Next Article in Journal
Revisiting the Role of Neurotrophic Factors in Inflammation
Previous Article in Journal
Adipose Stromal Cell Expansion and Exhaustion: Mechanisms and Consequences
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Extracellular Vesicles in Smoking-Mediated HIV Pathogenesis and their Potential Role in Biomarker Discovery and Therapeutic Interventions

College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Ave, Memphis, TN 38163, USA
*
Author to whom correspondence should be addressed.
Cells 2020, 9(4), 864; https://doi.org/10.3390/cells9040864
Submission received: 14 January 2020 / Revised: 16 March 2020 / Accepted: 30 March 2020 / Published: 2 April 2020
(This article belongs to the Section Cellular Pathology)

Abstract

:
In the last two decades, the mortality rate in people living with HIV/AIDS (PLWHA) has decreased significantly, resulting in an almost normal longevity in this population. However, a large portion of this population still endures a poor quality of life, mostly due to an increased inclination for substance abuse, including tobacco smoking. The prevalence of smoking in PLWHA is consistently higher than in HIV negative persons. A predisposition to cigarette smoking in the setting of HIV potentially leads to exacerbated HIV replication and a higher risk for developing neurocognitive and other CNS disorders. Oxidative stress and inflammation have been identified as mechanistic pathways in smoking-mediated HIV pathogenesis and HIV-associated neuropathogenesis. Extracellular vesicles (EVs), packaged with oxidative stress and inflammatory agents, show promise in understanding the underlying mechanisms of smoking-induced HIV pathogenesis via cell-cell interactions. This review focuses on recent advances in the field of EVs with an emphasis on smoking-mediated HIV pathogenesis and HIV-associated neuropathogenesis. This review also provides an overview of the potential applications of EVs in developing novel therapeutic carriers for the treatment of HIV-infected individuals who smoke, and in the discovery of novel biomarkers that are associated with HIV-smoking interactions in the CNS.

1. Introduction

Once a lethal pandemic, HIV has now taken the form of a chronic condition. As of 2018, at least 37.9 million people were living with HIV, with more than a million new cases each year [1]. The majority of these people living with HIV/AIDS (PLWHA) have a life expectancy comparable to healthy adults, which is attributed to remarkable advances in medicine, especially the introduction of combination anti-retroviral therapy (cART) [2,3,4,5]. However, a huge portion of PLWHA have a poor quality of life and suffer from high morbidity and mortality associated with drugs of abuse, including tobacco. More than 40% of PLWHA in the USA are cigarette smokers, which severely affect their life expectancy, reducing the average life span by over 6 years [6,7,8]. Mortality due to non-AIDS related malignancies is almost two-fold higher in HIV-positive smokers, irrespective of the use of cART [9]. Non-adherence to cART and/or attenuated treatment efficacy in HIV-positive smokers could possibly increase the risk of morbidity and mortality [10,11]. Smoking cessation can improve life expectancy, although studies have shown that smoking cessation is hard to achieve [12]. While the exact mechanistic pathway for smoking-mediated exacerbation of HIV pathogenesis is not fully understood, our studies have shown that tobacco smoke aggravates HIV pathogenesis, in part via the induction of cytochrome P450 (CYP)-mediated metabolism and activation of cigarette smoke constituents, resulting in oxidative stress [13,14,15,16,17,18]. In particular, we have demonstrated that benzo(a)pyrene (B(a)p), a potent component of cigarette smoke, exacerbates HIV replication via CYP-induced oxidative stress followed by the NF-κβ pathway [13].
Approximately 50% of PLWHA demonstrate a pattern of cognitive, motor, and behavioral dysfunction, cumulatively termed HIV-associated neurocognitive disorders (HAND) [19,20,21]. In the presence of cigarette smoke, the risk of peripheral neuropathy and HAND in PLWHA increases significantly [7,22,23,24,25]. Some reports demonstrate a conflicting impact of cigarette smoke on PLWHA in terms of neurocognitive disorders [26,27,28], which further strengthens the necessity to study whether cigarette smoking is a causative factor for HAND in PLWHA.
One possible mechanistic pathway of tobacco smoking-induced HIV pathogenesis and HAND could be the transportation of oxidative stress-related agents and inflammatory modulators via extracellular vesicles (EVs), commonly referred to as exosomes prior to 2018. EVs are biological nanoparticles and are released by almost all cells [29,30]. They are considered as both inter and intra-cellular messengers, able to modify their cargo according to the condition or stimulus affecting the parent cells [31,32], which upon internalization by recipient cells, can modulate the pathophysiological state in those cells [33,34]. EVs play an important role in HIV pathogenesis - either in improving or deteriorating the existing condition; however, the exact role of EVs in HIV pathogenesis is poorly understood [35,36]. Currently, only a handful of studies have investigated the role of EVs in smoking-mediated toxicity in the setting of HIV [37,38,39].
The complete eradication of HIV is not currently feasible, with a few exceptions [40,41], due to viral latency in cellular reservoirs, e.g. CD4 T cells, cells of the myeloid lineage (monocytes and macrophages) and dendritic cells [42,43,44,45]. Monocytes and macrophages are considered one of the most suitable cells for studying viral latency due to their long lifespan, as well as their ubiquitous presence throughout the body, including the brain [43,45,46]. EVs derived from monocytes and macrophages potentially have a profound effect on recipient cells [47,48]. For example, we have previously reported that EVs derived from uninfected monocytes protect recipient cells due to the specific packaging of protective elements [39], manuscript under revision). Conversely, HIV-infected macrophage-derived EVs lose this defense capacity, as evidenced by a higher viral load and increased cellular toxicity [39]. Proteomic and cytokine analyses of plasma EVs obtained from HIV-positive and negative smokers demonstrated a differential packaging of proteins in the EVs [37,49]. In addition, macrophage-derived EVs can readily cross the blood brain barrier, suggesting the potential role of EVs in either disseminating or alleviating HIV and HAND pathogenesis [30].
Evidently, there is a strong correlation between cigarette smoking and HIV and/or HAND pathogenesis as demonstrated via EVs. Nevertheless, there are unresolved questions to be answered. For example, what is the true nature/role of EVs in smoking-mediated HIV and HAND pathogenesis? Can we inhibit the viral transfection and oxidative stress through EVs? Is there any therapeutic application of EVs in this context? Can the components of EVs be used as biomarkers for HIV-tobacco smoking interactions that lead to HAND? Very recently, especially in the last five years, more and more studies are being conducted to answer these questions. This niche field has drawn researchers to connect the dots between HIV, cigarette smoking, HAND and EVs, and whether oxidative stress acts as a driving force to exacerbate the conditions. This review will explore possible answers to these questions, emphasizing investigations since 2015 and speculating on the potential role of EVs in HIV pathogenesis and HAND in tobacco smokers, as well as propose therapeutics to combat these conditions.

2. Extracellular Vesicles-a Sneak Peek

The discovery of extracellular vesicles (EVs), particularly exosomes-one particular type of EVs, dates back to the 1980s [50,51]. There are various classifications of EVs, including exosomes, extracellular vesicles, microsomes, ectosomes, prostasomes, oncosomes etc. The EVs that are generally formed in the multivesicular bodies within cells are exosomes. These are the nanosized vesicles (≤ 200nm), that capture the functional messages from cells and are thus the primary vesicles of interest [52,53,54,55,56,57,58]. Unfortunately, isolation and purification of exosomes from other sorts of extracellular vesicles released by the cells remain challenging. Thus, most of the isolated vesicles are a mixed population of exosomes, small extracellular vesicles, and microvesicles. Therefore, according to the guidelines provided by the Minimal information for studies of extracellular vesicles (MISEV), it is more appropriate to use the generic term extracellular vesicles (EVs) to designate these vesicles [29].
Ultracentrifugation is considered the gold standard method for the isolation of EVs from cell culture media and biological fluid [59]. However, this method has its own limitations that include the requirement for a large volume of starting fluid, co-precipitation of unwanted materials etc. [60]. Several isolation techniques have been developed to overcome these limitations, such as- sucrose gradient centrifugation with iodixanol, size-exclusion chromatography, polymer-based precipitation, antibody-specific EVs separation technique etc. [60,61,62,63,64]. Due to persistent discrepancies in EVs isolation procedures, the MISEV 2014 guideline recommended choosing the isolation method according to the need of the downstream analysis [65]. After successful isolation of EVs, it is equally important to characterize the particles. According to the MISEV 2014 guideline, molecular techniques to identify the presence of at least three or more exosome-enriched proteins (e.g. tetraspanin membrane proteins, heat shock proteins, membrane transporters and lipid-bound proteins), absence of cell-specific proteins (e.g. β-actin, GAPDH) [65]. At least two different biophysical techniques should be performed for biophysical characterization of EVs. These techniques could be optical particle tracking, including Transmission Electron Microscopy and atomic force microscopy [66,67,68]. Comparable data from nanoparticle-tracking analysis, dynamic light scattering, or resistive pulse sensing, are the basic requirements for EVs [65]. The contents of EVs vary greatly depending upon the condition of the parent cell. Thus, apart from characterizing the vesicles, identifying these contents reveals a breadth of information regarding the parent cells.
The unique nature of EVs is their role as intercellular messengers. The speculation started with the finding that EVs were involved in indirect activation of CD4-T cells by carrying major histocompatibility complex (MHC) and T cell costimulatory molecules [69,70]. In 2007, Valadi et al. took the EV field one-step forward by demonstrating that these vesicles packaged and shuttled mRNA and miRNA to recipient cells for translation to functional proteins [55,70]. Since then, extensive studies have revealed that EVs are released from nearly all kinds of mammalian cells [31,71,72,73,74]. The intricate role of EVs in disease pathogenesis is thus a matter of deep interest. The most studied is perhaps the role of EVs in cancer or tumor pathogenesis [75,76,77,78,79,80,81]. Additionally, EVs have been demonstrated to participate in disseminating pathogens like HIV, HCV, EBV, prions etc. [82,83,84,85,86]. The focus of this review will be in the field of research around EVs in HIV.

2.1. Advances Since 2015

2.1.1. Isolation Techniques and Characterization Methods

Recently, the isolation procedure of EVs has seen tremendous development. Rong et al. have demonstrated a sequential ultrafiltration method to isolate two subtypes of EVs (exosomes and microvesicles) from cancer cells. Although the sequential ultracentrifugation method allows for the separation of more purified exosomes, it requires a substantial amount of starting material [87]. Mengxi et al. have developed an integrated acoustics and microfluidics method to isolate EVs from undiluted whole blood, with high purity and yield in an automated system [88]. Furthermore, a commercial column method to isolate EVs is also currently available [89]. However, there is still potential for improvement, in terms of purity, yield, and the cost of the isolation procedure [90]. For the characterization of EVs, MISEV 2018 updated the 2014 guideline and suggested, “Both the source of EVs and the EV preparation must be described quantitatively”, with global quantification. With other guidelines being revised and updated, MISEV stressed that since the research field of EVs is highly diverse and continuously evolving, investigators have the flexibility to divert from the suggested guidelines with appropriate reasoning [29].

2.1.2. EVs as Intercellular Messengers

In addition to isolation techniques, knowledge of the role of EVs in cell-to-cell communication is also evolving rapidly. New insights are coming forward in terms of understanding cellular uptake of EVs. Some EVs demonstrate cell-specific uptake, while others are universally internalized by all cells. The exact reasons for this semi-selective nature is still to be discovered, however, some studies have identified specific receptor binding molecules on the surface of EVs that are responsible for intercellular communication [91]. Depending on the uptake process, the EVs can release their cargos at the plasma membrane of the recipient cells or go through endocytosis to be re-released within cells, with modified characteristics. Whether the EVs will have a functional or phenotypical impact on the recipient cells largely depends on the process by which EVs release their components. Therefore, there is a huge gap in knowledge to fully comprehend the true impact of EV-mediated intercellular communication. However, after internalization and release of cargos, the role of EVs in the physiological modulation of recipient cells is well-established [39,92,93,94,95,96,97]. Interestingly, the physiological effects of EVs are as diverse as their contents. Moreover, the potential of using EVs in biomarker identification has rapidly grown, which expanded from cancer biomarker identification to liquid biopsy in ADME (absorption, distribution, metabolism, and excretion) phenotyping of a drug [98,99,100]. Figure 1 is a diagram depicting basic characteristics of EVs and their potential applications.

3. Role of EVs in HIV

EVs have vital role in HIV pathogenesis. To begin with, EVs and HIV both share a multitude of similarities in terms of size, origination, and cellular release mechanisms [101]. EVs carry viral proteins like Negative Regulatory Factor (nef), transactivator of transcription (Tat) encoding protein, entry receptors like CCR5 etc. [102,103,104]. On the other hand, HIV can utilize the release pathway of EVs to disseminate viral transmission [103]. Again, it is well established that oxidative stress is involved in various stages of the HIV life cycle, including viral replication and the inflammatory response, ultimately leading to the destruction of T cells, as well nuclear and mitochondrial DNA in brain tissue [105,106,107,108,109,110]. However, it is still not clearly understood how these oxidative and immunomodulatory factors are translocated from cells of origin to neighboring cells or even to distant organs. Based on the role of EVs in cell-cell communication, it is hypothesized that EVs carry these agents from parent cells to recipient cells in distant places, including the brain, by crossing the blood brain barrier (BBB). Since 2015, studies have been exploring the potential role of EVs in transporting oxidative stress and immunomodulatory agents in the pathogenesis of HIV and HAND, which will be discussed in detail in the following section.

Advances Since 2015

It is well documented that HIV-infected populations have elevated levels of EVs in their plasma in comparison to healthy individuals [111]. EVs originated from HIV-infected cells can reveal important information about these cells as well as the phase of the infection. Most importantly, they can provide information about the cell’s exposure to stressors [112,113,114]. Sukrutha Chettimada et al. have shown that the EV cargos provide a wealth of information about immune responses and oxidative stress. Their study not only confirmed the increased level of plasma EVs in the HIV positive patients, but also showed that this increase directly correlated with the increase in oxidative stress marker (cystine, oxidized cys-gly). They also detected other oxidative stress markers such as CAT, PRDX1, PRDX2, and TXN, markers of EVs, immune markers, and Notch4 in plasma EVs by untargeted proteomic analysis [115]. Moreover, they showed reduction in the anti-inflammatory polyunsaturated fatty acid (PUFA) level, which are known to be responsible for sensitizing cells to oxidative stress and eventually leading to cell death and apoptosis [115].
The contents within EVs can be used as biomarkers for diagnostic purposes, which would be an excellent non-invasive method [116]. The therapeutic use of EVs as biomarkers has been utilized for cancer therapy for a while now [117]; however, it is still an emerging field in the case of HIV. For instance, EVs from PLWHA are enriched with miR-122 and miR-200a, which are known markers for liver disease [118]. HIV-associated neurocognitive disorder, demonstrated by Aβ deposition, is highly common in PLWHA, which is a well-established biomarker for Alzheimer’s disease. Interestingly, neuron derived EVs from HIV populations show high levels of Aβ, which can serve as a possible biomarker for Alzheimer’s disease in PLWHA [119,120]. Markers of general neurodegeneration, e.g. HMGB1 and NF-L, are also found in these EVs, suggesting their potential role in biomarker identification [120,121].
Taken together, EVs shuttle various biomarkers that could prove to be useful in clinical application. Moreover, as EVs can be collected in a noninvasive method without any surgical procedure, this would make them an ideal candidate for diagnostic applications.

4. Role of EVs in HIV and Tobacco-Smoking

Cigarette smoke contributes to the deterioration of pre-existing conditions of HIV by various mechanisms, one of them being the CYP-induced oxidative stress-mediated NFκβ pathway [17,18,122,123]. However, other studies challenge these observations, which prompts further investigation [124,125]. It is thought that oxidative stress conditions induced by cigarette smoking trigger the release of EVs, as EVs may play a key role in inflammatory pathways [126] and in some cases may mediate protective signals during oxidative stress, reducing cell death [127]. Our earlier studies revealed a higher expression of CYP1A1 and antioxidant enzyme mRNAs in EVs from CSC-exposed monocytes and point toward a protective role of EVs against CSC-mediated cytotoxicity [39]. Although the exacerbation of HIV pathogenesis via cigarette smoke constituents inducing oxidative stress has been frequently proposed in literatures, the role of EVs in this pathway is scarcely studied until 2015. The next section will discuss about the advances in this field in the last five years.

Advances Since 2015

Earlier studies have demonstrated that EVs are involved in the shuttling of biomarkers, which could be beneficial as a diagnostic tool. However, it is still largely unknown if a specific biomarker or agent is directly linked to cigarette smoking-mediated HIV pathogenesis. Our recent study with plasma EVs from HIV positive smokers has revealed some exciting information. First, we have shown the downregulation of properdin in the plasma EVs from HIV positive smokers, a protein in the human complementary system that interacts with viral proteins, suggesting a relatively high risk of secondary infections [49]. We also observed an alteration in the levels of CD9, CETP, and vitronectin (VTN) in plasma EVs from the HIV positive population; however, further studies are required to reach to any conclusion. We have also looked at the differential cytokine packaging within the same population [37]. We identified two cytokine/chemokines, IL-6 and MCP-1, which were significantly packaged within plasma EVs from HIV-infected smokers [37]. We further confirmed this finding by measuring their concentrations in the EVs derived from macrophages exposed to CSC and HIV, where IL-6 showed a similar trend (manuscript under review). These findings suggest that: a) EVs can potentially be used as a biomarker for smoking-mediated HIV pathogenesis and b) modulating EV-mediated HIV pathogenesis has potential for therapeutic intervention. Several other studies are also looking at potential biomarkers to indicate the possible association between HIV and tobacco smoking, as well as to predict the development of non-AIDS-related illnesses in HIV positive smokers [128,129,130]. Further, Il-6 stands out as a pro-inflammatory cytokine, which is elevated in HIV smokers [131]. In addition, Steel at el. have suggested that the chemokine RANTES could be one of the potential candidate biomarkers, which was elevated in their study with HIV positive smokers [128]. Our studies have also demonstrated that these agents are also highly packaged in EVs in the presence of HIV and smoking. Moreover, soluble CD14 and transforming growth factor beta (TGF-β1) are also reported to be elevated in virally suppressive HIV positive smokers [129]. Soluble CD14 is a biomarker for monocyte activation, which is also associated with cardiovascular disease [132]. On the other hand, TGF-β1 has a crucial role in regulating cells of immune system, in which increasing TGF-β1 was correlated to a defective T cell proliferation in an HIV-infected population [133]. Among these two, TGF-β1 is reported to be in EVs, however confirming its potential as a biomarker for smoking-mediated HIV progression warrants further investigation [134]. In recent years, more studies with regard to EVs in HAND have revealed that EVs influence the CNS, ultimately deteriorating HAND [21,120,135,136,137,138]. For example, HIV-induced microRNA-7 and cathepsin B were shown to be transported by EVs to neurons, resulting into neuronal damage [21,135]. Additionally, as previously mentioned, Pulliam et al. have identified several other biomarkers of neurodegeneration, which include L1CAM, Aβ, NF-L, and HMGB1, in the neuron-derived EVs from individuals with HAND [120]. Although cigarette smoking is postulated to be associated with exacerbated HIV and HAND pathogenesis, to our knowledge, to date no biomarker has been identified that could indicate the development of HAND in HIV positive smokers. A summary of the discussions above is summarized in Table 1.

5. Therapeutic Potential of Using EVs in HIV-Tobacco Smoking Comorbidity

We have already demonstrated the role of EVs in carrying pro- and anti-oxidant contents, functional molecules relevant to smoking and exacerbation of HIV and HAND. Evidently, EVs are gaining importance as biological vehicles to carry various molecules, including drugs. EVs are a natural nanocarrier, low immunogenic in nature, with semi-selective targeting capacity. Thus, EVs are capable of evading phagocytosis or engulfment by macrophages and lysosomal degradation and vascular occlusion, thereby circulating within the body for a longer time [139,140]. Moreover, as EVs potentially evade first pass metabolism, they have a greater potential of reaching target organs [138]. Therefore, the therapeutic potential of EVs in smoking and HIV can be discussed from two perspectives: a) EVs as drug carriers and b) EVs in biomarker identification.

5.1. EVs as Drug Carriers

5.1.1. In the Periphery

EV drug loading capacity and efficiency has been explored, mostly in the case of anti-cancer drugs, to induce immunity against tuberculosis, toxoplasmosis, and diptheria toxoid, for delivery of small interfering RNA (siRNA) and much more [139,141,142,143,144,145,146]. Undoubtedly, EVs are a highly promising option as a drug delivery system. However, loading EVs with anti-retroviral drugs (ART) to target HIV is barely studied. Recently, Zou et al. have demonstrated the efficacy of engineered EVs loaded with curcumin (chemodietary agent that suppresses HIV replication) or miR-143 (apoptosis-inducing miRNA) to suppress HIV replication in vitro and ex vivo systems of latent HIV reservoirs, which also successfully suppressed HIV envelope protein-expressing solid tissue tumor in a humanized mouse model [147]. siRNA-based therapy is also being largely explored to target the latent HIV reservoirs. Although highly promising, siRNA are short-lived and not well-equipped to penetrate major reservoirs [148]. Packaging siRNA in EVs can overcome these issues. In fact, mesenchymal stem cell and dendritic cell-derived EVs have already been utilized to deliver siRNA for other purposes [149,150]. Again, based on earlier studies, EVs can package CYP enzymes, which could play a major role in drug metabolism and oxidative stress [97,151]. Therefore, targeting heightened CYP levels in EVs by selective CYP inhibitors could be a way to neutralize the increased drug metabolism/drug resistance and subsequent oxidative stress. In a similar way, EVs loaded with AOEs can reduce smoking-exacerbated HIV replication mediated by oxidative stress. EVs could also be used in formulating personalized therapy by isolating EVs from the patient, which will significantly reduce the risk of immunogenicity.

5.1.2. In CNS

The ability of EVs to cross the BBB makes them efficient vehicles to reduce the burden of HAND. EVs, especially derived from macrophages, can readily cross BBB and deliver their cargos to resident macrophages [30]. EVs can be loaded with cART drugs to improve their efficacy in smoking-exacerbated, HIV-associated comorbidities, including HAND. Again, long-term usage of cART drugs can lead to CNS toxicity, in part via oxidative stress pathway [152,153]. Loading of EVs with the AOE, catalase, has already shown neuroprotective effects in in vitro and in vivo models of Parkinson’s disease [154]. Similarly, antioxidants in combination with ART can be loaded in EVs, which could be used as potential interventions in smoking-exacerbated HAND. Additionally, dendritic cell-derived EVs have been modified to deliver siRNA therapy to cross the BBB, which could provide a promising means of targeting HIV reservoirs in the brain [150].

5.1.3. Targeting EVs

EVs possess naturally semi-selective targeting properties, depending on their origin and composition. Although not completely determined yet, adhesion molecules like integrins, major histocompatibility complex class II molecules, tetraspanin complexes, lipid compositions, glycans, and liposomes found on the surface or inside the vesicles, are demonstrated to aid in targeting [91,155,156,157]. Several studies have suggested that EVs attain the homing pattern of their originating cells, suggesting that it is crucial to consider this natural tropism of EVs while developing therapeutic carriers [158,159]. Initiatives have been taken to develop engineered EVs to more specifically bind to the target cells by enriching with specific antibodies, nanobodies, peptides, nucleic acids, as well as using the pseudotyping method commonly used in altering viral tropism [145,160], further discussed in the review by Murphy et al. [91]. However, not many studies were performed to target EVs to combat HIV viral infection in the periphery and in CNS cells, let alone under the influence of cigarette smoking. Recently, the study performed by Zou et al. designed an HIV specific monoclonal antibody containing EVs, loaded them with curcumin/miRNA, which successfully targeted HIV reservoirs (in vitro and ex vivo) as well as an in vivo study to reduce viral replication [147]. Similarly, modifying EVs to target cellular reservoirs e.g. CCR5, CXCR4 co-receptors, and CD4, could be the next step to deliver targeted cART in these cells. As eradication of viral reservoirs remains a big challenge in HIV, EVs could potentially play a significant role in specifically targeting these populations.

5.2. EVs in Biomarker Identification

5.2.1. EVs in HIV

EV contents could serve as biomarkers for HIV, either for diagnosis or prognosis. Recognition of specific protein and RNA contents in plasma EVs from HIV positive population has been used for diagnostic and prognostic purposes, as well as for the determination of treatment efficacy [161]. Identifying specific miRNA levels in EVs has shown significant promise in this regard. EVs isolated from a small group of HIV-suppressed patients demonstrated that miR-21 was downregulated in HIV controlled patients, with a decreasing pattern of CD4 cell count [162]. In addition, EVs obtained from an HIV positive population without antiretroviral therapy are larger in size and greater in quantity, with a lower amount of miRNA-155 and miRNA-223 [111,163]. These findings suggest that EVs could potentially be used to indicate the stages of infection, as well as markers of response towards treatment. As mentioned earlier, neuron-derived EVs, isolated from the plasma, could serve as an excellent source of liquid biopsy biomarkers for identifying neurocognitive disorders in HIV [121,164]. Additionally, urinary EVs collected from HIV positive individuals may provide a good source of HIV biomarkers, including p24 [165]. In fact, commercial diagnostic tools are also being developed to identify HIV in patients [161,166]. Moreover, PLWHA have a significantly higher rate of neurocognitive disorders. As mentioned, Aβ deposition is an indicator of Alzheimer’s disease, which was also identified in the EVs of HIV positive individuals, suggesting the possibility of using Aβ in EVs as a biomarker [119].

5.2.2. EVs in HIV and Smoking Comorbidity

EVs have significant potential to be used as biomarkers of smoking-induced HIV-progression in a non-invasive way. EVs obtained from smokers could potentially be used in the diagnosis of smoking-induced diseases. Ryu et al. have summarized the potential biomarkers for lung cancer, chronic obstructive pulmonary disease, cardiovascular disease, and Non-small cell lung cancer, in the presence of cigarette smoke, which could be indicated by identifying miRNAs and protein content present in EVs derived from plasma, serum, bronchoalveolar lavage (BAL), and urine collected from patients [167].
In addition, a few possible markers, IL-6, RANTES, soluble CD14, and properdin have been identified within EVs isolated from the plasma of HIV smokers [37,49,128,129,131,132,133,134]. However, further studies are warranted to establish their true potential as biomarkers of smoking-induced HIV pathogenesis. Moreover, future investigations to discover biomarkers in plasma EVs, which are altered only in the presence of both HIV and cigarette smoking, will also be highly beneficial. In addition, studies to link cigarette smoking and HIV with HAND progression, as well as biomarker discovery, will strengthen our understanding of the gravity of the impact that smoking has on HIV.
Taken together, understanding the role of EVs in smoking-exacerbated conditions in HIV patients may provide opportunities to develop new therapeutic interventions for this subgroup of the HIV population. Figure 2 briefly summarizes the therapeutic potential of EVs.

6. Conclusion and Future Prospects

To address the questions initially proposed in this review, EVs are undoubtedly a major player in smoking-mediated HIV and HAND progression. It is yet to be determined how and to what extent EVs carry oxidative stress and inflammatory agents across the BBB and within the CNS as well as cause HIV pathogenesis and neuronal damage, especially in the presence of tobacco smoking. The prospect of EVs as biomarkers and therapeutic carriers is highly promising. However, in terms of the knowledge and potential of EVs in influencing smoking-mediated HIV pathogenesis and HAND, investigations are still at the very early stages. Therefore, there is a tremendous scope of research in this field.
EVs, particularly from macrophages, have the advantage of penetrating the highly selective, semipermeable BBB, allowing them a unique exposure to the resident cells of the CNS [30]. EVs from uninfected and HIV-infected macrophages, in the absence and/or presence of tobacco exposure, can potentially deliver oxidative (e.g. CYPs) and inflammatory elements (cytokines/chemokines) to the brain and exacerbate HIV pathogenesis and HAND. Therefore, future studies to investigate the role of EVs in CNS cells would be desirable. Further investigations to identify the specific role of EVs containing cytokines and chemokines (especially IL-6), as well as CYPs, AOEs, other proteins, mRNA, and miRNA on HIV and HIV-associated neuropathogenesis, are highly desirable.
Researchers have demonstrated the potential of EV-carrying AOEs to negate the mediation of oxidative stress in a CNS disease. However, in the case of inhibiting viral transfection by EVs-carrying agent, more studies are warranted. Thus, the use of EVs as therapeutic carriers in HIV viral suppression or targeting latency reservoirs is highly promising.
Similarly, EVs have a high potential in HIV infection stage diagnosis, prognosis, as well as biomarker discovery for smoking-mediated HIV and HAND progression. The potential of EVs is already under investigation in terms of biomarker identification, diagnosis, and prognosis of various conditions, and as therapeutic carriers of drugs in the periphery as well as in the CNS. Our studies and others have postulated the prospect of IL-6, RANTES, soluble CD14, and properdin being potential biomarkers for tobacco-enhanced HIV-pathogenesis [37,49,128,129,131,132,133,134]. Whether they could be used in actual therapeutic settings will require further study.
Therefore, knowledge about EVs can help us in devising novel therapeutic agents for treating tobacco-induced HIV and HIV-associated pathologies more efficiently. Future studies should be directed towards finding the mechanistic pathways to fully comprehend the role of EVs in smoking-mediated HIV and HAND pathogenesis, which can be employed to develop the clinical and therapeutic potentials of EVs.

Funding

This study is supported by the funding opportunity from the National Institute of Health (DA047178).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. HIV.Gov. The Global HIV/AIDS Epidemic. Available online: https://www.hiv.gov/hiv-basics/overview/data-and-trends/global-statistics (accessed on 31 March 2020).
  2. Teeraananchai, S.; Kerr, S.J.; Amin, J.; Ruxrungtham, K.; Law, M.G. Life expectancy of HIV-positive people after starting combination antiretroviral therapy: A meta-analysis. Hiv Med. 2017, 18, 256–266. [Google Scholar] [CrossRef] [PubMed]
  3. Samji, H.; Cescon, A.; Hogg, R.S.; Modur, S.P.; Althoff, K.N.; Buchacz, K.; Burchell, A.N.; Cohen, M.; Gebo, K.A.; Gill, M.J.; et al. Closing the gap: Increases in life expectancy among treated HIV-positive individuals in the United States and Canada. PLoS ONE 2013, 8, e81355. [Google Scholar] [CrossRef] [PubMed]
  4. Ray, M.; Logan, R.; Sterne, J.A.; Hernandez-Diaz, S.; Robins, J.M.; Sabin, C.; Bansi, L.; van Sighem, A.; de Wolf, F.; Costagliola, D.; et al. The effect of combined antiretroviral therapy on the overall mortality of HIV-infected individuals. Aids 2010, 24, 123–137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Romley, J.A.; Juday, T.; Solomon, M.D.; Seekins, D.; Brookmeyer, R.; Goldman, D.P. Early HIV treatment led to life expectancy gains valued at $80 billion for people infected in 1996-2009. Health Aff. 2014, 33, 370–377. [Google Scholar] [CrossRef] [Green Version]
  6. Reddy, K.P.; Parker, R.A.; Losina, E.; Baggett, T.P.; Paltiel, A.D.; Rigotti, N.A.; Weinstein, M.C.; Freedberg, K.A.; Walensky, R.P. Impact of Cigarette Smoking and Smoking Cessation on Life Expectancy Among People With HIV: A US-Based Modeling Study. J. Infect. Dis. 2016, 214, 1672–1681. [Google Scholar] [CrossRef]
  7. Reynolds, N.R. Cigarette smoking and HIV: More evidence for action. Aids Educ. Prev. Off. Publ. Int. Soc. Aids Educ. 2009, 21, 106–121. [Google Scholar] [CrossRef]
  8. Helleberg, M.; Afzal, S.; Kronborg, G.; Larsen, C.S.; Pedersen, G.; Pedersen, C.; Gerstoft, J.; Nordestgaard, B.G.; Obel, N. Mortality attributable to smoking among HIV-1-infected individuals: A nationwide, population-based cohort study. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2013, 56, 727–734. [Google Scholar] [CrossRef] [Green Version]
  9. Helleberg, M.; May, M.T.; Ingle, S.M.; Dabis, F.; Reiss, P.; Fatkenheuer, G.; Costagliola, D.; d’Arminio, A.; Cavassini, M.; Smith, C.; et al. Smoking and life expectancy among HIV-infected individuals on antiretroviral therapy in Europe and North America. Aids 2015, 29, 221–229. [Google Scholar] [CrossRef] [Green Version]
  10. Shuter, J.; Bernstein, S.L. Cigarette smoking is an independent predictor of nonadherence in HIV-infected individuals receiving highly active antiretroviral therapy. Nicotine Tob. Res. Off. J. Soc. Res. Nicotine Tob. 2008, 10, 731–736. [Google Scholar] [CrossRef]
  11. Kariuki, W.; Manuel, J.I.; Kariuki, N.; Tuchman, E.; O’Neal, J.; Lalanne, G.A. HIV and smoking: Associated risks and prevention strategies. Hiv Aids 2016, 8, 17–36. [Google Scholar] [CrossRef] [Green Version]
  12. Brath, H.; Grabovac, I.; Schalk, H.; Degen, O.; Dorner, T.E. Prevalence and Correlates of Smoking and Readiness to Quit Smoking in People Living with HIV in Austria and Germany. PLoS ONE 2016, 11, e0150553. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Ranjit, S.; Sinha, N.; Kodidela, S.; Kumar, S. Benzo(a)pyrene in Cigarette Smoke Enhances HIV-1 Replication through NF-kappaB Activation via CYP-Mediated Oxidative Stress Pathway. Sci. Rep. 2018, 8, 10394. [Google Scholar] [CrossRef] [PubMed]
  14. Rao, P.; Ande, A.; Sinha, N.; Kumar, A.; Kumar, S. Effects of Cigarette Smoke Condensate on Oxidative Stress, Apoptotic Cell Death, and HIV Replication in Human Monocytic Cells. PLoS ONE 2016, 11, e0155791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Kumar, S.; Rao, P.; Sinha, N.; Midde, N.M. Cytochrome P450 and Oxidative Stress as Possible Pathways for Alcohol- and Tobacco-Mediated HIV Pathogenesis and NeuroAIDS. In Neuropathology of Drug Addictions and Substance Misuse; V.R., P., Ed.; 2016; Volume 1, pp. 179–188. [Google Scholar]
  16. Ranjit, S.; Midde, N.M.; Sinha, N.; Patters, B.J.; Rahman, M.A.; Cory, T.J.; Rao, P.S.; Kumar, S. Effect of Polyaryl Hydrocarbons on Cytotoxicity in Monocytic Cells: Potential Role of Cytochromes P450 and Oxidative Stress Pathways. PLoS ONE 2016, 11, e0163827. [Google Scholar] [CrossRef]
  17. Ande, A.; McArthur, C.; Ayuk, L.; Awasom, C.; Achu, P.N.; Njinda, A.; Sinha, N.; Rao, P.S.; Agudelo, M.; Nookala, A.R.; et al. Effect of mild-to-moderate smoking on viral load, cytokines, oxidative stress, and cytochrome P450 enzymes in HIV-infected individuals. PLoS ONE 2015, 10, e0122402. [Google Scholar] [CrossRef] [Green Version]
  18. Ande, A.; McArthur, C.; Kumar, A.; Kumar, S. Tobacco smoking effect on HIV-1 pathogenesis: Role of cytochrome P450 isozymes. Expert Opin. Drug Metab. Toxicol. 2013, 9, 1453–1464. [Google Scholar] [CrossRef]
  19. Rumbaugh, J.A.; Tyor, W. HIV-associated neurocognitive disorders: Five new things. Neurology. Clin. Pract. 2015, 5, 224–231. [Google Scholar] [CrossRef] [Green Version]
  20. Sacktor, N.; McDermott, M.P.; Marder, K.; Schifitto, G.; Selnes, O.A.; McArthur, J.C.; Stern, Y.; Albert, S.; Palumbo, D.; Kieburtz, K.; et al. HIV-associated cognitive impairment before and after the advent of combination therapy. J. Neurovirology 2002, 8, 136–142. [Google Scholar] [CrossRef]
  21. Cantres-Rosario, Y.M.; Ortiz-Rodriguez, S.C.; Santos-Figueroa, A.G.; Plaud, M.; Negron, K.; Cotto, B.; Langford, D.; Melendez, L.M. HIV Infection Induces Extracellular Cathepsin B Uptake and Damage to Neurons. Sci. Rep. 2019, 9, 8006. [Google Scholar] [CrossRef]
  22. Strazza, M.; Pirrone, V.; Wigdahl, B.; Nonnemacher, M.R. Breaking down the barrier: The effects of HIV-1 on the blood-brain barrier. Brain Res. 2011, 1399, 96–115. [Google Scholar] [CrossRef] [Green Version]
  23. Atluri, V.S.; Hidalgo, M.; Samikkannu, T.; Kurapati, K.R.; Jayant, R.D.; Sagar, V.; Nair, M.P. Effect of human immunodeficiency virus on blood-brain barrier integrity and function: An update. Front. Cell. Neurosci. 2015, 9, 212. [Google Scholar] [CrossRef] [PubMed]
  24. Harrison, J.D.; Dochney, J.A.; Blazekovic, S.; Leone, F.; Metzger, D.; Frank, I.; Gross, R.; Hole, A.; Mounzer, K.; Siegel, S.; et al. The nature and consequences of cognitive deficits among tobacco smokers with HIV: A comparison to tobacco smokers without HIV. J. Neurovirology 2017, 23, 550–557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Chang, L.; Lim, A.; Lau, E.; Alicata, D. Chronic Tobacco-Smoking on Psychopathological Symptoms, Impulsivity and Cognitive Deficits in HIV-Infected Individuals. J. Neuroimmune Pharmacol. Off. J. Soc. Neuroimmune Pharmacol. 2017, 12, 389–401. [Google Scholar] [CrossRef] [PubMed]
  26. Bryant, V.E.; Kahler, C.W.; Devlin, K.N.; Monti, P.M.; Cohen, R.A. The effects of cigarette smoking on learning and memory performance among people living with HIV/AIDS. Aids Care 2013, 25, 1308–1316. [Google Scholar] [CrossRef] [Green Version]
  27. Wojna, V.; Robles, L.; Skolasky, R.L.; Mayo, R.; Selnes, O.; de la Torre, T.; Maldonado, E.; Nath, A.; Melendez, L.M.; Lasalde-Dominicci, J. Associations of cigarette smoking with viral immune and cognitive function in human immunodeficiency virus-seropositive women. J. Neurovirology 2007, 13, 561–568. [Google Scholar] [CrossRef] [Green Version]
  28. Tsima, B.; Ratcliffe, S.J.; Schnoll, R.; Frank, I.; Kolson, D.L.; Gross, R. Is Tobacco Use Associated with Neurocognitive Dysfunction in Individuals with HIV? J. Int. Assoc. Provid. Aids Care 2018, 17, 2325958218768018. [Google Scholar] [CrossRef] [Green Version]
  29. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Yuan, D.; Zhao, Y.; Banks, W.A.; Bullock, K.M.; Haney, M.; Batrakova, E.; Kabanov, A.V. Macrophage exosomes as natural nanocarriers for protein delivery to inflamed brain. Biomaterials 2017, 142, 1–12. [Google Scholar] [CrossRef]
  31. 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. [Google Scholar] [CrossRef]
  32. Greening, D.W.; Simpson, R.J. Understanding extracellular vesicle diversity-current status. Expert Rev. Proteom. 2018, 15, 887–910. [Google Scholar] [CrossRef]
  33. Latifkar, A.; Hur, Y.H.; Sanchez, J.C.; Cerione, R.A.; Antonyak, M.A. New insights into extracellular vesicle biogenesis and function. J. Cell Sci. 2019, 132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Kucharzewska, P.; Belting, M. Emerging roles of extracellular vesicles in the adaptive response of tumour cells to microenvironmental stress. J. Extracell. Vesicles 2013, 2. [Google Scholar] [CrossRef] [PubMed]
  35. Welch, J.L.; Stapleton, J.T.; Okeoma, C.M. Vehicles of intercellular communication: Exosomes and HIV-1. J. Gen. Virol. 2019, 100, 350–366. [Google Scholar] [CrossRef]
  36. Urbanelli, L.; Buratta, S.; Tancini, B.; Sagini, K.; Delo, F.; Porcellati, S.; Emiliani, C. The Role of Extracellular Vesicles in Viral Infection and Transmission. Vaccines 2019, 7, 102. [Google Scholar] [CrossRef] [Green Version]
  37. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. 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. [Google Scholar] [CrossRef] [PubMed]
  39. 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. [Google Scholar] [CrossRef] [Green Version]
  40. Hutter, G.; Nowak, D.; Mossner, M.; Ganepola, S.; Mussig, A.; Allers, K.; Schneider, T.; Hofmann, J.; Kucherer, C.; Blau, O.; et al. Long-term control of HIV by CCR5 Delta32/Delta32 stem-cell transplantation. New Engl. J. Med. 2009, 360, 692–698. [Google Scholar] [CrossRef] [Green Version]
  41. Gupta, R.K.; Abdul-Jawad, S.; McCoy, L.E.; Mok, H.P.; Peppa, D.; Salgado, M.; Martinez-Picado, J.; Nijhuis, M.; Wensing, A.M.J.; Lee, H.; et al. HIV-1 remission following CCR5Delta32/Delta32 haematopoietic stem-cell transplantation. Nature 2019, 568, 244–248. [Google Scholar] [CrossRef]
  42. Vanhamel, J.; Bruggemans, A.; Debyser, Z. Establishment of latent HIV-1 reservoirs: What do we really know? J. Virus Erad. 2019, 5, 3–9. [Google Scholar]
  43. Kruize, Z.; Kootstra, N.A. The Role of Macrophages in HIV-1 Persistence and Pathogenesis. Front. Microbiol. 2019, 10, 2828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Van Marle, G.; Church, D.L.; van der Meer, F.; Gill, M.J. Combating the HIV reservoirs. Biotechnol. Genet. Eng. Rev. 2018, 34, 76–89. [Google Scholar] [CrossRef] [PubMed]
  45. Wacleche, V.S.; Tremblay, C.L.; Routy, J.P.; Ancuta, P. The Biology of Monocytes and Dendritic Cells: Contribution to HIV Pathogenesis. Viruses 2018, 10, 65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Sung, J.M.; Margolis, D.M. HIV Persistence on Antiretroviral Therapy and Barriers to a Cure. Adv. Exp. Med. Biol. 2018, 1075, 165–185. [Google Scholar] [CrossRef] [PubMed]
  47. 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. Off. Publ. Fed. Am. Soc. Exp. Biol. 2018, 32, 5174–5185. [Google Scholar] [CrossRef]
  48. Tang, N.; Sun, B.; Gupta, A.; Rempel, H.; Pulliam, L. Monocyte exosomes induce adhesion molecules and cytokines via activation of NF-kappaB in endothelial cells. Faseb J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2016, 30, 3097–3106. [Google Scholar] [CrossRef] [Green Version]
  49. 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. [Google Scholar] [CrossRef]
  50. Pan, B.T.; Johnstone, R.M. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: Selective externalization of the receptor. Cell 1983, 33, 967–978. [Google Scholar] [CrossRef]
  51. Harding, C.; Stahl, P. Transferrin recycling in reticulocytes: pH and iron are important determinants of ligand binding and processing. Biochem. Biophys. Res. Commun. 1983, 113, 650–658. [Google Scholar] [CrossRef]
  52. Johnstone, R.M.; Adam, M.; Hammond, J.R.; Orr, L.; Turbide, C. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J. Biol. Chem. 1987, 262, 9412–9420. [Google Scholar]
  53. Raposo, G.; Nijman, H.W.; Stoorvogel, W.; Liejendekker, R.; Harding, C.V.; Melief, C.J.; Geuze, H.J. B lymphocytes secrete antigen-presenting vesicles. J. Exp. Med. 1996, 183, 1161–1172. [Google Scholar] [CrossRef] [PubMed]
  54. Zitvogel, L.; Regnault, A.; Lozier, A.; Wolfers, J.; Flament, C.; Tenza, D.; Ricciardi-Castagnoli, P.; Raposo, G.; Amigorena, S. Eradication of established murine tumors using a novel cell-free vaccine: Dendritic cell-derived exosomes. Nat. Med. 1998, 4, 594–600. [Google Scholar] [CrossRef] [PubMed]
  55. 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. [Google Scholar] [CrossRef] [Green Version]
  56. Rahman, M.A.; Patters, B.J.; Kodidela, S.; Kumar, S. Extracellular Vesicles: Intercellular Mediators in Alcohol-Induced Pathologies. J. Neuroimmune Pharmacol. Off. J. Soc. Neuroimmune Pharmacol. 2019. [Google Scholar] [CrossRef] [PubMed]
  57. Gheinani, A.H.; Vögeli, M.; Baumgartner, U.; Vassella, E.; Draeger, A.; Burkhard, F.C.; Monastyrskaya, K. Improved isolation strategies to increase the yield and purity of human urinary exosomes for biomarker discovery. Sci. Rep. 2018, 8, 3945. [Google Scholar] [CrossRef]
  58. Hessvik, N.P.; Llorente, A. Current knowledge on exosome biogenesis and release. Cell. Mol. Life Sci. Cmls 2018, 75, 193–208. [Google Scholar] [CrossRef] [Green Version]
  59. Heinemann, M.L.; Ilmer, M.; Silva, L.P.; Hawke, D.H.; Recio, A.; Vorontsova, M.A.; Alt, E.; Vykoukal, J. Benchtop isolation and characterization of functional exosomes by sequential filtration. J. Chromatography. A 2014, 1371, 125–135. [Google Scholar] [CrossRef]
  60. Momen-Heravi, F.; Balaj, L.; Alian, S.; Mantel, P.Y.; Halleck, A.E.; Trachtenberg, A.J.; Soria, C.E.; Oquin, S.; Bonebreak, C.M.; Saracoglu, E.; et al. Current methods for the isolation of extracellular vesicles. Biol. Chem. 2013, 394, 1253–1262. [Google Scholar] [CrossRef]
  61. Cantin, R.; Diou, J.; Belanger, D.; Tremblay, A.M.; Gilbert, C. Discrimination between exosomes and HIV-1: Purification of both vesicles from cell-free supernatants. J. Immunol. Methods 2008, 338, 21–30. [Google Scholar] [CrossRef]
  62. Thery, C.; Amigorena, S.; Raposo, G.; Clayton, A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr. Protoc. Cell Biol. 2006, Chapter 3, Unit 3.22. [Google Scholar] [CrossRef]
  63. Taylor, D.D.; Zacharias, W.; Gercel-Taylor, C. Exosome isolation for proteomic analyses and RNA profiling. Methods Mol. Biol. (Cliftonn. J.) 2011, 728, 235–246. [Google Scholar] [CrossRef]
  64. Tauro, B.J.; Greening, D.W.; Mathias, R.A.; Ji, H.; Mathivanan, S.; Scott, A.M.; Simpson, R.J. Comparison of ultracentrifugation, density gradient separation, and immunoaffinity capture methods for isolating human colon cancer cell line LIM1863-derived exosomes. Methods 2012, 56, 293–304. [Google Scholar] [CrossRef] [PubMed]
  65. Lotvall, J.; Hill, A.F.; Hochberg, F.; Buzas, E.I.; Di Vizio, D.; Gardiner, C.; Gho, Y.S.; Kurochkin, I.V.; Mathivanan, S.; Quesenberry, P.; et al. Minimal experimental requirements for definition of extracellular vesicles and their functions: A position statement from the International Society for Extracellular Vesicles. J. Extracell. Vesicles 2014, 3, 26913. [Google Scholar] [CrossRef] [PubMed]
  66. Dragovic, R.A.; Gardiner, C.; Brooks, A.S.; Tannetta, D.S.; Ferguson, D.J.; Hole, P.; Carr, B.; Redman, C.W.; Harris, A.L.; Dobson, P.J.; et al. Sizing and phenotyping of cellular vesicles using Nanoparticle Tracking Analysis. Nanomed. Nanotechnol. Biol. Med. 2011, 7, 780–788. [Google Scholar] [CrossRef] [Green Version]
  67. Aras, O.; Shet, A.; Bach, R.R.; Hysjulien, J.L.; Slungaard, A.; Hebbel, R.P.; Escolar, G.; Jilma, B.; Key, N.S. Induction of microparticle- and cell-associated intravascular tissue factor in human endotoxemia. Blood 2004, 103, 4545–4553. [Google Scholar] [CrossRef]
  68. Sharma, S.; Gillespie, B.M.; Palanisamy, V.; Gimzewski, J.K. Quantitative nanostructural and single-molecule force spectroscopy biomolecular analysis of human-saliva-derived exosomes. Langmuir Acs J. Surf. Colloids 2011, 27, 14394–14400. [Google Scholar] [CrossRef] [Green Version]
  69. Thery, C.; Duban, L.; Segura, E.; Veron, P.; Lantz, O.; Amigorena, S. Indirect activation of naive CD4+ T cells by dendritic cell-derived exosomes. Nat. Immunol. 2002, 3, 1156–1162. [Google Scholar] [CrossRef]
  70. Thery, C. Exosomes: Secreted vesicles and intercellular communications. F1000 biology reports 2011, 3, 15. [Google Scholar] [CrossRef]
  71. Van Niel, G.; Raposo, G.; Candalh, C.; Boussac, M.; Hershberg, R.; Cerf-Bensussan, N.; Heyman, M. Intestinal epithelial cells secrete exosome-like vesicles. Gastroenterology 2001, 121, 337–349. [Google Scholar] [CrossRef]
  72. Faure, J.; Lachenal, G.; Court, M.; Hirrlinger, J.; Chatellard-Causse, C.; Blot, B.; Grange, J.; Schoehn, G.; Goldberg, Y.; Boyer, V.; et al. Exosomes are released by cultured cortical neurones. Mol. Cell. Neurosci. 2006, 31, 642–648. [Google Scholar] [CrossRef]
  73. Wolfers, J.; Lozier, A.; Raposo, G.; Regnault, A.; Thery, C.; Masurier, C.; Flament, C.; Pouzieux, S.; Faure, F.; Tursz, T.; et al. Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming. Nat. Med. 2001, 7, 297–303. [Google Scholar] [CrossRef] [PubMed]
  74. Cossetti, C.; Iraci, N.; Mercer, T.R.; Leonardi, T.; Alpi, E.; Drago, D.; Alfaro-Cervello, C.; Saini, H.K.; Davis, M.P.; Schaeffer, J.; et al. Extracellular vesicles from neural stem cells transfer IFN-gamma via Ifngr1 to activate Stat1 signaling in target cells. Mol. Cell 2014, 56, 193–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Klein-Scory, S.; Tehrani, M.M.; Eilert-Micus, C.; Adamczyk, K.A.; Wojtalewicz, N.; Schnolzer, M.; Hahn, S.A.; Schmiegel, W.; Schwarte-Waldhoff, I. New insights in the composition of extracellular vesicles from pancreatic cancer cells: Implications for biomarkers and functions. Proteome Sci. 2014, 12, 50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Mrizak, D.; Martin, N.; Barjon, C.; Jimenez-Pailhes, A.S.; Mustapha, R.; Niki, T.; Guigay, J.; Pancre, V.; de Launoit, Y.; Busson, P.; et al. Effect of nasopharyngeal carcinoma-derived exosomes on human regulatory T cells. J. Natl. Cancer Inst. 2015, 107, 363. [Google Scholar] [CrossRef] [Green Version]
  77. Rolfo, C.; Castiglia, M.; Hong, D.; Alessandro, R.; Mertens, I.; Baggerman, G.; Zwaenepoel, K.; Gil-Bazo, I.; Passiglia, F.; Carreca, A.P.; et al. Liquid biopsies in lung cancer: The new ambrosia of researchers. Biochim. Et Biophys. Acta 2014, 1846, 539–546. [Google Scholar] [CrossRef] [Green Version]
  78. Wojtowicz, A.; Baj-Krzyworzeka, M.; Baran, J. Characterization and biological role of extracellular vesicles. Postepy Hig. I Med. Dosw. 2014, 68, 1421–1432. [Google Scholar] [CrossRef]
  79. Vella, L.J. The emerging role of exosomes in epithelial-mesenchymal-transition in cancer. Front. Oncol. 2014, 4, 361. [Google Scholar] [CrossRef] [Green Version]
  80. Villanueva, M.T. Small containers, important cargo. Nat. Rev. Cancer 2014, 14, 764–765. [Google Scholar] [CrossRef]
  81. Andaloussi, S.E.; Mager, I.; Breakefield, X.O.; Wood, M.J. Extracellular vesicles: Biology and emerging therapeutic opportunities. Nat. Reviews. Drug Discov. 2013, 12, 347–357. [Google Scholar] [CrossRef]
  82. Liu, Z.; Zhang, X.; Yu, Q.; He, J.J. Exosome-associated hepatitis C virus in cell cultures and patient plasma. Biochem. Biophys. Res. Commun. 2014, 455, 218–222. [Google Scholar] [CrossRef]
  83. Narayanan, A.; Iordanskiy, S.; Das, R.; Van Duyne, R.; Santos, S.; Jaworski, E.; Guendel, I.; Sampey, G.; Dalby, E.; Iglesias-Ussel, M.; et al. Exosomes derived from HIV-1-infected cells contain trans-activation response element RNA. J. Biol. Chem. 2013, 288, 20014–20033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Sampey, G.C.; Meyering, S.S.; Zadeh, M.A.; Saifuddin, M.; Hakami, R.M.; Kashanchi, F. Exosomes and their role in CNS viral infections. J. Neurovirology 2014, 20, 199–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Patman, G. Hepatitis: Exosomal route of HCV transmission exists in patients. Nat. Reviews. Gastroenterol. Hepatol. 2014, 11, 704. [Google Scholar] [CrossRef] [PubMed]
  86. Pegtel, D.M.; Peferoen, L.; Amor, S. Extracellular vesicles as modulators of cell-to-cell communication in the healthy and diseased brain. Philos. Trans. R. Soc. London. Ser. Bbiological Sci. 2014, 369. [Google Scholar] [CrossRef] [Green Version]
  87. Xu, R.; Simpson, R.J.; Greening, D.W. A Protocol for Isolation and Proteomic Characterization of Distinct Extracellular Vesicle Subtypes by Sequential Centrifugal Ultrafiltration. Methods Mol. Biol. 2017, 1545, 91–116. [Google Scholar] [CrossRef]
  88. Wu, M.; Ouyang, Y.; Wang, Z.; Zhang, R.; Huang, P.H.; Chen, C.; Li, H.; Li, P.; Quinn, D.; Dao, M.; et al. Isolation of exosomes from whole blood by integrating acoustics and microfluidics. Proc. Natl. Acad. Sci. USA 2017, 114, 10584–10589. [Google Scholar] [CrossRef] [Green Version]
  89. Welton, J.L.; Webber, J.P.; Botos, L.A.; Jones, M.; Clayton, A. Ready-made chromatography columns for extracellular vesicle isolation from plasma. J. Extracell. Vesicles 2015, 4, 27269. [Google Scholar] [CrossRef]
  90. Yamamoto, T.; Kosaka, N.; Ochiya, T. Latest advances in extracellular vesicles: From bench to bedside. Sci. Technol. Adv. Mater. 2019, 20, 746–757. [Google Scholar] [CrossRef] [Green Version]
  91. Murphy, D.E.; de Jong, O.G.; Brouwer, M.; Wood, M.J.; Lavieu, G.; Schiffelers, R.M.; Vader, P. Extracellular vesicle-based therapeutics: Natural versus engineered targeting and trafficking. Exp. Mol. Med. 2019, 51, 32. [Google Scholar] [CrossRef]
  92. Mathieu, M.; Martin-Jaular, L.; Lavieu, G.; Thery, C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat. Cell Biol. 2019, 21, 9–17. [Google Scholar] [CrossRef]
  93. Freeman, D.W.; Noren Hooten, N.; Eitan, E.; Green, J.; Mode, N.A.; Bodogai, M.; Zhang, Y.; Lehrmann, E.; Zonderman, A.B.; Biragyn, A.; et al. Altered Extracellular Vesicle Concentration, Cargo, and Function in Diabetes. Diabetes 2018, 67, 2377–2388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Juan, T.; Furthauer, M. Biogenesis and function of ESCRT-dependent extracellular vesicles. Semin. Cell Dev. Biol. 2018, 74, 66–77. [Google Scholar] [CrossRef] [PubMed]
  95. Macia, L.; Nanan, R.; Hosseini-Beheshti, E.; Grau, G.E. Host- and Microbiota-Derived Extracellular Vesicles, Immune Function, and Disease Development. Int. J. Mol. Sci. 2019, 21, 107. [Google Scholar] [CrossRef] [Green Version]
  96. Shahjin, F.; Guda, R.S.; Schaal, V.L.; Odegaard, K.; Clark, A.; Gowen, A.; Xiao, P.; Lisco, S.J.; Pendyala, G.; Yelamanchili, S.V. Brain-Derived Extracellular Vesicle microRNA Signatures Associated with In Utero and Postnatal Oxycodone Exposure. Cells 2019, 9, 21. [Google Scholar] [CrossRef] [Green Version]
  97. Rahman, M.A.; Kodidela, S.; Sinha, N.; Haque, S.; Shukla, P.K.; Rao, R.; Kumar, S. Plasma exosomes exacerbate alcohol- and acetaminophen-induced toxicity via CYP2E1 pathway. Sci. Rep. 2019, 9, 6571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Huang, C.; Liu, S.; Tong, X.; Fan, H. Extracellular vesicles and ctDNA in lung cancer: Biomarker sources and therapeutic applications. Cancer Chemother. Pharmacol. 2018, 82, 171–183. [Google Scholar] [CrossRef]
  99. Xu, R.; Rai, A.; Chen, M.; Suwakulsiri, W.; Greening, D.W.; Simpson, R.J. Extracellular vesicles in cancer - implications for future improvements in cancer care. Nat. Reviews. Clin. Oncol. 2018, 15, 617–638. [Google Scholar] [CrossRef]
  100. Rowland, A.; Ruanglertboon, W.; van Dyk, M.; Wijayakumara, D.; Wood, L.S.; Meech, R.; Mackenzie, P.I.; Rodrigues, A.D.; Marshall, J.C.; Sorich, M.J. Plasma extracellular nanovesicle (exosome)-derived biomarkers for drug metabolism pathways: A novel approach to characterize variability in drug exposure. Br. J. Clin. Pharmacol. 2019, 85, 216–226. [Google Scholar] [CrossRef] [Green Version]
  101. Dias, M.V.S.; Costa, C.S.; daSilva, L.L.P. The Ambiguous Roles of Extracellular Vesicles in HIV Replication and Pathogenesis. Front. Microbiol. 2018, 9, 2411. [Google Scholar] [CrossRef] [Green Version]
  102. Li, M.; Aliotta, J.M.; Asara, J.M.; Tucker, L.; Quesenberry, P.; Lally, M.; Ramratnam, B. Quantitative proteomic analysis of exosomes from HIV-1-infected lymphocytic cells. Proteomics 2012, 12, 2203–2211. [Google Scholar] [CrossRef]
  103. Soares, H. HIV-1 Intersection with CD4 T Cell Vesicle Exocytosis: Intercellular Communication Goes Viral. Front. Immunol. 2014, 5, 454. [Google Scholar] [CrossRef] [Green Version]
  104. Qiao, X.; He, B.; Chiu, A.; Knowles, D.M.; Chadburn, A.; Cerutti, A. Human immunodeficiency virus 1 Nef suppresses CD40-dependent immunoglobulin class switching in bystander B cells. Nat. Immunol. 2006, 7, 302–310. [Google Scholar] [CrossRef]
  105. Aquaro, S.; Scopelliti, F.; Pollicita, M.; Perno, C.F. Oxidative stress and HIV infection: Target pathways for novel therapies? Future Hiv Ther. 2008, 2, 327–338. [Google Scholar] [CrossRef]
  106. Gil, L.; Martinez, G.; Gonzalez, I.; Tarinas, A.; Alvarez, A.; Giuliani, A.; Molina, R.; Tapanes, R.; Perez, J.; Leon, O.S. Contribution to characterization of oxidative stress in HIV/AIDS patients. Pharmacol. Res. 2003, 47, 217–224. [Google Scholar] [CrossRef]
  107. Elbim, C.; Pillet, S.; Prevost, M.H.; Preira, A.; Girard, P.M.; Rogine, N.; Matusani, H.; Hakim, J.; Israel, N.; Gougerot-Pocidalo, M.A. Redox and activation status of monocytes from human immunodeficiency virus-infected patients: Relationship with viral load. J. Virol. 1999, 73, 4561–4566. [Google Scholar] [CrossRef] [Green Version]
  108. Kalinowska, M.; Bazdar, D.A.; Lederman, M.M.; Funderburg, N.; Sieg, S.F. Decreased IL-7 responsiveness is related to oxidative stress in HIV disease. PLoS ONE 2013, 8, e58764. [Google Scholar] [CrossRef] [Green Version]
  109. Aukrust, P.; Luna, L.; Ueland, T.; Johansen, R.F.; Muller, F.; Froland, S.S.; Seeberg, E.C.; Bjoras, M. Impaired base excision repair and accumulation of oxidative base lesions in CD4+ T cells of HIV-infected patients. Blood 2005, 105, 4730–4735. [Google Scholar] [CrossRef]
  110. Zhang, Y.; Wang, M.; Li, H.; Zhang, H.; Shi, Y.; Wei, F.; Liu, D.; Liu, K.; Chen, D. Accumulation of nuclear and mitochondrial DNA damage in the frontal cortex cells of patients with HIV-associated neurocognitive disorders. Brain Res. 2012, 1458, 1–11. [Google Scholar] [CrossRef]
  111. Hubert, A.; Subra, C.; Jenabian, M.A.; Tremblay Labrecque, P.F.; Tremblay, C.; Laffont, B.; Provost, P.; Routy, J.P.; Gilbert, C. Elevated Abundance, Size, and MicroRNA Content of Plasma Extracellular Vesicles in Viremic HIV-1+ Patients: Correlations With Known Markers of Disease Progression. Journal of acquired immune deficiency syndromes (1999) 2015, 70, 219–227. [Google Scholar] [CrossRef] [Green Version]
  112. De Toro, J.; Herschlik, L.; Waldner, C.; Mongini, C. Emerging roles of exosomes in normal and pathological conditions: New insights for diagnosis and therapeutic applications. Front. Immunol. 2015, 6, 203. [Google Scholar] [CrossRef] [Green Version]
  113. Madison, M.N.; Okeoma, C.M. Exosomes: Implications in HIV-1 Pathogenesis. Viruses 2015, 7, 4093–4118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Buzas, E.I.; György, B.; Nagy, G.; Falus, A.; Gay, S. Emerging role of extracellular vesicles in inflammatory diseases. Nat. Rev. Rheumatol. 2014, 10, 356–364. [Google Scholar] [CrossRef] [PubMed]
  115. Chettimada, S.; Lorenz, D.R.; Misra, V.; Dillon, S.T.; Reeves, R.K.; Manickam, C.; Morgello, S.; Kirk, G.D.; Mehta, S.H.; Gabuzda, D. Exosome markers associated with immune activation and oxidative stress in HIV patients on antiretroviral therapy. Sci. Rep. 2018, 8, 7227. [Google Scholar] [CrossRef] [PubMed]
  116. Lin, J.; Li, J.; Huang, B.; Liu, J.; Chen, X.; Chen, X.M.; Xu, Y.M.; Huang, L.F.; Wang, X.Z. Exosomes: Novel biomarkers for clinical diagnosis. TheScientificWorldJournal 2015, 2015, 657086. [Google Scholar] [CrossRef] [PubMed]
  117. Kumar, D.; Gupta, D.; Shankar, S.; Srivastava, R.K. Biomolecular characterization of exosomes released from cancer stem cells: Possible implications for biomarker and treatment of cancer. Oncotarget 2015, 6, 3280–3291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Murray, D.D.; Suzuki, K.; Law, M.; Trebicka, J.; Neuhaus Nordwall, J.; Johnson, M.; Vjecha, M.J.; Kelleher, A.D.; Emery, S. Circulating miR-122 and miR-200a as biomarkers for fatal liver disease in ART-treated, HIV-1-infected individuals. Sci. Rep. 2017, 7, 10934. [Google Scholar] [CrossRef] [PubMed]
  119. Kodidela, S.; Gerth, K.; Haque, S.; Gong, Y.; Ismael, S.; Singh, A.; Tauheed, I.; Kumar, S. Extracellular Vesicles: A Possible Link between HIV and Alzheimer’s Disease-Like Pathology in HIV Subjects? Cells 2019, 8, 968. [Google Scholar] [CrossRef] [Green Version]
  120. Pulliam, L.; Sun, B.; Mustapic, M.; Chawla, S.; Kapogiannis, D. Plasma neuronal exosomes serve as biomarkers of cognitive impairment in HIV infection and Alzheimer’s disease. J. Neurovirology 2019, 25, 702–709. [Google Scholar] [CrossRef]
  121. Sun, B.; Dalvi, P.; Abadjian, L.; Tang, N.; Pulliam, L. Blood neuron-derived exosomes as biomarkers of cognitive impairment in HIV. Aids (Lond. Engl.) 2017, 31, F9–F17. [Google Scholar] [CrossRef]
  122. Buhl, R.; Jaffe, H.A.; Holroyd, K.J.; Wells, F.B.; Mastrangeli, A.; Saltini, C.; Cantin, A.M.; Crystal, R.G. Systemic glutathione deficiency in symptom-free HIV-seropositive individuals. Lancet (Lond. Engl.) 1989, 2, 1294–1298. [Google Scholar] [CrossRef]
  123. Morris, A.; George, M.P.; Crothers, K.; Huang, L.; Lucht, L.; Kessinger, C.; Kleerup, E.C. HIV and chronic obstructive pulmonary disease: Is it worse and why? Proc. Am. Thorac. Soc. 2011, 8, 320–325. [Google Scholar] [CrossRef] [PubMed]
  124. Cole, S.B.; Langkamp-Henken, B.; Bender, B.S.; Findley, K.; Herrlinger-Garcia, K.A.; Uphold, C.R. Oxidative stress and antioxidant capacity in smoking and nonsmoking men with HIV/acquired immunodeficiency syndrome. Nutr. Clin. Pract. Off. Publ. Am. Soc. Parenter. Enter. Nutr. 2005, 20, 662–667. [Google Scholar] [CrossRef] [PubMed]
  125. Zhao, L.; Li, F.; Zhang, Y.; Elbourkadi, N.; Wang, Z.; Yu, C.; Taylor, E.W. Mechanisms and genes involved in enhancement of HIV infectivity by tobacco smoke. Toxicology 2010, 278, 242–248. [Google Scholar] [CrossRef]
  126. Camussi, G.; Deregibus, M.C.; Bruno, S.; Cantaluppi, V.; Biancone, L. Exosomes/microvesicles as a mechanism of cell-to-cell communication. Kidney Int. 2010, 78, 838–848. [Google Scholar] [CrossRef] [Green Version]
  127. Eldh, M.; Ekstrom, K.; Valadi, H.; Sjostrand, M.; Olsson, B.; Jernas, M.; Lotvall, J. Exosomes communicate protective messages during oxidative stress; possible role of exosomal shuttle RNA. PLoS ONE 2010, 5, e15353. [Google Scholar] [CrossRef] [Green Version]
  128. Steel, H.C.; Venter, W.D.F.; Theron, A.J.; Anderson, R.; Feldman, C.; Kwofie, L.; Cronje, T.; Arullapan, N.; Rossouw, T.M. Effects of Tobacco Usage and Antiretroviral Therapy on Biomarkers of Systemic Immune Activation in HIV-Infected Participants. Mediat. Inflamm. 2018, 2018, 8357109. [Google Scholar] [CrossRef] [Green Version]
  129. Rossouw, T.M.; Anderson, R.; Feldman, C. Impact of HIV infection and smoking on lung immunity and related disorders. Eur. Respir. J. 2015, 46, 1781–1795. [Google Scholar] [CrossRef] [Green Version]
  130. Agus Somia, I.K.; Merati, T.P.; Bakta, I.M.; Putra Manuaba, I.B.; Yasa, W.P.S.; Sukrama, I.D.M.; Suryana, K.; Wisaksana, R. High levels of serum IL-6 and serum hepcidin and low CD4 cell count were risk factors of anemia of chronic disease in HIV patients on the combination of antiretroviral therapy. Hiv/Aids 2019, 11, 133–139. [Google Scholar] [CrossRef] [Green Version]
  131. Borges, A.H.; O’Connor, J.L.; Phillips, A.N.; Ronsholt, F.F.; Pett, S.; Vjecha, M.J.; French, M.A.; Lundgren, J.D. Factors Associated With Plasma IL-6 Levels During HIV Infection. J. Infect. Dis. 2015, 212, 585–595. [Google Scholar] [CrossRef] [Green Version]
  132. Cioe, P.A.; Baker, J.; Hammer, J.; Kojic, E.M.; Onen, N.; Patel, P.; Kahler, C.W. Soluble CD14 and D-dimer are associated with smoking and heavy alcohol use in HIV-Infected dults. In Proceedings of Conference on Retroviruses and Opportunistic Infections, Boston, Massachusetts, 3–6 March 2014. [Google Scholar]
  133. Theron, A.J.; Anderson, R.; Rossouw, T.M.; Steel, H.C. The Role of Transforming Growth Factor Beta-1 in the Progression of HIV/AIDS and Development of Non-AIDS-Defining Fibrotic Disorders. Front. Immunol. 2017, 8, 1461. [Google Scholar] [CrossRef]
  134. Shelke, G.V.; Yin, Y.; Jang, S.C.; Lasser, C.; Wennmalm, S.; Hoffmann, H.J.; Li, L.; Gho, Y.S.; Nilsson, J.A.; Lotvall, J. Endosomal signalling via exosome surface TGFbeta-1. J. Extracell. Vesicles 2019, 8, 1650458. [Google Scholar] [CrossRef] [Green Version]
  135. Hu, G.; Niu, F.; Liao, K.; Periyasamy, P.; Sil, S.; Liu, J.; Dravid, S.M.; Buch, S. HIV-1 Tat-Induced Astrocytic Extracellular Vesicle miR-7 Impairs Synaptic Architecture. J. Neuroimmune Pharmacol. Off. J. Soc. Neuroimmune Pharmacol. 2019. [Google Scholar] [CrossRef]
  136. Yang, L.; Niu, F.; Yao, H.; Liao, K.; Chen, X.; Kook, Y.; Ma, R.; Hu, G.; Buch, S. Exosomal miR-9 Released from HIV Tat Stimulated Astrocytes Mediates Microglial Migration. J. Neuroimmune Pharmacol. Off. J. Soc. Neuroimmune Pharmacol. 2018, 13, 330–344. [Google Scholar] [CrossRef]
  137. Ojha, C.R.; Lapierre, J.; Rodriguez, M.; Dever, S.M.; Zadeh, M.A.; DeMarino, C.; Pleet, M.L.; Kashanchi, F.; El-Hage, N. Interplay between Autophagy, Exosomes and HIV-1 Associated Neurological Disorders: New Insights for Diagnosis and Therapeutic Applications. Viruses 2017, 9, 176. [Google Scholar] [CrossRef] [Green Version]
  138. Ha, D.; Yang, N.; Nadithe, V. Exosomes as therapeutic drug carriers and delivery vehicles across biological membranes: Current perspectives and future challenges. Acta Pharm. Sinica. B 2016, 6, 287–296. [Google Scholar] [CrossRef] [Green Version]
  139. Armstrong, J.P.; Holme, M.N.; Stevens, M.M. Re-Engineering Extracellular Vesicles as Smart Nanoscale Therapeutics. Acs Nano 2017, 11, 69–83. [Google Scholar] [CrossRef] [Green Version]
  140. Jiang, X.C.; Gao, J.Q. Exosomes as novel bio-carriers for gene and drug delivery. Int. J. Pharm. 2017, 521, 167–175. [Google Scholar] [CrossRef]
  141. Pascucci, L.; Cocce, V.; Bonomi, A.; Ami, D.; Ceccarelli, P.; Ciusani, E.; Vigano, L.; Locatelli, A.; Sisto, F.; Doglia, S.M.; et al. Paclitaxel is incorporated by mesenchymal stromal cells and released in exosomes that inhibit in vitro tumor growth: A new approach for drug delivery. J. Control. Release Off. J. Control. Release Soc. 2014, 192, 262–270. [Google Scholar] [CrossRef]
  142. Cocce, V.; Franze, S.; Brini, A.T.; Gianni, A.B.; Pascucci, L.; Ciusani, E.; Alessandri, G.; Farronato, G.; Cavicchini, L.; Sordi, V.; et al. In Vitro Anticancer Activity of Extracellular Vesicles (EVs) Secreted by Gingival Mesenchymal Stromal Cells Primed with Paclitaxel. Pharmaceutics 2019, 11, 61. [Google Scholar] [CrossRef] [Green Version]
  143. Garofalo, M.; Saari, H.; Somersalo, P.; Crescenti, D.; Kuryk, L.; Aksela, L.; Capasso, C.; Madetoja, M.; Koskinen, K.; Oksanen, T.; et al. Antitumor effect of oncolytic virus and paclitaxel encapsulated in extracellular vesicles for lung cancer treatment. J. Control. Release Off. J. Control. Release Soc. 2018, 283, 223–234. [Google Scholar] [CrossRef] [PubMed]
  144. Shtam, T.A.; Kovalev, R.A.; Varfolomeeva, E.Y.; Makarov, E.M.; Kil, Y.V.; Filatov, M.V. Exosomes are natural carriers of exogenous siRNA to human cells in vitro. Cell Commun. Signal. Ccs 2013, 11, 88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. 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. [Google Scholar] [CrossRef] [PubMed]
  146. Wahlgren, J.; De, L.K.T.; Brisslert, M.; Vaziri Sani, F.; Telemo, E.; Sunnerhagen, P.; Valadi, H. Plasma exosomes can deliver exogenous short interfering RNA to monocytes and lymphocytes. Nucleic Acids Res. 2012, 40, e130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Zou, X.; Yuan, M.; Zhang, T.; Wei, H.; Xu, S.; Jiang, N.; Zheng, N.; Wu, Z. Extracellular vesicles expressing a single-chain variable fragment of an HIV-1 specific antibody selectively target Env(+) tissues. Theranostics 2019, 9, 5657–5671. [Google Scholar] [CrossRef]
  148. Rao, D.D.; Vorhies, J.S.; Senzer, N.; Nemunaitis, J. siRNA vs. shRNA: Similarities and differences. Adv. Drug Deliv. Rev. 2009, 61, 746–759. [Google Scholar] [CrossRef]
  149. Munoz, J.L.; Bliss, S.A.; Greco, S.J.; Ramkissoon, S.H.; Ligon, K.L.; Rameshwar, P. Delivery of Functional Anti-miR-9 by Mesenchymal Stem Cell-derived Exosomes to Glioblastoma Multiforme Cells Conferred Chemosensitivity. Mol. Therapy. Nucleic Acids 2013, 2, e126. [Google Scholar] [CrossRef]
  150. 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. [Google Scholar] [CrossRef]
  151. Kumar, S.; Sinha, N.; Gerth, K.A.; Rahman, M.A.; Yallapu, M.M.; Midde, N.M. Specific packaging and circulation of cytochromes P450, especially 2E1 isozyme, in human plasma exosomes and their implications in cellular communications. Biochem. Biophys. Res. Commun. 2017, 491, 675–680. [Google Scholar] [CrossRef]
  152. Sharma, B. Oxidative stress in HIV patients receiving antiretroviral therapy. Curr. Hiv Res. 2014, 12, 13–21. [Google Scholar] [CrossRef]
  153. Akay, C.; Cooper, M.; Odeleye, A.; Jensen, B.K.; White, M.G.; Vassoler, F.; Gannon, P.J.; Mankowski, J.; Dorsey, J.L.; Buch, A.M.; et al. Antiretroviral drugs induce oxidative stress and neuronal damage in the central nervous system. J. Neurovirology 2014, 20, 39–53. [Google Scholar] [CrossRef] [Green Version]
  154. Haney, M.J.; Klyachko, N.L.; Zhao, Y.; Gupta, R.; Plotnikova, E.G.; He, Z.; Patel, T.; Piroyan, A.; Sokolsky, M.; Kabanov, A.V.; et al. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J. Control. Release Off. J. Control. Release Soc. 2015, 207, 18–30. [Google Scholar] [CrossRef] [Green Version]
  155. Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef] [Green Version]
  156. Mallegol, J.; Van Niel, G.; Lebreton, C.; Lepelletier, Y.; Candalh, C.; Dugave, C.; Heath, J.K.; Raposo, G.; Cerf-Bensussan, N.; Heyman, M. T84-intestinal epithelial exosomes bear MHC class II/peptide complexes potentiating antigen presentation by dendritic cells. Gastroenterology 2007, 132, 1866–1876. [Google Scholar] [CrossRef] [Green Version]
  157. Rana, S.; Yue, S.; Stadel, D.; Zoller, M. Toward tailored exosomes: The exosomal tetraspanin web contributes to target cell selection. Int. J. Biochem. Cell Biol. 2012, 44, 1574–1584. [Google Scholar] [CrossRef] [PubMed]
  158. Wiklander, O.P.; Nordin, J.Z.; O’Loughlin, A.; Gustafsson, Y.; Corso, G.; Mager, I.; Vader, P.; Lee, Y.; Sork, H.; Seow, Y.; et al. Extracellular vesicle in vivo biodistribution is determined by cell source, route of administration and targeting. J. Extracell. Vesicles 2015, 4, 26316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Kim, M.S.; Haney, M.J.; Zhao, Y.; Mahajan, V.; Deygen, I.; Klyachko, N.L.; Inskoe, E.; Piroyan, A.; Sokolsky, M.; Okolie, O.; et al. Development of exosome-encapsulated paclitaxel to overcome MDR in cancer cells. Nanomed. Nanotechnol. Biol. Med. 2016, 12, 655–664. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Kooijmans, S.A.; Aleza, C.G.; Roffler, S.R.; van Solinge, W.W.; Vader, P.; Schiffelers, R.M. Display of GPI-anchored anti-EGFR nanobodies on extracellular vesicles promotes tumour cell targeting. J. Extracell. Vesicles 2016, 5, 31053. [Google Scholar] [CrossRef] [PubMed]
  161. Vlassov, A.V.; Magdaleno, S.; Setterquist, R.; Conrad, R. Exosomes: Current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials. Biochim. Et Biophys. Acta 2012, 1820, 940–948. [Google Scholar] [CrossRef]
  162. Ruiz-de-Leon, M.J.; Jimenez-Sousa, M.A.; Moreno, S.; Garcia, M.; Gutierrez-Rivas, M.; Leon, A.; Montero-Alonso, M.; Gonzalez-Garcia, J.; Resino, S.; Rallon, N.; et al. Lower expression of plasma-derived exosome miR-21 levels in HIV-1 elite controllers with decreasing CD4 T cell count. J. Microbiol. Immunol. Infect. = Wei Mian Yu Gan Ran Za Zhi 2019, 52, 667–671. [Google Scholar] [CrossRef]
  163. Qiu, Y.; Ma, J.; Zeng, Y. Therapeutic Potential of Anti-HIV RNA-loaded Exosomes. Biomed. Environ. Sci. Bes 2018, 31, 215–226. [Google Scholar] [CrossRef]
  164. Hu, G.; Yelamanchili, S.; Kashanchi, F.; Haughey, N.; Bond, V.C.; Witwer, K.W.; Pulliam, L.; Buch, S. Proceedings of the 2017 ISEV symposium on "HIV, NeuroHIV, drug abuse, & EVs". J. Neurovirology 2017, 23, 935–940. [Google Scholar] [CrossRef]
  165. Dimov, I.; Jankovic Velickovic, L.; Stefanovic, V. Urinary exosomes. TheScientificWorldJournal 2009, 9, 1107–1118. [Google Scholar] [CrossRef] [PubMed]
  166. Aethlon. Available online: https://www.aethlonmedical.com/ (accessed on 31 March 2020).
  167. Ryu, A.R.; Kim, D.H.; Kim, E.; Lee, M.Y. The Potential Roles of Extracellular Vesicles in Cigarette Smoke-Associated Diseases. Oxidative Med. Cell. Longev. 2018, 2018, 4692081. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Basic characteristics and potential applications of extracellular vesicles upon exposure to HIV and tobacco smoke.
Figure 1. Basic characteristics and potential applications of extracellular vesicles upon exposure to HIV and tobacco smoke.
Cells 09 00864 g001
Figure 2. Therapeutic potential of using EVs in HIV-tobacco smoking comorbidity. siRNA: Small interfering RNA; miRNA: microRNA; MHCII: major histocompatibility complex II; L1CAM: L1 cell adhesion molecule; Aβ:Amyloid beta; Neurofilament Light; HMGB1: High mobility group box 1; IL-6: Interleukin 6; RANTES: Regulated on activation, normal T cell expressed and secreted; CD14: Cluster of differentiation14
Figure 2. Therapeutic potential of using EVs in HIV-tobacco smoking comorbidity. siRNA: Small interfering RNA; miRNA: microRNA; MHCII: major histocompatibility complex II; L1CAM: L1 cell adhesion molecule; Aβ:Amyloid beta; Neurofilament Light; HMGB1: High mobility group box 1; IL-6: Interleukin 6; RANTES: Regulated on activation, normal T cell expressed and secreted; CD14: Cluster of differentiation14
Cells 09 00864 g002
Table 1. Role of extracellular vesicles in HIV, tobacco smoking, and HIV and smoking.
Table 1. Role of extracellular vesicles in HIV, tobacco smoking, and HIV and smoking.
HIV-Associated EV ComponentHIV + Smoking-Associated Component
TypeSpecificTypeSpecific
Viral proteinsNef, Tat [104]CytokinesIL-6 10, MCP-1 11, RANTES 12 [37,128,131]
Viral entry receptorsCCR5 1 [103]OthersProperdin [49], soluble CD144, TGF-β1 13 [129]
Oxidative stress markerscystine, oxidized cys-gly, Catalase, PRDX1 2, PRDX2 2, and TXN 3 [115]
Anti-inflammatory markerPUFA [115]
Immune activation markersCD14 4, CRP 5, HLA-A, and HLA-B 6 [115]
NeurodegenerationMarkers7, HMGB1 8 and NF-L 9 [119,120,121]
1 C-C chemokine receptor type 5; 2 Peroxiredoxin 1 and 2; 3 Thioredoxin-1; 4 Cluster of differentiation14; 5 C-reactive protein; 6 human leukocyte antigen major histocompatibility complex, class I, A & B; 7 Amyloid beta; 8 High mobility group box 1 protein; 9 Neurofilament light; 10 Interleukin 6; 11 Monocyte chemoattractant protein-1; 12 Regulated on activation, normal T cell expressed and secreted; 13 Transforming growth factor beta-1.

Share and Cite

MDPI and ACS Style

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. https://doi.org/10.3390/cells9040864

AMA Style

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(4):864. https://doi.org/10.3390/cells9040864

Chicago/Turabian Style

Haque, Sanjana, Sunitha Kodidela, Kelli Gerth, Elham Hatami, Neha Verma, and Santosh Kumar. 2020. "Extracellular Vesicles in Smoking-Mediated HIV Pathogenesis and their Potential Role in Biomarker Discovery and Therapeutic Interventions" Cells 9, no. 4: 864. https://doi.org/10.3390/cells9040864

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