EVs, Substance Abuse, and HIV: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: , , , , , , , , ,

While extracellular vesicles (EVs) have been shown to play a role in CNS disorders, the intersection of EVs, drug use, and HIV is of particular interest. The interactions of HIV and drugs of abuse are a growing concern given the increasing incidence of HIV transmission via shared needles in illicit drug use. As a drug commonly taken through shared needles, METH is being investigated due to its role in exacerbating HIV-mediated inflammation through both increased vesicular shedding and extracellular release. In vivo experiments have shown that cocaine-induced EV release impacts synaptic plasticity through noncoding RNA. Nicotine studies have also highlighted how the differential packaging of antioxidant enzyme cargoes into EVs affects nicotine-mediated HIV pathogenesis. Additionally, studies of both morphine and heroin have demonstrated differences in the miRNA cargoes of EVs, potentially impacting gene expression and exacerbating HIV. Studies of alcohol use in combination with HIV have shown that EV cargoes such as cytokines are affected in HIV-infected subjects who use alcohol. Investigating EV cargo alterations in all forms of substance abuse studies may allow the EV, HIV, and addiction fields to progress towards diagnosis and remedies for substance-abuse-induced toxicity in HIV patients.

  • Extracellular vesicles (EVs)
  • substance abuse
  • HIV
  • cocaine
  • methamphetamine (METH)
  • alcohol
  • heroin
  • nicotine

1. Introduction

1.1. Extracellular Vesicles

Extracellular vesicles (EVs) are a broad, heterogeneous class of membranous lipid-bilayer vesicles that facilitate intercellular communication throughout the body. Secreted from all cell types, these cargo carriers have become important targets of investigation in various fields of study for their potential role in disease pathologies, drug-delivery systems, and therapeutics [1][2]. For the purpose of this review, all three classes of EVs—exosomes (30–150 nm), microvesicles (100–500 nm), and apoptotic bodies (500–5000 nm)—are collectively referred to as EVs, as endorsed by the International Society for Extracellular Vesicles [3]. EVs carry a variety of cargo types, including proteins, lipids, DNA fragments, and a variety of small noncoding RNAs, including miRNAs, mRNAs, and siRNAs [4][5]. The contents of EVs are reflective of the intracellular environments of their host cells, and EVs are released by both healthy and diseased cells [6]. EVs can transfer these cargoes from host cells to recipient cells, inducing functional transformations within recipient cells [7][8][9]. Regulation of EV secretion remains an active area of study, although certain stimuli and cellular conditions have been implicated in triggering EV release from different cell types [10].

EVs play a role in various aspects of healthy physiology, including immune responses [11][12], embryonic stem-cell communication during embryo implantation [13], and exercise [14][15]. EVs also shuttle essential biomolecules between cells that are critical for intercellular communication [16], antigen presentation [17], and signal transduction [18]. Moreover, EVs derived from mesenchymal stem cells have garnered interest in the fields of tissue repair, inflammation, anticancer therapy [19], and stroke [20][21]. Further, compelling evidence marks EVs as a potential drug-delivery system [1][22][23][24]; indeed, engineered EVs are capable of passing through the blood–brain barrier (BBB) [25], which has traditionally been a roadblock for efficient drug delivery to the brain [26][27][28][29].

Besides their beneficial role in the maintenance of physiological homeostasis and potentially therapeutic, diagnostic, and drug-delivery capabilities, EVs have been implicated in many pathogenies, including cardiovascular disease [30], neurodegenerative disorders [31][32][33][34], traumatic brain injury [35][36], HIV [37][38], and a wide range of cancers [39][40][41][42][43]. For instance, EVs may contribute to cancerous proliferation through angiogenesis, migratory and invasive capacities, and formation of metastatic lesions [44]. Dissecting the role and effects of EVs in these disease pathologies presents an ongoing challenge and an opportunity to progress understanding of the mechanisms underlying a diverse array of pressing health issues. Specifically, EV contents may indicate pathological changes in the body, and analysis of the molecular cargoes of the EVs may contribute to the advancement of diagnostic and treatment methods for these diseases.

1.2. Extracellular Vesicles in CNS Disorders and Addiction

1.2.1. EVs and CNS Disorders

Central nervous system (CNS) cells like neurons, microglia, astrocytes, oligodendrocytes, ependymal, and brain endothelial cells communicate by releasing EVs containing signaling molecules [45][46]. EVs aid in the signal transmission between neurons and glial cells, along with communication between CNS and peripheral body systems [47][48][49]. EVs maintain cellular homeostasis and clear abnormal aggregates; however, they also contribute to pathogenesis by delivering toxic substances to healthy cells, leading to inflammation and neurodegeneration [50] and thereby perpetuating CNS-associated neurodegenerative disorders [51][52]. Such CNS disorders include lysosomal storage disorders, Parkinson’s disease (PD) [53], Alzheimer’s disease (AD) [54][55][56][57], Huntington’s disease, amyotrophic lateral sclerosis [58], epilepsy, and multiple sclerosis [59][60][61][62][63]. EVs exacerbate disease pathogenesis by providing transportation to abnormally folded proteins and disease factors like α-synuclein [64], amyloid beta (Aβ) and Tau [65][66], huntingtin, and superoxide dismutase 1 [52][58].

EVs in diseased states differ significantly in their morphology and function, making them ideal biomarker candidates [67] as they contain unique proteins depending on the healthy or diseased microenvironment conditions [68][69]. The ability of EVs to cross the BBB, combined with their prevalence in bodily fluids, makes it possible to detect certain biomarkers found in difficult-to-assess regions like the CNS and spleen [70]. EVs may also contribute to neuroprotection; in AD, EVs sequester Aβ in vitro and promote its clearance, thus reducing neurotoxicity [71][72][73]. Moreover, neuronal EVs carry extracellular RNAs [74][75], including disease miRNA signatures that could be used as biomarkers to diagnose CNS disorders [58][76][77][78].

Additionally, EVs are potential candidates as therapeutic delivery agents as they can be easily loaded with therapeutic drugs, are minimally degraded, maintain their morphology and function, and can cross the BBB [2][79][80][81][82]. Due to their ability to carry functional small miRNA, tRNAs, lipids, and proteins [83], EVs are excellent carriers of the therapeutic agents. Besides acting as protective barriers against degradation and immunoreactivity, EVs can also increase the efficiency of delivery to targets, further aiding drug delivery and therapy for CNS diseases.

1.2.2. EVs and Substance Abuse

Investigations into the role of EVs in drug addiction and as future therapeutics for addiction are currently represented by a small but developing body of work [84]. Recent evidence points to a role of EV cargoes, specifically noncoding regulatory miRNAs [85], in mediating the body’s response to a variety of addictive substances, including cocaine [86][87], cannabinoids [88], nicotine [89], alcohol [90], and opioids [91][92]. These studies indicate that EVs and their cargoes may play a significant role in modulating addiction to a variety of substances, but further investigation is required to understand the full impact of EVs on addictive pathways and of addictive substances on EV secretion, uptake, and cargo content. There is a significant gap in the knowledge connecting substance abuse and our understanding of EVs and their cargoes in those addiction pathologies, although many investigators are currently working to close that gap.

2. EVs, Substance Abuse, and HIV

The Centers for Disease Control and Prevention (CDC) reports that out of 38,739 HIV infected individuals in the United States, 9% (3641) are individuals who inject drugs (http://www.cdc.gov/hiv/group/hiv-idu.html). As EVs can cross the BBB, the presence of HIV components in EVs can contribute to neuroinflammation [93] and neurodegeneration [6]. The interactions of HIV and drugs of abuse are of growing interest given the growing incidence of HIV transmission via shared needles during illicit drug use [94][95][96]. HIV exposure may also perpetuate addiction to stimulants [97]. Studies of HIV suggest that neuropathologies and substance abuse disorders often have a complex relationship that cannot be classified in one direction [98][99][100]; HIV and substance use together frequently result in the exacerbation of CNS disorders [101]. EVs are likely a key communication factor causing this exacerbation and interrelationship between HIV and substance abuse [102], however further research needs to be performed.

HIV is particularly hard to treat due to its ability to amass beyond the blood–brain barrier; it has a wide variety of impacts on the brain, including increased EV release [103][104]. Recently, research has investigated the role EVs play in the progression of microglia-mediated inflammation of HIV-infected subjects [93][103][105]. This inflammatory state is not resolved by combination antiretroviral therapy (cART) and remains a persisting issue [93]. Currently, METH is being investigated for its potential role in exacerbating HIV-mediated inflammation due to its ability to increase vesicular shedding and extracellular release [106][101][107][108]. Additionally, macrophage-derived EVs from primary human pulmonary arterial smooth muscle cells have been shown to be critically regulated by cocaine addiction and HIV infection [109].

Much like cocaine and METH, nicotine exacerbates HIV pathogenesis through the oxidative stress pathway [94][110]. Interestingly, EVs have revealed a strong correlation between cigarette smoking and HIV [110]. A recent study found that cigarette smoke condensate (CSC) reduced the total protein and antioxidant capacity in EVs isolated from HIV-infected and uninfected macrophages [111]. The EVs isolated from CSC-treated uninfected cells exhibited a protective property against cytotoxicity and viral replication in HIV-infected macrophages. Intriguingly, EVs isolated from HIV-infected cells lost their protective capacity. Further, levels of catalase and PRDX6, antioxidant enzyme cargoes, were decreased in EVs derived from HIV-infected cells. These results highlight the role of antioxidant enzymes in HIV replication and how the differential packaging of these cargoes into EVs affects nicotine-mediated HIV pathogenesis [111]. Indeed, Ranjit et al. suggest that because neurons have a weak antioxidant defense capacity and therefore rely on astrocytes to supply antioxidants, synthetically developed EVs loaded with antioxidant cargoes may be an efficient strategy for offsetting smoking-induced oxidative stress and HIV replication in the CNS [112].

Previous studies suggest that opioids may also play a role in exacerbating HIV-related neurological dysfunction and neuropathogenesis [113]. In simian immunodeficiency virus (SIV)-infected macaque monkeys, a model of HIV, opioid dependency has been demonstrated to increase mortality and exacerbate viral replication [114]. A 2012 study built upon previous studies of the consequences of HIV infection and opioid use by investigating the role of EV-delivered miR-29b in the regulation of PDGF-B gene expression in opioid-dependent SIV-infected macaques [115]. PDGF-B plays a crucial role in neuronal homeostasis, primarily via the protection of hippocampal neurons from glutamate-induced damage. The results of this study indicated that morphine exposure led to increased miR-29b secretion from astrocytes via EVs and demonstrated that increased miR-29b presentation decreased cell viability via decreased PDGF-B expression. This early study was the first to demonstrate that ADEVs can deliver miRNA cargoes to neurons and, in turn, these cargoes can induce functional changes in gene expression in the recipient neurons.

Similarly, a 2019 study investigated the effects of HIV infection and heroin use on inflammation-associated EV miRNA [116]. This study found that HIV-infected heroin users had significantly upregulated levels of miR-146a, miR-126, miR-21, and miR-let-7a, all of which are implicated in neuroinflammation. Interestingly, only the HIV-infected heroin-using group displayed this upregulation; neither uninfected heroin users nor heroin-free HIV-infected patients displayed significant levels of these miRNAs. Further, several members of the let-7 family of miRNA were significantly upregulated within the group of heroin users without HIV infection, namely miRNA-let-7a, -7d, -7e, -7f, -7g, and -7i. The let-7 family is highly conserved across animal species, including humans and mice, and is known to promote cell differentiation [117]. Interestingly, another group noted that morphine significantly increased expression levels of miRNA-let-7a, 7c, and 7g [91]. These results further indicate the importance of understanding the implications of the combination of HIV infection and opioid use as it relates to EV miRNA cargo.

As opioids and needle-sharing are associated with increased risk of HIV infection, alcohol also increases the risk of infection and aggravates HIV replication. Further, alcohol diminishes the adherence to and the efficiency of antiretroviral therapy (ART), which may further enhance HIV replication. HIV infection is correlated with enhanced expression of pro-inflammatory cytokines and chemokines, consequently promoting the pathogenesis of HIV [118]. In the search for a prospective biomarker for alcohol-stimulated toxicity in HIV patients, Kodidela et al. found that HIV-positive alcohol users had substantially lower levels of EV IL-1ra compared to HIV-negative alcohol drinkers. Additionally, no change in the levels of EV IL-1ra was found in the nondrinker HIV-positive subjects. IL-10 was also present in EVs of HIV-positive drinkers. Furthermore, compared to plasma, the percentages of TNF-α, IL-8, and IL-1ra packaged in the EVs isolated from HIV-positive alcohol users were 15%, 10%, and 10%, respectively [118].

In addition to cytokine EV cargo changes, hemopexin (HPX), a protein that binds to free heme, was found in reduced concentrations in the EVs of HIV-positive drinkers, possibly aggravating or contributing to neuroAIDS in those patients [119]. Although unchanged in alcohol drinkers and HIV patients, HPX was substantially downregulated in alcohol users with HIV. HPX may possess an anti-inflammatory function through the negative regulation of TNF-α and IL-6 secretion by macrophages. Additionally, HPX is an extracellular antioxidant, and its diminished level in the EVs of HIV-positive drinkers is consistent with its protective role against alcohol-induced oxidative stress. Additionally, Kodidela et al. found that GFAP expression was significantly enhanced in plasma EVs obtained from HIV-positive subjects and alcohol users, suggesting increased astrocyte activation in those subjects [120]. Exploring EV cargo alterations, such as those listed in Table 1, may allow the field to progress towards diagnosis of and remedies for alcohol-induced toxicity in HIV patients.

Table 1. Differentially regulated EV cargoes identified in studies of substance abuse and HIV.

Cargo Condition EV Source Model Up/Down Reference
miRNA 29b Morphine + HIV Astrocyte Rat primary cultures Up [115]
21 Heroin + HIV Plasma Human Up [116]
146a Heroin + HIV Plasma Human Up [116][121]
126 Heroin + HIV Plasma Human Up [116]
let-7a Heroin + HIV Plasma Human Up [116]
let-7b Alcohol Microglia BV2 cell line Up [122]
276 Methamphetamine (METH) Plasma Rat Up [123]
218b METH Plasma Rat Up [123]
194-5p METH Plasma Rat Up [123]
152-3p METH Plasma Rat Up [123]
25 METH Plasma Rat Down [123]
276 Ketamine Plasma Rat Down [123]
22-3p METH/Bipolar Plasma Rat Up [123][124]
107 Nicotine Bronchoalveolar lavage fluid (BLF) Human Up [125]
126 Nicotine BLF Human Up [125]
19a-3p Nicotine BLF Human Up [125]
200a-3p Nicotine BLF Human Up [125]
21-3p Nicotine Macrophage RAW264.7 cell line Up [126]
21 SIV Brain Monkey Up [102]
182 Alcohol Astrocyte Mouse primary culture Up [121]
200b Alcohol Astrocyte Mouse primary culture Down [121]
155 Alcohol Microglia BV2 cell line Up [122]
140-3p Alcohol Fetal neural stem cells (fNSC) Mouse Up [127]
15b-3p Alcohol fNSC Mouse Up [127]
340-5p Alcohol fNSC Mouse Up [127]
674-5p Alcohol fNSC Mouse Up [127]
130a HIV/Cocaine Monocytes Monomac-1 cell line Up [109]
lncRNA MALAT1 Nicotine BLF Human Up [125]
HOTAIR Nicotine BLF Human Up [125]
HOTTIP Nicotine BLF Human Up [125]
AGAP-AS1 Nicotine BLF Human Up [125]
ATB Nicotine BLF Human Up [125]
TCF7 Nicotine BLF Human Up [125]
FOXD2-AS1 Nicotine BLF Human Up [125]
HOXA11-AS Nicotine BLF Human Up [125]
PCAF1 Nicotine BLF Human Up [125]
BCAR4 Nicotine BLF Human Up [125]
mRNA EGFR Nicotine BLF Human Up [125]
KRAS Nicotine BLF Human Up [125]
ALK Nicotine BLF Human Up [125]
MET Nicotine BLF Human Up [125]
LKB1 Nicotine BLF Human Up [125]
BRAF Nicotine BLF Human Up [125]
PIK3CA Nicotine BLF Human Up [125]
RET Nicotine BLF Human Up [125]
ROS1 Nicotine BLF Human Up [125]
Cytokines 130a HIV/Cocaine Monocytes; Plasma Monomac-1 cell line; Human Up [109][118]
IL6/IL-8 Smoking + HIV Plasma Human Up [118]
IL-6 Smoking + HIV Plasma Human Up [118]
IL-1ra Alcohol/ Nicotine + HIV Plasma Human Up [118]
IL-10 Alcohol/Nicotine
HIV
Plasma Human Up [118]
Proteins Amyloid beta (Aβ) HIV Brain Human Up [103]
GFAP HIV + Alcohol Plasma Human Up [120]
L1CAM Nicotine Plasma Human Up [120]
α-synuclein METH Neuroblastoma cells SH-SY5Y cell line Up [128]
TLR4 Alcohol Astrocyte Mouse primary culture Up [121]
NFκB-p65 Alcohol Astrocyte Mouse primary culture Up [121]
IL-1R Alcohol Astrocyte Mouse primary culture Up [121]
Caspase-1 Alcohol Astrocyte Mouse primary culture Up [121]
CPM HIV Plasma Human Up [129]
CDH3 HIV Plasma Human Up [129]
HPX HIV + alcohol Plasma Human Down [119]
BAGE Nicotine Lung Human Up [125]
PD-L1 Nicotine Lung Human Up [125]
PRDX6 HIV + Nicotine Macrophage U937 cells Down [111]
Catalase HIV + Nicotine Macrophage U937 cells Down [111]
CSF2RA HIV Plasma Human Up [129]
MANF HIV Plasma Human Up [129]

This entry is adapted from the peer-reviewed paper 10.3390/ijms21186765

References

  1. Shahjin, F.; Chand, S.; Yelamanchili, S.V. Extracellular Vesicles as Drug Delivery Vehicles to the Central Nervous System. J. Neuroimmune Pharmacol. Off. J. Soc. Neuroimmune Pharmacol. 2019, doi:10.1007/s11481-019-09875-w.
  2. Rufino-Ramos, D.; Albuquerque, P.R.; Carmona, V.; Perfeito, R.; Nobre, R.J.; Pereira de Almeida, L. Extracellular vesicles: Novel promising delivery systems for therapy of brain diseases. J Control Release 2017, 262, 247–258.
  3. Thery, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell. Vesicles 2018, 7, 1535750.
  4. Cocucci, E.; Meldolesi, J. Ectosomes and exosomes: Shedding the confusion between extracellular vesicles. Trends Cell Biol. 2015, 25, 364–372.
  5. Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, doi:10.1126/science.aau6977.
  6. Hu, G.; Yang, L.; Cai, Y.; Niu, F.; Mezzacappa, F.; Callen, S.; Fox, H.S.; Buch, S. Emerging roles of extracellular vesicles in neurodegenerative disorders: Focus on HIV-associated neurological complications. Cell Death Dis. 2016, 7, e2481.
  7. Valadi, H.; Ekstrom, K.; Bossios, A.; Sjostrand, M.; Lee, J.J.; Lotvall, J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659.
  8. Antonyak, M.A.; Li, B.; Boroughs, L.K.; Johnson, J.L.; Druso, J.E.; Bryant, K.L.; Holowka, D.A.; Cerione, R.A. Cancer cell-derived microvesicles induce transformation by transferring tissue transglutaminase and fibronectin to recipient cells. Proc. Natl. Acad. Sci. USA 2011, 108, 4852–4857.
  9. Novak, C.M.; Ozen, M.; McLane, M.; Alqutub, S.; Lee, J.Y.; Lei, J.; Burd, I. Progesterone improves perinatal neuromotor outcomes in a mouse model of intrauterine inflammation via immunomodulation of the placenta. Am. J. Reprod. Immunol. 2018, 79, e12842.
  10. Colombo, M.; Raposo, G.; Thery, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289.
  11. Greening, D.W.; Gopal, S.K.; Xu, R.; Simpson, R.J.; Chen, W. Exosomes and their roles in immune regulation and cancer. Semin. Cell Dev. Biol. 2015, 40, 72–81.
  12. Robbins, P.D.; Morelli, A.E. Regulation of immune responses by extracellular vesicles. Nat. Rev. Immunol. 2014, 14, 195–208.
  13. Desrochers, L.M.; Bordeleau, F.; Reinhart-King, C.A.; Cerione, R.A.; Antonyak, M.A. Microvesicles provide a mechanism for intercellular communication by embryonic stem cells during embryo implantation. Nat. Commun. 2016, 7, 11958.
  14. Frühbeis, C.; Helmig, S.; Tug, S.; Simon, P.; Krämer-Albers, E.M. Physical exercise induces rapid release of small extracellular vesicles into the circulation. J. Extracell. Vesicles 2015, 4, 28239.
  15. Lovett, J.A.C.; Durcan, P.J.; Myburgh, K.H. Investigation of Circulating Extracellular Vesicle MicroRNA Following Two Consecutive Bouts of Muscle-Damaging Exercise. Front. Physiol. 2018, 9, 1149.
  16. Shi, M.; Sheng, L.; Stewart, T.; Zabetian, C.P.; Zhang, J. New windows into the brain: Central nervous system-derived extracellular vesicles in blood. Prog. Neurobiol. 2019, 175, 96–106.
  17. Lindenbergh, M.F.S.; Stoorvogel, W. Antigen Presentation by Extracellular Vesicles from Professional Antigen-Presenting Cells. Annu. Rev. Immunol. 2018, 36, 435–459.
  18. Sun, Z.; Wang, L.; Dong, L.; Wang, X. Emerging role of exosome signalling in maintaining cancer stem cell dynamic equilibrium. J. Cell. Mol. Med. 2018, doi:10.1111/jcmm.13676.
  19. Baek, G.; Choi, H.; Kim, Y.; Lee, H.-C.; Choi, C. Mesenchymal Stem Cell-Derived Extracellular Vesicles as Therapeutics and as a Drug Delivery Platform. Stem Cells Transl. Med. 2019, 8, 880–886.
  20. Zhang, Y.; Chopp, M.; Meng, Y.; Katakowski, M.; Xin, H.; Mahmood, A.; Xiong, Y. Effect of exosomes derived from multipluripotent mesenchymal stromal cells on functional recovery and neurovascular plasticity in rats after traumatic brain injury. J. Neurosurg. 2015, 122, 856–867.
  21. Xin, H.; Li, Y.; Cui, Y.; Yang, J.J.; Zhang, Z.G.; Chopp, M. Systemic administration of exosomes released from mesenchymal stromal cells promote functional recovery and neurovascular plasticity after stroke in rats. J. Cereb. Blood Flow Metab. 2013, 33, 1711–1715.
  22. Lamichhane, T.N.; Jay, S.M. Production of Extracellular Vesicles Loaded with Therapeutic Cargo. Methods Mol. Biol. 2018, 1831, 37–47.
  23. O’Loughlin, A.J.; Mager, I.; de Jong, O.G.; Varela, M.A.; Schiffelers, R.M.; El Andaloussi, S.; Wood, M.J.A.; Vader, P. Functional Delivery of Lipid-Conjugated siRNA by Extracellular Vesicles. Mol. Ther. 2017, 25, 1580–1587.
  24. Villa, F.; Quarto, R.; Tasso, R. Extracellular Vesicles as Natural, Safe and Efficient Drug Delivery Systems. Pharmaceutics 2019, 11, 557.
  25. Saeedi, S.; Israel, S.; Nagy, C.; Turecki, G. The emerging role of exosomes in mental disorders. Transl. Psychiatry 2019, 9, 122.
  26. Ha, D.; Yang, N.; Nadithe, V. Exosomes as therapeutic drug carriers and delivery vehicles across biological membranes: Current perspectives and future challenges. Acta Pharm. Sin. B 2016, 6, 287–296.
  27. Alvarez-Erviti, L.; Seow, Y.; Yin, H.; Betts, C.; Lakhal, S.; Wood, M.J. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol. 2011, 29, 341–345.
  28. Yang, T.; Martin, P.; Fogarty, B.; Brown, A.; Schurman, K.; Phipps, R.; Yin, V.P.; Lockman, P.; Bai, S. Exosome delivered anticancer drugs across the blood-brain barrier for brain cancer therapy in Danio rerio. Pharm. Res. 2015, 32, 2003–2014.
  29. Bagchi, S.; Chhibber, T.; Lahooti, B.; Verma, A.; Borse, V.; Jayant, R.D. In-vitro blood-brain barrier models for drug screening and permeation studies: An overview. Drug Des. Dev. Ther. 2019, 13, 3591–3605.
  30. Zhang, Y.; Hu, Y.W.; Zheng, L.; Wang, Q. Characteristics and Roles of Exosomes in Cardiovascular Disease. DNA Cell Biol. 2017, 36, 202–211.
  31. Xiao, T.; Zhang, W.; Jiao, B.; Pan, C.-Z.; Liu, X.; Shen, L. The role of exosomes in the pathogenesis of Alzheimer’ disease. Transl. Neurodegener. 2017, 6, 3–3.
  32. Watson, L.S.; Hamlett, E.D.; Stone, T.D.; Sims-Robinson, C. Neuronally derived extracellular vesicles: An emerging tool for understanding Alzheimer’s disease. Mol. Neurodegener. 2019, 14, 22.
  33. Kanninen, K.M.; Bister, N.; Koistinaho, J.; Malm, T. Exosomes as new diagnostic tools in CNS diseases. Biochim. Et Biophys. Acta 2016, 1862, 403–410.
  34. Lee, S.; Mankhong, S.; Kang, J.-H. Extracellular Vesicle as a Source of Alzheimer’s Biomarkers: Opportunities and Challenges. Int. J. Mol. Sci. 2019, 20, 1728.
  35. Osier, N.; Motamedi, V.; Edwards, K.; Puccio, A.; Diaz-Arrastia, R.; Kenney, K.; Gill, J. Exosomes in Acquired Neurological Disorders: New Insights into Pathophysiology and Treatment. Mol. Neurobiol. 2018, 55, 9280–9293.
  36. Huang, S.; Ge, X.; Yu, J.; Han, Z.; Yin, Z.; Li, Y.; Chen, F.; Wang, H.; Zhang, J.; Lei, P. Increased miR-124-3p in microglial exosomes following traumatic brain injury inhibits neuronal inflammation and contributes to neurite outgrowth via their transfer into neurons. FASEB J. 2018, 32, 512–528.
  37. Patters, B.J.; Kumar, S. The role of exosomal transport of viral agents in persistent HIV pathogenesis. Retrovirology 2018, 15, 79.
  38. Schorey, J.S.; Cheng, Y.; Singh, P.P.; Smith, V.L. Exosomes and other extracellular vesicles in host-pathogen interactions. EMBO Rep. 2015, 16, 24–43.
  39. Melo, S.A.; Sugimoto, H.; O’Connell, J.T.; Kato, N.; Villanueva, A.; Vidal, A.; Qiu, L.; Vitkin, E.; Perelman, L.T.; Melo, C.A.; et al. Cancer exosomes perform cell-independent microRNA biogenesis and promote tumorigenesis. Cancer Cell 2014, 26, 707–721.
  40. Nedaeinia, R.; Manian, M.; Jazayeri, M.H.; Ranjbar, M.; Salehi, R.; Sharifi, M.; Mohaghegh, F.; Goli, M.; Jahednia, S.H.; Avan, A.; et al. Circulating exosomes and exosomal microRNAs as biomarkers in gastrointestinal cancer. Cancer Gene Ther. 2017, 24, 48–56.
  41. Jalalian, S.H.; Ramezani, M.; Jalalian, S.A.; Abnous, K.; Taghdisi, S.M. Exosomes, new biomarkers in early cancer detection. Anal. Biochem. 2019, 571, 1–13.
  42. Taylor, D.D.; Gercel-Taylor, C. MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol. Oncol. 2008, 110, 13–21.
  43. Muralidharan-Chari, V.; Kohan, H.G.; Asimakopoulos, A.G.; Sudha, T.; Sell, S.; Kannan, K.; Boroujerdi, M.; Davis, P.J.; Mousa, S.A. Microvesicle removal of anticancer drugs contributes to drug resistance in human pancreatic cancer cells. Oncotarget 2016, 7, 50365–50379.
  44. Osaki, M.; Okada, F. Exosomes and Their Role in Cancer Progression. Yonago Acta Med. 2019, 62, 182–190.
  45. Paolicelli, R.C.; Bergamini, G.; Rajendran, L. Cell-to-cell Communication by Extracellular Vesicles: Focus on Microglia. Neuroscience 2019, 405, 148–157.
  46. Chhibber, T.; Bagchi, S.; Lahooti, B.; Verma, A.; Al-Ahmad, A.; Paul, M.K.; Pendyala, G.; Jayant, R.D. CNS organoids: An innovative tool for neurological disease modeling and drug neurotoxicity screening. Drug Discov. Today 2020, 25, 456–465.
  47. Chulpanova, D.S.; Kitaeva, K.V.; James, V.; Rizvanov, A.A.; Solovyeva, V.V. Therapeutic Prospects of Extracellular Vesicles in Cancer Treatment. Front. Immunol. 2018, 9, 1534.
  48. Jiang, L.; Vader, P.; Schiffelers, R.M. Extracellular vesicles for nucleic acid delivery: Progress and prospects for safe RNA-based gene therapy. Gene Ther. 2017, 24, 157–166.
  49. Galieva, L.R.; James, V.; Mukhamedshina, Y.O.; Rizvanov, A.A. Therapeutic Potential of Extracellular Vesicles for the Treatment of Nerve Disorders. Front. Neurosci. 2019, 13, 163.
  50. Hill, A.F. Extracellular Vesicles and Neurodegenerative Diseases. J. Neurosci. 2019, 39, 9269–9273.
  51. Caruso Bavisotto, C.; Scalia, F.; Marino Gammazza, A.; Carlisi, D.; Bucchieri, F.; Conway de Macario, E.; Macario, A.J.L.; Cappello, F.; Campanella, C. Extracellular Vesicle-Mediated Cell⁻Cell Communication in the Nervous System: Focus on Neurological Diseases. Int. J. Mol. Sci. 2019, 20, 434.
  52. Croese, T.; Furlan, R. Extracellular vesicles in neurodegenerative diseases. Mol. Asp. Med. 2018, 60, 52–61.
  53. Shi, M.; Liu, C.; Cook, T.J.; Bullock, K.M.; Zhao, Y.; Ginghina, C.; Li, Y.; Aro, P.; Dator, R.; He, C.; et al. Plasma exosomal α-synuclein is likely CNS-derived and increased in Parkinson’s disease. Acta Neuropathol. 2014, 128, 639–650.
  54. Sardar Sinha, M.; Ansell-Schultz, A.; Civitelli, L.; Hildesjö, C.; Larsson, M.; Lannfelt, L.; Ingelsson, M.; Hallbeck, M. Alzheimer’s disease pathology propagation by exosomes containing toxic amyloid-beta oligomers. Acta Neuropathol. 2018, 136, 41–56.
  55. Brites, D.; Fernandes, A. Neuroinflammation and Depression: Microglia Activation, Extracellular Microvesicles and microRNA Dysregulation. Front. Cell. Neurosci. 2015, 9, 476.
  56. Mustapic, M.; Eitan, E.; Werner, J.K., Jr.; Berkowitz, S.T.; Lazaropoulos, M.P.; Tran, J.; Goetzl, E.J.; Kapogiannis, D. Plasma Extracellular Vesicles Enriched for Neuronal Origin: A Potential Window into Brain Pathologic Processes. Front. Neurosci. 2017, 11, 278.
  57. Trotta, T.; Panaro, M.A.; Cianciulli, A.; Mori, G.; Di Benedetto, A.; Porro, C. Microglia-derived extracellular vesicles in Alzheimer’s Disease: A double-edged sword. Biochem. Pharm. 2018, 148, 184–192.
  58. Shaimardanova, A.A.; Solovyeva, V.V.; Chulpanova, D.S.; James, V.; Kitaeva, K.V.; Rizvanov, A.A. Extracellular vesicles in the diagnosis and treatment of central nervous system diseases. Neural. Regen. Res. 2020, 15, 586–596.
  59. Selmaj, I.; Mycko, M.P.; Raine, C.S.; Selmaj, K.W. The role of exosomes in CNS inflammation and their involvement in multiple sclerosis. J. Neuroimmunol. 2017, 306, 1–10.
  60. Bonafede, R.; Mariotti, R. ALS Pathogenesis and Therapeutic Approaches: The Role of Mesenchymal Stem Cells and Extracellular Vesicles. Front. Cell. Neurosci. 2017, 11, 80.
  61. Moyano, A.L.; Li, G.; Boullerne, A.I.; Feinstein, D.L.; Hartman, E.; Skias, D.; Balavanov, R.; van Breemen, R.B.; Bongarzone, E.R.; Månsson, J.E.; et al. Sulfatides in extracellular vesicles isolated from plasma of multiple sclerosis patients. J. Neurosci. Res. 2016, 94, 1579–1587.
  62. Pieragostino, D.; Lanuti, P.; Cicalini, I.; Cufaro, M.C.; Ciccocioppo, F.; Ronci, M.; Simeone, P.; Onofrj, M.; van der Pol, E.; Fontana, A.; et al. Proteomics characterization of extracellular vesicles sorted by flow cytometry reveals a disease-specific molecular cross-talk from cerebrospinal fluid and tears in multiple sclerosis. J. Proteom. 2019, 204, 103403.
  63. Ulivieri, C.; Baldari, C.T. Regulation of T Cell Activation and Differentiation by Extracellular Vesicles and Their Pathogenic Role in Systemic Lupus Erythematosus and Multiple Sclerosis. Molecules 2017, 22, 225.
  64. Emmanouilidou, E.; Melachroinou, K.; Roumeliotis, T.; Garbis, S.D.; Ntzouni, M.; Margaritis, L.H.; Stefanis, L.; Vekrellis, K. Cell-produced α-synuclein is secreted in a calcium-dependent manner by exosomes and impacts neuronal survival. J. Neurosci. 2010, 30, 6838–6851.
  65. Medina, M.; Avila, J. The role of extracellular Tau in the spreading of neurofibrillary pathology. Front. Cell. Neurosci. 2014, 8, 113.
  66. Iba, M.; Guo, J.L.; McBride, J.D.; Zhang, B.; Trojanowski, J.Q.; Lee, V.M.-Y. Synthetic tau fibrils mediate transmission of neurofibrillary tangles in a transgenic mouse model of Alzheimer’s-like tauopathy. J. Neurosci. 2013, 33, 1024–1037.
  67. Chand, S.; Jo, A.; Vellichirammal, N.N.; Gowen, A.; Guda, C.; Schaal, V.; Odegaard, K.; Lee, H.; Pendyala, G.; Yelamanchili, S.V. Comprehensive Characterization of Nanosized Extracellular Vesicles from Central and Peripheral Organs : Implications for Preclinical and Clinical Applications. ACS Appl. Nano Mater. 2020, doi:10.1021/acsanm.0c01654.
  68. Wong, C.H.; Chen, Y.C. Clinical significance of exosomes as potential biomarkers in cancer. World J. Clin. Cases 2019, 7, 171–190.
  69. Haraszti, R.A.; Didiot, M.C.; Sapp, E.; Leszyk, J.; Shaffer, S.A.; Rockwell, H.E.; Gao, F.; Narain, N.R.; DiFiglia, M.; Kiebish, M.A.; et al. High-resolution proteomic and lipidomic analysis of exosomes and microvesicles from different cell sources. J. Extracell. Vesicles 2016, 5, 32570.
  70. Manek, R.; Moghieb, A.; Yang, Z.; Kumar, D.; Kobessiy, F.; Sarkis, G.A.; Raghavan, V.; Wang, K.K.W. Protein Biomarkers and Neuroproteomics Characterization of Microvesicles/Exosomes from Human Cerebrospinal Fluid Following Traumatic Brain Injury. Mol. Neurobiol. 2018, 55, 6112–6128.
  71. Yuyama, K.; Sun, H.; Sakai, S.; Mitsutake, S.; Okada, M.; Tahara, H.; Furukawa, J.-I.; Fujitani, N.; Shinohara, Y.; Igarashi, Y. Decreased amyloid-β pathologies by intracerebral loading of glycosphingolipid-enriched exosomes in Alzheimer model mice. J. Biol. Chem. 2014, 289, 24488–24498.
  72. Yuyama, K.; Sun, H.; Usuki, S.; Sakai, S.; Hanamatsu, H.; Mioka, T.; Kimura, N.; Okada, M.; Tahara, H.; Furukawa, J.-I. A potential function for neuronal exosomes: Sequestering intracerebral amyloid‐β peptide. FEBS Lett. 2015, 589, 84–88.
  73. Thompson, A.G.; Gray, E.; Heman-Ackah, S.M.; Mäger, I.; Talbot, K.; El Andaloussi, S.; Wood, M.J.; Turner, M.R. Extracellular vesicles in neurodegenerative disease—Pathogenesis to biomarkers. Nat. Rev. Neurol. 2016, 12, 346.
  74. Saugstad, J.A.; Lusardi, T.A.; Van Keuren-Jensen, K.R.; Phillips, J.I.; Lind, B.; Harrington, C.A.; McFarland, T.J.; Courtright, A.L.; Reiman, R.A.; Yeri, A.S.; et al. Analysis of extracellular RNA in cerebrospinal fluid. J Extracell. Vesicles 2017, 6, 1317577.
  75. Kim, K.M.; Abdelmohsen, K.; Mustapic, M.; Kapogiannis, D.; Gorospe, M. RNA in extracellular vesicles. Wiley Interdiscip. Rev. RNA 2017, 8, e1413.
  76. Delpech, J.C.; Herron, S.; Botros, M.B.; Ikezu, T. Neuroimmune Crosstalk through Extracellular Vesicles in Health and Disease. Trends Neurosci. 2019, 42, 361–372.
  77. Wang, X.; Botchway, B.O.A.; Zhang, Y.; Yuan, J.; Liu, X. Combinational Treatment of Bioscaffolds and Extracellular Vesicles in Spinal Cord Injury. Front. Mol. Neurosci. 2019, 12, 81.
  78. Gui, Y.; Liu, H.; Zhang, L.; Lv, W.; Hu, X. Altered microRNA profiles in cerebrospinal fluid exosome in Parkinson disease and Alzheimer disease. Oncotarget 2015, 6, 37043.
  79. Matsumoto, J.; Stewart, T.; Banks, W.A.; Zhang, J. The Transport Mechanism of Extracellular Vesicles at the Blood-Brain Barrier. Curr. Pharm. Des. 2017, 23, 6206–6214.
  80. Ramirez, S.H.; Andrews, A.M.; Paul, D.; Pachter, J.S. Extracellular vesicles: Mediators and biomarkers of pathology along CNS barriers. Fluids Barriers CNS 2018, 15, 19.
  81. Saint-Pol, J.; Gosselet, F.; Duban-Deweer, S.; Pottiez, G.; Karamanos, Y. Targeting and Crossing the Blood-Brain Barrier with Extracellular Vesicles. Cells 2020, 9, 851.
  82. Xu, G.; Ao, R.; Zhi, Z.; Jia, J.; Yu, B. miR-21 and miR-19b delivered by hMSC-derived EVs regulate the apoptosis and differentiation of neurons in patients with spinal cord injury. J. Cell. Physiol. 2019, 234, 10205–10217.
  83. Barile, L.; Vassalli, G. Exosomes: Therapy delivery tools and biomarkers of diseases. Pharm. Ther. 2017, 174, 63–78.
  84. Rao, P.S.S.; O’Connell, K.; Finnerty, T.K. Potential Role of Extracellular Vesicles in the Pathophysiology of Drug Addiction. Mol. Neurobiol. 2018, 55, 6906–6913.
  85. Nunez, Y.O.; Mayfield, R.D. Understanding Alcoholism Through microRNA Signatures in Brains of Human Alcoholics. Front. Genet. 2012, 3, 43.
  86. Quinn, R.K.; Brown, A.L.; Goldie, B.J.; Levi, E.M.; Dickson, P.W.; Smith, D.W.; Cairns, M.J.; Dayas, C.V. Distinct miRNA expression in dorsal striatal subregions is associated with risk for addiction in rats. Transl. Psychiatry 2015, 5, e503.
  87. Hollander, J.A.; Im, H.I.; Amelio, A.L.; Kocerha, J.; Bali, P.; Lu, Q.; Willoughby, D.; Wahlestedt, C.; Conkright, M.D.; Kenny, P.J. Striatal microRNA controls cocaine intake through CREB signalling. Nature 2010, 466, 197–202.
  88. Chiarlone, A.; Börner, C.; Martín-Gómez, L.; Jiménez-González, A.; García-Concejo, A.; García-Bermejo, M.L.; Lorente, M.; Blázquez, C.; García-Taboada, E.; de Haro, A.; et al. MicroRNA let-7d is a target of cannabinoid CB1 receptor and controls cannabinoid signaling. Neuropharmacology 2016, 108, 345–352.
  89. Lee, S.; Woo, J.; Kim, Y.S.; Im, H.I. Integrated miRNA-mRNA analysis in the habenula nuclei of mice intravenously self-administering nicotine. Sci. Rep. 2015, 5, 12909.
  90. Pietrzykowski, A.Z.; Friesen, R.M.; Martin, G.E.; Puig, S.I.; Nowak, C.L.; Wynne, P.M.; Siegelmann, H.T.; Treistman, S.N. Posttranscriptional regulation of BK channel splice variant stability by miR-9 underlies neuroadaptation to alcohol. Neuron 2008, 59, 274–287.
  91. He, Y.; Yang, C.; Kirkmire, C.M.; Wang, Z.J. Regulation of opioid tolerance by let-7 family microRNA targeting the mu opioid receptor. J. Neurosci. 2010, 30, 10251–10258.
  92. Barbierato, M.; Zusso, M.; Skaper, S.D.; Giusti, P. MicroRNAs: Emerging role in the endogenous mu opioid system. CNS Neurol. Disord. Drug Targets 2015, 14, 239–250.
  93. Pérez, P.S.; Romaniuk, M.A.; Duette, G.A.; Zhao, Z.; Huang, Y.; Martin-Jaular, L.; Witwer, K.W.; Théry, C.; Ostrowski, M. Extracellular vesicles and chronic inflammation during HIV infection. J. Extracell. Vesicles 2019, 8, 1687275.
  94. Jayant, R.D.; Tiwari, S.; Atluri, V.; Kaushik, A.; Tomitaka, A.; Yndart, A.; Colon-Perez, L.; Febo, M.; Nair, M. Multifunctional Nanotherapeutics for the Treatment of neuroAIDS in Drug Abusers. Sci. Rep. 2018, 8, 12991.
  95. Nair, M.; Jayant, R.D.; Kaushik, A.; Sagar, V. Getting into the brain: Potential of nanotechnology in the management of NeuroAIDS. Adv. Drug Deliv. Rev. 2016, 103, 202–217.
  96. Jayant, R.D.; Atluri, V.S.; Agudelo, M.; Sagar, V.; Kaushik, A.; Nair, M. Sustained-release nanoART formulation for the treatment of neuroAIDS. Int. J. Nanomed. 2015, 10, 1077–1093.
  97. Blackstone, K.; Iudicello, J.E.; Morgan, E.E.; Weber, E.; Moore, D.J.; Franklin, D.R.; Ellis, R.J.; Grant, I.; Woods, S.P.; Translational Methamphetamine, A.R.C.G. Human immunodeficiency virus infection heightens concurrent risk of functional dependence in persons with long-term methamphetamine use. J. Addict. Med. 2013, 7, 255–263.
  98. Sanchez, A.B.; Kaul, M. Neuronal stress and injury caused by HIV-1, cART and drug abuse: Converging contributions to HAND. Brain Sci. 2017, 7, 25.
  99. MacDuffie, K.E.; Brown, G.G.; McKenna, B.S.; Liu, T.T.; Meloy, M.J.; Tawa, B.; Archibald, S.; Fennema-Notestine, C.; Atkinson, J.H.; Ellis, R.J.; et al. Effects of HIV Infection, methamphetamine dependence and age on cortical thickness, area and volume. Neuroimage Clin. 2018, 20, 1044–1052.
  100. Napier, T.C. Impact on Cortical Function of Cocaine Abuse Co-Occurring with HIV. Neuropsychopharmacology 2017, 42, 365–365.
  101. Rippeth, J.D.; Heaton, R.K.; Carey, C.L.; Marcotte, T.D.; Moore, D.J.; Gonzalez, R.; Wolfson, T.; Grant, I.; Group, H. Methamphetamine dependence increases risk of neuropsychological impairment in HIV infected persons. J. Int. Neuropsychol. Soc. 2004, 10, 1–14.
  102. Yelamanchili, S.V.; Lamberty, B.G.; Rennard, D.A.; Morsey, B.M.; Hochfelder, C.G.; Meays, B.M.; Levy, E.; Fox, H.S. MiR-21 in Extracellular Vesicles Leads to Neurotoxicity via TLR7 Signaling in SIV Neurological Disease. PLoS Pathog. 2015, 11, e1005032.
  103. András, I.E.; Leda, A.; Contreras, M.G.; Bertrand, L.; Park, M.; Skowronska, M.; Toborek, M. Extracellular vesicles of the blood-brain barrier: Role in the HIV-1 associated amyloid beta pathology. Mol. Cell. Neurosci. 2017, 79, 12–22.
  104. Tian, J.; Casella, G.; Zhang, Y.; Rostami, A.; Li, X. Potential roles of extracellular vesicles in the pathophysiology, diagnosis, and treatment of autoimmune diseases. Int. J. Biol. Sci. 2020, 16, 620–632.
  105. Lee, J.H.; Ostalecki, C.; Zhao, Z.; Kesti, T.; Bruns, H.; Simon, B.; Harrer, T.; Saksela, K.; Baur, A.S. HIV Activates the Tyrosine Kinase Hck to Secrete ADAM Protease-Containing Extracellular Vesicles. EBioMedicine 2018, 28, 151–161.
  106. Nazari, A.; Zahmatkesh, M.; Mortaz, E.; Hosseinzadeh, S. Effect of methamphetamine exposure on the plasma levels of endothelial-derived microparticles. Drug Alcohol Depend. 2018, 186, 219–225.
  107. Théry, C.; Witwer, K. ISEV2018 abstract book. J. Extracell. Vesicles 2018, 7, 1461450.
  108. Lemaire, Q.; Lefebvre, C.; Salzet, M.; Raffo-Romero, A.; Arab, T.; Van Camp, C.; LeMarrec-Crocq, F.; Vizioli, J.; Sautière, P.-E. Study of exosomal microRNAs from microglia involved in neuroprotection in Hirudo medicinalis. J. Extracell. Vesicles 2018, 7, 108.
  109. Sharma, H.; Chinnappan, M.; Agarwal, S.; Dalvi, P.; Gunewardena, S.; O’Brien-Ladner, A.; Dhillon, N.K. Macrophage-derived extracellular vesicles mediate smooth muscle hyperplasia: Role of altered miRNA cargo in response to HIV infection and substance abuse. FASEB J. 2018, 32, 5174–5185.
  110. Haque, S.; Kodidela, S.; Gerth, K.; Hatami, E.; Verma, N.; Kumar, S. Extracellular Vesicles in Smoking-Mediated HIV Pathogenesis and their Potential Role in Biomarker Discovery and Therapeutic Interventions. Cells 2020, 9, 864.
  111. Haque, S.; Sinha, N.; Ranjit, S.; Midde, N.M.; Kashanchi, F.; Kumar, S. Monocyte-derived exosomes upon exposure to cigarette smoke condensate alter their characteristics and show protective effect against cytotoxicity and HIV-1 replication. Sci. Rep. 2017, 7, 16120.
  112. Ranjit, S.; Patters, B.J.; Gerth, K.A.; Haque, S.; Choudhary, S.; Kumar, S. Potential neuroprotective role of astroglial exosomes against smoking-induced oxidative stress and HIV-1 replication in the central nervous system. Expert Opin. Ther. Targets 2018, 22, 703–714.
  113. Murphy, A.; Barbaro, J.; Martínez-Aguado, P.; Chilunda, V.; Jaureguiberry-Bravo, M.; Berman, J.W. The Effects of Opioids on HIV Neuropathogenesis. Front. Immunol. 2019, 10, 2445.
  114. Bokhari, S.M.; Hegde, R.; Callen, S.; Yao, H.; Adany, I.; Li, Q.; Li, Z.; Pinson, D.; Yeh, H.W.; Cheney, P.D.; et al. Morphine potentiates neuropathogenesis of SIV infection in rhesus macaques. J. Neuroimmune Pharmacol. Off. J. Soc. Neuroimmune Pharmacol. 2011, 6, 626–639.
  115. Hu, G.; Yao, H.; Chaudhuri, A.D.; Duan, M.; Yelamanchili, S.V.; Wen, H.; Cheney, P.D.; Fox, H.S.; Buch, S. Exosome-mediated shuttling of microRNA-29 regulates HIV Tat and morphine-mediated neuronal dysfunction. Cell. Death Dis. 2012, 3, e381.
  116. Wang, X.; Sun, L.; Zhou, Y.; Su, Q.J.; Li, J.L.; Ye, L.; Liu, M.Q.; Zhou, W.; Ho, W.Z. Heroin Abuse and/or HIV Infection Dysregulate Plasma Exosomal miRNAs. J. Neuroimmune Pharmacol. Off. J. Soc. Neuroimmune Pharmacol. 2019, doi:10.1007/s11481-019-09892-9.
  117. Roush, S.; Slack, F.J. The let-7 family of microRNAs. Trends Cell Biol. 2008, 18, 505–516.
  118. Kodidela, S.; Ranjit, S.; Sinha, N.; McArthur, C.; Kumar, A.; Kumar, S. Cytokine profiling of exosomes derived from the plasma of HIV-infected alcohol drinkers and cigarette smokers. PLoS ONE 2018, 13, e0201144.
  119. Kodidela, S.; Wang, Y.; Patters, B.J.; Gong, Y.; Sinha, N.; Ranjit, S.; Gerth, K.; Haque, S.; Cory, T.; McArthur, C.; et al. Proteomic Profiling of Exosomes Derived from Plasma of HIV-Infected Alcohol Drinkers and Cigarette Smokers. J. Neuroimmune Pharmacol. Off. J. Soc. Neuroimmune Pharmacol. 2019, doi:10.1007/s11481-019-09853-2.
  120. Kodidela, S.; Gerth, K.; Sinha, N.; Kumar, A.; Kumar, P.; Kumar, S. Circulatory Astrocyte and Neuronal EVs as Potential Biomarkers of Neurological Dysfunction in HIV-Infected Subjects and Alcohol/Tobacco Users. Diagnostics (Basel) 2020, 10, 349.
  121. Ibáñez, F.; Montesinos, J.; Ureña-Peralta, J.R.; Guerri, C.; Pascual, M. TLR4 participates in the transmission of ethanol-induced neuroinflammation via astrocyte-derived extracellular vesicles. J. Neuroinflamm. 2019, 16, 136.
  122. Coleman, L.G.; Zou, J.; Crews, F.T. Microglial-derived miRNA let-7 and HMGB1 contribute to ethanol-induced neurotoxicity via TLR7. J. Neuroinflamm. 2017, 14, 22.
  123. Li, H.; Li, C.; Zhou, Y.; Luo, C.; Ou, J.; Li, J.; Mo, Z. Expression of microRNAs in the serum exosomes of methamphetamine-dependent rats vs. ketamine-dependent rats. Exp. Ther. Med. 2018, 15, 3369–3375.
  124. Fries, G.R.; Lima, C.N.; Valvassori, S.S.; Zunta-Soares, G.; Soares, J.C.; Quevedo, J. Preliminary investigation of peripheral extracellular vesicles’ microRNAs in bipolar disorder. J. Affect. Disord. 2019, 255, 10–14.
  125. Wu, F.; Yin, Z.; Yang, L.; Fan, J.; Xu, J.; Jin, Y.; Yu, J.; Zhang, D.; Yang, G. Smoking Induced Extracellular Vesicles Release and Their Distinct Properties in Non-Small Cell Lung Cancer. J. Cancer 2019, 10, 3435–3443.
  126. Zhu, J.; Liu, B.; Wang, Z.; Wang, D.; Ni, H.; Zhang, L.; Wang, Y. Exosomes from nicotine-stimulated macrophages accelerate atherosclerosis through miR-21-3p/PTEN-mediated VSMC migration and proliferation. Theranostics 2019, 9, 6901–6919.
  127. Tseng, A.M.; Chung, D.D.; Pinson, M.R.; Salem, N.A.; Eaves, S.E.; Miranda, R.C. Ethanol Exposure Increases miR-140 in Extracellular Vesicles: Implications for Fetal Neural Stem Cell Proliferation and Maturation. Alcohol. Clin. Exp. Res. 2019, 43, 1414–1426.
  128. Meng, Y.; Ding, J.; Li, C.; Fan, H.; He, Y.; Qiu, P. Transfer of pathological alpha-synuclein from neurons to astrocytes via exosomes causes inflammatory responses after METH exposure. Toxicol. Lett. 2020, 331, 188–199.
  129. Sun, B.; Fernandes, N.; Pulliam, L. Profile of neuronal exosomes in HIV cognitive impairment exposes sex differences. AIDS 2019, 33, 1683–1692.
More
This entry is offline, you can click here to edit this entry!