Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Immunol Invest ; : 1-56, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38836373

ABSTRACT

Studying various microglial phenotypes and their functions in neurodegenerative diseases is crucial due to the intricate nature of their phenomics and their vital immunological role. Microglia undergo substantial phenomic changes, encompassing morphological, transcriptional, and functional aspects, resulting in distinct cell types with diverse structures, functions, properties, and implications. The traditional classification of microglia as ramified, M1 (proinflammatory), or M2 (anti-inflammatory) phenotypes is overly simplistic, failing to capture the wide range of recently identified microglial phenotypes in various brain regions affected by neurodegenerative diseases. Altered and activated microglial phenotypes deviating from the typical ramified structure are significant features of many neurodegenerative conditions. Understanding the precise role of each microglial phenotype is intricate and sometimes contradictory. This review specifically focuses on elucidating recent modifications in microglial phenotypes within neurodegenerative diseases. Recognizing the heterogeneity of microglial phenotypes in diseased states can unveil novel therapeutic strategies for targeting microglia in neurodegenerative diseases. Moreover, the exploration of the use of healthy isolated microglia to mitigate disease progression has provided an innovative perspective. In conclusion, this review discusses the dynamic landscape of mysterious microglial phenotypes, emphasizing the need for a nuanced understanding to pave the way for innovative therapeutic strategies for neurodegenerative diseases.

2.
Inflammopharmacology ; 31(5): 2303-2310, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37405587

ABSTRACT

Alzheimer's disease (AD) is a progressive type of neurodegenerative disease characterized by successive loss of the conventional structure and functions of neurons. In addition to dead neurons type detected within AD brain tissues, there are a predominantly varying number of deteriorating neurons (DTNs). As the number of deteriorating neurons increases, they exaggerate the release of inflammatory factors and oxidative stress that trigger the cascade of neuroinflammation. Triggering receptor expressed on myeloid cells 1 (TREM-1) which is a transmembrane immune receptor type regularly expressed by phagocytic cells, may act as a stimulating factor for neuroinflammation. Once TREM-1 is activated, it directly activates spleen tyrosine kinase (SYK) downstream signaling cascades, which can be considered an initiating phase for AD pathology and AD progression. Sequentially, SYK activates the pro-inflammatory microglia M1 phenotype which executes several inflammatory actions, leading to neurotoxicity. These released neurotoxins promote neuronal cell death, synaptic dysfunctions, and memory impairments. Thus, the current review outlines the direct etiological and pathologic features of Alzheimer's disease linked with deteriorating neurons, TREM-1, and SYK.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Humans , Alzheimer Disease/metabolism , Triggering Receptor Expressed on Myeloid Cells-1/metabolism , Neurodegenerative Diseases/metabolism , Neuroinflammatory Diseases , Neurons/pathology , Microglia/metabolism , Syk Kinase/metabolism
3.
Inflammopharmacology ; 31(3): 1257-1277, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37017850

ABSTRACT

Neuroinflammation is a devastating predisposing factor for Alzheimer's disease (AD). A number of clinical findings have reported peripheral disorders among AD patients. Amyloid beta (Aß) is a toxic physiological aggregate that serves as a triggering factor for hepatic and cardiac disorders related to neurotoxicity. As a drawback of Aß excessive accumulation in the brain, part of Aß is believed to readily cross the blood-brain barrier (BBB) into the peripheral circulation resulting in serious inflammatory and toxic cascades acting as a direct bridge to cardiac and hepatic pathophysiology. The main aim is to find out whether neuroinflammation-related AD may result in cardiac and liver dysfunctions. Potential therapeutic interventions are also suggested to alleviate AD's cardiac and hepatic defects. Male rats were divided into: control group I, lipopolysaccharide (LPS)-neuroinflammatory-induced group II, LPS-neuroinflammatory-induced group treated with sodium hydrogen sulphide donor (NaHS) (group III), and LPS-neuroinflammatory-induced group treated with mesenchymal stem cells (MSCs) (group IV). Behavior and histopathological studies were conducted in addition to the estimation of different biological biomarkers. It was revealed that the increased toxic Aß level in blood resulted in cardiac and hepatic malfunctions as a drawback of exaggerated inflammatory cascades. The administration of NaHS and MSCs proved their efficiency in combating neuroinflammatory drawbacks by hindering cardiac and hepatic dysfunctions. The consistent direct association of decreased heart and liver functions with increased Aß levels highlights the direct involvement of AD in other organ complications. Thereby, these findings will open new avenues for combating neuroinflammatory-related AD and long-term asymptomatic toxicity.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Rats , Male , Animals , Lipopolysaccharides/toxicity , Neuroinflammatory Diseases , Alzheimer Disease/chemically induced , Liver/pathology
4.
Eur J Neurosci ; 56(9): 5453-5475, 2022 11.
Article in English | MEDLINE | ID: mdl-34182602

ABSTRACT

The brain is a highly vascularized tissue protected by the blood-brain barrier (BBB), a complex structure allowing only necessary substances to pass through into the brain while limiting the entrance of harmful toxins. The BBB comprises several components, and the most prominent features are tight junctions between endothelial cells (ECs), which are further wrapped in a layer of pericytes. Pericytes are multitasked cells embedded in a thick basement membrane (BM) that consists of a fibrous extracellular matrix (ECM) and are surrounded by astrocytic endfeet. The primary function of astrocytes and pericytes is to provide essential blood supply and vital nutrients to the brain. In Alzheimer's disease (AD), long-term neuroinflammatory cascades associated with infiltration of harmful neurotoxic proteins may lead to BBB dysfunction and altered ECM components resulting in brain homeostatic imbalance, synaptic damage, and declined cognitive functions. Moreover, BBB structure and functional integrity may be lost due to induced ECM alterations, astrocyte damage, and pericytes dysfunction, leading to amyloid-beta (Aß) hallmarks deposition in different brain regions. Herein, we highlight how BBB, ECM, astrocytes, and pericytes dysfunction can play a leading role in AD's pathogenesis and discuss their impact on brain functions.


Subject(s)
Alzheimer Disease , Pericytes , Humans , Pericytes/metabolism , Pericytes/pathology , Astrocytes/metabolism , Alzheimer Disease/metabolism , Endothelial Cells/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/pathology
5.
Cell Biochem Funct ; 40(1): 17-27, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34716723

ABSTRACT

Neurologists have highly observed a frequent increasing number of elderly patients with Alzheimer's disease (AD) without any relevant evidence of any genetic or known AD-linked predisposing factors in the past few years. Those patients are characterized by continuous and irreversible neuron cells loss along with declined cognitive functions. Numerous studies have suggested that the exaggerated release of reactive oxygen species (ROS) within the brain may develop late-onset neurodegenerative disorders, especially AD-neuroinflammatory type. However, the central nervous system is vitally linked with whole-brain chemical integrity and its related healthy state, the cascade by which ROS may result in AD's development has not been highly justified or even maintained. It is widely known that the brain consumes a vast amount of oxygen and is characterized by being rich in lipid polyunsaturated fatty acids content, explaining why it is a prone region to oxidative stress (OS) and ROS damage. The formed OS-AD cytoskeletal protein aggregates can be considered a main predisposing factor for amyloid-beta (Aß) hallmarks precipitation. Herein, this review aims to provide a detailed information on how oxidative stress can play a pathogenic role in activating damage-associated molecular patterns (DAMPs)-related toll-like receptor-4 inflammatory (TLR-4) cascades resulting in the deposition of Aß hallmarks in brain tissues ending with irreversible cognitive dysfunction. It also explains how microglia can be activated via ROS, which may significantly release several pro-inflammatory cascades ending with general brain atrophy. Furthermore, different types of suggested antioxidant therapies will be discussed to combat AD-related pathological disorders and hallmarks.


Subject(s)
Alzheimer Disease , Aged , Amyloid beta-Peptides/metabolism , Brain/metabolism , Humans , Oxidative Stress , Reactive Oxygen Species
6.
J Immunoassay Immunochem ; 41(6): 960-975, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-33393415

ABSTRACT

In December 2019, an outbreak of pandemic severe respiratory distress syndrome coronavirus disease 2019 (COVID-19) initially occurred in China, has spread the world resulted in serious threats to human public health. Uncommon neurological manifestations with pathophysiological symptoms were observed in infected patients including headache, seizures, and neuroimmunological disorders. Regardless of whether these neurological symptoms are direct or indirect casual infection relationship, this novel viral infection has a relevant impact on the neuroimmune system that requires a neurologist's careful assessment. Recently, the use of immunotherapy has been emerged in fighting against COVID-19 infection despite the uncertain efficiency in managing COVID-19 related disorders or even its proven failure by increasing its severity. Herein, the author is addressing the first approaches in using immunotherapies in controlling COVID-19 viral impact on the brain by highlighting their role in decreasing or increasing infection risks among subjects. This point of view review article supports the use of immunotherapies in managing COVID-19 neurological disorders but in optimal timing and duration to ensure the maximum therapeutic outcome by reducing morbidity and mortality rate. Based on recently published data, the current review article highlights the beneficial effects and drawbacks of using immunotherapies to combat COVID-19 and its neurological symptoms.


Subject(s)
COVID-19/immunology , COVID-19/therapy , Cytokine Release Syndrome/virology , Immunotherapy/methods , Nervous System Diseases/immunology , Nervous System Diseases/therapy , Brain/pathology , COVID-19/complications , Cladribine/therapeutic use , Cytokine Release Syndrome/therapy , Cytokines/immunology , Glucocorticoids/therapeutic use , Headache/virology , Humans , Immunoglobulins, Intravenous/therapeutic use , Interferon-beta/immunology , Interleukin-6/immunology , Nervous System Diseases/virology , Pandemics
7.
BMC Complement Med Ther ; 24(1): 153, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38581023

ABSTRACT

BACKGROUND: Vortioxetine (VORTX) is a potent and selective type of selective serotonin reuptake inhibitor (SSRI) that is mainly prescribed for treating major depression along with mood disorders as the first drug of choice. Limited previous findings have indicated evidence of liver injury and hepatotoxicity associated with daily VORTX treatment. Rutin (RUT), which is known for its antioxidant properties, has demonstrated several beneficial health actions, including hepatoprotection. Therefore the current study aimed to evaluate and assess the ameliorative effect of RUT against the hepatotoxic actions of daily low and high-dose VORTX administration. METHODS: The experimental design included six groups of rats, each divided equally. Control, rats exposed to RUT (25 mg/kg), rats exposed to VORTX (28 mg/kg), rats exposed to VORTX (28 mg/kg) + RUT (25 mg/kg), rats exposed to VORTX (80 mg/kg), and rats exposed to VORTX (80 mg/kg) + RUT (25 mg/kg). After 30 days from the daily exposure period, assessments were conducted for serum liver enzyme activities, hepatotoxicity biomarkers, liver antioxidant endogenous enzymes, DNA fragmentation, and histopathological studies of liver tissue. RESULTS: Interestingly, the risk of liver damage and hepatotoxicity related to VORTX was attenuated by the daily co-administration of RUT. Significant improvements were observed among all detected liver functions, oxidative stress, and inflammatory biomarkers including aspartate aminotransferase (AST), alanine transaminase (ALT), lactate dehydrogenase (LDH), albumin, malondialdehyde (MDA), superoxide dismutase (SOD), glutathione (GSH), glutathione S-transferase (GST), total protein, acid phosphatase, N-Acetyl-/ß-glucosaminidase (ß-NAG), ß-Galactosidase (ß-Gal), alpha-fetoprotein (AFP), caspase 3, and cytochrom-C along with histopathological studies, compared to the control and sole RUT group. CONCLUSION: Thus, RUT can be considered a potential and effective complementary therapy in preventing hepatotoxicity and liver injury induced by the daily or prolonged administration of VORTX.


Subject(s)
Antioxidants , Chemical and Drug Induced Liver Injury , Rats , Animals , Antioxidants/pharmacology , Rutin/pharmacology , Vortioxetine , Inflammation/drug therapy , Glutathione/metabolism , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/prevention & control , Biomarkers
8.
Sci Rep ; 14(1): 5113, 2024 03 01.
Article in English | MEDLINE | ID: mdl-38429312

ABSTRACT

Inflammation is a part of the body's intricate biological reaction to noxious stimuli and defensive reactions. So, the aim of this investigation was to study the anti-inflammatory activity of exopolysaccharide (EPSSM) using carrageenan-induced paw edema in rats. A halophilic bacterial strain was isolated from marine sediments in the Red Sea in Egypt. The isolate has been visually and physiologically recognized, as well as by analyzing its 16S rRNA gene, which confirms Kocuria sp. clone Asker4. This particular isolate can be referenced using the accession number OL798051.1. EPSSM was subjected to purification and fractionation by a DEAE-cellulose column. Preliminary chemical analysis of EPSSM indicated that the monosaccharides were fructose, glucuronic acid, and xylose, with 2.0, 0.5, and 1.0, respectively. The antioxidant potential of EPSSM was investigated, and it was discovered that the level of activity increased independently of the concentrations, reaching a maximum threshold of 94.13% at 100 µg/mL of EPSSM for 120 min. Also, EPSSM at 50 mg/kg orally produced a significant anti-inflammatory effect on the carrageenan model at 2, 3, and 4 intervals. The EPSSM intervention resulted in reductions in the levels of catalase and superoxide dismutase enzymes, as well as a decrease in glutathione. Furthermore, the levels of nitric oxide, lipid peroxidation, and reactive oxygen species resulting from carrageenan-induced edema showed a significant reduction subsequent to the administration of EPSSM. Moreover, the findings indicated that the protein expression levels of cyclooxygenase-2 and interleukin-6 were reduced following treatment with EPSSM, resulting in a reduction of paw edema.


Subject(s)
Antioxidants , Bacteria , Animals , Rats , Antioxidants/pharmacology , Carrageenan , RNA, Ribosomal, 16S , Edema/chemically induced , Edema/drug therapy , Inflammation , Nitric Oxide
9.
Ibrain ; 9(1): 63-71, 2023.
Article in English | MEDLINE | ID: mdl-37786515

ABSTRACT

Despite decades of repeated and intense research, the etiology of sudden Alzheimer's disease (AD) symptoms is still unclear. AD progressive pathology mainly involves neuron damage, depositions of amyloid-beta (Aß), and hyperphosphorylated tau protein. All these defects are manifested by exaggerated cytokine storm and neuroinflammation leading to irreversible brain damage in the long term. Despite the numerous risks and drawbacks associated with AD, it is believed that there is a hidden unknown causative and predisposing factors for AD. Extracellular vesicles (EVs) are small vesicles released by cells as a type of intercellular communication. Several pieces of evidence support the inclusion of viral components within EVs facilitating their penetration into the blood-brain barrier leading to neuroinflammation. In light of the SARS-CoV-19 pandemic and its related neurological complications, it is mandatory to highlight the possibility and viability of viral infections such as varicella-zoster virus (VZV) and herpes simplex virus (HSV) on the onset of AD. Herein, the author is investigating the potential role of VZV and HSV along with highlighting the suggested route of pathogenesis entry resulting in AD manifestations. Additionally, this review aims to summarize the role of EVs in mediating the central nervous system viral infections leading to AD.

10.
Neurodegener Dis Manag ; 13(2): 85-99, 2023 04.
Article in English | MEDLINE | ID: mdl-36644988

ABSTRACT

Aim: To investigate whether the estimation of cerebrospinal fluid (CSF) and brain YKL-40 levels may be used as an efficient biomarker for Parkinson's disease (PD). Methods: Lipopolysaccharides (LPS) was injected into the right substantia nigra pars compacta (SNpc). Rats were divided into: control group, early LPS-induced PD group (14 days), and advanced LPS-induced PD group (28 days). YKL-40 and other related factors were detected in CSF and brain tissue. Results: Increased expression of YKL-40 was observed in brain tissue and CSF of PD-induced rats associated with triggered inflammatory cytokine release. Conclusion: The current study was limited to detecting YKL-40 and other inflammatory factors in brain and CSF. YKL-40 may be considered as an early biomarker and therapeutic target for PD.


Parkinson's disease (PD) is one of the most devastating fast-spreading neurological disorders with a high prevalence and incidence of death. Progress in developing efficient therapeutic interventions in PD may only be achieved through reliable early diagnostic approaches along with regular monitoring of PD progression. Although the current clinical diagnostic approach to PD relies mainly on late motor symptoms, the rate of misdiagnosis is considered high due to the overlap of PD with other etiologies. Novel and reliable biological biomarkers are highly required among researchers with the aim of early detection of the disease pathogenesis during the premotor stages to avoid debilitating PD stages. Thereby, identifying and highlighting novel biological biomarkers are highly required in the early diagnosis of neurological disorders.


Subject(s)
Parkinson Disease , Humans , Rats , Animals , Parkinson Disease/diagnosis , Chitinase-3-Like Protein 1/metabolism , Lipopolysaccharides/metabolism , Biomarkers/metabolism , Brain/metabolism
11.
Nat Prod Res ; 37(5): 847-851, 2023 Mar.
Article in English | MEDLINE | ID: mdl-35730634

ABSTRACT

Flavonoids are polyphenolic natural compounds with various biological actions and limited toxicity including diosmin (DM) which is considered a safe flavonoid natural type with anti-inflammatory and antioxidant activities. Tramadol (TM) is a centrally long-acting analgesic class of opioids extensively being used among the population. It was reported that long-term exposure to TM triggers the releases of oxidative stress, inflammatory factors, and nitric oxides resulting in organs damage. This study aimed to investigate the possible ameliorative and restorative actions of DM against tramadol-induced testicular damage. Rats were divided into: GI: control; GII: Rats received TM, GIII: Rats received DM, GIV: Rats received TM + DM; GV: Rats received DM + TM. Rat's testicular tissue and blood samples were collected. A relevant improvement in all examined parameters was observed among GIV and GV groups. Thereby, it was highlighted that diosmin has beneficial natural actions against tramadol-induced testicular injury via suppressing triggered oxidative stress, and inflammatory factors.


Subject(s)
Diosmin , Infertility , Tramadol , Rats , Male , Animals , Tramadol/pharmacology , Rats, Wistar , Diosmin/pharmacology , Flavonoids/pharmacology , Antioxidants/pharmacology , Antioxidants/metabolism , Oxidative Stress
12.
BMC Complement Med Ther ; 22(1): 327, 2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36482339

ABSTRACT

BACKGROUND: Among the most commonly consumed non-steroidal anti-inflammatory drugs (NSAID) is Diclofenac (Dic), especially in low-income countries due to its high efficiency and affordable price. However, the continuous administration of Diclofenac may induce toxic effects on various body organs including the liver and kidney. Caffeine (Caf) (1,3,7-trimethylxanthine) is a pharmacologically active alkaloid type with antioxidant and anti-inflammatory actions. AIM: The current study aims to evaluate the ameliorative effect of Caffeine against Dic-induced hepato-renal toxicity and damage. METHODS: Twenty-four male albino rats type were assigned randomly into four groups (n = 6): (Group 1): Control group, (Group 2): Six male rats were exposed to Dic 10 mg/kg intraperitoneally (I.P) for 28 days, (Group 3): Six male rats were exposed to Caf (15 mg/kg orally) for 28 days; (Groups 4): Six male rats were exposed to Dic (10 mg/kg, i.p) + Caf (15 mg/kg, orally) for 28 days. Histopathological study and various biological parameters were estimated among the four groups including hemoglobin (Hb%) red blood cells (RBCs), Hematocrit (HT%), total leucocyte count (WBCs), lipid peroxidation (LPO), glutathione peroxidase (GPx), alanine aminotransferase (ALT), aspartate aminotransferase (AST), urea, creatinine, tumor necrosis factor-α (TNF-α), and nitric oxide (NO). RESULTS: The administration of Diclofenac resulted in significant deteriorations in the histopathological findings and estimated biological parameters. Whereas, daily Caffeine administration ameliorated Diclofenac-induced toxicity in the kidney and liver by three mechanisms including antioxidant, anti-inflammatory, and DNA damage inhibition. CONCLUSION: The current study demonstrated the promising ameliorative and protective effects of Caffeine against Diclofenac-induced hepatic and renal injury.


Subject(s)
Caffeine , Diclofenac , Male , Rats , Animals , Diclofenac/toxicity , Caffeine/pharmacology , Liver , Chromosome Aberrations , Anti-Inflammatory Agents
13.
Front Neurosci ; 16: 1041461, 2022.
Article in English | MEDLINE | ID: mdl-36704003

ABSTRACT

Alzheimer's disease (AD) is a predominantly heterogeneous disease with a highly complex pathobiology. The presence of amyloid-beta (Aß) depositions and the accumulation of hyperphosphorylated tau protein remain the characteristic hallmarks of AD. These hallmarks can be detected throughout the brain and other regions, including cerebrospinal fluid (CSF) and the spinal cord. Microglia cells, the brain-resident macrophage type of the brain, are implicated in maintaining healthy brain homeostasis. The localized administration of primary healthy microglia (PHM) is suggested to play a role in mitigating AD hallmark depositions and associated cognitive dysfunction. Carbenoxolone (CBX) is the most common gap junction blocker. It cannot effectively cross the blood-brain barrier (BBB) under systemic administration. Therefore, localized administration of CBX may be a recommended intervention against AD by acting as an antioxidant and anti-inflammatory agent. This study aims to determine whether the localized intracerebroventricular (ICV) administration of PHM and CBX may act as an effective therapeutic intervention for AD neuroinflammatory type. In addition, this study also aims to reveal whether detecting AD hallmarks in the spinal cord and CSF can be considered functional and effective during AD early diagnosis. Male albino rats were divided into four groups: control (group 1), lipopolysaccharide (LPS)-induced AD neuroinflammatory type (group 2), ICV injection of LPS + isolated PHM (group 3), and ICV injection of LPS + CBX (group 4). Morris water maze (MWM) was conducted to evaluate spatial working memory. The brain and spinal cord were isolated from each rat with the collection of CSF. Our findings demonstrate that the localized administration of PHM and CBX can act as promising therapeutic approaches against AD. Additionally, Aß and tau toxic aggregates were detected in the spinal cord and the CSF of the induced AD model concomitant with the brain tissues. Overall, it is suggested that the ICV administration of PHM and CBX can restore normal brain functions and alleviate AD hallmark depositions. Detecting these depositions in the spinal cord and CSF may be considered in AD early diagnosis. As such, conducting clinical research is recommended to reveal the benefits of related therapeutic approaches compared with preclinical findings.

14.
Curr Res Physiol ; 4: 216-222, 2021.
Article in English | MEDLINE | ID: mdl-34746841

ABSTRACT

Early diagnosis of neurodegenerative diseases, especially Alzheimer's disease (AD), is essential for implementing the appropriate treatment protocols and controlling disease progression. Early AD diagnosis helps patients achieve the best therapeutic outcomes, lessening irreversible neurodegenerative damage and severe cognitive decline. The measurement of brain waves and structural modifications, including gray/white matter and brain volume, have recently been considered a promising approach for brain biometrics because of the inherent specificity, degree of confidentiality, and reproducibility. Brain printing biometrics (BPB) is thus becoming more commonly considered as tool for early AD detection. This review proposes using BPB as a tool for the detection of AD prior to the appearance of persistent hallmark depositions, including Aß and tau protein aggregations in different brain regions. It also describes BPB authentication, a method of implementation, as well as potential outcomes.

15.
eNeurologicalSci ; 21: 100290, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33200104

ABSTRACT

Serious of unpredictable drawbacks of Coronavirus 2019 (COVID-19) infectious disease caused by SARS-COV-2 on the nervous system, have been widely noticed among the huge number of infected people. It was found that this type of newly revolving pandemic infection mainly infects the human respiratory tract causing mild to moderate symptoms, however, the hidden door side of COVID-19 is via penetrating the brain, revealing a huge threat especially to elderly people who are more susceptible to its severe side effects and even death to more extent. Almost 80% of COVID-19 patients suffer from severe neurological manifestations including dizziness, headache, unconscious, irritability, dysfunction in smell, and taste accompanied by muscle fatigue. Herein, we are trying to address the direct neuroinvasive pathway of COVID-19 into human brain cells which is mainly through the olfactory route leading to long-term neurological complications. In addition to highlighting the ability of COVID-19 infection to intensify a pre-existing AD to a more prominent severe stage. The other thing to emphasize is whether AD patients with a highly prominent activation of local immune responses are more or less exposed to getting infected with COVID-19. Along with underlying the hypothesis that the susceptibility to COVID-19 infection may lead to a future risk for neurodegenerative diseases including Alzheimer's disease (AD).

SELECTION OF CITATIONS
SEARCH DETAIL