Skip to main content
Erschienen in: Virology Journal 1/2024

Open Access 01.12.2024 | Review

Exosome-mediated regulation of inflammatory pathway during respiratory viral disease

verfasst von: Hamidreza Gheitasi, Mohammad Sabbaghian, Ali Akbar Shekarchi, Amir Ali Mirmazhary, Vahdat Poortahmasebi

Erschienen in: Virology Journal | Ausgabe 1/2024

Abstract

Viruses have developed many mechanisms by which they can stimulate or inhibit inflammation and cause various diseases, including viral respiratory diseases that kill many people every year. One of the mechanisms that viruses use to induce or inhibit inflammation is exosomes. Exosomes are small membrane nanovesicles (30–150 nm) released from cells that contain proteins, DNA, and coding and non-coding RNA species. They are a group of extracellular vesicles that cells can take up to produce and mediate communication. Intercellular effect exosomes can deliver a broad confine of biological molecules, containing nucleic acids, proteins, and lipids, to the target cell, where they can convey therapeutic or pathogenic consequences through the modulation of inflammation and immune processes. Recent research has shown that exosomes can deliver entire virus genomes or virions to distant target cells, then the delivered viruses can escape the immune system and infect cells. Adenoviruses, orthomyxoviruses, paramyxoviruses, respiratory syncytial viruses, picornaviruses, coronaviruses, and rhinoviruses are mostly related to respiratory diseases. In this article, we will first discuss the current knowledge of exosomes. We will learn about the relationship between exosomes and viral infections, and We mention the inflammations caused by viruses in the airways, the role of exosomes in them, and finally, we examine the relationship between the viruses as mentioned earlier, and the regulation of inflammatory pathways that play a role in causing the disease.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

The respiratory system includes a set of organs of the human body that perform gas exchange between the human body and the outside air, organs such as the nose, pharynx, larynx, trachea, bronchi, lungs, etc. are among the organs. Which are widely used in the human respiratory system and are among the vital human organs [1]. Respiratory diseases including chronic obstructive pulmonary disease (COPD), asthma and obstructive sleep apnea (OSA), and acute conditions such as pneumonia have a high prevalence in the world population, and numerous reports show their gradual increase and have health and economic consequences. It has attracted the attention of many doctors and researchers [2]. Therefore, it is essential to identify and control the factors involved in respiratory diseases. Considering that various antiviral drugs such as oseltamivir and zanamivir have been proposed for respiratory diseases caused by the influenza virus, but the main problem here is that there is no clinically approved antiviral drug for common respiratory viruses, and there is also a practical vaccine does not have [3, 4]. Mutual recognition between viral respiratory infections and host responses and their associated molecular mechanisms has recently been established [5]. For example, during virus replication, infected cells may secrete cytokines that alter the local inflammation. Inflammation in the airways creates an anti-viral state, called type 1 inflammation, which can ultimately lead to the virus being removed [6, 7].
Microorganisms play a significant function in respiratory disease, for example, viruses and bacteria can cause respiratory diseases [8]. Respiratory syncytial viruses, influenza, parainfluenza, rhinoviruses, bocaviruses, and viruses of the coronavirus can cause respiratory diseases [914]. These viruses can cause respiratory problems in different ways; for example, they can transfer their DNA or RNA to exosomes [15].
Since viruses are tiny organisms with parasitic life, research has found a close relationship between viruses and exosomes [16]. Exosomes are phospholipid nanovesicles with cup-shaped morphology and diameters between 30 and 150 nm, found in blood, urine, and almost all eukaryotic fluids and cell culture media, and can be isolated using it [17]. Recent research has shown that exosomes act in two ways during viral infections. They can deliver the viral genome to the target cell or they can change the physiology of the target cell to facilitate infection [18].
In general, exosomes show an attractive field of research that promises a wide range of applications in medicine, and investigating the relationship between viruses and infections caused by them with blisters can be very helpful in exosome-based treatments and diagnostics. In this review, we do our best to provide an overview of the effects of respiratory infectious viruses and exosomes.

Biogenesis, composition, and function of exosomes

The term exosome was first used for extracellular vesicles (EVs) in 1981. In 1983, research showed that exosomes derived from reticulocytes can fuse with the plasma membrane and release their contents through exocytosis [19, 20]. EV is a general term for particles that are naturally removed from the cell and are bound by a lipid bilayer and cannot replicate, i.e. they do not contain a functional nucleus [21]. As mentioned, exosomes are particles between 30 and 150 nm that are secreted by most cells, and they are small extracellular vesicles that are derived from endosomes. Before being released into the external environment, they are formed and stored inside multivesicular bodies (MVBs) [22], as seen in Fig. 1. The different proteins and lipids that exist in the membrane of exosomes are caused by the phospholipid membrane that exosomes have acquired from the parental cells [23]. Phosphatidylcholine, phosphatidylethanolamines, phosphatidylinositol, phosphatidylserine, and sphingomyelin are present in the exosome membrane [24]. The high stability of exosomes in body fluids and at different pH depends on the composition and levels of these lipid molecules. This is due to the presence of high levels of sphingomyelin and phosphatidylinositol in their membranes [25]. Exosomes can be secreted from most cells in both physiological and pathological conditions, and by transferring vital genetic materials such as miRNA, mRNA, and DNA, as well as proteins and microorganisms such as viruses, they play an essential role in cellular communication and epigenetic regulation. As a result, they can be used to diagnose diseases based on exosomes and treatment methods [26, 27]. In culture, exosomes are also released from several cells, including B cells, dendritic cells (DCs), T cells, mast cells, and epithelial cells [28].
During biogenesis, the early exosomes formed by the endosomal system mature into late endosomes; during, this process, the contents of the endosomes construction due to the action of the v-ATPase booster pump, as well as the replacement of late endosome markers Rab 7 and -9. Rab. with mannose 6-phosphate receptor instead of 5-Rab [30]. After the formation of mature late endosomes, intraluminal vesicles (ILVs) are formed and accumulate in their lumen. There are two mechanisms for forming ILVs: endosomal sorting complex required for transport (ESCRT)-dependent and ESCRT-independent [30].
ESCRT-related mechanism: several mechanisms and molecules are involved in the formation of ILV, and the known ESCRT-related tool is the most ubiquitin-related [31]. ESCRT machines with SNARE producers and their effectors such as RAB GTPase play a prominent role in this process [32]. ESCRT consists of four complexes, ESCRT -0, ESCRT -I, ESCRT-II, and ESCRT-III, each having related functions (VPS4, Tsg101, and ALIX—classified lipid domains. ESCRT-I and ESCRT-II cause membrane deformation to form a stable membrane neck, and the use of the Vps4 complex in ESCRT-III causes vesicle neck cleavage and the separation of the ESCRT-III complex [33].
Non-ESCRT-dependent mechanism: Research shows that exosome biogenesis can have an EST-independent pathway, for example, showing that 4 ESCRT complexes are silenced. Intraluminal vesicles can still form in multivesicular bodies [34]. Non-ESCRT-dependent mechanism: research shows that exosome biogenesis can have an EST-independent pathway, for example, showing that 4 ESCRT complexes are silenced. Intraluminal vesicles can still form in multivesicular bodies [35]. Generally, non-ESCRT-dependent mechanisms can be investigated with two categories: lipid-mediated biogenesis and tetraspanin-mediated biogenesis.
Lipid-Mediated Biogenesis: Exosomes are enriched with cholesterol, sphingolipids, and ceramide. Neutral sphingomyelinase two and phospholipase D2 converts sphingomyelin to ceramide and phosphatidylcholine to phosphatidic acid (PA), respectively [36]. Ceramide and PA then form a cone-like structure that eventually leads to the formation of ILVs released as EVs [37].
Tetraspanin-mediated biogenesis: One of the ESCRT-independent regulators of exosome biogenesis that are highly enriched within exosomes and have been explicitly used as exosome biomarkers over the years is the tetraspanin family, of which CD9 is specifically used as an exosome marker [38].
The composition of exosomes depends on various factors, and it can also partially depend on the type of cell and is affected by different cell states; it, can also depend on its biogenesis, and it can also be determined by biological processes such as reverse budding of the plasma membrane [39]. According to the origin and biogenesis of exosomes, the composition of exosomes includes three major types of biomolecules: (a) proteins, such as proteins involved in membrane transport and fusion, heat shock proteins, Alix proteins, TSG101 and lipid-related proteins and phospholipases. (b) Lipids: lipids such as ceramides, cholesterol, long chain glycerophospholipids and sphingolipids and types of RNAs including mRNA, miRNA and other non-coding RNAs, tRNA and rRNA [4042].
Exosome has a wide range of functions, the most important of which is the exchange of information and transfer of materials between cells. Exosomes communicate with cells by three main mechanisms: either they bind to the receptor of the target cell, or they combine directly with the membrane of the target cell, or they enter the target cell by endocytosis, which endocytosis can be clathrin-dependent or clathrin-independent. To occur, exosomes are heterogeneous in terms of size and content are released by different cell types and can regulate the biological activity of target cells by transferring proteins, lipids, and nucleic acids, for example, they are involved in various biological processes such as angiogenesis, antigen presentation, apoptosis, and inflammation play a role. Furthermore, research has demonstrated a strong correlation between illnesses and the molecular constituents present within exosomes [4347].

Role of exosome in the regulation of inflammation

In the last decade, studies reported that exosomes generated by various cell types have promising biological activity in not only promoting cell proliferation, angiogenesis, cancer cell metastasis, and invasion, but also reprogramming metabolic processes, maintaining physiological homeostasis, regulating inflammation, and modulating the immune evasion, etc. [48, 49]. This evidence sheds light on the potential therapeutic effects of exosomes for different chronic inflammatory diseases and tumors [50]. As summarized in Table 1, exosomes can play dual pro-inflammatory and anti-inflammatory roles during inflammation.
Table 1
Regulation of inflammation by exosome cargo
Exosome derived from
Disease
Mechanism
Pathway
Effect
References
Monocytes
Tumor cell
Exosomes derived from monocytes secrete various inflammatory cytokines, including IL6, IL-1β, IL-8, and TNF-α
STAT3 and NFκB
Pro-inflammatory effects
[51]
Macrophages
Breast and stomach tumor
Exosomes derived from macrophages in breast and stomach tumors stimulate the production of inflammatory cytokines, including GCSF, IL-6, IL-8, IL1β, CCL2, and TNF-α
NF-κB
Pro-inflammatory effects
[52]
Dendritic cells
Tumor cell
Induction of IL6 and Secretion of TGFβ
STAT3
Pro-inflammatory effects
[51, 53]
Myeloid-derived suppressor cells (MDSC)
Tumor cell
Induction of the proinflammatory cytokine Cox 2 and an increase in inflammatory cytokines such as IL-6, TNF-α, VEGF, CCL2
STAT3
Pro-inflammatory effects
[54]
Natural killer cells
Tumor
Inhibition of NK cell activation mediated by IL-2
By inducing Smad phosphorylation, it can disrupt cytotoxicity and reduce NKG2D receptor expression
Anti-inflammatory effects
[55]
Regulatory T cells (Treg)
Tumor
Expansion phosphorylation of relevant transcription factors IL-10 and TGF- β1
immunosuppression
Anti-inflammatory effects
[56]
Macrophages
_
Decreased expression of IL-6
Decreased expression of IL-6
It can also act as a negative regulator in the JAK/STAT pathway
Anti-inflammatory effects
[57, 58]
Today, inflammation is considered the leading cause of many respiratory diseases, which is associated with a high rate of complications and mortality [59]. Many blood vessels in the lung can cause the release of membrane mucins from exosomes derived from lung endothelial cells, which facilitates the innate defense of the airway [60]. As mentioned, nowadays, exosomes have received a lot of attention in various inflammatory lung diseases such as COPD, asthma, acute lung injury (ALI), and COVID-19. In the meantime, COPD is obtained from the damage of endothelial cells and epithelial cells along with epithelial-mesenchymal transition (EMT) in the lung parenchyma [61, 62]. The secretion of exosomes from lung endothelial cells significantly increases with exposure to smoke and infection in the lungs of COPD subjects, which leads to increased production of IL-8, followed by continuous damage and inflammation in the lung tissue [63]. After injury has occurred, the production of large numbers of exosomes derived from lung endothelial cells in the bronchoalveolar lavage fluid (BALF) can alter the function of receptor cells, which can predict the level of COPD injury [64]. In research by Lee et al., they indicated that alveolar macrophages and alveolar epithelial cells type I are the primary sources of exosomes in BALF of ARDS and ALI [65]. The production of more exosomes, as well as the increase of eosinophils in the airways, can improve asthma in most cases [66, 67]. COVID-19 is another cause of severe lung inflammation. Based on the analysis, exosomes derived from COVID-19 patients contain some molecules, such as complement subunits C1r and C1s, which can stimulate inflammatory responses [68]. Exosomes may be used in the production of antiviral vaccines as well as pharmaceutical platforms, which are due to the intercellular communication activity and the distinct structure of the two lipid layers of exosomes. Also, the development of lung cancer can also be one of the cases in which exosomes are used in. They play a role in the way that exosomes secreted by the tumor can affect angiogenesis, EMT, etc. [6971]. All the above shows the high importance of exosomes and shows that exosomes can have a dual role that can be very useful for therapeutic work.

Regulation of exosome biogenesis by viral proteins

Virus regulates exosome production

Millions of people worldwide are affected by microbial diseases such as bacterial, parasitic, fungal, and viral diseases. In recent years, the role of exosomes has been much noticed, and the discovery of their relationship with viruses has progressed a lot [72, 73]. According to previous studies, it has been found that viruses can hijack the path of exosomes to facilitate the germination, accumulation, or release of the virus, and these hijacked exosomes help the virus escape from the immune system and spread in the body [74]. As mentioned, exosomes can act as viral carriers and directly contribute to virus replication; the first evidence to understand this issue came when researchers realized the importance of ESCRT components during viral capsid packaging and the maturation of several enveloped viruses [17]. Research has proven that viruses can regulate the biogenesis of exosomes, which play an essential role in virus and pathogen transmission. For example, the nef protein of HIV mediates the hijacking of exosome biogenesis. It directs proteins for integration into exosomes, or during infection with respiratory syncytial virus (RSV), it causes the cell to secrete more exosomes and change exosome components that are in N, F, G proteins of RSV virus are involved in this. Also, during hepatitis C virus infection, the viral genome can enter ILVs and later be secreted in exosomes. In the case of Epstein-Barr virus (EBV), exosomes may contain viral proteins that cause infection, or herpes simplex virus-1 (HSV-1)-derived exosomes may contain microRNAs that produce host-associated proteins. Virus-induced exosomes modulate antiviral immunity [72, 7477].
Based on recent research, it has been found that exosome production from cells can be increased by exposure to viruses [78]. In a way, the number of late endosomes/MVBs could be enhanced by viral protein expression in different cells, such as human T lymphocytes [79], CIITA, HeLa, and Mel JuSo cells [80], resulting in raised release of ILVs. These results support the involvement of viral protein in exosome biogenesis in different cell types, such as astrocytes [81], CD4 + T cells, and U937 cells [82]. Considering the discovery of the viral protein domain and amino acid sequence responsible for stimulation of vesicle release [83], it has been found that N, G, and F proteins of respiratory syncytial virus (RSV) are involved in the secretion of more exosomes and changes in exosome components during infection [84]. According to the research of Wu and his colleagues, it was found that infection with enterovirus A71 (EV-A71) increases the secretion of exosomes both in vitro and in vivo [85]. It also increases the production of exosomes from macrophages infected by HIV-1 [86]. In general, some viruses can increase the production of exosomes and use them for their benefit by increasing the production of exosomes.

Virus regulates exosome composition

Also, viral infection changes the exosomal proteome [87]. As mentioned, viral proteins have a substantial impact on changing the exosome’s composition. For instance, the incorporation of the Nef protein of human immunodeficiency virus (HIV) into the exosomes derived from HIV-infected cells [88], as well as Nef-transformed T cells [84, 85], stimulates the release of Nef from cells [89] and cell death of bystander CD4 + T lymphocytes. Of note, these T lymphocytes are essential and involved in the AIDS progression [89] and derive inflammation mediated by destroying cholesterol metabolism and elevating lipid rafts in bystander cells [90]. In addition, increased expression of ADP-ribosyl cyclase 1 (CD38) by exosomes released by HIV-infected T-lymphocytes occurs, which decreases annexin A5 (ANXA5) and L-lactate dehydrogenase B (LDHB) chain levels, which The above interact with Tat and P24, HIV viral proteins, and affect the cellular processes of proliferation and apoptosis [87]. In a research conducted in 2023, it was found that the exosomes of HBV-infected hepatocytes activate stellate cells, which can aggravate liver fibrosis [91].
Despite all the above information, the specific roles, and mechanisms of exosomal sorting of these proteins have not been fully discovered during viral infection and require more extensive research in this field.

Regulation of inflammation by respiratory viruses through exosomes

Respiratory syncytial viruses

In 1956, the respiratory syncytial virus (RSV) was discovered. Over the past years, researchers realized that it could cause lower respiratory tract disease at all ages and is the leading cause of death in children under five years of age, for which there is no vaccine. RSV is one of the RNA viruses that, according to statistics presented in 2019, causes 3.6 million hospital beds and causes the death of about 101,400 children aged 0–60 months [9295]. RSV encodes eleven separate proteins, including structural proteins such as membrane envelope glycoproteins (F and G), two non-structural proteins 1 and 2 (NS1 and NS2), and matrix proteins (M), which are considered crucial pathogenic agents to stimulate airway hyperreactivity (AHR), such as Th2-type overexpression, cytokines immune disorder, and inflammatory disequilibrium [9698]. RSV infection can affect the airway epithelium, polarized and differentiated cells release chemokines such as IL-8 and CCL5 and cause inflammatory cells to invade infected tissues [99]. Chemokines removed from the respiratory tract of RSV-infected children such as β-chemokines CCL2 (monocyte chemoattractant protein-1 [MCP-1]), CCL3 (macrophage inflammatory protein-1 [MIP-1α]), CCL11 (eotaxin), and CCL5 in the RSV-infected respiratory tract has been shown to not only predict disease severity, possibly by enhancing inflammation in response to RSV [100]. Reportedly, the RSV-infected cells could aggravate the release rates of exosomes carrying the RSV N protein, attachment G protein, fusion F protein, and a wide range of RNAs (e.g., mRNA, small noncoding-RNAs, ribosomal RNA segments, and RSV RNA) [101]. Chahar et al. reported that RSV-infected cells produced exosomes with a significant potential for enhanced release of the chemokines, IP-10, RANTES, and MCP-1, in monocytes compared to control cell exosomes. Similar to monocytes, RSV exosomes in comparison to mock exosomes enhanced the secretion of RANTES, IP-10, and TNF-α in the A549 cells, indicative of RSV exosomes ability in the generation of biological signals to alter innate immunity and inflammation during RSV infection [101]. Also, RSV can induce inflammation in different ways, and exosome is one of these ways; for, example, according to research on exosomes isolated from A549 cells infected with RSV in the laboratory, it was shown that the secretion of RANTES, Il-10, and TNF-α increases significantly and can induce inflammation [101]. Identifying these pathways can be very useful for treatment.

Influenza virus

Influenza viruses, the members of the family Orthomyxoviridae, are negative-sense RNA viruses classified into A, B, C, and D types. Amongst, only type A and B cause seasonal epidemics in people. Influenza A virus (IAV) is subtyped based on the two surface glycoproteins expression, neuraminidase (NA) and hemagglutinin (HA). Infection begins with the direct deposition of influenza particles into the epithelium or alveoli of the upper respiratory tract. The virus then binds to the cellular receptors via HA on its surface [102, 103]. According to the CDC, research on all common types of influenza between 2010 and 2022 showed that children under four years are more likely to be infected with almost all pathogenic influenza types than other age groups. Each year, severe influenza infection affects about 3–5 million people, of whom nearly 250,000–500,000 give in to the disease [104]. It has been reported that mice infected with influenza virus have various microRNAs (miRNAs), such as miR-483-3p, in their BALF exosomes. It has been hypothesized that miR-483p may boost the innate immunity in MLE-12 epithelial cells rather than other lung epithelial cells due to the miR-483-3p potential in enhanced activation of the transcription factors NF-kB and IRF3 in MLE-12 cells [105]. Based on this, miR-483-3p could provide body protection against the flu. Also, one of the critical receptors used by the influenza virus to bind to the target cell is sialic acid, related to a2,3 and a2,6, which are expressed by exosomes released in the airways. According to these ways, it competes with the sialylated cell surface receptor to which HA binds and is harmful to subsequent infections. In this way, it is clear that influenza virus infectivity can be neutralized by airway exosomes [106]. Also, BALF exosomes infected with the virus have been found to produce IL-6, MCP-1, and TNF and can induce inflammation [106]. To use exosomes as therapeutic or vaccine platforms, it is helpful to identify the different pathways by which exosomes impose their effects.

Parainfluenza virus

Human parainfluenza virus (HPIV), belonging to the family paramyxoviridae, is an enveloped, single-stranded, negative-sense RNA virus that can infect various host species. HPIV has four types [1, 107, 3] and two subtypes (4a and 4b) with different clinical and epidemiological characteristics [108]. HPIV is vital because there is no vaccine available for it to prevent the infection. Infections with parainfluenza viruses may result in various illnesses, such as conjunctivitis, pharyngitis, tracheobronchitis, otitis media, croup, and pneumonia. In children under five years, HPIV is the second leading reason for acute respiratory illness causing hospitalization after RSV [109]. HPIV infection increases the secretion of exosomes containing a massive load of viral proteins and RNA which can be transferred to other cells, causing productive infection and promoting viral replication. It has been shown that exosomes derived from HPIV-infected cells contain a different range of RNA species, including piRNA and miRNA. Autophagy inhibited by HPIV exosomes has also been demonstrated in other studies. The miR-126-3 p_2 was an essential regulatory factor in this process [110]. Probably, autophagy inhibition is one of the reasons for developing efficient exosome-mediated replication of HPIV infection. Also, miRNAs delivered to recipient cells through exosomes subsequently cause gene silencing, which has different effects depending on the cell type and stage [111]. Overall, the knowledge about inflammation caused by exosome and parainfluenza is minimal and needs further investigation.

Rhinoviruses

Rhinoviruses are linear single-stranded RNA viruses that belong to the picornaviridae family and belong to the enterovirus genus [112]. So far, more than 150 serotypes of rhinoviruses have been identified, which primarily cause mild and self-limiting diseases in healthy people, but cause significant complications in people with lung diseases [113, 114]. More than half of upper respiratory tract infections include human rhinoviruses, which are known as colds, and eventually recover after a week [115]. Rhinoviruses cause inflammation in the airways by activating of interleukins such as IL1 [116]. Also, through Exosomal miRNAs (ExoMiRNAs), which are non-coding RNAs with a length of 18 to 25 nucleotides, they can affect the inflammation process [117]. According to research, it has been determined that exosomes derived from rhinoviruses that contain MiR155 can induce inflammation because miR-155 Th2-mediated eosinophilic inflammation is essential in the lung [118]. Also, various studies show the induction of inflammation by exosomes derived from rhinoviruses, which have different mechanisms; for, example they can induce inflammation by producing cytokines and chemokines such as CXCL8 [119].

Coronaviruses

The coronaviruses family can cause common colds or acute and mild infections in the upper respiratory tract of both animals and humans. Coronavirus is a single-stranded (positive sense) RNA virus enclosed in an envelope with several protein molecules. The viral envelope consists of a lipid bilayer made of structural proteins: the membrane (M), coat (E), and spike (S). Coronaviruses are classified by the appearance of the corona or halo-like envelope glycoproteins, and chemical and replication characteristics. Coronaviruses enter the body through the respiratory system and attach to host cells using spike proteins. Then, they penetrate the cells and multiply and produce more viral particles. Upon internalization of the cells, an immune response is created to control the infection. However, an excessive immune response can lead to tissue damage and inflammation, especially in the respiratory system, called a cytokine storm [120, 121]. Coronaviruses can also affect other organs and methods of the body. The severity of the disease can range from mild respiratory signs to severe pneumonia and multi-organ failure [19]. The relationship between exosomes and coronavirus can be investigated in three ways: 1-entry of the virus, 2-immune modulation, and 3-detection potential [122124].
Exosomes potentially facilitate the coronavirus entry into the target cells [15]. Coronaviruses, including SARS-CoV-2, modulate the host immune response. During coronavirus infection, exosomes carrying viral components or immunoregulatory molecules are released, which affect immune cell function and inflammatory responses [125]. Probably, exosomes derived from virus-infected cells contain various viral details, including viral protein and RNA [125]. As a diagnostic tool, there are many biomarkers for detecting coronaviruses through exosomes. According to the Fujita et al. study, these biomarkers can be classified into three categories as follows; 1-antiviral response-related biomarkers, 2-coagulation-related markers, and 3- liver damage-related biomarkers [126]. Much research has been conducted on coronaviruses, especially after the spread and epidemic of SARS-CoV-2, which shows interesting results. It has been found that these types of exosomes have stimulating effects on inflammation. Therefore, exosomes can be used in different ways to treat and prevent respiratory diseases.
During the tests on exosomes derived from lung macrophages, it was found that Nsp12 and Nsp13 in exosomes derived from them can lead to the activation of nuclear factor κB (NF-κB) and subsequent induction of an array of inflammatory cytokines [127]. Also, in another study, it was found that pro-inflammatory cytokines can be produced through the interaction of the NF-kB inflammatory signaling pathway with Fibrinogen-β (FGB) and Tenascin-C (TNC) and plasma exosomes can cause the transfer of FGB, which ultimately causes pro-inflammatory cytokine signals are removed in organ cells [128].

Adenoviruses

Adenoviruses are a group of viruses that can cause various illnesses in humans. They can infect different tissues and are transmitted through close personal contact, respiratory droplets, or contaminated objects. Adenoviruses can provoke respiratory infections (common cold, sore throat, and pneumonia), gastrointestinal infections (diarrhea and vomiting), conjunctivitis (pink eye), and urinary tract infections [129, 130]. Most infected cases resolve independently, but severe conditions can occur in individuals with weakened immune systems. Prevention involves good hygiene practices and vaccination against certain adenovirus types. Consulting a healthcare professional is advised for diagnosis and treatment. The pathogenesis of adenovirus infection involves several steps, including entering the body through the respiratory or gastrointestinal tract, attaching to the specific receptors on host cells, releasing their DNA into the cell's nucleus followed by replication and transcription of viral genetic material, alongside synthesizing viral proteins [30]. From the infected cell, this is released to create new viral particles, these components assemble [131]. Adenoviruses can spread within the body and target various organs. The host immune system could be activated to eliminate the infection. Adenoviruses can evade immunity, resulting in prolonged or persistent disease, especially in individuals with weakened immune systems. The severity of adenovirus infections can vary, and different serotypes cause different clinical manifestations [132].
It has been revealed that adenoviruses use exosomes during their reproduction in the host cells. During infection, they can infect neighboring cells by release of exosomes from infected cells. These exosomes can contain viral components or factors that help the spread of the virus. Also, other studies have shown that adenoviruses can use exosomes to escape from the immune system [132].
In addition, exosomes released by uninfected cells can contain antiviral agents that have the potential to inhibit virus replication or stimulate immune responses against the virus [133]. In the research conducted in the laboratory on A549 cells infected with adenovirus type 3, it was found that the exosomes derived from these infected cells significantly increased IL-1β, which caused the presence of the condition to become inflammatory [134]. In contrast to other studies, other studies indicate that DC/FasL-derived exosomes can be used clinically to treat inflammatory and autoimmune diseases [135].
In summary, adenoviruses can manipulate the exosome pathway during infection and stimulate the release of virus-induced exosomes from infected cells [16]. These exosomes can help spread the virus, modulate the immune system, and promote disease progression. In addition, exosomes derived from uninfected cells can influence adenovirus infection by transferring antiviral agents [15]. Exosomes from adenovirus-infected cells have been shown to both induce and inhibit inflammation.

Conclusion and outlook

One of the things that plays an essential role in anti-virus immunity and virus promotion and is also an important vector for the transmission of viruses is exosome. The role of exosomes in the transmission of viruses and their role in inducing or inhibiting airway inflammation is critical, and as shown in Table 2 and Fig. 2, viruses that cause respiratory diseases can regulate inflammatory pathways through Exosomes that play a role in causing disease.
Table 2
Regulation of inflammatory pathways by viruses through exosomes
Virus
Exosome source
Exosome cargo
Sample (in vitro, in vivo)
Induction or inhibition of inflammation
Note
References
Respiratory syncytial viruses
RSV-infected A549 cells
In vitro (RSV-infected A549 cells)
Induction
Exosomes derived from RSV-infected A549 cells secrete significantly higher levels of RANTES, IP-10, and TNF-α, which activate the innate immune response and may through the release of pro-inflammatory cytokines and apoptosis in receptor cells have antiviral effects
[101]
Influenza
Respiratory epithelial cell lines
NP, NS1, M1, HA
In vitro (Respiratory epithelial cell lines)
Inhibition
Exosomes derived from respiratory epithelial cell lines infected with influenza virus showed an anti-viral response, which may inhibit inflammation in the early stages
[106]
Influenza
Bronchial alveolar lavage fluid (BALf) of influenza virus
In vitro (bronchial alveolar lavage fluid (BALf) of influenza virus)
Induction
Exosomes derived from bronchial alveolar lavage fluid (BALf) of influenza virus produce IL-6, MCP-1, and TNF, which are inflammatory cytokines and increase inflammation
[106]
Influenza
Macrophages, fibroblasts, T cell, and B cell lines
Nucleoprotein (NP) and non-structural protein (NS1)
In vitro (macrophages, fibroblasts, T cell, and B cell lines)
Induction
Levels of pro-inflammatory cytokines, such as IFN-γ, IL-1β, and CXCL8, were also elevated by the exosomes
[136]
Influenza
Bronchoalveolar lavage fluid (BALF)
miR-483-3p
In vitro (bronchoalveolar lavage fluid (BALF))
Induction
The transfer of miR-483-3p from bronchoalveolar lavage fluid (BALF) caused the expression of type I interferon and proinflammatory cytokine genes and strengthened their face
[137]
Influenza
Exosomes derived from H5N1-infected chickens
Viral proteins, NP and NS1
In vitro (exosomes derived from H5N1-infected chickens)
Induction
The presence of viral proteins such as NP and NS1 in exosomes derived from H5N1-infected chickens increases the level of pro-inflammatory cytokines such as IFN-γ, IL-1β, and IL-8
[119]
Influenza
Bronchoalveolar lavage fluid (BALF)
miR-483-3p, miR-374c-5p, miR-466i-5p, miR-203-3p
In vitro (bronchoalveolar lavage fluid (BALF))
Induction
miR-483-3p, miR-374c-5p, miR-466i-5p, miR-203-3p in exosomes derived from bronchoalveolar lavage fluid (BALF) significantly increased IFN-β expression, proinflammatory cytokine gene expression, and upregulates interferon-stimulated genes (ISGs), including IL6, CCL2, TNF-α, and SP110
[138]
Parainfluenza
Madin–Darby bovine kidney (MDBK) cells inoculated with CPIV3
miRNA 11
In vitro (Madin–Darby bovine kidney (MDBK) cells inoculated with CPIV3)
Unknown
These exosomes could transfer CPIV3 genetic materials to recipient cells to establish a productive infection and promote viral replication
[139]
Rhinoviruses
Primary bone marrow-derived DCs (BMDCs)
MiR155
in vitro and in vivo
Induction
miR-155 in exosomes derived from primary bone marrow-derived DCs (BMDCs) is essential for Th2-mediated eosinophilic inflammation in the lung and induces inflammatory conditions in the body
[118]
Rhinoviruses
RV-infected AECs
In vitro
Induction
Exosomes from RV-infected AECs yielded significant inflammatory cytokines/chemokines such as CXCL8, which induced an inflammatory state in the body
[140]
Rhinoviruses
Bronchoalveolar lavage fluid
In vitro
Induction
[141]
Coronaviruses
Lung macrophages
Nsp12 and Nsp13
In vitro
Induction
Nsp12 and Nsp13 in exosomes derived from lung macrophages lead to the activation of nuclear factor κB (NF-κB) and subsequent induction of an array of inflammatory cytokines
[127]
Coronaviruses
Patient plasma
Tenascin-C (TNC) and fibrinogen-β (FGB)
In vitro
Induction
Tenascin-C (TNC) and fibrinogen-β (FGB) induce the production of pro-inflammatory cytokines through interaction with the NF-κB inflammatory signaling pathway. FGB is transported via plasma exosomes and potentially induces pro-inflammatory cytokine signals in distant organ cells
[128]
Coronaviruses
A549 cell
Viral protein E, Nsp7, Nsp10, Nsp12, Nsp13, and slight protein M
In vitro
Induction
The RNA polymerase, Nsp12, alone inhibits tumor necrosis factor (TNF)-α and interleukin (IL)-6. Furthermore, a synergistic effect of Nsp12 working with Nsp13 was observed where, compared to Nsp12 alone, there was a significant induction of TNF-α, IL-1β, and IL-6, which are inflammatory cytokines. In contrast, M protein, Nsp13 alone, or Nsp10 did not
[127]
Coronaviruses
Endothelial cells
In vitro
Induction
Exosomes derived from endothelial cells infected with coronaviruses lead to NLRP3 inflammasome activation in endothelial cells of distant organs, which ultimately leads to IL-1β secretion and inflammatory response
[128]
Adenoviruses
Bone marrow-derived dendritic cells (DCs)
Expressing viral IL-10
In vitro
Inhibition
 
[142]
Adenoviruses
Derived from FasL-expressing DC
In vitro
Inhibition
Mice are immunized with a specific antigen, keyhole limpet hemocyanin (KLH). Then a Th1-mediated inflammatory response is induced 10 to 14 days after immunization by injection of the particular antigen into the hind pads. Mice immunized with KLH 106 DC or 1 μg exosome received in one hind paw 12 h before KLH booster injection into both hind paws. Delivery of DC/FasL or DC/FasL-derived exosomes significantly reduced paw swelling not only in the treated paw but also in the contralateral paw. Untreated suppressed 24, 48, and 72 h after antigen injection. These results show that genetically modified DC-expressing FasL as well as DC/FasL-derived exosomes are equally effective in suppressing the DTH response not only in the treated paw, but they are also in the untreated opposite paw
[135]
Adenovirus
Infected A549
In vitro
Induction
A549-derived exosomes infected with adenoviruses significantly increased IL-1β protein, which induced inflammatory conditions in the body
[134]

Acknowledgements

The authors thank the Department of Medical Virology, Tabriz University of Medical Sciences, for supporting this project.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors declare no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Ding L, Qi H, Wang Y, Zhang Z, Liu Q, Guo C, et al. Recent advances in ginsenosides against respiratory diseases: therapeutic targets and potential mechanisms. Biomed Pharmacother. 2023;158:114096.PubMedCrossRef Ding L, Qi H, Wang Y, Zhang Z, Liu Q, Guo C, et al. Recent advances in ginsenosides against respiratory diseases: therapeutic targets and potential mechanisms. Biomed Pharmacother. 2023;158:114096.PubMedCrossRef
3.
Zurück zum Zitat Chan-Zapata I, Borges-Argáez R, Ayora-Talavera G. Quinones as promising compounds against respiratory viruses: a review. Molecules. 2023;28(4):1981.PubMedPubMedCentralCrossRef Chan-Zapata I, Borges-Argáez R, Ayora-Talavera G. Quinones as promising compounds against respiratory viruses: a review. Molecules. 2023;28(4):1981.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Behl T, Rocchetti G, Chadha S, Zengin G, Bungau S, Kumar A, et al. Phytochemicals from plant foods as potential source of antiviral agents: an overview. Pharmaceuticals. 2021;14(4):381.PubMedPubMedCentralCrossRef Behl T, Rocchetti G, Chadha S, Zengin G, Bungau S, Kumar A, et al. Phytochemicals from plant foods as potential source of antiviral agents: an overview. Pharmaceuticals. 2021;14(4):381.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Schoborg RV, Borel N. Porcine epidemic diarrhea virus (PEDV) co-infection induced chlamydial persistence/stress does not require viral replication. Front Cell Infect Microbiol. 2014;4:20.PubMedPubMedCentralCrossRef Schoborg RV, Borel N. Porcine epidemic diarrhea virus (PEDV) co-infection induced chlamydial persistence/stress does not require viral replication. Front Cell Infect Microbiol. 2014;4:20.PubMedPubMedCentralCrossRef
7.
9.
10.
Zurück zum Zitat Chotpitayasunondh T, Fischer TK, Heraud JM, Hurt AC, Monto AS, Osterhaus A, et al. Influenza and COVID-19: what does co-existence mean? Influenza Other Respir Viruses. 2021;15(3):407–12.PubMedCrossRef Chotpitayasunondh T, Fischer TK, Heraud JM, Hurt AC, Monto AS, Osterhaus A, et al. Influenza and COVID-19: what does co-existence mean? Influenza Other Respir Viruses. 2021;15(3):407–12.PubMedCrossRef
11.
Zurück zum Zitat Cordisco M, Lucente MS, Sposato A, Cardone R, Pellegrini F, Franchini D, et al. Canine parainfluenza virus infection in a dog with acute respiratory disease. Vet Sci. 2022;9(7):346.PubMedPubMedCentral Cordisco M, Lucente MS, Sposato A, Cardone R, Pellegrini F, Franchini D, et al. Canine parainfluenza virus infection in a dog with acute respiratory disease. Vet Sci. 2022;9(7):346.PubMedPubMedCentral
12.
Zurück zum Zitat Ortega H, Nickle D, Carter L. Rhinovirus and asthma: challenges and opportunities. Rev Med Virol. 2021;31(4):e2193.PubMedCrossRef Ortega H, Nickle D, Carter L. Rhinovirus and asthma: challenges and opportunities. Rev Med Virol. 2021;31(4):e2193.PubMedCrossRef
13.
Zurück zum Zitat He Y, Zhang J, Feng J, Mei Z, Qian L, Huang Q, et al. Viral spectrum and clinical features in adult inpatients with community-acquired pneumonia and respiratory viral infection. Chin Gen Pract. 2021;24(26):3323. He Y, Zhang J, Feng J, Mei Z, Qian L, Huang Q, et al. Viral spectrum and clinical features in adult inpatients with community-acquired pneumonia and respiratory viral infection. Chin Gen Pract. 2021;24(26):3323.
14.
Zurück zum Zitat Hu B, Guo H, Zhou P, Shi Z-L. Characteristics of SARS-CoV-2 and COVID-19. Nat Rev Microbiol. 2021;19(3):141–54.PubMedCrossRef Hu B, Guo H, Zhou P, Shi Z-L. Characteristics of SARS-CoV-2 and COVID-19. Nat Rev Microbiol. 2021;19(3):141–54.PubMedCrossRef
15.
Zurück zum Zitat Chaudhari P, Ghate V, Nampoothiri M, Lewis S. Multifunctional role of exosomes in viral diseases: from transmission to diagnosis and therapy. Cell Signal. 2022;94:110325.PubMedPubMedCentralCrossRef Chaudhari P, Ghate V, Nampoothiri M, Lewis S. Multifunctional role of exosomes in viral diseases: from transmission to diagnosis and therapy. Cell Signal. 2022;94:110325.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Shi Y, Du L, Lv D, Li Y, Zhang Z, Huang X, et al. Emerging role and therapeutic application of exosome in hepatitis virus infection and associated diseases. J Gastroenterol. 2021;56:336–49.PubMedPubMedCentralCrossRef Shi Y, Du L, Lv D, Li Y, Zhang Z, Huang X, et al. Emerging role and therapeutic application of exosome in hepatitis virus infection and associated diseases. J Gastroenterol. 2021;56:336–49.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Fu Y, Zi R, Xiong S. Infection by exosome-carried Coxsackievirus B3 induces immune escape resulting in an aggravated pathogenesis. Microbes Infect. 2023:105148. Fu Y, Zi R, Xiong S. Infection by exosome-carried Coxsackievirus B3 induces immune escape resulting in an aggravated pathogenesis. Microbes Infect. 2023:105148.
20.
21.
Zurück zum Zitat Théry C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, 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(1):1535750.PubMedPubMedCentralCrossRef Théry C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, 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(1):1535750.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Jeppesen DK, Fenix AM, Franklin JL, Higginbotham JN, Zhang Q, Zimmerman LJ, et al. Reassessment of exosome composition. Cell. 2019;177(2):428–45.PubMedPubMedCentralCrossRef Jeppesen DK, Fenix AM, Franklin JL, Higginbotham JN, Zhang Q, Zimmerman LJ, et al. Reassessment of exosome composition. Cell. 2019;177(2):428–45.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Jiang Y, Wang F, Wang K, Zhong Y, Wei X, Wang Q, et al. Engineered exosomes: a promising drug delivery strategy for brain diseases. Curr Med Chem. 2022;29(17):3111–24.PubMedCrossRef Jiang Y, Wang F, Wang K, Zhong Y, Wei X, Wang Q, et al. Engineered exosomes: a promising drug delivery strategy for brain diseases. Curr Med Chem. 2022;29(17):3111–24.PubMedCrossRef
24.
Zurück zum Zitat Ding D, He X, Agarry IE, Wang Y, Zhou F, Li Y, et al. Profile of human milk phospholipids at different lactation stages with UPLC/Q-TOF-MS: characterization, distribution, and differences. J Agric Food Chem. 2023;71(16):6326–37.PubMedCrossRef Ding D, He X, Agarry IE, Wang Y, Zhou F, Li Y, et al. Profile of human milk phospholipids at different lactation stages with UPLC/Q-TOF-MS: characterization, distribution, and differences. J Agric Food Chem. 2023;71(16):6326–37.PubMedCrossRef
25.
Zurück zum Zitat Paskeh MDA, Entezari M, Mirzaei S, Zabolian A, Saleki H, Naghdi MJ, et al. Emerging role of exosomes in cancer progression and tumor microenvironment remodeling. J Hematol Oncol. 2022;15(1):1–39.CrossRef Paskeh MDA, Entezari M, Mirzaei S, Zabolian A, Saleki H, Naghdi MJ, et al. Emerging role of exosomes in cancer progression and tumor microenvironment remodeling. J Hematol Oncol. 2022;15(1):1–39.CrossRef
26.
Zurück zum Zitat Yang D, Zhang W, Zhang H, Zhang F, Chen L, Ma L, et al. Progress, opportunity, and perspective on exosome isolation-efforts for efficient exosome-based theranostics. Theranostics. 2020;10(8):3684.PubMedPubMedCentralCrossRef Yang D, Zhang W, Zhang H, Zhang F, Chen L, Ma L, et al. Progress, opportunity, and perspective on exosome isolation-efforts for efficient exosome-based theranostics. Theranostics. 2020;10(8):3684.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Sims B, Gu L, Krendelchtchikov A, Matthews QL. Neural stem cell-derived exosomes mediate viral entry. Int J Nanomed. 2014;9:4893.CrossRef Sims B, Gu L, Krendelchtchikov A, Matthews QL. Neural stem cell-derived exosomes mediate viral entry. Int J Nanomed. 2014;9:4893.CrossRef
28.
Zurück zum Zitat Admyre C, Telemo E, Almqvist N, Lötvall J, Lahesmaa R, Scheynius A, et al. Exosomes–nanovesicles with possible roles in allergic inflammation. Allergy. 2008;63(4):404–8.PubMedCrossRef Admyre C, Telemo E, Almqvist N, Lötvall J, Lahesmaa R, Scheynius A, et al. Exosomes–nanovesicles with possible roles in allergic inflammation. Allergy. 2008;63(4):404–8.PubMedCrossRef
29.
Zurück zum Zitat Than UTT, Guanzon D, Leavesley D, Parker T. Association of extracellular membrane vesicles with cutaneous wound healing. Int J Mol Sci. 2017;18(5):956.PubMedPubMedCentralCrossRef Than UTT, Guanzon D, Leavesley D, Parker T. Association of extracellular membrane vesicles with cutaneous wound healing. Int J Mol Sci. 2017;18(5):956.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Burkova EE, Sedykh SE, Nevinsky GA. Human placenta exosomes: biogenesis, isolation, composition, and prospects for use in diagnostics. Int J Mol Sci. 2021;22(4):2158.PubMedPubMedCentralCrossRef Burkova EE, Sedykh SE, Nevinsky GA. Human placenta exosomes: biogenesis, isolation, composition, and prospects for use in diagnostics. Int J Mol Sci. 2021;22(4):2158.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Tschuschke M, Kocherova I, Bryja A, Mozdziak P, Angelova Volponi A, Janowicz K, et al. Inclusion biogenesis, methods of isolation and clinical application of human cellular exosomes. J Clin Med. 2020;9(2):436.PubMedPubMedCentralCrossRef Tschuschke M, Kocherova I, Bryja A, Mozdziak P, Angelova Volponi A, Janowicz K, et al. Inclusion biogenesis, methods of isolation and clinical application of human cellular exosomes. J Clin Med. 2020;9(2):436.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Gurung S, Perocheau D, Touramanidou L, Baruteau J. The exosome journey: from biogenesis to uptake and intracellular signalling. Cell Commun Signal. 2021;19(1):1–19.CrossRef Gurung S, Perocheau D, Touramanidou L, Baruteau J. The exosome journey: from biogenesis to uptake and intracellular signalling. Cell Commun Signal. 2021;19(1):1–19.CrossRef
33.
Zurück zum Zitat Yue B, Yang H, Wang J, Ru W, Wu J, Huang Y, et al. Exosome biogenesis, secretion and function of exosomal miRNAs in skeletal muscle myogenesis. Cell Prolif. 2020;53(7):e12857.PubMedPubMedCentralCrossRef Yue B, Yang H, Wang J, Ru W, Wu J, Huang Y, et al. Exosome biogenesis, secretion and function of exosomal miRNAs in skeletal muscle myogenesis. Cell Prolif. 2020;53(7):e12857.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Jia G, Sowers JR. Targeting endothelial exosomes for the prevention of cardiovascular disease. Biochim et Biophys Acta (BBA)-Mol Basis Dis. 2020;1866(8):165833.CrossRef Jia G, Sowers JR. Targeting endothelial exosomes for the prevention of cardiovascular disease. Biochim et Biophys Acta (BBA)-Mol Basis Dis. 2020;1866(8):165833.CrossRef
35.
Zurück zum Zitat Anakor E, Le Gall L, Dumonceaux J, Duddy WJ, Duguez S. Exosomes in ageing and motor neurone disease: biogenesis, uptake mechanisms, modifications in disease and uses in the development of biomarkers and therapeutics. Cells. 2021;10(11):2930.PubMedPubMedCentralCrossRef Anakor E, Le Gall L, Dumonceaux J, Duddy WJ, Duguez S. Exosomes in ageing and motor neurone disease: biogenesis, uptake mechanisms, modifications in disease and uses in the development of biomarkers and therapeutics. Cells. 2021;10(11):2930.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Ghossoub R, Lembo F, Rubio A, Gaillard CB, Bouchet J, Vitale N, et al. Syntenin-ALIX exosome biogenesis and budding into multivesicular bodies are controlled by ARF6 and PLD2. Nat Commun. 2014;5(1):3477.PubMedCrossRef Ghossoub R, Lembo F, Rubio A, Gaillard CB, Bouchet J, Vitale N, et al. Syntenin-ALIX exosome biogenesis and budding into multivesicular bodies are controlled by ARF6 and PLD2. Nat Commun. 2014;5(1):3477.PubMedCrossRef
38.
Zurück zum Zitat Charrin S, Jouannet S, Boucheix C, Rubinstein E. Tetraspanins at a glance. J Cell Sci. 2014;127(17):3641–8.PubMed Charrin S, Jouannet S, Boucheix C, Rubinstein E. Tetraspanins at a glance. J Cell Sci. 2014;127(17):3641–8.PubMed
39.
Zurück zum Zitat Palmulli R, van Niel G. To be or not to be… secreted as exosomes, a balance finely tuned by the mechanisms of biogenesis. Essays Biochem. 2018;62(2):177–91.PubMedCrossRef Palmulli R, van Niel G. To be or not to be… secreted as exosomes, a balance finely tuned by the mechanisms of biogenesis. Essays Biochem. 2018;62(2):177–91.PubMedCrossRef
40.
Zurück zum Zitat Zhao R, Zhao T, He Z, Cai R, Pang W. Composition, isolation, identification and function of adipose tissue-derived exosomes. Adipocyte. 2021;10(1):587–604.PubMedPubMedCentralCrossRef Zhao R, Zhao T, He Z, Cai R, Pang W. Composition, isolation, identification and function of adipose tissue-derived exosomes. Adipocyte. 2021;10(1):587–604.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Lai JJ, Chau ZL, Chen SY, Hill JJ, Korpany KV, Liang NW, et al. Exosome processing and characterization approaches for research and technology development. Adv Sci. 2022;9(15):2103222.CrossRef Lai JJ, Chau ZL, Chen SY, Hill JJ, Korpany KV, Liang NW, et al. Exosome processing and characterization approaches for research and technology development. Adv Sci. 2022;9(15):2103222.CrossRef
42.
Zurück zum Zitat Huang J, Xiong J, Yang L, Zhang J, Sun S, Liang Y. Cell-free exosome-laden scaffolds for tissue repair. Nanoscale. 2021;13(19):8740–50.PubMedCrossRef Huang J, Xiong J, Yang L, Zhang J, Sun S, Liang Y. Cell-free exosome-laden scaffolds for tissue repair. Nanoscale. 2021;13(19):8740–50.PubMedCrossRef
43.
44.
Zurück zum Zitat Masyuk AI, Masyuk TV, LaRusso NF. Exosomes in the pathogenesis, diagnostics and therapeutics of liver diseases. J Hepatol. 2013;59(3):621–5.PubMedCrossRef Masyuk AI, Masyuk TV, LaRusso NF. Exosomes in the pathogenesis, diagnostics and therapeutics of liver diseases. J Hepatol. 2013;59(3):621–5.PubMedCrossRef
45.
Zurück zum Zitat Mao W, Wang K, Wu Z, Xu B, Chen M. Current status of research on exosomes in general, and for the diagnosis and treatment of kidney cancer in particular. J Exp Clin Cancer Res. 2021;40(1):1–13.CrossRef Mao W, Wang K, Wu Z, Xu B, Chen M. Current status of research on exosomes in general, and for the diagnosis and treatment of kidney cancer in particular. J Exp Clin Cancer Res. 2021;40(1):1–13.CrossRef
46.
Zurück zum Zitat Beatriz M, Vilaça R, Lopes C. Exosomes: innocent bystanders or critical culprits in neurodegenerative diseases. Front Cell Dev Biol. 2021;9:635104.PubMedPubMedCentralCrossRef Beatriz M, Vilaça R, Lopes C. Exosomes: innocent bystanders or critical culprits in neurodegenerative diseases. Front Cell Dev Biol. 2021;9:635104.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Jiao Y, Xu P, Shi H, Chen D, Shi H. Advances on liver cell-derived exosomes in liver diseases. J Cell Mol Med. 2021;25(1):15–26.PubMedCrossRef Jiao Y, Xu P, Shi H, Chen D, Shi H. Advances on liver cell-derived exosomes in liver diseases. J Cell Mol Med. 2021;25(1):15–26.PubMedCrossRef
48.
Zurück zum Zitat Efthymiou G, Saint A, Ruff M, Rekad Z, Ciais D, Van Obberghen-Schilling E. Shaping up the tumor microenvironment with cellular fibronectin. Front Oncol. 2020;10:641.PubMedPubMedCentralCrossRef Efthymiou G, Saint A, Ruff M, Rekad Z, Ciais D, Van Obberghen-Schilling E. Shaping up the tumor microenvironment with cellular fibronectin. Front Oncol. 2020;10:641.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Santiago-Sánchez GS, Pita-Grisanti V, Quiñones-Díaz B, Gumpper K, Cruz-Monserrate Z, Vivas-Mejía PE. Biological functions and therapeutic potential of Lipocalin 2 in cancer. Int J Mol Sci. 2020;21(12):4365.PubMedPubMedCentralCrossRef Santiago-Sánchez GS, Pita-Grisanti V, Quiñones-Díaz B, Gumpper K, Cruz-Monserrate Z, Vivas-Mejía PE. Biological functions and therapeutic potential of Lipocalin 2 in cancer. Int J Mol Sci. 2020;21(12):4365.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Chan BD, Wong WY, Lee MM, Cho WC, Yee BK, Kwan YW, et al. Exosomes in inflammation and inflammatory disease. Proteomics. 2019;19(8):e1800149.PubMedCrossRef Chan BD, Wong WY, Lee MM, Cho WC, Yee BK, Kwan YW, et al. Exosomes in inflammation and inflammatory disease. Proteomics. 2019;19(8):e1800149.PubMedCrossRef
52.
Zurück zum Zitat Chow A, Zhou W, Liu L, Fong M, Champer J, Van Haute D, et al. Macrophage immunomodulation by breast cancer-derived exosomes requires Toll-like receptor 2-mediated activation of NF-ĸB. Sci Rep. 2014;4:5750.PubMedPubMedCentralCrossRef Chow A, Zhou W, Liu L, Fong M, Champer J, Van Haute D, et al. Macrophage immunomodulation by breast cancer-derived exosomes requires Toll-like receptor 2-mediated activation of NF-ĸB. Sci Rep. 2014;4:5750.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Valenti R, Huber V, Filipazzi P, Pilla L, Sovena G, Villa A, et al. Human tumor-released microvesicles promote the differentiation of myeloid cells with transforming growth factor-β–mediated suppressive activity on T lymphocytes. Can Res. 2006;66(18):9290–8.CrossRef Valenti R, Huber V, Filipazzi P, Pilla L, Sovena G, Villa A, et al. Human tumor-released microvesicles promote the differentiation of myeloid cells with transforming growth factor-β–mediated suppressive activity on T lymphocytes. Can Res. 2006;66(18):9290–8.CrossRef
54.
Zurück zum Zitat Chalmin F, Ladoire S, Mignot G, Vincent J, Bruchard M, Remy-Martin J-P, et al. Membrane-associated Hsp72 from tumor-derived exosomes mediates STAT3-dependent immunosuppressive function of mouse and human myeloid-derived suppressor cells. J Clin Investig. 2010;120(2):457–71.PubMedPubMedCentral Chalmin F, Ladoire S, Mignot G, Vincent J, Bruchard M, Remy-Martin J-P, et al. Membrane-associated Hsp72 from tumor-derived exosomes mediates STAT3-dependent immunosuppressive function of mouse and human myeloid-derived suppressor cells. J Clin Investig. 2010;120(2):457–71.PubMedPubMedCentral
55.
Zurück zum Zitat Szczepanski MJ, Szajnik M, Welsh A, Whiteside TL, Boyiadzis M. Blast-derived microvesicles in sera from patients with acute myeloid leukemia suppress natural killer cell function via membrane-associated transforming growth factor-β1. Haematologica. 2011;96(9):1302–9.PubMedPubMedCentralCrossRef Szczepanski MJ, Szajnik M, Welsh A, Whiteside TL, Boyiadzis M. Blast-derived microvesicles in sera from patients with acute myeloid leukemia suppress natural killer cell function via membrane-associated transforming growth factor-β1. Haematologica. 2011;96(9):1302–9.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Szajnik M, Czystowska M, Szczepanski MJ, Mandapathil M, Whiteside TL. Tumor-derived microvesicles induce, expand and up-regulate biological activities of human regulatory T cells (Treg). PLoS ONE. 2010;5(7):e11469.PubMedPubMedCentralCrossRef Szajnik M, Czystowska M, Szczepanski MJ, Mandapathil M, Whiteside TL. Tumor-derived microvesicles induce, expand and up-regulate biological activities of human regulatory T cells (Treg). PLoS ONE. 2010;5(7):e11469.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Yoshimura A, Naka T, Kubo M. SOCS proteins, cytokine signalling and immune regulation. Nat Rev Immunol. 2007;7(6):454–65.PubMedCrossRef Yoshimura A, Naka T, Kubo M. SOCS proteins, cytokine signalling and immune regulation. Nat Rev Immunol. 2007;7(6):454–65.PubMedCrossRef
58.
Zurück zum Zitat Li D, Jia H, Zhang H, Lv M, Liu J, Zhang Y, et al. TLR4 signaling induces the release of microparticles by tumor cells that regulate inflammatory cytokine IL-6 of macrophages via microRNA let-7b. Oncoimmunology. 2012;1(5):687–93.PubMedPubMedCentralCrossRef Li D, Jia H, Zhang H, Lv M, Liu J, Zhang Y, et al. TLR4 signaling induces the release of microparticles by tumor cells that regulate inflammatory cytokine IL-6 of macrophages via microRNA let-7b. Oncoimmunology. 2012;1(5):687–93.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Calderón MA. Cardiopulmonary axis and cardiovascular mortality in patients with COPD. SEMERGEN. 2023;49(4):101928. Calderón MA. Cardiopulmonary axis and cardiovascular mortality in patients with COPD. SEMERGEN. 2023;49(4):101928.
60.
Zurück zum Zitat Kesimer M, Gupta R. Physical characterization and profiling of airway epithelial derived exosomes using light scattering. Methods (San Diego Calif). 2015;87:59–63.PubMedCrossRef Kesimer M, Gupta R. Physical characterization and profiling of airway epithelial derived exosomes using light scattering. Methods (San Diego Calif). 2015;87:59–63.PubMedCrossRef
61.
Zurück zum Zitat De SK. Novel 4-chloro-N-phenyl benzamide derivatives as p38α mitogenactivated protein kinase inhibitors for treating cancer, COVID-19, and other diseases. Recent Pat Anti-Cancer Drug Discov. 2023;18(4):549–51.CrossRef De SK. Novel 4-chloro-N-phenyl benzamide derivatives as p38α mitogenactivated protein kinase inhibitors for treating cancer, COVID-19, and other diseases. Recent Pat Anti-Cancer Drug Discov. 2023;18(4):549–51.CrossRef
62.
Zurück zum Zitat Nishioka M, Venkatesan N, Dessalle K, Mogas A, Kyoh S, Lin TY, et al. Fibroblast-epithelial cell interactions drive epithelial-mesenchymal transition differently in cells from normal and COPD patients. Respir Res. 2015;16(1):72.PubMedPubMedCentralCrossRef Nishioka M, Venkatesan N, Dessalle K, Mogas A, Kyoh S, Lin TY, et al. Fibroblast-epithelial cell interactions drive epithelial-mesenchymal transition differently in cells from normal and COPD patients. Respir Res. 2015;16(1):72.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Chang Y, Al-Alwan L, Alshakfa S, Audusseau S, Mogas AK, Chouiali F, et al. Upregulation of IL-17A/F from human lung tissue explants with cigarette smoke exposure: implications for COPD. Respir Res. 2014;15(1):145.PubMedPubMedCentralCrossRef Chang Y, Al-Alwan L, Alshakfa S, Audusseau S, Mogas AK, Chouiali F, et al. Upregulation of IL-17A/F from human lung tissue explants with cigarette smoke exposure: implications for COPD. Respir Res. 2014;15(1):145.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Liu Z, Yan J, Tong L, Liu S, Zhang Y. The role of exosomes from BALF in lung disease. J Cell Physiol. 2022;237(1):161–8.PubMedCrossRef Liu Z, Yan J, Tong L, Liu S, Zhang Y. The role of exosomes from BALF in lung disease. J Cell Physiol. 2022;237(1):161–8.PubMedCrossRef
65.
Zurück zum Zitat Lee H, Zhang D, Laskin DL, Jin Y. Functional evidence of pulmonary extracellular vesicles in infectious and noninfectious lung inflammation. J Immunol (Baltim Md: 1950). 1950;201(5):1500–9. Lee H, Zhang D, Laskin DL, Jin Y. Functional evidence of pulmonary extracellular vesicles in infectious and noninfectious lung inflammation. J Immunol (Baltim Md: 1950). 1950;201(5):1500–9.
66.
Zurück zum Zitat Gauvreau GM, El-Gammal AI, O’Byrne PM. Allergen-induced airway responses. Eur Respir J. 2015;46(3):819–31.PubMedCrossRef Gauvreau GM, El-Gammal AI, O’Byrne PM. Allergen-induced airway responses. Eur Respir J. 2015;46(3):819–31.PubMedCrossRef
67.
Zurück zum Zitat Esnault S, Kelly EA, Johnson SH, DeLain LP, Haedt MJ, Noll AL, et al. Matrix metalloproteinase-9-dependent release of IL-1β by human eosinophils. Mediators Inflamm. 2019;2019:7479107.PubMedPubMedCentralCrossRef Esnault S, Kelly EA, Johnson SH, DeLain LP, Haedt MJ, Noll AL, et al. Matrix metalloproteinase-9-dependent release of IL-1β by human eosinophils. Mediators Inflamm. 2019;2019:7479107.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Kapferer-Seebacher I, Pepin M, Werner R, Aitman TJ, Nordgren A, Stoiber H, et al. Periodontal Ehlers-Danlos syndrome is caused by mutations in C1R and C1S, which encode subcomponents C1r and C1s of complement. Am J Hum Genet. 2016;99(5):1005–14.PubMedPubMedCentralCrossRef Kapferer-Seebacher I, Pepin M, Werner R, Aitman TJ, Nordgren A, Stoiber H, et al. Periodontal Ehlers-Danlos syndrome is caused by mutations in C1R and C1S, which encode subcomponents C1r and C1s of complement. Am J Hum Genet. 2016;99(5):1005–14.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Frydrychowicz M, Kolecka-Bednarczyk A, Madejczyk M, Yasar S, Dworacki G. Exosomes—structure, biogenesis and biological role in non-small-cell lung cancer. Scand J Immunol. 2015;81(1):2–10.PubMedCrossRef Frydrychowicz M, Kolecka-Bednarczyk A, Madejczyk M, Yasar S, Dworacki G. Exosomes—structure, biogenesis and biological role in non-small-cell lung cancer. Scand J Immunol. 2015;81(1):2–10.PubMedCrossRef
70.
Zurück zum Zitat Wang B, Tan Z, Guan F. Tumor-derived exosomes mediate the instability of cadherins and promote tumor progression. Int J Mol Sci. 2019;20(15):3652.PubMedPubMedCentralCrossRef Wang B, Tan Z, Guan F. Tumor-derived exosomes mediate the instability of cadherins and promote tumor progression. Int J Mol Sci. 2019;20(15):3652.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Xu K, Zhang C, Du T, Gabriel ANA, Wang X, Li X, et al. Progress of exosomes in the diagnosis and treatment of lung cancer. Biomed Pharmacother Biomed Pharmacother. 2021;134:111111.PubMedCrossRef Xu K, Zhang C, Du T, Gabriel ANA, Wang X, Li X, et al. Progress of exosomes in the diagnosis and treatment of lung cancer. Biomed Pharmacother Biomed Pharmacother. 2021;134:111111.PubMedCrossRef
72.
73.
Zurück zum Zitat Saad MH, Badierah R, Redwan EM, El-Fakharany EM. A comprehensive insight into the role of exosomes in viral infection: dual faces bearing different functions. Pharmaceutics. 2021;13(9):1405.PubMedPubMedCentralCrossRef Saad MH, Badierah R, Redwan EM, El-Fakharany EM. A comprehensive insight into the role of exosomes in viral infection: dual faces bearing different functions. Pharmaceutics. 2021;13(9):1405.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Vlachakis D, Mitsis Τ, Nicolaides N, Efthimiadou A, Giannakakis A, Bacopoulou F, et al. Functions, pathophysiology and current insights of exosomal endocrinology. Mol Med Rep. 2021;23(1):1.CrossRef Vlachakis D, Mitsis Τ, Nicolaides N, Efthimiadou A, Giannakakis A, Bacopoulou F, et al. Functions, pathophysiology and current insights of exosomal endocrinology. Mol Med Rep. 2021;23(1):1.CrossRef
75.
Zurück zum Zitat Dyball LE, Smales CM. Exosomes: biogenesis, targeting, characterization and their potential as “Plug & Play” vaccine platforms. Biotechnol J. 2022;17(11):2100646.CrossRef Dyball LE, Smales CM. Exosomes: biogenesis, targeting, characterization and their potential as “Plug & Play” vaccine platforms. Biotechnol J. 2022;17(11):2100646.CrossRef
76.
Zurück zum Zitat Mosquera-Heredia MI, Morales LC, Vidal OM, Barceló E, Silvera-Redondo C, Vélez JI, et al. Exosomes: potential disease biomarkers and new therapeutic targets. Biomedicines. 2021;9(8):1061.PubMedPubMedCentralCrossRef Mosquera-Heredia MI, Morales LC, Vidal OM, Barceló E, Silvera-Redondo C, Vélez JI, et al. Exosomes: potential disease biomarkers and new therapeutic targets. Biomedicines. 2021;9(8):1061.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Gurunathan S, Kang M-H, Kim J-H. A comprehensive review on factors influences biogenesis, functions, therapeutic and clinical implications of exosomes. Int J Nanomed. 2021;16:1281.CrossRef Gurunathan S, Kang M-H, Kim J-H. A comprehensive review on factors influences biogenesis, functions, therapeutic and clinical implications of exosomes. Int J Nanomed. 2021;16:1281.CrossRef
79.
Zurück zum Zitat Sanfridson A, Hester S, Doyle C. Nef proteins encoded by human and simian immunodeficiency viruses induce the accumulation of endosomes and lysosomes in human T cells. Proc Natl Acad Sci USA. 1997;94(3):873–8.PubMedPubMedCentralCrossRef Sanfridson A, Hester S, Doyle C. Nef proteins encoded by human and simian immunodeficiency viruses induce the accumulation of endosomes and lysosomes in human T cells. Proc Natl Acad Sci USA. 1997;94(3):873–8.PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Stumptner-Cuvelette P, Jouve M, Helft J, Dugast M, Glouzman AS, Jooss K, et al. Human immunodeficiency virus-1 Nef expression induces intracellular accumulation of multivesicular bodies and major histocompatibility complex class II complexes: potential role of phosphatidylinositol 3-kinase. Mol Biol Cell. 2003;14(12):4857–70.PubMedPubMedCentralCrossRef Stumptner-Cuvelette P, Jouve M, Helft J, Dugast M, Glouzman AS, Jooss K, et al. Human immunodeficiency virus-1 Nef expression induces intracellular accumulation of multivesicular bodies and major histocompatibility complex class II complexes: potential role of phosphatidylinositol 3-kinase. Mol Biol Cell. 2003;14(12):4857–70.PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Pužar Dominkuš P, Ferdin J, Plemenitaš A, Peterlin BM, Lenassi M. Nef is secreted in exosomes from Nef.GFP-expressing and HIV-1-infected human astrocytes. J Neurovirol. 2017;23(5):713–24.PubMedCrossRef Pužar Dominkuš P, Ferdin J, Plemenitaš A, Peterlin BM, Lenassi M. Nef is secreted in exosomes from Nef.GFP-expressing and HIV-1-infected human astrocytes. J Neurovirol. 2017;23(5):713–24.PubMedCrossRef
82.
Zurück zum Zitat Aqil M, Naqvi AR, Mallik S, Bandyopadhyay S, Maulik U, Jameel S. The HIV Nef protein modulates cellular and exosomal miRNA profiles in human monocytic cells. J Extracell Vesicles. 2014;3:23129.CrossRef Aqil M, Naqvi AR, Mallik S, Bandyopadhyay S, Maulik U, Jameel S. The HIV Nef protein modulates cellular and exosomal miRNA profiles in human monocytic cells. J Extracell Vesicles. 2014;3:23129.CrossRef
83.
Zurück zum Zitat Ali SA, Huang MB, Campbell PE, Roth WW, Campbell T, Khan M, et al. Genetic characterization of HIV type 1 Nef-induced vesicle secretion. AIDS Res Hum Retroviruses. 2010;26(2):173–92.PubMedPubMedCentralCrossRef Ali SA, Huang MB, Campbell PE, Roth WW, Campbell T, Khan M, et al. Genetic characterization of HIV type 1 Nef-induced vesicle secretion. AIDS Res Hum Retroviruses. 2010;26(2):173–92.PubMedPubMedCentralCrossRef
84.
Zurück zum Zitat Chahar HS, Corsello T, Kudlicki AS, Komaravelli N, Casola A. Respiratory syncytial virus infection changes cargo composition of exosome released from airway epithelial cells. Sci Rep. 2018;8(1):387.PubMedPubMedCentralCrossRef Chahar HS, Corsello T, Kudlicki AS, Komaravelli N, Casola A. Respiratory syncytial virus infection changes cargo composition of exosome released from airway epithelial cells. Sci Rep. 2018;8(1):387.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Wu J, Zhao Y, Chen Q, Chen Y, Gu J, Mao L. Enterovirus A71 promotes exosome secretion by the nonstructural protein 3A interacting with Rab27a. Microbiol Spectrum. 2023;11(2):e03446-e3522.CrossRef Wu J, Zhao Y, Chen Q, Chen Y, Gu J, Mao L. Enterovirus A71 promotes exosome secretion by the nonstructural protein 3A interacting with Rab27a. Microbiol Spectrum. 2023;11(2):e03446-e3522.CrossRef
86.
Zurück zum Zitat Kadiu I, Narayanasamy P, Dash PK, Zhang W, Gendelman HE. Biochemical and biologic characterization of exosomes and microvesicles as facilitators of HIV-1 infection in macrophages. J Immunol. 2012;189(2):744–54.PubMedCrossRef Kadiu I, Narayanasamy P, Dash PK, Zhang W, Gendelman HE. Biochemical and biologic characterization of exosomes and microvesicles as facilitators of HIV-1 infection in macrophages. J Immunol. 2012;189(2):744–54.PubMedCrossRef
87.
Zurück zum Zitat Li M, Aliotta JM, Asara JM, Tucker L, Quesenberry P, Lally M, et al. Quantitative proteomic analysis of exosomes from HIV-1-infected lymphocytic cells. Proteomics. 2012;12(13):2203–11.PubMedCrossRef Li M, Aliotta JM, Asara JM, Tucker L, Quesenberry P, Lally M, et al. Quantitative proteomic analysis of exosomes from HIV-1-infected lymphocytic cells. Proteomics. 2012;12(13):2203–11.PubMedCrossRef
88.
Zurück zum Zitat Muratori C, Cavallin LE, Krätzel K, Tinari A, De Milito A, Fais S, et al. Massive secretion by T cells is caused by HIV Nef in infected cells and by Nef transfer to bystander cells. Cell Host Microbe. 2009;6(3):218–30.PubMedCrossRef Muratori C, Cavallin LE, Krätzel K, Tinari A, De Milito A, Fais S, et al. Massive secretion by T cells is caused by HIV Nef in infected cells and by Nef transfer to bystander cells. Cell Host Microbe. 2009;6(3):218–30.PubMedCrossRef
89.
Zurück zum Zitat Lenassi M, Cagney G, Liao M, Vaupotic T, Bartholomeeusen K, Cheng Y, et al. HIV Nef is secreted in exosomes and triggers apoptosis in bystander CD4+ T cells. Traffic. 2010;11(1):110–22.PubMedPubMedCentralCrossRef Lenassi M, Cagney G, Liao M, Vaupotic T, Bartholomeeusen K, Cheng Y, et al. HIV Nef is secreted in exosomes and triggers apoptosis in bystander CD4+ T cells. Traffic. 2010;11(1):110–22.PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat Mukhamedova N, Hoang A, Dragoljevic D, Dubrovsky L, Pushkarsky T, Low H, et al. Exosomes containing HIV protein Nef reorganize lipid rafts potentiating inflammatory response in bystander cells. PLoS Pathog. 2019;15(7):e1007907.PubMedPubMedCentralCrossRef Mukhamedova N, Hoang A, Dragoljevic D, Dubrovsky L, Pushkarsky T, Low H, et al. Exosomes containing HIV protein Nef reorganize lipid rafts potentiating inflammatory response in bystander cells. PLoS Pathog. 2019;15(7):e1007907.PubMedPubMedCentralCrossRef
91.
Zurück zum Zitat Yin M, Ding X, Yin S, Wang L, Zhang K, Chen Y, et al. Exosomes from hepatitis B virus‐infected hepatocytes activate hepatic stellate cells and aggravate liver fibrosis through the miR‐506‐3p/Nur77 pathway. J Biochem Mol Toxicol. 2023:e23432. Yin M, Ding X, Yin S, Wang L, Zhang K, Chen Y, et al. Exosomes from hepatitis B virus‐infected hepatocytes activate hepatic stellate cells and aggravate liver fibrosis through the miR‐506‐3p/Nur77 pathway. J Biochem Mol Toxicol. 2023:e23432.
92.
Zurück zum Zitat Cao D, Gooneratne I, Mera C, Vy J, Royal M, Huang B, et al. Analysis of template variations on RNA synthesis by respiratory syncytial virus polymerase. Viruses. 2023;15(1):47.CrossRef Cao D, Gooneratne I, Mera C, Vy J, Royal M, Huang B, et al. Analysis of template variations on RNA synthesis by respiratory syncytial virus polymerase. Viruses. 2023;15(1):47.CrossRef
93.
Zurück zum Zitat Wittenauer R, Pecenka C, Baral R. Cost of childhood RSV management and cost-effectiveness of RSV interventions: a systematic review from a low-and middle-income country perspective. BMC Med. 2023;21(1):121.PubMedPubMedCentralCrossRef Wittenauer R, Pecenka C, Baral R. Cost of childhood RSV management and cost-effectiveness of RSV interventions: a systematic review from a low-and middle-income country perspective. BMC Med. 2023;21(1):121.PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Falsey AR, Williams K, Gymnopoulou E, Bart S, Ervin J, Bastian AR, et al. Efficacy and safety of an Ad26. RSV. preF-RSV preF protein vaccine in older adults. N Engl J Med. 2023;388(7):609–20.PubMedCrossRef Falsey AR, Williams K, Gymnopoulou E, Bart S, Ervin J, Bastian AR, et al. Efficacy and safety of an Ad26. RSV. preF-RSV preF protein vaccine in older adults. N Engl J Med. 2023;388(7):609–20.PubMedCrossRef
95.
Zurück zum Zitat Graham BS. The journey to RSV vaccines—heralding an era of structure-based design. N Engl J Med. 2023;388(7):579–81.PubMedCrossRef Graham BS. The journey to RSV vaccines—heralding an era of structure-based design. N Engl J Med. 2023;388(7):579–81.PubMedCrossRef
96.
Zurück zum Zitat Caidi H, Miao C, Thornburg NJ, Tripp RA, Anderson LJ, Haynes LM. Anti-respiratory syncytial virus (RSV) G monoclonal antibodies reduce lung inflammation and viral lung titers when delivered therapeutically in a BALB/c mouse model. Antiviral Res. 2018;154:149–57.PubMedPubMedCentralCrossRef Caidi H, Miao C, Thornburg NJ, Tripp RA, Anderson LJ, Haynes LM. Anti-respiratory syncytial virus (RSV) G monoclonal antibodies reduce lung inflammation and viral lung titers when delivered therapeutically in a BALB/c mouse model. Antiviral Res. 2018;154:149–57.PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat Hu M, Bogoyevitch MA, Jans DA. Impact of respiratory syncytial virus infection on host functions: implications for antiviral strategies. Physiol Rev. 2020;100(4):1527–94.PubMedCrossRef Hu M, Bogoyevitch MA, Jans DA. Impact of respiratory syncytial virus infection on host functions: implications for antiviral strategies. Physiol Rev. 2020;100(4):1527–94.PubMedCrossRef
98.
Zurück zum Zitat Walker JA, McKenzie AN. TH2 cell development and function. Nat Rev Immunol. 2018;18(2):121–33.PubMedCrossRef Walker JA, McKenzie AN. TH2 cell development and function. Nat Rev Immunol. 2018;18(2):121–33.PubMedCrossRef
99.
Zurück zum Zitat Linssen RSN, Sridhar A, Moreni G, van der Wel NN, van Woensel JBM, Wolthers KC, et al. Neutrophil extracellular traps do not induce injury and inflammation in well-differentiated RSV-infected airway epithelium. Cells. 2022;11(5):785.PubMedPubMedCentralCrossRef Linssen RSN, Sridhar A, Moreni G, van der Wel NN, van Woensel JBM, Wolthers KC, et al. Neutrophil extracellular traps do not induce injury and inflammation in well-differentiated RSV-infected airway epithelium. Cells. 2022;11(5):785.PubMedPubMedCentralCrossRef
100.
Zurück zum Zitat Lin HY, Chang KT, Hung CC, Kuo CH, Hwang SJ, Chen HC, et al. Effects of the mTOR inhibitor rapamycin on monocyte-secreted chemokines. BMC Immunol. 2014;15:37.PubMedPubMedCentralCrossRef Lin HY, Chang KT, Hung CC, Kuo CH, Hwang SJ, Chen HC, et al. Effects of the mTOR inhibitor rapamycin on monocyte-secreted chemokines. BMC Immunol. 2014;15:37.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Chahar HS, Corsello T, Kudlicki AS, Komaravelli N, Casola A. Respiratory syncytial virus infection changes cargo composition of exosome released from airway epithelial cells. Sci Rep. 2018;8(1):1–18.CrossRef Chahar HS, Corsello T, Kudlicki AS, Komaravelli N, Casola A. Respiratory syncytial virus infection changes cargo composition of exosome released from airway epithelial cells. Sci Rep. 2018;8(1):1–18.CrossRef
102.
Zurück zum Zitat Londrigan SL, Short KR, Ma J, Gillespie L, Rockman SP, Brooks AG, Reading PC. Infection of mouse macrophages by seasonal influenza viruses can be restricted at the level of virus entry and at a late stage in the virus life cycle. ABSTRACT J Virol. 2015;89(24):12319–29. https://doi.org/10.1128/JVI.01455-15.CrossRef Londrigan SL, Short KR, Ma J, Gillespie L, Rockman SP, Brooks AG, Reading PC. Infection of mouse macrophages by seasonal influenza viruses can be restricted at the level of virus entry and at a late stage in the virus life cycle. ABSTRACT J Virol. 2015;89(24):12319–29. https://​doi.​org/​10.​1128/​JVI.​01455-15.CrossRef
103.
Zurück zum Zitat Laporte M, Stevaert A, Raeymaekers V, Boogaerts T, Nehlmeier I, Chiu W, Benkheil M, Vanaudenaerde B, Pöhlmann S, Naesens L. Hemagglutinin cleavability acid stability and temperature dependence optimize influenza B virus for replication in human airways. ABSTRACT J Virol. 2019;94(1):e01430-19. https://doi.org/10.1128/JVI.01430-19.CrossRef Laporte M, Stevaert A, Raeymaekers V, Boogaerts T, Nehlmeier I, Chiu W, Benkheil M, Vanaudenaerde B, Pöhlmann S, Naesens L. Hemagglutinin cleavability acid stability and temperature dependence optimize influenza B virus for replication in human airways. ABSTRACT J Virol. 2019;94(1):e01430-19. https://​doi.​org/​10.​1128/​JVI.​01430-19.CrossRef
104.
Zurück zum Zitat Gounder AP, Boon A. Influenza pathogenesis: the effect of host factors on severity of disease. J Immunol. 2019;202(2):341–50.PubMedCrossRef Gounder AP, Boon A. Influenza pathogenesis: the effect of host factors on severity of disease. J Immunol. 2019;202(2):341–50.PubMedCrossRef
105.
Zurück zum Zitat Maemura T, Fukuyama S, Kawaoka Y. High levels of miR-483-3p are present in serum exosomes upon infection of mice with highly pathogenic avian influenza virus. Front Microbiol. 2020;11:144.PubMedPubMedCentralCrossRef Maemura T, Fukuyama S, Kawaoka Y. High levels of miR-483-3p are present in serum exosomes upon infection of mice with highly pathogenic avian influenza virus. Front Microbiol. 2020;11:144.PubMedPubMedCentralCrossRef
106.
Zurück zum Zitat Bedford JG, Infusini G, Dagley LF, Villalon-Letelier F, Zheng MZ, Bennett-Wood V, et al. Airway exosomes released during influenza virus infection serve as a key component of the antiviral innate immune response. Front Immunol. 2020;11:887.PubMedPubMedCentralCrossRef Bedford JG, Infusini G, Dagley LF, Villalon-Letelier F, Zheng MZ, Bennett-Wood V, et al. Airway exosomes released during influenza virus infection serve as a key component of the antiviral innate immune response. Front Immunol. 2020;11:887.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Allegra A, Murdaca G, Gammeri L, Ettari R, Gangemi S. Alarmins and MicroRNAs, a new axis in the genesis of respiratory diseases: possible therapeutic implications. Int J Mol Sci. 2023;24(2):1783.PubMedPubMedCentralCrossRef Allegra A, Murdaca G, Gammeri L, Ettari R, Gangemi S. Alarmins and MicroRNAs, a new axis in the genesis of respiratory diseases: possible therapeutic implications. Int J Mol Sci. 2023;24(2):1783.PubMedPubMedCentralCrossRef
108.
Zurück zum Zitat Welch M, Park J, Harmon K, Zhang J, Piñeyro P, Giménez-Lirola L, et al. Pathogenesis of a novel porcine parainfluenza virus type 1 isolate in conventional and colostrum deprived/caesarean derived pigs. Virology. 2021;563:88–97.PubMedCrossRef Welch M, Park J, Harmon K, Zhang J, Piñeyro P, Giménez-Lirola L, et al. Pathogenesis of a novel porcine parainfluenza virus type 1 isolate in conventional and colostrum deprived/caesarean derived pigs. Virology. 2021;563:88–97.PubMedCrossRef
109.
Zurück zum Zitat Schomacker H, Schaap-Nutt A, Collins PL, Schmidt AC. Pathogenesis of acute respiratory illness caused by human parainfluenza viruses. Curr Opin Virol. 2012;2(3):294–9.PubMedPubMedCentralCrossRef Schomacker H, Schaap-Nutt A, Collins PL, Schmidt AC. Pathogenesis of acute respiratory illness caused by human parainfluenza viruses. Curr Opin Virol. 2012;2(3):294–9.PubMedPubMedCentralCrossRef
110.
Zurück zum Zitat Tang F, Yang T-L. MicroRNA-126 alleviates endothelial cells injury in atherosclerosis by restoring autophagic flux via inhibiting of PI3K/Akt/mTOR pathway. Biochem Biophys Res Commun. 2018;495(1):1482–9.PubMedCrossRef Tang F, Yang T-L. MicroRNA-126 alleviates endothelial cells injury in atherosclerosis by restoring autophagic flux via inhibiting of PI3K/Akt/mTOR pathway. Biochem Biophys Res Commun. 2018;495(1):1482–9.PubMedCrossRef
111.
Zurück zum Zitat Pegtel DM, Cosmopoulos K, Thorley-Lawson DA, van Eijndhoven MA, Hopmans ES, Lindenberg JL, et al. Functional delivery of viral miRNAs via exosomes. Proc Natl Acad Sci. 2010;107(14):6328–33.PubMedPubMedCentralCrossRef Pegtel DM, Cosmopoulos K, Thorley-Lawson DA, van Eijndhoven MA, Hopmans ES, Lindenberg JL, et al. Functional delivery of viral miRNAs via exosomes. Proc Natl Acad Sci. 2010;107(14):6328–33.PubMedPubMedCentralCrossRef
112.
Zurück zum Zitat Real-Hohn A, Groznica M, Kontaxis G, Zhu R, Chaves OA, Vazquez L, et al. Stabilization of the quadruplex-forming G-rich sequences in the rhinovirus genome inhibits uncoating— role of Na+ and K+. Viruses. 2023;15(4):1003.PubMedPubMedCentralCrossRef Real-Hohn A, Groznica M, Kontaxis G, Zhu R, Chaves OA, Vazquez L, et al. Stabilization of the quadruplex-forming G-rich sequences in the rhinovirus genome inhibits uncoating— role of Na+ and K+. Viruses. 2023;15(4):1003.PubMedPubMedCentralCrossRef
113.
Zurück zum Zitat Coultas JA, Cafferkey J, Mallia P, Johnston SL. Experimental antiviral therapeutic studies for human rhinovirus infections. J Exp Pharmacol. 2021:645–59. Coultas JA, Cafferkey J, Mallia P, Johnston SL. Experimental antiviral therapeutic studies for human rhinovirus infections. J Exp Pharmacol. 2021:645–59.
115.
Zurück zum Zitat Blaas D, Fuchs R. Mechanism of human rhinovirus infections. Mol Cell Pediatr. 2016;3(1):1–4.CrossRef Blaas D, Fuchs R. Mechanism of human rhinovirus infections. Mol Cell Pediatr. 2016;3(1):1–4.CrossRef
116.
Zurück zum Zitat Montgomery ST, Frey DL, Mall MA, Stick SM, Kicic A, Arest CF. Rhinovirus infection is associated with airway epithelial cell necrosis and inflammation via interleukin-1 in young children with cystic fibrosis. Front Immunol. 2020;11:596.PubMedPubMedCentralCrossRef Montgomery ST, Frey DL, Mall MA, Stick SM, Kicic A, Arest CF. Rhinovirus infection is associated with airway epithelial cell necrosis and inflammation via interleukin-1 in young children with cystic fibrosis. Front Immunol. 2020;11:596.PubMedPubMedCentralCrossRef
117.
118.
Zurück zum Zitat Gutierrez MJ, Gomez JL, Perez GF, Pancham K, Val S, Pillai DK, et al. Airway secretory microRNAome changes during rhinovirus infection in early childhood. PLoS ONE. 2016;11(9):e0162244.PubMedPubMedCentralCrossRef Gutierrez MJ, Gomez JL, Perez GF, Pancham K, Val S, Pillai DK, et al. Airway secretory microRNAome changes during rhinovirus infection in early childhood. PLoS ONE. 2016;11(9):e0162244.PubMedPubMedCentralCrossRef
119.
Zurück zum Zitat Jantaratrirat S, Boonarkart C, Ruangrung K, Suptawiwat O, Auewarakul P. Microparticle release from cell lines and its anti-influenza activity. Viral Immunol. 2018;31(6):447–56.PubMedCrossRef Jantaratrirat S, Boonarkart C, Ruangrung K, Suptawiwat O, Auewarakul P. Microparticle release from cell lines and its anti-influenza activity. Viral Immunol. 2018;31(6):447–56.PubMedCrossRef
122.
Zurück zum Zitat Lanyu Z, Feilong H. Emerging role of extracellular vesicles in lung injury and inflammation. Biomed Pharmacother. 2019;113:108748.PubMedCrossRef Lanyu Z, Feilong H. Emerging role of extracellular vesicles in lung injury and inflammation. Biomed Pharmacother. 2019;113:108748.PubMedCrossRef
123.
Zurück zum Zitat Martinez-Bravo M-J, Wahlund CJ, Qazi KR, Moulder R, Lukic A, Rådmark O, et al. Pulmonary sarcoidosis is associated with exosomal vitamin D-binding protein and inflammatory molecules. J Allergy Clin Immunol. 2017;139(4):1186–94.PubMedCrossRef Martinez-Bravo M-J, Wahlund CJ, Qazi KR, Moulder R, Lukic A, Rådmark O, et al. Pulmonary sarcoidosis is associated with exosomal vitamin D-binding protein and inflammatory molecules. J Allergy Clin Immunol. 2017;139(4):1186–94.PubMedCrossRef
124.
Zurück zum Zitat Askenase PW. COVID-19 therapy with mesenchymal stromal cells (MSC) and convalescent plasma must consider exosome involvement: do the exosomes in convalescent plasma antagonize the weak immune antibodies? J Extracell Vesicles. 2020;10(1):e12004.PubMedPubMedCentralCrossRef Askenase PW. COVID-19 therapy with mesenchymal stromal cells (MSC) and convalescent plasma must consider exosome involvement: do the exosomes in convalescent plasma antagonize the weak immune antibodies? J Extracell Vesicles. 2020;10(1):e12004.PubMedPubMedCentralCrossRef
125.
Zurück zum Zitat Pesce E, Manfrini N, Cordiglieri C, Santi S, Bandera A, Gobbini A, et al. Exosomes recovered from the plasma of COVID-19 patients expose SARS-CoV-2 spike-derived fragments and contribute to the adaptive immune response. Front Immunol. 2022;12:5775.CrossRef Pesce E, Manfrini N, Cordiglieri C, Santi S, Bandera A, Gobbini A, et al. Exosomes recovered from the plasma of COVID-19 patients expose SARS-CoV-2 spike-derived fragments and contribute to the adaptive immune response. Front Immunol. 2022;12:5775.CrossRef
126.
Zurück zum Zitat Rosell A, Havervall S, Von Meijenfeldt F, Hisada Y, Aguilera K, Grover SP, et al. Patients with COVID-19 have elevated levels of circulating extracellular vesicle tissue factor activity that is associated with severity and mortality—brief report. Arterioscler Thromb Vasc Biol. 2021;41(2):878–82.PubMedCrossRef Rosell A, Havervall S, Von Meijenfeldt F, Hisada Y, Aguilera K, Grover SP, et al. Patients with COVID-19 have elevated levels of circulating extracellular vesicle tissue factor activity that is associated with severity and mortality—brief report. Arterioscler Thromb Vasc Biol. 2021;41(2):878–82.PubMedCrossRef
127.
Zurück zum Zitat Teng Y, Xu F, Zhang X, Mu J, Sayed M, Hu X, et al. Plant-derived exosomal microRNAs inhibit lung inflammation induced by exosomes SARS-CoV-2 Nsp12. Mol Ther. 2021;29(8):2424–40.PubMedPubMedCentralCrossRef Teng Y, Xu F, Zhang X, Mu J, Sayed M, Hu X, et al. Plant-derived exosomal microRNAs inhibit lung inflammation induced by exosomes SARS-CoV-2 Nsp12. Mol Ther. 2021;29(8):2424–40.PubMedPubMedCentralCrossRef
128.
Zurück zum Zitat Sur S, Khatun M, Steele R, Isbell TS, Ray R, Ray RB. Exosomes from COVID-19 patients carry tenascin-C and fibrinogen-β in triggering inflammatory signals in cells of distant organ. Int J Mol Sci. 2021;22(6):3184.PubMedPubMedCentralCrossRef Sur S, Khatun M, Steele R, Isbell TS, Ray R, Ray RB. Exosomes from COVID-19 patients carry tenascin-C and fibrinogen-β in triggering inflammatory signals in cells of distant organ. Int J Mol Sci. 2021;22(6):3184.PubMedPubMedCentralCrossRef
130.
Zurück zum Zitat Stawicki SP, Jeanmonod R, Miller AC, et al. The 2019–2020 novel coronavirus (severe acute respiratory syndrome coronavirus 2) pandemic: A joint american college of academic international medicine-world academic council of emergency medicine multidisciplinary COVID-19 working group consensus paper. J Global Infect Dis. 2020;12(2):47. https://doi.org/10.4103/jgid.jgid_86_20.CrossRef Stawicki SP, Jeanmonod R, Miller AC, et al. The 2019–2020 novel coronavirus (severe acute respiratory syndrome coronavirus 2) pandemic: A joint american college of academic international medicine-world academic council of emergency medicine multidisciplinary COVID-19 working group consensus paper. J Global Infect Dis. 2020;12(2):47. https://​doi.​org/​10.​4103/​jgid.​jgid_​86_​20.CrossRef
131.
Zurück zum Zitat Zeng X, Carlin CR. Host cell autophagy modulates early stages of adenovirus infections in airway epithelial cells. J Virol. 2013;87(4):2307–19.PubMedPubMedCentralCrossRef Zeng X, Carlin CR. Host cell autophagy modulates early stages of adenovirus infections in airway epithelial cells. J Virol. 2013;87(4):2307–19.PubMedPubMedCentralCrossRef
132.
Zurück zum Zitat Ipinmoroti AO, Matthews QL. Extracellular vesicles: roles in human viral infections, immune-diagnostic, and therapeutic applications. Pathogens. 2020;9(12):1056.PubMedPubMedCentralCrossRef Ipinmoroti AO, Matthews QL. Extracellular vesicles: roles in human viral infections, immune-diagnostic, and therapeutic applications. Pathogens. 2020;9(12):1056.PubMedPubMedCentralCrossRef
134.
Zurück zum Zitat Ipinmoroti AO, Crenshaw BJ, Pandit R, Kumar S, Sims B, Matthews QL. Human adenovirus serotype 3 infection modulates the biogenesis and composition of lung cell-derived extracellular vesicles. J Immunol Res. 2021;2021. Ipinmoroti AO, Crenshaw BJ, Pandit R, Kumar S, Sims B, Matthews QL. Human adenovirus serotype 3 infection modulates the biogenesis and composition of lung cell-derived extracellular vesicles. J Immunol Res. 2021;2021.
135.
Zurück zum Zitat Kim SH, Bianco N, Menon R, Lechman ER, Shufesky WJ, Morelli AE, et al. Exosomes derived from genetically modified DC expressing FasL are anti-inflammatory and immunosuppressive. Mol Ther. 2006;13(2):289–300.PubMedCrossRef Kim SH, Bianco N, Menon R, Lechman ER, Shufesky WJ, Morelli AE, et al. Exosomes derived from genetically modified DC expressing FasL are anti-inflammatory and immunosuppressive. Mol Ther. 2006;13(2):289–300.PubMedCrossRef
136.
Zurück zum Zitat Hong Y, Truong AD, Vu TH, Lee S, Heo J, Kang S, et al. Exosomes from H5N1 avian influenza virus-infected chickens regulate antiviral immune responses of chicken immune cells. Dev Comp Immunol. 2022;130:104368.PubMedCrossRef Hong Y, Truong AD, Vu TH, Lee S, Heo J, Kang S, et al. Exosomes from H5N1 avian influenza virus-infected chickens regulate antiviral immune responses of chicken immune cells. Dev Comp Immunol. 2022;130:104368.PubMedCrossRef
137.
Zurück zum Zitat Maemura T, Fukuyama S, Sugita Y, Lopes TJS, Nakao T, Noda T, et al. Lung-derived exosomal miR-483-3p regulates the innate immune response to influenza virus infection. J Infect Dis. 2018;217(9):1372–82.PubMedCrossRef Maemura T, Fukuyama S, Sugita Y, Lopes TJS, Nakao T, Noda T, et al. Lung-derived exosomal miR-483-3p regulates the innate immune response to influenza virus infection. J Infect Dis. 2018;217(9):1372–82.PubMedCrossRef
138.
Zurück zum Zitat Zabrodskaya Y, Plotnikova M, Gavrilova N, Lozhkov A, Klotchenko S, Kiselev A, et al. Exosomes released by influenza-virus-infected cells carry factors capable of suppressing immune defense genes in Naïve cells. Viruses. 2022;14(12):2690.PubMedPubMedCentralCrossRef Zabrodskaya Y, Plotnikova M, Gavrilova N, Lozhkov A, Klotchenko S, Kiselev A, et al. Exosomes released by influenza-virus-infected cells carry factors capable of suppressing immune defense genes in Naïve cells. Viruses. 2022;14(12):2690.PubMedPubMedCentralCrossRef
139.
Zurück zum Zitat Mao L, Liang P, Li W, Zhang S, Liu M, Yang L, et al. Exosomes promote caprine parainfluenza virus type 3 infection by inhibiting autophagy. J Gen Virol. 2020;101(7):717–34.PubMedCrossRef Mao L, Liang P, Li W, Zhang S, Liu M, Yang L, et al. Exosomes promote caprine parainfluenza virus type 3 infection by inhibiting autophagy. J Gen Virol. 2020;101(7):717–34.PubMedCrossRef
140.
Zurück zum Zitat Mills J. Investigating the role of tenascin-C and extracellular vesicles in human rhinovirus induced exacerbations of asthma. Sheffield: University of Sheffield; 2018. Mills J. Investigating the role of tenascin-C and extracellular vesicles in human rhinovirus induced exacerbations of asthma. Sheffield: University of Sheffield; 2018.
141.
Zurück zum Zitat Mills JT, Schwenzer A, Marsh EK, Edwards MR, Sabroe I, Midwood KS, et al. Airway epithelial cells generate pro-inflammatory tenascin-C and small extracellular vesicles in response to TLR3 stimuli and rhinovirus infection. Front Immunol. 2019;10:1987.PubMedPubMedCentralCrossRef Mills JT, Schwenzer A, Marsh EK, Edwards MR, Sabroe I, Midwood KS, et al. Airway epithelial cells generate pro-inflammatory tenascin-C and small extracellular vesicles in response to TLR3 stimuli and rhinovirus infection. Front Immunol. 2019;10:1987.PubMedPubMedCentralCrossRef
142.
Zurück zum Zitat Kim S-H, Lechman ER, Bianco N, Menon R, Keravala A, Nash J, et al. Exosomes derived from IL-10-treated dendritic cells can suppress inflammation and collagen-induced arthritis 1. J Immunol. 2005;174(10):6440–8.PubMedCrossRef Kim S-H, Lechman ER, Bianco N, Menon R, Keravala A, Nash J, et al. Exosomes derived from IL-10-treated dendritic cells can suppress inflammation and collagen-induced arthritis 1. J Immunol. 2005;174(10):6440–8.PubMedCrossRef
Metadaten
Titel
Exosome-mediated regulation of inflammatory pathway during respiratory viral disease
verfasst von
Hamidreza Gheitasi
Mohammad Sabbaghian
Ali Akbar Shekarchi
Amir Ali Mirmazhary
Vahdat Poortahmasebi
Publikationsdatum
01.12.2024
Verlag
BioMed Central
Erschienen in
Virology Journal / Ausgabe 1/2024
Elektronische ISSN: 1743-422X
DOI
https://doi.org/10.1186/s12985-024-02297-y

Weitere Artikel der Ausgabe 1/2024

Virology Journal 1/2024 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Eingreifen von Umstehenden rettet vor Erstickungstod!

15.05.2024 Fremdkörperaspiration Nachrichten

Wer sich an einem Essensrest verschluckt und um Luft ringt, benötigt vor allem rasche Hilfe. Dass Umstehende nur in jedem zweiten Erstickungsnotfall bereit waren, diese zu leisten, ist das ernüchternde Ergebnis einer Beobachtungsstudie aus Japan. Doch es gibt auch eine gute Nachricht.

Neue S3-Leitlinie zur unkomplizierten Zystitis: Auf Antibiotika verzichten?

15.05.2024 Harnwegsinfektionen Nachrichten

Welche Antibiotika darf man bei unkomplizierter Zystitis verwenden und wovon sollte man die Finger lassen? Welche pflanzlichen Präparate können helfen? Was taugt der zugelassene Impfstoff? Antworten vom Koordinator der frisch überarbeiteten S3-Leitlinie, Prof. Florian Wagenlehner.

Schadet Ärger den Gefäßen?

14.05.2024 Arteriosklerose Nachrichten

In einer Studie aus New York wirkte sich Ärger kurzfristig deutlich negativ auf die Endothelfunktion gesunder Probanden aus. Möglicherweise hat dies Einfluss auf die kardiovaskuläre Gesundheit.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.