1
|
Mazarati A. Gut-microbiota-brain Axis and post-traumatic epilepsy. Epilepsia Open 2024. [PMID: 39688879 DOI: 10.1002/epi4.13113] [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: 10/04/2024] [Revised: 11/22/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
There has been growing evidence that perturbations in gut-microbiota-brain axis (GMBA) are involved in mechanisms of chronic sequelae of traumatic brain injury (TBI). This review discusses the connection between GMBA and post-traumatic epilepsy (PTE), the latter being a common outcome of TBI. The focus is on two aspects of post-TBI GMBA dysfunction that are relevant to epilepsy. First are impairments in intestinal permeability with subsequent translocation of gut bacteria into the bloodstream. Specifically, endotoxemia following TBI may have a serendipitous protective effect against PTE through lipopolysaccharide conditioning, which may be leveraged for the development of therapeutic interventions. Second are changes in microbial composition (i.e., dysbiosis). Here, the GMBA-PTE connection is explored from predictive biomarker perspective, whereby the risk of PTE can be stratified based on specific microbial profiles. Finally, microbiota transplantation is discussed both as a tool to examine the role of gut microbiota in PTE and as a prelude to novel approaches for PTE therapy and prevention.
Collapse
Affiliation(s)
- Andrey Mazarati
- Department of Pediatrics and Children's Discovery and Innovation Institute, David Geffen School of Medicine at the University of California, Los Angeles, California, USA
| |
Collapse
|
2
|
Hooshmand M, Sadeghi MR, Asoodeh A, Pourbadie HG, Mehni MK, Sayyah M. Administration of monophosphoryl lipid A shortly after traumatic brain injury blocks the following spatial and avoidance memory loss and neuroinflammation. Sci Rep 2024; 14:29408. [PMID: 39592660 PMCID: PMC11599587 DOI: 10.1038/s41598-024-80331-3] [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: 08/27/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
Traumatic brain injury (TBI) frequently leads to cognitive impairments. The toll-like receptor 4 (TLR4) ligand, Monophosphoryl lipid A (MPL), has shown promise in modulating neuroinflammatory responses after TBI. We investigated the effects of MPL on spatial memory, passive avoidance memory, neuronal survival, and inflammatory/anti-inflammatory cytokines in rat brain following mild-to-moderate TBI. Rats underwent a learning period in the Morris water maze and shuttle box, followed by TBI induction by controlled cortical impact. MPL was administered into the cerebral ventricle 20 min after TBI. Spatial memory was assessed 7 and 28 days later. Passive avoidance memory was assessed 2 and 6 days after TBI. MPL significantly improved the spatial memory deficit at 7 days but not 28 days after TBI. It also improved impairment of the avoidance memory at both 2 and 6 days after TBI. MPL prohibited the TBI-induced TNF-α increase and IL-10 decrease in the injured region at 7 days post-TBI period. MPL prevented the neuronal loss induced by TBI in the hippocampus. A single administration of MPL shortly after TBI alleviates short-term memory deficits, through anti-inflammatory and anti-cell loss activities. Repeated MPL administration may also inhibit the long-term memory deficits after TBI.
Collapse
Affiliation(s)
- Maryam Hooshmand
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad Reza Sadeghi
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
- Department of Biochemistry, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Ahmad Asoodeh
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
| | | | - Mahbobeh Kamrani Mehni
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
- Department of Physiology, Faculty of Veterinary Medicine, Tehran University, Tehran, Iran
| | - Mohamad Sayyah
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
3
|
Ye M, Zhu H, Lu X, Yang R, Wang H, Peng J, Pan H, Fang Y, Shi R, Li F, Chen Z, Hu W, Huang C. Central innate immunization induces tolerance against post-traumatic stress disorder-like behavior and neuroinflammatory responses in male mice. Brain Behav Immun 2024; 122:368-387. [PMID: 39197543 DOI: 10.1016/j.bbi.2024.08.049] [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: 01/07/2024] [Revised: 08/23/2024] [Accepted: 08/24/2024] [Indexed: 09/01/2024] Open
Abstract
Post-traumatic stress disorder (PTSD) is a severe psychiatric disorder associated with abnormally elevated neuroinflammatory responses. Suppression of neuroinflammation is considered to be effective in ameliorating PTSD-like behaviors in rodents. Since pre-stimulation of microglia prior to stress exposure can prevent neuroinflammation, we hypothesized that pre-stimulation of microglia may prevent PTSD in animals. The results show that a single injection of a classical immune stimulant, lipopolysaccharide (LPS), at 50, 100 or 500, but not 10 μg/kg, one day before stress exposure, prevented the anxiety- and fear-like behaviors induced by modified single prolonged stress (mSPS). The time-dependent analysis shows that a single injection of LPS (100 μg/kg) either one or five, but not ten, days before stress prevented mSPS-induced anxiety- and fear-like behaviors. A second low-dose LPS injection 10 days after the first injection or a repeated LPS injection (4 × ) 10 days before stress induced tolerance to mSPS. Mechanistic studies show that a single injection of LPS one day before stress stimulation prevented mSPS-induced increases in levels of interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and IL-6 mRNA in the hippocampus and medial prefrontal cortex. Inhibition of microglia by pretreatment with minocycline or depletion of microglia by PLX3397 abolished the preventive effect of low-dose LPS pre-injection on mSPS-induced anxiety- and fear-like behavior and neuroinflammatory responses. These results suggest that pre-stimulation of microglia may prevent the development of PTSD-like behaviors by attenuating the development of neuroinflammatory responses. This could help to develop new strategies to prevent the damaging effects of harmful stress on the brain.
Collapse
Affiliation(s)
- Minxiu Ye
- Department of Pharmacy, Kunshan Hospital of Traditional Chinese Medicine, #388 Zuchongzhi South Road, Kunshan, Suzhou 215300, Jiangsu, China
| | - Haojie Zhu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Rongrong Yang
- Department of Anesthesiology, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001, Jiangsu, China
| | - Hanxiao Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Jie Peng
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Hainan Pan
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Yunli Fang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Ruiting Shi
- Faculty of Humanities and Social Sciences, City University of Macau, Av. Parde Tomas Pereira, Macau, Taipa 999078, China
| | - Fu Li
- Department of Pharmacy, Changzhou Geriatric Hospital Affiliated to Soochow University, Changzhou No.7 People's Hospital, 288# Yanling East Road, Changzhou 213000, Jiangsu, China
| | - Zhuo Chen
- Invasive Technology Department, the Second Affiliated Hospital of Nantong University, First People's Hospital of Nantong City, #666 Shengli Road, Nantong 226006, Jiangsu, China
| | - Wenfeng Hu
- Department of Pharmacy, Affiliated Maternal and Child Health Hospital of Nantong University, #399 Shijidadao, Nantong 226007, Jiangsu, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China.
| |
Collapse
|
4
|
Gaire S, Yang H, Dumre M, Lee EJ, Park SM, Joe EH. Systemic Inflammation Decreases Initial Brain Injury but Attenuates Neurite Extension and Synapse Formation during the Repair of Injured Brains. Exp Neurobiol 2024; 33:251-262. [PMID: 39568181 PMCID: PMC11581824 DOI: 10.5607/en24018] [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: 08/14/2024] [Revised: 10/13/2024] [Accepted: 10/21/2024] [Indexed: 11/22/2024] Open
Abstract
In this study, we explored the impact of systemic inflammation on initial brain injury and repair processes, including neurite extension and synapse formation. For this purpose, we established a brain injury model by administering adenosine triphosphate (ATP), a component of damage-associated molecular patterns (DAMPs), through stereotaxic injection into the striatum of mice. Systemic inflammation was induced by intraperitoneal injection of lipopolysaccharide (LPS-ip). Bulk RNA-sequencing (RNA-seq) analyses and immunostaining for microtubule-associated protein 2 (MAP2) and tyrosine hydroxylase (TH) showed that LPS-ip led to a reduction in initial brain injury, but inhibited neurite extension into the damaged brain. LPS-ip upregulated expression of defense response genes and anti-apoptotic genes, but decreased expression of genes associated with repair and regeneration. In addition, LPS-ip reduced levels of vGlut1 and PSD95 (markers for excitatory pre and post synapses, respectively), but had little effect on vGAT and gephyrin (markers for inhibitory pre and post synapses, respectively). Taken together, these findings suggest that systemic inflammation reduce initial damage but impede subsequent repair process.
Collapse
Affiliation(s)
- Sushil Gaire
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, Korea
- Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Korea
| | - Haijie Yang
- Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon 16499, Korea
| | - Manisha Dumre
- Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon 16499, Korea
| | - Eun Jeong Lee
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, Korea
- Department of Brain Science, Ajou University School of Medicine, Suwon 16499, Korea
| | - Sang-Myun Park
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, Korea
- Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon 16499, Korea
| | - Eun-Hye Joe
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, Korea
- Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon 16499, Korea
| |
Collapse
|
5
|
Cucarian J, Raposo P, Vavrek R, Nguyen A, Nelson B, Monnier P, Torres-Espin A, Fenrich K, Fouad K. No impact of anti-inflammatory medication on inflammation-driven recovery following cervical spinal cord injury in rats. Exp Neurol 2024; 383:115039. [PMID: 39481514 DOI: 10.1016/j.expneurol.2024.115039] [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: 06/13/2024] [Revised: 10/24/2024] [Accepted: 10/27/2024] [Indexed: 11/02/2024]
Abstract
Following spinal cord injury (SCI), inflammation is associated with the exacerbation of damage to spinal tissue. Consequently, managing inflammation during the acute and subacute phases is a common target in SCI treatment. However, inflammation may also induce potential benefits, including the stimulation of neuroplasticity and repair. This positive role of inflammation in spinal cord healing and functional recovery is not fully understood. To address this knowledge gap, we examined the effects of two common anti-inflammatory medications, Diphenhydramine and Methylprednisolone, on the efficacy of rehabilitative motor training on recovery from subacute cervical SCI in adult rats. Training depends critically on neuroplasticity thus if inflammation is a key regulator, we propose that anti-inflammatory drugs will reduce subsequent recovery. Both drugs were administered orally over one month, alongside task-specific reaching and grasping training. After treatment, no substantial changes in motor recovery or lesion size between the treated and control groups were observed. Treated animals also did not show any discernible changes in sensory function or anxiety-like behavior. Taken together, our data indicate that the prolonged use of these anti-inflammatory agents at commonly used doses did not profoundly impact recovery following an SCI. Therefore, considering earlier reports of the benefits of pro-inflammatory stimuli on plasticity, further studies in this area are imperative to elucidate the true impact of treating inflammation and its implications for recovery after spinal cord injuries.
Collapse
Affiliation(s)
- Jaison Cucarian
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada.
| | - Pamela Raposo
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Romana Vavrek
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Antoinette Nguyen
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Brooklynn Nelson
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Philippe Monnier
- Department of Ophthalmology and Vision Science, University of Toronto, Toronto, ON, Canada; Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Abel Torres-Espin
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada; School of Public Health Sciences, Faculty of Health, University of Waterloo, Waterloo, ON, Canada; Department of Neurological Surgery and Brain and Spinal Injury Center (BASIC), Faculty of Medicine, University of California San Francisco, San Francisco, USA
| | - Keith Fenrich
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada; Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Karim Fouad
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada; Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| |
Collapse
|
6
|
Khoshkroodian B, Javid H, Pourbadie HG, Sayyah M. Toll-Like Receptor 1/2 Postconditioning by the Ligand Pam3cys Tempers Posttraumatic Hyperexcitability, Neuroinflammation, and Microglial Response: A Potential Candidate for Posttraumatic Epilepsy. Inflammation 2024:10.1007/s10753-024-02109-z. [PMID: 39044002 DOI: 10.1007/s10753-024-02109-z] [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: 04/20/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024]
Abstract
Toll-like receptors (TLRs) are activated by endogenous molecules released from damaged cells and contribute to neuroinflammation following traumatic brain injury (TBI) and epilepsy. TLR1/2 agonist tri-palmitoyl-S-glyceryl-cysteine (Pam3cys) is a vaccine adjuvant with confirmed safety in humans. We assessed impact of TLR1/2 postconditioning by Pam3cys on epileptogenesis and neuroinflammation in male rats, 6, 24, and 48 h after mild-to-moderate TBI. Pam3cys was injected into cerebral ventricles 30 min after controlled cortical impact (CCI) injury. After 24 h, rats underwent chemical kindling by once every other day injections of pentylenetetrazole (PTZ) 35 mg/kg until development of generalized seizures. Number of intact neurons, brain expression of proinflammatory cytokine TNF-α, anti-inflammatory cytokine IL-10, and marker of anti-inflammatory microglia arginase1 (Arg1) were determined by immunoblotting. Astrocytes and macrophage/microglia activation/polarization at the contused area was assessed by double immunostaining with Iba1/Arg1, Iba1/iNOS and GFAP/iNOS, specific antibodies. The CCI-injured rats became kindled by less number of PTZ injections than sham-operated rats (9 versus 14 injections, p < 0.0001). Pam3cys treatment returned the accelerated rate of epileptogenesis in TBI state to the sham level. Pam3cys decreased neural death 48 h after TBI. It decreased TNF-α (6 h post-TBI, p < 0.01), and up-regulated IL-10 (p < 0.01) and Arg1 (p < 0.05) 48 h after TBI. The iNOS-positive cells decreased (p < 0.001) whereas Iba1/Arg1-positive cells enhanced (p < 0.01) after Pam3cys treatment. Pam3cys inhibits TBI-accelerated acquisition of seizures. Pam3cys reprograms microglia and up-regulates anti-inflammatory cytokines during the first few days after TBI. This capacity along with the clinical safety, makes Pam3cys a potential candidate for development of effective medications against posttraumatic epilepsy.
Collapse
Affiliation(s)
- Bahar Khoshkroodian
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Pasteur Street, Tehran, 13169455, Iran
| | - Hanieh Javid
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Pasteur Street, Tehran, 13169455, Iran
- Department of Neuroscience and Addition, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Gholami Pourbadie
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Pasteur Street, Tehran, 13169455, Iran
| | - Mohammad Sayyah
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Pasteur Street, Tehran, 13169455, Iran.
| |
Collapse
|
7
|
Liu H, Zhu T, Zhang L, Li F, Zheng M, Chen B, Zhu H, Ren J, Lu X, Huang C. Immunization with a low dose of zymosan A confers resistance to depression-like behavior and neuroinflammatory responses in chronically stressed mice. Behav Pharmacol 2024; 35:211-226. [PMID: 38651984 DOI: 10.1097/fbp.0000000000000774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Stimulation of the innate immune system prior to stress exposure is a possible strategy to prevent depression under stressful conditions. Based on the innate immune system stimulating activities of zymosan A, we hypothesize that zymosan A may prevent the development of chronic stress-induced depression-like behavior. Our results showed that a single injection of zymosan A 1 day before stress exposure at a dose of 2 or 4 mg/kg, but not at a dose of 1 mg/kg, prevented the development of depression-like behaviors in mice treated with chronic social defeat stress (CSDS). The prophylactic effect of a single zymosan A injection (2 mg/kg) on CSDS-induced depression-like behaviors disappeared when the time interval between zymosan A and stress exposure was extended from 1 day or 5 days to 10 days, which was rescued by a second zymosan A injection 10 days after the first zymosan A injection and 4 days (4×, once daily) of zymosan A injections 10 days before stress exposure. Further analysis showed that a single zymosan A injection (2 mg/kg) 1 day before stress exposure could prevent the CSDS-induced increase in pro-inflammatory cytokines in the hippocampus and prefrontal cortex. Inhibition of the innate immune system by pretreatment with minocycline (40 mg/kg) abolished the preventive effect of zymosan A on CSDS-induced depression-like behaviors and CSDS-induced increase in pro-inflammatory cytokines in the brain. These results suggest that activation of the innate immune system triggered by zymosan A prevents the depression-like behaviors and neuroinflammatory responses in the brain induced by chronic stress.
Collapse
Affiliation(s)
- Huijun Liu
- Department of Pharmacy, The First People's Hospital of Yancheng, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng
| | - Tao Zhu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong
| | - Linlin Zhang
- Department of Pharmacy, The First People's Hospital of Yancheng, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng
| | - Fu Li
- Department of Pharmacy, Changzhou Geriatric Hospital Affiliated to Soochow University, Changzhou No. 7 People's Hospital, Changzhou, Jiangsu, China
| | - Meng Zheng
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong
| | - Bingran Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong
| | - Haojie Zhu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong
| | - Jie Ren
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong
| |
Collapse
|
8
|
Boland R, Kokiko-Cochran ON. Deplete and repeat: microglial CSF1R inhibition and traumatic brain injury. Front Cell Neurosci 2024; 18:1352790. [PMID: 38450286 PMCID: PMC10915023 DOI: 10.3389/fncel.2024.1352790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 01/25/2024] [Indexed: 03/08/2024] Open
Abstract
Traumatic brain injury (TBI) is a public health burden affecting millions of people. Sustained neuroinflammation after TBI is often associated with poor outcome. As a result, increased attention has been placed on the role of immune cells in post-injury recovery. Microglia are highly dynamic after TBI and play a key role in the post-injury neuroinflammatory response. Therefore, microglia represent a malleable post-injury target that could substantially influence long-term outcome after TBI. This review highlights the cell specific role of microglia in TBI pathophysiology. Microglia have been manipulated via genetic deletion, drug inhibition, and pharmacological depletion in various pre-clinical TBI models. Notably, colony stimulating factor 1 (CSF1) and its receptor (CSF1R) have gained much traction in recent years as a pharmacological target on microglia. CSF1R is a transmembrane tyrosine kinase receptor that is essential for microglia proliferation, differentiation, and survival. Small molecule inhibitors targeting CSF1R result in a swift and effective depletion of microglia in rodents. Moreover, discontinuation of the inhibitors is sufficient for microglia repopulation. Attention is placed on summarizing studies that incorporate CSF1R inhibition of microglia. Indeed, microglia depletion affects multiple aspects of TBI pathophysiology, including neuroinflammation, oxidative stress, and functional recovery with measurable influence on astrocytes, peripheral immune cells, and neurons. Taken together, the data highlight an important role for microglia in sustaining neuroinflammation and increasing risk of oxidative stress, which lends to neuronal damage and behavioral deficits chronically after TBI. Ultimately, the insights gained from CSF1R depletion of microglia are critical for understanding the temporospatial role that microglia develop in mediating TBI pathophysiology and recovery.
Collapse
Affiliation(s)
- Rebecca Boland
- Department of Neuroscience, College of Medicine, Chronic Brain Injury Program, Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
| | - Olga N Kokiko-Cochran
- Department of Neuroscience, College of Medicine, Chronic Brain Injury Program, Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
9
|
Ren J, Zhang Y, Pan H, Shi R, Zhu H, Yang R, Zhang L, Chen B, Zhu T, Lu X, Huang C. Mobilization of the innate immune response by a specific immunostimulant β-glucan confers resistance to chronic stress-induced depression-like behavior by preventing neuroinflammatory responses. Int Immunopharmacol 2024; 127:111405. [PMID: 38118316 DOI: 10.1016/j.intimp.2023.111405] [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: 09/07/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 12/22/2023]
Abstract
Pre-stimulation of the innate immune response is an effective strategy to prevent depression-like phenotypes in animals. However, the use of conventional immunostimulants may cause adverse effects. Therefore, the search for agents that stimulate the innate immune response but do not induce a pro-inflammatory response could be a new research direction for the prevention of depression. β-glucan is a polysaccharide from Saccharomyces cerevisiae with unique immunomodulatory activity in microglia without eliciting a pro-inflammatory response that could lead to tissue damage. This suggests that β-glucan may be a suitable drug that can be used to prevent depression-like phenotypes. Our results showed that a single injection of β-glucan 1 day before stress exposure at a dose of 10 or 20 mg/kg, but notat a dose of 5 mg/kg, prevented depression-like behavior in mice treated with chronic unpredictable stress (CUS). This effect of β-glucan disappeared when the time interval between β-glucan and stress was extended from 1 day or 5 days to 10 days, which was rescued by a second injection 10 days after the first injection or by a repeated injection (4×, once daily) 10 days before stress exposure. A single β-glucan injection (20 mg/kg) 1 day before stress exposure prevented the CUS-induced increase in brain pro-inflammatory cytokines, and inhibition of the innate immune response by minocycline (40 mg/kg) abolished the preventive effect of β-glucan on CUS-induced depression-like behaviors and neuroinflammatory responses. These results suggest that β-glucan may prevent chronic stress-induced depression-like phenotypes and neuroinflammatory responses by stimulating the innate immune response.
Collapse
Affiliation(s)
- Jie Ren
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Yi Zhang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Hainan Pan
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Ruiting Shi
- Faculty of Humanities and Social Sciences, City University of Macau, Av. Parde Tomas Pereira, Taipa 999078, Macau
| | - Haojie Zhu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Rongrong Yang
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Lin Zhang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China; Department of Pharmacy, Affiliated Maternal and Child Health Hospital of Nantong University, #399 Shiji Dadao, Nantong 226007, Jiangsu, China
| | - Bingran Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Tao Zhu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China.
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China.
| |
Collapse
|
10
|
Zhu H, Guan A, Liu J, Peng L, Zhang Z, Wang S. Noteworthy perspectives on microglia in neuropsychiatric disorders. J Neuroinflammation 2023; 20:223. [PMID: 37794488 PMCID: PMC10548593 DOI: 10.1186/s12974-023-02901-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 09/22/2023] [Indexed: 10/06/2023] Open
Abstract
Microglia are so versatile that they not only provide immune surveillance for central nervous system, but participate in neural circuitry development, brain blood vessels formation, blood-brain barrier architecture, and intriguingly, the regulation of emotions and behaviors. Microglia have a profound impact on neuronal survival, brain wiring and synaptic plasticity. As professional phagocytic cells in the brain, they remove dead cell debris and neurotoxic agents via an elaborate mechanism. The functional profile of microglia varies considerately depending on age, gender, disease context and other internal or external environmental factors. Numerous studies have demonstrated a pivotal involvement of microglia in neuropsychiatric disorders, including negative affection, social deficit, compulsive behavior, fear memory, pain and other symptoms associated with major depression disorder, anxiety disorder, autism spectrum disorder and schizophrenia. In this review, we summarized the latest discoveries regarding microglial ontogeny, cell subtypes or state spectrum, biological functions and mechanistic underpinnings of emotional and behavioral disorders. Furthermore, we highlight the potential of microglia-targeted therapies of neuropsychiatric disorders, and propose outstanding questions to be addressed in future research of human microglia.
Collapse
Affiliation(s)
- Hongrui Zhu
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Ao Guan
- School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Jiayuan Liu
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Li Peng
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Zhi Zhang
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
- Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Sheng Wang
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| |
Collapse
|
11
|
Fu A, Qiao F, Feng H, Luo Q. Inhibition of TREM-1 Ameliorates Lipopolysaccharide-induced Depressive-like Behaviors by Alleviating Neuroinflammation in the PFC via PI3K/Akt Signaling Pathway. Behav Brain Res 2023; 449:114464. [PMID: 37142164 DOI: 10.1016/j.bbr.2023.114464] [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: 02/10/2023] [Revised: 04/19/2023] [Accepted: 05/01/2023] [Indexed: 05/06/2023]
Abstract
Neuroinflammation is closely related to depression and is a key pathophysiological process of depression. Triggering receptor expressed on myeloid cells 1 (TREM-1) has been proven to exert proinflammatory effects in various diseases. However, the role of TREM-1 in depression has not been elucidated. Thus, we hypothesized that TREM-1 inhibition might have protective effects in depression. Here, lipopolysaccharide (LPS) was used to induce depressive-like behaviors in mice, LP17 was treated to inhibit TREM-1, and LY294002 was administrated to inhibit phosphatidylinositol 3-kinase (PI3K) which is one of the downstream of TREM-1. Physical and neurobehavioral tests, Western blot analysis, and immunofluorescence staining were performed in this study. We found that LPS caused significant depressive-like behaviors in mice, including body weight decline, anodynia (sucrose preference decrease), lack of locomotor activity, and desperation in tail suspension test (TST) and forced swimming test (FST). Next, we revealed that TREM-1 was expressed on microglia, neurons, and astrocytes in the prefrontal cortex (PFC) after LPS administration. TREM-1 inhibition by LP17 suppressed the expression of TREM-1 in the PFC. In addition, LP17 could alleviate neuroinflammation and microglial activation in the PFC. Meanwhile, LP17 could prevent damage of LPS to neuronal primary cilia and neuronal activity. Finally, we revealed that PI3K/Akt might exert crucial role in the protective effects of TREM-1 inhibition to depressive-like behaviors induced by LPS. Taken together, TREM-1 inhibition by LP17 could alleviate depressive-like behaviors induced by LPS by mitigating neuroinflammation in the PFC via PI3K/Akt signaling pathway. Finally, we demonstrated that TREM-1 might be a promising therapeutic target for treatment of depression.
Collapse
Affiliation(s)
- Anhui Fu
- Department of Neurosurgery, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Fei Qiao
- Department of Neurosurgery, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Hao Feng
- Department of Neurosurgery, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Qing Luo
- Department of Ultrasound, Chonggang general hospital, Chongqing, China.
| |
Collapse
|
12
|
Innate immune tolerance against adolescent intermittent alcohol exposure-induced behavioral abnormalities in adult mice. Int Immunopharmacol 2022; 113:109250. [DOI: 10.1016/j.intimp.2022.109250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/23/2022] [Accepted: 09/09/2022] [Indexed: 11/05/2022]
|
13
|
Wang Y, Hu Z, Liu H, Gu Y, Ye M, Lu Q, Lu X, Huang C. Adolescent microglia stimulation produces long-lasting protection against chronic stress-induced behavioral abnormalities in adult male mice. Brain Behav Immun 2022; 105:44-66. [PMID: 35781008 DOI: 10.1016/j.bbi.2022.06.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/05/2022] [Accepted: 06/28/2022] [Indexed: 11/18/2022] Open
Abstract
Our previous studies had reported that microglia activation one day before stress exposure prevented the behavioral abnormalities induced by chronic stress in adult mice, and a 10-day interval between microglia stimulation and stress exposure can abolish the prophylactic effect of LPS preinjection on the behavioral abnormalities induced by chronic stress, which, however, could be rescued by repeated LPS injection. This suggests that increased stimulation of microglia results in animals developing a strong ability to prevent deleterious stress stimuli. Because microglia in the adolescent brain exhibit flexible immunological plasticity, we hypothesize that a single low-dose LPS injection during adolescence may provide long-lasting protection against behavioral abnormalities induced by chronic stress in adult mice. As expected, our results showed that a single injection of LPS (100 μg/kg) at post-natal day 28 (PND 28) prevented the development of abnormal behaviors and shifted neuroinflammatory responses toward an anti-inflammatory phenotype in adult mice treated with CSDS at their different stages of the age (PND 56, 140, and 252). Moreover, pretreatment with minocycline or PLX3397 to inhibit microglial activation abolished the prophylactic effect of LPS preinjection after PND 28 on behavioral abnormalities and neuroinflammatory responses induced by CSDS in adult mice at their different stages of the age, PND 56, 140, and 252. These results indicate that stimulation of microglia in adolescence may confer long-lasting protection against neuroinflammatory responses and behavioral abnormalities induced by chronic stress in adult mice. This may offer the potential for the development of a "vaccine-like strategy" to prevent mental disorders.
Collapse
Affiliation(s)
- Yue Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Zhichao Hu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Huijun Liu
- Department of Pharmacy, Yancheng First Hospital, the Fourth Affiliated Hospital of Nantong University, #66 Renmin South Road, Yancheng 224006, Jiangsu, China
| | - Yue Gu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Minxiu Ye
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Qun Lu
- Department of Pharmacy, Nantong Third Hospital Affiliated to Nantong University, #60 Middle Qingnian Road, Nantong 226006, Jiangsu, China
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China.
| |
Collapse
|
14
|
Innate immune stimulation prevents chronic stress-induced depressive and anxiogenic-like behaviors in female mice. Int Immunopharmacol 2022; 111:109126. [PMID: 35973368 DOI: 10.1016/j.intimp.2022.109126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/04/2022] [Accepted: 08/01/2022] [Indexed: 11/05/2022]
Abstract
It has been reported that pre-stimulation of the innate immune system can prevent depressive and anxiogenic-like behaviors in chronically stressed male mice. However, it is unclear whether similar effects can be observed in female animals. In the present study, we investigated this question in female mice. Our results showed that a single injection of lipopolysaccharide (LPS; 100 μg/kg) one day before stress exposure prevented increased immobility time in the tail suspension test and forced swimming test and decreased sucrose intake in the sucrose preference test in chronic unpredictable stress (CUS)-treated female mice. The single LPS pre-injection (100 μg/kg) prevented the CUS-induced decrease in (i) time spent in open arms and number of entries into open arms in the elevated plus maze test, (ii) time spent in lit side in the light-dark test, and (iii) time spent in the central region of the open field in the open field test, along with no changes in locomotor activity. It was also found that the single LPS pre-injection in female mice prevented the CUS-induced increase in the expression levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and IL-6 mRNA in the hippocampus and medial prefrontal cortex. Inhibition of innate immune system stimulation by minocycline pretreatment abrogated the preventive effect of LPS on CUS-induced depressive and anxiogenic-like behaviors and neuroinflammatory responses in the hippocampus and medial prefrontal cortex in female mice. These results suggest that pre-stimulation of the innate immune system by LPS injection may prevent the development of behavioral abnormalities in female mice.
Collapse
|
15
|
Activating toll-like receptor 4 after traumatic brain injury inhibits neuroinflammation and the accelerated development of seizures in rats. Exp Neurol 2022; 357:114202. [PMID: 35970203 DOI: 10.1016/j.expneurol.2022.114202] [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: 07/25/2022] [Accepted: 08/09/2022] [Indexed: 01/01/2023]
Abstract
Toll-like receptor 4 (TLR4) signaling plays a detrimental role in traumatic brain injury (TBI) pathology. Pharmacologic or genetic inactivating TLR4 diminish TBI inflammation and neurological complications. Nonetheless, TLR4 priming alleviates TBI inflammation and seizure susceptibility. We investigated impact of postconditioning with TLR4 agonist monophosphoryl lipid A (MPL) on TBI neuroinflammation and epileptogenesis in rats. TBI was induced in temporo-parietal cortex of rats by Controlled Cortical Impact device. Then rats received a single dose (0.1 μg/rat) of MPL by intracerebroventricular injection. After 24 h, CCI-injured rats received intraperitoneal injection of pentylenetetrazole 35 mg/kg once every other day until acquisition of generalized seizures. The injury size, number of survived neurons, and brain protein level of TNF-α, TGF-β, IL-10, and arginase1 (Arg1) were determined. Astrocytes and macrophage/microglia activation/polarization was assessed by double immunostaining with anti GFAP/Arg1 or anti Iba1/Arg1 antibodies. The CCI-injured rats developed generalized seizures after 5.9 ± 1.3 pentylenetetrazole injections (p < 0.001, compared to 12.3 ± 1.4 injections for sham-operated rats). MPL treatment returned the accelerated rate of epileptogenesis in TBI state to the sham-operated level. MPL did not change damage volume but attenuated number of dead neurons (p < 0.01). MPL decreased TNF-α overexpression (6 h post-TBI p < 0.0001), upregulated expression of TGF-β (48 h post-TBI, p < 0.0001), and IL-10 (48 h post-TBI, p < 0.0001) but did not change Arg1 expression. GFAP/Arg1 and Iba1/Arg1 positive cells were detected in TBI area with no significant change following MPL administration. MPL administration after TBI reduces vulnerability to seizure acquisition through down regulating neural death and inflammation, and up-regulating anti-inflammatory cytokines. This capacity along with the clinical safety, makes MPL a potential candidate for development of drugs against neurological deficits of TBI.
Collapse
|
16
|
Chen Z, Liu H, Ye Y, Chen D, Lu Q, Lu X, Huang C. Granulocyte-macrophage colony-stimulating factor-triggered innate immune tolerance against chronic stress-induced behavioral abnormalities in mice. Int Immunopharmacol 2022; 109:108924. [PMID: 35704970 DOI: 10.1016/j.intimp.2022.108924] [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: 04/26/2022] [Revised: 05/25/2022] [Accepted: 06/02/2022] [Indexed: 11/05/2022]
Abstract
Pre-stimulation of the innate immune is considered a potential strategy to prevent chronic stress-induced behavioral abnormalities in animals. In this study, we investigated whether granulocyte-macrophage colony-stimulating factor (GM-CSF), an immunostimulant used in the clinic to treat diseases of the hematopoietic system, can prevent chronic stress-induced behavioral abnormalities in mice. Our results showed that a single intraperitoneal injection of GM-CSF (100 μg/kg) one day before stress exposure prevented the depression- and anxiety-like behaviors induced by chronic social defeat stress (CSDS) in mice, including preventing the CSDS-induced increase in the immobility time in the tail suspension test and forced swimming test and decrease in the time spent in the interaction zone in the social interaction test, as well as preventing the CSDS-induced decrease in the time spent (i) in open arms in the elevated plus maze test, (ii) on the illuminated side in the light-dark test, and (iii) in the central region of the open field test. The single GM-CSF preinjection (100 μg/kg) also prevented the CSDS-induced increase in the expression levels of interleukin-6 (IL-6), IL-1β, and tumor necrosis factor-α (TNF-α) mRNA in the hippocampus and medial prefrontal cortex of the mice. Further analysis showed that the preventive effect of GM-CSF on CSDS-induced depression- and anxiety-like behaviors and neuroinflammatory responses was abolished by pretreatment with minocycline (an innate immune inhibitor). These results indicate that a single low dose of GM-CSF before injection could be a potential way to prevent behavioral abnormalities induced by chronic stress in mice.
Collapse
Affiliation(s)
- Zhuo Chen
- Invasive Technology Department, Affiliated Hospital 2 of Nantong University, First People's Hospital of Nantong City, No. 6 Haierxiang North Road, Nantong, 226001, China.
| | - Huijun Liu
- Department of Pharmacy, Yancheng First Hopital, the Fourth Affiliated Hospital of Nantong University, #66 Renmin South Road, Yancheng 224008, Jiangsu, China
| | - Ying Ye
- Department of Ultrasound, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001, Jiangsu, China
| | - Dongjian Chen
- Invasive Technology Department, Affiliated Hospital 2 of Nantong University, First People's Hospital of Nantong City, No. 6 Haierxiang North Road, Nantong, 226001, China
| | - Qun Lu
- Department of Pharmacy, Nantong Third Hospital Affiliated to Nantong University, #60 Middle Qingnian Road, Nantong 226006, Jiangsu, China
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China.
| |
Collapse
|
17
|
Lu J, Huang C, Lu Q, Lu X. Therapeutic and Prophylactic Effects of Amphotericin B Liposomes on Chronic Social Defeat Stress-Induced Behavioral Abnormalities in Mice. Front Pharmacol 2022; 13:918177. [PMID: 35910388 PMCID: PMC9335357 DOI: 10.3389/fphar.2022.918177] [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: 04/12/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Recently, innate immune system stimulants, such as lipopolysaccharide (LPS) and macrophage-colony stimulating factor (M-CSF), were reported to prevent and reverse chronic stress-induced behavioral abnormalities, suggesting that innate immune stimulation could be a potential strategy for the treatment and prevention of mental disorders. Amphotericin B liposome is a clinically available antifungal medication that can stimulate macrophages and microglia. We hypothesize that amphotericin B liposome may be used to prevent and reverse behavioral abnormalities triggered by chronic stress. As expected, our results showed that a single injection of amphotericin B liposome (1 mg/kg) immediately after stress cessation reversed the decrease in time spent in the interaction zone in the social interaction test (SIT) and the increase in immobility time in the tail suspension test (TST) and forced swimming test (FST) in mice caused by chronic social defeat stress (CSDS). In addition, a single injection of amphotericin B liposomes (1 mg/kg) 1 day before stress exposure was found to prevent the CSDS-induced decrease in time spent in the interaction zone in the SIT and the increase in immobility time in the TST and FST in mice. Pretreatment with minocycline to inhibit the innate immune response was able to abolish the reversal effect of post-stress injection of amphotericin B liposomes on CSDS-induced behavioral abnormalities and the prophylactic effect of pre-stress injection of amphotericin B liposomes on CSDS-induced behavioral abnormalities. These results demonstrate that amphotericin B liposomes have both therapeutic and prophylactic effects on chronic stress-induced behavioral abnormalities in mice by mobilizing the innate immune response.
Collapse
Affiliation(s)
- Jiashu Lu
- Department of Pharmacy, The People’s Hospital of Taizhou, The Fifth Affiliated Hospital of Nantong University, Taizhou, China
- *Correspondence: Jiashu Lu,
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
| | - Qun Lu
- Department of Pharmacy, Nantong Third Hospital Affiliated to Nantong University, Nantong, China
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
| |
Collapse
|
18
|
Yang QY, Li XW, Yang R, Qin TY, Long H, Zhang SB, Zhang F. Effects of intraperitoneal injection of lipopolysaccharide-induced peripheral inflammation on dopamine neuron damage in rat midbrain. CNS Neurosci Ther 2022; 28:1624-1636. [PMID: 35789066 PMCID: PMC9437226 DOI: 10.1111/cns.13906] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/24/2022] [Accepted: 06/04/2022] [Indexed: 11/26/2022] Open
Abstract
Introduction Current studies have documented neuroinflammation is implicated in Parkinson's disease. Recently, growing evidence indicated peripheral inflammation plays an important role in regulation of neuroinflammation and thus conferring protection against dopamine (DA) neuronal damage. However, the underlying mechanisms are not clearly illuminated. Methods The effects of intraperitoneal injection of LPS (LPS[i.p.])‐induced peripheral inflammation on substantia nigra (SN) injection of LPS (LPS[SN])‐elicited DA neuronal damage in rat midbrain were investigated. Rats were intraperitoneally injected with LPS (0.5 mg/kg) daily for 4 consecutive days and then given single injection of LPS (8 μg) into SN with an interval of 0 (LPS(i.p.) 0 day ± LPS(SN)), 30 (LPS(i.p.) 30 days ± LPS(SN)), and 90 (LPS(i.p.) 90 days ± LPS(SN)) days after LPS(i.p.) administration. Results LPS(i.p.) increased the levels of inflammatory factors in peripheral blood in (LPS(i.p.) 0 day ± LPS(SN)). Importantly, in (LPS(i.p.) 0 day ± LPS(SN)) and (LPS(i.p.) 30 days ± LPS(SN)), LPS(i.p.) attenuated LPS(SN)‐induced DA neuronal loss in SN. Besides, LPS(i.p.) reduced LPS(SN)‐induced microglia and astrocytes activation in SN. Furtherly, LPS(i.p.) reduced pro‐inflammatory M1 microglia markers mRNA levels and increased anti‐inflammatory M2 microglia markers mRNA levels. In addition, the increased T‐cell marker expression and the decreased M1 microglia marker expression and more DA neuronal survival were discerned at the same area of rat midbrain in LPS(SN)‐induced DA neuronal damage 30 days after LPS(i.p.) application. Conclusion This study suggested LPS(i.p.)‐induced peripheral inflammation might cause T cells to infiltrate the brain to regulate microglia‐mediated neuroinflammation, thereby protecting DA neurons.
Collapse
Affiliation(s)
- Qiu-Yu Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Center, Zunyi Medical University, Zunyi, China
| | - Xian-Wei Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Center, Zunyi Medical University, Zunyi, China
| | - Rong Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Center, Zunyi Medical University, Zunyi, China
| | - Ting-Yang Qin
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Center, Zunyi Medical University, Zunyi, China
| | - Hong Long
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Center, Zunyi Medical University, Zunyi, China
| | - Shi-Bin Zhang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Center, Zunyi Medical University, Zunyi, China
| | - Feng Zhang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Center, Zunyi Medical University, Zunyi, China
| |
Collapse
|
19
|
Jiang T, He Y. Recent Advances in the Role of Nuclear Factor Erythroid-2-Related Factor 2 in Spinal Cord Injury: Regulatory Mechanisms and Therapeutic Options. Front Aging Neurosci 2022; 14:851257. [PMID: 35754957 PMCID: PMC9226435 DOI: 10.3389/fnagi.2022.851257] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 05/09/2022] [Indexed: 01/09/2023] Open
Abstract
Nuclear factor erythroid-2-related factor 2 (Nrf2) is a pleiotropic transcription factor, and it has been documented that it can induce defense mechanisms both oxidative stress and inflammatory injury. At present, more and more evidences show that the Nrf2 signaling pathway is a key pharmacological target for the treatment of spinal cord injury (SCI), and activating the Nrf2 signaling pathway can effectively treat the inflammatory injury and oxidative stress after SCI. This article firstly introduces the biological studies of the Nrf2 pathway. Meanwhile, it is more powerful to explain that activating the Nrf2 signaling pathway can effectively treat SCI by deeply exploring the relationship between Nrf2 and oxidative stress, inflammatory injury, and SCI. In addition, several potential drugs for the treatment of SCI by promoting Nrf2 activation and Nrf2-dependent gene expression are reviewed. And some other treatment strategies of SCI by modulating the Nrf2 pathway are also summarized. It will provide new ideas and directions for the treatment of SCI.
Collapse
Affiliation(s)
- Tianqi Jiang
- Graduate School of Inner Mongolia Medical University, Hohhot, China,Spine Surgery, Inner Mongolia People’s Hospital, Hohhot, China
| | - Yongxiong He
- Spine Surgery, Inner Mongolia People’s Hospital, Hohhot, China,*Correspondence: Yongxiong He,
| |
Collapse
|
20
|
Zhang X, Kracht L, Lerario AM, Dubbelaar ML, Brouwer N, Wesseling EM, Boddeke EWGM, Eggen BJL, Kooistra SM. Epigenetic regulation of innate immune memory in microglia. J Neuroinflammation 2022; 19:111. [PMID: 35568856 PMCID: PMC9107649 DOI: 10.1186/s12974-022-02463-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 04/16/2022] [Indexed: 12/13/2022] Open
Abstract
Background Microglia are the tissue-resident macrophages of the CNS. They originate in the yolk sac, colonize the CNS during embryonic development and form a self-sustaining population with limited turnover. A consequence of their relative slow turnover is that microglia can serve as a long-term memory for inflammatory or neurodegenerative events. Methods Using ATAC-, ChIP- and RNA-sequencing, we characterized the epigenomes and transcriptomes of FACS-purified microglia from mice exposed to different stimuli. A repeated endotoxin challenge (LPS) was used to induce tolerance in microglia, while genotoxic stress (DNA repair deficiency-induced accelerated aging through Ercc1 deficiency) resulted in primed (hypersensitive) microglia. Results Whereas the enrichment of permissive epigenetic marks at enhancer regions could explain training (hyper-responsiveness) of primed microglia to an LPS challenge, the tolerized response of microglia seems to be regulated by loss of permissive epigenetic marks. We identify that inflammatory stimuli and accelerated aging as a result of genotoxic stress activate distinct gene networks. These gene networks and associated biological processes are partially overlapping, which is likely driven by specific transcription factor networks, resulting in altered epigenetic signatures and distinct functional (desensitized vs. primed) microglia phenotypes. Conclusion This study provides insight into epigenetic profiles and transcription factor networks associated with transcriptional signatures of tolerized and trained microglia in vivo, leading to a better understanding of innate immune memory of microglia. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02463-5.
Collapse
Affiliation(s)
- Xiaoming Zhang
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, Hpc-FB43, 9713 AV, Groningen, The Netherlands
| | - Laura Kracht
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, Hpc-FB43, 9713 AV, Groningen, The Netherlands
| | - Antonio M Lerario
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Marissa L Dubbelaar
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, Hpc-FB43, 9713 AV, Groningen, The Netherlands
| | - Nieske Brouwer
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, Hpc-FB43, 9713 AV, Groningen, The Netherlands
| | - Evelyn M Wesseling
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, Hpc-FB43, 9713 AV, Groningen, The Netherlands
| | - Erik W G M Boddeke
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, Hpc-FB43, 9713 AV, Groningen, The Netherlands.,Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Bart J L Eggen
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, Hpc-FB43, 9713 AV, Groningen, The Netherlands.
| | - Susanne M Kooistra
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, Hpc-FB43, 9713 AV, Groningen, The Netherlands.
| |
Collapse
|
21
|
Vonder Haar C, Wampler SK, Bhatia HS, Ozga JE, Toegel C, Lake AD, Iames CW, Cabral CE, Martens KM. Repeat Closed-Head Injury in Male Rats Impairs Attention but Causes Heterogeneous Outcomes in Multiple Measures of Impulsivity and Glial Pathology. Front Behav Neurosci 2022; 16:809249. [PMID: 35359588 PMCID: PMC8963781 DOI: 10.3389/fnbeh.2022.809249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/16/2022] [Indexed: 01/31/2023] Open
Abstract
Repetitive mild traumatic brain injury, or concussion, can lead to the development of long-term psychiatric impairments. However, modeling these deficits is challenging in animal models and necessitates sophisticated behavioral approaches. The current set of studies were designed to evaluate whether a rubberized versus metal impact tip would cause functional deficits, the number of injuries required to generate such deficits, and whether different psychiatric domains would be affected. Across two studies, male rats were trained in either the 5-choice serial reaction time task (5CSRT; Experiment 1) to assess attention and motor impulsivity or concurrently on the 5CSRT and the delay discounting task (Experiment 2) to also assess choice impulsivity. After behavior was stable, brain injuries were delivered with the Closed-head Injury Model of Engineered Rotational Acceleration (CHIMERA) either once per week or twice per week (Experiment 1) or just once per week (Experiment 2). Astrocyte and microglia pathology was also assayed in relevant regions of interest. CHIMERA injury caused attentional deficits across both experiments, but only increased motor impulsivity in Experiment 1. Surprisingly, choice impulsivity was actually reduced on the Delay Discounting Task after repeat injuries. However, subsequent analyses suggested potential visual issues which could alter interpretation of these and attentional data. Subtle changes in glial pathology immediately after the injury (Experiment 1) were attenuated after 4 weeks recovery (Experiment 2). Given the heterogenous findings between experiments, additional research is needed to determine the root causes of psychiatric disturbances which may arise as a results of repeated brain injuries.
Collapse
Affiliation(s)
- Cole Vonder Haar
- Injury and Recovery Laboratory, Department of Neuroscience, Ohio State University, Columbus, OH, United States
- Injury and Recovery Laboratory, Department of Psychology, West Virginia University, Morgantown, WV, United States
| | - Sarah K. Wampler
- Injury and Recovery Laboratory, Department of Neuroscience, Ohio State University, Columbus, OH, United States
- Injury and Recovery Laboratory, Department of Psychology, West Virginia University, Morgantown, WV, United States
| | - Henna S. Bhatia
- Injury and Recovery Laboratory, Department of Psychology, West Virginia University, Morgantown, WV, United States
| | - Jenny E. Ozga
- Injury and Recovery Laboratory, Department of Psychology, West Virginia University, Morgantown, WV, United States
| | - Cory Toegel
- Injury and Recovery Laboratory, Department of Psychology, West Virginia University, Morgantown, WV, United States
| | - Anastasios D. Lake
- Injury and Recovery Laboratory, Department of Psychology, West Virginia University, Morgantown, WV, United States
| | - Christopher W. Iames
- Injury and Recovery Laboratory, Department of Psychology, West Virginia University, Morgantown, WV, United States
| | - Caitlyn E. Cabral
- Injury and Recovery Laboratory, Department of Psychology, West Virginia University, Morgantown, WV, United States
| | - Kris M. Martens
- Injury and Recovery Laboratory, Department of Neuroscience, Ohio State University, Columbus, OH, United States
- Injury and Recovery Laboratory, Department of Psychology, West Virginia University, Morgantown, WV, United States
- *Correspondence: Kris M. Martens,
| |
Collapse
|
22
|
Yao L, Li P, Chen Q, Hu A, Wu Y, Li B. Protective effects of endotoxin tolerance on peripheral lipopolysaccharide-induced neuroinflammation and dopaminergic neuronal injury. Immunopharmacol Immunotoxicol 2022; 44:326-337. [PMID: 35260024 DOI: 10.1080/08923973.2022.2043900] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
CONTEXT Parkinson's disease is a common chronic neurodegenerative disease characterized by massive loss of dopaminergic neurons in the substantia nigra. Neuroinflammation has been shown to play an important role in the pathogenesis of neurodegenerative diseases such as Parkinson's disease. The role of immune tolerance in neuroinflammation and neurodegenerative diseases induced by peripheral factors is unclear. OBJECTIVE This study established a model of endotoxin tolerance to explore the protective effect of endotoxin tolerance on Parkinson-like changes induced by repeated peripheral injections of high-dose LPS, and to explore its inflammatory mechanism. MATERIALS AND METHODS In this study, mice were injected intraperitoneally with low dose (0.5 mg/kg) LPS for 4 days to induce endotoxin tolerance (ET). Then, high-dose (1 mg/kg) LPS was injected continuously intraperitoneally for 4 days to induce Parkinson-like changes. Cytokines were detected by enzyme-linked immunosorbent assay (ELISA) and quantitative real-time polymerase chain reaction (qRT-PCR). Activation of microglial cells was detected by protein expression of CD68 and ionized calcium binding adapter molecule 1(Iba-1) by Western blotting and immunofluorescence. Hematoxylin and eosin staining and expression of tyrosine hydroxylase (TH) and dopamine (DA) were used to assess dopaminergic neuronal injury. The open field test and muscle tension test were used to assess behavioral disorders. RESULTS As expected, compared with non-ET animals, ET preconditioning significantly reduced the production of inflammatory cytokines in the substantia nigra, inhibited microglial activation, and alleviated the pathological changes of dopaminergic neurons. CONCLUSIONS ET may be a promising intervention method for neurodegenerative diseases.HighlightsET was successfully induced by continuous low-dose intraperitoneal LPS injection in mice.ET pretreatment inhibited neuroinflammation in the SN induced by continuous peripheral high doses of LPS.ET pretreatment inhibited continuous peripheral high-dose LPS injection-induced microglial activation in the SN.ET pretreatment decreased LPS-induced functional impairment of dopaminergic neurons.ET reversed the morphological changes of dopaminergic neurons induced by peripheral high-dose LPS.ET pretreatment improved continuous peripheral high-dose LPS injection-induced behavioral impairment.
Collapse
Affiliation(s)
- Liyan Yao
- Department of Toxicology, School of Public Health, Harbin Medical University, Harbin, People's Republic of China.,Department of Toxicology, School of Public Health, Mudanjiang Medical University, Mudanjiang, People's Republic of China
| | - Peng Li
- Department of Toxicology, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Qilei Chen
- Department of Toxicology, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Anxue Hu
- Department of Toxicology, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Yanping Wu
- Department of Toxicology, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Baixiang Li
- Department of Toxicology, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| |
Collapse
|
23
|
Li F, Lu X, Ma Y, Gu Y, Ye T, Huang C. Monophosphoryl Lipid A Tolerance Against Chronic Stress-Induced Depression-Like Behaviors in Mice. Int J Neuropsychopharmacol 2022; 25:399-411. [PMID: 35015863 PMCID: PMC9154281 DOI: 10.1093/ijnp/pyab097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/29/2021] [Accepted: 01/06/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUNDS Our recent studies reported that a single injection with lipopolysaccharide (LPS) before stress exposure prevents depression-like behaviors in stressed mice. Monophosphoryl lipid A (MPL) is a derivative of LPS that lacks the undesirable properties of LPS. We hypothesize that MPL can exert a prophylactic effect on depression. METHODS The experimental mice were pre-injected with MPL before stress exposure. Depression in mice was induced through chronic social defeat stress (CSDS). Behavioral tests were conducted to identify depression-like behaviors. Real-time polymerase chain reaction and biochemical assays were performed to examine the gene and protein expression levels of pro-inflammatory cytokines. RESULTS A single MPL injection 1 day before stress exposure at the dosages of 400, 800, and 1600 μg/kg but not 200 μg/kg prevented CSDS-induced depression-like behaviors in mice. This effect of MPL, however, vanished with the extension of the interval time between drug injection and stress exposure from 1 day or 5 days to 10 days, which was rescued by a second MPL injection 10 days after the first MPL injection or by a 4× MPL injection 10 days before stress exposure. A single MPL injection (800 μg/kg) before stress exposure prevented CSDS-induced increases in the gene expression levels of pro-inflammatory cytokines in the hippocampus and prefrontal cortex. Pre-inhibiting the innate immune stimulation by minocycline pretreatment (40 mg/kg) abrogated the preventive effect of MPL on CSDS-induced depression-like behaviors and neuroinflammatory responses in animal brains. CONCLUSIONS MPL, through innate immune stimulation, prevents stress-induced depression-like behaviors in mice by preventing neuroinflammatory responses.
Collapse
Affiliation(s)
| | | | | | | | - Ting Ye
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Chao Huang
- Correspondence: Chao Huang, Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu Province, China ()
| |
Collapse
|
24
|
Lu Q, Xiang H, Zhu H, Chen Y, Lu X, Huang C. Intranasal lipopolysaccharide administration prevents chronic stress-induced depression- and anxiety-like behaviors in mice. Neuropharmacology 2021; 200:108816. [PMID: 34599975 DOI: 10.1016/j.neuropharm.2021.108816] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/24/2021] [Accepted: 09/26/2021] [Indexed: 02/06/2023]
Abstract
We recently reported that intraperitoneal injection of a low dose of lipopolysaccharide (LPS) prevents chronic stress-induced depression-like behaviors in mice. In this study, we reported that a single intranasal LPS administration (10 μg/mouse) one day prior to stress exposure produced prophylactic effects on chronic social defeat stress (CSDS)-induced depression-like behaviors, which was indicated by the reduction in social interaction time in the social interaction test and the decrease in immobility time in the tail suspension test and forced swimming test. The single intranasal LPS administration prior to stress exposure was also found to prevent CSDS-induced anxiety-like behaviors, including prevention of CSDS-induced decrease in the time spent in open arms in the elevated plus maze test, decrease in the time spent in lit side in the light-dark test, and decrease in the time spent in central regions in the open field test, along with no changes in locomotor activity. Further analysis showed that the single intranasal LPS administration one day prior to stress exposure prevented CSDS-induced increase in levels of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and IL-1β mRNA in the hippocampus and prefrontal cortex. Inhibition of innate immune stimulation by minocycline pretreatment not only abrogated the preventive effect of intranasal LPS administration on CSDS-induced depression- and anxiety-like behaviors, but also abrogated the preventive effect of intranasal LPS administration on CSDS-induced neuroinflammatory responses in the hippocampus and prefrontal cortex. These results demonstrate that intranasal administration of innate immune stimulants could be a potential approach for the prevention of depression and anxiety.
Collapse
Affiliation(s)
- Qun Lu
- Department of Pharmacy, Nantong Third Hospital Affiliated to Nantong University, #60 Middle Qingnian Road, Nantong, 226006, Jiangsu, China
| | - Haitao Xiang
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, #118 Wansheng Street, Suzhou, 215028, Jiangsu, China
| | - Haojie Zhu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Yifan Chen
- Department of Medical Imaging, School of Medicine, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu, China.
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
25
|
Mizobuchi H, Soma GI. Low-dose lipopolysaccharide as an immune regulator for homeostasis maintenance in the central nervous system through transformation to neuroprotective microglia. Neural Regen Res 2021; 16:1928-1934. [PMID: 33642362 PMCID: PMC8343302 DOI: 10.4103/1673-5374.308067] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/26/2020] [Accepted: 12/16/2020] [Indexed: 12/25/2022] Open
Abstract
Microglia, which are tissue-resident macrophages in the brain, play a central role in the brain innate immunity and contribute to the maintenance of brain homeostasis. Lipopolysaccharide is a component of the outer membrane of gram-negative bacteria, and activates immune cells including microglia via Toll-like receptor 4 signaling. Lipopolysaccharide is generally known as an endotoxin, as administration of high-dose lipopolysaccharide induces potent systemic inflammation. Also, it has long been recognized that lipopolysaccharide exacerbates neuroinflammation. In contrast, our study revealed that oral administration of lipopolysaccharide ameliorates Alzheimer's disease pathology and suggested that neuroprotective microglia are involved in this phenomenon. Additionally, other recent studies have accumulated evidence demonstrating that controlled immune training with low-dose lipopolysaccharide prevents neuronal damage by transforming the microglia into a neuroprotective phenotype. Therefore, lipopolysaccharide may not a mere inflammatory inducer, but an immunomodulator that can lead to neuroprotective effects in the brain. In this review, we summarized current studies regarding neuroprotective microglia transformed by immune training with lipopolysaccharide. We state that microglia transformed by lipopolysaccharide preconditioning cannot simply be characterized by their general suppression of proinflammatory mediators and general promotion of anti-inflammatory mediators, but instead must be described by their complex profile comprising various molecules related to inflammatory regulation, phagocytosis, neuroprotection, anti-apoptosis, and antioxidation. In addition, microglial transformation seems to depend on the dose of lipopolysaccharide used during immune training. Immune training of neuroprotective microglia using low-dose lipopolysaccharide, especially through oral lipopolysaccharide administration, may represent an innovative prevention or treatment for neurological diseases; however more vigorous studies are still required to properly modulate these treatments.
Collapse
Affiliation(s)
- Haruka Mizobuchi
- Control of Innate Immunity, Technology Research Association, Kagawa, Japan
| | - Gen-Ichiro Soma
- Control of Innate Immunity, Technology Research Association, Kagawa, Japan
- Macrophi Inc., Kagawa, Japan
- Research Institute for Healthy Living, Niigata University of Pharmacy and Applied Life Sciences, Niigata, Japan
| |
Collapse
|
26
|
Li WC, Yao SP, Zhang J, Liu WB, Liu J, Geng CK. Low-dose lipopolysaccharide protects nerve cells against spinal cord injury via regulating the PI3K-AKT-Nrf2 signaling pathway. Biochem Cell Biol 2021; 99:527-535. [PMID: 34424795 DOI: 10.1139/bcb-2020-0641] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This study explored the molecular mechanism behind the protective effects from low-dose lipopolysaccharide (LPS) on an in-vitro model of spinal cord injury (SCI). For this, PC12 cells were treated with different concentrations of LPS and the cell counting kit-8 assay was used to measure the toxicity of LPS to the cells. Next, we used immunofluorescence to measure nuclear translocation of Nrf2 in PC12 cells. PC12 cells were then treated with IGF-1 (PI3K agonist) and LY294002 (PI3K inhibitor). An in-vitro model of SCI was then established via oxygen-glucose deprivation/reoxygenation. Rates of apoptosis were measured using flow cytometry and the TUNEL assay. Low-dose LPS increased the expression levels of Nrf2, p-PI3K/PI3K, and p-AKT/AKT, and facilitated nuclear translocation of Nrf2. The activation of PI3K-AKT signaling by IGF-1 significantly increased the expression of Nrf2, whereas inhibition of PI3K-AKT signaling significantly decreased the expression of Nrf2. Low-dose LPS reduced the apoptotic ratio of PC12 cells, decreased the expression levels of caspase 3 and caspase 9, and increased the expression levels of HO-1, NQO1, and γ-GCS. Low-dose LPS also reduced the rate of apoptosis and oxidative stress by activating the PI3K-AKT-Nrf2 signaling pathway. Collectively, the results indicate that PI3K-AKT-Nrf2 signaling participates in the protective effects from low-dose LPS in an in-vitro PC12 cell model of SCI.
Collapse
Affiliation(s)
- Wei-Chao Li
- Department of Orthopedic Surgery, The First People's Hospital of Yunnan province, Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China.,Faculty of Medical Science, Kunming University of Science and Technology, Kunming 650500, China
| | - Shao-Ping Yao
- Department of Orthopedic Surgery, The First People's Hospital of Yunnan province, Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| | - Jun Zhang
- Department of Orthopedic Surgery, The First People's Hospital of Yunnan province, Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| | - Wei-Bing Liu
- Department of Orthopedic Surgery, The First People's Hospital of Yunnan province, Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| | - Jie Liu
- Department of Orthopedic Surgery, The First People's Hospital of Yunnan province, Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China.,Yunnan Key Laboratory of Digital Orthopaedics, The First People's Hospital of Yunnan province, Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| | - Cheng-Kui Geng
- Department of orthopedics, Yan'an Hospital, Kunming Medical University, Kunming 650035, China
| |
Collapse
|
27
|
McDonald SJ, Sharkey JM, Sun M, Kaukas LM, Shultz SR, Turner RJ, Leonard AV, Brady RD, Corrigan F. Beyond the Brain: Peripheral Interactions after Traumatic Brain Injury. J Neurotrauma 2021; 37:770-781. [PMID: 32041478 DOI: 10.1089/neu.2019.6885] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability, and there are currently no pharmacological treatments known to improve patient outcomes. Unquestionably, contributing toward a lack of effective treatments is the highly complex and heterogenous nature of TBI. In this review, we highlight the recent surge of research that has demonstrated various central interactions with the periphery as a potential major contributor toward this heterogeneity and, in particular, the breadth of research from Australia. We describe the growing evidence of how extracranial factors, such as polytrauma and infection, can significantly alter TBI neuropathology. In addition, we highlight how dysregulation of the autonomic nervous system and the systemic inflammatory response induced by TBI can have profound pathophysiological effects on peripheral organs, such as the heart, lung, gastrointestinal tract, liver, kidney, spleen, and bone. Collectively, this review firmly establishes TBI as a systemic condition. Further, the central and peripheral interactions that can occur after TBI must be further explored and accounted for in the ongoing search for effective treatments.
Collapse
Affiliation(s)
- Stuart J McDonald
- Department Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Jessica M Sharkey
- Discipline of Anatomy and Pathology, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Mujun Sun
- Department Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Lola M Kaukas
- School of Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Sandy R Shultz
- Department Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Renee J Turner
- Discipline of Anatomy and Pathology, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Anna V Leonard
- Discipline of Anatomy and Pathology, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Rhys D Brady
- Department Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Frances Corrigan
- School of Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
28
|
Sheng R, Chen JL, Qin ZH. Cerebral conditioning: Mechanisms and potential clinical implications. BRAIN HEMORRHAGES 2021. [DOI: 10.1016/j.hest.2021.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
29
|
Zhang R, Wang J, Huang L, Wang TJ, Huang Y, Li Z, He J, Sun C, Wang J, Chen X, Wang J. The pros and cons of motor, memory, and emotion-related behavioral tests in the mouse traumatic brain injury model. Neurol Res 2021; 44:65-89. [PMID: 34308784 DOI: 10.1080/01616412.2021.1956290] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Traumatic brain injury (TBI) is a medical emergency with high morbidity and mortality. Motor, memory, and emotion-related deficits are common symptoms following TBI, yet treatment is very limited. To develop new drugs and find new therapeutic avenues, a wide variety of TBI models have been established to mimic the heterogeneity of TBI. In this regard, along with histologic measures, behavioral functional outcomes provide valuable insight into the underlying neuropathology and guide neurorehabilitation efforts for neuropsychiatric impairment after TBI. Development, characterization, and application of behavioral tests that can assess functional neurologic deficits are essential to the development of translational therapies. This comprehensive review aims to summarize 19 common behavioral tests from three aspects (motor, memory, and emotion-related) that are associated with TBI pathology. Discussion covers the apparatus, the test steps, the evaluation indexes, data collection and analysis, animal performance and applications, advantages and disadvantages as well as precautions to eliminate bias wherever possible. We discussed recent studies on TBI-related preconditioning, biomarkers, and optimized behavioral protocols. The neuropsychologic tests employed in clinics were correlated with those used in mouse TBI models. In summary, this review provides a comprehensive, up-to-date reference for TBI researchers to choose the right neurobehavioral protocol according to the research objectives of their translational investigation.
Collapse
Affiliation(s)
- Ruoyu Zhang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Junming Wang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Leo Huang
- Department of Psychology, University of Toronto, Toronto, Canada
| | - Tom J Wang
- Winston Churchill High School, Potomac, Maryland, USA
| | - Yinrou Huang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zefu Li
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jinxin He
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Chen Sun
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jing Wang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xuemei Chen
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jian Wang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
30
|
Ji J, Xiang H, Lu X, Tan P, Yang R, Ye T, Chen Z, Chen D, He H, Chen J, Ma Y, Huang C. A prophylactic effect of macrophage-colony stimulating factor on chronic stress-induced depression-like behaviors in mice. Neuropharmacology 2021; 193:108621. [PMID: 34062163 DOI: 10.1016/j.neuropharm.2021.108621] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 04/22/2021] [Accepted: 05/17/2021] [Indexed: 11/18/2022]
Abstract
Innate immune activation has been shown to reduce the severity of nervous system disorders such as brain ischemia and traumatic brain damage. Macrophage-colony stimulating factor (M-CSF), a drug that is used to treat hematological system disease, is an enhancer of the innate immune response. In the present study, we evaluated the effect of M-CSF preconditioning on chronic social defeat stress (CSDS)-induced depression-like behaviors in mice. Results showed that a single M-CSF injection 1 day before stress exposure at the dose of 100 and 500 μg/kg, or a single M-CSF injection (100 μg/kg) 1 or 5 days but not 10 days before stress exposure prevented CSDS-induced depression-like behaviors in mice. Further analysis showed that a second M-CSF injection 10 days after the first M-CSF injection and a 2 × or 4 × M-CSF injections 10 days before stress exposure also prevented CSDS-induced depression-like behaviors. Molecular studies revealed that a single M-CSF injection prior to stress exposure skewed the neuroinflammatory responses in the brain in CSDS-exposed mice towards an anti-inflammatory phenotype. These behavioral and molecular actions of M-CSF were correlated with innate immune stimulation, as pre-inhibiting the innate immune activation by minocycline pretreatment (40 mg/kg) abrogated the preventive effect of M-CSF on CSDS-induced depression-like behaviors and neuroinflammatory responses. These results provide evidence to show that innate immune activation by M-CSF pretreatment may prevent chronic stress-induced depression-like behaviors via preventing the development of neuroinflammatory response in the brain, which may help to develop novel strategies for the prevention of depression.
Collapse
Affiliation(s)
- Jianlin Ji
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Haitao Xiang
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, #118 Wansheng Street, Suzhou, 215028, Jiangsu, China
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Pingping Tan
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Rongrong Yang
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Jiangsu Province, #20Xisi Road, Nantong, Jiangsu, 226001, China
| | - Ting Ye
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Zhuo Chen
- Invasive Technology Department, Nantong First People's Hospital, The Second Affiliated Hospital of Nantong University, #6 North Road Hai'er Xiang, Nantong, 226001, Jiangsu, China
| | - Dongjian Chen
- Invasive Technology Department, Nantong First People's Hospital, The Second Affiliated Hospital of Nantong University, #6 North Road Hai'er Xiang, Nantong, 226001, Jiangsu, China
| | - Haiyan He
- Department of Respiratory Medicine, Nantong First People's Hospital, The Second Affiliated Hospital of Nantong University, #6 North Road Hai'er Xiang, Nantong, 226001, Jiangsu, China
| | - Jinliang Chen
- Department of Respiratory Medicine, Nantong First People's Hospital, The Second Affiliated Hospital of Nantong University, #6 North Road Hai'er Xiang, Nantong, 226001, Jiangsu, China
| | - Yaoying Ma
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu, China.
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
31
|
Postolache TT, Wadhawan A, Can A, Lowry CA, Woodbury M, Makkar H, Hoisington AJ, Scott AJ, Potocki E, Benros ME, Stiller JW. Inflammation in Traumatic Brain Injury. J Alzheimers Dis 2021; 74:1-28. [PMID: 32176646 DOI: 10.3233/jad-191150] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
There is an increasing evidence that inflammation contributes to clinical and functional outcomes in traumatic brain injury (TBI). Many successful target-engaging, lesion-reducing, symptom-alleviating, and function-improving interventions in animal models of TBI have failed to show efficacy in clinical trials. Timing and immunological context are paramount for the direction, quality, and intensity of immune responses to TBI and the resulting neuroanatomical, clinical, and functional course. We present components of the immune system implicated in TBI, potential immune targets, and target-engaging interventions. The main objective of our article is to point toward modifiable molecular and cellular mechanisms that may modify the outcomes in TBI, and contribute to increasing the translational value of interventions that have been identified in animal models of TBI.
Collapse
Affiliation(s)
- Teodor T Postolache
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Aurora, CO, USA.,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO, USA.,Mental Illness Research, Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 5, VA Capitol Health Care Network, Baltimore, MD, USA
| | - Abhishek Wadhawan
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,Saint Elizabeths Hospital, Department of Psychiatry, Washington, DC, USA
| | - Adem Can
- School of Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Christopher A Lowry
- Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Aurora, CO, USA.,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO, USA.,Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA.,Department of Physical Medicine and Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Margaret Woodbury
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,VA Maryland Healthcare System, Baltimore VA Medical Center, Baltimore, MD, USA
| | - Hina Makkar
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Andrew J Hoisington
- Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Aurora, CO, USA.,Systems Engineering and Management, Air Force Institute of Technology, Wright-Patterson AFB, OH, USA
| | - Alison J Scott
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD, USA
| | - Eileen Potocki
- VA Maryland Healthcare System, Baltimore VA Medical Center, Baltimore, MD, USA
| | - Michael E Benros
- Copenhagen Research Center for Mental Health-CORE, Mental Health Centre Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - John W Stiller
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,Maryland State Athletic Commission, Baltimore, MD, USA.,Saint Elizabeths Hospital, Neurology Consultation Services, Washington, DC, USA
| |
Collapse
|
32
|
High-frequency head impact causes chronic synaptic adaptation and long-term cognitive impairment in mice. Nat Commun 2021; 12:2613. [PMID: 33972519 PMCID: PMC8110563 DOI: 10.1038/s41467-021-22744-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 03/24/2021] [Indexed: 02/03/2023] Open
Abstract
Repeated head impact exposure can cause memory and behavioral impairments. Here, we report that exposure to non-damaging, but high frequency, head impacts can alter brain function in mice through synaptic adaptation. High frequency head impact mice develop chronic cognitive impairments in the absence of traditional brain trauma pathology, and transcriptomic profiling of mouse and human chronic traumatic encephalopathy brain reveal that synapses are strongly affected by head impact. Electrophysiological analysis shows that high frequency head impacts cause chronic modification of the AMPA/NMDA ratio in neurons that underlie the changes to cognition. To demonstrate that synaptic adaptation is caused by head impact-induced glutamate release, we pretreated mice with memantine prior to head impact. Memantine prevents the development of the key transcriptomic and electrophysiological signatures of high frequency head impact, and averts cognitive dysfunction. These data reveal synapses as a target of high frequency head impact in human and mouse brain, and that this physiological adaptation in response to head impact is sufficient to induce chronic cognitive impairment in mice.
Collapse
|
33
|
Hodoodi F, Allah-Tavakoli M, Tajik F, Fatemi I, Moghadam Ahmadi A. The effect of head cooling and remote ischemic conditioning on patients with traumatic brain injury. iScience 2021; 24:102472. [PMID: 34169235 PMCID: PMC8207229 DOI: 10.1016/j.isci.2021.102472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/12/2020] [Accepted: 04/22/2021] [Indexed: 11/17/2022] Open
Abstract
Cerebral impairment caused by an external force to the head is known as traumatic brain injury (TBI). The aim of this study was to determine the role of local hypothermia and remote ischemic conditioning (RIC) on oxidative stress, inflammatory response after TBI, and other involved variables. The present study is a clinical trial on 84 patients with TBI who were divided into 4 groups. The head cooling for 1.5 to 6 hr was performed in the first three days after TBI. RIC intervention was performed within the golden time after TBI in the form of four 5-min cycles with full cuff and 5 min of emptying of cuff. The group receiving the head cooling technique recovered better than the group receiving the RIC technique. Generally, combination of the two interventions of head cooling and RIC techniques is more effective on the improvement of clinical status of patients than each separate technique. The effect of the head cooling method in controlling secondary injury in patients with TBI. The effect of the RIC method in controlling secondary injury in patients with TBI. Comparison of two interventions of head cooling and RIC. Evaluation of clinical and paraclinical parameters.
Collapse
Affiliation(s)
- Fardin Hodoodi
- Department of Physiology and Pharmacology, Schoole of Medicine, Rafsanjan University of Medical Science, Rafsanjan, Iran
| | - Mohammad Allah-Tavakoli
- Department of Physiology and Pharmacology, Schoole of Medicine, Rafsanjan University of Medical Science, Rafsanjan, Iran
- Physiology-pharmacology Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Farzad Tajik
- Department of Clinical Research Sciences, Department of Medicine, Rafsanjan University of Medical Science, Rafsanjan, Iran
- Department of Neurology, Department of Medicine, Rafsanjan University of Medical Science, Rafsanjan, Iran
| | - Iman Fatemi
- Research Center of Tropical and Infectious Diseases, Kerman University of Medical Sciences, Kerman, Iran
| | - Amir Moghadam Ahmadi
- Department of Neurology, Department of Medicine, Rafsanjan University of Medical Science, Rafsanjan, Iran
- Non-Communicable Diseases Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Corresponding author
| |
Collapse
|
34
|
Gao P, Tang S, Chen H, Zhou X, Ou Y, Shen R, He Y. Preconditioning increases brain resistance against acute brain injury via neuroinflammation modulation. Exp Neurol 2021; 341:113712. [PMID: 33819449 DOI: 10.1016/j.expneurol.2021.113712] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/16/2021] [Accepted: 03/26/2021] [Indexed: 01/10/2023]
Abstract
Acute brain injury (ABI) is a broad concept mainly comprised of sudden parenchymal brain injury. Acute brain injury outcomes are dependent not only on the severity of the primary injury, but the delayed secondary injury that subsequently follows as well. These are both taken into consideration when determining the patient's prognosis. Growing clinical and experimental evidence demonstrates that "preconditioning," a prophylactic approach in which the brain is exposed to various pre-injury stressors, can induce varying degrees of "tolerance" against the impact of the ABI by modulating neuroinflammation. In this review, we will summarize the pathophysiology of ABI, and discuss the involved mechanisms of neuroinflammation in ABI, as well as existing experimental and clinical studies demonstrating the efficacy of preconditioning methods in various types of ABI by modulating neuroinflammation.
Collapse
Affiliation(s)
- Pan Gao
- Department of Translational Neurodegeneration, German Centre for Neurodegenerative Diseases (DZNE), Munich 81377, Germany.
| | - Sicheng Tang
- Medical Clinic and Polyclinic IV, Ludwig-Maximilians University Munich (LMU), Munich 80336, Germany
| | - Hanmin Chen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Xiangyue Zhou
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Yibo Ou
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Ronghua Shen
- Department of Psychological Rehabilitation, Hankou Hospital, Wuhan, Hubei 430010, PR China.
| | - Yue He
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China.
| |
Collapse
|
35
|
Ebeid MA, Habib MZ, Mohamed AM, Faramawy YE, Saad SST, El-Kharashi OA, El Magdoub HM, Abd-Alkhalek HA, Aboul-Fotouh S, Abdel-Tawab AM. Cognitive effects of the GSK-3 inhibitor "lithium" in LPS/chronic mild stress rat model of depression: Hippocampal and cortical neuroinflammation and tauopathy. Neurotoxicology 2021; 83:77-88. [PMID: 33417987 DOI: 10.1016/j.neuro.2020.12.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/18/2020] [Accepted: 12/30/2020] [Indexed: 11/16/2022]
Abstract
Low-dose repeated lipopolysaccharide pre-challenge followed by chronic mild stress (LPS/CMS) protocol has been introduced as a rodent model of depression combining the roles of immune activation and chronic psychological stress. However, the impact of this paradigm on cognitive functioning has not been investigated hitherto. METHODS This study evaluated LPS/CMS-induced cognitive effects and the role of glycogen synthase kinase-3β (GSK-3β) activation with subsequent neuroinflammation and pathological tau deposition in the pathogenesis of these effects using lithium (Li) as a tool for GSK-3 inhibition. RESULTS LPS pre-challenge reduced CMS-induced neuroinflammation, depressive-like behavior and cognitive inflexibility. It also improved spatial learning but increased GSK-3β expression and exaggerated hyperphosphorylated tau accumulation in hippocampus and prefrontal cortex. Li ameliorated CMS and LPS/CMS-induced depressive and cognitive deficits, reduced GSK-3β over-expression and tau hyperphosphorylation, impeded neuroinflammation and enhanced neuronal survival. CONCLUSION This study draws attention to LPS/CMS-triggered cognitive changes and highlights how prior low-dose immune challenge could develop an adaptive capacity to buffer inflammatory damage and maintain the cognitive abilities necessary to withstand threats. This work also underscores the favorable effect of Li (as a GSK-3β inhibitor) in impeding exaggerated tauopathy and neuroinflammation, rescuing neuronal survival and preserving cognitive functions. Yet, further in-depth studies utilizing different low-dose LPS challenge schedules are needed to elucidate the complex interactions between immune activation and chronic stress exposure.
Collapse
Affiliation(s)
- Mai A Ebeid
- Department of Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mohamed Z Habib
- Department of Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Ahmed M Mohamed
- Department of Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Yasser El Faramawy
- Department of Geriatrics, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sherin S T Saad
- Department of Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Omnyah A El-Kharashi
- Department of Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Hekmat M El Magdoub
- Department of Biochemistry, Faculty of Pharmacy, Misr International University, Cairo, Egypt
| | - Hadwa A Abd-Alkhalek
- Department of Histology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sawsan Aboul-Fotouh
- Department of Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt; Clinical Pharmacology Unit, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ahmed M Abdel-Tawab
- Department of Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt; Clinical Pharmacology Unit, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
36
|
Sangaran PG, Ibrahim ZA, Chik Z, Mohamed Z, Ahmadiani A. LPS Preconditioning Attenuates Apoptosis Mechanism by Inhibiting NF-κB and Caspase-3 Activity: TLR4 Pre-activation in the Signaling Pathway of LPS-Induced Neuroprotection. Mol Neurobiol 2021; 58:2407-2422. [PMID: 33421016 DOI: 10.1007/s12035-020-02227-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022]
Abstract
Neuroinflammation, an inflammatory response within the nervous system, has been shown to be implicated in the progression of various neurodegenerative diseases. Recent in vivo studies showed that lipopolysaccharide (LPS) preconditioning provides neuroprotection by activating Toll-like receptor 4 (TLR4), one of the members for pattern recognition receptor (PRR) family that play critical role in host response to tissue injury, infection, and inflammation. Pre-exposure to low dose of LPS could confer a protective state against cellular apoptosis following subsequent stimulation with LPS at higher concentration, suggesting a role for TLR4 pre-activation in the signaling pathway of LPS-induced neuroprotection. However, the precise molecular mechanism associated with this protective effect is not well understood. In this article, we provide an overall review of the current state of our knowledge about LPS preconditioning in attenuating apoptosis mechanism and conferring neuroprotection via TLR4 signaling pathway.
Collapse
Affiliation(s)
- Pushpa Gandi Sangaran
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Zaridatul Aini Ibrahim
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Zamri Chik
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Zahurin Mohamed
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Abolhassan Ahmadiani
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Evin, PO Box 19839-63113, Tehran, Iran.
| |
Collapse
|
37
|
Fleischmann C, Shohami E, Trembovler V, Heled Y, Horowitz M. Cognitive Effects of Astaxanthin Pretreatment on Recovery From Traumatic Brain Injury. Front Neurol 2020; 11:999. [PMID: 33178093 PMCID: PMC7593578 DOI: 10.3389/fneur.2020.00999] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/29/2020] [Indexed: 01/12/2023] Open
Abstract
Traumatic brain injury (TBI), caused by mechanical impact to the brain, is a leading cause of death and disability among young adults, with slow and often incomplete recovery. Preemptive treatment strategies may increase the injury resilience of high-risk populations such as soldiers and athletes. In this work, the xanthophyll carotenoid Astaxanthin was examined as a potential nutritional preconditioning method in mice (sabra strain) to increase their resilience prior to TBI in a closed head injury (CHI) model. The effect of Astaxanthin pretreatment on heat shock protein (HSP) dynamics and functional outcome after CHI was explored by gavage or free eating (in pellet form) for 2 weeks before CHI. Assessment of neuromotor function by the neurological severity score (NSS) revealed significant improvement in the Astaxanthin gavage-treated group (100 mg/kg, ATX) during recovery compared to the gavage-treated olive oil group (OIL), beginning at 24 h post-CHI and lasting throughout 28 days (p < 0.007). Astaxanthin pretreatment in pellet form produced a smaller improvement in NSS vs. posttreatment at 7 days post-CHI (p < 0.05). Cognitive and behavioral evaluation using the novel object recognition test (ORT) and the Y Maze test revealed an advantage for Astaxanthin administration via free eating vs. standard chow during recovery post-CHI (ORT at 3 days, p < 0.035; improvement in Y Maze score from 2 to 29 days, p < 0.02). HSP profile and anxiety (open field test) were not significantly affected by Astaxanthin. In conclusion, astaxanthin pretreatment may contribute to improved recovery post-TBI in mice and is influenced by the form of administration.
Collapse
Affiliation(s)
- Chen Fleischmann
- The Institute of Military Physiology, IDF Medical Corps, Tel-Hashomer, Israel.,Heller Institute of Medical Research, Sheba Medical Center, Ramat Gan, Israel.,Laboratory of Environmental Physiology, Hebrew University, Jerusalem, Israel
| | - Esther Shohami
- Department of Pharmacology, Institute for Drug Research, Hebrew University, Jerusalem, Israel
| | - Victoria Trembovler
- Department of Pharmacology, Institute for Drug Research, Hebrew University, Jerusalem, Israel
| | - Yuval Heled
- Heller Institute of Medical Research, Sheba Medical Center, Ramat Gan, Israel.,Kibbutzim College, Tel Aviv, Israel
| | - Michal Horowitz
- Laboratory of Environmental Physiology, Hebrew University, Jerusalem, Israel
| |
Collapse
|
38
|
Trained Innate Immunity by Repeated Low-Dose Lipopolysaccharide Injections Displays Long-Term Neuroprotective Effects. Mediators Inflamm 2020; 2020:8191079. [PMID: 33061831 PMCID: PMC7547335 DOI: 10.1155/2020/8191079] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 12/20/2022] Open
Abstract
Disrupted immune response is an important feature of many neurodegenerative conditions, including sepsis-associated cognitive impairment. Accumulating evidence has demonstrated that immune memory occurs in microglia, which has a significant impact on pathological hallmarks of neurological diseases. However, it remains unclear whether immune memory can cause subsequent alterations in the brain immune response and affect neurobehavioral outcomes in sepsis survivors. In the present study, mice received daily intraperitoneal injection of low-dose lipopolysaccharide (LPS, 0.1 mg/kg) for three consecutive days to induce immune memory (immune tolerance) and then were subjected to sham operation or cecal ligation and puncture (CLP) 9 months later, followed by a battery of neurobehavioral and biochemical studies. Here, we showed that repeated low-dose LPS injection-induced immune memory protected mice from sepsis-induced cognitive and affective impairments, which were accompanied by significantly decreased brain proinflammatory cytokines and immune response. In conclusion, our study suggests that modulation of brain immune responses by repeated LPS injections confers neuroprotective effects by preventing overactivated immune response in response to subsequent septic insult.
Collapse
|
39
|
Florentino D, Della Giustina A, de Souza Goldim MP, Danielski LG, de Oliveira Junior AN, Joaquim L, Bonfante S, Biehl E, da Rosa N, Fernandes D, Gava FF, Michels M, Fortunato JJ, Réus GZ, S Valvassori S, Quevedo J, Dal-Pizzol F, Barichello T, Petronilho F. Early life neuroimmune challenge protects the brain after sepsis in adult rats. Neurochem Int 2020; 135:104712. [DOI: 10.1016/j.neuint.2020.104712] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 01/24/2020] [Accepted: 02/27/2020] [Indexed: 12/20/2022]
|
40
|
Eslami M, Alizadeh L, Morteza-Zadeh P, Sayyah M. The effect of Lipopolysaccharide (LPS) pretreatment on hippocampal apoptosis in traumatic rats. Neurol Res 2020; 42:91-98. [PMID: 31914902 DOI: 10.1080/01616412.2019.1709139] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Objectives: Traumatic brain injury (TBI) is a serious medical problem that affects the quality of life. Apoptosis is a form of programmed cell death that happens after trauma. Effector caspases are responsible for initiating apoptosis.Methods: In the present study, we examined the effect of LPS preconditioning (0.1 and 0.5 mg/kg, ip; 5 days prior controlled cortical injury) on apoptosis, 4 and 12 hours after trauma. We investigated possible mechanisms on the expression of caspase3 and caspase7 in hippocampal CA1 and CA3 areas by using immunohistochemistry and Western blotting techniques and also TUNEL-positive cells.Results: Higher expression of caspase3 and caspase7 were accompanied by a higher number of dead neurons in traumatic rats 4 and 12 hours after trauma(P < 0.05). LPS preconditioning decreased caspase3 and caspase7over-expression and the number of dead neurons in the hippocampus(P < 0.05).Discussion: Our data indicate that LPS preconditioning inhibits neural damage and apoptosis induced by trauma in the hippocampus.
Collapse
Affiliation(s)
- Mansoureh Eslami
- Department of Basic Sciences, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Alizadeh
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | | | - Mohammad Sayyah
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
41
|
Loring HS, Thompson PR. Emergence of SARM1 as a Potential Therapeutic Target for Wallerian-type Diseases. Cell Chem Biol 2019; 27:1-13. [PMID: 31761689 DOI: 10.1016/j.chembiol.2019.11.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/24/2019] [Accepted: 11/04/2019] [Indexed: 11/30/2022]
Abstract
Wallerian degeneration is a neuronal death pathway that is triggered in response to injury or disease. Death was thought to occur passively until the discovery of a mouse strain, i.e., Wallerian degeneration slow (WLDS), which was resistant to degeneration. Given that the WLDS mouse encodes a gain-of-function fusion protein, its relevance to human disease was limited. The later discovery that SARM1 (sterile alpha and toll/interleukin receptor [TIR] motif-containing protein 1) promotes Wallerian degeneration suggested the existence of a pathway that might be targeted therapeutically. More recently, SARM1 was found to execute degeneration by hydrolyzing NAD+. Notably, SARM1 knockdown or knockout prevents neuron degeneration in response to a range of insults that lead to peripheral neuropathy, traumatic brain injury, and neurodegenerative disease. Here, we discuss the role of SARM1 in Wallerian degeneration and the opportunities to target this enzyme therapeutically.
Collapse
Affiliation(s)
- Heather S Loring
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, LRB 826, 364 Plantation Street, Worcester, MA 01605, USA; Program in Chemical Biology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | - Paul R Thompson
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, LRB 826, 364 Plantation Street, Worcester, MA 01605, USA; Program in Chemical Biology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA.
| |
Collapse
|
42
|
Katharesan V, Deery S, Johnson IP. Neuroprotective effect of acute prior inflammation with lipopolysaccharide for adult male rat facial motoneurones. Brain Res 2018; 1696:56-62. [DOI: 10.1016/j.brainres.2018.05.039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 05/22/2018] [Accepted: 05/25/2018] [Indexed: 12/14/2022]
|
43
|
Monophosphoryl Lipid A and Pam3Cys Prevent the Increase in Seizure Susceptibility and Epileptogenesis in Rats Undergoing Traumatic Brain Injury. Neurochem Res 2018; 43:1978-1985. [PMID: 30173323 DOI: 10.1007/s11064-018-2619-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/23/2018] [Accepted: 08/18/2018] [Indexed: 12/19/2022]
Abstract
Five percent of all epilepsy cases are attributed to traumatic brain injury (TBI), which are known as post-traumatic epilepsy (PTE). Finding preventive strategies for PTE is valuable. Remarkable feature of TBI is activation of microglia and subsequent neuroinflammation, which provokes epileptogenesis. The toll-like receptor agonists monophosphoryl lipid A (MPL) and tri-palmitoyl-S-glyceryl-cysteine (Pam3Cys) are safe, well-tolerated and effective adjuvants existing in prophylactic human vaccines. We examined the impact of early injection of MPL and Pam3Cys to rats, on the rate of kindled seizures acquisition following TBI. Rats received a single dose (1 µg/rat) of MPL or Pam3Cys through intracerebroventricular injection. 5 days later, trauma was exerted to temporo-parietal cortex of rats by controlled cortical impact device. After 24 h, traumatic rats underwent amygdala kindling. Brain level of the inflammatory cytokine tumor necrosis factor-alpha (TNF-α) was also measured in traumatic rats by immunoblotting. Compared to non-traumatic (sham-operated) rats, traumatic rats showed three times lower seizure threshold (133 ± 5 µA vs. 416.3 ± 16 µA, p < 0.001); about three times less number of stimuli to become kindled (5 ± 1 vs. 14 ± 2, p < 0.01); longer duration of kindled seizure parameters including entire seizure behavior, generalized seizures, and afterdischarges (p < 0.001); and a two times increase in the TNF-α level. MPL and Pam3Cys did not change kindling rate and the seizure parameters in sham-operated rats. The MPL- and Pam3Cys-pretreated traumatic rats displayed seizure threshold, speed of kindling, and duration of kindled seizure parameters, similar to the non-traumatic rats. Pretreatment by MPL and Pam3Cys prevented the increase in TNF-α level by trauma. Given that MPL and Pam3Cys currently have clinical use as well-tolerated vaccines with reliable safety, they have the potential to be used in prevention of PTE.
Collapse
|
44
|
Zhang J, Zhang Z, Zhang W, Li X, Wu T, Li T, Cai M, Yu Z, Xiang J, Cai D. Jia-Jian-Di-Huang-Yin-Zi decoction exerts neuroprotective effects on dopaminergic neurons and their microenvironment. Sci Rep 2018; 8:9886. [PMID: 29959371 PMCID: PMC6026152 DOI: 10.1038/s41598-018-27852-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 06/07/2018] [Indexed: 11/09/2022] Open
Abstract
As a classical prescription of Traditional Chinese medicine, the Jia-Jian-Di-Huang-Yin-Zi (JJDHYZ) decoction has long been used to treat movement disorders. The present study evaluated the effects of JJDHYZ on dopaminergic (DA) neurons and their survival-enhancing microenvironment as well as the possible mechanisms involved using a mouse model of Parkinson's disease. In MPTP-lesioned mice, a high dosage of JJDHYZ (34 g/kg/day) attenuated the loss of DA neurons, reversed the dopamine depletion, and improved the expression of glial-derived neurotrophic factor (GDNF) compared to the untreated model group. JJDHYZ also protected the ultrastructure of the blood-brain barrier (BBB) and tight junction proteins by inhibiting the activation of microglia and astrocytes besides the increase in three types of matrix metalloproteinases in the substantia nigra. In conclusion, the JJDHYZ-high dosage (JJDHYZ-H) group exhibited the neuroprotection of DA neurons, and the underlying mechanism may be related to the survival-enhancing microenvironment of the DA neurons.
Collapse
Affiliation(s)
- Jingsi Zhang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhennian Zhang
- Department of Neurology, Nanjing Hospital of Traditional Chinese Medicine, Nanjing, 210000, China
| | - Wen Zhang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiangting Li
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ting Wu
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Tingting Li
- Department of Neurology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Min Cai
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhonghai Yu
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jun Xiang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Dingfang Cai
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
45
|
Kokiko-Cochran ON, Godbout JP. The Inflammatory Continuum of Traumatic Brain Injury and Alzheimer's Disease. Front Immunol 2018; 9:672. [PMID: 29686672 PMCID: PMC5900037 DOI: 10.3389/fimmu.2018.00672] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/19/2018] [Indexed: 12/23/2022] Open
Abstract
The post-injury inflammatory response is a key mediator in long-term recovery from traumatic brain injury (TBI). Moreover, the immune response to TBI, mediated by microglia and macrophages, is influenced by existing brain pathology and by secondary immune challenges. For example, recent evidence shows that the presence of beta-amyloid and phosphorylated tau protein, two hallmark features of AD that increase during normal aging, substantially alter the macrophage response to TBI. Additional data demonstrate that post-injury microglia are “primed” and become hyper-reactive following a subsequent acute immune challenge thereby worsening recovery. These alterations may increase the incidence of neuropsychiatric complications after TBI and may also increase the frequency of neurodegenerative pathology. Therefore, the purpose of this review is to summarize experimental studies examining the relationship between TBI and development of AD-like pathology with an emphasis on the acute and chronic microglial and macrophage response following injury. Furthermore, studies will be highlighted that examine the degree to which beta-amyloid and tau accumulation as well as pre- and post-injury immune stressors influence outcome after TBI. Collectively, the studies described in this review suggest that the brain’s immune response to injury is a key mediator in recovery, and if compromised by previous, coincident, or subsequent immune stressors, post-injury pathology and behavioral recovery will be altered.
Collapse
Affiliation(s)
- Olga N Kokiko-Cochran
- Department of Neuroscience, Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Jonathan P Godbout
- Department of Neuroscience, Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
46
|
Chong SA, Balosso S, Vandenplas C, Szczesny G, Hanon E, Claes K, Van Damme X, Danis B, Van Eyll J, Wolff C, Vezzani A, Kaminski RM, Niespodziany I. Intrinsic Inflammation Is a Potential Anti-Epileptogenic Target in the Organotypic Hippocampal Slice Model. Neurotherapeutics 2018; 15:470-488. [PMID: 29464573 PMCID: PMC5935638 DOI: 10.1007/s13311-018-0607-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Understanding the mechanisms of epileptogenesis is essential to develop novel drugs that could prevent or modify the disease. Neuroinflammation has been proposed as a promising target for therapeutic interventions to inhibit the epileptogenic process that evolves from traumatic brain injury. However, it remains unclear whether cytokine-related pathways, particularly TNFα signaling, have a critical role in the development of epilepsy. In this study, we investigated the role of innate inflammation in an in vitro model of post-traumatic epileptogenesis. We combined organotypic hippocampal slice cultures, representing an in vitro model of post-traumatic epilepsy, with multi-electrode array recordings to directly monitor the development of epileptiform activity and to examine the concomitant changes in cytokine release, cell death, and glial cell activation. We report that synchronized ictal- and interictal-like activities spontaneously evolve in this culture. Dynamic changes in the release of the pro-inflammatory cytokines IL-1β, TNFα, and IL-6 were observed throughout the culture period (3 to 21 days in vitro) with persistent activation of microglia and astrocytes. We found that neutralizing TNFα with a polyclonal antibody significantly reduced ictal discharges, and this effect lasted for 1 week after antibody washout. Neither phenytoin nor an anti-IL-6 polyclonal antibody was efficacious in inhibiting the development of epileptiform activity. Our data show a sustained effect of the anti-TNFα antibody on the ictal progression in organotypic hippocampal slice cultures supporting the critical role of inflammatory mediators in epilepsy and establishing a proof-of-principle evidence for the utility of this preparation to test the therapeutic effects of anti-inflammatory treatments.
Collapse
Affiliation(s)
- Seon-Ah Chong
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium.
| | - Silvia Balosso
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, 20156, Italy
| | | | - Gregory Szczesny
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Etienne Hanon
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Kasper Claes
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Xavier Van Damme
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Bénédicte Danis
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Jonathan Van Eyll
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Christian Wolff
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Annamaria Vezzani
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, 20156, Italy
| | - Rafal M Kaminski
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | | |
Collapse
|
47
|
Chong SA, Balosso S, Vandenplas C, Szczesny G, Hanon E, Claes K, Van Damme X, Danis B, Van Eyll J, Wolff C, Vezzani A, Kaminski RM, Niespodziany I. Intrinsic Inflammation Is a Potential Anti-Epileptogenic Target in the Organotypic Hippocampal Slice Model. Neurotherapeutics 2018; 15:470-488. [PMID: 29464573 DOI: 10.1007/s13311-018-0607-6/figures/7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2023] Open
Abstract
Understanding the mechanisms of epileptogenesis is essential to develop novel drugs that could prevent or modify the disease. Neuroinflammation has been proposed as a promising target for therapeutic interventions to inhibit the epileptogenic process that evolves from traumatic brain injury. However, it remains unclear whether cytokine-related pathways, particularly TNFα signaling, have a critical role in the development of epilepsy. In this study, we investigated the role of innate inflammation in an in vitro model of post-traumatic epileptogenesis. We combined organotypic hippocampal slice cultures, representing an in vitro model of post-traumatic epilepsy, with multi-electrode array recordings to directly monitor the development of epileptiform activity and to examine the concomitant changes in cytokine release, cell death, and glial cell activation. We report that synchronized ictal- and interictal-like activities spontaneously evolve in this culture. Dynamic changes in the release of the pro-inflammatory cytokines IL-1β, TNFα, and IL-6 were observed throughout the culture period (3 to 21 days in vitro) with persistent activation of microglia and astrocytes. We found that neutralizing TNFα with a polyclonal antibody significantly reduced ictal discharges, and this effect lasted for 1 week after antibody washout. Neither phenytoin nor an anti-IL-6 polyclonal antibody was efficacious in inhibiting the development of epileptiform activity. Our data show a sustained effect of the anti-TNFα antibody on the ictal progression in organotypic hippocampal slice cultures supporting the critical role of inflammatory mediators in epilepsy and establishing a proof-of-principle evidence for the utility of this preparation to test the therapeutic effects of anti-inflammatory treatments.
Collapse
Affiliation(s)
- Seon-Ah Chong
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium.
| | - Silvia Balosso
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, 20156, Italy
| | | | - Gregory Szczesny
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Etienne Hanon
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Kasper Claes
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Xavier Van Damme
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Bénédicte Danis
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Jonathan Van Eyll
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Christian Wolff
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Annamaria Vezzani
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, 20156, Italy
| | - Rafal M Kaminski
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | | |
Collapse
|
48
|
Sun M, McDonald SJ, Brady RD, O'Brien TJ, Shultz SR. The influence of immunological stressors on traumatic brain injury. Brain Behav Immun 2018; 69:618-628. [PMID: 29355823 DOI: 10.1016/j.bbi.2018.01.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 01/13/2018] [Accepted: 01/14/2018] [Indexed: 12/15/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability worldwide, and typically involves a robust immune response. Although a great deal of preclinical research has been conducted to identify an effective treatment, all phase III clinical trials have been unsuccessful to date. These translational shortcomings are in part due to a failure to recognize and account for the heterogeneity of TBI, including how extracranial factors can influence the aftermath of TBI. For example, most preclinical studies have utilized isolated TBI models in young adult males, while clinical trials typically involve highly heterogeneous patient populations (e.g., different mechanisms of injury, a range of ages, presence of polytrauma or infection). This paper will review the current, albeit limited literature related to how TBI is affected by common concomitant immunological stressors. In particular, discussion will focus on whether extracranial trauma (i.e., polytrauma), infection, and age/immunosenescence can influence TBI pathophysiology, and thereby may result in a different brain injury than what would have occurred in an isolated TBI. It is concluded that these immunological stressors are all likely to be TBI modifiers that should be further studied and could impact translational treatment strategies.
Collapse
Affiliation(s)
- Mujun Sun
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Stuart J McDonald
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC 3086, Australia
| | - Rhys D Brady
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC 3052, Australia; Departments of Neuroscience and Medicine, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Terence J O'Brien
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC 3052, Australia; Departments of Neuroscience and Medicine, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Sandy R Shultz
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC 3052, Australia; Departments of Neuroscience and Medicine, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia.
| |
Collapse
|
49
|
Liu Y, Xie X, Xia LP, Lv H, Lou F, Ren Y, He ZY, Luo XG. Peripheral immune tolerance alleviates the intracranial lipopolysaccharide injection-induced neuroinflammation and protects the dopaminergic neurons from neuroinflammation-related neurotoxicity. J Neuroinflammation 2017; 14:223. [PMID: 29145874 PMCID: PMC5693474 DOI: 10.1186/s12974-017-0994-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 11/02/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Neuroinflammation plays a critical role in the onset and development of neurodegeneration disorders such as Parkinson's disease. The immune activities of the central nervous system are profoundly affected by peripheral immune activities. Immune tolerance refers to the unresponsiveness of the immune system to continuous or repeated stimulation to avoid excessive inflammation and unnecessary by-stander injury in the face of continuous antigen threat. It has been proved that the immune tolerance could suppress the development of various peripheral inflammation-related diseases. However, the role of immune tolerance in neuroinflammation and neurodegenerative diseases was not clear. METHODS Rats were injected with repeated low-dose lipopolysaccharide (LPS, 0.3 mg/kg) intraperitoneally for 4 days to induce peripheral immune tolerance. Neuroinflammation was produced using intracranial LPS (15 μg) injection. Inflammation cytokines were measured using enzyme-linked immunosorbent assay (ELISA) and quantitative real-time polymerase chain reaction (qRT-PCR). Microglial activation were measured using immunostaining of Iba-1 and ED-1. Dopaminergic neuronal damage was evaluated using immunochemistry staining and stereological counting of TH-positive neurons. Behavioral impairment was evaluated using amphetamine-induced rotational behavioral assessment. RESULTS Compared with the non-immune tolerated animals, pre-treatment of peripheral immune tolerance significantly decreased the production of inflammatory cytokines, suppressed the microglial activation, and increased the number of dopaminergic neuronal survival in the substantia nigra. CONCLUSIONS Our results indicated that peripheral immune tolerance attenuated neuroinflammation and inhibited neuroinflammation-induced dopaminergic neuronal death.
Collapse
Affiliation(s)
- Yang Liu
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Xin Xie
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Li-Ping Xia
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Hong Lv
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Fan Lou
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Yan Ren
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Zhi-Yi He
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Xiao-Guang Luo
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China.
| |
Collapse
|
50
|
De Blasio D, Fumagalli S, Longhi L, Orsini F, Palmioli A, Stravalaci M, Vegliante G, Zanier ER, Bernardi A, Gobbi M, De Simoni MG. Pharmacological inhibition of mannose-binding lectin ameliorates neurobehavioral dysfunction following experimental traumatic brain injury. J Cereb Blood Flow Metab 2017; 37:938-950. [PMID: 27165013 PMCID: PMC5363468 DOI: 10.1177/0271678x16647397] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Mannose-binding lectin is present in the contusion area of traumatic brain-injured patients and in that of traumatic brain-injured mice, where mannose-binding lectin-C exceeds mannose-binding lectin-A. The reduced susceptibility to traumatic brain injury of mannose-binding lectin double knock-out mice (mannose-binding lectin-/-) when compared to wild type mice suggests that mannose-binding lectin may be a therapeutic target following traumatic brain injury. Here, we evaluated the effects of a multivalent glycomimetic mannose-binding lectin ligand, Polyman9, following traumatic brain injury in mice. In vitro surface plasmon resonance assay indicated that Polyman9 dose-dependently inhibits the binding to immobilized mannose residues of plasma mannose-binding lectin-C selectively over that of mannose-binding lectin-A. Male C57Bl/6 mice underwent sham/controlled cortical impact traumatic brain injury and intravenous treatment with Polyman9/saline. Ex-vivo surface plasmon resonance studies confirmed that Polyman9 effectively reduces the binding of plasma mannose-binding lectin-C to immobilized mannose residues. In vivo studies up to four weeks post injury, showed that Polyman9 induces significant improvement in sensorimotor deficits (by neuroscore and beam walk), promotes neurogenesis (73% increase in doublecortin immunoreactivity), and astrogliosis (28% increase in glial fibrillary acid protein). Polyman9 administration in brain-injured mannose-binding lectin-/- mice had no effect on post-traumatic brain-injured functional deficits, suggestive of the specificity of its neuroprotective effects. The neurobehavioral efficacy of Polyman9 implicates mannose-binding lectin-C as a novel therapeutic target for traumatic brain injury.
Collapse
Affiliation(s)
- Daiana De Blasio
- 1 IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy.,2 Department of Anesthesia and Critical Care Medicine, Fondazione IRCCS Ca'Granda - Ospedale Maggiore Policlinico, Milano, Italy
| | - Stefano Fumagalli
- 1 IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy.,2 Department of Anesthesia and Critical Care Medicine, Fondazione IRCCS Ca'Granda - Ospedale Maggiore Policlinico, Milano, Italy
| | - Luca Longhi
- 3 Department of Anesthesia and Critical Care Medicine, Neurosurgical Intensive Care Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Franca Orsini
- 1 IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy
| | | | - Matteo Stravalaci
- 1 IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy
| | - Gloria Vegliante
- 1 IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy
| | - Elisa R Zanier
- 1 IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy
| | - Anna Bernardi
- 4 Department of Chemistry, Università degli Studi di Milano, Milano, Italy
| | - Marco Gobbi
- 1 IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy
| | | |
Collapse
|