1
|
Denver P, Cunningham C. Microglial activation and neuroinflammation in acute and chronic cognitive deficits in sepsis. Neuropharmacology 2025; 267:110285. [PMID: 39746541 DOI: 10.1016/j.neuropharm.2024.110285] [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: 09/20/2024] [Revised: 12/11/2024] [Accepted: 12/26/2024] [Indexed: 01/04/2025]
Abstract
Sepsis is characterised by dysregulated immune responses to infection, leading to multi-organ dysfunction and high rates of mortality. With increasing survival rates in recent years long-term neurological and psychiatric consequences have become more apparent in survivors. Many patients develop sepsis associated encephalopathy (SAE) which encompasses the profound but usually transient neuropsychiatric syndrome delirium but also new brain injury that emerges in the months and years post-sepsis. It is now clear that systemic inflammatory signals reach the brain during sepsis and that very significant neuroinflammation ensues. The major brain resident immune cell population, the microglia, has been implicated in acute and chronic cognitive dysfunction in animal models of sepsis based on a growing number of studies using bacterial endotoxin and in polymicrobial sepsis models such as cecal ligation and puncture. The current review explores the effects of sepsis on the brain, focussing on how systemic insults translate to microglial activation and neuroinflammation and how this disrupts neuronal function and integrity. We examine what has been demonstrated specifically with respect to microglial activation, revealing robust evidence for a role for neuroinflammation in sepsis-induced brain sequelae but less clear information on the extent of the specific microglial contribution to this, arising from findings using global knockout mice, non-selective drugs and treatments that equally target peripheral and central compartments. There is, nonetheless, clear evidence that microglia do become activated and do contribute to brain consequences of sepsis thus arguing for improved understanding of these neuroinflammatory processes toward the prevention and treatment of sepsis-induced brain dysfunction.
Collapse
Affiliation(s)
- Paul Denver
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin 2, Ireland
| | - Colm Cunningham
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin 2, Ireland.
| |
Collapse
|
2
|
Ye F, Wei C, Wu A. The potential mechanism of mitochondrial homeostasis in postoperative neurocognitive disorders: an in-depth review. Ann Med 2024; 56:2411012. [PMID: 39450938 PMCID: PMC11514427 DOI: 10.1080/07853890.2024.2411012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 10/26/2024] Open
Abstract
Postoperative neurocognitive disorders (PND) are the most common neurological disorders following surgery and anaesthesia before and within 12 months after surgery, with a high prevalence in the geriatric population. PND can severely deteriorate the quality of life of patients, especially among the elderly, mainly manifested as memory loss, attention, decline and language comprehension disorders, mostly in elderly patients, with an incidence as high as 31%. Previous studies have also raised the possibility of accelerated cognitive decline and underlying neuropathological processes associated with diseases that affect cognitive performance (e.g. Alzheimer's dementia) for reasons related to anaesthesia and surgery. Currently, most research on PND has focused on various molecular pathways, especially in the geriatric population. The various hypotheses that have been proposed regarding the mechanisms imply peripheral neuroinflammation, oxidative stress, mitochondrial homeostasis, synaptic function, autophagy disorder, blood-brain barrier dysfunction, the microbiota-gut-brain axis and lack of neurotrophic support. However, the underlying pathogenesis and molecular mechanisms of PND have not yet been uncovered. Recent research has focused on mitochondrial homeostasis. In this paper, we present a review of various studies to better understand and characterize the mechanisms of associated cognitive dysfunction. As the biochemical basis of PND becomes more clearly defined, future treatments based on mitochondrial homeostasis modulation can prove to be very promising.
Collapse
Affiliation(s)
- Fan Ye
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Changwei Wei
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Anshi Wu
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
3
|
Tezer D, Başay BK, Başay Ö, Yener GO, Yüksel S. Cognitive performance, psychiatric comorbidities, and quality of life in pediatric patients with juvenile idiopathic arthritis: a comparative analysis with healthy controls. Child Neuropsychol 2024:1-19. [PMID: 39569588 DOI: 10.1080/09297049.2024.2426272] [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: 01/12/2024] [Accepted: 10/31/2024] [Indexed: 11/22/2024]
Abstract
This study aimed to assess the extent of cognitive impairment in children and adolescents with Juvenile Idiopathic Arthritis (JIA). While cognitive deficits are recognized in other systemic rheumatic diseases, exploration within the pediatric JIA population remains limited. The investigation utilized a comprehensive approach to examine neuropsychological test performance. A cohort of 160 participants (79 JIA, 81 healthy controls aged 8-17) underwent evaluations using the Pediatric Quality of Life Inventory (PedsQL), Schedule for Affective Disorders and Schizophrenia for School Age Children-Present and Lifetime Version (K-SADS-PL), and the computerized neurocognitive test battery Central Nervous System Vital Signs (CNSVS). Children with JIA exhibited statistically significant cognitive deficits across various parameters (p < .05). This was associated with an increased prevalence of lifelong psychiatric illnesses and diminished overall quality of life compared to healthy counterparts (p < .05). Analysis highlighted that specific JIA subtypes, excluding Oligoarthritis, significantly elevated the risk of neurocognitive impairments, emphasizing the impact on various cognitive outcomes (OR range: 3.1-5.1, 95% CI: 1.163-19.980). Additionally, the active disease stage was identified as a specific risk factor, amplifying the likelihood of low executive functions by 4.3 times (OR: 4.363, 95% CI: 1.095-17.378). This study underscores the critical importance of recognizing and addressing neurocognitive impairments in children with JIA. Specific attention to disease subtypes and activity levels is crucial, with the potential for targeted interventions to enhance overall cognitive well-being and quality of life in this vulnerable population.
Collapse
Affiliation(s)
- Damla Tezer
- Department of Child and Adolescent Psychiatry, Niğde Ömer Halis Demir University Education and Research Hospital, Niğde, Turkey
| | - Bürge Kabukçu Başay
- Faculty of Medicine, Department of Child and Adolescent Psychiatry, Pamukkale University, Denizli, Turkey
| | - Ömer Başay
- Faculty of Medicine, Department of Child and Adolescent Psychiatry, Pamukkale University, Denizli, Turkey
| | - Gülçin Otar Yener
- Department of Pediatrics, Section of Pediatric Rheumatology, Eskişehir State Hospital, Eskişehir, Turkey
| | - Selçuk Yüksel
- Faculty of Medicine, Department of Pediatric Rheumatology, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| |
Collapse
|
4
|
Schrock JM. Accelerated aging in people living with HIV: The neuroimmune feedback model. Brain Behav Immun Health 2024; 36:100737. [PMID: 38356933 PMCID: PMC10864877 DOI: 10.1016/j.bbih.2024.100737] [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: 08/18/2023] [Revised: 01/02/2024] [Accepted: 02/04/2024] [Indexed: 02/16/2024] Open
Abstract
People living with HIV (PLWH) experience earlier onset of aging-related comorbidities compared to their counterparts without HIV. This paper lays out a theoretical model to explain why PLWH experience accelerated aging. Briefly, the model is structured as follows. PLWH experience disproportionately heavy burdens of psychosocial stress across the life course. This psychosocial stress increases risks for depressive symptoms and problematic substance use. Depressive symptoms and problematic substance use interfere with long-term adherence to antiretroviral therapy (ART). Lower ART adherence, in turn, exacerbates the elevated systemic inflammation stemming from HIV infection. This inflammation increases risks for aging-related comorbidities. Systemic inflammation also reduces connectivity in the brain's central executive network (CEN), a large-scale brain network that is critical for coping with stressful circumstances. This reduced capacity for coping with stress leads to further increases in depressive symptoms and problematic substance use. Together, these changes form a neuroimmune feedback loop that amplifies the impact of psychosocial stress on aging-related comorbidities. In this paper, I review the existing evidence relevant to this model and highlight directions for future research.
Collapse
Affiliation(s)
- Joshua M. Schrock
- Institute for Sexual and Gender Minority Health and Wellbeing, Northwestern University, 625 N. Michigan Avenue, Suite 1400, Chicago, IL, 60611, United states
| |
Collapse
|
5
|
McKay TB, Khawaja ZQ, Freedman IG, Turco I, Wiredu K, Colecchi T, Akeju O. Exploring the Pathophysiology of Delirium: An Overview of Biomarker Studies, Animal Models, and Tissue-Engineered Models. Anesth Analg 2023; 137:1186-1197. [PMID: 37851904 PMCID: PMC10840625 DOI: 10.1213/ane.0000000000006715] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
Delirium is an acute brain disorder associated with disorganized thinking, difficulty focusing, and confusion that commonly follows major surgery, severe infection, and illness. Older patients are at high risk for developing delirium during hospitalization, which may contribute to increased morbidity, longer hospitalization, and increased risk of institutionalization following discharge. The pathophysiology underlying delirium remains poorly studied. This review delves into the findings from biomarker studies and animal models, and highlights the potential for tissue-engineered models of the brain in studying this condition. The aim is to bring together the existing knowledge in the field and provide insight into the future direction of delirium research.
Collapse
Affiliation(s)
- Tina B. McKay
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Zain Q. Khawaja
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Isaac G. Freedman
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Isabella Turco
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Kwame Wiredu
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Talia Colecchi
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Oluwaseun Akeju
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
6
|
Dulko E, Jedrusiak M, Osuru HP, Atluri N, Illendula M, Davis EM, Beenhakker MP, Lunardi N. Sleep Fragmentation, Electroencephalographic Slowing, and Circadian Disarray in a Mouse Model for Intensive Care Unit Delirium. Anesth Analg 2023; 137:209-220. [PMID: 37192134 DOI: 10.1213/ane.0000000000006524] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
BACKGROUND We aimed to further validate our previously published animal model for delirium by testing the hypothesis that in aged mice, Anesthesia, Surgery and simulated ICU conditions (ASI) induce sleep fragmentation, electroencephalographic (EEG) slowing, and circadian disarray consistent with intensive care unit (ICU) patients with delirium. METHODS A total of 41 mice were used. Mice were implanted with EEG electrodes and randomized to ASI or control groups. ASI mice received laparotomy, anesthesia, and simulated ICU conditions. Controls did not receive ASI. Sleep was recorded at the end of ICU conditions, and hippocampal tissue was collected on EEG recording. Arousals, EEG dynamics, and circadian gene expression were compared with t tests. Two-way repeated measures analysis of variance (RM ANOVA) was used to assess sleep according to light. RESULTS ASI mice experienced frequent arousals (36.6 ± 3.2 vs 26.5 ± 3.4; P = .044; 95% confidence interval [CI], 0.29-19.79; difference in mean ± SEM, 10.04 ± 4.62) and EEG slowing (frontal theta ratio, 0.223 ± 0.010 vs 0.272 ± 0.019; P = .026; 95% CI, -0.091 to -0.007; difference in mean ± SEM, -0.05 ± 0.02) relative to controls. In ASI mice with low theta ratio, EEG slowing was associated with a higher percentage of quiet wakefulness (38.2 ± 3.6 vs 13.4 ± 3.8; P = .0002; 95% CI, -35.87 to -13.84; difference in mean ± SEM, -24.86 ± 5.19). ASI mice slept longer during the dark phases of the circadian cycle (nonrapid eye movement [NREM], dark phase 1 [D1]: 138.9 ± 8.1 minutes vs 79.6 ± 9.6 minutes, P = .0003, 95% CI, -95.87 to -22.69, predicted mean difference ± SE: -59.28 ± 13.89; NREM, dark phase 2 (D2): 159.3 ± 7.3 minutes vs 112.6 ± 15.5 minutes, P = .006, 95% CI, -83.25 to -10.07, mean difference ± SE, -46.66 ± 13.89; rapid eye movement (REM), D1: 20.5 ± 2.1 minutes vs 5.8 ± 0.8 minutes, P = .001, 95% CI, -24.60 to -4.71, mean difference ± SE, -14. 65 ± 3.77; REM, D2: 21.0 ± 2.2 minutes vs 10.3 ± 1.4 minutes, P = .029, 95% CI, -20.64 to -0.76, mean difference ± SE, -10.70 ± 3.77). The expression of essential circadian genes was also lower in ASI mice (basic helix-loop-helix ARNT like [BMAL1] : -1.3 fold change; circadian locomotor output cycles protein kaput [CLOCK] : -1.2). CONCLUSIONS ASI mice experienced EEG and circadian changes mimicking those of delirious ICU patients. These findings support further exploration of this mouse approach to characterize the neurobiology of delirium.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mark P Beenhakker
- Pharmacology, University of Virginia Health, Charlottesville, Virginia
| | | |
Collapse
|
7
|
Vasunilashorn SM, Lunardi N, Newman JC, Crosby G, Acker L, Abel T, Bhatnagar S, Cunningham C, de Cabo R, Dugan L, Hippensteel JA, Ishizawa Y, Lahiri S, Marcantonio ER, Xie Z, Inouye SK, Terrando N, Eckenhoff RG. Preclinical and translational models for delirium: Recommendations for future research from the NIDUS delirium network. Alzheimers Dement 2023; 19:2150-2174. [PMID: 36799408 PMCID: PMC10576242 DOI: 10.1002/alz.12941] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 02/18/2023]
Abstract
Delirium is a common, morbid, and costly syndrome that is closely linked to Alzheimer's disease (AD) and AD-related dementias (ADRD) as a risk factor and outcome. Human studies of delirium have advanced our knowledge of delirium incidence and prevalence, risk factors, biomarkers, outcomes, prevention, and management. However, understanding of delirium neurobiology remains limited. Preclinical and translational models for delirium, while challenging to develop, could advance our knowledge of delirium neurobiology and inform the development of new prevention and treatment approaches. We discuss the use of preclinical and translational animal models in delirium, focusing on (1) a review of current animal models, (2) challenges and strategies for replicating elements of human delirium in animals, and (3) the utility of biofluid, neurophysiology, and neuroimaging translational markers in animals. We conclude with recommendations for the development and validation of preclinical and translational models for delirium, with the goal of advancing awareness in this important field.
Collapse
Affiliation(s)
- Sarinnapha M Vasunilashorn
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Nadia Lunardi
- Department of Anesthesiology, University of Virginia, Charlottesville, Virginia, USA
| | - John C Newman
- Department of Medicine, University of California, San Francisco, California, USA
- Buck Institute for Research on Aging, Novato, California, USA
| | - Gregory Crosby
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Anesthesiology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Leah Acker
- Department of Anesthesiology, Duke University, Durham, Massachusetts, USA
| | - Ted Abel
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Seema Bhatnagar
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Colm Cunningham
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Rafael de Cabo
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, Baltimore, Maryland, USA
| | - Laura Dugan
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA
- Division of Geriatric Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- VA Tennessee Valley Geriatric Research, Education, and Clinical Center (GRECC), Nashville, Tennessee, USA
| | - Joseph A Hippensteel
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Yumiko Ishizawa
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Shouri Lahiri
- Department of Neurology, Neurosurgery, and Biomedical Sciences, Cedar-Sinai Medical Center, Los Angeles, California, USA
| | - Edward R Marcantonio
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts, USA
| | - Zhongcong Xie
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sharon K Inouye
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts, USA
| | - Niccolò Terrando
- Department of Anesthesiology, Duke University, Durham, North Carolina, USA
- Department of Cell Biology, Duke University, Durham, North Carolina, USA
- Department of Immunology, Duke University, Durham, North Carolina, USA
- Duke Center for the Study of Aging and Human Development, Duke University School of Medicine, Durham, USA
| | - Roderic G Eckenhoff
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
8
|
Brier LM, Chen S, Sherafati A, Bice AR, Lee JM, Culver JP. Transient disruption of functional connectivity and depression of neural fluctuations in a mouse model of acute septic encephalopathy. Cereb Cortex 2023; 33:3548-3561. [PMID: 35972424 PMCID: PMC10068285 DOI: 10.1093/cercor/bhac291] [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: 01/03/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
Septic encephalopathy leads to major and costly burdens for a large percentage of admitted hospital patients. Elderly patients are at an increased risk, especially those with dementia. Current treatments are aimed at sedation to combat mental status changes and are not aimed at the underlying cause of encephalopathy. Indeed, the underlying pathology linking together peripheral infection and altered neural function has not been established, largely because good, acutely accessible readouts of encephalopathy in animal models do not exist. Behavioral testing in animals lasts multiple days, outlasting the time frame of acute encephalopathy. Here, we propose optical fluorescent imaging of neural functional connectivity (FC) as a readout of encephalopathy in a mouse model of acute sepsis. Imaging and basic behavioral assessment were performed at baseline, Hr8, Hr24, and Hr72 following injection of either lipopolysaccharide or phosphate buffered saline. Neural FC strength decreased at Hr8 and returned to baseline by Hr72 in motor, somatosensory, parietal, and visual cortical regions. Additionally, neural fluctuations transiently declined at Hr8 and returned to baseline by Hr72. Both FC strength and fluctuation tone correlated with neuroscore indicating this imaging methodology is a sensitive and acute readout of encephalopathy.
Collapse
Affiliation(s)
- L M Brier
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - S Chen
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - A Sherafati
- Department of Physics, Washington University School of Arts and Science, St. Louis, MO 63110, USA
| | - A R Bice
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - J M Lee
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - J P Culver
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Physics, Washington University School of Arts and Science, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University School of Engineering, St. Louis, MO 63110, USA
- Department of Electrical and Systems Engineering, Washington University School of Engineering, St. Louis, MO 63110, USA
| |
Collapse
|
9
|
Xie Z. Microfluidics-assisted blood-brain barrier device: a powerful tool to study perioperative neurocognitive disorder. Br J Anaesth 2023; 130:e212-e214. [PMID: 36182556 DOI: 10.1016/j.bja.2022.08.024] [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: 08/08/2022] [Revised: 08/28/2022] [Accepted: 08/29/2022] [Indexed: 01/25/2023] Open
Abstract
The dysfunction of the blood-brain barrier could contribute to the pathogenesis of the perioperative neurocognitive disorder. In a recent study published in the British Journal of Anaesthesia, Yang and colleagues developed an innovative microfluidics-assisted blood-brain barrier device to investigate the effects of neuroimmune interactions on blood-brain barrier opening. The findings are important and timely to understanding the mechanistic insights of perioperative neurocognitive disorder.
Collapse
Affiliation(s)
- Zhongcong Xie
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
10
|
Liu LF, Hu Y, Liu YN, Shi DW, Liu C, Da X, Zhu SH, Zhu QY, Zhang JQ, Xu GH. Reactive oxygen species contribute to delirium-like behavior by activating CypA/MMP9 signaling and inducing blood-brain barrier impairment in aged mice following anesthesia and surgery. Front Aging Neurosci 2022; 14:1021129. [PMID: 36337710 PMCID: PMC9629746 DOI: 10.3389/fnagi.2022.1021129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
Postoperative delirium (POD) is common in the elderly and is associated with poor clinical outcomes. Reactive oxygen species (ROS) and blood-brain barrier (BBB) damage have been implicated in the development of POD, but the association between these two factors and the potential mechanism is not clear. Cyclophilin A (CypA) is a specifically chemotactic leukocyte factor that can be secreted in response to ROS, which activates matrix metalloproteinase 9 (MMP9) and mediates BBB breakdown. We, therefore, hypothesized that ROS may contribute to anesthesia/surgery-induced BBB damage and delirium-like behavior via the CypA/MMP9 pathway. To test these hypotheses, 16-month-old mice were subjected to laparotomy under 3% sevoflurane anesthesia (anesthesia/surgery) for 3 h. ROS scavenger (N-acetyl-cysteine) and CypA inhibitor (Cyclosporin A) were used 0.5 h before anesthesia/surgery. A battery of behavior tests (buried food test, open field test, and Y maze test) was employed to evaluate behavioral changes at 24 h before and after surgery in the mice. Levels of tight junction proteins, CypA, MMP9, postsynaptic density protein (PSD)-95, and synaptophysin in the prefrontal cortex were assessed by western blotting. The amounts of ROS and IgG in the cortex of mice were observed by fluorescent staining. The concentration of S100β in the serum was detected by ELISA. ROS scavenger prevented the reduction in TJ proteins and restored the permeability of BBB as well as reduced the levels of CypA/MMP9, and further alleviated delirium-like behavior induced by anesthesia/surgery. Furthermore, the CypA inhibitor abolished the increased levels of CypA/MMP, which reversed BBB damage and ameliorated delirium-like behavior caused by ROS accumulation. Our findings demonstrated that ROS may participate in regulating BBB permeability in aged mice with POD via the CypA/MMP9 pathway, suggesting that CypA may be a potential molecular target for preventing POD.
Collapse
Affiliation(s)
- Li-fang Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, China
| | - Yun Hu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, China
| | - Yi-nuo Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, China
| | - De-wen Shi
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, China
| | - Chang Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, China
| | - Xin Da
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, China
| | - Si-hui Zhu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, China
| | - Qian-yun Zhu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, China
| | - Ji-qian Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, China
| | - Guang-hong Xu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, China
- *Correspondence: Guang-hong Xu,
| |
Collapse
|
11
|
Mu JL, Liu XD, Dong YH, Fang YY, Qiu SD, Zhang F, Liu KX. Peripheral interleukin-6-associated microglial QUIN elevation in basolateral amygdala contributed to cognitive dysfunction in a mouse model of postoperative delirium. Front Med (Lausanne) 2022; 9:998397. [PMID: 36160165 PMCID: PMC9500157 DOI: 10.3389/fmed.2022.998397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/16/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Developing effective approaches for postoperative delirium has been hampered due to the lack of a pathophysiologically similar animal model to offer insights into the pathogenesis. The study, therefore, aimed to develop a delirium-like mouse model and explore the underlying mechanism. METHODS The three cycles of 10-min clamp following 5-min reopening of the superior mesenteric artery (SMA) were performed in adult male C57BL/6 mice to induce a delirium-like phenotype. Composite Z score calculated based on the results of Open Field, Y Maze and Buried Food Tests was employed to assess the delirium phenotype in mice. Microglia activities were monitored by immunofluorescence staining and comprehensive morphological analysis. Systemic administration of minocycline (MINO), IL-6 antibody or IL-6 neutralizing antibody, was applied to manipulate microglia. The expressions of Indoleamine 2,3-dioxygenase-1 (IDO-1) and quinolinic acid (QUIN) were examined by RT-PCR and High-Performance Liquid Chromatography/Mass Spectrometry, respectively. Cytokines were measured using fluorescence activated cell sorting method. RESULTS The repeated ischemia/reperfusion (I/R) surgery caused significant anxiety (P < 0.05) and cognition decline in working memory and orientation (P < 0.05) in mice at postoperative 24 h. The composite Z score, indicating an overall disturbance of brain function, fluctuated over 24 h after I/R surgery (P < 0.001). Immunofluorescent staining showed that the percentage of microglia in the basolateral amygdala (BLA) (P < 0.05) was reactivated after I/R surgery and was negatively correlated with dwell time at Y maze (R = -0.759, P = 0.035). Inhibiting microglia activities by MINO reduced QUIN productions (P < 0.01) that improved cognitive deficits (P < 0.05). The peripheral IL-6 might cause IL-6 elevation in the BLA. Systemic administration of IL-6 antibodies suppressed I/R-induced IL-6 elevations (P < 0.05), microglial reactivations (P < 0.05), IDO-1 expressions (P < 0.01), and neuroactive metabolite QUIN productions (P < 0.05) in the BLA, resulting in a recovery of cognitive deficits (P < 0.05). Injection of IL-6 exerted opposite effects. CONCLUSION The repeated intestinal I/R surgery-induced mouse model is a simple and reproducible one of postoperative delirium. Peripheral IL-6-associated microglial QUIN elevations in the BLA contributed to cognitive dysfunction in the model of postoperative delirium.
Collapse
Affiliation(s)
- Jing-Lan Mu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiao-Dong Liu
- Department of Anesthesia and Intensive Care, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ye-Hong Dong
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ying-Ying Fang
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shi-Da Qiu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fu Zhang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ke-Xuan Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
12
|
Chen M, Zhang L, Shao M, Du J, Xiao Y, Zhang F, Zhang T, Li Y, Zhou Q, Liu K, Wang Z, Wu B. E4BP4 Coordinates Circadian Control of Cognition in Delirium. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200559. [PMID: 35713240 PMCID: PMC9376827 DOI: 10.1002/advs.202200559] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/24/2022] [Indexed: 05/07/2023]
Abstract
Improved understanding of the etiologies of delirium, a common and severe neuropsychiatric syndrome, would facilitate the disease prevention and treatment. Here, the authors invesitgate the role of circadian rhythms in the pathogenesis of delirium. They observe perturbance of circadian rhythms in mouse models of delirium and disrupted clock gene expression in patients with delirium. In turn, physiological and genetic circadian disruptions sensitize mice to delirium with aggravated cognitive impairment. Likewise, global deletion of E4bp4 (E4 promoter-binding protein), a clock gene markedly altered in delirium conditions, results in exacerbated delirium-associated cognitive decline. Cognitive decline in delirium models is attributed to microglial activation and impaired long-term potentiation in the hippocampus. Single-cell RNA-sequencing reveals microglia as the regulatory target of E4bp4. E4bp4 restrains microglial activation via inhibiting the ERK1/2 signaling pathway. Supporting this, mice lacking in microglial E4bp4 are delirious prone, whereas mice with E4bp4 specifically deleted in hippocampal CA1 neurons have a normal phenotype. Mechanistically, E4bp4 inhibits ERK1/2 signaling by trans-repressing Mapk1/3 (genes encoding ERK1/2) via direct binding to a D-box element in the promoter region. These findings define a causal role of clock dysfunction in delirium development and indicate E4bp4 as a regulator of cognition at the crosstalk between circadian clock and delirium.
Collapse
Affiliation(s)
- Min Chen
- Institute of Molecular Rhythm and MetabolismGuangzhou University of Chinese MedicineGuangzhou510006China
- College of PharmacyJinan UniversityGuangzhou510632China
| | - Li Zhang
- College of PharmacyJinan UniversityGuangzhou510632China
| | - Mingting Shao
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhou510632China
| | - Jianhao Du
- College of PharmacyJinan UniversityGuangzhou510632China
| | - Yifei Xiao
- Institute of Molecular Rhythm and MetabolismGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Fugui Zhang
- Institute of Molecular Rhythm and MetabolismGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Tianpeng Zhang
- Institute of Molecular Rhythm and MetabolismGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Yifang Li
- College of PharmacyJinan UniversityGuangzhou510632China
| | - Qianqian Zhou
- Shenzhen People's Hospital (The Second Clinical Medical CollegeJinan University; The First Affiliated HospitalSouthern University of Science and Technology)Shenzhen518119China
| | - Kaisheng Liu
- Shenzhen People's Hospital (The Second Clinical Medical CollegeJinan University; The First Affiliated HospitalSouthern University of Science and Technology)Shenzhen518119China
| | - Zhigang Wang
- Department of Intensive Care UnitFirst Affiliated Hospital of Jinan UniversityGuangzhou510630China
| | - Baojian Wu
- Institute of Molecular Rhythm and MetabolismGuangzhou University of Chinese MedicineGuangzhou510006China
| |
Collapse
|
13
|
Aniwattanapong D, List JJ, Ramakrishnan N, Bhatti GS, Jorge R. Effect of Vagus Nerve Stimulation on Attention and Working Memory in Neuropsychiatric Disorders: A Systematic Review. Neuromodulation 2022; 25:343-355. [PMID: 35088719 DOI: 10.1016/j.neurom.2021.11.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND It has been suggested that vagus nerve stimulation (VNS) may enhance attention and working memory. The neuromodulator effects of VNS are thought to activate the release of neurotransmitters involving cognition and to promote neuronal plasticity. Therefore, VNS has been studied for its effects on attention and working memory impairment in neuropsychiatric disorders. OBJECTIVES This study aimed to assess the effects of VNS on attention and working memory among patients with neuropsychiatric disorders, examine stimulation parameters, provide mechanistic hypotheses, and propose future studies using VNS. MATERIALS AND METHODS We conducted a systematic review using electronic databases MEDLINE (Ovid), Embase (Ovid), Cochrane library, and PsycINFO (Ovid). Narrative analysis was used to describe the therapeutic effects of VNS on attention and working memory, describe stimulation parameters, and propose explanatory mechanisms. RESULTS We identified 20 studies reporting VNS effects on attention and working memory in patients with epilepsy or mood disorders. For epilepsy, there was one randomized controlled trial from all 18 studies. It demonstrated no statistically significant differences in the cognitive tasks between active and control VNS. From a within-subject experimental design, significant improvement of working memory after VNS was demonstrated. One of three nonrandomized controlled trials found significantly improved attentional performance after VNS. The cohort studies compared VNS and surgery and found attentional improvement in both groups. Nine of 12 pretest-posttest studies showed improvement of attention or working memory after VNS. For mood disorders, although one study showed significant improvement of attention following VNS, the other did not. CONCLUSIONS This review suggests that, although we identified some positive results from eligible studies, there is insufficient good-quality evidence to establish VNS as an effective intervention to enhance attention and working memory in persons with neuropsychiatric disorders. Further studies assessing the efficacy of such intervention are needed.
Collapse
Affiliation(s)
- Daruj Aniwattanapong
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Chulalongkorn Cognitive, Clinical & Computational Neuroscience Lab, Chula Neuroscience Center, King Chulalongkorn Memorial Hospital, Bangkok, Thailand; Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA.
| | - Justine J List
- Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA; Mental Health Care Line, Michael E. DeBakey Veterans Affairs Medical Center, Baylor College of Medicine, Houston, TX, USA
| | - Nithya Ramakrishnan
- Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA; Mental Health Care Line, Michael E. DeBakey Veterans Affairs Medical Center, Baylor College of Medicine, Houston, TX, USA
| | - Gursimrat S Bhatti
- Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA; Mental Health Care Line, Michael E. DeBakey Veterans Affairs Medical Center, Baylor College of Medicine, Houston, TX, USA
| | - Ricardo Jorge
- Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA; Mental Health Care Line, Michael E. DeBakey Veterans Affairs Medical Center, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
14
|
Porter GA, O’Connor JC. Brain-derived neurotrophic factor and inflammation in depression: Pathogenic partners in crime? World J Psychiatry 2022; 12:77-97. [PMID: 35111580 PMCID: PMC8783167 DOI: 10.5498/wjp.v12.i1.77] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/21/2021] [Accepted: 12/02/2021] [Indexed: 02/06/2023] Open
Abstract
Major depressive disorder is a debilitating disorder affecting millions of people each year. Brain-derived neurotrophic factor (BDNF) and inflammation are two prominent biologic risk factors in the pathogenesis of depression that have received considerable attention. Many clinical and animal studies have highlighted associations between low levels of BDNF or high levels of inflammatory markers and the development of behavioral symptoms of depression. However, less is known about potential interaction between BDNF and inflammation, particularly within the central nervous system. Emerging evidence suggests that there is bidirectional regulation between these factors with important implications for the development of depressive symptoms and anti-depressant response. Elevated levels of inflammatory mediators have been shown to reduce expression of BDNF, and BDNF may play an important negative regulatory role on inflammation within the brain. Understanding this interaction more fully within the context of neuropsychiatric disease is important for both developing a fuller understanding of biological pathogenesis of depression and for identifying novel therapeutic opportunities. Here we review these two prominent risk factors for depression with a particular focus on pathogenic implications of their interaction.
Collapse
Affiliation(s)
- Grace A Porter
- Department of Pharmacology, UT Health San Antonio, San Antonio, TX 78229, United States
| | - Jason C O’Connor
- Department of Pharmacology, University of Texas Health San Antonio, San Antonio, TX 78229, United States
- Audie L. Murphy VA Hospital, South Texas Veterans Health System, San Antonio, TX 78229, United States
| |
Collapse
|
15
|
Cholinergic blockade of neuroinflammation – from tissue to RNA regulators. Neuronal Signal 2022; 6:NS20210035. [PMID: 35211331 PMCID: PMC8837817 DOI: 10.1042/ns20210035] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/06/2022] [Accepted: 01/06/2022] [Indexed: 11/17/2022] Open
Abstract
Inflammatory stimuli and consequent pro-inflammatory immune responses may facilitate neurodegeneration and threaten survival following pathogen infection or trauma, but potential controllers preventing these risks are incompletely understood. Here, we argue that small RNA regulators of acetylcholine (ACh) signaling, including microRNAs (miRs) and transfer RNA fragments (tRFs) may tilt the balance between innate and adaptive immunity, avoid chronic inflammation and prevent the neuroinflammation-mediated exacerbation of many neurological diseases. While the restrictive permeability of the blood–brain barrier (BBB) protects the brain from peripheral immune events, this barrier can be disrupted by inflammation and is weakened with age. The consequently dysregulated balance between pro- and anti-inflammatory processes may modify the immune activities of brain microglia, astrocytes, perivascular macrophages, oligodendrocytes and dendritic cells, leading to neuronal damage. Notably, the vagus nerve mediates the peripheral cholinergic anti-inflammatory reflex and underlines the consistent control of body–brain inflammation by pro-inflammatory cytokines, which affect cholinergic functions; therefore, the disruption of this reflex can exacerbate cognitive impairments such as attention deficits and delirium. RNA regulators can contribute to re-balancing the cholinergic network and avoiding its chronic deterioration, and their activities may differ between men and women and/or wear off with age. This can lead to hypersensitivity of aged patients to inflammation and higher risks of neuroinflammation-driven cholinergic impairments such as delirium and dementia following COVID-19 infection. The age- and sex-driven differences in post-transcriptional RNA regulators of cholinergic elements may hence indicate new personalized therapeutic options for neuroinflammatory diseases.
Collapse
|
16
|
Jaywant A, Vanderlind WM, Alexopoulos GS, Fridman CB, Perlis RH, Gunning FM. Frequency and profile of objective cognitive deficits in hospitalized patients recovering from COVID-19. Neuropsychopharmacology 2021; 46:2235-2240. [PMID: 33589778 PMCID: PMC7884062 DOI: 10.1038/s41386-021-00978-8] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/16/2021] [Accepted: 01/20/2021] [Indexed: 02/06/2023]
Abstract
Early reports and case series suggest cognitive deficits occurs in some patients with COVID-19. We evaluated the frequency, severity, and profile of cognitive dysfunction in patients recovering from prolonged COVID-19 hospitalization who required acute inpatient rehabilitation prior to discharge. We analyzed cross-sectional scores from the Brief Memory and Executive Test (BMET) in a cohort of N = 57 COVID-19 patients undergoing inpatient rehabilitation, calculating the frequency of impairment based on neuropsychologist diagnosis and by age-normed BMET subtests. In total, 43 patients (75%) were male, 35 (61%) were non-white, and mean age was 64.5 (SD = 13.9) years. In total, 48 (84%) were previously living at home independently. Two patients had documented preexisting cognitive dysfunction; none had known dementia. Patients were evaluated at a mean of 43.2 (SD = 19.2) days after initial admission. In total, 50 patients (88%) had documented hypoxemic respiratory failure and 44 (77%) required intubation. Forty-six patients (81%) had cognitive impairment, ranging from mild to severe. Deficits were common in working memory (26/47 [55%] of patients), set-shifting (21/44 [47%]), divided attention (18/39 [46%]), and processing speed (14/35 [40%]). Executive dysfunction was not significantly associated with intubation length or the time from extubation to assessment, psychiatric diagnosis, or preexisting cardiovascular/metabolic disease. Attention and executive functions are frequently impaired in COVID-19 patients who require acute rehabilitation prior to discharge. Though interpretation is limited by lack of a comparator group, these results provide an early benchmark for identifying and characterizing cognitive difficulties after COVID-19. Given the frequency and pattern of impairment, easy-to-disseminate interventions that target attention and executive dysfunctions may be beneficial to this population.
Collapse
Affiliation(s)
- Abhishek Jaywant
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, US
- Department of Rehabilitation Medicine, Weill Cornell Medicine, New York, NY, US
- NewYork-Presbyterian Hospital/Weill Cornell Medical Center, New York, NY, US
| | - W Michael Vanderlind
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, US
- NewYork-Presbyterian Hospital/Weill Cornell Medical Center, New York, NY, US
| | - George S Alexopoulos
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, US
- Institute of Geriatric Psychiatry, Weill Cornell Medicine, White Plains, NY, US
| | - Chaya B Fridman
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, US
- NewYork-Presbyterian Hospital/Weill Cornell Medical Center, New York, NY, US
| | - Roy H Perlis
- Massachusetts General Hospital/Harvard Medical School, Boston, MA, US
| | - Faith M Gunning
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, US.
- NewYork-Presbyterian Hospital/Weill Cornell Medical Center, New York, NY, US.
- Institute of Geriatric Psychiatry, Weill Cornell Medicine, White Plains, NY, US.
| |
Collapse
|
17
|
Li Y, Chen L, Li Z, Song Y, Yuan Y, Liu T, Hong J, Wang Q, Chang H, Kuang Z, He J, Li Y, Mi X, Han D, Yang N, Guo X. Potential Serum Biomarkers for Postoperative Neurocognitive Disorders Based on Proteomic Analysis of Cognitive-Related Brain Regions. Front Aging Neurosci 2021; 13:741263. [PMID: 34658843 PMCID: PMC8511679 DOI: 10.3389/fnagi.2021.741263] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/03/2021] [Indexed: 12/14/2022] Open
Abstract
Postoperative neurocognitive disorders (po-NCD), including postoperative delirium (POD) and delayed neurocognitive recovery (dNCR), are common in geriatric surgical patients. However, the ideal diagnostic biomarkers to predict individual risks of po-NCDs have not been identified. In this study, proteomic analysis was used to detect dysregulated proteins in three cognitive-related brain regions, the hippocampus, prefrontal cortex, and temporal lobe, of aged dNCR rats. The common affected proteins in these three brain regions were further verified by real-time polymerase chain reaction and western blotting. Furthermore, serum samples from aged rats with dNCR and elderly hip fracture patients with POD were also assessed with enzyme linked immunosorbent assays to investigate the biomarker potential of these dysregulated proteins. The increased expression levels of haptoglobin, caseinolytic protease (ClpP), and alpha-2 macroglobulin (A2M) as well as decreased expression levels of 14-3-3β/α and biliverdin reductase-A (BVR-A) were validated by proteomic analysis in the hippocampus, prefrontal cortex, and temporal lobe of aged dNCR rats. The increased expression of haptoglobin and decreased expression of 14-3-3β/α were further demonstrated in the three brain regions by western blotting. Moreover, increased levels of S100A6 and BVR-A in the hippocampus, S100A6 in the prefrontal cortex, and A2M in the temporal lobe were also observed. More intriguingly, both decreased serum 14-3-3β/α and increased A2M in geriatric POD patients as well as decreased serum ClpP in aged dNCR rats were verified. These results not only indicate potential diagnostic biomarkers for po-NCD but also provide directions for further pathological investigations. Clinical Trial Registration:www.ClinicalTrials.gov, identifier [ChiCTR1900027393].
Collapse
Affiliation(s)
- Yitong Li
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Lei Chen
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Zhengqian Li
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Yanan Song
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Yi Yuan
- Department of Anesthesiology, Beijing Jishuitan Hospital, Beijing, China
| | - Taotao Liu
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Jingshu Hong
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Qian Wang
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Huixian Chang
- School of Information Science and Engineering, Yanshan University, Qinhuangdao, China
| | - Zhongshen Kuang
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Jindan He
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Yue Li
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Xinning Mi
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Dengyang Han
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Ning Yang
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Xiangyang Guo
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
18
|
Dalmaz C, Barth B, Pokhvisneva I, Wang Z, Patel S, Quillfeldt JA, Mendonça Filho EJ, de Lima RMS, Arcego DM, Sassi RB, Hall GBC, Kobor MS, Meaney MJ, Silveira PP. Prefrontal cortex VAMP1 gene network moderates the effect of the early environment on cognitive flexibility in children. Neurobiol Learn Mem 2021; 185:107509. [PMID: 34454100 DOI: 10.1016/j.nlm.2021.107509] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 08/10/2021] [Accepted: 08/20/2021] [Indexed: 01/07/2023]
Abstract
During development, genetic and environmental factors interact to modify specific phenotypes. Both in humans and in animal models, early adversities influence cognitive flexibility, an important brain function related to behavioral adaptation to variations in the environment. Abnormalities in cognitive functions are related to changes in synaptic connectivity in the prefrontal cortex (PFC), and altered levels of synaptic proteins. We investigated if individual variations in the expression of a network of genes co-expressed with the synaptic protein VAMP1 in the prefrontal cortex moderate the effect of early environmental quality on the performance of children in cognitive flexibility tasks. Genes overexpressed in early childhood and co-expressed with the VAMP1 gene in the PFC were selected for study. SNPs from these genes (post-clumping) were compiled in an expression-based polygenic score (PFC-ePRS-VAMP1). We evaluated cognitive performance of the 4 years-old children in two cohorts using similar cognitive flexibility tasks. In the first cohort (MAVAN) we utilized two CANTAB tasks: (a) the Intra-/Extra-dimensional Set Shift (IED) task, and (b) the Spatial Working Memory (SWM) task. In the second cohort, GUSTO, we used the Dimensional Change Card Sort (DCCS) task. The results show that in 4 years-old children, the PFC-ePRS-VAMP1 network moderates responsiveness to the effects of early adversities on the performance in attentional flexibility tests. The same result was observed for a spatial working memory task. Compared to attentional flexibility, reversal learning showed opposite effects of the environment, as moderated by the ePRS. A parallel ICA analysis was performed to identify relationships between whole-brain voxel based gray matter density and SNPs that comprise the PFC-ePRS-VAMP1. The early environment predicts differences in gray matter content in regions such as prefrontal and temporal cortices, significantly associated with a genetic component related to Wnt signaling pathways. Our data suggest that a network of genes co-expressed with VAMP1 in the PFC moderates the influence of early environment on cognitive function in children.
Collapse
Affiliation(s)
- Carla Dalmaz
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada; Depto Bioquimica e PPG CB Bioquimica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; PPG Neurociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Barbara Barth
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada; Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Irina Pokhvisneva
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Zihan Wang
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Sachin Patel
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Jorge A Quillfeldt
- PPG Neurociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Depto Biofisica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Euclides J Mendonça Filho
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Randriely Merscher Sobreira de Lima
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada; PPG Neurociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Danusa M Arcego
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada; Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Roberto Britto Sassi
- Mood Disorders Program, Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Geoffrey B C Hall
- Department of Psychology, Neuroscience & Behaviour, McMaster University, Hamilton, ON, Canada
| | - Michael S Kobor
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, Department of Medical Genetics, The University of British Columbia, 938 West 28th Avenue, Vancouver, BC V5Z 4H4, Canada
| | - Michael J Meaney
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada; Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, QC, Canada; Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Patrícia P Silveira
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada; Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, QC, Canada; PPG Neurociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
19
|
Increased peripheral inflammation in schizophrenia is associated with worse cognitive performance and related cortical thickness reductions. Eur Arch Psychiatry Clin Neurosci 2021; 271:595-607. [PMID: 33760971 DOI: 10.1007/s00406-021-01237-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 02/03/2021] [Indexed: 12/16/2022]
Abstract
While the biological substrates of brain and behavioural changes in persons with schizophrenia remain unclear, increasing evidence implicates that inflammation is involved. In schizophrenia, including first-episode psychosis and anti-psychotic naïve patients, there are numerous reports of increased peripheral inflammation, cognitive deficits and neuropathologies such as cortical thinning. Research defining the relationship between inflammation and schizophrenia symptomatology and neuropathology is needed. Therefore, we analysed the level of C-reactive protein (CRP), a peripheral inflammation marker, and its relationship with cognitive functioning in a cohort of 644 controls and 499 schizophrenia patients. In a subset of individuals who underwent MRI scanning (99 controls and 194 schizophrenia cases), we tested if serum CRP was associated with cortical thickness. CRP was significantly increased in schizophrenia patients compared to controls, co-varying for age, sex, overweight/obesity and diabetes (p < 0.006E-10). In schizophrenia, increased CRP was mildly associated with worse performance in attention, controlling for age, sex and education (R =- 0.15, p = 0.001). Further, increased CRP was associated with reduced cortical thickness in three regions related to attention: the caudal middle frontal, the pars opercularis and the posterior cingulate cortices, which remained significant after controlling for multiple comparisons (all p < 0.05). Together, these findings indicate that increased peripheral inflammation is associated with deficits in cognitive function and brain structure in schizophrenia, especially reduced attention and reduced cortical thickness in associated brain regions. Using CRP as a biomarker of peripheral inflammation in persons with schizophrenia may help to identify vulnerable patients and those that may benefit from adjunctive anti-inflammatory treatments.
Collapse
|
20
|
Ibala R, Mekonnen J, Gitlin J, Hahm EY, Ethridge BR, Colon KM, Marota S, Ortega C, Pedemonte JC, Cobanaj M, Chamadia S, Qu J, Gao L, Barbieri R, Akeju O. A polysomnography study examining the association between sleep and postoperative delirium in older hospitalized cardiac surgical patients. J Sleep Res 2021; 30:e13322. [PMID: 33759264 DOI: 10.1111/jsr.13322] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/13/2020] [Accepted: 02/09/2021] [Indexed: 12/01/2022]
Abstract
Hospitalized older patients who undergo elective cardiac surgery with cardiopulmonary bypass are prone to postoperative delirium. Self-reported shorter sleep and longer sleep have been associated with impaired cognition. Few data exist to guide us on whether shorter or longer sleep is associated with postoperative delirium in this hospitalized cohort. This was a prospective, single-site, observational study of hospitalized patients (>60 years) scheduled to undergo elective major cardiac surgery with cardiopulmonary bypass (n = 16). We collected and analysed overnight polysomnography data using the Somté PSG device and assessed for delirium twice a day until postoperative day 3 using the long version of the confusion assessment method and a structured chart review. We also assessed subjective sleep quality using the Pittsburg Sleep Quality Index. The delirium median preoperative hospital stay of 9 [Q1, Q3: 7, 11] days was similar to the non-delirium preoperative hospital stay of 7 [4, 9] days (p = .154). The incidence of delirium was 45.5% (10/22) in the entire study cohort and 50% (8/16) in the final cohort with clean polysomnography data. The preoperative delirium median total sleep time of 323.8 [Q1, Q3: 280.3, 382.1] min was longer than the non-delirium median total sleep time of 254.3 [210.9, 278.1] min (p = .046). This was accounted for by a longer delirium median non-rapid eye movement (REM) stage 2 sleep duration of 282.3 [229.8, 328.8] min compared to the non-delirium median non-REM stage 2 sleep duration of 202.5 [174.4, 208.9] min (p = .012). Markov chain modelling confirmed these findings. There were no differences in measures of sleep quality assessed by the Pittsburg Sleep Quality Index. Polysomnography measures of sleep obtained the night preceding surgery in hospitalized older patients scheduled for elective major cardiac surgery with cardiopulmonary bypass are suggestive of an association between longer sleep duration and postoperative delirium.
Collapse
Affiliation(s)
- Reine Ibala
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jennifer Mekonnen
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jacob Gitlin
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Eunice Y Hahm
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Breanna R Ethridge
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Katia M Colon
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Sophia Marota
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Cristy Ortega
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Juan C Pedemonte
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA.,División de Anestesiología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marisa Cobanaj
- Department of Electronics Informatics and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Shubham Chamadia
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jason Qu
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Lei Gao
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Riccardo Barbieri
- Department of Electronics Informatics and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Oluwaseun Akeju
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA.,Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
21
|
Nashi R, Misra D. Special Considerations in Geriatric Populations. Arthritis Care Res (Hoboken) 2020; 72 Suppl 10:731-737. [DOI: 10.1002/acr.24342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 05/22/2020] [Indexed: 11/06/2022]
Affiliation(s)
- Rand Nashi
- Beth Israel Deaconess Medical Center Harvard Medical School Boston Massachusetts
| | - Devyani Misra
- Beth Israel Deaconess Medical Center Harvard Medical School Boston Massachusetts
| |
Collapse
|
22
|
Illendula M, Osuru HP, Ferrarese B, Atluri N, Dulko E, Zuo Z, Lunardi N. Surgery, Anesthesia and Intensive Care Environment Induce Delirium-Like Behaviors and Impairment of Synaptic Function-Related Gene Expression in Aged Mice. Front Aging Neurosci 2020; 12:542421. [PMID: 33088271 PMCID: PMC7544741 DOI: 10.3389/fnagi.2020.542421] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 09/07/2020] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE To establish a clinically relevant mouse model of perioperative delirium. METHODS Aged C57BL/6J mice were tested at baseline in the Y-maze novel arm preference, buried food, simple discrimination task of the attentional set-shifting test, and open field tests. They were subsequently randomized to insult (anesthesia, surgery, and Intensive Care Unit environment) or control group. Insult-exposed mice received laparotomy under sevoflurane anesthesia, propofol sedation and exposure to intermittent lights, sounds and cage shaking. Controls did not receive anesthesia, surgery, or intensive care environment. All mice were tested in the Y-maze novel arm preference, buried food, attentional, and open field tests at the end of intensive care environment (0 h) and every 6 h up to 24 h. Mouse hippocampi were collected at 24 h for gene expression analyses. RESULTS Surgery, anesthesia and Intensive Care environment decreased the entries in the Y-maze novel arm at 0 h (P = 0.001), 6 h (P < 0.001), 18 h (P = 0.002), and 24 h (P = 0.029). Insult exposure increased the latency to find a buried cereal reward at 18 h (P = 0.035) and 24 h (P = 0.027), and increased the trials to criterion in the reverse compound discrimination (P = 0.013) and extradimensional shift (P < 0.001) tasks of the attentional test. The overall incidence of delirium was 72% in A/S/I mice. Messenger RNA levels of synuclein alpha (-3.785 fold change relative to controls), Neurotrophic Receptor Tyrosine Kinase1 (-2.267), and syntaxin1a (-1.498) were decreased in the hippocampus of mice 24 h after insult exposure. Protein levels of syntaxin 1a (P = 0.012), Neurotrophic Receptor Tyrosine Kinase1 (P = 0.039), synuclein alpha (P = 0.017), phosphorylated synuclein alpha (P = 0.008), synaptophysin (P = 0.002), postsynaptic density protein 95 (P = 0.003), and microtubule-associated protein 2 (P = 0.013) were also decreased, relative to controls. CONCLUSION Surgery, anesthesia and Intensive Care environment impaired mouse behaviors that depend on attention, memory, and thought organization. The changes were acute in onset and fluctuating in time. Mice with delirium exhibited decreased expression of key synaptic function-related genes. The behavioral changes induced by anesthesia, surgery, and Intensive Care environment in aged mice are consistent with the clinical features of human delirium, and support the use of this animal model for future mechanistic studies of perioperative delirium.
Collapse
Affiliation(s)
- Meghana Illendula
- Department of Anesthesiology, University of Virginia Health System, Charlottesville, VA, United States
| | - Hari Prasad Osuru
- Department of Anesthesiology, University of Virginia Health System, Charlottesville, VA, United States
| | - Bianca Ferrarese
- Department of Anesthesiology and Intensive Care Medicine, University of Padova, Padua, Italy
| | - Navya Atluri
- Department of Anesthesiology, University of Virginia Health System, Charlottesville, VA, United States
| | - Elzbieta Dulko
- Department of Anesthesiology, University of Virginia Health System, Charlottesville, VA, United States
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia Health System, Charlottesville, VA, United States
| | - Nadia Lunardi
- Department of Anesthesiology, University of Virginia Health System, Charlottesville, VA, United States
| |
Collapse
|
23
|
The association between systemic inflammation and cognitive performance in healthy adults. J Neuroimmunol 2020; 345:577272. [PMID: 32534387 DOI: 10.1016/j.jneuroim.2020.577272] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 05/22/2020] [Accepted: 05/22/2020] [Indexed: 01/02/2023]
Abstract
BACKGROUND Several studies have indicated that mild systemic inflammation is associated with the risk of cognitive impairment. However, not every cognitive domain has been evaluated to have a correlation with peripheral inflammation in healthy individuals. Therefore, we aimed to investigate the association between C-reactive protein (CRP) as a marker of peripheral inflammation with various domains of cognition in healthy adults. METHOD This study consisted of 216 healthy native German adults (138 males and 78 females, mean age: 39.12 ± 20.19 years) from "Leipzig Study for Mind-Body-Emotion Interactions" (LEMON) database. After the initial assessment and conducting the cognitive battery, a blood sample was collected and CRP level was evaluated. Patients were categorized into three groups based on their CRP level. Subsequently, demographic and cognitive features were compared across three groups and to confirm the association between CRP level and cognitive performance, general linear models (GLM) were applied. RESULTS All California Verbal Learning Task (CVLT)-evaluated aspects of memory performance were inversely associated with CRP level, some of which remained significant after the adjustment for age, gender, education, smoking status and body mass index. Moreover, GLM analysis indicated that mean reaction time of the Test of Attentional Performance-Alertness (TAP-A) test (with and without signal) was also significantly associated with CRP level. CONCLUSION The current study indicated that healthy subjects with higher levels of CRP exhibit poorer performance in verbal learning memory and general wakefulness domains of cognition.
Collapse
|
24
|
High-Pressure Pneumoperitoneum Aggravates Surgery-Induced Neuroinflammation and Cognitive Dysfunction in Aged Mice. Mediators Inflamm 2020; 2020:6983193. [PMID: 32655313 PMCID: PMC7321510 DOI: 10.1155/2020/6983193] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/13/2020] [Accepted: 05/19/2020] [Indexed: 12/15/2022] Open
Abstract
Postoperative cognitive dysfunction (POCD) is a common complication after surgery, especially in aged patients. Neuroinflammation has been closely associated with the development of POCD. While the contribution of pneumoperitoneum to the systemic inflammation has been well documented, the effect of pneumoperitoneal pressure on neuroinflammation and postoperative cognitive function remains unclear. In this study, we showed that high-pressure pneumoperitoneum promoted the postoperative neuroinflammation and microglial activation in the hippocampus and aggravated the postoperative cognitive impairment in aged mice. These results support the requirement to implement interventions with lower intra-abdominal pressure, which allows for adequate exposure of the operative field rather than a routine pressure.
Collapse
|
25
|
Deiner S, Baxter MG, Mincer JS, Sano M, Hall J, Mohammed I, O'Bryant S, Zetterberg H, Blennow K, Eckenhoff R. Human plasma biomarker responses to inhalational general anaesthesia without surgery. Br J Anaesth 2020; 125:282-290. [PMID: 32536445 DOI: 10.1016/j.bja.2020.04.085] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/07/2020] [Accepted: 04/22/2020] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Postoperative neurocognitive disorders may arise in part from adverse effects of general anaesthetics on the CNS, especially in older patients or individuals otherwise vulnerable to neurotoxicity because of systemic disease or the presence of pre-existing neuropathology. Previous studies have documented cytokine and injury biomarker responses to surgical procedures that included general anaesthesia, but it is not clear to what degree anaesthetics contribute to these responses. METHODS We performed a prospective cohort study of 59 healthy volunteers aged 40-80 yr who did not undergo surgery. Plasma markers of neurological injury and inflammation were measured immediately before and 5 h after induction of general anaesthesia with 1 minimum alveolar concentration of sevoflurane. Biomarkers included interleukin-6 (IL-6), tumour necrosis factor alpha (TNF-α), C-reactive protein (CRP), and neural injury (tau, neurofilament light [NF-L], and glial fibrillary acidic protein [GFAP]). RESULTS Baseline biomarkers were in the normal range, although NF-L and GFAP were elevated as a function of age. At 5 h after induction of anaesthesia, plasma tau, NF-L, and GFAP were significantly decreased relative to baseline. Plasma IL-6 was significantly increased after anaesthesia, but by a biologically insignificant degree (<1 pg ml-1); plasma TNF-α and CRP were unchanged. CONCLUSIONS Sevoflurane general anaesthesia without surgery, even in older adults, did not provoke an inflammatory state or neuronal injury at a concentration that is detectable by an acute elevation of measured plasma biomarkers in the early hours after exposure. CLINICAL TRIAL REGISTRATION NCT02275026.
Collapse
Affiliation(s)
- Stacie Deiner
- Department of Anesthesiology, Perioperative and Pain Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Mark G Baxter
- Department of Anesthesiology, Perioperative and Pain Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Anesthesiology, Perioperative and Pain Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joshua S Mincer
- Department of Anesthesiology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medicine, New York, NY, USA
| | - Mary Sano
- Department of Psychiatry, James J. Peters VA Medical Center, Bronx, NY, USA
| | - James Hall
- University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Ismail Mohammed
- Department of Anesthesiology, Perioperative and Pain Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sid O'Bryant
- University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; UK Dementia Research Institute at UCL, London, UK; Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Roderic Eckenhoff
- Department of Anesthesiology and Critical Care, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
26
|
Abstract
Neuroinflammation has become a key hallmark of neurological complications including perioperative pathologies such as postoperative delirium and longer-lasting postoperative cognitive dysfunction. Dysregulated inflammation and neuronal injury are emerging from clinical studies as key features of perioperative neurocognitive disorders. These findings are paralleled by a growing body of preclinical investigations aimed at better understanding how surgery and anesthesia affect the central nervous system and possibly contribute to cognitive decline. Herein, we review the role of postoperative neuroinflammation and underlying mechanisms in immune-to-brain signaling after peripheral surgery.
Collapse
Affiliation(s)
- Saraswathi Subramaniyan
- From the Center for Translational Pain Medicine, Department of Anesthe siology, Duke University Medical Center, Durham, North Carolina
| | | |
Collapse
|
27
|
Velagapudi R, Subramaniyan S, Xiong C, Porkka F, Rodriguiz RM, Wetsel WC, Terrando N. Orthopedic Surgery Triggers Attention Deficits in a Delirium-Like Mouse Model. Front Immunol 2019; 10:2675. [PMID: 31911786 PMCID: PMC6918861 DOI: 10.3389/fimmu.2019.02675] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 10/30/2019] [Indexed: 01/15/2023] Open
Abstract
Postoperative delirium is a frequent and debilitating complication, especially amongst high risk procedures such as orthopedic surgery, and its pathogenesis remains unclear. Inattention is often reported in the clinical diagnosis of delirium, however limited attempts have been made to study this cognitive domain in preclinical models. Here we implemented the 5-choice serial reaction time task (5-CSRTT) to evaluate attention in a clinically relevant mouse model following orthopedic surgery. The 5-CSRTT showed a time-dependent impairment in the number of responses made by the mice acutely after orthopedic surgery, with maximum impairment at 24 h and returning to pre-surgical performance by day 5. Similarly, the latency to the response was also delayed during this time period but returned to pre-surgical levels within several days. While correct responses decreased following surgery, the accuracy of the response (e.g., selection of the correct nose-poke) remained relatively unchanged. In a separate cohort we evaluated neuroinflammation and blood-brain barrier (BBB) dysfunction using clarified brain tissue with light-sheet microscopy. CLARITY revealed significant changes in microglial morphology and impaired astrocytic-tight junction interactions using high-resolution 3D reconstructions of the neurovascular unit. Deposition of IgG, fibrinogen, and autophagy markers (TFEB and LAMP1) were also altered in the hippocampus 24 h after surgery. Together, these results provide translational evidence for the role of peripheral surgery contributing to delirium-like behavior and disrupted neuroimmunity in adult mice.
Collapse
Affiliation(s)
- Ravikanth Velagapudi
- Department of Anesthesiology, Center for Translational Pain Medicine, Duke University Medical Center, Durham, NC, United States
| | - Saraswathi Subramaniyan
- Department of Anesthesiology, Center for Translational Pain Medicine, Duke University Medical Center, Durham, NC, United States
| | - Chao Xiong
- Department of Anesthesiology, Center for Translational Pain Medicine, Duke University Medical Center, Durham, NC, United States
| | - Fiona Porkka
- Department of Psychiatry and Behavioral Sciences, Mouse Behavioral and Neuroendocrine Analysis Core Facility, Duke University Medical Center, Durham, NC, United States
| | - Ramona M. Rodriguiz
- Department of Psychiatry and Behavioral Sciences, Mouse Behavioral and Neuroendocrine Analysis Core Facility, Duke University Medical Center, Durham, NC, United States
| | - William C. Wetsel
- Department of Psychiatry and Behavioral Sciences, Mouse Behavioral and Neuroendocrine Analysis Core Facility, Duke University Medical Center, Durham, NC, United States
- Departments of Neurobiology and Cell Biology, Duke University Medical Center, Durham, NC, United States
| | - Niccolò Terrando
- Department of Anesthesiology, Center for Translational Pain Medicine, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
28
|
Yegla B, Foster T. Effect of Systemic Inflammation on Rat Attentional Function and Neuroinflammation: Possible Protective Role for Food Restriction. Front Aging Neurosci 2019; 11:296. [PMID: 31708767 PMCID: PMC6823289 DOI: 10.3389/fnagi.2019.00296] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/14/2019] [Indexed: 12/11/2022] Open
Abstract
Background: Aging is characterized by subtle cognitive decline, which correlates with increased peripheral inflammation. Acute activation of the peripheral immune system, via lipopolysaccharide (LPS) injection, elicits deficits in hippocampal-dependent spatial memory. Little is known concerning the effect of chronic inflammation on prefrontal cortex (PFC)-dependent vigilance. We examined the impact of repeated LPS injections in young and middle-age rats on the 5-choice serial reaction time task (5-CSRTT), expecting repeated LPS treatment to induce attentional deficits with greater disruption in middle-age. Methods: Male Fischer-344 rats, 4- and 12-months-old, were food restricted and trained on the 5-CSRTT. Once rats reached criterion, they were injected with LPS (1 mg/kg, i.p.) weekly for 4 weeks and testing started 48 h after each injection. To examine the possibility that mild food restriction inherent to the behavioral task influenced inflammation markers, a second group of food-restricted or ad-lib-fed rats was assessed for cytokine changes 48 h after one injection. Results: Performing LPS-treated rats exhibited a sickness response, manifesting as reduced initiated and completed trials during the first week but recovered by the second week of testing. After the first week, LPS-treated rats continued to exhibit longer response latencies, despite no change in food retrieval latency, suggestive of LPS-induced cognitive slowing. Similarly, LPS-induced impairment of attention was observed as increased omissions with heightened cognitive demand and increased age. Repeated LPS-treatment increased the level of PFC IL-1α, and PFC IL-6 was marginally higher in middle-age rats. No effect of age or treatment was observed for plasma cytokines in performing rats. Histological examination of microglia indicated increased colocalization of Iba1+ and CD68+ cells from middle-age relative to young rats. Examination of food restriction demonstrated an attenuation of age- and LPS-related increases in plasma cytokine levels. Conclusions: Systemic inflammation, induced through LPS treatment, impaired attentional function, which was independent of sickness and exacerbated by increased cognitive demand and increased age. Additional studies revealed that food restriction, associated with the task, attenuated markers of neuroinflammation and plasma cytokines. The results emphasize the need for improved methods for modeling low-level chronic systemic inflammation to effectively examine its impact on attention during aging.
Collapse
Affiliation(s)
- Brittney Yegla
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Thomas Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
- Genetics and Genomics Program, University of Florida, Gainesville, FL, United States
| |
Collapse
|
29
|
Ahmed A, Zeng G, Jiang D, Lin H, Azhar M, Farooq AD, Choudhary MI, Liu X, Wang Q. Time-dependent impairments in learning and memory in Streptozotocin-induced hyperglycemic rats. Metab Brain Dis 2019; 34:1431-1446. [PMID: 31286327 DOI: 10.1007/s11011-019-00448-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 06/10/2019] [Indexed: 12/17/2022]
Abstract
The sedentary lifestyle is responsible for the high prevalence of diabetes which also impairs cognition including learning and memory. Various studies have highlighted the learning and memory impairments in rodent models but data regarding the timeline of their development and their correlation to biochemical parameters are scarce. So, the present study was designed to investigate the type of memory which is more susceptible to hyperglycemia and its correlation with biochemical parameters such as inflammatory cytokines, cAMP response element binding (CREB) and protein kinase B (Akt) activation. Hyperglycemia was induced using streptozotocin (STZ, 45 mg/kg i.p.) and confirmed by measuring fasting blood glucose levels after 1 week of STZ injection. Learning and memory deficits were evaluated using the Novel Object Recognition Test (NORT) and Morris water maze (MWM), and correlated with biochemical parameters (TNF-α, IL-1β, and dopamine) at 3, 6 and 9 weeks. STZ-injected rats after 3 weeks of injection demonstrated moderate hyperglycemia (blood glucose = 7.99 ± 0.62 mM) with intact learning and reference memory; however, their working memory was impaired in MWM. Severe hyperglycemia (blood glucose = 11.51 ± 0.69 mM) accompanied by impaired short, long, and working memory was evident after 6 weeks whereas learning was intact. After 9 weeks of STZ injection, hyperglycemia was more pronounced (13.69 ± 1.43 mM) and accompanied by a learning deficit in addition to short, long, and working memory impairments. The extent of hyperglycemia either in terms of duration or severity resulted in enhanced inflammation, down-regulation of the level of dopamine, protein expression of AKT and CREB, which possibly affected learning and memory negatively.
Collapse
Affiliation(s)
- Ayaz Ahmed
- Affiliated TCM hospital/ Sino-Portugal TCM International Cooperation Center / Department of Physiology in School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
- Hunan Provincial Research Center for Safety Evaluation of Drugs, Changsha, China
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Guirong Zeng
- Hunan Provincial Research Center for Safety Evaluation of Drugs, Changsha, China
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dejiang Jiang
- Hunan Provincial Research Center for Safety Evaluation of Drugs, Changsha, China
| | - Haiying Lin
- Affiliated TCM hospital/ Sino-Portugal TCM International Cooperation Center / Department of Physiology in School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Mudassar Azhar
- Hunan Provincial Research Center for Safety Evaluation of Drugs, Changsha, China
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Ahsana Dar Farooq
- Hamdard Al-Majeed College of Eastern Medicine, Hamdard University, Karachi, 74600, Pakistan
| | - Muhammad Iqbal Choudhary
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
- Department of Biochemistry, College of Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Xinmin Liu
- Affiliated TCM hospital/ Sino-Portugal TCM International Cooperation Center / Department of Physiology in School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China.
- Hunan Provincial Research Center for Safety Evaluation of Drugs, Changsha, China.
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Qiong Wang
- Affiliated TCM hospital/ Sino-Portugal TCM International Cooperation Center / Department of Physiology in School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
30
|
A Systematic Review and Meta-Analysis Examining the Impact of Sleep Disturbance on Postoperative Delirium. Crit Care Med 2019; 46:e1204-e1212. [PMID: 30222634 DOI: 10.1097/ccm.0000000000003400] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Basic science and clinical studies suggest that sleep disturbance may be a modifiable risk factor for postoperative delirium. We aimed to assess the association between preoperative sleep disturbance and postoperative delirium. DATA SOURCES We searched PubMed, Embase, CINAHL, Web of Science, and Cochrane from inception until May 31, 2017. STUDY SELECTION We performed a systematic search of the literature for all studies that reported on sleep disruption and postoperative delirium excluding cross-sectional studies, case reports, and studies not reported in English language. DATA EXTRACTION Two authors independently performed study selection and data extraction. We calculated pooled effects estimates with a random-effects model constructed in Stata and evaluated the risk of bias by formal testing (Stata Corp V.14, College Station, TX), DATA SYNTHESIS:: We included 12 studies, from 1,238 citations that met our inclusion criteria. The pooled odds ratio for the association between sleep disturbance and postoperative delirium was 5.24 (95% CI, 3.61-7.60; p < 0.001 and I = 0.0%; p = 0.76). The pooled risk ratio for the association between sleep disturbance and postoperative delirium in prospective studies (n = 6) was 2.90 (95% CI, 2.28-3.69; p < 0.001 and I = 0.0%; p = 0.89). The odds ratio associated with obstructive sleep apnea and unspecified types of sleep disorder were 4.75 (95% CI, 2.65-8.54; p < 0.001 and I = 0.0%; p = 0.85) and 5.60 (95% CI, 3.46-9.07; p < 0.001 and I = 0.0%; p = 0.41), respectively. We performed Begg's and Egger's tests for publication bias and confirmed a null result for publication bias (p = 0.371 and 0.103, respectively). CONCLUSIONS Preexisting sleep disturbances are likely associated with postoperative delirium. Whether system-level initiatives targeting patients with preoperative sleep disturbance may help reduce the prevalence, morbidity, and healthcare costs associated with postoperative delirium remains to be determined.
Collapse
|
31
|
Banqueri M, Méndez M, Gómez-Lázaro E, Arias JL. Early life stress by repeated maternal separation induces long-term neuroinflammatory response in glial cells of male rats. Stress 2019; 22:563-570. [PMID: 31007117 DOI: 10.1080/10253890.2019.1604666] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Childhood maltreatment and neglect lead to a wide range of mental disorders highlighted by hormone and immune alterations in neglected children. This social-health challenge has led to the creation of early stress models such as maternal separation (MS) in rodents. We performed a MS model (4 h per day, 21 days; n = 16 MS and n = 16 control), and then measured three parameters in adult male rat brains, in order to look for long-term effects of early life stress. We used immunocytochemistry to mark glial fibrillary acidic protein (GFAP)-positive cells, which indicates changes in astroglia, and ionized calcium binding adaptor molecule 1 (Iba-1)-positive cells, which inform about reactive microglia. In order to study mRNA levels of some immune mediators, interleukin determination (interleukin-6, IL-6; tumor necrosis factor, TNFα) mRNAs were evaluated by real-time polymerase chain reaction (rt-PCR) in discrete brain regions. Measurements of numbers of GFAP-positive cells, and expression of Iba-1, IL-6 and TNFα mRNAs were performed in prefrontal cortex (PFC): cingulate cortex (CG), prelimbic cortex (PL) and infralimbic cortex (IL), striatal areas (dorsal striatum, STD; and nucleus accumbens, ACC), and dorsal hippocampus (HC: CA1, CA3 and dentate gyrus (DG)). We found that MS produces a dramatic and sustained decrease in the astroglial population in all the areas measured (from -25% in CA1 to -85.7% in ACC), whereas increased numbers of microglia were found, in more restricted regions: STD (72.6%), ACC (31%) and CA3 (33.3%) areas. Regarding mRNA measurements, we found increased IL-6 mRNA expression in HC (104.2%), and after MS.
Collapse
Affiliation(s)
- María Banqueri
- a Laboratory of Neuroscience, Department of Psychology, University of Oviedo , Oviedo , Spain
- b Instituto de Neurociencias del Principado de Asturias (INEUROPA)
| | - Marta Méndez
- a Laboratory of Neuroscience, Department of Psychology, University of Oviedo , Oviedo , Spain
- b Instituto de Neurociencias del Principado de Asturias (INEUROPA)
| | - Eneritz Gómez-Lázaro
- c Department of Basic Psychological Processes and their Development, Basque Country University , San Sebastian , Spain
| | - Jorge L Arias
- a Laboratory of Neuroscience, Department of Psychology, University of Oviedo , Oviedo , Spain
- b Instituto de Neurociencias del Principado de Asturias (INEUROPA)
| |
Collapse
|
32
|
Carr ZJ, Miller L, Ruiz-Velasco V, Kunselman AR, Karamchandani K. In a Model of Neuroinflammation Designed to Mimic Delirium, Quetiapine Reduces Cortisol Secretion and Preserves Reversal Learning in the Attentional Set Shifting Task. J Neuroimmune Pharmacol 2019; 14:383-390. [PMID: 31119596 DOI: 10.1007/s11481-019-09857-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 05/13/2019] [Indexed: 01/21/2023]
Abstract
Quetiapine, an atypical antipsychotic medication has lacked pre-clinical validation for its purported benefits in the treatment of delirium. This laboratory investigation examined the effects of quetiapine on the attentional set shifting task (ASST), a measure of cognitive flexibility and executive functioning, in a rodent model of lipopolysaccharide (LPS) mediated neuroinflammation. 19 Sprague Dawley female rats were randomly selected to receive intraperitoneal placebo (N = 5), LPS and placebo (N = 7) or LPS and quetiapine (n = 7) and performed the ASST. We measured trials to criterion, errors, non-locomotion episodes and latency to criterion, serum cortisol and tumor necrosis factor alpha (TNF-α) levels. TNF-α levels were not different between groups at 24 h. Cortisol levels in the LPS + Quetiapine group were reduced compared to LPS + Placebo (P < 0.001) and did not differ from the placebo group (P = 0.15). Analysis between LPS + Quetiapine and LPS + Placebo treated rats demonstrated improvement in the compound discrimination reversal (CD Rev1) (P = 0.016) and the intra-dimensional reversal (ID Rev2) (P = 0.007) discriminations on trials to criterion. LPS + Quetiapine treated rats had fewer errors than LPS + Placebo treated animals in the compound discrimination (CD) (P = 0.007), CD Rev1 (P = 0.005), ID Rev2 (P < 0.001) discriminations. There was no difference in non-locomotion frequency or latency to criterion between the three groups in all discriminations (P > 0.0167). We demonstrated preserved reversal learning, no effect on attentional set shifting and normalized cortisol levels in quetiapine-treated rats in this neuroinflammatory model of delirium. This suggests that quetiapine's beneficial effects in delirium may be related to the preservation of reversal learning and potential downstream effects related to reduction in cortisol production. Graphical Abstract.
Collapse
Affiliation(s)
- Zyad J Carr
- Department of Anesthesiology & Perioperative Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, 17033, USA. .,Department of Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, 17033, USA. .,Department of Anesthesiology & Perioperative Medicine, H187, 500 University Dr., Hershey, PA, 17078, USA.
| | - Lauren Miller
- Department of Anesthesiology & Perioperative Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, 17033, USA
| | - Victor Ruiz-Velasco
- Department of Anesthesiology & Perioperative Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, 17033, USA.,Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Allen R Kunselman
- Department of Public Health Sciences, Penn State College of Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, 17033, USA
| | - Kunal Karamchandani
- Department of Anesthesiology & Perioperative Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, 17033, USA
| |
Collapse
|
33
|
Xu J, Zhu XY, Sun H, Xu XQ, Xu SA, Suo Y, Cao LJ, Zhou Q, Yu HJ, Cao WZ. Low vitamin D levels are associated with cognitive impairment in patients with Hashimoto thyroiditis. BMC Endocr Disord 2018; 18:87. [PMID: 30477467 PMCID: PMC6260768 DOI: 10.1186/s12902-018-0314-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 11/08/2018] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Cognitive impairment is commonly observed in patients with Hashimoto thyroiditis (HT). Low levels of vitamin D have been correlated with cognitive impairment in non-HT population. We examined the association of vitamin D levels with cognitive impairment in patients with HT. METHODS We recruited 194 patients with HT and 200 healthy volunteers. Levels of serum 25-hydroxyvitamin D (25(OH)D) were measured using a competitive protein-binding assay. Cognitive funtion was assessed using Montreal Cognitive Assessment score (MoCA). Subjects with a MoCA scores < 26 are considered as having mild cognitive impairment (MCI). Multivariate analysis was performed using logistic regression models. RESULTS Fifty-five HT patients (28.4%) were diagnosed as having MCI. Patients with MCI had significantly lower 25(OH)D levels when compared with patients without MCI (33.9 ± 6.2 vs. 44.3 ± 9.6 nmol/L, P < 0.001). Significant differences in 25(OH)D quartiles of HT patients were observed between the patients with MCI and the patients without MCI (P < 0.001). In multivariate analyses, serum 25(OH)D levels (≤ 34.0 and ≥ 47.1 nmol/L) were significantly associated with cognitive impairment in patients with HT (OR 6.279, 95% CI 2.673-14.834, P < 0.001; OR 0.061, 95% CI 0.008-0.491, P = 0.009, respectively). CONCLUSION Our results demonstrate an important association between serum vitamin D levels and cognitive impairment in patients with HT.
Collapse
Affiliation(s)
- Jun Xu
- Department of Emergency, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang Province China
| | - Xiang-yun Zhu
- Department of Emergency, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang Province China
| | - Hui Sun
- Department of Emergency, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang Province China
| | - Xiao-qin Xu
- Department of Emergency, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang Province China
| | - Song-ao Xu
- Department of Emergency, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang Province China
| | - Yuan Suo
- Department of Emergency, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang Province China
| | - Li-jun Cao
- Department of Emergency, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang Province China
| | - Qiang Zhou
- Department of Endocrinology, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang Province China
| | - Hui-jie Yu
- Department of Emergency, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang Province China
| | - Wei-zhong Cao
- Department of Emergency, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang Province China
| |
Collapse
|
34
|
Berger M, Terrando N, Smith SK, Browndyke JN, Newman MF, Mathew JP. Neurocognitive Function after Cardiac Surgery: From Phenotypes to Mechanisms. Anesthesiology 2018; 129:829-851. [PMID: 29621031 PMCID: PMC6148379 DOI: 10.1097/aln.0000000000002194] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
For half a century, it has been known that some patients experience neurocognitive dysfunction after cardiac surgery; however, defining its incidence, course, and causes remains challenging and controversial. Various terms have been used to describe neurocognitive dysfunction at different times after cardiac surgery, ranging from "postoperative delirium" to "postoperative cognitive dysfunction or decline." Delirium is a clinical diagnosis included in the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5). Postoperative cognitive dysfunction is not included in the DSM-5 and has been heterogeneously defined, though a recent international nomenclature effort has proposed standardized definitions for it. Here, the authors discuss pathophysiologic mechanisms that may underlie these complications, review the literature on methods to prevent them, and discuss novel approaches to understand their etiology that may lead to novel treatment strategies. Future studies should measure both delirium and postoperative cognitive dysfunction to help clarify the relationship between these important postoperative complications.
Collapse
Affiliation(s)
- Miles Berger
- Assistant Professor, Department of Anesthesiology, Duke University Medical Center, Durham, NC
| | - Niccolò Terrando
- Assistant Professor, Department of Anesthesiology, Duke University Medical Center, Durham, NC
| | - S. Kendall Smith
- Critical Care Fellow, Department of Anesthesiology, Duke University Medical Center, Durham, NC
| | - Jeffrey N. Browndyke
- Assistant Professor, Division of Geriatric Behavioral Health, Department of Psychiatry & Behavioral Sciences, Duke University Medical Center, Durham, NC
| | - Mark F. Newman
- Merel H. Harmel Professor of Anesthesiology, and President of the Private Diagnostic Clinic, Duke University Medical Center, Durham, NC
| | - Joseph P. Mathew
- Jerry Reves, MD Professor and Chair, Department of Anesthesiology, Duke University Medical Center, Durham, NC
| |
Collapse
|
35
|
Ji RR, Nackley A, Huh Y, Terrando N, Maixner W. Neuroinflammation and Central Sensitization in Chronic and Widespread Pain. Anesthesiology 2018; 129:343-366. [PMID: 29462012 PMCID: PMC6051899 DOI: 10.1097/aln.0000000000002130] [Citation(s) in RCA: 866] [Impact Index Per Article: 123.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chronic pain is maintained in part by central sensitization, a phenomenon of synaptic plasticity, and increased neuronal responsiveness in central pain pathways after painful insults. Accumulating evidence suggests that central sensitization is also driven by neuroinflammation in the peripheral and central nervous system. A characteristic feature of neuroinflammation is the activation of glial cells, such as microglia and astrocytes, in the spinal cord and brain, leading to the release of proinflammatory cytokines and chemokines. Recent studies suggest that central cytokines and chemokines are powerful neuromodulators and play a sufficient role in inducing hyperalgesia and allodynia after central nervous system administration. Sustained increase of cytokines and chemokines in the central nervous system also promotes chronic widespread pain that affects multiple body sites. Thus, neuroinflammation drives widespread chronic pain via central sensitization. We also discuss sex-dependent glial/immune signaling in chronic pain and new therapeutic approaches that control neuroinflammation for the resolution of chronic pain.
Collapse
Affiliation(s)
- Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710
| | - Andrea Nackley
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710
| | - Yul Huh
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710
| | - Niccolò Terrando
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710
| | - William Maixner
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|
36
|
Shelton KT, Qu J, Bilotta F, Brown EN, Cudemus G, D’Alessandro DA, Deng H, DiBiasio A, Gitlin JA, Hahm EY, Hobbs LE, Houle TT, Ibala R, Loggia M, Pavone KJ, Shaefi S, Tolis G, Westover MB, Akeju O. Minimizing ICU Neurological Dysfunction with Dexmedetomidine-induced Sleep (MINDDS): protocol for a randomised, double-blind, parallel-arm, placebo-controlled trial. BMJ Open 2018; 8:e020316. [PMID: 29678977 PMCID: PMC5914725 DOI: 10.1136/bmjopen-2017-020316] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Delirium, which is prevalent in postcardiac surgical patients, is an acute brain dysfunction characterised by disturbances in attention, awareness and cognition not explained by a pre-existing neurocognitive disorder. The pathophysiology of delirium remains poorly understood. However, basic science and clinical studies suggest that sleep disturbance may be a modifiable risk factor for the development of delirium. Dexmedetomidine is a α-2A adrenergic receptor agonist medication that patterns the activity of various arousal nuclei similar to sleep. A single night-time loading dose of dexmedetomidine promotes non-rapid eye movement sleep stages N2 and N3 sleep. This trial hypothesises dexmedetomidine-induced sleep as pre-emptive therapy for postoperative delirium. METHODS AND ANALYSIS The MINDDS (Minimizing ICU Neurological Dysfunction with Dexmedetomidine-induced Sleep) trial is a 370-patient block-randomised, placebo-controlled, double-blinded, single-site, parallel-arm superiority trial. Patients over 60 years old, undergoing cardiac surgery with planned cardiopulmonary bypass, will be randomised to receive a sleep-inducing dose of dexmedetomidine or placebo. The primary outcome is the incidence of delirium on postoperative day 1, assessed with the Confusion Assessment Method by staff blinded to the treatment assignment. To ensure that the study is appropriately powered for the primary outcome measure, patients will be recruited and randomised into the study until 370 patients receive the study intervention on postoperative day 0. Secondary outcomes will be evaluated by in-person assessments and medical record review for in-hospital end points, and by telephone interview for 30-day, 90-day and 180-day end points. All trial outcomes will be evaluated using an intention-to-treat analysis plan. Hypothesis testing will be performed using a two-sided significance level (type I error) of α=0.05. Sensitivity analyses using the actual treatment received will be performed and compared with the intention-to-treat analysis results. Additional sensitivity analyses will assess the potential impact of missing data due to loss of follow-up. ETHICS AND DISSEMINATION The Partners Human Research Committee approved the MINDDS trial. Recruitment began in March 2017. Dissemination plans include presentations at scientific conferences, scientific publications and popular media. TRIAL REGISTRATION NUMBER NCT02856594.
Collapse
Affiliation(s)
- Kenneth T Shelton
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jason Qu
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Federico Bilotta
- Department of Anaesthesia and Critical Care Medicine, Sapienza University of Rome, Rome, Italy
| | - Emery N Brown
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Institute for Medical Engineering and Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Gaston Cudemus
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - David A D’Alessandro
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Hao Deng
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alan DiBiasio
- Department of Pharmacy, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jacob A Gitlin
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Eunice Y Hahm
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Lauren E Hobbs
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Timothy T Houle
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Reine Ibala
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Marco Loggia
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kara J Pavone
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Shahzad Shaefi
- Department of Anesthesia and Critical Care, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - George Tolis
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - M. Brandon Westover
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Oluwaseun Akeju
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
37
|
Mamad O, Islam MN, Cunningham C, Tsanov M. Differential response of hippocampal and prefrontal oscillations to systemic LPS application. Brain Res 2017; 1681:64-74. [PMID: 29294350 PMCID: PMC5792247 DOI: 10.1016/j.brainres.2017.12.036] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 11/06/2017] [Accepted: 12/27/2017] [Indexed: 01/10/2023]
Abstract
1 mg/kg LPS i.p. injection robustly suppresses theta frequency in hippocampus. LPS administration augments delta frequency in hippocampus but not mPFC. LPS injection triggers hippocampal spike-wave discharges.
The early electrophysiological phenomena linked to systemic inflammation are largely underexplored. We developed here local field analyses to detect prodromal oscillatory abnormalities. We identified early band-specific patterns in local field potential recorded from freely-moving rats injected intraperitoneally with lipopolysaccharide (LPS, 1 mg/kg). Theta frequency was significantly reduced and this effect was not related to the decreased locomotion of the animal. Furthermore, LPS-induced alterations show a region-specific response when compared between the hippocampal region and medial prefrontal cortex. Delta mean frequency increased in the hippocampal region but not in the prefrontal cortex. We explored also the hypothesis that systemic inflammation increases the propensity of abnormally synchronized brain activity. Our data indicate that the LPS-evoked alteration of delta and theta frequency parameters reflects the formation of abnormal synchronization in similar frequency ranges. The onset of abnormal brain activity was indicated by spike-wave discharges in the range of 1–10 Hz with three main frequency domains. Importantly, the occurrence of spike-wave discharges was observed in the hippocampus but not in the cortex. In summary, the hippocampal theta rhythm is an accurate indicator of the oscillatory changes evoked by LPS application. The findings offer clear patterns of altered brain function that will facilitate mechanistic investigations of brain dysfunction and delirium occurring during sepsis.
Collapse
Affiliation(s)
- Omar Mamad
- Trinity College Institute of Neuroscience, Trinity College Dublin, Ireland; School of Psychology, Trinity College Dublin, Ireland
| | - Md Nurul Islam
- Trinity College Institute of Neuroscience, Trinity College Dublin, Ireland; School of Psychology, Trinity College Dublin, Ireland
| | - Colm Cunningham
- Trinity College Institute of Neuroscience, Trinity College Dublin, Ireland; School of Biochemistry and Immunology, Trinity College Dublin, Ireland; Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - Marian Tsanov
- Trinity College Institute of Neuroscience, Trinity College Dublin, Ireland; School of Psychology, Trinity College Dublin, Ireland.
| |
Collapse
|
38
|
Bharani KL, Derex R, Granholm AC, Ledreux A. A noradrenergic lesion aggravates the effects of systemic inflammation on the hippocampus of aged rats. PLoS One 2017; 12:e0189821. [PMID: 29261743 PMCID: PMC5736222 DOI: 10.1371/journal.pone.0189821] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 12/02/2017] [Indexed: 12/18/2022] Open
Abstract
Neuroinflammation is potentiated by early degeneration of the locus coeruleus noradrenergic pathway (LC-NE) commonly seen in aging-related neurodegenerative diseases such as Alzheimer’s disease and Parkinson’s disease. In animal models, lipopolysaccharide (LPS) induces strong peripheral immune responses that can cause cognitive changes secondary to neuroinflammation. The influence of the peripheral immune response on cognition might be exacerbated by LC-NE degeneration, but this has not been well characterized previously. In this study, we investigated how systemic inflammation affects neuroinflammation and cognition in aged rats that have had either normal or damaged LC-NE transmitter systems. Rats were first exposed to the selective noradrenergic (NE) neurotoxin N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP4) to induce degeneration of central NE pathways. Two weeks later, the rats received a low dose of LPS. This resulted in 3 treatment groups (Control, LPS-, and DSP4+LPS-treated rats) studied at 4 hours (short-term subgroup) and 7 days (long-term subgroup) following the LPS injection. DSP4+LPS-treated rats exhibited increased serum levels of several pro-inflammatory cytokines, increased astroglial and microglial activation in the hippocampus, and poorer performance in the novel object recognition task (NORT) compared to controls and LPS-treated rats. Additionally, serum and brain tissue levels of brain-derived neurotrophic factor (BDNF) were modulated over time in the DSP4+LPS group compared to the other two groups. Specifically, DSP4+LPS-treated rats in the short-term subgroup had lower hippocampal BDNF levels (~25%) than controls and LPS-treated rats, which negatively correlated with hippocampal astrogliosis and positively correlated with hippocampal IL-1β levels. Serum and hippocampal BDNF levels in the DSP4+LPS-treated rats in the long-term subgroup returned to levels similar to the control group. These results show that systemic inflammation in LC-NE-lesioned aged rats promotes an exacerbated systemic and central inflammatory response compared to LC-NE-intact rats and alters BDNF levels, indicating the important role of this neurotransmitter system in response to neuroinflammation.
Collapse
Affiliation(s)
- Krishna L. Bharani
- Department of Neurosciences, Medical University of South Carolina, BSB, Charleston, SC, United States of America
| | - Rebecca Derex
- Department of Neurosciences, Medical University of South Carolina, BSB, Charleston, SC, United States of America
| | - Ann-Charlotte Granholm
- Department of Neurosciences, Medical University of South Carolina, BSB, Charleston, SC, United States of America
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, United States of America
| | - Aurélie Ledreux
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, United States of America
- * E-mail:
| |
Collapse
|
39
|
Xin X, Xin F, Chen X, Zhang Q, Li Y, Huo S, Chang C, Wang Q. Hypertonic saline for prevention of delirium in geriatric patients who underwent hip surgery. J Neuroinflammation 2017; 14:221. [PMID: 29137628 PMCID: PMC5686947 DOI: 10.1186/s12974-017-0999-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 11/08/2017] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Postoperative delirium (POD) is a common disorder in the elderly patients, and neuroinflammation is the possible underlying mechanism. This study is designed to determine whether or not hypertonic saline (HS) pre-injection can alleviate POD in aged patients. METHODS This prospective study recruited 120 geriatric patients who underwent hip surgery. The patients were randomly divided into two groups: control group (NS group) and HS group. Patients in the NS group were pre-injected with 4 mL/kg isotonic saline, and those in the HS group were pre-injected with 4 mL/kg 7.5% HS. All 120 patients were then subjected to general anesthesia. Blood samples were extracted to detect the concentration of inflammatory factors, namely, IL-1β, IL-6, IL-10, and TNF-α, and the nerve injury factor S100β. Flow cytometry was used to detect the number of monocytes in peripheral venous blood and evaluate the relationship of inflammation to delirium. The nursing delirium screening scale (Nu-DESC) was used to determine cognitive function 1 to 3 days postoperatively. RESULTS Analysis using random-effect multivariable logistic regression indicated that HS administration before anesthesia was associated with a low risk of POD (odds ratio [OR], 0.13; 95% CI, 0.04 to 0.41; P = 0.001) and few CD14 + CD16+ monocytes (β = - 0.61; 95% CI, - 0.74 to - 0.48; P = 0.000) the following day. When the association between HS and delirium was controlled for CD14 + CD16+ monocytes, the effect size became nonsignificant (odds ratio [OR], 0.86; 95% CI, 0.14 to 5.33; P = 0.874). TNF-α was significantly associated with POD (odds ratio [OR], 1.10; 95% CI, 1.05 to 1.16; P = 0.000). However, IL-1β, IL-6, IL-10, and S100β were not significantly related to POD. CONCLUSION HS can alleviate POD in geriatric patients and may inhibit the secretion of inflammatory factors by monocytes.
Collapse
Affiliation(s)
- Xi Xin
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, No 139, Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Fei Xin
- Department of Respiration, Tianjin Institute of Respiratory Diseases, Tianjin Haihe Hospital, Tianjin Medical University, Tianjin, 300350, People's Republic of China
| | - Xuguang Chen
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, No 139, Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Qi Zhang
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, No 139, Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Yanan Li
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, No 139, Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Shuping Huo
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, No 139, Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Chongfu Chang
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, No 139, Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Qiujun Wang
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, No 139, Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China.
| |
Collapse
|
40
|
Kempuraj D, Thangavel R, Selvakumar GP, Zaheer S, Ahmed ME, Raikwar SP, Zahoor H, Saeed D, Natteru PA, Iyer S, Zaheer A. Brain and Peripheral Atypical Inflammatory Mediators Potentiate Neuroinflammation and Neurodegeneration. Front Cell Neurosci 2017; 11:216. [PMID: 28790893 PMCID: PMC5522882 DOI: 10.3389/fncel.2017.00216] [Citation(s) in RCA: 263] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 07/05/2017] [Indexed: 12/18/2022] Open
Abstract
Neuroinflammatory response is primarily a protective mechanism in the brain. However, excessive and chronic inflammatory responses can lead to deleterious effects involving immune cells, brain cells and signaling molecules. Neuroinflammation induces and accelerates pathogenesis of Parkinson’s disease (PD), Alzheimer’s disease (AD) and Multiple sclerosis (MS). Neuroinflammatory pathways are indicated as novel therapeutic targets for these diseases. Mast cells are immune cells of hematopoietic origin that regulate inflammation and upon activation release many proinflammatory mediators in systemic and central nervous system (CNS) inflammatory conditions. In addition, inflammatory mediators released from activated glial cells induce neurodegeneration in the brain. Systemic inflammation-derived proinflammatory cytokines/chemokines and other factors cause a breach in the blood brain-barrier (BBB) thereby allowing for the entry of immune/inflammatory cells including mast cell progenitors, mast cells and proinflammatory cytokines and chemokines into the brain. These peripheral-derived factors and intrinsically generated cytokines/chemokines, α-synuclein, corticotropin-releasing hormone (CRH), substance P (SP), beta amyloid 1–42 (Aβ1–42) peptide and amyloid precursor proteins can activate glial cells, T-cells and mast cells in the brain can induce additional release of inflammatory and neurotoxic molecules contributing to chronic neuroinflammation and neuronal death. The glia maturation factor (GMF), a proinflammatory protein discovered in our laboratory released from glia, activates mast cells to release inflammatory cytokines and chemokines. Chronic increase in the proinflammatory mediators induces neurotoxic Aβ and plaque formation in AD brains and neurodegeneration in PD brains. Glial cells, mast cells and T-cells can reactivate each other in neuroinflammatory conditions in the brain and augment neuroinflammation. Further, inflammatory mediators from the brain can also enter into the peripheral system through defective BBB, recruit immune cells into the brain, and exacerbate neuroinflammation. We suggest that mast cell-associated inflammatory mediators from systemic inflammation and brain could augment neuroinflammation and neurodegeneration in the brain. This review article addresses the role of some atypical inflammatory mediators that are associated with mast cell inflammation and their activation of glial cells to induce neurodegeneration.
Collapse
Affiliation(s)
- Duraisamy Kempuraj
- Harry S. Truman Memorial Veteran's Hospital, U.S. Department of Veterans AffairsColumbia, MO, United States.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Ramasamy Thangavel
- Harry S. Truman Memorial Veteran's Hospital, U.S. Department of Veterans AffairsColumbia, MO, United States.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Govindhasamy P Selvakumar
- Harry S. Truman Memorial Veteran's Hospital, U.S. Department of Veterans AffairsColumbia, MO, United States.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Smita Zaheer
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Mohammad E Ahmed
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Sudhanshu P Raikwar
- Harry S. Truman Memorial Veteran's Hospital, U.S. Department of Veterans AffairsColumbia, MO, United States.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Haris Zahoor
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Daniyal Saeed
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Prashant A Natteru
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Shankar Iyer
- Harry S. Truman Memorial Veteran's Hospital, U.S. Department of Veterans AffairsColumbia, MO, United States.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Asgar Zaheer
- Harry S. Truman Memorial Veteran's Hospital, U.S. Department of Veterans AffairsColumbia, MO, United States.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| |
Collapse
|
41
|
Schreuder L, Eggen BJ, Biber K, Schoemaker RG, Laman JD, de Rooij SE. Pathophysiological and behavioral effects of systemic inflammation in aged and diseased rodents with relevance to delirium: A systematic review. Brain Behav Immun 2017; 62:362-381. [PMID: 28088641 DOI: 10.1016/j.bbi.2017.01.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 12/26/2016] [Accepted: 01/10/2017] [Indexed: 01/20/2023] Open
Abstract
Delirium is a frequent outcome for aged and demented patients that suffer a systemic inflammatory insult. Animal models that reconstruct these etiological processes have potential to provide a better understanding of the pathophysiology of delirium. Therefore, we systematically reviewed animal studies in which systemic inflammation was superimposed on aged or diseased animal models. In total, 77 studies were identified. Aged animals were challenged with a bacterial endotoxin in 29 studies, 25 studies superimposed surgery on aged animals, and in 6 studies a bacterial infection, Escherichia coli (E. coli), was used. Diseased animals were challenged with a bacterial endotoxin in 15 studies, two studies examined effects of the cytokine IL-1β, and one study used polyinosinic:polycytidilic acid (poly I:C). This systematic review analyzed the impact of systemic inflammation on the production of inflammatory and neurotoxic mediators in peripheral blood, cerebrospinal fluid (CSF), and on the central nervous system (CNS). Moreover, concomitant behavioral and cognitive symptoms were also evaluated. Finally, outcomes of behavioral and cognitive tests from animal studies were compared to features and symptoms present in delirious patients.
Collapse
Affiliation(s)
- Leroy Schreuder
- University of Groningen, University Medical Center Groningen, University Center for Geriatric Medicine, Groningen, The Netherlands.
| | - B J Eggen
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Knut Biber
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Psychiatry and Psychotherapy, Section of Molecular Psychiatry, University of Freiburg, Freiburg, Germany.
| | - Regien G Schoemaker
- Department of Neurobiology, GELIFES, University of Groningen, Groningen, The Netherlands.
| | - Jon D Laman
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Sophia E de Rooij
- University of Groningen, University Medical Center Groningen, University Center for Geriatric Medicine, Groningen, The Netherlands.
| |
Collapse
|
42
|
Lamotrigine Reduces Inflammatory Response and Ameliorates Executive Function Deterioration in an Alzheimer's-Like Mouse Model. BIOMED RESEARCH INTERNATIONAL 2016; 2016:7810196. [PMID: 28042572 PMCID: PMC5155079 DOI: 10.1155/2016/7810196] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 10/21/2016] [Accepted: 11/14/2016] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) has been described in the literature, to be associated with impairment of executive function which develops early in the course of disease, and an effective treatment for this clinical feature remains elusive. Preclinical studies have implied that lamotrigine, an antiepileptic agent, could be a potential treatment for executive dysfunction in AD patients. Although there have been promising results in previous studies with lamotrigine, executive function has never been measured using animal models. The aim of the present study was to evaluate the effects of lamotrigine on executive function and determine whether lamotrigine can attenuate inflammatory response in an AD mouse model. Nontransgenic and transgenic mice were treated with lamotrigine (0 or 30 mg/kg/day) in a standard laboratory chow diet starting at 3 months of age. After 6 months of continuous lamotrigine administration, there was a marked improvement in executive function and a significant attenuation in the expression of proinflammatory cytokines. These results suggest that lamotrigine could ameliorate executive dysfunction and brain inflammatory response in the mouse model of AD and early lamotrigine intervention may be a promising therapeutic strategy for AD.
Collapse
|
43
|
Kolanowski A, Fick D, Litaker M, Mulhall P, Clare L, Hill N, Mogle J, Boustani M, Gill D, Yevchak-Sillner A. Effect of Cognitively Stimulating Activities on Symptom Management of Delirium Superimposed on Dementia: A Randomized Controlled Trial. J Am Geriatr Soc 2016; 64:2424-2432. [PMID: 27861718 DOI: 10.1111/jgs.14511] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To determine whether cognitively stimulating activities would reduce duration and severity of delirium and improve cognitive and physical function to a greater extent than usual care. DESIGN Single-blind randomized clinical trial. SETTING Eight post-acute care (PAC) facilities. PARTICIPANTS Community-dwelling older adults with dementia and delirium (N = 283). INTERVENTION Research staff provided cognitively stimulating activities daily for up to 30 days. MEASUREMENTS Primary outcomes were delirium duration (Confusion Assessment Method) and delirium severity (Delirium Rating Scale). Secondary outcomes were cognitive function (Digits Forward, Montreal Cognitive Assessment, CLOX) and physical function (Barthel Index). RESULTS Mean percentage of delirium-free days (intervention: 64.8%, 95% confidence interval (CI) = 59.6-70.1; control: 68.7%, 95% CI = 63.9-73.6; P = .37, Wilcoxon rank sum test) and delirium severity (range 0-39: intervention: 10.77, 95% CI = 10.10-11.45; control: 11.15, 95% CI = 10.50-11.80; difference 0.37, 95% CI = 0.56-1.31, P = .43) were similar in both groups. Significant differences for secondary outcomes favoring intervention were found (executive function (range 0-15): intervention: 6.58, 95% CI = 6.12-7.04; control: 5.89, 95% CI = 5.45-6.33; difference -0.69, 95% CI = 1.33 to -0.06, P = .03; constructional praxis (range 0-15): intervention: 8.84, 95% CI = 8.83-9.34; control: 7.53, 95% CI = 7.04-8.01; difference -1.31, 95% CI = 2.01 to -0.61, P < .001). After adjusting for baseline constructional praxis, the group comparison was no longer significant. Average length of stay was shorter in the intervention (36.09 days) than the control (53.13 days) group (standard error = 0.15, P = .01, negative binomial regression). CONCLUSION Cognitively stimulating activities did not improve delirium but improved executive function and reduced length of stay. Resolution of delirium may require more-intense nonpharmacological management when the individual has dementia.
Collapse
Affiliation(s)
- Ann Kolanowski
- College of Nursing, Pennsylvania State University, University Park, Pennsylvania
| | - Donna Fick
- College of Nursing, Pennsylvania State University, University Park, Pennsylvania
| | - Mark Litaker
- School of Dentistry, University of Alabama, Birmingham, Alabama
| | - Paula Mulhall
- Prevention Research Center, College of Health and Human Development, Pennsylvania State University, University Park, Pennsylvania
| | - Linda Clare
- Centre for Research in Ageing and Cognitive Health, Washington Singer Laboratories, Exeter, United Kingdom
| | - Nikki Hill
- College of Nursing, Pennsylvania State University, University Park, Pennsylvania
| | - Jacqueline Mogle
- College of Nursing, Pennsylvania State University, University Park, Pennsylvania
| | - Malaz Boustani
- School of Medicine and Aging Brain Center, Indiana University, Indianapolis, Indiana
| | - David Gill
- Rochester Regional Health, University of Rochester, Rochester, New York.,Department of Neurology, University of Rochester, Rochester, New York
| | | |
Collapse
|
44
|
Deferoxamine regulates neuroinflammation and iron homeostasis in a mouse model of postoperative cognitive dysfunction. J Neuroinflammation 2016; 13:268. [PMID: 27733186 PMCID: PMC5062909 DOI: 10.1186/s12974-016-0740-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 09/30/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Postoperative cognitive dysfunction (POCD) is a common complication after surgery, especially amongst elderly patients. Neuroinflammation and iron homeostasis are key hallmarks of several neurological disorders. In this study, we investigated the role of deferoxamine (DFO), a clinically used iron chelator, in a mouse model of surgery-induced cognitive dysfunction and assessed its neuroprotective effects on neuroinflammation, oxidative stress, and memory function. METHODS A model of laparotomy under general anesthesia and analgesia was used to study POCD. Twelve to 14 months C57BL/6J male mice were treated with DFO, and changes in iron signaling, microglia activity, oxidative stress, inflammatory cytokines, and neurotrophic factors were assessed in the hippocampus on postoperative days 3, 7, and 14. Memory function was evaluated using fear conditioning and Morris water maze tests. BV2 microglia cells were used to test the anti-inflammatory and neuroprotective effects of DFO. RESULTS Peripheral surgical trauma triggered changes in hippocampal iron homeostasis including ferric iron deposition, increase in hepcidin and divalent metal transporter-1, reduction in ferroportin and ferritin, and oxidative stress. Microglia activation, inflammatory cytokines, brain-derived neurotropic factor impairments, and cognitive dysfunction were found up to day 14 after surgery. Treatment with DFO significantly reduced neuroinflammation and improved cognitive decline by modulating p38 MAPK signaling, reactive oxygen species, and pro-inflammatory cytokines release. CONCLUSIONS Iron imbalance represents a novel mechanism underlying surgery-induced neuroinflammation and cognitive decline. DFO treatment regulates neuroinflammation and microglia activity after surgery.
Collapse
|
45
|
Akeju O, Kim SE, Vazquez R, Rhee J, Pavone KJ, Hobbs LE, Purdon PL, Brown EN. Spatiotemporal Dynamics of Dexmedetomidine-Induced Electroencephalogram Oscillations. PLoS One 2016; 11:e0163431. [PMID: 27711165 PMCID: PMC5053525 DOI: 10.1371/journal.pone.0163431] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 09/08/2016] [Indexed: 12/19/2022] Open
Abstract
An improved understanding of the neural correlates of altered arousal states is fundamental for precise brain state targeting in clinical settings. More specifically, electroencephalogram recordings are now increasingly being used to relate drug-specific oscillatory dynamics to clinically desired altered arousal states. Dexmedetomidine is an anesthetic adjunct typically administered in operating rooms and intensive care units to produce and maintain a sedative brain state. However, a high-density electroencephalogram characterization of the neural correlates of the dexmedetomidine-induced altered arousal state has not been previously accomplished. Therefore, we administered dexmedetomidine (1mcg/kg bolus over 10 minutes, followed by 0.7mcg/kg/hr over 50 minutes) and recorded high-density electroencephalogram signals in healthy volunteers, 18–36 years old (n = 8). We analyzed the data with multitaper spectral and global coherence methods. We found that dexmedetomidine was associated with increased slow-delta oscillations across the entire scalp, increased theta oscillations in occipital regions, increased spindle oscillations in frontal regions, and decreased beta oscillations across the entire scalp. The theta and spindle oscillations were globally coherent. During recovery from this state, these electroencephalogram signatures reverted towards baseline signatures. We report that dexmedetomidine-induced electroencephalogram signatures more closely approximate the human sleep onset process than previously appreciated. We suggest that these signatures may be targeted by real time visualization of the electroencephalogram or spectrogram in clinical settings. Additionally, these signatures may aid the development of control systems for principled neurophysiological based brain-state targeting.
Collapse
Affiliation(s)
- Oluwaseun Akeju
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
- * E-mail:
| | - Seong-Eun Kim
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Rafael Vazquez
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - James Rhee
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Kara J. Pavone
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Lauren E. Hobbs
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Patrick L. Purdon
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Emery N. Brown
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| |
Collapse
|
46
|
Mawanda F, Wallace RB, McCoy K, Abrams TE. Systemic and localized extra-central nervous system bacterial infections and the risk of dementia among US veterans: A retrospective cohort study. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2016; 4:109-117. [PMID: 27752534 PMCID: PMC5061465 DOI: 10.1016/j.dadm.2016.08.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Emerging evidence indicates associations between extra-central nervous system (CNS) bacterial infections and an increased risk for dementia; however, epidemiological evidence is still very limited. METHODS This study involved a retrospective cohort of a national sample of US veterans (N = 417,172) aged ≥56 years. Extended Cox proportional hazard models adjusted for demographic characteristics and medical and psychiatric comorbidities determined the associations between systemic and localized extra-CNS bacterial infections occurring >2 years before the initial dementia diagnosis and the risk for dementia. RESULTS Exposure to any extra-CNS bacterial infection was associated with a significantly increased risk for dementia (hazard ratio [HR] = 1.20 [95% confidence interval = 1.16-1.24]). Independently, septicemia (HR = 1.39 [1.16-1.66]), bacteremia (HR = 1.22 [1.00-1.49]), osteomyelitis (HR = 1.20 [1.06-1.37]), pneumonia (HR = 1.10 [1.02-1.19]), urinary tract infections (HR = 1.13 [1.08-1.18]), and cellulitis (HR = 1.14 [1.09-1.20]) were associated with a significantly increased risk for dementia. DISCUSSION Both systemic and localized extra-CNS bacterial infections are associated with an increased risk for developing dementia.
Collapse
Affiliation(s)
- Francis Mawanda
- Department of Epidemiology, University of Iowa, Iowa City, IA, USA
| | - Robert B. Wallace
- Department of Epidemiology, University of Iowa, Iowa City, IA, USA
- Department of Medicine, University of Iowa, Iowa City, IA, USA
| | - Kimberly McCoy
- Center for Comprehensive Access & Delivery Research and Evaluation, Iowa City VA Health Care System, Iowa City, IA, USA
| | - Thad E. Abrams
- Department of Epidemiology, University of Iowa, Iowa City, IA, USA
- Department of Medicine, University of Iowa, Iowa City, IA, USA
- Center for Comprehensive Access & Delivery Research and Evaluation, Iowa City VA Health Care System, Iowa City, IA, USA
| |
Collapse
|
47
|
Peng M, Zhang C, Dong Y, Zhang Y, Nakazawa H, Kaneki M, Zheng H, Shen Y, Marcantonio ER, Xie Z. Battery of behavioral tests in mice to study postoperative delirium. Sci Rep 2016; 6:29874. [PMID: 27435513 PMCID: PMC4951688 DOI: 10.1038/srep29874] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 06/23/2016] [Indexed: 12/31/2022] Open
Abstract
Postoperative delirium is associated with increased morbidity, mortality and cost. However, its neuropathogenesis remains largely unknown, partially owing to lack of animal model(s). We therefore set out to employ a battery of behavior tests, including natural and learned behavior, in mice to determine the effects of laparotomy under isoflurane anesthesia (Anesthesia/Surgery) on these behaviors. The mice were tested at 24 hours before and at 6, 9 and 24 hours after the Anesthesia/Surgery. Composite Z scores were calculated. Cyclosporine A, an inhibitor of mitochondria permeability transient pore, was used to determine potential mitochondria-associated mechanisms of these behavioral changes. Anesthesia/Surgery selectively impaired behaviors, including latency to eat food in buried food test, freezing time and time spent in the center in open field test, and entries and duration in the novel arm of Y maze test, with acute onset and various timecourse. The composite Z scores quantitatively demonstrated the Anesthesia/Surgery-induced behavior impairment in mice. Cyclosporine A selectively ameliorated the Anesthesia/Surgery-induced reduction in ATP levels, the increases in latency to eat food, and the decreases in entries in the novel arm. These findings suggest that we could use a battery of behavior tests to establish a mouse model to study postoperative delirium.
Collapse
Affiliation(s)
- Mian Peng
- Department of Anesthesia, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129-2060, USA
| | - Ce Zhang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129-2060, USA
- Department of Anesthesia, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033, P. R. China
| | - Yuanlin Dong
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129-2060, USA
| | - Yiying Zhang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129-2060, USA
| | - Harumasa Nakazawa
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children and Harvard Medical School, Charlestown, MA 02129-2060, USA
| | - Masao Kaneki
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children and Harvard Medical School, Charlestown, MA 02129-2060, USA
| | - Hui Zheng
- Massachusetts General Hospital Biostatistics Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Yuan Shen
- Department of Psychiatry, Tenth People’s Hospital of Tongji University, Shanghai, 200072, P. R. China
| | - Edward R. Marcantonio
- Divisions of General Medicine and Primary Care and Gerontology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215
| | - Zhongcong Xie
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129-2060, USA
| |
Collapse
|
48
|
Özer S, Bozkurt H, Yılmaz R, Sönmezgöz E, Bütün I. Evaluation of executive functions in children and adolescents with familial Mediterranean fever. Child Neuropsychol 2015; 23:332-342. [DOI: 10.1080/09297049.2015.1108397] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
49
|
Ren Q, Peng M, Dong Y, Zhang Y, Chen M, Yin N, Marcantonio ER, Xie Z. Surgery plus anesthesia induces loss of attention in mice. Front Cell Neurosci 2015; 9:346. [PMID: 26441522 PMCID: PMC4561675 DOI: 10.3389/fncel.2015.00346] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 08/19/2015] [Indexed: 01/12/2023] Open
Abstract
There is a need to develop animal models to study postoperative delirium. Inattention is one of the symptoms of delirium. Increases in the levels of α-synuclein and S100β have been reported to be associated with delirium. Therefore, we set out to determine the effects of surgery plus general anesthesia on the behavioral changes (including loss of attention) in mice and on the levels of α-synuclein and S100β in the brain tissues of these mice. C57BL/6J mice (2- to 8-months-old) had a simple laparotomy plus isoflurane anesthesia. The behavioral changes, including attention level and the speed of movements, were determined 12, 24, and 48 h after the surgery plus anesthesia in the mice. The levels of α-synuclein and S100β in the cortex of these mice following the surgery plus anesthesia were determined by Western blot analysis. We found that there was a loss of attention at 24, but not 12 or 48 h following the surgery plus anesthesia (49% ± 5 vs. 33% ± 2.9, P = 0.011, N = 12) in the mice without significantly affecting the speed of their movements. There were increases in the levels of total α-synuclein (139% ± 33.5 vs. 100% ± 13.7, P = 0.037, N = 6) and S100β (142% ± 7.7 vs. 100% ± 6, P = 0.002, N = 6) in the cortex of the mice 12 h following the surgery plus anesthesia. These findings suggested that the surgery plus isoflurane anesthesia might induce behavioral and biochemical/cellular changes associated with delirium. We could use the surgery plus anesthesia in mice to develop an animal model to study postoperative delirium.
Collapse
Affiliation(s)
- Quan Ren
- Department of Anesthesia, Zhongda Hospital, Southeast University Nanjing, China ; Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School Charlestown, MA, USA
| | - Mian Peng
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School Charlestown, MA, USA ; Department of Anesthesia, Zhongnan Hospital of Wuhan University Wuhan, China
| | - Yuanlin Dong
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School Charlestown, MA, USA
| | - Yiying Zhang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School Charlestown, MA, USA
| | - Ming Chen
- Urology Department, Zhongda Hospital, Southeast University Nanjing, China
| | - Ning Yin
- Department of Anesthesia, Zhongda Hospital, Southeast University Nanjing, China
| | - Edward R Marcantonio
- Divisions of General Medicine and Primary Care and Gerontology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School Boston, MA, USA
| | - Zhongcong Xie
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School Charlestown, MA, USA
| |
Collapse
|
50
|
Worsening cognitive impairment and neurodegenerative pathology progressively increase risk for delirium. Am J Geriatr Psychiatry 2015; 23:403-415. [PMID: 25239680 PMCID: PMC4278840 DOI: 10.1016/j.jagp.2014.08.005] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 08/06/2014] [Accepted: 08/08/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND Delirium is a profound neuropsychiatric disturbance precipitated by acute illness. Although dementia is the major risk factor this has typically been considered a binary quantity (i.e., cognitively impaired versus cognitively normal) with respect to delirium risk. We used humans and mice to address the hypothesis that the severity of underlying neurodegenerative changes and/or cognitive impairment progressively alters delirium risk. METHODS Humans in a population-based longitudinal study, Vantaa 85+, were followed for incident delirium. Odds for reporting delirium at follow-up (outcome) were modeled using random-effects logistic regression, where prior cognitive impairment measured by Mini-Mental State Exam (MMSE) (exposure) was considered. To address whether underlying neurodegenerative pathology increased susceptibility to acute cognitive change, mice at three stages of neurodegenerative disease progression (ME7 model of neurodegeneration: controls, 12 weeks, and 16 weeks) were assessed for acute cognitive dysfunction upon systemic inflammation induced by bacterial lipopolysaccharide (LPS; 100 μg/kg). Synaptic and axonal correlates of susceptibility to acute dysfunction were assessed using immunohistochemistry. RESULTS In the Vantaa cohort, 465 persons (88.4 ± 2.8 years) completed MMSE at baseline. For every MMSE point lost, risk of incident delirium increased by 5% (p = 0.02). LPS precipitated severe and fluctuating cognitive deficits in 16-week ME7 mice but lower incidence or no deficits in 12-week ME7 and controls, respectively. This was associated with progressive thalamic synaptic loss and axonal pathology. CONCLUSION A human population-based cohort with graded severity of existing cognitive impairment and a mouse model with progressing neurodegeneration both indicate that the risk of delirium increases with greater severity of pre-existing cognitive impairment and neuropathology.
Collapse
|