Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25
1.
Nature ; 620(7976): 1071-1079, 2023 Aug.
Article En | MEDLINE | ID: mdl-37587343

Identifying therapeutics to delay, and potentially reverse, age-related cognitive decline is critical in light of the increased incidence of dementia-related disorders forecasted in the growing older population1. Here we show that platelet factors transfer the benefits of young blood to the ageing brain. Systemic exposure of aged male mice to a fraction of blood plasma from young mice containing platelets decreased neuroinflammation in the hippocampus at the transcriptional and cellular level and ameliorated hippocampal-dependent cognitive impairments. Circulating levels of the platelet-derived chemokine platelet factor 4 (PF4) (also known as CXCL4) were elevated in blood plasma preparations of young mice and humans relative to older individuals. Systemic administration of exogenous PF4 attenuated age-related hippocampal neuroinflammation, elicited synaptic-plasticity-related molecular changes and improved cognition in aged mice. We implicate decreased levels of circulating pro-ageing immune factors and restoration of the ageing peripheral immune system in the beneficial effects of systemic PF4 on the aged brain. Mechanistically, we identified CXCR3 as a chemokine receptor that, in part, mediates the cellular, molecular and cognitive benefits of systemic PF4 on the aged brain. Together, our data identify platelet-derived factors as potential therapeutic targets to abate inflammation and rescue cognition in old age.


Aging , Cognition , Cognitive Dysfunction , Neuroinflammatory Diseases , Nootropic Agents , Platelet Factor 4 , Animals , Male , Mice , Aging/blood , Aging/drug effects , Aging/physiology , Cognition/drug effects , Cognition/physiology , Neuroinflammatory Diseases/blood , Neuroinflammatory Diseases/complications , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/prevention & control , Platelet Factor 4/blood , Platelet Factor 4/metabolism , Platelet Factor 4/pharmacology , Platelet Factor 4/therapeutic use , Nootropic Agents/blood , Nootropic Agents/metabolism , Nootropic Agents/pharmacology , Nootropic Agents/therapeutic use , Plasma/chemistry , Hippocampus/drug effects , Hippocampus/physiology , Cognitive Dysfunction/blood , Cognitive Dysfunction/complications , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/prevention & control , Transcription, Genetic/drug effects , Neuronal Plasticity/drug effects
2.
Neurology ; 101(4): e455-e458, 2023 07 25.
Article En | MEDLINE | ID: mdl-37487758

While it was previously believed that neuromyelitis optic spectrum disorder (NMOSD) mostly affected the optic nerves and the spinal cord, it is increasingly recognized that NMOSD can involve any area of the CNS where aquaporin-4 is highly expressed. These other areas can include the hypothalamus and the circumventricular organs that surround the third and fourth ventricles, serving as osmoregulators. The syndrome of inappropriate antidiuretic hormone secretion (SIADH) is one of the most common causes of hyponatremia and has been associated with NMOSD due to these lesions. In this report, we present a case of a patient with known NMOSD, who presented with dizziness, fatigue, and generalized weakness and whose workup revealed hyponatremia in the setting of SIADH and hypothalamic demyelinating lesions. This case illustrates an atypical presentation of NMOSD and the importance of looking for syndromes, such as SIADH. This can guide diagnostic testing, such as getting thin MRI cuts through the hypothalamus and brainstem, as well as advanced management techniques such as immunotherapy.


Hyponatremia , Inappropriate ADH Syndrome , Neuroinflammatory Diseases , Neuromyelitis Optica , Adult , Female , Humans , Dizziness/complications , Fatigue/complications , Hyponatremia/complications , Hyponatremia/diagnosis , Hyponatremia/therapy , Hypothalamus/pathology , Inappropriate ADH Syndrome/complications , Inappropriate ADH Syndrome/diagnosis , Inappropriate ADH Syndrome/therapy , Magnetic Resonance Imaging , Neuroinflammatory Diseases/complications , Neuroinflammatory Diseases/pathology , Neuromyelitis Optica/complications , Neuromyelitis Optica/pathology , Immunotherapy
3.
Exp Neurol ; 367: 114470, 2023 09.
Article En | MEDLINE | ID: mdl-37327964

Clinical evidence indicates that major depression is a common comorbidity of chronic pain, including neuropathic pain; however, the cellular basis for chronic pain-mediated major depression remains unclear. Mitochondrial dysfunction induces neuroinflammation and has been implicated in various neurological diseases, including depression. Nevertheless, the relationship between mitochondrial dysfunction and anxiodepressive-like behaviors in the neuropathic pain state remains unclear. The current study examined whether hippocampal mitochondrial dysfunction and downstream neuroinflammation are involved in anxiodepressive-like behaviors in mice with neuropathic pain, which was induced by partial sciatic nerve ligation (PSNL). At 8 weeks after surgery, there was decreased levels of mitochondrial damage-associated molecular patterns, such as cytochrome c and mitochondrial transcription factor A, and increased level of cytosolic mitochondrial DNA in the contralateral hippocampus, suggesting the development of mitochondrial dysfunction. Type I interferon (IFN) mRNA expression in the hippocampus was also increased at 8 weeks after PSNL surgery. The restoration of mitochondrial function by curcumin blocked the increased cytosolic mitochondrial DNA and type I IFN expression in PSNL mice and improved anxiodepressive-like behaviors. Blockade of type I IFN signaling by anti-IFN alpha/beta receptor 1 antibody also improved anxiodepressive-like behaviors in PSNL mice. Together, these findings suggest that neuropathic pain induces hippocampal mitochondrial dysfunction followed by neuroinflammation, which may contribute to anxiodepressive-behaviors in the neuropathic pain state. Improving mitochondrial dysfunction and inhibiting type I IFN signaling in the hippocampus might be a novel approach to reducing comorbidities associated with neuropathic pain, such as depression and anxiety.


Anxiety , Depression , Interferon Type I , Mitochondria , Neuralgia , Animals , Male , Mice , Anxiety/complications , Anxiety/drug therapy , Anxiety/metabolism , Chronic Pain/complications , Chronic Pain/metabolism , Chronic Pain/pathology , Chronic Pain/psychology , Curcumin/pharmacology , Curcumin/therapeutic use , Cytosol/drug effects , Cytosol/metabolism , Depression/complications , Depression/drug therapy , Depression/metabolism , DNA, Mitochondrial/metabolism , Frontal Lobe/metabolism , Frontal Lobe/pathology , Hippocampus/drug effects , Hippocampus/immunology , Hippocampus/metabolism , Hippocampus/pathology , Interferon Type I/antagonists & inhibitors , Interferon Type I/genetics , Interferon Type I/metabolism , Microglia/drug effects , Microglia/immunology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Neuralgia/complications , Neuralgia/metabolism , Neuralgia/pathology , Neuralgia/psychology , Neuroinflammatory Diseases/complications , Sciatic Nerve/surgery
4.
Front Immunol ; 14: 1065226, 2023.
Article En | MEDLINE | ID: mdl-37197666

Mostly, pain has been studied in association with inflammation, until recent studies which indicate that during bacterial infections, pain mechanisms could be independent of the inflammation. Chronic pain can sustain long after the healing from the injury, even in the absence of any visible inflammation. However, the mechanism behind this is not known. We tested inflammation in lysozyme-injected mice foot paw. Interestingly, we observed no inflammation in mice foot paw. Yet, lysozyme injections induced pain in these mice. Lysozyme induces pain in a TLR4-dependent manner and TLR4 activation by its ligands such as LPS leads to inflammatory response. We compared the intracellular signaling of MyD88 and TRIF pathways upon TLR4 activation by lysozyme and LPS to understand the underlying mechanism behind the absence of an inflammatory response upon lysozyme treatment. We observed a TLR4 induced selective TRIF and not MyD88 pathway activation upon lysozyme treatment. This is unlike any other previously known endogenous TLR4 activators. A selective activation of TRIF pathway by lysozyme induces weak inflammatory cytokine response devoid of inflammation. However, lysozyme activates glutamate oxaloacetate transaminase-2 (GOT2) in neurons in a TRIF-dependent manner, resulting in enhanced glutamate response. We propose that this enhanced glutaminergic response could lead to neuronal activation resulting in pain sensation upon lysozyme injections. Collectively we identify that TLR4 activation by lysozyme can induce pain in absence of a significant inflammation. Also, unlike other known TLR4 endogenous activators, lysozyme does not activate MyD88 signaling. These findings uncover a mechanism of selective activation of TRIF pathway by TLR4. This selective TRIF activation induces pain with negligible inflammation, constituting a chronic pain homeostatic mechanism.


Chronic Pain , Neuroinflammatory Diseases , Toll-Like Receptor 4 , Animals , Mice , Muramidase/pharmacology , Chronic Pain/chemically induced , Chronic Pain/complications , Chronic Pain/metabolism , Toll-Like Receptor 4/metabolism , Myeloid Differentiation Factor 88/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Neuroinflammatory Diseases/complications , Neuroinflammatory Diseases/metabolism , Lipopolysaccharides , Neurons/drug effects , Neurons/metabolism
5.
BMC Med ; 21(1): 7, 2023 01 05.
Article En | MEDLINE | ID: mdl-36600274

BACKGROUND: Perioperative neurocognitive disorders (PND) with a high incidence frequently occur in elderly surgical patients closely associated with prolonged anesthesia-induced neurotoxicity. The neuromorphopathological underpinnings of anesthesia-induced neurotoxicity have remained elusive. METHODS: Prolonged anesthesia with sevoflurane was used to establish the sevoflurane-induced neurotoxicity (SIN) animal model. Morris water maze, elevated plus maze, and open field test were employed to track SIN rats' cognitive behavior and anxiety-like behaviors. We investigated the neuropathological basis of SIN through techniques such as transcriptomic, electrophysiology, molecular biology, scanning electron microscope, Golgi staining, TUNEL assay, and morphological analysis. Our work further clarifies the pathological mechanism of SIN by depleting microglia, inhibiting neuroinflammation, and C1q neutralization. RESULTS: This study shows that prolonged anesthesia triggers activation of the NF-κB inflammatory pathway, neuroinflammation, inhibition of neuronal excitability, cognitive dysfunction, and anxiety-like behaviors. RNA sequencing found that genes of different types of synapses were downregulated after prolonged anesthesia. Microglial migration, activation, and phagocytosis were enhanced. Microglial morphological alterations were also observed. C1qa, the initiator of the complement cascade, and C3 were increased, and C1qa tagging synapses were also elevated. Then, we found that the "Eat Me" complement pathway mediated microglial synaptic engulfment in the hippocampus after prolonged anesthesia. Afterward, synapses were remarkably lost in the hippocampus. Furthermore, dendritic spines were reduced, and their genes were also downregulated. Depleting microglia ameliorated the activation of neuroinflammation and complement and rescued synaptic loss, cognitive dysfunction, and anxiety-like behaviors. When neuroinflammatory inhibition or C1q neutralization occurred, complement was also decreased, and synaptic elimination was interrupted. CONCLUSIONS: These findings illustrated that prolonged anesthesia triggered neuroinflammation and complement-mediated microglial synaptic engulfment that pathologically caused synaptic elimination in SIN. We have demonstrated the neuromorphopathological underpinnings of SIN, which have direct therapeutic relevance for PND patients.


Anesthesia , Cognitive Dysfunction , Neuroinflammatory Diseases , Animals , Rats , Anesthesia/adverse effects , Anxiety/etiology , Anxiety/metabolism , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Complement C1q/metabolism , Hippocampus/metabolism , Microglia/drug effects , Microglia/physiology , Neuroinflammatory Diseases/chemically induced , Neuroinflammatory Diseases/complications , Sevoflurane/adverse effects , Sevoflurane/metabolism
6.
ACS Chem Neurosci ; 13(23): 3291-3302, 2022 12 07.
Article En | MEDLINE | ID: mdl-36399525

Depression is a serious mental illness, mainly characterized as large mood swings and sleep, diet, and cognitive function disorders. NLPR3, one of the inflammasomes that can be activated by a variety of stimuli to promote the maturation and secretion of pro-inflammatory cytokines, has been considered to be involved in the pathophysiology of depression. In this study, the putative role of CY-09, a selective and direct inhibitor of NLRP3, was evaluated in the lipopolysaccharide (LPS)-induced mice. The results of the study indicated that CY-09 significantly decreased the levels of NLRP3 in the hippocampus of LPS-induced mice. In addition, CY-09 increased the sucrose preference and shortened the immobility time in LPS-induced mice, suggesting the antidepressant-like effects of inhibiting NLRP3 inflammasome. Biochemical analysis showed that LPS significantly activated the NLRP3/ASC/cytokine signaling pathway and caused microglial activation, while CY-09 prevented the changes. Moreover, CY-09 increased the brain-derived neurotrophic factor (BDNF) only in microglia but not in the whole hippocampus. Meanwhile, CY-09 did not promote neurogenesis in the hippocampus of LPS mice. In conclusion, the results of the study showed that the antidepressant-like effects of NLRP3 inhibitor CY-09 were mediated by alleviating neuroinflammation in microglia and independent of the neurotrophic function in the hippocampus.


Depression , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Neuroinflammatory Diseases , Thiazolidines , Thiones , Animals , Mice , Inflammasomes/drug effects , Lipopolysaccharides/toxicity , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Thiones/pharmacology , Thiones/therapeutic use , Thiazolidines/pharmacology , Thiazolidines/therapeutic use , Neuroinflammatory Diseases/complications , Depression/drug therapy , Depression/etiology , Hippocampus/drug effects , Hippocampus/metabolism
7.
J Virol ; 96(17): e0006522, 2022 09 14.
Article En | MEDLINE | ID: mdl-35993737

Swine acute diarrhea syndrome coronavirus (SADS-CoV) is a recently emerging bat-borne coronavirus responsible for high mortality rates in piglets. In vitro studies have indicated that SADS-CoV has a wide tissue tropism in different hosts, including humans. However, whether this virus potentially threatens other animals remains unclear. Here, we report the experimental infection of wild-type BALB/c and C57BL/6J suckling mice with SADS-CoV. We found that mice less than 7 days old are susceptible to the virus, which caused notable multitissue infections and damage. The mortality rate was the highest in 2-day-old mice and decreased in older mice. Moreover, a preliminary neuroinflammatory response was observed in 7-day-old SADS-CoV-infected mice. Thus, our results indicate that SADS-CoV has potential pathogenicity in young hosts. IMPORTANCE SADS-CoV, which likely has originated from bat coronaviruses, is highly pathogenic to piglets and poses a threat to the swine industry. Little is known about its potential to disseminate to other animals. No efficient treatment is available, and the quarantine strategy is the only preventive measure. In this study, we demonstrated that SADS-CoV can efficiently replicate in suckling mice younger than 7 days. In contrast to infected piglets, in which intestinal tropism is shown, SADS-CoV caused infection and damage in all murine tissues evaluated in this study. In addition, neuroinflammatory responses were detected in some of the infected mice. Our work provides a preliminary cost-effective model for the screening of antiviral drugs against SADS-CoV infection.


Alphacoronavirus , Coronavirus Infections , Diarrhea , Mice , Swine Diseases , Alphacoronavirus/pathogenicity , Animals , Chiroptera/virology , Coronavirus Infections/complications , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Diarrhea/complications , Diarrhea/veterinary , Diarrhea/virology , Humans , Mice/virology , Mice, Inbred BALB C , Mice, Inbred C57BL , Neuroinflammatory Diseases/complications , Neuroinflammatory Diseases/veterinary , Neuroinflammatory Diseases/virology , Swine/virology , Swine Diseases/virology
8.
Int J Mol Sci ; 23(6)2022 Mar 13.
Article En | MEDLINE | ID: mdl-35328506

Cerebrovascular events, notably acute ischemic strokes (AIS), have been reported in the setting of novel coronavirus disease (COVID-19) infection. Commonly regarded as cryptogenic, to date, the etiology is thought to be multifactorial and remains obscure; it is linked either to a direct viral invasion or to an indirect virus-induced prothrombotic state, with or without the presence of conventional cerebrovascular risk factors. In addition, patients are at a greater risk of developing long-term negative sequelae, i.e., long-COVID-related neurological problems, when compared to non-COVID-19 stroke patients. Central to the underlying neurobiology of stroke recovery in the context of COVID-19 infection is reduced angiotensin-converting enzyme 2 (ACE2) expression, which is known to lead to thrombo-inflammation and ACE2/angiotensin-(1-7)/mitochondrial assembly receptor (MasR) (ACE2/Ang-(1-7)/MasR) axis inhibition. Moreover, after AIS, the activated nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome may heighten the production of numerous proinflammatory cytokines, mediating neuro-glial cell dysfunction, ultimately leading to nerve-cell death. Therefore, potential neuroprotective therapies targeting the molecular mechanisms of the aforementioned mediators may help to inform rehabilitation strategies to improve brain reorganization (i.e., neuro-gliogenesis and synaptogenesis) and secondary prevention among AIS patients with or without COVID-19. Therefore, this narrative review aims to evaluate the mediating role of the ACE2/Ang- (1-7)/MasR axis and NLRP3 inflammasome in COVID-19-mediated AIS, as well as the prospects of these neuroinflammation mediators for brain repair and in secondary prevention strategies against AIS in stroke rehabilitation.


Angiotensin-Converting Enzyme 2/metabolism , COVID-19/metabolism , Inflammasomes/metabolism , Ischemic Stroke/metabolism , Neuroinflammatory Diseases/metabolism , Proteins/metabolism , Angiotensin I/metabolism , COVID-19/complications , COVID-19/virology , Humans , Ischemic Stroke/complications , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuroinflammatory Diseases/complications , Peptide Fragments/metabolism , Proto-Oncogene Mas/metabolism , SARS-CoV-2/metabolism , SARS-CoV-2/physiology , Signal Transduction
9.
J Neuroinflammation ; 19(1): 4, 2022 Jan 04.
Article En | MEDLINE | ID: mdl-34983592

Patients with inflammatory bowel disease (IBD) suffer from depression at higher rates than the general population. An etiological trigger of depressive symptoms is theorised to be inflammation within the central nervous system. It is believed that heightened intestinal inflammation and dysfunction of the enteric nervous system (ENS) contribute to impaired intestinal permeability, which facilitates the translocation of intestinal enterotoxins into the blood circulation. Consequently, these may compromise the immunological and physiological functioning of distant non-intestinal tissues such as the brain. In vivo models of colitis provide evidence of increased blood-brain barrier permeability and enhanced central nervous system (CNS) immune activity triggered by intestinal enterotoxins and blood-borne inflammatory mediators. Understanding the immunological, physiological, and structural changes associated with IBD and neuroinflammation may aid in the development of more tailored and suitable pharmaceutical treatment for IBD-associated depression.


Brain-Gut Axis/physiology , Depression/etiology , Inflammatory Bowel Diseases/etiology , Neuroinflammatory Diseases/complications , Depression/physiopathology , Humans , Inflammatory Bowel Diseases/physiopathology , Neuroinflammatory Diseases/physiopathology
10.
Nutr Neurosci ; 25(1): 180-191, 2022 Jan.
Article En | MEDLINE | ID: mdl-32124682

Parkinson's disease (PD) is characterized by dysfunction of the nigrostriatal system, loss of dopamine neurons and intracellular aggregation of α-synuclein. Recently, both clinical and experimental studies have reported that neuroinflammation and oxidative stress markedly contribute to the etiology of PD. Current clinical pharmacotherapies only temporarily relieve the symptoms of PD, accompanied by many side effects. Hence, searching for natural anti-inflammatory, anti-oxidative and neuroprotective agents has received great attention. Polyunsaturated fatty acids (PUFAs), especially omega (n)-3, are essential lipid nutrients in the human diet and important components of cell membranes. Together by competing with the production of n-6 PUFAs, the precursors of inflammatory mediators, n-3 PUFAs can inhibit microglial activity and neuroinflammation, protect astrocyte function to produce neurotrophins, thereby normalizing neurotransmission and improving neurodegeneration. Thus, with regard to the hypotheses of PD, our and other's recent studies have demonstrated that n-3 PUFAs may improve PD by inhibiting proinflammatory cytokine release, promoting neurotrophic factor expression, recovering mitochondrial function and membrane fluidity, decreasing the levels of oxidant production, maintaining α-synuclein proteostasis, calcium homeostasis, axonal transport, and reducing endoplasmic reticulum stress. This review mainly introduces and analyzes the effect of n-3 PUFA treatments on PD-related behavioral and neuropathological abnormalities in clinical patients and different cellular and animal models of PD. Finally, the limitations and future work in n-3 PUFAs anti-PD area are discussed.


Fatty Acids, Omega-3/therapeutic use , Parkinson Disease/drug therapy , Animals , Anti-Inflammatory Agents , Antioxidants , Astrocytes/physiology , Humans , Microglia/drug effects , Microglia/physiology , Nerve Growth Factors/biosynthesis , Neuroinflammatory Diseases/complications , Neuroinflammatory Diseases/prevention & control , Neuroprotective Agents , Oxidative Stress , Parkinson Disease/etiology , alpha-Synuclein/metabolism
11.
Neurobiol Aging ; 109: 52-63, 2022 01.
Article En | MEDLINE | ID: mdl-34655981

Pathological aggregation of tau and neuroinflammatory changes mark the clinical course of Alzheimer's disease and related tauopathies. To understand the correlation between these pathological hallmarks and functional deficits, we assessed behavioral and physiological deficits in the PS19 mouse model, a broadly utilized model of tauopathy. At 9 months, PS19 mice have characteristic hyperactive behavior, a decline in motor strength, and deterioration in physiological conditions marked by lower body temperature, reduced body weight, and an increase in measures of frailty. Correlation of these deficits with different pathological hallmarks revealed that pathological tau species, characterized by soluble p-tau species, and tau seeding bioactivity correlated with impairment in grip strength and thermal regulation. On the other hand, astrocyte reactivity showed a positive correlation with the hyperactive behavior of the PS19 mice. These results suggest that a diverse spectrum of soluble pathological tau species could be responsible for different symptoms and that neuroinflammation could contribute to functional deficits independently from tau pathology. These observations enhance the necessity of a multi-targeted approach for the treatment of neurodegenerative tauopathies.


Gliosis/etiology , Neuroinflammatory Diseases/complications , Protein Aggregation, Pathological/complications , Tauopathies/etiology , tau Proteins/metabolism , Animals , Behavior, Animal , Body Temperature Regulation , Disease Models, Animal , Female , Frailty/etiology , Hand Strength , Humans , Male , Mice, Transgenic , Motor Activity , Tauopathies/pathology , Tauopathies/physiopathology , Tauopathies/psychology
12.
Clin. biomed. res ; 42(4): 397-404, 2022.
Article Pt | LILACS | ID: biblio-1516673

A Doença de Alzheimer (DA) consiste em um grande problema de saúde pública no Brasil e no mundo. Trata-se de uma doença neurodegenerativa, em que ocorre perda progressiva de neurônios e atrofia das regiões cerebrais. Essa degeneração está associada principalmente ao depósito de duas proteínas tóxicas: a proteína beta-amiloide e a proteína Tau, uma vez que estas proteínas se encontram acumuladas, elas prejudicam a ocorrência de sinapses nervosas. Apesar de extremamente prevalente na população mais idosa, suas causas ainda não estão bem esclarecidas, sendo que vários fatores já foram apontados como possíveis motivos para o surgimento do depósito destas proteínas, levando assim a neurodegeneração. Recentemente, tem se estudado o papel da inflamação, que é fundamental durante todo o curso da doença, tanto para a eliminação das proteínas tóxicas quanto para a proteção de neurônios. Um funcionamento anormal do processo inflamatório poderia dificultar a eliminação das proteínas e acentuar a perda neuronal. Com isso essa revisão de literatura tem como objetivo descrever os principais fatores imunológico que se encontram alterados na Doença de Alzheimer e como isso pode contribuir para o quadro neurodegenerativo.


Alzheimer's Disease (AD) is a major public health problem in Brazil and worldwide. It is a neurodegenerative disease, in which there is a progressive loss of neurons and atrophy of brain regions. This degeneration is mainly associated with the deposition of two toxic proteins, the beta-amyloid protein and the Tau protein, once these proteins are accumulated, they impair the occurrence of nerve synapses. Despite being extremely prevalent in the older population, its causes are still not well understood, and several factors have already been pointed out as possible reasons for the emergence of the deposit of these proteins, thus leading to neurodegeneration. Recently, the role of inflammation, which is fundamental throughout the course of the disease, has been studied, both for the elimination of toxic proteins and for the protection of neurons. An abnormal functioning of the inflammatory process could hinder the elimination of proteins and accentuate the neuronal loss Thus, this literature review aims to describe the main immunological factors that are altered in Alzheimer's Disease and how this can contribute to the neurodegenerative picture.


Alzheimer Disease/physiopathology , Neuroinflammatory Diseases/complications , Astrocytes , Microglia
13.
Open Heart ; 8(2)2021 11.
Article En | MEDLINE | ID: mdl-34819349

BACKGROUND: Myocardial infarction (MI) is associated with mental health disorders, in which neuroinflammation and cerebral microvascular dysfunction may play a role. Previously, we have shown that the proinflammatory factors Nε-(carboxymethyl)lysine (CML) and NADPH oxidase 2 (NOX2) are increased in the human infarcted heart microvasculature. The aim of this study was to analyse the presence of CML and NOX2 in the cerebral microvasculature of patients with MI. METHODS: Brain tissue was obtained at autopsy from 24 patients with MI and nine control patients. According to their infarct age, patients with MI were divided into three groups: 3-6 hours old (phase I), 6 hours-5 days old (phase II) and 5-14 days old (phase III). CML and NOX2 in the microvasculature were studied through immunohistochemical analysis. RESULTS: We observed a 2.5-fold increase in cerebral microvascular CML in patients with phase II and phase III MI (phase II: 21.39±7.91, p=0.004; phase III: 24.21±10.37, p=0.0007) compared with non-MI controls (8.55±2.98). NOX2 was increased in microvessels in patients with phase II MI (p=0.002) and phase III MI (p=0.04) compared with controls. No correlation was found between CML and NOX2 (r=0.58, p=0.13). CONCLUSIONS: MI coincides with an increased presence of CML and NOX2 in the brain microvasculature. These data point to proinflammatory alterations in the brain microvasculature that may underlie MI-associated mental health disorders.


Cerebral Arteries/enzymology , Lysine/analogs & derivatives , Microvessels/enzymology , Myocardial Infarction/enzymology , NADPH Oxidase 2/biosynthesis , Neuroinflammatory Diseases/enzymology , Aged , Biomarkers/metabolism , Cerebral Arteries/pathology , Female , Humans , Immunohistochemistry , Lysine/biosynthesis , Male , Microvessels/pathology , Middle Aged , Myocardial Infarction/complications , Myocardial Infarction/pathology , Neuroinflammatory Diseases/complications , Neuroinflammatory Diseases/pathology
14.
Cells ; 10(11)2021 11 05.
Article En | MEDLINE | ID: mdl-34831266

Cerebral complications in preeclampsia are leading causes of maternal mortality. Animal models suggest that an injured blood-brain barrier and neuroinflammation may be important but there is paucity of data from human studies. Therefore, we aimed to evaluate this in women with preeclampsia and eclampsia. We included women recruited to the South African Preeclampsia Obstetric Adverse Events (PROVE) biobank. Blood and cerebrospinal fluid (CSF) were collected around delivery. CSF was analyzed for neuroinflammatory markers interleukin 1ß, interleukin 6, interleukin-8 and tumor necrosis factor alpha (TNF-alpha). The CSF to plasma albumin ratio was measured to assess blood-brain barrier function. Women with eclampsia (n = 4) showed increased CSF concentrations of all pro-inflammatory cytokines and TNF-alpha compared to women with normotensive pregnancies (n = 7) and also for interleukin-6 and TNF-alpha compared to women with preeclampsia (n = 4). Women with preeclampsia also showed increases in pro-inflammatory cytokines IL-6 and IL-8 but not TNF-alpha in the CSF compared to women with normotensive pregnancies. In particular, women with eclampsia but also women with preeclampsia showed an increase in the CSF to plasma albumin ratio compared to normotensive women. In conclusion, women with preeclampsia and eclampsia show evidence of neuroinflammation and an injured blood-brain barrier. These findings are seen in particular among women with eclampsia.


Blood-Brain Barrier/pathology , Eclampsia/blood , Neuroinflammatory Diseases/blood , Neuroinflammatory Diseases/complications , Pre-Eclampsia/blood , Adult , Albumins/metabolism , Biomarkers/cerebrospinal fluid , Eclampsia/cerebrospinal fluid , Female , Humans , Neuroinflammatory Diseases/cerebrospinal fluid , Pre-Eclampsia/cerebrospinal fluid , Pregnancy
15.
Neurotox Res ; 39(6): 1800-1811, 2021 Dec.
Article En | MEDLINE | ID: mdl-34655374

Microglia-mediated neuroinflammation in response to injurious self and non-self-stimuli exerts detrimental effects on neurons, which may lead to cognitive impairment. Luteolin, a typical kind of natural flavonoid in honeysuckle, chrysanthemum, and Herba Schizonepetae, is widely recognized to be anti-inflammatory and antioxidant against peripheral inflammation. However, its protective effect against inflammation-induced cognitive impairment is currently unknown. In this paper, we investigated the relief potential of luteolin against lipopolysaccharide (LPS)-induced cognitive impairment and neuroinflammation and its possible anti-inflammatory mechanisms in lipopolysaccharide-stimulated BV2 microglia cells. In this study, luteolin ameliorated LPS-induced cognitive impairments, indicated by behavioral performance of neuroinflammatory model mice in Morris water maze tests. Protein analyses and histological examination also revealed protective effect of luteolin against neuronal damage, through inhibiting overproduction of inflammatory cytokines in both hippocampus and cortex of mice. We also observed luteolin in vitro significantly suppressed the levels of pro-inflammatory cytokines, such as tumor necrosis factor alpha (TNF-α) and interleukin-1 ß (IL-1ß), and inflammatory mediators like nitric oxide. Taken together, these results demonstrated luteolin was effective in alleviating cognitive impairment and limited neuronal damage via inhibiting the release of inflammatory mediators, suggesting luteolin is potential for further therapeutic research of neuroinflammation-related neurodegenerative diseases.


Cognitive Dysfunction/drug therapy , Luteolin/therapeutic use , Microglia/drug effects , Neuroinflammatory Diseases/drug therapy , Animals , Blotting, Western , Cell Line , Cognitive Dysfunction/etiology , Enzyme-Linked Immunosorbent Assay , Female , Male , Mice , Mice, Inbred C57BL , Morris Water Maze Test , Neuroinflammatory Diseases/complications
16.
Eur J Pharmacol ; 910: 174506, 2021 Nov 05.
Article En | MEDLINE | ID: mdl-34534533

This study aimed to investigate the effect of flavonoid morin on oxidative/nitrosative stress, neuroinflammation, and histological, molecular, and behavioral changes caused by amyloid-beta (Aß)1-42 in male Wistar rats (Alzheimer's disease model). Rats received morin (20 mg/kg, oral gavage) for 14 consecutive days after intrahippocampal injection of Aß1-42. Morin decreased the levels of malondialdehyde and nitric oxide, increased glutathione content, and enhanced catalase activity in the hippocampus of animals receiving Aß1-42. It also reduced the expression of tumor necrosis factor-α, interleukin-1ß, interleukin-6, nuclear factor-kappa B, and N-methyl-D-aspartate receptor subunits 2A and 2B and increased the expression of brain-derived neurotrophic factor and α7 nicotinic acetylcholine receptor in the hippocampus of Aß1-42-injected rats. Besides, morin modified neuronal loss and histological changes in the CA1 region of the hippocampus. Morin allowed Aß1-42-infused rats to swim more time in the target quadrant in the Morris water maze test. It is concluded that morin may be suitable for the prevention and treatment of Alzheimer's disease by strengthening the antioxidant system, inhibiting neuroinflammation, preventing neuronal death, and enhancing memory function.


Alzheimer Disease/drug therapy , Antioxidants/pharmacology , Flavonoids/pharmacology , Memory Disorders/drug therapy , Neuroinflammatory Diseases/drug therapy , Alzheimer Disease/complications , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Amyloid beta-Peptides/administration & dosage , Animals , Antioxidants/therapeutic use , Disease Models, Animal , Flavonoids/therapeutic use , Hippocampus/immunology , Hippocampus/pathology , Humans , Male , Memory Disorders/immunology , Memory Disorders/pathology , Neuroinflammatory Diseases/complications , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/pathology , Oxidative Stress/drug effects , Oxidative Stress/immunology , Peptide Fragments/administration & dosage , Rats , Rats, Wistar
17.
Exp Mol Med ; 53(9): 1251-1267, 2021 09.
Article En | MEDLINE | ID: mdl-34489558

Recent research into meningeal lymphatics has revealed a never-before appreciated role of type II innate lymphoid cells (ILC2s) in modulating neuroinflammation in the central nervous system (CNS). To date, the role of ILC2-mediated inflammation in the periphery has been well studied. However, the exact distribution of ILC2s in the CNS and therefore their putative role in modulating neuroinflammation in neurodegenerative diseases such as Alzheimer's disease (AD), multiple sclerosis (MS), Parkinson's disease (PD), and major depressive disorder (MDD) remain highly elusive. Here, we review the current evidence of ILC2-mediated modulation of neuroinflammatory cues (i.e., IL-33, IL-25, IL-5, IL-13, IL-10, TNFα, and CXCL16-CXCR6) within the CNS, highlight the distribution of ILC2s in both the periphery and CNS, and discuss some challenges associated with cell type-specific targeting that are important for therapeutics. A comprehensive understanding of the roles of ILC2s in mediating and responding to inflammatory cues may provide valuable insight into potential therapeutic strategies for many dementia-related disorders.


Immunity, Innate , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Meninges/metabolism , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neuroinflammatory Diseases/complications , Animals , Biomarkers , Brain/metabolism , Cytokines/metabolism , Diagnosis, Differential , Disease Susceptibility , Humans , Immunomodulation , Lymphatic System/immunology , Lymphatic System/metabolism , Meninges/immunology , Neurodegenerative Diseases/diagnosis , Neuroimmunomodulation , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/metabolism
18.
Retrovirology ; 18(1): 26, 2021 09 16.
Article En | MEDLINE | ID: mdl-34530855

BACKGROUND: Extracellular Vesicles (EV) recently have been implicated in the pathogenesis of HIV-1 syndromes, including neuroinflammation and HIV-1 associated neurological disorder (HAND). Cocaine, an illicit stimulant drug used worldwide is known to exacerbate these HIV-1 associated neurological syndromes. However, the effects of cocaine on EV biogenesis and roles of EVs in enhancing HIV-1 pathogenesis are not yet well defined. RESULTS: Here, we investigated the effects of cocaine on EV biogenesis and release in HIV-1 infected immune cells and explored their roles in elicitation of neuroinflammation. We found that cocaine significantly augmented the release of EVs from uninfected and HIV-1 infected T-cells, DCs and macrophages. Further analysis of the molecular components of EVs revealed enhanced expression of adhesion molecules integrin ß1 and LFA-1 in those EVs derived from cocaine treated cells. Intriguingly, in EVs derived from HIV-1 infected cells, cocaine treatment significantly increased the levels of viral genes in EVs released from macrophages and DCs, but not in T-cells. Exploring the molecular mechanism to account for this, we found that DCs and macrophages showed enhanced expression of the cocaine receptor Sigma 1-Receptor compared to T-cells. In addition, we found that cocaine significantly altered the integrity of the RNA-induced silencing complex (RISC) in HIV-1 infected macrophages and DCs compared to untreated HIV-1 infected cells. Characterizing further the molecular mechanisms involved in how cocaine increased EV release, we found that cocaine decreased the expression of the interferon-inducible protein BST-2; this resulted in altered trafficking of intracellular virus containing vesicles and EV biogenesis and release. We also observed EVs released from cocaine treated HIV-1 infected macrophages and DCs enhanced HIV-1 trans-infection to T-cells compared to those from untreated and HIV-1 infected cells. These EVs triggered release of proinflammatory cytokines in human brain microvascular endothelial cells (HBMECs) and altered monolayer integrity. CONCLUSIONS: Taken together, our results provide a novel mechanism which helps to elucidate the enhanced prevalence of neurological disorders in cocaine using HIV-1 infected individuals and offers insights into developing novel therapeutic strategies against HAND in these hosts.


Cocaine/adverse effects , Cocaine/immunology , Dendritic Cells/drug effects , Extracellular Vesicles/drug effects , HIV-1/immunology , Macrophages/drug effects , Neuroinflammatory Diseases/complications , Brain/cytology , Cocaine/pharmacology , Cytokines/immunology , Dendritic Cells/virology , Endothelial Cells/drug effects , Endothelial Cells/immunology , Endothelial Cells/virology , Extracellular Vesicles/immunology , Extracellular Vesicles/virology , HIV-1/drug effects , HIV-1/pathogenicity , Humans , Inflammation , Macrophages/immunology , Macrophages/virology , Organelle Biogenesis
19.
Metab Brain Dis ; 36(7): 1591-1626, 2021 10.
Article En | MEDLINE | ID: mdl-34387831

Neuroinflammation is one of the host defensive mechanisms through which the nervous system protects itself from pathogenic and or infectious insults. Moreover, neuroinflammation occurs as one of the most common pathological outcomes in various neurological disorders, makes it the promising target. The present review focuses on elaborating the recent advancement in understanding molecular mechanisms of neuroinflammation and its role in the etiopathogenesis of various neurological disorders, especially Alzheimer's disease (AD), Parkinson's disease (PD), and Epilepsy. Furthermore, the current status of anti-inflammatory agents in neurological diseases has been summarized in light of different preclinical and clinical studies. Finally, possible limitations and future directions for the effective use of anti-inflammatory agents in neurological disorders have been discussed.


Nervous System Diseases/etiology , Neuroinflammatory Diseases/etiology , Alzheimer Disease/etiology , Anti-Inflammatory Agents/therapeutic use , Autophagy , Cytokines/physiology , Humans , Inflammasomes/physiology , Monocytes/physiology , Nervous System Diseases/drug therapy , Neuroglia/physiology , Neuroinflammatory Diseases/complications , Oxidative Stress , T-Lymphocytes/physiology
20.
J Neuroinflammation ; 18(1): 128, 2021 Jun 06.
Article En | MEDLINE | ID: mdl-34092247

BACKGROUND: Patients with prior illness are more vulnerable to heat stroke-induced injury, but the underlying mechanism is unknown. Recent studies suggested that NLRP3 inflammasome played an important role in the pathophysiology of heat stroke. METHODS: In this study, we used a classic animal heat stroke model. Prior infection was mimicked by using lipopolysaccharide (LPS) or lipoteichoic acid (LTA) injection before heat stroke (LPS/LTA 1 mg/kg). Mice survival analysis curve and core temperature (TC) elevation curve were produced. NLRP3 inflammasome activation was measured by using real-time PCR and Western blot. Mice hypothalamus was dissected and neuroinflammation level was measured. To further demonstrate the role of NLRP3 inflammasome, Nlrp3 knockout mice were used. In addition, IL-1ß neutralizing antibody was injected to test potential therapeutic effect on heat stroke. RESULTS: Prior infection simulated by LPS/LTA injection resulted in latent inflammation status presented by high levels of cytokines in peripheral serum. However, LPS/LTA failed to cause any change in animal survival rate or body temperature. In the absence of LPS/LTA, heat treatment induced heat stroke and animal death without significant systemic or neuroinflammation. Despite a decreased level of IL-1ß in hypothalamus, Nlrp3 knockout mice demonstrated no survival advantage under mere heat exposure. In animals with prior infection, their heat tolerance was severely impaired and NLRP3 inflammasome induced neuroinflammation was detected. The use of Nlrp3 knockout mice enhanced heat tolerance and alleviated heat stroke-induced death by reducing mice hypothalamus IL-1ß production with prior infection condition. Furthermore, IL-1ß neutralizing antibody injection significantly extended endotoxemic mice survival under heat stroke. CONCLUSIONS: Based on the above results, NLRP3/IL-1ß induced neuroinflammation might be an important mechanistic factor in heat stroke pathology, especially with prior infection. IL-1ß may serve as a biomarker for heat stroke severity and potential therapeutic method.


Brain/metabolism , Brain/pathology , Heat Stroke/complications , Heat Stroke/physiopathology , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuroinflammatory Diseases/complications , Neuroinflammatory Diseases/metabolism , Animals , Antibodies, Neutralizing/therapeutic use , Disease Models, Animal , Heat Stroke/drug therapy , Heat Stroke/pathology , Inflammasomes/metabolism , Interleukin-1beta/immunology , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Signal Transduction , Teichoic Acids , Thermotolerance
...