Next Article in Journal
Molecular Cloning and Functional Analysis of GmLACS2-3 Reveals Its Involvement in Cutin and Suberin Biosynthesis along with Abiotic Stress Tolerance
Next Article in Special Issue
Circulation of Nor98 Atypical Scrapie in Portuguese Sheep Confirmed by Transmission of Isolates into Transgenic Ovine ARQ-PrP Mice
Previous Article in Journal
Physicochemical Characterization and Drug Release Properties of Methyl-Substituted Silica Xerogels Made Using Sol–Gel Process
Previous Article in Special Issue
How an Infection of Sheep Revealed Prion Mechanisms in Alzheimer’s Disease and Other Neurodegenerative Disorders
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

microRNA-146a-5p, Neurotropic Viral Infection and Prion Disease (PrD)

by
Aileen I. Pogue
1 and
Walter J. Lukiw
2,3,4,*
1
Alchem Biotech, Toronto, ON M5S 1A8, Canada
2
LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans, LA 70112, USA
3
Department of Ophthalmology, Louisiana State University Health Science Center, New Orleans, LA 70112, USA
4
Department of Neurology, Louisiana State University Health Science Center, New Orleans, LA 70112, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(17), 9198; https://doi.org/10.3390/ijms22179198
Submission received: 1 July 2021 / Revised: 9 August 2021 / Accepted: 16 August 2021 / Published: 25 August 2021
(This article belongs to the Special Issue Prions and Prion Diseases 2.0)

Abstract

:
The human brain and central nervous system (CNS) harbor a select sub-group of potentially pathogenic microRNAs (miRNAs), including a well-characterized NF-kB-sensitive Homo sapiens microRNA hsa-miRNA-146a-5p (miRNA-146a). miRNA-146a is significantly over-expressed in progressive and often lethal viral- and prion-mediated and related neurological syndromes associated with progressive inflammatory neurodegeneration. These include ~18 different viral-induced encephalopathies for which data are available, at least ~10 known prion diseases (PrD) of animals and humans, Alzheimer’s disease (AD) and other sporadic and progressive age-related neurological disorders. Despite the apparent lack of nucleic acids in prions, both DNA- and RNA-containing viruses along with prions significantly induce miRNA-146a in the infected host, but whether this represents part of the host’s adaptive immunity, innate-immune response or a mechanism to enable the invading prion or virus a successful infection is not well understood. Current findings suggest an early and highly interactive role for miRNA-146a: (i) as a major small noncoding RNA (sncRNA) regulator of innate-immune responses and inflammatory signaling in cells of the human brain and CNS; (ii) as a critical component of the complement system and immune-related neurological dysfunction; (iii) as an inducible sncRNA of the brain and CNS that lies at a critical intersection of several important neurobiological adaptive immune response processes with highly interactive associations involving complement factor H (CFH), Toll-like receptor pathways, the innate-immunity, cytokine production, apoptosis and neural cell decline; and (iv) as a potential biomarker for viral infection, TSE and AD and other neurological diseases in both animals and humans. In this report, we review the recent data supporting the idea that miRNA-146a may represent a novel and unique sncRNA-based biomarker for inflammatory neurodegeneration in multiple species. This paper further reviews the current state of knowledge regarding the nature and mechanism of miRNA-146a in viral and prion infection of the human brain and CNS with reference to AD wherever possible.

1. Introduction and Overview

Multiple independent research laboratories have reported the significant upregulation of a specific sub-group of pathogenic microRNAs (miRNAs) in progressive, neuro-inflammatory, incapacitating and often lethal neurodegenerative diseases of the human brain and CNS. As soluble, amphipathic regulatory molecules, miRNAs are important post-transcriptional and epigenetic regulators of messenger RNA (mRNA) abundance, speciation and complexity [1,2]. These microRNAs: (i) exist as ~18- to ~25-ribonucleotide (nt), single-stranded noncoding RNAs (sncRNAs) whose sequences are both unique and highly selected over evolution; (ii) represent the smallest information-carrying ribonucleic acids yet defined; (iii) have been repeatedly shown to play critical and determinant roles in the onset and propagation of many human CNS disorders, including progressive, incapacitating and terminal neurological syndromes; and (iv) are fascinating molecular entities because of their somewhat unconventional origin, their ribonucleotide sequence characteristics, their ability to regulate multiple cellular processes in health and disease, their immense potential in disease therapeutics and evolutionary dynamics [1,3,4,5,6,7]. Regarding the evolutionary aspects of miRNA and miRNA sequence complexity and selection, from mathematical considerations alone, a ~22 nt sncRNA with the possibility of four ribonucleotides at each position (A, C, G and U—adenine, cytosine, guanine and uracil, respectively) has the potential to generate an exponentiation of 4 to the power of 22 or about 1.76 × 1013 unique miRNA species. However, genome-wide analysis of all known human miRNAs, with a current total of ~2650 individual species, indicates that the RNA sequence of human miRNAs has been highly selected from many different RNA sequence possibilities. Interestingly, some miRNAs have been shown to maintain their exact or highly homologous miRNA ribonucleotide sequence between plants and animals over ~1.5 billion years of evolution (the Arabidopsis thalianaHomo sapiens divergence), indicating that highly conserved ribonucleotide sequence-mediated genetic regulatory functions are attributable to the same miRNAs and miRNA binding proteins over vast periods of time [1,8,9]. Further RNA-sequencing and array-based analyses have indicated that only certain miRNAs, probably about 25–30 individual miRNA species: (i) are abundant in the cytoplasm of the human brain as well as retinal and other CNS cells; (ii) are inducible by pathological factors, such as pro-inflammatory cytokines and chemokines; and (iii) are upregulated by different types of pathogenic microbes, including viral gene-encoded products and highly neurotoxic secreted bacterial exudates such as lipopolysaccharide (LPS) [5,10,11] see below.

2. Homo sapiens miRNA-146a-5p (hsa-miRNA-146a-5p) and Mechanism of Action

Amongst these brain- and CNS-expressed microRNAs, one of the most well-studied microRNA species known to be involved in the pathogenesis of progressive, age-related neurological diseases is an inducible Homo sapiens microRNA-146a-5p (hsa-miRNA-146a-5p; hsa-miRNA-146a; or simply miRNA-146a) whose significant upregulation is currently implicated in every prion disease (PrD) in humans and animals analyzed to date [12,13,14] (see below); in at least 18 viral-induced encephalopathies, Alzheimer’s disease (AD) [15,16,17] and multiple neurological disorders that include cerebrovascular disease (CVD) [3,18], traumatic brain injury (TBI) [19], temporal lobe epilepsy (TLE) [20], age-related macular degeneration (AMD) [10,11,21,22], amyotrophic lateral sclerosis (ALS) [3,23], peripheral neuropathies and neurological tumors of the CNS [3] and neuro-immune diseases such as myasthenia gravis (MG) and multiple sclerosis (MS) [24] (Table 1).
In both Homo sapiens and Mus musculus microRNA-146a-5p (hsa-miRNA-146a and mmu-miRNA-146a, respectively) are 22-nucleotide (40.9%G+C) single-stranded small noncoding RNAs (sncRNA; 5′-UGAGAACUGAAUUCCAUGGGUU-3′; miRBase Acc-ession MIMAT0000449 and MI0000170, respectively; https://www.mirbase.org/cgi-bin/mature.pl?mature_acc=MIMAT0000449; http://www.mirbase.org/cgi-bin/mirnaentry.pl?acc=MI0000170; https://www.Genecards.org/cgi-bin/carddisp.pl?gene=MIR-146A last accessed on 23 August 2021; ad-ditional hsa miRNA-146a-5p sequence and associated disease-relevant biological data in AD are also available at Alzheimer Disease disease: Malacards—Research Articles, Drugs, Genes, Clinical Trials; https://www.malacards.org/card/alzheimer_disease?search=146a; last accessed on 23 August 2021).
Human and murine miRNA-146a are amongst the most intensively studied sncRNAs in all human and murine neurobiology, neuropathology, neurodegenerative disease and transgenic (Tg) murine models of these diseases. The equivalent and preserved 100% identical RNA sequence of miRNA-146a between Homo sapiens and Mus musculus: (i) attests to the basal importance of this sncRNA from an evolutionary perspective and (ii) is highly useful in molecular-genetic comparative and modelling studies and considerations, because this sncRNA has an identical RNA sequence in both species. Encoded from a single locus at chromosome 5q33.3 in humans (https://www.genecards.org/cgi-bin/carddisp.pl?gene=MIR146A accessed on 16 August 2021; human genome version GRCh38/hg38) and at chromosome 11q in mice, human miRNA-146a-5p has three tandem NF-KB (p50/p65) binding sites in its immediate upstream 5′ promoter. This miRNA can be rapidly induced by NF-kB (50/p65) and pathological mechanisms that upregulate NF-kB, such as biophysical and biochemical inducers, including hypoxia, reactive oxygen species (ROS), bacterial endotoxins and lipopolysaccharides (LPS), amyloid peptides, cytokines and other cell stressors [63,64,65]. Importantly miRNA-146a has a relatively short half-life of about 1.5–2 h in human brain cells and tissues; however, the stability of this sncRNA may be extended under nonphysiological circumstances and/or in certain subcellular compartments [39,65].
miRNA-146a, a unique, relatively abundant and distinctive member of the microRNA gene family, was originally described as being significantly induced and upregulated after microbial endotoxin, lipopolysaccharide (LPS) and/or cytokine stimulation of human THP1 cells, monocytes originally derived and immortalized from an acute monocytic leukemia patient of the M5 subtype [7,66]. THP1 cells are microglia-like and phagocytic for both latex beads and sensitized erythrocytes, lack cell surface and cytoplasmic immunoglobulins and are involved in the clearance of the 42 amino acid amyloid beta (Aβ42) peptide, a major biomarker for AD. It was subsequently found that the induction of this endotoxin-responsive microRNA is under transcriptional control by NF-kB (p50/p65); shortly thereafter, this inducible, pro-inflammatory miRNA-146a was found to be upregulated by metal sulfate-generated reactive oxygen species (ROS); by pro-inflammatory cytokines, such as IL-1β and TNFα; by bacterial endotoxins, such as LPS and fragilysin; by Aβ42 peptides; by inflammatory cocktails containing IL-1β and Aβ42 peptides together, in stressed human primary neuronal–glial (HNG) cells in primary culture, in stressed human brain-derived microglial (HMG) cells; and by many different strains of prions and neurotropic viruses [7,16,21,40,53,63,66] (see Table 1). Importantly, miRNA-146a is also found to be moderately abundant in the aging human brain and CNS and the immune cells of mice and humans where its over-expression during neurodegenerative disease contributes to astroglial proliferation and astrogliosis, cytokine overexpression, deficits in the innate-immune response and the initiation of inflammatory events leading to dysfunctional neurons, synaptic deficits and eventually neuronal cell atrophy and brain cell death [13,21,40,55,67].
Similar to all microRNAs, the major mechanism of miRNA genetic and neurobiological activity is to ‘seek out’ and interact via base pair recognition, complementarity and noncovalent hydrogen binding within the 3′-untranslated region (3′-UTR) of its target mRNA 3′-UTRs, and, in doing so, it decrease the capability of the specific mRNA to be expressed [2,68,69]. All metazoan miRNAs appear to initially recognize their target mRNAs by recognition of a ‘seed region’ or ‘seed sequence’ in the 3′-UTR; this ‘seed region’ is a conserved heptametrical sequence co-localized at positions 2–7 from the 5′-end of the miRNA. Even though base pairing of miRNA and its target mRNA often does not perfectly match, the ‘seed sequence’ is perfectly complementary [2,5,11]. In human neurodegenerative disease, miRNA-146a has several known relevant and verified targets that include the 3′-UTRs of: (i) the complement factor H (CFH) mRNA involved in inflammation and the innate immune response; (ii) the membrane-spanning TSPAN-12 protein involved in amyloidogenesis and the clearance of amyloid beta peptides from brain cells; and (iii) the interleukin-1 receptor kinase IRAK-1 (with a compensatory increase in IRAK-2) involved in pathological NF-kB accumulation, signaling and neuro-inflammation [2,68,69,70,71]. Upregulated miRNA-146a has been definitively linked to the downregulation in the expression of CFH, TSPAN-12 and IRAK-1 both in HNG cells in primary culture and in AD brain [10,11,64,69].

3. Neurotropic Viral-mediated Induction of hsa-miRNA-146a-5p

One truly remarkable and widely observed phenomenon concerning hsa-miRNA-146a-5p is that this inducible sncRNA is significantly upregulated by at least 18 neurotropic DNA and RNA viruses that infect the human brain, CNS, immune, lymphatic and hepatic and/or circulatory systems. In alphabetical order, these include (i) Borna encephalitis disease virus 1 (BoEDV-1; BDV; Mononegavirales; (−)ssRNA genome; [26,27]; (ii) Chikungunya virus (CHIKV; Togaviridae; (+)ssRNA genome; [28]; (iii) enterovirus 71 (EV71; Picornaviridae; (+)ssRNA genome; [29]; (iv) Epstein–Barr virus (EBV; Herpesviridae; dsDNA genome; [30]; (v) hantavirus (HTV; Bunyaviridae; (−)ssRNA genome; [31]; (vi) hepatitis A virus (HAV; Picornaviridae; (+)ssRNA genome; [32]; (vii) hepatitis B virus (HBV; Hepadnaviridae; dsDNA genome; [33,34,35,36,72]; (viii) hepatitis C virus (HCV; Flaviviridae; (+)ssRNA genome; [37]; (ix) herpes simplex virus-1 (HSV-1; Herpesviridae; dsDNA genome; [38,39,41]; (x) Hendra (Henipavirus) virus (HeV; Paramyxoviridae; (−)ssRNA genome; [42]; (xi) human immunodeficiency virus (HIV; Retroviridae; (+)ssRNA genome; [43]; (xii) human influenza A viruses (H1N1/H3N2; Orthomyxoviridae; (+)ssRNA genome; [44,45]; (xiii) early human papillomavirus virus 16 (eHPV-16; Papillomaviridae; dsDNA genome; [46]; (xiv) human severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2; the causative agent of COVID-19 disease; genus Betacoronavirus of the family Coronaviridae; (+)ssRNA genome; [47]; unpublished data; (xv) human T-cell leukemia (lymphotropic) virus type 1 (HTLV-1; Retroviridae; (+)ssRNA genome; [48]; (xvi) Japanese encephalitis virus (JEV; Flaviviridae; (+)ssRNA genome; [49,50]; (xvii) Kaposi’s sarcoma-associated herpesvirus (KSHV; Herpesviridae; dsDNA genome; [51]; and (xviii) severe fever with thrombocytopenia syndrome virus (SFTSV; Bunyaviridae; (−)ssRNA genome; [27,52] (Table 1). Whether the significant induction of host miRNA-146a after viral invasion and/or successful infection is a protective mechanism of the human host cell or a strategy used by the virus for invasion and productive replication is currently not well understood [16,21,35,40,49,53,73,74]. Regardless of its innate-immune, inflammatory signaling, pro- or anti-viral signaling function miRNA-146a has been shown to be in part compartmentalized and packaged into exosomes (EXs) and/or extracellular microvesicles (EMVs); after these EXs and/or EMVs are released from donor cells, they may be taken up by recipient cells and function in the modulation and/or spread of miRNA-146a-mediated gene expression signaling during either viral or prion infection [35,53]. Interestingly, all of the many types of viral infections that induce miRNA-146a are associated with specific neurological disease symptoms and/or syndromes that are progressive, age-related, insidious, incapacitating and often lethal. It is clear that the ubiquity of miRNA-146a upregulation in neurotropic viral infection indicates that this pro-inflammatory miRNA lies at a critical intersection of several important neuro-biological immune-response processes with highly interactive associations affecting the following: (i) Toll-like receptor signaling pathways; (ii) the inflammatory and innate- immune response; (iii) cytokine storms involving cytokine IL-1β, TNFα and chemokine production, most often as complex pro-inflammatory cocktails in neural cells and tissues; (iv) morphological change in neuronal cytoarchitecture and apoptosis; and (v) neural cell decline and neuronal cell demise [4,16,45,53,75,76].
Recent evidence has indicated that, in general, endogenous human miRNAs (small, single-stranded ~22 nt RNAs, such as miRNA-146a) might have a significant antiviral role and hence may be useful to the host in an innate-immune defense by targeting the single-stranded viral ribonucleic acid (ssvRNA) genomes of several different neurotropic viruses, such as SARS-CoV-2, and function to downregulate or modulate their expression [77,78]. To this end, and to quote just one recent example, RNA sequencing and other analytical genetics in RNA-based sequence studies have shown that at least ~160 of the ~2650 of the 18–22 nt naturally occurring human single-stranded miRNAs have perfect complementarity within the miRNA-mRNA ‘seed region’ of the SARS-CoV-2 ssvRNA genome, and these include the unique hsa-miRNA-146a ribonucleotide sequence and those of other related microRNA gene families [47,73,79,80,81]. Interestingly, the apparent lack of nucleic acids detectable within the prion particle indicates that some yet poorly understood pathological mechanism is responsible for the prion-mediated upregulation of miRNA-146a when neurons are confronted with infection by these neurological-disease inducing sialoglycoproteins [16,53,60,75,82,83].

4. Prion Disease (PrD) Upregulates hsa-miRNA-146a-5p

Prion diseases (PrD), coined from the term ‘proteinaceous infectious particle’ represent a group of progressive, transmissible and incurable spongiform encephalopathies (TSEs) comprising a small family of relatively rare, rapid onset and consistently fatal neurodegenerative disorders affecting both animals and humans [25,75]. TSEs are characterized by: (i) decreased activity of cholinergic and gamma-amino butyric acid (GABA) pathway-related enzymes, while adrenergic pathways are relatively spared; (ii) distinctive spongiform changes in the neocortex and other anatomical areas of brain tissues; (iii) association with long prodromal and incubation periods; (iv) synaptic and dendritic damage and dysfunctional connectivity in the neocortex; (v) progressive neuronal atrophy and loss; (vi) gliosis and the unusually rapid proliferation of neuroglial cells; (vii) an atypical inflammatory response in neural tissues, which further stimulates spongiform change; and (viii) the induction of sncRNAs, including miRNA-146a [3,16,53,59,60,75,83,84,85]. Naturally occurring TSEs of herbivorous mammals of the orders Ruminantia and Artiodactyla have been considered the ‘prototype’ of prion disease, and these include scrapie of sheep and goats (both of the family Bovidae, subfamily Caprinae) and bovine spongiform encephalopathy (BSE, or ‘mad cow disease’) of cattle (family Bovidae, subfamily Bovinae). These animal diseases of the family Bovidae are closely related in their molecular and genetic neurobiology, pathology and clinical presentation to the human neurological disorders Gerstmann–Sträussler–Scheinker syndrome (GSS) and Creutzfeldt–Jakob disease (CJD) [3,16,25,53,56,57,58,59,75,86,87]; https://www.nhs.uk/conditions/creutzfeldt-jakob-disease-cjd/; last accessed on 23 August 2021; see Table 1).
Largely because of their unusual and novel nature, PrDs have been intensively studied and are known to be caused by a misfolded isoform of a ubiquitous and highly conserved brain-, CNS- and PNS-enriched cellular prion sialoglycoprotein known as PrPc. The PrPc monomer is a ~209 amino acid (~200 kDa) glycosylated cell surface polypeptide, containing a predominant internal α-helical region, encoded in humans at chr 20p13. Normally, the constitutively expressed PrPc appears to be involved in neuritogenesis, neuronal homeostasis, cell signaling, cell–cell adhesion and interaction and intercellular communication; moreover, it may provide a protective role against multiple forms of induced physiological stress [56,60,75,84,86,88,89]. The misfolded, abnormal and insoluble isoform of PrPc known as PrPsc self-associates into pro-inflammatory, protease-resistant aggregates that are insoluble in most detergents and chaotropic agents [59,75,84,89,90,91]. The molecular mechanisms of PrPsc neurotoxicity that drive the initiation, development and progression of PrD are highly complex and, similar to the case of AD, increased oxidative stress and chronic inflammation appear to be critically involved in the initiation and progression of PrD [5,73,86,92,93,94,95]. Typically, activated microglia accumulate within the immediate vicinity of abnormal PrPsc aggregates, and they release cytokines such as IL-1β that play important roles in the inflammatory pathogenesis of PrD, including the upregulation of genes that promote pro-inflammatory signaling and innate-immune system deficits [86,90]. As PrPsc aggregates progressively form over time, they further induce inevitably fatal neurodegenerative disease conditions, including neuroinflammation, which is typically discernable by massive microglial activation and proliferation and the subsequent and self-reinforcing upregulation of cytokines, such as TNF-alpha (TNFα), interleukin 1 alpha (IL-1α) and glial fibrillary acidic protein (GFAP) as well as astrogliosis accompanied by multiple additional pathogenic alterations in the neuronal transcriptome [25,59,67,85,86,96]. The unique primary amino acid sequence of the heavily glycosylated cellular prion PrPc sialoglycoprotein appears to predispose this relatively small molecule to non-homeostatic and pathological secondary and/or tertiary folding changing or ‘flipping’ its spatial conformation into the disease-causing PrPsc protease-resistant isotype [56,75,97]. An analogous situation may occur in the unique primary amino acid sequence of the 42 amino acid human amyloid-beta (Aβ42) peptide that also accumulates as a protease-resistant polymer that progressively accumulates in AD-affected brain [98,99].
Human prion diseases currently include Creutzfeldt–Jacob disease (CJD), stratified into sporadic CJD (sCJD) and variant CJD (vCJD) clinical subtypes, Gerstmann–Sträussler–Scheinker syndrome (GSS), fatal familial insomnia (FFI) and kuru, a fatal neurological disease found among natives from New Guinea who practiced a form of ritual cannibalism in which they consumed the brains of their predecessors [56,57,58,59,60,97]. Animal prion diseases include scrapie in sheep and goats (Ruminants of the order Artiodactyla, family Bovidae, subfamily Caprinae), bovine spongiform encephalopathy (BSE, also known as mad cow disease) in cattle (also of the family Bovidae), chronic wasting disease (CWD) in cervids (family Cervidae), transmissible mink encephalopathy (TME) in mink (family Mustelidae) and feline spongiform encephalopathy (FSE) in cats (family Felidae; [56,67,75]). Atypical and novel human prion diseases in Chordata continue to emerge, such as the recently identified camel prion disease (CPD) in dromedary camels observed for the first time in Algeria (order Artiodactyla, family Camelidae) [100]). Prion diseases can also be experimentally studied via the inoculation of brain and other PrPsc-containing extracts into laboratory animals such as mice, voles, gerbils and hamsters, causing a recapitulation of the PrD and TLE in sensitive animals that can be further studied, analyzed and carefully investigated in a biohazard safety level 2 or 3 (BSL-2 or BSL-3) laboratory (see Handling Prions-Environmental Health and Safety, Michigan State University (msu.edu); www.ehs.msu.edu/lab-clinic/bio/handling-prions.html; last accessed on 23 August 2021; [25,67,82,83,84,90]; (Table 1).
Shortly after the first reports of a significantly upregulated miRNA-146a in AD-affected brain and IL-1β-, TNFα- and/or Aβ42 peptide-stressed human neuronal–glial (HNG) cells (transplantation grade) in primary co-culture, the increased abundance of this same pro-inflammatory sncRNA was reported by multiple groups in animal and human nervous tissues affected with PrD [10,54,55,63,75,101]. As a pro-inflammatory sncRNA, over the last ~15 years, miRNA-146a has been repeatedly shown to participate in the regulation of adaptive and innate-immune systems and cytokine-mediated pro-inflammatory responses that potentially culminate in uncontrolled neural tissue damage [24,33,67,69,102,103]. miRNA-146a upregulation in transfected co-cultures of neuronal–glial cells can downregulate both CFH mRNA and protein levels via miRNA-146a pairing with 3′-UTR of human CFH, a finding also observed in multiple murine transgenic models for neural degeneration and in human AD, AMD, MS and TLE [10,22,24,104]. Given that CFH downregulation in neural tissues plays an important role in complement system regulation, immunological signaling and neural cell demise, and since miRNA-146a is the main sncRNA-regulator of neuro-inflammatory responses in neural tissues: (i) it is generally accepted that miRNA-146a is critical in the pathogenesis of inflammatory neurodegeneration in multiple forms of immune-related prion diseases, AD and neurotrophic viral infection; and (ii) this may be informative for using this microRNA as a general early diagnostic biomarker for multiple forms of these insidious brain diseases [2,4,55].

5. Unanswered Questions—Looking Backward and Forward

Since the initial description of scrapie in goats and sheep in about 1750 AD [57,105], there has been a steady emergence of the recognition, identification and characterization of a series of TSEs that now include (in alphabetical order) BSE, CPD, CWD and TME in animals and BSE-like disease, CJD, FFI, GSS and kuru in humans [25,56,60,72,75,82,96,106]. It has been about 64 years since kuru was initially reported to Western medicine, the first described TSE of humans for which the world renown anthropologist, biochemist and virologist D. Carleton Gajdusek was awarded the Nobel Prize in Medicine in 1976 [57,60,72]. At that time, both scrapie and kuru were thought to be caused by a ‘slow virus’ or ‘infectious protein’ that took many years or decades for these lethal neurological disorders to develop in the mammalian brain and CNS [56,59,105]. Twenty-one years later, the biochemist and neurologist Stanley B. Prusiner was awarded the Nobel prize in Medicine in 1997 for the isolation, characterization and proof of transmissibility of novel nucleic-acid-free ‘prion particles’ in susceptible animal models [25,56,72,75]. The relatively recent discovery of camel prion disease (CPD; ‘mad camel disease’) in the Middle East suggests: (i) that there may be yet other novel TSEs of animals and humans awaiting our discovery and/or; (ii) that mammalian central nervous systems are still in the process of evolving prion-like entities in the expanding spectrum of prion-induced disease [56,75,100]. Serious incapacitating, progressive, age-related and lethal neurological diseases such as AD, first described by the neurologist and physician Alois Alzheimer in 1906, have been suggested to be caused by transmissible, ‘infectious’ prion particles, which, depending on multiple biophysical factors, may adopt alternative conformations that are both self-propagating and found in a very wide array of organisms ranging from yeast to humans [56,75].
Many questions remain unanswered concerning the role of the inducible host miRNA-146a-5p that is upregulated during prion infection as well as by many other types of DNA and RNA viruses and neurodegenerative disease syndromes that include AMD, AD, ALS, CVD, peripheral neuropathies and tumors of the CNS, TLE and TBI (Table 1). Interestingly, the related human microRNA hsa-miRNA-146a-3p does not appear to be significantly upregulated under similar conditions. Our understanding of the potential pathological role of miRNA-146a in AD, prion disease and/or neurotropic viral infection began only about 15 years ago [54,55,61,101]. The list of miRNA-146a participation in prion disease, viral infections of the brain and CNS and related neurodegenerative diseases such as AD is an expanding one. Very fundamental neurobiological questions requiring additional research investigation include the following: (i) Does miRNA-146a always contribute to neuro-pathological, neuro-inflammatory and altered neuro-immunological aspects of PrD, viral infection and AD, and/or are other sncRNAs or transmissible particulate species involved?; (ii) As pathological and molecular genetic processes associated with neurological disorders often precede clinical symptoms, might miRNA-146a be useful as an early clinical diagnostic and/or prognostic biomarker for viral and/or prion disease and/or AD or other related neurological disorders?; (iii) Is the rapid increase in miRNA-146a upon prion or viral infection advantageous to the host via an innate-immunity-mediated mechanism, are upregulated miRNAs part of the invading prion or a viral strategy for a more efficient invasion and infection, or are both of these biomolecular and/or immunological scenarios plausible?; (iv) Would both of these possibilities be expected to involve miRNA-146a-specific mRNA targets and modulation of gene expression signaling to ultimately alter the transcriptome of the neuron and/or neural support cells?; (v) Are other transcription factors besides NF-κB and other pro-inflammatory microRNAs besides miRNA-146a involved in initiating, driving or modulating prion- and/or viral-directed neuro-degeneration?; (vi) Are anti-NF-kB (p50/p65)- and/or anti-miRNA-146a-5p-based therapeutic strategies clinically feasible to be deployed, and are they suitable to address and successfully treat the broad spectrum of human neurological disorders and other neurodegenerative disease syndromes initiated by neurotropic DNA and RNA viruses and/or prions?; and (vii) Would these same pharmacological strategies and treatments be useful in the clinical management of viral infection, prion disease, AD and/or perhaps other progressive, neurological disorders associated with aging and progressive degeneration of the brain and CNS?

6. Conclusions

Among the most significant advances in human neuroscience, neurology and molecular neurogenetics over the last fifteen years are: (i) the discovery of a family of small noncoding single-stranded RNAs called microRNAs in the mammalian brain and central nervous system (CNS) and (ii) the analysis and categorization of their abundance, speciation and complexity in development, aging and in neurological health and CNS disease [2,5,6,9,70,71,93,101,107]. A growing body of evidence indicates that select species of the 2650 member human miRNA gene family are brain-abundant and participate in the initiation, propagation and development of insidious age-related neurological disorders of the mammalian brain and CNS. This includes the involvement of a unique pro-inflammatory miRNA-146a in a broad spectrum of viral- and prion-induced encephalopathies and related progressive age-related neurodegenerations of the human brain that include, prominently, AD, ALS, AMD, MS, TLE, scrapie and BSE (mad cow disease) as well as CJD, GSS and kuru. miRNA-146a’s role and significance in viral-induced encephalopathies and prion disease appear to be expanding. Several attractive and all-encompassing recently proposed theories suggest: (i) that there is a contribution of prions and misfolded proteins in human degenerative diseases that include AD and AMD and other miRNA-146a-associated neurological disorders [56,75,108]; and (ii) that there may be a gastrointestinal (GI)-tract-sourced contribution of microbes or microbial neurotoxins to AD and related neurodegenerative disorders that critically involve miRNA-146a-mediated immunological and/or pro-inflammatory signaling components [108,109,110,111].

Author Contributions

A.I.P. and W.J.L. compiled and refined the results from all laboratory experiments and those of many collaborators, both domestic and international, and performed literature searches of recent peer-reviewed publications in the microRNA area of neurobiological research; W.J.L. wrote the article. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported through an unrestricted grant to the LSU Eye Center from Research to Prevent Blindness (RPB), The Brown Foundation, Joe and Dorothy Dorsett Innovation in Science Healthy Aging Award; the Louisiana Biotechnology Research Network (LBRN); and NIH grants NEI EY006311, NIA AG18031 and NIA AG038834 (W.J.L.).

Institutional Review Board Statement

The scientific material in this review paper were obtained, compiled and synthesized from the 111 References listed at the end of this paper; ethical review and approval were waived in this study as this review papers is simply a compilation of scientific facts.

Data Availability Statement

All data used in this review are openly available and freely accessible on MedLine (www.ncbi.nlm.nih.gov accessed on 16 August 2021) where they are listed by the last names of the individual authors.

Acknowledgments

The research in this article was presented in part at the Society for Neuroscience (SFN) Annual Meeting, Chicago, IL, USA, November 2019. Sincere thanks are extended to L Cong, C Eicken, K Navel, W Poon, T Bond, E Head, Y Zhao and the late PN Alexandrov, JM Hill and HM Wisniewski for helpful discussions in this research area and to D Guillot for expert technical assistance and medical artwork and general organization. Research on microRNAs, ethnobiology, botanical neurotoxins, pro-inflammatory and pathogenic signaling in the Lukiw laboratory involving the microbiome’ the innate immune response, amyloidogenesis, synaptogenesis and neuro-inflammation in AD, prion and in other human neurological- and plant-viroid-based diseases was supported through an unrestricted grant to the LSU Eye Center from Research to Prevent Blindness (RPB), The Brown Foundation, Joe and Dorothy Dorsett Innovation in Science Healthy Aging Award; the Louisiana Biotechnology Research Network (LBRN); and NIH grants NEI EY006311, NIA AG18031 and NIA AG038834 (WJL). The content of this manuscript is solely the responsibility of the authors and does not necessarily represent the official views of the National Institute on Aging, the National Center for Research Resources or the National Institutes of Health.

Conflicts of Interest

The authors declare that this research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Arteaga-Vázquez, M.; Caballero-Pérez, J.; Vielle-Calzada, J.P. A family of microRNAs present in plants and animals. Plant Cell 2006, 18, 3355–3369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Bartel, D.P. Metazoan microRNAs. Cell 2018, 173, 20–51. [Google Scholar] [CrossRef] [Green Version]
  3. Fan, W.; Liang, C.; Ou, M.; Zou, T.; Sun, F.; Zhou, H.; Cui, L. microRNA-146a is a wide-reaching neuro-inflammatory regulator and potential treatment target in neurological diseases. Front. Mol. Neurosci. 2020, 13, 90. [Google Scholar] [CrossRef] [PubMed]
  4. Gaudet, A.D.; Fonken, L.K.; Watkins, L.R.; Nelson, R.J.; Popovich, P.G. MicroRNAs: Roles in regulating neuroinflammation. Neuroscientist 2017, 24, 221–245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Khan, I.; Preeti, K.; Fernandes, V.; Khatri, D.K.; Singh, S.B. Role of MicroRNAs, aptamers in neuroinflammation and neurodegenerative disorders. Cell. Mol. Neurobiol. 2021, 1–21. [Google Scholar] [CrossRef]
  6. Kinser, H.E.; Pincus, Z. MicroRNAs as modulators of longevity and the aging process. Qual. Life Res. 2019, 139, 291–308. [Google Scholar] [CrossRef]
  7. Taganov, K.D.; Boldin, M.; Chang, K.-J.; Baltimore, D. NF- B-dependent induction of microRNA miRNA-146a, an inhibitor targeted to signaling proteins of innate immune responses. Proc. Natl. Acad. Sci. USA 2006, 103, 12481–12486. [Google Scholar] [CrossRef] [Green Version]
  8. Avsar, B.; Zhao, Y.; Li, W.; Lukiw, W.J. Atropa belladonna expresses a microRNA (aba-miRNA-9497) highly homologous to Homo sapiens miRNA-378 (hsa-miRNA-378); both miRNAs target the 3′-untranslated region (3′-UTR) of the mRNA encoding the neurologically relevant, zinc-finger transcription factor ZNF-691. Cell. Mol. Neurobiol. 2019, 40, 179–188. [Google Scholar] [CrossRef]
  9. Kinoshita, C.; Kubota, N.; Aoyama, K. Interplay of RNA-binding proteins and microRNAs in neurodegenerative diseases. Int. J. Mol. Sci. 2021, 22, 5292. [Google Scholar] [CrossRef]
  10. Alexandrov, P.N.; Dua, P.; Lukiw, W.J. Up-regulation of miRNA-146a in progressive, age-related inflammatory neurodegen-erative disorders of the human CNS. Front. Neurol. 2014, 5, 181. [Google Scholar] [CrossRef] [Green Version]
  11. Juźwik, C.A.; Drake, S.S.; Zhang, Y.; Paradis-Isler, N.; Sylvester, A.; Amar-Zifkin, A.; Douglas, C.; Morquette, B.; Moore, C.S.; Fournier, A.E. microRNA dysregulation in neurodegenerative diseases: A systematic review. Prog. Neurobiol. 2019, 182, 101664. [Google Scholar] [CrossRef]
  12. Asher, D.M.; Gregori, L. Human transmissible spongiform encephalopathies: Historic view. Handb. Clin. Neurol. 2018, 153, 1–17. [Google Scholar] [CrossRef] [PubMed]
  13. Lukiw, W.J.; Dua, P.; Pogue, A.I.; Eicken, C.; Hill, J.M. Upregulation of microRNA-146a (miRNA-146a), a marker for inflammatory neurodegeneration, in sporadic Creutzfeldt–Jakob disease (sCJD) and Gerstmann–Straussler–Scheinker (GSS) syndrome. J. Toxicol. Environ. Health Part A 2011, 74, 1460–1468. [Google Scholar] [CrossRef] [Green Version]
  14. Orge, L.; Lima, C.; Machado, C.; Tavares, P.; Mendonça, P.; Carvalho, P.; Silva, J.; Pinto, M.; Bastos, E.; Pereira, J.; et al. Neuropathology of animal prion diseases. Biomolecules 2021, 11, 466. [Google Scholar] [CrossRef]
  15. Stelzmann, R.A.; Schnitzlein, H.N.; Murtagh, F.R. An english translation of Alzheimer’s 1907 paper? Uber eine eigenartige erkankung der hirnrinde? Clin. Anat. 1995, 8, 429–431. [Google Scholar] [CrossRef]
  16. Lukiw, W.J. microRNA-146a signaling in Alzheimer’s disease (AD) and prion disease (PrD). Front. Neurol. 2020, 11, 462. [Google Scholar] [CrossRef]
  17. Yang, J.; Malone, F.; Go, M.; Kou, J.; Lim, J.-E.; Caughey, R.C.; Fukuchi, K.-I. Lipopolysaccharide-induced exosomal miR-146a is involved in altered expression of Alzheimer’s risk genes via suppression of TLR4 signaling. J. Mol. Neurosci. 2020, 71, 1245–1255. [Google Scholar] [CrossRef] [PubMed]
  18. Dirnagl, U.; Iadecola, C.; Moskowitz, M.A. Pathobiology of ischaemic stroke: An integrated view. Trends Neurosci. 1999, 22, 391–397. [Google Scholar] [CrossRef]
  19. Ma, S.-Q.; Xu, X.-X.; He, Z.-Z.; Li, X.-H.; Luo, J.-M. Dynamic changes in peripheral blood-targeted miRNA expression profiles in patients with severe traumatic brain injury at high altitude. Mil. Med. Res. 2019, 6, 12. [Google Scholar] [CrossRef]
  20. Roncon, P.; Soukupovà, M.; Binaschi, A.; Falcicchia, C.; Zucchini, S.; Ferracin, M.; Langley, S.R.; Petretto, E.; Johnson, M.R.; Marucci, G.; et al. MicroRNA profiles in hippocampal granule cells and plasma of rats with pilocarpine-induced epilepsy—Comparison with human epileptic samples. Sci. Rep. 2015, 5, 14143. [Google Scholar] [CrossRef] [Green Version]
  21. Hill, J.M.; Pogue, A.I.; Lukiw, W.J. Pathogenic microRNAs common to brain and retinal degeneration; recent observations in Alzheimer’s disease and age-related macular degeneration. Front. Neurol. 2015, 6. [Google Scholar] [CrossRef] [Green Version]
  22. Pogue, A.I.; Lukiw, W.J. Up-regulated pro-inflammatory microRNAs (miRNAs) in Alzheimer’s disease (AD) and age-related macular degeneration (AMD). Cell. Mol. Neurobiol. 2018, 38, 1021–1031. [Google Scholar] [CrossRef] [PubMed]
  23. Pegoraro, V.; Merico, A.; Angelini, C. Micro-RNAs in ALS muscle: Differences in gender, age at onset and disease duration. J. Neurol. Sci. 2017, 380, 58–63. [Google Scholar] [CrossRef] [Green Version]
  24. Devier, D.J.; Lovera, J.F.; Lukiw, W.J. Increase in NF-κB-sensitive miRNA-146a and miRNA-155 in multiple sclerosis (MS) and pro-inflammatory neurodegeneration. Front. Mol. Neurosci. 2015, 8, 5. [Google Scholar] [CrossRef] [Green Version]
  25. Prusiner, S. Novel proteinaceous infectious particles cause scrapie. Science 1982, 216, 136–144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Carbone, K.M. Borna disease virus and human disease. Clin. Microbiol. Rev. 2001, 14, 513–527. [Google Scholar] [CrossRef] [Green Version]
  27. Zhang, X.; Guo, Y.; Xu, X.; Tang, T.; Sun, L.; Wang, H.; Zhou, W.; Fang, L.; Li, Q.; Xie, P. miR-146a promotes Borna disease virus 1 replication through IRAK1/TRAF6/NF-κB signaling pathway. Virus Res. 2019, 271, 197671. [Google Scholar] [CrossRef] [PubMed]
  28. Selvamani, S.P.; Mishra, R.; Singh, S.K. Chikungunya virus exploits miR-146a to regulate NF-κB pathway in human synovial fibroblasts. PLoS ONE 2014, 9, e103624. [Google Scholar] [CrossRef]
  29. Ho, B.-C.; Yu, I.-S.; Lu, L.-F.; Rudensky, A.; Chen, H.-Y.; Tsai, C.-W.; Chang, Y.-L.; Wu, C.-T.; Chang, L.-Y.; Shih, S.-R.; et al. Inhibition of miR-146a prevents enterovirus-induced death by restoring the production of type I interferon. Nat. Commun. 2014, 5, 3344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Jonigk, D.; Izykowski, N.; Maegel, L.; Schormann, E.; Maecker-Kolhoff, B.; Laenger, F.; Kreipe, H.; Hussein, K. MicroRNA expression in Epstein-Barr virus-associated post-transplant smooth muscle tumours is related to leiomyomatous phenotype. Clin. Sarcoma Res. 2013, 3, 9. [Google Scholar] [CrossRef] [Green Version]
  31. Shin, O.S.; Kumar, M.; Yanagihara, R.; Song, J.-W. Hantaviruses induce cell type- and viral species-specific host microRNA expression signatures. Virology 2013, 446, 217–224. [Google Scholar] [CrossRef] [Green Version]
  32. Mo, L.; Zeng, Z.; Deng, R.; Li, Z.; Sun, J.; Hu, N.; Shi, J.; Hu, Y. Hepatitis A virus-induced hsa-miR-146a-5p attenuates IFN-β signaling by targeting adaptor protein TRAF6. Arch. Virol. 2021, 166, 789–799. [Google Scholar] [CrossRef]
  33. Li, J.-F.; Dai, X.-P.; Zhang, W.; Sun, S.-H.; Zeng, Y.; Zhao, G.-Y.; Kou, Z.-H.; Guo, Y.; Yu, H.; Du, L.-Y.; et al. Upregulation of microRNA-146a by hepatitis B virus X protein contributes to hepatitis development by downregulating complement factor h. mBio 2015, 6, e02459-14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Liu, Y.; Zhao, J.J.; Wang, C.M.; Li, M.Y.; Han, P.; Wang, L.; Cheng, Y.Q.; Zoulim, F.; Ma, X.; Xu, D.P. Altered expression profiles of microRNAs in a stable hepatitis B virus-expressing cell line. Chin. Med. J. 2009, 122, 10–14. [Google Scholar] [CrossRef] [PubMed]
  35. Nahand, J.S.; Karimzadeh, M.R.; Nezamnia, M.; Fatemipour, M.; Khatami, A.; Jamshidi, S.; Moghoofei, M.; Taghizadieh, M.; Hajighadimi, S.; Shafiee, A.; et al. The role of miRNA-146a in viral infection. IUBMB Life 2019, 72, 343–360. [Google Scholar] [CrossRef] [PubMed]
  36. Wang, S.; Zhang, X.; Ju, Y.; Zhao, B.; Yan, X.; Hu, J.; Shi, L.; Yang, L.; Ma, Z.; Chen, L.; et al. MicroRNA-146a feedback suppresses T cell immune function by targeting Stat1 in patients with chronic hepatitis. J. Immunol. 2013, 191, 293–301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Joshi, D.; Salehi, S.; Brereton, H.; Arno, M.; Quaglia, A.; Heaton, N.; O’Grady, J.; Agarwal, K.; Aluvihare, V. Distinct microRNA profiles are associated with the severity of hepatitis C virus recurrence and acute cellular rejection after liver transplantation. Liver Transpl. 2013, 19, 383–394. [Google Scholar] [CrossRef]
  38. Higaki, S.; Gebhardt, B.M.; Lukiw, W.J.; Thompson, H.W.; Hill, J.M. Gene expression profiling in the HSV-1 latently infected mouse trigeminal ganglia following hyperthermic stress. Curr. Eye Res. 2003, 26, 231–238. [Google Scholar] [CrossRef]
  39. Hill, J.M.; Zhao, Y.; Clement, C.; Neumann, D.M.; Lukiw, W.J. HSV-1 infection of human brain cells induces miRNA-146a and Alzheimer-type inflammatory signaling. Neuroreport 2009, 20, 1500–1505. [Google Scholar] [CrossRef] [Green Version]
  40. Hill, J.M.; Clement, C.; Zhao, Y.; Lukiw, W.J. Induction of the pro-inflammatory NF-kB-sensitive miRNA-146a by human neurotrophic viruses. Front. Microbiol. 2015, 6, 43. [Google Scholar] [CrossRef]
  41. Lukiw, W.J.; Cui, J.G.; Yuan, L.Y.; Bhattacharjee, P.S.; Corkern, M.; Clement, C.; Kammerman, E.M.; Ball, M.; Zhao, Y.; Sullivan, P.M.; et al. Acyclovir or Aβ42 peptides attenuate HSV-1-induced miRNA-146a levels in human primary brain cells. NeuroReport 2010, 21, 922–927. [Google Scholar] [CrossRef] [Green Version]
  42. Stewart, C.R.; Marsh, G.A.; Jenkins, K.A.; Gantier, M.; Tizard, M.; Middleton, D.; Lowenthal, J.W.; Haining, J.; Izzard, L.; Gough, T.J.; et al. Promotion of Hendra virus replication by microRNA 146a. J. Virol. 2013, 87, 3782–3791. [Google Scholar] [CrossRef] [Green Version]
  43. Duskova, K.; Nagilla, P.; Le, H.-S.; Iyer, P.; Thalamuthu, A.; Martinson, J.; Bar-Joseph, Z.; Buchanan, W.; Rinaldo, C.; Ayyavoo, V. MicroRNA regulation and its effects on cellular transcriptome in human immunodeficiency virus-1 (HIV-1) infected individuals with distinct viral load and CD4 cell counts. BMC Infect. Dis. 2013, 13, 250. [Google Scholar] [CrossRef] [Green Version]
  44. Chen, J.; Yuan, L.; Fan, Q.; Su, F.; Chen, Y.; Hu, S. Adjuvant effect of docetaxel on the immune responses to influenza A H1N1 vaccine in mice. BMC Immunol. 2012, 13, 36. [Google Scholar] [CrossRef] [Green Version]
  45. Terrier, O.; Textoris, J.; Carron, C.; Marcel, V.; Bourdon, J.-C.; Rosa-Calatrava, M. Host microRNA molecular signatures associated with human H1N1 and H3N2 influenza A viruses reveal an unanticipated antiviral activity for miR-146a. J. Gen. Virol. 2013, 94, 985–995. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Greco, D.; Kivi, N.; Qian, K.; Leivonen, S.-K.; Auvinen, P.; Auvinen, E. Human Papillomavirus 16 E5 modulates the expression of host microRNAs. PLoS ONE 2011, 6, e21646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Roganović, J.R. microRNA-146a and -155, upregulated by periodontitis and type 2 diabetes in oral fluids, are predicted to regulate SARS-CoV-2 oral receptor genes. J. Periodontol. 2020, 92. [Google Scholar] [CrossRef]
  48. Pichler, K.; Schneider, G.; Grassmann, R. MicroRNA miR-146a and further oncogenesis-related cellular microRNAs are dysregulated in HTLV-1-transformed T lymphocytes. Retrovirology 2008, 5, 100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Deng, M.; Du, G.; Zhao, J.; Du, X. miRNA-146a negatively regulates the induction of proinflammatory cytokines in response to Japanese encephalitis virus infection in microglial cells. Arch. Virol. 2017, 162, 1495–1505. [Google Scholar] [CrossRef]
  50. Pareek, S.; Roy, S.; Kumari, B.; Jain, P.; Banerjee, A.; Vrati, S. miR-155 induction in microglial cells suppresses Japanese encephalitis virus replication and negatively modulates innate immune responses. J. Neuroinflamm. 2014, 11, 97. [Google Scholar] [CrossRef] [Green Version]
  51. Punj, V.; Matta, H.; Schamus, S.; Tamewitz, A.; Anyang, B.; Chaudhary, P.M. Kaposi’s sarcoma-associated herpesvirus-encoded viral FLICE inhibitory protein (vFLIP) K13 suppresses CXCR4 expression by upregulating miRNA-146a. Oncogene 2009, 29, 1835–1844. [Google Scholar] [CrossRef] [Green Version]
  52. Zhang, L.; Fu, Y.; Wang, H.; Guan, Y.; Zhu, W.; Guo, M.; Zheng, N.; Wu, Z. Severe fever with thrombocytopenia syndrome virus-induced macrophage differentiation is regulated by miR-146. Front. Immunol. 2019, 10, 1095. [Google Scholar] [CrossRef]
  53. Lukiw, W.J.; Pogue, A.I. Vesicular transport of encapsulated microRNA between glial and neuronal cells. Int. J. Mol. Sci. 2020, 21, 5078. [Google Scholar] [CrossRef] [PubMed]
  54. Saba, R.; Goodman, C.D.; Huzarewich, R.L.C.H.; Robertson, C.; Booth, S.A. A miRNA signature of prion induced neurodegeneration. PLoS ONE 2008, 3, e3652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Slota, J.; Booth, S.A. MicroRNAs in Neuroinflammation: Implications in disease pathogenesis, biomarker discovery and therapeutic applications. Non-coding RNA 2019, 5, 35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Ayers, J.I.; Paras, N.A.; Prusiner, S.B. Expanding spectrum of prion diseases. Emerg. Top. Life Sci. 2020, 4. [Google Scholar] [CrossRef]
  57. Gajdusek, D.C. Slow-virus infections of the nervous system. N. Engl. J. Med. 1967, 276, 392–400. [Google Scholar] [CrossRef]
  58. Comoy, E.E.; Mikol, J.; Jaffré, N.; Lebon, V.; Levavasseur, E.; Streichenberger, N.; Sumian, C.; Perret-Liaudet, A.; Eloit, M.; Andreoletti, O.; et al. Experimental transfusion of variant CJD-infected blood reveals previously uncharacterized prion disorder in mice and macaque. Nat. Commun. 2017, 8, 1–13. [Google Scholar] [CrossRef]
  59. Gajdusek, D.C. Kuru and its contribution to medicine. Philos. Trans. R. Soc. B Biol. Sci. 2008, 363, 3697–3700. [Google Scholar] [CrossRef] [Green Version]
  60. Liberski, P.P.; Gajos, A.; Sikorska, B.; Lindenbaum, S. Kuru, the first human prion disease. Viruses 2019, 11, 232. [Google Scholar] [CrossRef] [Green Version]
  61. Saba, R.; Gushue, S.; Huzarewich, R.L.C.H.; Manguiat, K.; Medina, S.; Robertson, C.; Booth, S.A. MicroRNA 146a (miR-146a) is over-expressed during prion disease and modulates the innate immune response and the microglial activation state. PLoS ONE 2012, 7, e30832. [Google Scholar] [CrossRef] [PubMed]
  62. Clement, C.; Hill, J.M.; Dua, P.; Culicchia, F.; Lukiw, W.J. Analysis of RNA from Alzheimer’s disease post-mortem brain tissues. Mol. Neurobiol. 2015, 53, 1322–1328. [Google Scholar] [CrossRef] [Green Version]
  63. Lukiw, W.J.; Zhao, Y.; Cui, J.G. An NF-κB-sensitive micro RNA-146a-mediated inflammatory circuit in Alzheimer disease and in stressed human brain cells. J. Biol. Chem. 2008, 283, 31315–31322. [Google Scholar] [CrossRef] [Green Version]
  64. Cui, J.G.; Li, Y.Y.; Zhao, Y.; Bhattacharjee, S.; Lukiw, W.J. Differential regulation of interleukin-1 receptor-associated kinase-1 (IRAK-1) and IRAK-2 by microRNA-146a and NF-κB in stressed human astroglial cells and in Alzheimer disease. J. Biol. Chem. 2010, 285, 38951–38960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Pogue, A.I.; Li, Y.Y.; Cui, J.-G.; Zhao, Y.; Kruck, T.P.; Percy, M.E.; Tarr, M.A.; Lukiw, W.J. Characterization of an NF-κB-regulated, miRNA-146a-mediated down-regulation of complement factor H (CFH) in metal-sulfate-stressed human brain cells. J. Inorg. Biochem. 2009, 103, 1591–1595. [Google Scholar] [CrossRef] [PubMed]
  66. Tsuchiya, S.; Yamabe, M.; Yamaguchi, Y.; Kobayashi, Y.; Konno, T.; Tada, K. Establishment and characterization of a human acute monocytic leukemia cell line (THP-1). Int. J. Cancer 1980, 26, 171–176. [Google Scholar] [CrossRef] [PubMed]
  67. Li, B.; Chen, M.; Zhu, C. Neuroinflammation in prion disease. Int. J. Mol. Sci. 2021, 22, 2196. [Google Scholar] [CrossRef]
  68. Jaber, V.R.; Zhao, Y.; Sharfman, N.M.; Li, W.; Lukiw, W.J. Addressing Alzheimer’s disease (AD) neuropathology using anti-microRNA (AM) strategies. Mol. Neurobiol. 2019, 56, 8101–8108. [Google Scholar] [CrossRef]
  69. Li, Y.; Cui, J.; Hill, J.; Bhattacharjee, S.; Zhao, Y.; Lukiw, W. Increased expression of miRNA-146a in Alzheimer’s disease transgenic mouse models. Neurosci. Lett. 2011, 487, 94–98. [Google Scholar] [CrossRef] [Green Version]
  70. Roshan, R.; Ghosh, T.; Scaria, V.; Pillai, B. MicroRNAs: Novel therapeutic targets in neurodegenerative diseases. Drug Discov. Today 2009, 14, 1123–1129. [Google Scholar] [CrossRef]
  71. Saugstad, J. MicroRNAs as effectors of brain function with roles in ischemia and injury, neuroprotection, and neurodegeneration. Br. J. Pharmacol. 2010, 30, 1564–1576. [Google Scholar] [CrossRef] [PubMed]
  72. Liu, R.; Weng, Y. From the Scrapie syndrome of sheep and goat to the mad cow disease—The history of the discovery of prion. Zhonghua Yi Shi Za Zhi 2009, 39, 175–177. [Google Scholar]
  73. Jafarinejad-Farsangi, S.; Jazi, M.M.; Rostamzadeh, F.; Hadizadeh, M. High affinity of host human microRNAs to SARS-CoV-2 genome: An in silico analysis. Non-coding RNA Res. 2020, 5, 222–231. [Google Scholar] [CrossRef]
  74. Pierce, J.B.; Simion, V.; Icli, B.; Pérez-Cremades, D.; Cheng, H.S.; Feinberg, M.W. Computational analysis of targeting SARS-CoV-2, viral entry proteins ACE2 and TMPRSS2, and interferon genes by host microRNAs. Genes 2020, 11, 1354. [Google Scholar] [CrossRef]
  75. Carlson, G.; Prusiner, S. How an infection of sheep revealed prion mechanisms in Alzheimer’s disease and other neurodegenerative disorders. Int. J. Mol. Sci. 2021, 22, 4861. [Google Scholar] [CrossRef] [PubMed]
  76. Morris, G.; Bortolasci, C.C.; Puri, B.K.; Marx, W.; O’Neil, A.; Athan, E.; Walder, K.; Berk, M.; Olive, L.; Carvalho, A.F.; et al. The cytokine storms of COVID-19, H1N1 influenza, CRS and MAS compared. Can one sized treatment fit all? Cytokine 2021, 144, 155593. [Google Scholar] [CrossRef] [PubMed]
  77. Lukiw, W.J. microRNA heterogeneity, innate-immune defense and the efficacy of SARS-CoV-2 infection—A commentary. Non-coding RNA 2021, 7, 37. [Google Scholar] [CrossRef]
  78. Mirzaei, R.; Mahdavi, F.; Badrzadeh, F.; Hosseini-Fard, S.R.; Heidary, M.; Jeda, A.S.; Mohammadi, T.; Roshani, M.; Yousefimashouf, R.; Keyvani, H.; et al. The emerging role of microRNAs in the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Int. Immunopharmacol. 2020, 90, 107204. [Google Scholar] [CrossRef]
  79. Alam, T.; Lipovich, L. miRCOVID-19: Potential targets of human miRNAs in SARS-CoV-2 for RNA-based drug discovery. Non-coding RNA 2021, 7, 18. [Google Scholar] [CrossRef]
  80. Hu, C.; Chen, C.; Dong, X.-P. Impact of COVID-19 pandemic on patients with neurodegenerative diseases. Front. Aging Neurosci. 2021, 13. [Google Scholar] [CrossRef]
  81. Hu, J.; Stojanović, J.; Yasamineh, S.; Yasamineh, P.; Karuppannan, S.K.; Hussain Dowlath, M.J.; Serati-Nouri, H. The potential use of miRNAs as a therapeutic strategy for SARS-CoV-2 infection. Arch. Virol. 2021, 19, 1–24. [Google Scholar] [CrossRef]
  82. McLachlan, D.C.; Lukiw, W.; Cho, H.; Carp, R.; Wisniewski, H. Chromatin structure in scrapie and Alzheimer’s disease. Can. J. Neurol. Sci. 1986, 13, 427–431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Rubenstein, R.; Deng, H.; Race, R.; Ju, W.; Scalici, C.; Papini, M.; Kascsak, R.; Carp, R.; Rubenstein, A. Scrapie strain infection in vitro induces changes in neuronal cells. Mol. Neurobiol. 1994, 8, 129–138. [Google Scholar] [CrossRef] [PubMed]
  84. Carroll, J.A.; Chesebro, B. Neuroinflammation, microglia, and cell-association during prion disease. Viruses 2019, 11, 65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Wulf, M.-A.; Senatore, A.; Aguzzi, A.; Wulf, M.-A.; Senatore, A.; Aguzzi, A. The biological function of the cellular prion protein: An update. BMC Biol. 2017, 15, 1–13. [Google Scholar] [CrossRef] [Green Version]
  86. Jankovska, N.; Olejar, T.; Matej, R. Extracellular amyloid deposits in Alzheimer’s and Creutzfeldt–Jakob disease: Similar behavior of different proteins? Int. J. Mol. Sci. 2020, 22, 7. [Google Scholar] [CrossRef]
  87. Kordek, R.; Nerurkar, V.R.; Liberski, P.P.; Isaacson, S.; Yanagihara, R.; Gajdusek, D.C. Heightened expression of tumor necrosis factor alpha, interleukin 1 alpha, and glial fibrillary acidic protein in experimental Creutzfeldt-Jakob disease in mice. Proc. Natl. Acad. Sci. USA 1996, 93, 9754–9758. [Google Scholar] [CrossRef] [Green Version]
  88. Legname, G. Elucidating the function of the prion protein. PLoS Pathog. 2017, 13, e1006458. [Google Scholar] [CrossRef] [Green Version]
  89. Yoon, S.; Go, G.; Yoon, Y.; Lim, J.; Lee, G.; Lee, S. Harnessing the physiological functions of cellular prion protein in the kidneys: Applications for treating renal diseases. Biomolecules 2021, 11, 784. [Google Scholar] [CrossRef]
  90. Carroll, J.A.; Striebel, J.F.; Race, B.; Phillips, K.; Chesebro, B. Prion infection of mouse brain reveals multiple new upregulated genes involved in neuroinflammation or signal transduction. J. Virol. 2014, 89, 2388–2404. [Google Scholar] [CrossRef] [Green Version]
  91. Zabel, M.D.; Reid, C. A brief history of prions. Pathog. Dis. 2015, 73, ftv087. [Google Scholar] [CrossRef] [Green Version]
  92. Brunori, M. From Kuru to Alzheimer: A personal outlook. Protein Sci. 2021. [Google Scholar] [CrossRef]
  93. Khan, S.; Khan, S. Sporadic Creutzfeldt-Jakob disease: Diagnosing typical and atypical presentations under limited circumstances. Dement. Geriatr. Cogn. Disord. 2021, 50, 36–42. [Google Scholar] [CrossRef]
  94. Prasad, K.N. Oxidative and inflammatory events in prion diseases: Can they be therapeutic targets? Curr. Aging Sci. 2019, 11, 216–225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Soto, C.; Satani, N. The intricate mechanisms of neurodegeneration in prion diseases. Trends Mol. Med. 2011, 17, 14–24. [Google Scholar] [CrossRef] [Green Version]
  96. Kitamoto, T. A history for 50 years of the prion disease research. Rinsho Shinkeigaku 2009, 49, 936–938. [Google Scholar] [CrossRef] [PubMed]
  97. Brandel, J.-P. Les maladies à prions ou encéphalopathies spongiformes transmissibles. Rev. Med. Interne 2021. [Google Scholar] [CrossRef] [PubMed]
  98. Gkanatsiou, E.; Sahlin, C.; Portelius, E.; Johannesson, M.; Söderberg, L.; Fälting, J.; Basun, H.; Möller, C.; Odergren, T.; Zetterberg, H.; et al. Characterization of monomeric and soluble aggregated Aβ in Down’s syndrome and Alzheimer’s disease brains. Neurosci. Lett. 2021, 754, 135894. [Google Scholar] [CrossRef] [PubMed]
  99. Takahashi, R.H.; Nagao, T.; Gouras, G. Plaque formation and the intraneuronal accumulation of β-amyloid in Alzheimer’s disease. Pathol. Int. 2017, 67, 185–193. [Google Scholar] [CrossRef] [PubMed]
  100. Babelhadj, B.; Di Bari, M.A.; Pirisinu, L.; Chiappini, B.; Gaouar, S.B.S.; Riccardi, G.; Marcon, S.; Agrimi, U.; Nonno, R.; Vaccari, G. Prion disease in dromedary camels, Algeria. Emerg. Infect. Dis. 2018, 24, 1029–1036. [Google Scholar] [CrossRef] [Green Version]
  101. Lukiw, W.J. Micro-RNA speciation in fetal, adult and Alzheimer’s disease hippocampus. NeuroReport 2007, 18, 297–300. [Google Scholar] [CrossRef]
  102. Berger, A. Science commentary: Th1 and Th2 responses: What are they? BMJ 2000, 321, 424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Lescher, J.; Paap, F.; Schultz, V.; Redenbach, L.; Scheidt, U.; Rosewich, H.; Nessler, S.; Fuchs, E.; Gärtner, J.; Brück, W.; et al. MicroRNA regulation in experimental autoimmune encephalomyelitis in mice and marmosets resembles regulation in human multiple sclerosis lesions. J. Neuroimmunol. 2012, 246, 27–33. [Google Scholar] [CrossRef] [PubMed]
  104. He, F.; Liu, B.; Meng, Q.; Sun, Y.; Wang, W.; Wang, C. Modulation of miR-146a/complement factor H-mediated inflammatory responses in a rat model of temporal lobe epilepsy. Biosci. Rep. 2016, 36, e00433. [Google Scholar] [CrossRef]
  105. Schneider, K.; Fangerau, H.; Michaelsen, B.; Raab, W.H. The early history of the transmissible spongiform encephalopathies exemplified by scrapie. Brain Res. Bull. 2008, 77, 343–355. [Google Scholar] [CrossRef]
  106. Liberski, P.P.; Brown, P. Kuru—ffty years later. Neurol. Neurochir. Pol. 2007, 41, 548–556. [Google Scholar]
  107. Berezikov, E.; Cuppen, E.; Plasterk, R.H. Approaches to microRNA discovery. Nat. Genet. 2006, 38, S2–S7. [Google Scholar] [CrossRef] [PubMed]
  108. Zhao, Y.; Lukiw, W.J. Microbiome-mediated upregulation of microRNA-146a in sporadic Alzheimer’s disease. Front. Neurol. 2018, 9, 145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Guilherme, M.D.S.; Nguyen, V.T.T.; Reinhardt, C.; Endres, K. Impact of gut microbiome manipulation in 5xFAD mice on Alz-heimer’s disease-like pathology. Microorganisms 2021, 9, 815. [Google Scholar] [CrossRef]
  110. Van Olst, L.; Roks, S.J.; Kamermans, A.; Verhaar, B.J.H.; van der Geest, A.M.; Muller, M.; van der Flier, W.M.; de Vries, H.E. Contribution of gut microbiota to immunological changes in Alzheimer’s disease. Front. Immunol. 2021, 12. [Google Scholar] [CrossRef]
  111. Zhang, Y.; Geng, R.; Tu, Q. Gut microbial involvement in Alzheimer’s disease pathogenesis. Aging 2021, 13, 13359–13371. [Google Scholar] [CrossRef] [PubMed]
Table 1. Progressive, age-related neurological disorders in which miRNA-146a-5p is significantly upregulated in brain or CNS tissues; all neurotropic viruses indicated ultimately affect human brain or CNS function; (−)ssRNA = negative single-stranded RNA; (+)ssRNA = positive single-stranded RNA; dsDNA = double-stranded DNA genomes; prions contain no detectable nucleic acids [25]; natural hosts for prion infection are indicated; ‘interspecies transmission’ indicates disease transmissibility in experimental laboratory animals; there may be other neurotropic viruses or PrDs that involve miRNA-146a-5p remaining to be discovered; all neurotropic virus and prion diseases are alphabetically ordered; see also manuscript text.
Table 1. Progressive, age-related neurological disorders in which miRNA-146a-5p is significantly upregulated in brain or CNS tissues; all neurotropic viruses indicated ultimately affect human brain or CNS function; (−)ssRNA = negative single-stranded RNA; (+)ssRNA = positive single-stranded RNA; dsDNA = double-stranded DNA genomes; prions contain no detectable nucleic acids [25]; natural hosts for prion infection are indicated; ‘interspecies transmission’ indicates disease transmissibility in experimental laboratory animals; there may be other neurotropic viruses or PrDs that involve miRNA-146a-5p remaining to be discovered; all neurotropic virus and prion diseases are alphabetically ordered; see also manuscript text.
Neurotropic Viral PathogenGenusGenomeReference
Borna encephalitis disease virus 1 (BoEDV-1; BDV)Mononegavirales(−)ssRNA[26,27]
Chikungunya virus (CHIKV)Togaviridae(+)ssRNA[28]
enterovirus 71 (EV71)Picornaviridae(+)ssRNA[29]
Epstein-Barr virus (EBV)HerpesviridaedsDNA[30]
Hantavirus (HTV)Bunyaviridae(−)ssRNA[31]
hepatitis A virus (HAV)Picornaviridae(+)ssRNA[32]
hepatitis B virus (HBV)HepadnaviridaedsDNA[33,34,35,36]
hepatitis C virus (HCV)Flaviviridae(+)ssRNA[37];
herpes simplex virus-1 (HSV-1)HerpesviridaedsDNA[21,38,39,40,41]
Hendra virus (Henipavirus; HeV)Paramyxoviridae(−)ssRNA[42]
human immunodeficiency virus (HIV)Retrovirida(+)ssRNA[43]
human influenza A viruses (H1N1/H3N2)Orthomyxoviridae(+)ssRNA[44,45]
early human papillomavirus virus 16 (eHPV-16)PapillomaviridaedsDNA[46]
SARS-CoV-2 (agent for COVID-19 disease)Betacoronavirus(+)ssRNA[47]
human T-cell leukemia virus type 1 (HTLV-1)Retroviridae(+)ssRNA[48]
Japanese encephalitis virus (JEV)Flaviviridae(+)ssRNA[49,50]
Kaposi’s sarcoma-associated herpesvirus (KSHV)HerpesviridaedsDNA[51]
severe fever-thrombocytopenia syndrome virus (SFTSV)Bunyaviridae(−)ssRNA[52]
neurological disease
(natural or experimental)
natural host (family)interspecies transmissionReference
prion disease (PrD)
bovine spongiform encephalopathy (BSE)Bovidae; Hominidae+[16,41,53,54]
chronic wasting disease (CWD)Cervidae?[55]; Pogue & Lukiw, unpublished
sporadic Creutzfeldt–Jacob disease (sCJD)Hominidae+[10,13,14,56,57]
variant Creutzfeldt–Jacob disease (sCJD)Hominidae+[10,13,14,56,57,58]
Gerstmann–Sträussler–Scheinker syndrome (GSS)Hominidae+[10,13,14,56]
fatal familial insomnia (FFI)Hominidae+[16,53]; Pogue & Lukiw, unpublished
KuruHominidae+[59,60]
murine scrapie (experimental)Muridae+[14,54,61]
transmissible mink encephalopathy (TME)Mustelidae+[56]; Pogue & Lukiw, unpublished
feline spongiform encephalopathy (FSE)Felidae?Pogue and Lukiw, unpublished
camel prion disease (CPD)Camelidae?[2,56]
human neurodegenerative disease
age-related macular degeneration (AMD)Hominidae?[11,21,39]
Alzheimer’s disease (AD)Hominidae?[3,10,21,40,62,63]
amyotrophic lateral sclerosis (ALS)Hominidae?[3,23]
cerebrovascular disease (CVD)Hominidae?[3,18]
peripheral neuropathies/tumors of the CNSHominidae?[3,24]
temporal lobe epilepsy (TLE)Hominidae?[20]
traumatic brain injury (TBI)Hominidae?[19]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pogue, A.I.; Lukiw, W.J. microRNA-146a-5p, Neurotropic Viral Infection and Prion Disease (PrD). Int. J. Mol. Sci. 2021, 22, 9198. https://doi.org/10.3390/ijms22179198

AMA Style

Pogue AI, Lukiw WJ. microRNA-146a-5p, Neurotropic Viral Infection and Prion Disease (PrD). International Journal of Molecular Sciences. 2021; 22(17):9198. https://doi.org/10.3390/ijms22179198

Chicago/Turabian Style

Pogue, Aileen I., and Walter J. Lukiw. 2021. "microRNA-146a-5p, Neurotropic Viral Infection and Prion Disease (PrD)" International Journal of Molecular Sciences 22, no. 17: 9198. https://doi.org/10.3390/ijms22179198

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop