequent accumulation of tau tangles forming neurotoxic aggregates. These misfolded proteins are characteristic of traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease and can lead to downstream neuroinflammatory processes, including assembly and activation of the inflammasome complex. Inflammasomes refer to a family of multimeric protein units that, upon activation, release a cascade of signaling molecules resulting in caspase-induced cell death and inflammation mediated by the release of interleukin-1β cytokine. One specific inflammasome, the NOD-like receptor protein 3, has been proposed to be a key regulator of tau phosphorylation where it has been shown that prolonged NOD-like receptor protein 3 activation acts as a causal factor in pathological tau accumulation and spreading. This review begins by describing the epidemiology and pathophysiology of traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease. Next, we highlight neuroinflammation as an overriding theme and discuss the role of the NOD-like receptor protein 3 inflammasome in the formation of tau deposits and how such tauopathic entities spread throughout the brain. We then propose a novel framework linking traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease as inflammasome-dependent pathologies that exist along a temporal continuum. Finally, we discuss potential therapeutic targets that may intercept this pathway and ultimately minimize long-term neurological decline.
Collapse
Affiliation(s)
| | - Yazan Bouchi
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jennifer C. Munoz Pareja
- Division of Pediatric Critical Care, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Andrew Reisner
- Department of Pediatrics, Emory University, Atlanta, GA, USA
- Department of Neurosurgery, Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Laura Blackwell
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, USA
| | - Kevin K. Wang
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | | | - Binu Tharakan
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA
| | - Firas Kobeissy
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| |
Collapse
|
2
|
Miao HT, Wang J, Shao JJ, Song RX, Li WG, Sun JK, Jia SY, Zhang DX, Li XM, Zhao JY, Zhang LM. Astrocytic NLRP3 cKO mitigates depression-like behaviors induced by mild TBI in mice. Neurobiol Dis 2025; 205:106785. [PMID: 39793767 DOI: 10.1016/j.nbd.2024.106785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/31/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Reports indicate that depression is a common mental health issue following traumatic brain injury (TBI). Our prior research suggests that Nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3)-related neuroinflammation, modulated by glial cells such as astrocytes, is likely to play a crucial role in the progression of anxiety and cognitive dysfunction. However, there is limited understanding of the potential of astrocytic NLRP3 in treating depression under mild TBI condition. This study aimed to determine whether astrocytic NLRP3 knockout (KO) could mitigate depressive-like behaviors following mild TBI and explore potential variations in such behaviors between genders post-mild TBI. METHODS Mild TBI was induced in mice using Feeney's weight-drop method. Behavioral assessments included neurological severity scores (NSS), social interaction test (SI), tail suspension test (TST), and forced swimming test (FST). Pathological changes were evaluated through immunofluorescence and local field potential (LFP) recordings at various time points post-injury. RESULTS Our findings indicated that astrocyte-specific NLRP3 KO decreased cleaved caspase-1 colocalized with astrocytes, decreased pathogenic astrocytes and increased Postsynaptic density protein 95 (PSD95) intensity, and significantly alleviated mild TBI-induced depression-like behaviors. It also led to the upregulation of protective astrocytes and apoptosis-associated factors, including cleaved caspase-3 post-mild TBI. Additionally, astrocyte-specific NLRP3 deletion resulting in improved θ and γ power and θ-γ phase coupling in the social interaction test (SI). Notably, under mild TBI conditions, astrocyte-specific NLRP3 exhibited greater neuroprotective effects in female knockout mice compared to males. CONCLUSION Astrocyte NLRP3 knockout demonstrated a protective mechanism in mice subjected to mild TBI, possibly attributed to the inhibition of pyroptosis through the NLRP3 signaling pathway in astrocytes.
Collapse
Affiliation(s)
- Hui-Tao Miao
- Department of Anesthesiology, Hebei Province, Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China,; Hebei Province Key Laboratory of Integrated Traditional and Western Medicine in Neurological Rehabilitation, Cangzhou, China; Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research (Preparing), Cangzhou, China
| | - Jun Wang
- Department of Orthopaedics, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Jing-Jing Shao
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rong-Xin Song
- Department of Anesthesiology, Hebei Province, Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China
| | - Wen-Guang Li
- Graduated School, Hebei Medical University, Shijiazhuang, China
| | - Jian-Kai Sun
- Graduated School, Hebei Medical University, Shijiazhuang, China
| | - Shi-Yan Jia
- Hebei Province Key Laboratory of Integrated Traditional and Western Medicine in Neurological Rehabilitation, Cangzhou, China
| | - Dong-Xue Zhang
- Department of Gerontology, Cangzhou Central Hospital, Cangzhou, China
| | - Xiao-Ming Li
- Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research (Preparing), Cangzhou, China
| | - Jian-Yong Zhao
- Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research (Preparing), Cangzhou, China
| | - Li-Min Zhang
- Department of Anesthesiology, Hebei Province, Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China,.
| |
Collapse
|
3
|
Hong Z, Zuo Z, Zhao Y, Ai Y, Zhang L, Li L, He X, Luo J, Xu J, Yang X, Yi S, Zheng H, Tie C, Niu L, Hu X. Transcranial focused ultrasound stimulation alleviates NLRP3-related neuroinflammation induced by ischemic stroke via regulation of the Nespas/miR-383-3p/SHP2 pathway. Int Immunopharmacol 2025; 144:113680. [PMID: 39616858 DOI: 10.1016/j.intimp.2024.113680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/06/2024] [Accepted: 11/18/2024] [Indexed: 12/15/2024]
Abstract
Transcranial focused ultrasound stimulation (tFUS) has emerged as a promising therapeutic strategy for mitigating brain injury in animal models. In this study, the effects and mechanisms of tFUS on ischemic stroke were explored in a transient middle cerebral artery occlusion (MCAO) rat model. Low-intensity tFUS was administered to the ischemic hemisphere 24 h post-MCAO for seven consecutive days. Neurological function was evaluated through neurobehavioral assessments following tFUS treatment. Western blotting, immunofluorescence staining, and quantitative real-time PCR were performed to examine the impact of tFUS on NLRP3-related neuroinflammation using brain tissues from MCAO rats and BV2 cells subjected to oxygen glucose deprivation/reperfusion (OGD/R). Additionally, RNA sequencing and cell transient transfection were employed to elucidate the underlying mechanisms. The findings revealed that tFUS improved neurobehavioral performance, reduced infarct size, and suppressed NLRP3 inflammasome activation seven days post-MCAO. Notably, Nespas expression was significantly elevated in tFUS-treated rats, whereas Nespas silencing exacerbated neurological deficits and enhanced NLRP3 activation. Moreover, Nespas positively regulated src homology 2 domain-containing tyrosine phosphatase-2 (SHP2), and SHP2 inhibition significantly amplified NLRP3 activation. Mechanistic in vitro studies further demonstrated that Nespas attenuated microglial NLRP3 activation via the Nespas/miR-383-3p/SHP2 pathway. These results suggest that the neuroprotective effects of tFUS are likely mediated through the upregulation of Nespas and suppression of NLRP3 via the Nespas/miR-383-3p/SHP2 axis, offering new insights into the molecular mechanisms supporting tFUS as a potential therapeutic approach for stroke-induced brain injury.
Collapse
Affiliation(s)
- Zhongqiu Hong
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Zejie Zuo
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yun Zhao
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yinan Ai
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liying Zhang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lili Li
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaofei He
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jing Luo
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jinghui Xu
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaofeng Yang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shasha Yi
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Haiqing Zheng
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Changjun Tie
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Lili Niu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China.
| | - Xiquan Hu
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
4
|
Zhang P, Liu Y, Jin X, Hu Z, Yang J, Lu H, Hang T, Song M. Alzheimer's disease-like pathology induced by Porphyromonas gingivalis in middle-aged mice is mediated by NLRP3 inflammasome via the microbiota-gut-brain axis. J Alzheimers Dis 2025; 103:487-505. [PMID: 39639573 DOI: 10.1177/13872877241302498] [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: 12/07/2024]
Abstract
BACKGROUND Porphyromonas gingivalis (P. gingivalis) has been found to enter the brain and induce inflammation, contributing to Alzheimer's disease (AD). P. gingivalis is also closely linked to gut dysbiosis. However, does P. gingivalis induce AD-like pathology through the microbiota-gut-brain axis? There is limited literature on this topic. OBJECTIVE To determine the precise causal link among P. gingivalis, intestinal inflammation, and AD-related pathology. METHODS 12- to 13-month-old female C57BL/6J mice were subjected to ligature placement and oral administration of P. gingivalis over a 24-week period. Then, cognitive performance was evaluated with behavioral tests, while AD neuropathological changes, neuroinflammation, and intestinal inflammation were assessed through qPCR, immunofluorescence, and western blot, and gut microbiota was analyzed by 16S rRNA. RESULTS Mice exposed to P. gingivalis showed impaired behavior in open field test, novel object recognition, and Y-maze tests. The bacterium infiltrated their brains, increasing Aβ42, AβPP, and Aβ fragments, promoting tau phosphorylation and microglial activation, and reducing levels of ZO-1, PSD95, SYP, and NeuN proteins. Inflammatory factors like NLRP3, caspase-1, IL-1β, IL-6, and TNF-α were elevated in both brains and intestine, while ZO-1 and occludin levels decreased in intestine. P. gingivalis also altered gut microbial compositions. CONCLUSIONS P. gingivalis induced gut dysbiosis and activated the NLRP3 inflammasome in the intestine and brains of mice. This led to impairment of both the intestinal and brain-blood barriers, triggering neuroinflammation and promoting the progression of AD. These findings highlight the critical role of NLRP3 inflammasome activation in the microbiota-gut-brain axis in the AD-like pathology induced by P. gingivalis.
Collapse
Affiliation(s)
- Pei Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing, China
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Yan Liu
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing, China
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Xin Jin
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing, China
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Zhaoliang Hu
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing, China
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Jucui Yang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing, China
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Haotian Lu
- International Department of High School, AFF to Nanjing Normal University Jiangning Campus, Nanjing, China
| | - Taijun Hang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing, China
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Min Song
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing, China
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
5
|
Hong C, Wang L, Zhou X, Zou L, Xiang X, Deng H, Li Q, Wu Y, Liu L, Li T. Protective Effects of Mdivi-1 on Cognition Disturbance Following Sepsis in Mice via Alleviating Microglia Activation and Polarization. CNS Neurosci Ther 2025; 31:e70149. [PMID: 39791542 PMCID: PMC11719124 DOI: 10.1111/cns.70149] [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: 05/20/2024] [Revised: 10/09/2024] [Accepted: 10/20/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Neuroinflammation is one of the essential pathogeneses of cognitive damage suffering from sepsis-associated encephalopathy (SAE). Lots of evidences showed the microglia presented mitochondrial fragmentation during SAE. This study investigated the protective effects and novel mechanisms of inhibiting microglia mitochondrial fragmentation via mitochondrial division inhibitor 1 (Mdivi-1) on cognitive damage in SAE. METHODS The SAE model was performed by cecal ligation and puncture (CLP), and Mdivi-1 was administrated via intraperitoneal injection. Morris water maze was performed to assess cognitive function. Mitochondrial morphology was observed by electron microscope or MitoTracker staining. The qRT-PCR, immunofluorescence staining, and western blots were used to detect the inflammatory factors and protein content, respectively. Flow cytometry was used to detect the polarization of hippocampal microglia. Bioinformatics analysis was used to verify hypotheses. RESULTS Mdivi-1 administration alleviated sepsis-induced mitochondrial fragmentation, microglia activation, polarization, and cognitive damage. The mechanisms study showed neuroinflammation and oxidative stress were suppressed via NF-κB and Keap1/Nrf2/HO-1 pathways following Mdivi-1 administration; meanwhile, pyroptosis in microglia was reduced, which was associated with enhanced autophagosome formation via p62 elevation following Mdivi-1 administration. CONCLUSION Inhibition of microglia mitochondrial fragmentation is beneficial to SAE cognitive disturbance, the mechanisms are related to alleviating neuroinflammation, oxidative stress, and pyroptosis.
Collapse
Affiliation(s)
- Chen Hong
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Li Wang
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Xiaowei Zhou
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Liyong Zou
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Xinming Xiang
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Haoyue Deng
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Qinghui Li
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Yue Wu
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Liangming Liu
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Tao Li
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| |
Collapse
|
6
|
Calderone A, Latella D, Cardile D, Gangemi A, Corallo F, Rifici C, Quartarone A, Calabrò RS. The Role of Neuroinflammation in Shaping Neuroplasticity and Recovery Outcomes Following Traumatic Brain Injury: A Systematic Review. Int J Mol Sci 2024; 25:11708. [PMID: 39519259 PMCID: PMC11546226 DOI: 10.3390/ijms252111708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Neuroplasticity and neuroinflammation are variables seen during recovery from traumatic brain injury (TBI), while biomarkers are useful in monitoring injury and guiding rehabilitation efforts. This systematic review examines how neuroinflammation affects neuroplasticity and recovery following TBI in animal models and humans. Studies were identified from an online search of the PubMed, Web of Science, and Embase databases without any search time range. This review has been registered on Open OSF (n) UDWQM. Recent studies highlight the critical role of biomarkers like serum amyloid A1 (SAA1) and Toll-like receptor 4 (TLR4) in predicting TBI patients' injury severity and recovery outcomes, offering the potential for personalized treatment and improved neurorehabilitation strategies. Additionally, insights from animal studies reveal how neuroinflammation affects recovery, emphasizing targets such as NOD-like receptor family pyrin domain-containing 3 (NLRP3) and microglia for enhancing therapeutic interventions. This review emphasizes the central role of neuroinflammation in TBI, and its adverse impact on neuroplasticity and recovery, and suggests that targeted anti-inflammatory treatments and biomarker-based personalized approaches hold the key to improvement. Such approaches will need further development in future research by integrating neuromodulation and pharmacological interventions, along with biomarker validation, to optimize management in TBI.
Collapse
Affiliation(s)
- Andrea Calderone
- Department of Clinical and Experimental Medicine, University of Messina, Piazza Pugliatti 1, 98122 Messina, Italy
| | - Desirèe Latella
- IRCCS Centro Neurolesi Bonino-Pulejo, S.S. 113 Via Palermo, C.da Casazza, 98124 Messina, Italy
| | - Davide Cardile
- IRCCS Centro Neurolesi Bonino-Pulejo, S.S. 113 Via Palermo, C.da Casazza, 98124 Messina, Italy
| | - Antonio Gangemi
- IRCCS Centro Neurolesi Bonino-Pulejo, S.S. 113 Via Palermo, C.da Casazza, 98124 Messina, Italy
| | - Francesco Corallo
- IRCCS Centro Neurolesi Bonino-Pulejo, S.S. 113 Via Palermo, C.da Casazza, 98124 Messina, Italy
| | - Carmela Rifici
- IRCCS Centro Neurolesi Bonino-Pulejo, S.S. 113 Via Palermo, C.da Casazza, 98124 Messina, Italy
| | - Angelo Quartarone
- IRCCS Centro Neurolesi Bonino-Pulejo, S.S. 113 Via Palermo, C.da Casazza, 98124 Messina, Italy
| | - Rocco Salvatore Calabrò
- IRCCS Centro Neurolesi Bonino-Pulejo, S.S. 113 Via Palermo, C.da Casazza, 98124 Messina, Italy
| |
Collapse
|
7
|
Taylor RR, Keane RW, Guardiola B, López-Lage S, Moratinos L, Dietrich WD, Perez-Barcena J, de Rivero Vaccari JP. Inflammasome Proteins Are Reliable Biomarkers of the Inflammatory Response in Aneurysmal Subarachnoid Hemorrhage. Cells 2024; 13:1370. [PMID: 39195261 PMCID: PMC11353247 DOI: 10.3390/cells13161370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/02/2024] [Accepted: 08/15/2024] [Indexed: 08/29/2024] Open
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH) is caused by abnormal blood vessel dilation and subsequent rupture, resulting in blood pooling in the subarachnoid space. This neurological insult results in the activation of the inflammasome, a multiprotein complex that processes pro-inflammatory interleukin (IL)-1 cytokines leading to morbidity and mortality. Moreover, increases in inflammasome proteins are associated with clinical deterioration in many neurological diseases. Limited studies have investigated inflammasome protein expression following aSAH. Reliable markers of the inflammatory response associated with aSAH may allow for earlier detection of patients at risk for complications and aid in the identification of novel pharmacologic targets. Here, we investigated whether inflammasome signaling proteins may serve as potential biomarkers of the inflammatory response in aSAH. Serum and cerebrospinal fluid (CSF) from fifteen aSAH subjects and healthy age-matched controls and hydrocephalus (CSF) no-aneurysm controls were evaluated for levels of inflammasome signaling proteins and downstream pro-inflammatory cytokines. Protein measurements were carried out using Simple Plex and Single-Molecule Array (Simoa) technology. The area under the curve (AUC) was calculated using receiver operating characteristics (ROCs) to obtain information on biomarker reliability, specificity, sensitivity, cut-off points, and likelihood ratio. In addition, a Spearman r correlation matrix was performed to determine the correlation between inflammasome protein levels and clinical outcome measures. aSAH subjects demonstrated elevated caspase-1, apoptosis-associated speck-like protein with a caspase recruiting domain (ASC), IL-18 and IL-1β levels in serum, and CSF when compared to controls. Each of these proteins was found to be a promising biomarker of inflammation in aSAH in the CSF. In addition, ASC, caspase-1, and IL-1β were found to be promising biomarkers of inflammation in aSAH in serum. Furthermore, we found that elevated levels of inflammasome proteins in serum are useful to predict worse functional outcomes following aSAH. Thus, the determination of inflammasome protein levels in CSF and serum in aSAH may be utilized as reliable biomarkers of inflammation in aSAH and used clinically to monitor patient outcomes.
Collapse
Affiliation(s)
- Ruby R. Taylor
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (R.R.T.); (R.W.K.); (W.D.D.)
- Medical Scientist Training Program, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Robert W. Keane
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (R.R.T.); (R.W.K.); (W.D.D.)
- Department of Cellular Physiology and Molecular Biophysics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Begoña Guardiola
- Intensive Care Department, Son Espases University Hospital, 07120 Palma de Mallorca, Spain; (B.G.); (J.P.-B.)
| | - Sofía López-Lage
- Neurosurgical Department, Son Espases University Hospital, 07120 Palma de Mallorca, Spain (L.M.)
| | - Lesmes Moratinos
- Neurosurgical Department, Son Espases University Hospital, 07120 Palma de Mallorca, Spain (L.M.)
| | - W. Dalton Dietrich
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (R.R.T.); (R.W.K.); (W.D.D.)
| | - Jon Perez-Barcena
- Intensive Care Department, Son Espases University Hospital, 07120 Palma de Mallorca, Spain; (B.G.); (J.P.-B.)
| | - Juan Pablo de Rivero Vaccari
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (R.R.T.); (R.W.K.); (W.D.D.)
- Department of Cellular Physiology and Molecular Biophysics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
8
|
Şahin S, Şahin E, Esenülkü G, Renda G, Gürgen SG, Alver A, Abidin İ, Cansu A. Oleuropein Has Modulatory Effects on Systemic Lipopolysaccharide-Induced Neuroinflammation in Male Rats. J Nutr 2024; 154:1282-1297. [PMID: 38403251 DOI: 10.1016/j.tjnut.2024.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/20/2024] [Accepted: 02/15/2024] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND Neuroinflammation induced by systemic inflammation is a risk factor for developing chronic neurologic disorders. Oleuropein (OLE) has antioxidant and anti-inflammatory properties; however, its effect on systemic inflammation-related neuroinflammation is unknown. OBJECTIVES This study aimed to determine whether OLE protects against systemic lipopolysaccharide (LPS)-induced neuroinflammation in rats. METHODS Six-wk-old Wistar rats were randomly assigned to 1 of the following 5 groups: 1) control, 2) OLE-only, 3) LPS + vehicle, 4) OLE+LPS (O-LPS), and 5) a single-dose OLE + LPS (SO-LPS group). OLE 200 mg/kg or saline as a vehicle was administered via gavage for 7 d. On the seventh day, 2.5 mg/kg LPS was intraperitoneally administered. The rats were decapitated after 24 h of LPS treatment, and serum collection and tissue dissection were performed. The study assessed astrocyte and microglial activation using glial fibrillary acidic protein (GFAP) and CD11b immunohistochemistry, nod-like receptor protein-3, interleukin (IL)-1β, IL-17A, and IL-4 concentrations in prefrontal and hippocampal tissues via enzyme-linked immunosorbent assay, and total antioxidant/oxidant status (TAS/TOS) in serum and tissues via spectrophotometry. RESULTS In both the O-LPS and SO-LPS groups, LPS-related activation of microglia and astrocytes was suppressed in the cortex and hippocampus (P < 0.001), excluding cortical astrocyte activation, which was suppressed only in the SO-LPS group (P < 0.001). Hippocampal GFAP immunoreactivity and IL-17A concentrations in the dentate gyrus were higher in the OLE group than those in the control group, but LPS-related increases in these concentrations were suppressed in the O-LPS group. The O-LPS group had higher cortical TAS and IL-4 concentrations. CONCLUSIONS OLE suppressed LPS-related astrocyte and microglial activation in the hippocampus and cortex. The OLE-induced increase in cortical IL-4 concentrations indicates the induction of an anti-inflammatory phenotype of microglia. OLE may also modulate astrocyte and IL-17A functions, which could explain its opposing effects on hippocampal GFAP immunoreactivity and IL-17A concentrations when administered with or without LPS.
Collapse
Affiliation(s)
- Sevim Şahin
- Department of Pediatric Neurology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey.
| | - Elif Şahin
- Department of Medical Biochemistry, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Gülnur Esenülkü
- Department of Pediatric Neurology, Trabzon Kanuni Training, and Research Hospital, Trabzon, Turkey
| | - Gülin Renda
- Department of Pharmacognosy, Faculty of Pharmacy, Karadeniz Technical University, Trabzon, Turkey
| | - Seren Gülşen Gürgen
- Department of Histology and Embryology, School of Vocational Health Service, Manisa Celal Bayar University, Manisa, Turkey
| | - Ahmet Alver
- Department of Medical Biochemistry, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - İsmail Abidin
- Department of Biophysics, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Ali Cansu
- Department of Pediatric Neurology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| |
Collapse
|
9
|
He L, Zhang R, Yang M, Lu M. The role of astrocyte in neuroinflammation in traumatic brain injury. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166992. [PMID: 38128844 DOI: 10.1016/j.bbadis.2023.166992] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/30/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023]
Abstract
Traumatic brain injury (TBI), a significant contributor to mortality and morbidity worldwide, is a devastating condition characterized by initial mechanical damage followed by subsequent biochemical processes, including neuroinflammation. Astrocytes, the predominant glial cells in the central nervous system, play a vital role in maintaining brain homeostasis and supporting neuronal function. Nevertheless, in response to TBI, astrocytes undergo substantial phenotypic alternations and actively contribute to the neuroinflammatory response. This article explores the multifaceted involvement of astrocytes in neuroinflammation subsequent to TBI, with a particular emphasis on their activation, release of inflammatory mediators, modulation of the blood-brain barrier, and interactions with other immune cells. A comprehensive understanding the dynamic interplay between astrocytes and neuroinflammation in the condition of TBI can provide valuable insights into the development of innovative therapeutic approaches aimed at mitigating secondary damage and fostering neuroregeneration.
Collapse
Affiliation(s)
- Liang He
- Department of Anesthesiology, Yan'an Hospital of Kunming City, Kunming 650051, China.
| | - Ruqiang Zhang
- Department of Anesthesiology, Yan'an Hospital of Kunming City, Kunming 650051, China
| | - Maiqiao Yang
- Department of Anesthesiology, Yan'an Hospital of Kunming City, Kunming 650051, China
| | - Meilin Lu
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China.
| |
Collapse
|
10
|
Ravichandran KA, Heneka MT. Inflammasomes in neurological disorders - mechanisms and therapeutic potential. Nat Rev Neurol 2024; 20:67-83. [PMID: 38195712 DOI: 10.1038/s41582-023-00915-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2023] [Indexed: 01/11/2024]
Abstract
Inflammasomes are molecular scaffolds that are activated by damage-associated and pathogen-associated molecular patterns and form a key element of innate immune responses. Consequently, the involvement of inflammasomes in several diseases that are characterized by inflammatory processes, such as multiple sclerosis, is widely appreciated. However, many other neurological conditions, including Alzheimer disease, Parkinson disease, amyotrophic lateral sclerosis, stroke, epilepsy, traumatic brain injury, sepsis-associated encephalopathy and neurological sequelae of COVID-19, all involve persistent inflammation in the brain, and increasing evidence suggests that inflammasome activation contributes to disease progression in these conditions. Understanding the biology and mechanisms of inflammasome activation is, therefore, crucial for the development of inflammasome-targeted therapies for neurological conditions. In this Review, we present the current evidence for and understanding of inflammasome activation in neurological diseases and discuss current and potential interventional strategies that target inflammasome activation to mitigate its pathological consequences.
Collapse
Affiliation(s)
- Kishore Aravind Ravichandran
- Department of Neuroinflammation, Institute of innate immunity, University of Bonn Medical Center Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Esch-sur-Alzette, Luxembourg.
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, North Worcester, MA, USA.
| |
Collapse
|
11
|
Zhao K, Zhou X, Chen M, Gou L, Mei D, Gao C, Zhao S, Luo S, Wang X, Tan T, Zhang Y. Neuroprotective Effects of CXCR2 Antagonist SB332235 on Traumatic Brain Injury Through Suppressing NLRP3 Inflammasome. Neurochem Res 2024; 49:184-198. [PMID: 37702890 PMCID: PMC10776743 DOI: 10.1007/s11064-023-04021-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/19/2023] [Accepted: 08/24/2023] [Indexed: 09/14/2023]
Abstract
The inflammatory process mediated by nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain comprising 3 (NLRP3) inflammasome plays a predominant role in the neurological dysfunction following traumatic brain injury (TBI). SB332235, a highly selective antagonist of chemokine receptor 2 (CXCR2), has been demonstrated to exhibit anti-inflammatory properties and improve neurological outcomes in the central nervous system. We aimed to determine the neuroprotective effects of SB332235 in the acute phase after TBI in mice and to elucidate its underlying mechanisms. Male C57BL/6J animals were exposed to a controlled cortical impact, then received 4 doses of SB332235, with the first dose administered at 30 min after TBI, followed by additional doses at 6, 24, and 30 h. Neurological defects were assessed by the modified neurological severity score, while the motor function was evaluated using the beam balance and open field tests. Cognitive performance was evaluated using the novel object recognition test. Brain tissues were collected for pathological, Western blot, and immunohistochemical analyses. The results showed that SB332235 significantly ameliorated TBI-induced deficits, including motor and cognitive impairments. SB332235 administration suppressed expression of both CXCL1 and CXCR2 in TBI. Moreover, SB332235 substantially mitigated the augmented expression levels and activation of the NLRP3 inflammasome within the peri-contusional cortex induced by TBI. This was accompanied by the blocking of subsequent production of pro-inflammatory cytokines. Additionally, SB332235 hindered microglial activity induced by TBI. These findings confirmed the neuroprotective effects of SB332235 against TBI, and the involved mechanisms were in part due to the suppression of NLRP3 inflammasome activity. This study suggests that SB332235 may act as an anti-inflammatory agent to improve functional outcomes in brain injury when applied clinically.
Collapse
Affiliation(s)
- Ke Zhao
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Henan Children's Neurodevelopment Engineering Research Center, Zhengzhou, China
| | - Xinkui Zhou
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Henan Children's Neurodevelopment Engineering Research Center, Zhengzhou, China
| | - Mengyuan Chen
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Henan Children's Neurodevelopment Engineering Research Center, Zhengzhou, China
| | - Lingshan Gou
- Center for Genetic Medicine, Xuzhou Maternity and Child Health Care Hospital Affiliated to Xuzhou Medical University, Xuzhou, China
| | - Daoqi Mei
- Department of Neurology, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Chao Gao
- Department of Rehabilitation, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Shuai Zhao
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Henan Children's Neurodevelopment Engineering Research Center, Zhengzhou, China
| | - Shuying Luo
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Henan Children's Neurodevelopment Engineering Research Center, Zhengzhou, China
| | - Xiaona Wang
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Henan Children's Neurodevelopment Engineering Research Center, Zhengzhou, China.
| | - Tao Tan
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Yaodong Zhang
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Henan Children's Neurodevelopment Engineering Research Center, Zhengzhou, China.
| |
Collapse
|
12
|
Sergi CM. MASLD and aspartame: are new studies in the horizon? Front Med (Lausanne) 2023; 10:1266918. [PMID: 38143439 PMCID: PMC10739386 DOI: 10.3389/fmed.2023.1266918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/17/2023] [Indexed: 12/26/2023] Open
Abstract
Fatty liver disease has been on the rise in the past few decades, and there is no hope that it will stop. The terminology change that has been recently proposed may not be sufficient to advocate for a reduction of steatogenic foods and a change in lifestyle. A course change may be supported by the recent labeling of aspartame sweetener as a possible carcinogenic compound by the International Association for Research on Cancer (IARC), an agency of the World Health Organization (WHO). Aspartame sweeteners and other edulcorating molecular compounds besides colorings may trigger liver cancer other than fatty liver disease, despite limited data supporting it. An essential bias in human cohort studies is indeed the exclusion of all confounding factors, which may be barely impossible for human studies. In this perspective, we suggest that the activation of the NOD-like receptor-enclosing protein 3 (NLRP3) inflammasome and the stimulation of the tumor suppression gene TP53 may be critical in the progression from fatty liver to liver inflammation and liver cancer. Aspartame reduces a transcriptional coactivator, precisely the peroxisomal proliferator-initiated receptor-γ (gamma) coactivator 1-α (alpha) (or PGC1α). This coactivator upregulates mitochondrial bioformation, oxidative phosphorylation, respiratory capacity, and fatty acid β-oxidation. Aspartame acts in this way, probably through the activation of TP53. These events have been accountable for the variations in the lipid outline in serum and total lipid storage as well as for the impairment of gluconeogenesis in the liver, as supported by the downregulation of the gluconeogenic enzymes in experimental animals, and may be relevant in humans as well.
Collapse
Affiliation(s)
- Consolato M. Sergi
- Department of Laboratory Medicine, University of Alberta, Edmonton, AB, Canada
- Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
13
|
Ye J, Hu X, Wang Z, Li R, Gan L, Zhang M, Wang T. The role of mtDAMPs in the trauma-induced systemic inflammatory response syndrome. Front Immunol 2023; 14:1164187. [PMID: 37533869 PMCID: PMC10391641 DOI: 10.3389/fimmu.2023.1164187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 06/26/2023] [Indexed: 08/04/2023] Open
Abstract
Systemic inflammatory response syndrome (SIRS) is a non-specific exaggerated defense response caused by infectious or non-infectious stressors such as trauma, burn, surgery, ischemia and reperfusion, and malignancy, which can eventually lead to an uncontrolled inflammatory response. In addition to the early mortality due to the "first hits" after trauma, the trauma-induced SIRS and multiple organ dysfunction syndrome (MODS) are the main reasons for the poor prognosis of trauma patients as "second hits". Unlike infection-induced SIRS caused by pathogen-associated molecular patterns (PAMPs), trauma-induced SIRS is mainly mediated by damage-associated molecular patterns (DAMPs) including mitochondrial DAMPs (mtDAMPs). MtDAMPs released after trauma-induced mitochondrial injury, including mitochondrial DNA (mtDNA) and mitochondrial formyl peptides (mtFPs), can activate inflammatory response through multiple inflammatory signaling pathways. This review summarizes the role and mechanism of mtDAMPs in the occurrence and development of trauma-induced SIRS.
Collapse
Affiliation(s)
- Jingjing Ye
- Trauma Center, Peking University People’s Hospital, Key Laboratory of Trauma Treatment and Neural Regeneration (Peking University) Ministry of Education, National Center for Trauma Medicine of China, Beijing, China
| | - Xiaodan Hu
- Trauma Center, Peking University People’s Hospital, Key Laboratory of Trauma Treatment and Neural Regeneration (Peking University) Ministry of Education, National Center for Trauma Medicine of China, Beijing, China
- School of Basic Medicine, Peking University, Beijing, China
| | - Zhiwei Wang
- Orthopedics Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rui Li
- Trauma Center, Peking University People’s Hospital, Key Laboratory of Trauma Treatment and Neural Regeneration (Peking University) Ministry of Education, National Center for Trauma Medicine of China, Beijing, China
| | - Lebin Gan
- Trauma Center, Peking University People’s Hospital, Key Laboratory of Trauma Treatment and Neural Regeneration (Peking University) Ministry of Education, National Center for Trauma Medicine of China, Beijing, China
| | - Mengwei Zhang
- Trauma Center, Peking University People’s Hospital, Key Laboratory of Trauma Treatment and Neural Regeneration (Peking University) Ministry of Education, National Center for Trauma Medicine of China, Beijing, China
| | - Tianbing Wang
- Trauma Center, Peking University People’s Hospital, Key Laboratory of Trauma Treatment and Neural Regeneration (Peking University) Ministry of Education, National Center for Trauma Medicine of China, Beijing, China
| |
Collapse
|
14
|
Sergi CM. NLRP-3 Inflammasome: A Key Target, but Mostly Overlooked following SARS-CoV-2 Infection. Vaccines (Basel) 2022; 10:1307. [PMID: 36016195 PMCID: PMC9413552 DOI: 10.3390/vaccines10081307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 08/10/2022] [Accepted: 08/10/2022] [Indexed: 11/30/2022] Open
Abstract
The last two years have shown many political and scientific debates during the current Coronavirus Disease 2019 (COVID-19) pandemic [...].
Collapse
Affiliation(s)
- Consolato M. Sergi
- AP Division/Pathology Laboratories, Children’s Hospital of Eastern Ontario, University of Ottawa, Ottawa, ON K1H 8L1, Canada; ; Tel.: +613-737-7600; Fax: 613-738-4837
- Department of Laboratory Medicine and Pathology, University of Alberta, Stollery Children’s Hospital, University Alberta Hospital, Edmonton, AB T6G 2B7, Canada
| |
Collapse
|