Next Article in Journal
The Effect of Epimedium Isopentenyl Flavonoids on the Broiler Gut Health Using Microbiomic and Metabolomic Analyses
Next Article in Special Issue
Intrathecal Injection of Autologous Mesenchymal Stem-Cell-Derived Extracellular Vesicles in Spinal Cord Injury: A Feasibility Study in Pigs
Previous Article in Journal
The Wheel of p53 Helps to Drive the Immune System
Previous Article in Special Issue
HIV Promotes Atherosclerosis via Circulating Extracellular Vesicle MicroRNAs
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Leveraging Exosomes as the Next-Generation Bio-Shuttles: The Next Biggest Approach against Th17 Cell Catastrophe

1
Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
2
Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(8), 7647; https://doi.org/10.3390/ijms24087647
Submission received: 23 March 2023 / Revised: 12 April 2023 / Accepted: 18 April 2023 / Published: 21 April 2023
(This article belongs to the Special Issue Extracellular Vesicles: The Biology and Therapeutic Applications)

Abstract

:
In recent years, the launch of clinical-grade exosomes is rising expeditiously, as they represent a new powerful approach for the delivery of advanced therapies and for diagnostic purposes for various diseases. Exosomes are membrane-bound extracellular vesicles that can act as biological messengers between cells, in the context of health and disease. In comparison to several lab-based drug carriers, exosome exhibits high stability, accommodates diverse cargo loads, elicits low immunogenicity and toxicity, and therefore manifests tremendous perspectives in the development of therapeutics. The efforts made to spur exosomes in drugging the untreatable targets are encouraging. Currently, T helper (Th) 17 cells are considered the most prominent factor in the establishment of autoimmunity and several genetic disorders. Current reports have indicated the importance of targeting the development of Th17 cells and the secretion of its paracrine molecule, interleukin (IL)-17. However, the present-day targeted approaches exhibit drawbacks, such as high cost of production, rapid transformation, poor bioavailability, and importantly, causing opportunistic infections that ultimately hamper their clinical applications. To overcome this hurdle, the potential use of exosomes as vectors seem to be a promising approach for Th17 cell-targeted therapies. With this standpoint, this review discusses this new concept by providing a snapshot of exosome biogenesis, summarizes the current clinical trials of exosomes in several diseases, analyzes the prospect of exosomes as an established drug carrier and delineates the present challenges, with an emphasis on their practical applications in targeting Th17 cells in diseases. We further decode the possible future scope of exosome bioengineering for targeted drug delivery against Th17 cells and its catastrophe.

1. Introduction

The advancement of novel computational tools and approaches, such as computer-aided drug design, high-throughput screening and artificial intelligence, helps expedite drug discovery and identify an active pharmaceutical ingredient (API) as new lead compounds in several disease settings [1]. Howbeit, several drugs escape from reaching the target sites due to an individual’s biological barrier and immune defense mechanisms during the process of disease treatment. Moreover, unwanted toxicities due to their accumulation in off-target tissues have been a major bottleneck in their translation into clinics [2,3]. Thereupon, strategies that enhance tissue-specific drug delivery hold promise to overcome these limitations and demonstrate tremendous success in parallel with drug discovery.
Exosomes are exemplified as “membrane-anchored extracellular vesicles” derived from the endosomal compartment of almost all eukaryotic cells [4]. As a nanoscale-sized vesicle, exosomes are found in all biological fluids (urine, blood, saliva, semen, breast milk and cerebrospinal fluid), which fabricate them as an excellent biomarker in the diagnosis of cancer and related disorders [5]. At the forefront, the exosome adapts a specific mechanism that includes surface receptor-mediated endocytosis, pinocytosis and membrane fusion to transport cargo to recipient cells [6]. As exosomes are imprinted with the ability to facilitate cell–cell communication, it is an unbiased claim that they represent potent drug delivery vectors in clinics due to their characteristic properties of innate stability, low immunogenicity, negative zeta potential to escape the immune attack and good capacity to penetrate through biological barriers [7].
Exosomes are extensively explored as tools in the landscape of immune regulation and immunotherapies, harnessing their role in immune surveillance, antigen presentation, immunosuppression, and anti-tumor immunity [8]. Proteomic studies have demonstrated that exosomes are tagged with immune function-related proteins, such as human leukocyte antigen (HLA)-1, β2-microglobulin and sub-units of the T cell receptor (TCR)-CD3 complex, thus defining their essential involvement in immune regulation [9]. For instance, cancer cell-derived exosomes activate vascular endothelial growth factor (VEGF) signaling in endothelial cells and advance angiogenesis in the tumor microenvironment [10]. In addition, exosomes exhibit immunosuppressive function via inhibition of CD8+ T cells, programmed death ligand 1 (PD-L1) secretion and expansion of suppressive regulatory T cells [11,12]. Notwithstanding, exosomes in the synovial fluid encapsulate citrullinated proteins that promote the formation of auto-immunogenic determinants [13]. However, dendritic cells (DCs)-derived exosomes express major histocompatibility complex class (MHC)-I molecules, which contribute to antigen presentation and anti-tumor responses [14], making them a hopeful prospect from the therapeutic standpoint of exosomes. Thereupon, the engineering of exosomes to deliver peptides, vaccines or drugs into specific cells in a time-dependent manner can be a star therapeutic strategy to negotiate immune responses with regard to immune-related diseases.
In various diseases such as cancer, autoimmune diseases and most genetic disorders, there exists an intricate imbalance between the occurrence of inflammatory events and the suppression of inflammation, which instigates disease progression [15]. Of note, immune cells are important modulators that facilitate this imbalance. In this context, an increased frequency of Th17 cells, a subset of CD4+ T immune cells, has been studied to promote the initiation and development of several tumors and autoimmune/inflammatory disorders via distinct mechanisms [16,17]. Thus far, targeting Th17 cells is an unmet clinical need. At present, exosome-based immune therapy is a jackpot as an effective strategy to restore immune balance and halt disease pathogenesis. In cancer, for example, curcumin export by exosomes facilitates the differentiation of effector T cells to kill cancer cells [18]. Under this notion, there are a few discussions on how exosomes communicate and can target Th17 CD4+ T cells to impede the development of life-threatening diseases.
Will exosomes be a solid answer to this ongoing search for a promising Th17-targeted drug delivery platform in clinics? With this explorative question in mind, this review will present an overview of the prospects of exosomes as a delivery platform for Th17-targeted therapies. As numerous reviews are published on exosomes, our initial discussion will be generalized to contextualize the growing importance of exosomes as “postmasters” for drug delivery. However, this review will be the first to provide a layout of exosomal payloads that repress Th17 cell function. We will update our readers on the clinical trials of exosomes, with an emphasis on the Th17 paradigm and current practical challenges faced in the race. Addressing these challenges will provide a roadmap to figure out exosome engineering strategies and categorize them based on their benefit scores to best exploit them for Th17 cell-targeted therapy. Finally, we will highlight the prospects of incorporating different small molecules, peptides, vaccines, antibodies and genetic moieties as exogenous payloads into exosomes to maximize the desired therapeutic applications against Th17 cell-mediated diseases. This review is contemplated to set a significant benchmark for designing the exosomes for successful drug encapsulation and render them as a facile and promising approach in the field of Th17 cell-targeted therapy in clinics.

2. The Structure of Exosomes and Its Composition

Exosomes are defined as lipid-bilayered extracellular vesicles with a diameter ranging from 30 to 200 nm. Secreted naturally from almost all kinds of cells, these vesicles are formed through endosomal membrane budding and constitute the nucleic acids, proteins, carbohydrates and lipids of a cell [19]. Moreover, these nanosized biological vectors shuttle cell-derived nucleic acids, lipids and proteins as signaling molecules to facilitate signal transduction and communication between neighboring cells [19]. Previous researchers have demonstrated that exosomes are closely associated with viral responses, immune regulation, cardiovascular risks, central nervous system (CNS) disorders, pregnancy, cancer and autoimmune disease progression [4]. Numerous efforts are in progress to isolate circulating exosomes that import biological substances and dynamically expand them as biomarkers as they can manifest several pathophysiological conditions [20]. Emphatically, exosomes have been explored as drug delivery vectors due to their ability to inherit characteristic properties of a cell and exhibit a better safety profile as compared to cell-based therapy [21]. Instead, molecular insights to understand the structures of exosomes could be helpful to engineer exosomes for loading cargoes to ferry them into target cells/tissues.
The contents inside and on the surface of the exosomes mirror the configuration of the parent cell. However, irrespective of their parental origin, some features including tetraspanins (CD9, CD81, CD82 and CD63), syndecans, heat shock proteins (Hsp60, Hsp20, Hsp70, Hsp22, Hsp90 and alpha-B Crystallin), biogenesis-associated proteins (ESCRT proteins, CHMP4, TSG101, STAM1, VPS4, PLD2), membrane transport and fusion proteins (annexins, GTPases, and Rab molecules), nuclear acids (long non-coding RNAs, miRNA, mRNA and DNA) and lipids, commonly span the membrane of exosomes structure [22]. Tetraspanins are abundant on the exosomal membranes to form a complex with integrins and contribute to the tropism of exosomes [23]. Advanced proteomic studies revealed that heat shock, transport and fusion proteins mediate the sprouting of multicellular vesicular bodies (MVBs) [24], whereas the lipidomic data demonstrated that the lipid contents of exosomes are either conserved or relatively similar and are crucially involved in preserving the shape of exosomes and maintaining homeostasis in the recipient cells [25]. Moreover, the asymmetrical distribution of the lipid bilayer of exosomes allows for successful drug delivery. For example, exosomes that display α6β4 integrins are likely to target S100-A4 positive fibroblasts and surfactant protein C (SPC)-positive epithelial cells of the lungs [26]. Similarly, lymphocyte function-associated antigen 1 (LFA-1)-tagged exosomes act as a “molecular Trojan horse” that can cross the blood–brain barrier to treat central nervous system (CNS)-related disorders [27], and αvβ6 exosomes play a critical role in interstitial inflammation and fibrosis [28].
The membrane of the exosomes is composed on the basis of their origin and physiological environment during exosome biogenesis [4]. Accordingly, the target site depends on the differential expression of markers on the surface of exosomes [29]. As a proof of concept, exosomes derived from dendritic cells express MHC class I, MHC class II, ICAM, CD40 and CD86 [30]. Also, chimeric antigen receptor T cell immunotherapy (CAR-T) cell-based exosomes express CARs and contain cytotoxic molecules used for cancer therapy [31], and those released by natural killer (NK) cells express lymphocyte function-associated antigen 1 (LFA-1) and DNAX accessory molecule 1 (DNAM-1) [32], thus, mirroring the characteristic features of parent cells. Exosomes generated from γδ T cells carry death-inducing ligands (FasL and TRAIL) and CD80/86 and MHC class I/II with dual tumor-killing effects [33]. Information on the membrane structures of exosomes that can guide their destination is available in different web-based sources, including the International Society for Extracellular Vesicles, American Society for Exosomes and Microvesicles, Extracellular RNA Communication Program, ExoCarta, Vesiclepedia and EVpedia.

3. Mechanistic Insights into Exosome Biogenesis and Release

The biogenesis of exosomes begins with the formation of intraluminal vesicles (ILVs) that develop via inward budding of the plasma membrane (early endosomes), which then generate late endosomes composed of combined intraluminal vesicles (ILVs), in a clathrin- or caveolin-dependent or -independent manner, called multivesicular bodies (MVBs) [19]. Based on the pathway they follow, they are categorized as degradative multivesicular exosomes (MVE) which coalesce with the lysosomes to ensure degradation of intraluminal contents and secretory MVE, which pinches out of the membrane to discharge its contents into the extracellular space. Moreover, MVE also leads to the formation of endosomal-related organelle, melanosomes and Weibel–Palade bodies in pigment and endothelial cells, respectively.
The biogenesis and protein sorting of ILVs are tightly regulated processes requiring an endosomal sorting complex (ESCRT) or can be an independent process [19]. In the context of the ESCRT-dependent pathway, it is composed of ESCRT-0, ESCRT-I, ESCRT-II and ESCRT-III complexes and AAA ATPase VPS4, tumor susceptibility gene (Tsg101) and ALIX auxiliary proteins [34]. EXCRT-0 escorts mono-ubiquitinated proteins into the endosomal domain with the help of cytosolic protein hepatocyte responsive serum phosphoprotein (HRS) heterodimer, signal transducing adapter molecule (STAM)1/2, Eps15 and clathrin. In the next sequential step, ESCRT-I and ESCRT-II together with ESCRT-0 bind to ubiquitinated cargo with higher affinity to form a stable membrane neck. Finally, ESCRT-III assembles the complex for the excision of the membrane neck and internalization of buds into the endosomes. Eventually, the cargoes are de-ubiquitinated by de-ubiquitylating enzymes and, ATPase VPS4 dissociates and recycles the endosomal components for the next cycle [34]. However, if de-ubiquitination is restrained, ILVs are subjected to lysosomal degradation [35]. Alternatively, the marker protein of exosomes, Alix, binds to ESCRT-III and guides the un-ubiquitinated cargo to the ILVs [34,35]. Besides it, the ESCRT-independent mechanism occurs via the melanosomal protein Pmel17 in melanocytes [36]. The association of the luminal domains of Pmel17 along with lipids contributes to ILV formation. It has also been reported that tetraspanin mediates cargo sorting and exosome secretion in an ESCRT-independent manner [37]. For instance, tetraspanin CD63 helps the loading of pigment cell-specific protein 17 into the ILV during the synthesis of the melanosome [37]. Similarly, the loading of MHC-II occurs exclusively in tetraspanin CD9-enriched membrane domains [38]. In addition, as reported previously, ceramide-rich parts of endosomes are prone to inward invagination, and hence, defects in the conversion of sphingomyelin into ceramide by sphingomyelinase (SMase) abrogates ILV formation, wherein, cholesterol is necessary for the formation of curved membrane structures called caveolae [39]. This has been confirmed for lipid-based cargo sorting as well as exosome secretion. It is imperative to understand that the absence of ESCRT machinery did not hamper the MVB formation, but instead manipulated ILV number and size, thus exemplifying coordination between ESCRT-dependent and -independent pathways during exosome biogenesis.
In view of exosome release, the involvement of molecular switches and motors (small GTPase, dynein and kinesin), cytoskeletal elements (microtubule and microfilaments) and membrane fusion complexes (SNARE complex) has been made crystal clear [40]. Rab GTPases control the trafficking and fusion of MVB with the plasma membrane. During the transport of MVB, the microtubule cytoskeleton and molecular motors direct its polarized distribution in the cells [40]. Rightly, the soluble N-ethylmaleimide (NEM)-sensitive factor attachment protein receptor (SNARE) complex is a core protein complex that assists membrane fusion and secretion [41]. Descriptively, the fusion process is initiated through the interaction of the plasma membrane protein syntaxin with synaptotagmin, the calcium sensor located on MVBs. Then, v-SNAREs of MVBs pair with the t-SNAREs, thus driving SNARE complex formation and fusion of membranes to release exosomes to the outer environment [40,41] (Figure 1). Further understanding of the complex process of exosome biogenesis should pave the way for novel therapeutic avenues.
With much detailed apprehension on the biogenesis of exosomes, efforts are in progress to inhibit the production or release of an exosome subpopulation that is involved in the pathology. Nevertheless, the available inhibitors are listed in Figure 1.

4. Exosome Classification and Its Biodistribution: Current State-of-the-Art

At least two distinct sub-types of exosomes have been discovered based on their modifications—natural exosomes and bioengineered exosomes. Subsequently, natural exosomes are subdivided into animal-derived exosomes and plant-derived exosomes based on their parental sources. With reference to animal-derived exosomes, almost all normal cells such as mesenchymal stem cells (MSCs), macrophages, natural killer (NK) cells, T and B immune cells and umbilical cord endothelial cells produce exosomes. Also, normal exosomes are plentily available in biofluids, such as plasma, urine, milk and saliva. In essence, tumor-cell-derived exosomes, because of their specific expression of tetraspanins, have also been considered for the delivery of chemotherapeutic or anti-cancer drugs. However, tetraspanins can sometimes lead to tumor growth and an increase in metastasis in certain types of cancer [42]. Therefore, the alarming risk associated with tumor exosomes jeopardizes the suitable therapeutic effects and aggravates patients’ malignancies, thus providing an awakening call to weigh the benefit-to-risk ratio and choose the right exosomes for therapy purposes. Based on these perspectives, several plant-derived exosomes have been explored for clinical use, as they are derived from purified sources and are considerably safe [43]. Contrastingly, engineered exosomes fall under the category of exosomes that are surface modified and loaded with substances to efficiently target disease sites and negate adverse profiles associated with treatments. With no available information to date, further sub-divisions of exosomes based on organophilicity, biodistribution and clearance may be considered [42,43].
Irrespective of the parental origin, it has been stated that the exosomes are largely accumulated in the liver, spleen, kidney and lungs. However, there is evidence that states that exosomes have asymmetric biodistribution based on their parental cell and the route of administration. Like, glioma-derived exosomes are demonstrated to efficiently deliver the drug to the brain [44]. Mesenchymal stem cell (MSC)-derived exosomes preferentially target kidneys in patients with kidney disorders [45]. Besides the exosome origin being one of the striking reasons, the route of administration can also be equally considered to alter exosome biodistribution. For instance, intramuscular administration of exosomes increases their bioavailability in the gastrointestinal tract. Alternatively, the oral gavage method of administration localized it more in the intestine. However, the most common method of administration is intravenous, in which exosomes travel via the heart and are entrapped in the capillaries of the lungs [46]. This highlights that the method of administration can lead to extreme variability and an asymmetric biodistribution pattern for exosomes.

5. Exosome in Diseases: The Boon against Evil

Capitalizing on the role of exosomes in the progression or treatment of several diseases is an important aspect of this review. Earlier described as “waste disposal units”, exosomes are now a hot research topic as they proportionately function between physiological and several pathological states. Finding how exosomes communicate with or affect neighboring/distant cells may provide insights into their roles in disease regulation, exploit them for drug delivery or monitor disease state in a non-invasive procedure.
Investigating the darker side of the exosome reveals that it secretes β-amyloid and hyperphosphorylated, misfolded tau into the extracellular space and leads to the pathological spreading of tauopathy in Alzheimer’s patients [47]. Exosomal alpha-synuclein (α-syn) promotes the aggregation of misfolded α-syn, a phenotype predominant in Parkinson’s disease [48]. An increase in the levels of circulating exosomes loaded with pro-inflammatory cytokines leads to persistent inflammation, as has been reported in multiple sclerosis and autoimmune encephalomyelitis [49]. In autoimmune rheumatoid arthritis (RA), exosomes derived from synovial fibroblasts and serum-induced Th17 differentiation and production of pro-inflammatory cytokines aggravate disease conditions [50]. In genetic diseases such as autosomal dominant polycystic kidney disease (ADPKD), cystic cells and urinary exosomes promote cyst growth in ADPKD patients [51]. In addition, fibroblasts of breast cancer origin secrete exosomes that mediate epithelial-to-mesenchymal transition (EMT) and metastasis via altered expression of lncRNA. A detailed analysis of miRNAs, lncRNAs, circRNAs and proteins that promote several diseases has been categorized in Table 1.
Owing to their ability to deliver mRNA and proteins to recipient cells at distant sites, exosomes can promote tumor metastasis and exert immune suppression to evade attack by the immune microenvironment. Gastric cancer cell exosomes establish an immunosuppressive environment for metastatic niche formation and exacerbate lung tumor metastasis [66]. In type 2 diabetes, exosomes released from adipocytes carry thrombospondin 5, which induces metastatic properties associated with the mesenchymal phenotype of breast cancer cells [67]. It is important to note that exosomal PD-L1 exhibits immunosuppressive function in the tumor microenvironment and promotes resistance to immune checkpoint blockade [12]. To note, exosomal PD-L1 from MEL624 cells decreased the proliferation of CD8+ T cells and reduced granzyme B secretion in an in vitro model of melanoma [68]. Moreover, tumor-derived exosomes re-educated neutrophils to exhibit an immunosuppressive microenvironment to promote gastric cancer via high mobility group box-1 (HMGB1) activated STAT-3 expression and increase PD-L1 activity [69]. Exosomes derived from patients with non-small cell lung cancer expressed PD-L1 and exhibited tumor escape via decreased IL-2 and IFN-γ secretion [70]. In a syngeneic model of prostate cancer, exosomal PD-L1 was resistant to anti-PD-L1 therapy, and its genetic blockade improved systemic anti-tumor response, strongly indicating the disease-promoting effect of exosomal PD-L1 towards tumor progression [71].
In another instance, exosomes shed from colon cancer cells showcased cetuximab resistance via inhibiting PTEN expression and concurrently, increasing the functional activity of the Akt pathway [72]. Interestingly, exosomes release circular RNA (circSKA3) that potentiates the formation of large colonies and the maintenance of invasiveness in breast cancer [73]. As evident, exosomes from the serum of patients with prostate cancer transfer pyruvate kinase M2 into bone stromal cells and promote its metastasis [74]. Exosome RNF126 derived from tumor cells promoted PTEN ubiquitination and macrophage infiltration, which led to nasopharyngeal carcinoma progression [75]. Melonoma-based exosomes increased PD-1 expression and reprogrammed mesenchymal stem cells to activate oncogenic and survival signals toward tumor metastasis [76]. Tumor exosomes display Tspan8 and CD151 that stimulate angiogenesis and tumor progression [77]. Cancer cell-derived exosomes are reported to stabilize cadherins and promote epithelial to mesenchymal transition that progresses cancer [78]. Taken together, the accumulating evidence concludes the cancer-promoting function of exosomes by subverting immune regulation.
As a bystander in therapy, exosomes of different origins have intrinsic remedial properties that can treat diseases, including cardiovascular, respiratory, kidney and brain disorders. Exosomes have the ability to increase drug half-life and stability by protecting them from degradation by digestive enzymes, and hence, serve as better vehicles for drug delivery in the treatment of cancer and other related diseases. Owing to the numerous undergoing clinical trials of exosome-based therapy, it can be speculated that it might contribute to better responses in patients and can thus be included under standard-of-care therapy for several diseases in the near future (Table 2) [79,80,81,82]. The incorporation of paclitaxel into exosomes exhibited better therapeutic efficacy in pancreatic cancer cells and in NODscid mice (n = 12) [83]. Exosomes have also been used to increase the stability and bioavailability of curcumin to mitigate inflammation and brain tumor growth (n = 5 mice per group) [84]. In fact, curcumin pre-treated lung cancer cells increased exosomal transcriptional factor 21 (TCF21) to subside lung cancer [85]. Doxorubicin-loaded exosomes exhibited higher efficacy with minimal levels of cardiotoxicity [86]. Similarly, exosomes released withaferin at target sites have been tested to treat lung tumor growth [87]. Besides transporting natural cargoes, exosomes are best suited as nanocarriers for the delivery of RNA (mRNA, siRNA, miRNA)-based therapeutics (Table 2). Markedly, specific disease-associated proteins and mRNAs of exosomes can be used as diagnostic markers. Apart from its role in delivering drugs and diagnosis, exosomes can be labeled with CFSE dyes, GFP-expressing plasmids and lipophilic agents (PKH67, Di dyes) to monitor treatment efficacy [88]. In addition, the bioluminescence resonance energy transfer (BRET) imaging technique scans the homing of exosomes at the target tissues and organs, which can be used to guide the development of therapeutics [89]. As a therapeutic carrier, diagnostic tool and best imaging agent, exosomes as a comrade outshine their criminal story and prove to be a boon in the medical field.

6. T Cell Exosomes: Its Biogenesis and Applicability

Although the information about the biogenesis of exosomes from T cells is preliminary, the omnipresent expression of tetraspanins makes it amenable to be involved in T cell exosome biogenesis [90]. Certain lipid metabolizing enzymes are also plausible to promote the maturation and release of exosomes by T cells. For instance, diacylglycerol kinase α helps in the secretion of CD63 exosomes from T cells [91]. An important clue for the involvement of condensed membranes in exosome formation is derived from the fact that exosomes derived from T cells are enriched with sphingomyelin and cholesterol. In addition, the participation of Tsg101 and, reportedly, HRS (exosomal markers) in exosome biogenesis rules out ESCRT components in the T cell biogenesis process [90].

7. Exosome Shedding at the Crossroads of Immune Synapse

It has been postulated that mature immune synapse formation launches exosome release from T cells. In this context, T-cell receptor (TCR) activation on the effector T cell induces the secretion of exosomes bearing CD63 and TCR, wherein different subpopulations of exosomes are released with the presence/absence of T cell co-stimulatory molecule signaling [92]. During the synapse formation, the secretory vesicles are polarized towards the central region of the formed immune synapse, called the central supramolecular activation cluster (cSMAC) for fusion and subsequent secretion in the synaptic cleft [93]. Disruption of Tsg101 and VPS4 interrupts the sorting of TCR into the secretory vesicle and secretion respectively, thus, rendering the important role of ESCRT components (Tsg101 and VPS4) in T cell exosome biology [92,93]. Thus, exosome synthesis in T cells upon encountering an antigen is a unique approach, and several tetraspanins, lipid components and ESCRT machinery can be pharmacologically targeted to block exosome secretion by T cells.
Regarding the functional role of T cell-based exosomes, it has been evidenced that exosomes with Fas ligand derived from T cells are lethal and induce death in other T lymphocytes [94]. It is also observed to promote tumor invasion in the lungs via the secretion of matrix metalloproteinase (MMP)-9 [90]. CD3+ T cell-derived exosomes function in accordance with IL-2 and promote the multiplication of autologous resting cells [95]. In addition, CD73-tagged exosomes released from T regulatory (Treg) cells induce the production of adenosine, which exhibits an anti-inflammatory response [96]. Furthermore, Treg cell-based exosomes release let-7b/7d and miRNA-I55 and abrogate Th1 differentiation and interferon (IFN)-γ production [97], and secretion of miR-150-5p and miR-142-3p to DCs suppresses TNF-α and IL-6 production via the increased amount of IL-10 cytokine production [98]. The levels of inhibitory molecules PD-1 and TIM-3 are increased in normal CD8 T cells as a result of lncRNA KCNQ1OT1-loaded T cell exosomes [99]. Yet importantly, CAR-T cell exosomes could surpass the disadvantages of CAR-T cell therapy and tend to dominate as therapeutic agents [98].

8. Th17 Cell Biology

CD4+ T cells are the central tenet in the promotion and maintenance of immune response. Defined as helper cells with a heterogeneous population, CD4+ T cells are categorized based on the transcription factor and cytokine profile. T helper (Th)-17 cells are a distinct subset of CD4+ T cells that expresses lineage-defining transcription factor RAR-related orphan receptor gamma (RORγT) and secretes IL-17 cytokine [100]. The differentiation and commitment of CD4+ Th17 cells is an intricate process and involves the “non-cytokine” mechanisms and “cytokine-induced” transcriptional program. The non-cytokine processes include the nature of antigen-presenting cells and their microbial peptides, the strength of interaction between TCR and antigenic peptide on MHC molecules and the activation of co-stimulatory signals [101]. It has long been reported that antigens from the fungus Candida albicans and its associated PAMP β-glucan differentiate fungus-specific Th17 cells [102]. However, there is a lack of concrete evidence regarding the TCR signals toward Th17 cell commitment. It is of the opinion that weak TCR signals in the proximity of Th17-promoting cytokines curb Th17 cell differentiation [103]. But, strong TCR signaling mediated activation of Tec family tyrosine kinase, interleukin-2-inducible T cell kinase (Itk), that skews the differentiation of CD4 T cells to Th17 phenotype. Interestingly, the binding of the nuclear factor of activated T cells (NFATc1) to the promoter region of IL-17 is also essential to differentiate Th17 cells even in the presence of strong TCR signals [104]. Alongside, low CD28 and high CD40-CD40L co-stimulatory signals with impaired IL-2 levels strengthen the Th17 developmental axis [105]. In sum, the nature of the antigen and the strength of TCR and co-stimulatory signals are instrumental in the biased commitment of CD4+ T cells to a distinct Th phenotype.
Along several lines, the inclusion of several cytokines and transcription factors (TFs) have been well reported that guide Th17 differentiation and lineage commitment. Particularly, TGF-β and IL-6 are the important cytokines that prime Th17 differentiation [106]. Furthermore, IL-21, IL-23 and IL-1β contribute to Th17 cell phenotype commitment [107]. Likewise, some TFs also have been demonstrated to participate to regulate the Th17 development process. Notably, in response to IL-6 cytokine stimulation, the STAT-3/ROR-γT axis is activated that mediates the formation of Th17 cells and the IL-17 transcriptional program [107]. Unlike ROR-γT, IL-23 promotes AhR transcription factor activation to expand the Th17 population [108]. It is likely that the induction of Th17 cell plasticity is also governed by interferon regulatory factor 4 (IRF4) TF, and IRF4 deficient cells impair IL-17 production and Th17 responses under Th17 differentiation conditions [109]. Furthermore, another transcription factor basic leucine zipper ATF-like transcription factor (BATF) forms a complex with IRF4 on the IL-17 promoter region downstream of TCR signaling [110]. However, the independent function of these transcription factors is yet to be determined along the Th17 differentiation axis. Though not much explored, the importance of Ying Yang 1 (YY1) has been demonstrated to bind to the T-bet promoter region and influence Th17 pathogenicity [111]. Integrated genome studies have further revealed that Zinc finger E-box binding homeobox 1 (ZEB1) necessitates JAK-2/STAT-3 dependent Th17 cell fate [112].
In addition, RNA-binding proteins also control Th17 differentiation and function. Reportedly, human antigen R (HuR) post-transcriptionally promotes ROR-γT expression and Th17 cell responses [113]. Very recently, the role of IGF2 mRNA-binding protein 2 (IMP2) RNA-binding protein drives the Th17 cell differentiation program and infiltration into the lymph nodes of the inflamed brain [114]. In sum, a detailed understanding of the factors that regulate Th17 biology will help open new avenues for exosome-targeted therapies in Th17-mediated diseases (Figure 2).

9. Th17 Cell Pathologies: A Snap on Th17 Exosomes

The pace in the use of IL-17 and ROR-γT inhibitors in clinics has resurfaced the critical role of Th17 in the exacerbation of several cancers, autoimmune diseases, and genetic disorders. Recently, Th17 cell populations are aggressively increased in COVID-19 patients with pulmonary complications. There are reports that correlate Th17 with neutrophilia and NETosis pathologies in COVID patients. Anti-IL-17 monoclonal antibodies (mAbs) netakimab administered to COVID patients mitigated lung lesions and demand for oxygen support [115]. Also, influenza virus-induced lung injury was associated with increased levels of IL-17 and Th17 population [116]. IL-17 contributes to liver fibrosis and necrosis of hepatocytes in mice infected with the herpes virus [117]. Notwithstanding, stromal keratitis in mice with corneal infection is an outcome of Th17 cells [118]. Furthermore, Th17 cells mediate myocarditis in coxsackievirus B3-infected mice [119], suggesting that targeting Th17 cells and IL-17 may be a better choice to control viral infections.
The pathological phenomenon of Th17 cells has also been reflected in autoimmune and inflammatory diseases, including psoriasis, rheumatoid arthritis, ankylosing spondylitis, SLE and multiple sclerosis [17]. Altered levels of Th17 cells have been the biggest disease-promoting factor in psoriasis [120]. It has been documented that Th17 in the dermis of psoriatic skin elicits self-lipid antigen production and inflammation [121]. Several mAbs (secukinumab, ixekizumab, bimekizumab, brodalumab, risankizumab, trldrakizumab and guselkumab) that target IL-17, IL-23 and IL-17RA have proven efficacious in the treatment of psoriasis [122]. Similarly, Th17 cells were shown to play a crucial role in joint inflammation and cartilage damage in autoimmune arthritis [123]. Despite the role of Th17 cells in SLE and multiple sclerosis remains skeptical, the fact that it bypasses the blood–brain barrier and accumulates at lesional sites showcases its participation in disease development [124]. It has also been portrayed that Huntington’s disease is a Th17 inflammatory reaction-mediated autoimmune disorder [125]. Besides, research on the active role of Th17 cells in autoimmune uveitis and Type I diabetes is in progress where inflammation is one of the core pathologies involved in disease progression.
On the side of genetic disorders, though much attention has not been drawn to studying Th17 pathology, it has still been acknowledged that Th17 are instructive cells that impact the pathological phenotype of sickle cell disease [126]. The levels of IL-17 are also upregulated in autosomal dominant polycystic kidney disease (ADPKD) [127], whereas its definitive role is yet to be explored. Certainly, a clear picture of the pathogenesis of Th17 cells and its differentiation process can help us use exosome engineering strategies to cope with the disease’s detrimental process.

10. Th17 Cell-Derived Exosomes in Disease Pathologies

Little is known about the molecular basis of exosomes in the regulation of Th17 differentiation toward disease progression. Recently, a study has demonstrated that contactin-1 in exosomes derived from epithelial cell signals could activate Th17 cell responses and promote asthma pathology [128]. A similar study has reported the relay of lncRNA CRNDE-h via tumor exosomes to escalate ROR-γT expression, Th17 differentiation and the activity of IL-17 cytokines [129]. In addition, LPS-stimulated human thymic mesenchymal stromal cells release exosomes that help differentiate CD4+ T cells into a proinflammatory Th17 cells phenotype [130]. In preeclampsia (PE) patients, exosomes significantly increase Th17 cell numbers and exacerbate PE pathogenesis [131]. Exosomes accumulated from tumor-infiltrating cells redistribute miR-451 from cancer cells to T cells, thus prompting its differentiation to the Th17 cell phenotype [132]. Noticeably, synovial fibroblast exosomes promoted Th17 differentiation under hypoxic conditions that potentiate RA pathogenesis [50]. Despite the adverse influence of exosomes in diseases, their potential in drug delivery and treatment cannot be circumvented and is currently of great research interest.

11. Can Exosomes Potentiate as a Bona Fide Therapeutic Candidate against Th17 Biology?

Fortunately, the ability of exosomes derived from different cells that act as drug decoys to modulate disease response has now been reported. Remarkably, granulocytic exosomes derived from bone marrow overexpress miR-29a-3p and miR-93-5P inhibit Th17 differentiation and attenuate autoimmune arthritis [133]. On the same grounds, human bone marrow mesenchymal stem cells-derived exosomes could downregulate the Th17 population and subside periodontal inflammation via modulating YAP1/Hippo signaling pathway [134]. Yet importantly, IFN-γ-stimulated mesenchymal stem cell exosomes could boost the levels of miR-125a and miR-125b that directly interfered with STAT-3 transcription and repressed Th17 differentiation in colitis mice [135]. Also, 3D exosomes improved colitis and periodontitis via restoration of Th17/Treg balance via the miR-1246/Nfat5 axis [136]. Th17 cell responses could also be strongly nullified by olfactory ecto-mesenchymal stem cells exosome in an experimental mice model of colitis [137]. Similarly, mesenchymal stem cell-derived exosomes from the human umbilical cord (UC) could negate Th17 differentiation and treat inflammatory bowel disease [138]. UC-derived blood mesenchymal stem cell exosomes could restore miR-19B/KLF13 expression to suppress Th17 cell proportion in SLE patients [139]. Interestingly, mesenchymal stem cell exosomes derived from labial glands could inhibit Th17 differentiation, and subsequently suppress IL-17 production in the treatment of Sjogren’s syndrome [140]. Of note, dendritic cell (DC) derived exosomes block effector Th17 function and protect the liver from ischemia reperfusion [141]. Bone marrow-derived mesenchymal stem cell exosomes deliver miR-23a-3p to balance the proportion of Th17 cells and halt aplastic anemia [142]. On the highlight, gene-modified DC-derived exosomes loaded with FOXP3 results in an obvious decrease in Th17 cells and ameliorate autoimmune encephalomyelitis [143]. And, TGF-β gene-modified DCs release exosomes that have been proven more efficacious in treating Th17-mediated inflammatory bowel disease [144]. Alternatively, a study has demonstrated that heat-stressed tumor-cell-derived exosomes exhibit anti-tumor effects via biased differentiation of Th17 cells [145]. In patients with hypertrophic scaring, adipose-derived stem cell exosomes could attenuate ROR-γT expression and Th17 differentiation, thus, defining its role in disease suppression [146]. Taken together, it is conceivable and of the opinion that exosomes can be used as promising bio-shuttles against Th17-associated diseases.
To further claim exosomes to be a powerful candidate as drug delivery vehicles against Th17 cells, methods of drug loading, exosome uptake efficiency and the ease to release the payload are further extrapolated. In general, drugs can be directly loaded into the exosomes or loaded into the parental cells, which are then released into the exosomes. In spite of several challenges such as nucleic acid degradation associated with electroporation and ultrasound loading techniques, they are considered the most efficient methods for exosomal drug loading. Moreover, techniques such as thermal shock and extrusion demand high-end equipment, which increases costs. Again, the addition of saponins and transfection reagents can increase the chances of drug toxicity. To highlight, though incubation methods result in low productivity, it protects the membrane from degradation and has the advantages of low cost and high safety. Howbeit, to overcome low drug yield via the incubation method, strategies of increasing drug concentration and continuous stirring during incubation can be implied [147]. Findings have demonstrated a new strategy (late domain-ubiquitin and ESCRT vesicle trafficking technique) for the successful loading of biologically active molecules that, however [148], needs further investigation for loading drugs against Th17 cells. Once drugs are sequestered inside the exosomes, the process of uptake occurs via direct membrane fusion, pinocytosis, and endocytosis (Figure 1). The uptake efficiency can be manipulated via surface engineering methods. For instance, a previous study made use of electrostatic interactions to bind cationic lipids onto exosome surfaces to increase their uptake efficiency [19]. Another modification approach includes the attachment of cell-penetrating peptides (CPPs), in particular arginine-rich CPPs, that increase exosome internalization by micropinocytosis [149]. Also, an increase in membrane rigidity via enrichment in sphingolipids and cholesterol improves fusion efficiency between exosomes and recipient cells [19]. Post internalization, a fraction of exosomes fuses with the limiting membrane of endosomes/lysosomes to release the cargo to the cell cytosol in an acidification-dependent manner. An interesting aspect studied previously demonstrated that exosome membrane integration with connexin 43 necessitates hexametric channel formation and provides a direct route for cytoplasmic transfer of exosomal payload [150]. Further detailed investigation of these methods in relation to exosomal loading of anti-Th17 drugs and their release inside Th17 cells needs to be clarified.

12. Exosomes as a Budding Bio-Shuttle: A Perspective in the Landscape of Th17 Cell

Several delivery units, such as liposomes, nano emulsions, nano gels and micelles, are underway for the targeted transfer of molecules in diseases. However, it cannot be overlooked that it faces several challenges, including poor solubility, stability, loading efficiency and, last but not the least, its clearance from the reticuloendothelial system [151]. By contrast, exosomes are naturally biocompatible cargo delivery “trucks” across many biological membranes. Recently, experiments have also been conducted to delineate the source of exosomes and their isolation techniques. Immature dendritic cells, for example, are the most potent source for the isolation of therapeutic exosomes as it lacks CD86 and MHC markers, and thus, possess low immunogenicity [152]. Moreover, mesenchymal stem cells are self-renewal and are a good source of exosomes as they exhibit immunosuppressive effects [153]. Above all, the exosomes from cancer cells possess significant tetraspanin content that facilitates ligand interaction in several tissues [154]. Among several biofluids, peripheral blood is a good source of exosomes obtained from different cells [155]. Other sources of exosomes include plants and fruits that can target cancer and inflammatory diseases.
Although the scope of exosome as therapeutics has been well described, its usage in ferrying drug cargo is not much explored in Th17 biology. At the cutting edge, the rationale for its design to deliver bio-shuttles against the powerful Th17 cells is to strengthen drug efficacy, overcome drug resistance and limit off-target side effects. This section of the review highlights the possible bio-shuttles that can specifically be delivered via the use of exosomes to hamper Th17 cells. A reasonable explanation will definitely be in hand to claim these bio-shuttles as anti-IL-17 therapies.

13. Packaging of Small Molecules

Small molecules are selective drugs that specifically function as enzyme or receptor inhibitors and allosteric modulators in order to disrupt protein–protein interactions. These drugs are prescribed with numerous warnings and advice to discontinue if the side effects are strong. Moreover, natural compounds that are small molecules show limited efficacy because of their low bioavailability. These limitations might prompt one to think that exosomes can be a prospective delivery platform for small molecules to dynamically increase their uptake, improve their availability at target sites and exhibit superior therapeutic indices. No reports have yet studied the application of exosomes in improving the delivery and efficacy of small-molecule drugs against the Th17 catastrophe. In light of these shortcomings, we list here the small molecules cargo that can be packaged into the exosomes for specific targeting of Th17 differentiation and function (Table 3).

14. Packaging of Proteins

Several investigations have made use of exosomes in the distribution of large molecules like peptides and proteins to treat diseases. The high potency, low toxicity and high selectivity make them outstanding therapeutic candidates over the small molecule. Howbeit, rapid clearance from the body, poor membrane permeability and poor metabolic stability demand a delivery vehicle [169]. Exosomes have been proven as a better vehicle for autoimmune diseases such as SLE, where tolerance therapy using the autoantigenic peptide, for instance, histone peptide H471–94, is a highly opted therapeutic option [170]. In another instance, exosome can be a carrier for a peptide derived from the globular C-terminal domain of adiponectin (e.g., KS23) that has characteristic anti-inflammatory properties and decreases the proportion of Th17 cells as reported in experimental autoimmune uveitis [171]. Interestingly, packaging antagonistic peptides that target the first and second extracellular loops of chemokine CCR5 and p19 subunit of IL-23 into exosomes may be effective in suppressing the IL-23/Th17 axis [172]. As αv integrin is present in dendritic cell populations, it necessitates Th17 development, whereas an antagonist peptide, VnP-16, when packaged inside exosomes and administered, could result in impairing Th17 differentiation and exhibit better clinical outcomes [173]. Notwithstanding, it is tempting to speculate that N-formyl peptide receptor 2 (FPR2) agonists, such as scolopendrine, Cpd43 and WKYMVm, could be loaded into exosomes to suppress dendritic cell maturation and inhibit Th17 responses, as demonstrated in a mouse model of autoimmune arthritis [174]. Of note, an exosome-released peptide agonist to ADAM17 that is responsible for membrane shedding of IL-23R can block Th17 functions in several Th17-associated diseases [175]. Critically, an antagonist peptide as a drug delivered by exosomes to CD71 can disrupt Th17 differentiation [176]. Furthermore, exosome delivery of antagonistic peptides, which target miRNA (e.g., miR-21), that incite Th17 pathogenesis can be a strategy for a peptide-based therapeutic approach.

15. Packaging of Genetic Substances

As exosomes are efficient in preventing nucleic acids (siRNAs and miRNAs) from degradation, they represent ideal vectors for gene therapy approaches. Besides, exosomes are naturally produced transporters of RNAs intercellularly. In our perspective, Phosphatase and tensin homolog (PTEN) regulatory miR-22 can be loaded into exosomes to repress Th17 cell responses [177]. In another instance, exosome-based delivery of miR-221-5p could be used to efficiently target the suppressor of cytokine signaling (SOCS)1 to inhibit Th17 cells in asthma [178]. On the upper hand, exosome-based delivery of miR-340 might be capable of attenuating psoriasis by directly targeting IL-17A [179]. In addition, nuclear receptor 4A2 (NR4A2) specific siRNA can be used as a drug to be delivered by exosomes and decrease Th17 effector function [180].
More recently, the introduction of CRISPR/Cas9 technology has revolutionized the treatment of several diseases. It has been an attractive approach due to the high precision and flexibility of manipulating a specific gene. However, finding a suitable vehicle to deliver CRISPR/Cas9 is a major hurdle [181]. Exosomes, as natural vehicles, possess low immunogenicity and can therefore be a potent approach in delivering CRISPR/Cas9 system into cells. It is true that exosomes can be exploited for CRISPR-mediated blockade of glycolysis, which can result in the loss of Th17 cells [182]. As a key point, targeting oxidative phosphorylation via exosome-based delivery of CRISPR/Cas9 might abrogate Th17 cells’ resistance to apoptosis [183], which can be a promising approach. It is probable that CRISPR/Cas9 release via exosomes to manipulate RNA binding proteins, such as HuR, can also help protect against Th17 responses [113,184].

16. Strategies to Engineer Exosomes for Specific Targeting of Th17 Cells

Nowadays, therapeutic exosomes are delivered to specific sites via active targeting strategies that utilize techniques to modulate the surface of exosomes. There are two ways for active targeting of exosomes, a non-genetic approach that defines direct manipulation of exosomal surface and genetic approaches that indirectly modulates the exosomes via genetic manipulation of its cellular source [29]. Here, we brief on the exosomal surface engineering that can be successfully accomplished for the targeted delivery of exosomes to Th17 cells.
The non-genetic/direct method utilizing chemical (click chemistry) or physical techniques (hydrophobic insertion and receptor–ligand interaction) can help acquire targetability in therapeutic exosomes. Click chemistry is a covalent bonding strategy in which the ligand molecules form covalent bonds with exosomes [185]. For instance, designed peptides for Th17 cell-specific receptors or integrins can be integrated into exosomes via click chemistry to generate Th17 cell-targeting exosomes [186]. It is a matter of debate whether αvβ3 targeted peptide can be conjugated to exosome surface via click chemistry to target Th17 cells. However, the method of click chemistry can be detrimental as it necessitates toxic chemicals for stabilizing bonds, thus, alarming for its use in therapeutics. Apart from the covalent binding method, physical methods can opt as one of the options for allowing the binding of ligand moieties to exosome membranes. Attachment of CD30 aptamer to exosomes by hydrophobic interactions can be employed for Th17 pathological condition as CD30 is actively expressed by activated T cells [187] (Figure 3).
Further, genetically engineered exosomes warrant attention in the active targeting of Th17 cells. To deliver exosomes carrying therapeutics to Th17 cells, targeting entities, such as antibodies or peptides that could bind specifically to integrins or receptors expressed on Th17 cells, should be showcased on the exosome surface through a genetic modification approach. Exosomes expressing anti-prohibitins attached to GPI-anchored proteins or the C1C2 domain of cadherin in exosomes can be targeted to Th17 cells, as prohibitins are highly expressed on the surface of murine and human Th17 cells [188]. In another instance, exosomes can be genetically manipulated to express a disintegrin and metalloproteinase 15 that can bind to αvβ3 overexpressing Th17 cells [189]. Overall, it is undeniable that efforts to express Th17-targeting entities on the surface of exosomes via conjugation with the GP1 and C1C2 domains and tetraspanins associated with exosome membranes can be a promising approach for the active targeting of the Th17 cells.

17. Present Challenges and Future Perspectives

Numerous exosomes are filed for clinical trials to assess the risk-benefit ratio, and their discovery as an outstanding drug delivery platform is opening the door toward the cure of several diseases. Unfortunately, cells produce a small number of exosomes that are insufficient for clinical studies, and miRNA that is loaded as therapeutic cargo, if anionic, fails to cross biological membranes and accumulates in kidneys and livers due to its short half-life. Specifically, little knowledge of the targeting entities of Th17 cells shields the exploration of exosomes against their pathogenicity. Despite numerous challenges, such as inadequate characterization methods, a lack of specific markers and stability issues, these naturally derived vesicles have tremendous potential in biomedical research. Therefore, it is suggestible to find ways to overcome the shortcomings and obstacles to label exosome drug delivery as a safe and efficient approach in clinics.
Several leading companies across the globe are conscious of overcoming the scientific and technological challenges faced during exosome clinical trials. As an alternative, synthetic exosome mimics or cell membrane-cloaked nanoparticles have been fabricated for drug delivery with the potency to increase drug half-life while still sustaining equal biodistribution and clearance rates as that of natural exosomes. The development of cost-effective strategies and isolation and exosome characterization methods with high sensitivity for large-scale production is in progress. Exciting new approaches for effective drug loading are being explored. Certainly, this will position exosomes as an attractive “waste disposal unit” that can be exploited for therapeutic usage in clinics.
In conclusion, it is envisaged that the integration of reliable isolation methods and drug packaging techniques will gear up its validation in in vivo studies that may lay a solid foundation to solve the puzzle of its clinical translation.

Funding

This research was funded by National Institutes of Health grant R01 DK129241 and DK126662 (X. Li).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Agamah, F.E.; Mazandu, G.K.; Hassan, R.; Bope, C.D.; Thomford, N.E.; Ghansah, A.; Chimusa, E.R. Computational/in silico methods in drug target and lead prediction. Brief. Bioinform. 2020, 21, 1663–1675. [Google Scholar] [CrossRef]
  2. Lin, A.; Giuliano, C.J.; Palladino, A.; John, K.M.; Abramowicz, C.; Yuan, M.L.; Sausville, E.L.; Lukow, D.A.; Liu, L.; Chait, A.R.; et al. Off-target toxicity is a common mechanism of action of cancer drugs undergoing clinical trials. Sci. Transl. Med. 2019, 11, eaaw8412. [Google Scholar] [CrossRef]
  3. Zhao, Z.; Ukidve, A.; Kim, J.; Mitragotri, S. Targeting Strategies for Tissue-Specific Drug Delivery. Cell 2020, 181, 151–167. [Google Scholar] [CrossRef]
  4. Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef] [PubMed]
  5. Boukouris, S.; Mathivanan, S. Exosomes in bodily fluids are a highly stable resource of disease biomarkers. Proteom. Clin. Appl. 2015, 9, 358–367. [Google Scholar] [CrossRef] [PubMed]
  6. Wei, H.; Chen, Q.; Lin, L.; Sha, C.; Li, T.; Liu, Y.; Yin, X.; Xu, Y.; Chen, L.; Gao, W.; et al. Regulation of exosome production and cargo sorting. Int. J. Biol. Sci. 2021, 17, 163–177. [Google Scholar] [CrossRef] [PubMed]
  7. Lin, Y.; Lu, Y.; Li, X. Biological characteristics of exosomes and genetically engineered exosomes for the targeted delivery of therapeutic agents. J. Drug Target. 2019, 28, 129–141. [Google Scholar] [CrossRef] [PubMed]
  8. Kugeratski, F.G.; Kalluri, R. Exosomes as mediators of immune regulation and immunotherapy in cancer. FEBS J. 2020, 288, 10–35. [Google Scholar] [CrossRef] [PubMed]
  9. Anel, A.; Gallego-Lleyda, A.; de Miguel, D.; Naval, J.; Martínez-Lostao, L. Role of Exosomes in the Regulation of T-Cell Mediated Immune Responses and in Autoimmune Disease. Cells 2019, 8, 154. [Google Scholar] [CrossRef]
  10. Ahmadi, M.; Rezaie, J. Tumor cells derived-exosomes as angiogenenic agents: Possible therapeutic implications. J. Transl. Med. 2020, 18, 1–17. [Google Scholar] [CrossRef]
  11. Maybruck, B.T.; Pfannenstiel, L.W.; Diaz-Montero, M.; Gastman, B.R. Tumor-derived exosomes induce CD8+ T cell suppressors. J. Immunother. Cancer 2017, 5, 65. [Google Scholar] [CrossRef]
  12. Ye, L.; Zhu, Z.; Chen, X.; Zhang, H.; Huang, J.; Gu, S.; Zhao, X. The Importance of Exosomal PD-L1 in Cancer Progression and Its Potential as a Therapeutic Target. Cells 2021, 10, 3247. [Google Scholar] [CrossRef] [PubMed]
  13. Skriner, K.; Adolph, K.; Jungblut, P.R.; Burmester, G.R. Association of citrullinated proteins with synovial exosomes. Arthr. Rheum. 2006, 54, 3809–3814. [Google Scholar] [CrossRef] [PubMed]
  14. Hao, S.; Bai, O.; Yuan, J.; Qureshi, M.; Xiang, J. Dendritic cell-derived exosomes stimulate stronger CD8+ CTL responses and anti-tumor immunity than tumor cell-derived exosomes. Cell Mol. Immunol. 2006, 3, 205–211. [Google Scholar] [PubMed]
  15. Chen, L.; Deng, H.; Cui, H.; Fang, J.; Zuo, Z.; Deng, J.; Li, Y.; Wang, X.; Zhao, L. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget 2017, 9, 7204–7218. [Google Scholar] [CrossRef] [PubMed]
  16. Wilke, C.M.; Kryczek, I.; Wei, S.; Zhao, E.; Wu, K.; Wang, G.; Zou, W. Th17 cells in cancer: Help or hindrance? Carcinog 2011, 32, 643–649. [Google Scholar] [CrossRef]
  17. Yang, J.; Sundrud, M.S.; Skepner, J.; Yamagata, T. Targeting Th17 cells in autoimmune diseases. Trends Pharmacol. Sci. 2014, 35, 493–500. [Google Scholar] [CrossRef]
  18. Zou, J.Y.; Su, C.H.; Luo, H.H.; Lei, Y.Y.; Zeng, B.; Zhu, H.S.; Chen, Z.G. Curcumin converts Foxp3+ regulatory T cells to T helper 1 cells in patients with lung cancer. J. Cell Biochem. 2017, 119, 1420–1428. [Google Scholar] [CrossRef]
  19. Gurung, S.; Perocheau, D.; Touramanidou, L.; Baruteau, J. The exosome journey: From biogenesis to uptake and intracellular sig-nalling. Cell Commun. Signal. 2021, 19, 47. [Google Scholar] [CrossRef]
  20. Gheinani, A.H.; Vögeli, M.; Baumgartner, U.; Vassella, E.; Draeger, A.; Burkhard, F.C.; Monastyrskaya, K. Improved isolation strategies to increase the yield and purity of human urinary exosomes for biomarker discovery. Sci. Rep. 2018, 8, 3945. [Google Scholar] [CrossRef]
  21. Sadeghi, S.; Tehrani, F.R.; Tahmasebi, S.; Shafiee, A.; Hashemi, S.M. Exosome engineering in cell therapy and drug delivery. Inflammopharmacology 2023, 31, 145–169. [Google Scholar] [CrossRef] [PubMed]
  22. Chen, H.; Wang, L.; Zeng, X.; Schwarz, H.; Nanda, H.S.; Peng, X.; Zhou, Y. Exosomes, a new star for targeted delivery. Front. Cell Dev. Biol. 2021, 9, 2827. [Google Scholar] [CrossRef] [PubMed]
  23. Andreu, Z.; Yáñez-Mó, M. Tetraspanins in extracellular vesicle formation and function. Front. Immunol. 2014, 5, 442. [Google Scholar] [CrossRef] [PubMed]
  24. Wubbolts, R.; Leckie, R.S.; Veenhuizen, P.T.; Schwarzmann, G.; Mȯbius, W.; Hoernschemeyer, J.; Slot, J.W.; Geuze, H.J.; Stoorvogel, W. Proteomic and biochemical analyses of human B cell-derived exosomes: Potential implications for their function and mul-tivesicular body formation. J. Biol. Chem. 2003, 278, 10963–10972. [Google Scholar] [CrossRef]
  25. Skotland, T.; Hessvik, N.P.; Sandvig, K.; Llorente, A. Exosomal lipid composition and the role of ether lipids and phosphoinositides in exosome biology. J. Lipid Res. 2019, 60, 9–18. [Google Scholar] [CrossRef]
  26. Zhao, L.; Ma, X.; Yu, J. Exosomes and organ-specific metastasis. Mol. Therapy-Methods Clin. Dev. 2021, 22, 133–147. [Google Scholar] [CrossRef]
  27. Xu, M.; Feng, T.; Liu, B.; Qiu, F.; Xu, Y.; Zhao, Y.; Zheng, Y. Engineered exosomes: Desirable target-tracking characteristics for cere-brovascular and neurodegenerative disease therapies. Theranostics 2021, 11, 8926. [Google Scholar] [CrossRef]
  28. Shen, A.-R.; Zhong, X.; Tang, T.-T.; Wang, C.; Jing, J.; Liu, B.-C.; Lv, L.-L. Integrin, Exosome and Kidney Disease. Front. Physiol. 2021, 11, 627800. [Google Scholar] [CrossRef]
  29. Liang, Y.; Duan, L.; Lu, J.; Xia, J. Engineering exosomes for targeted drug delivery. Theranostics 2021, 11, 3183–3195. [Google Scholar] [CrossRef]
  30. Wahlund, C.J.E.; Güclüler, G.; Hiltbrunner, S.; Veerman, R.E.; Näslund, T.I.; Gabrielsson, S. Exosomes from antigen-pulsed dendritic cells induce stronger antigen-specific immune responses than microvesicles in vivo. Sci. Rep. 2017, 7, 1–9. [Google Scholar] [CrossRef]
  31. Fu, W.; Lei, C.; Liu, S.; Cui, Y.; Wang, C.; Qian, K.; Li, T.; Shen, Y.; Fan, X.; Lin, F.; et al. CAR exosomes derived from effector CAR-T cells have potent antitumour effects and low toxicity. Nat. Commun. 2019, 10, 4355. [Google Scholar] [CrossRef]
  32. Enqvist, M.; Ask, E.H.; Forslund, E.; Carlsten, M.; Abrahamsen, G.; Béziat, V.; Andersson, S.; Schaffer, M.; Spurkland, A.; Bryceson, Y.; et al. Coordinated Expression of DNAM-1 and LFA-1 in Educated NK Cells. J. Immunol. 2015, 194, 4518–4527. [Google Scholar] [CrossRef]
  33. Wang, S.; Shi, Y. Exosomes Derived from Immune Cells: The New Role of Tumor Immune Microenvironment and Tumor Therapy. Int. J. Nanomed. 2022, 17, 6527–6550. [Google Scholar] [CrossRef] [PubMed]
  34. Larios, J.; Mercier, V.; Roux, A.; Gruenberg, J. ALIX- and ESCRT-III–dependent sorting of tetraspanins to exosomes. J. Cell Biol. 2020, 219, e201904113. [Google Scholar] [CrossRef] [PubMed]
  35. Krylova, S.V.; Feng, D. The Machinery of Exosomes: Biogenesis, Release, and Uptake. Int. J. Mol. Sci. 2023, 24, 1337. [Google Scholar] [CrossRef]
  36. Colombo, M.; Moita, C.; van Niel, G.; Kowal, J.; Vigneron, J.; Benaroch, P.; Manel, N.; Moita, L.F.; Théry, C.; Raposo, G. Analysis of ESCRT functions in exosome biogenesis, composition and secretion highlights the heterogeneity of extracellular vesicles. J. Cell Sci. 2013, 126, 5553–5565. [Google Scholar] [CrossRef]
  37. Van Niel, G.; Charrin, S.; Simoes, S.; Romao, M.; Rochin, L.; Saftig, P.; Marks, M.S.; Rubinstein, E.; Raposo, G. The tetraspanin CD63 regulates ESCRT-independent and-dependent endosomal sorting during melanogenesis. Dev. Cell 2011, 21, 708–721. [Google Scholar] [CrossRef] [PubMed]
  38. Brosseau, C.; Colas, L.; Magnan, A.; Brouard, S. CD9 Tetraspanin: A New Pathway for the Regulation of Inflammation? Front. Immunol. 2018, 9, 2316. [Google Scholar] [CrossRef] [PubMed]
  39. Albacete-Albacete, L.; Navarro-Lérida, I.; López, J.A.; Martín-Padura, I.; Astudillo, A.M.; Ferrarini, A.; Van-Der-Heyden, M.; Balsinde, J.; Orend, G.; Vázquez, J.; et al. ECM deposition is driven by caveolin-1–dependent regulation of exosomal biogenesis and cargo sorting. J. Cell Biol. 2020, 219, e202006178. [Google Scholar] [CrossRef]
  40. Hessvik, N.P.; Llorente, A. Current knowledge on exosome biogenesis and release. Cell Mol. Life Sci. 2018, 75, 193–208. [Google Scholar] [CrossRef]
  41. Mashouri, L.; Yousefi, H.; Aref, A.R.; Ahadi, A.M.; Molaei, F.; Alahari, S.K. Exosomes: Composition, biogenesis, and mechanisms in cancer metastasis and drug resistance. Mol. Cancer 2019, 18, 75. [Google Scholar] [CrossRef]
  42. Zhang, Y.; Bi, J.; Huang, J.; Tang, Y.; Du, S.; Li, P. Exosome: A Review of Its Classification, Isolation Techniques, Storage, Diagnostic and Targeted Therapy Applications. Int. J. Nanomed. 2020, 15, 6917–6934. [Google Scholar] [CrossRef] [PubMed]
  43. Suharta, S.; Barlian, A.; Hidajah, A.C.; Notobroto, H.B.; Ana, I.D.; Indariani, S.; Wungu, T.D.; Wijaya, C.H. Plant-derived exosome-like nanoparticles: A concise review on its extraction methods, content, bioactivities, and potential as functional food ingredient. J. Food Sci. 2021, 86, 2350–2838. [Google Scholar] [CrossRef] [PubMed]
  44. Lee, H.; Bae, K.; Baek, A.-R.; Kwon, E.-B.; Kim, Y.-H.; Nam, S.-W.; Lee, G.H.; Chang, Y. Glioblastoma-Derived Exosomes as Nanopharmaceutics for Improved Glioma Treatment. Pharmaceutics 2022, 14, 1002. [Google Scholar] [CrossRef]
  45. Cao, Q.; Huang, C.; Chen, X.-M.; Pollock, C.A. Mesenchymal Stem Cell-Derived Exosomes: Toward Cell-Free Therapeutic Strategies in Chronic Kidney Disease. Front. Med. 2022, 9, 816656. [Google Scholar] [CrossRef] [PubMed]
  46. Kang, M.; Jordan, V.; Blenkiron, C.; Chamley, L.W. Biodistribution of extracellular vesicles following administration into animals: A systematic review. J. Extracell. Vesicles 2021, 10, e12085. [Google Scholar] [CrossRef] [PubMed]
  47. Miyoshi, E.; Bilousova, T.; Melnik, M.; Fakhrutdinov, D.; Poon, W.W.; Vinters, H.V.; Miller, C.A.; Corrada, M.; Kawas, C.; Bohannan, R.; et al. Exosomal tau with seeding activity is released from Alzheimer’s disease synapses, and seeding potential is associ-ated with amyloid beta. Lab. Investig. 2021, 101, 1605–1617. [Google Scholar] [CrossRef]
  48. Guo, M.; Wang, J.; Zhao, Y.; Feng, Y.; Han, S.; Dong, Q.; Cui, M.; Tieu, K. Microglial exosomes facilitate α-synuclein transmission in Parkinson’s disease. Brain 2020, 143, 1476–1497. [Google Scholar] [CrossRef]
  49. Lee, J.Y.; Park, J.K.; Lee, E.Y.; Lee, E.B.; Song, Y.W. Circulating exosomes from patients with systemic lupus erythematosus induce a proinflammatory immune response. Arthritis Res. Ther. 2016, 18, 264. [Google Scholar] [CrossRef]
  50. Ding, Y.; Wang, L.; Wu, H.; Zhao, Q.; Wu, S. Exosomes derived from synovial fibroblasts under hypoxia aggravate rheumatoid arthritis by regulating Treg/Th17 balance. Exp. Biol. Med. 2020, 245, 1177–1186. [Google Scholar] [CrossRef]
  51. Ding, H.; Li, L.X.; Harris, P.C.; Yang, J.; Li, X. Extracellular vesicles and exosomes generated from cystic renal epithelial cells promote cyst growth in autosomal dominant polycystic kidney disease. Nat. Commun. 2021, 12, 1–18. [Google Scholar] [CrossRef] [PubMed]
  52. Wang, J.; Yue, B.-L.; Huang, Y.-Z.; Lan, X.-Y.; Liu, W.-J.; Chen, H. Exosomal RNAs: Novel Potential Biomarkers for Diseases—A Review. Int. J. Mol. Sci. 2022, 23, 2461. [Google Scholar] [CrossRef] [PubMed]
  53. Gao, C.; Wang, B.; Chen, Q.; Wang, M.; Fei, X.; Zhao, N. Serum exosomes from diabetic kidney disease patients promote pyroptosis and oxidative stress through the miR-4449/HIC1 pathway. Nutr. Diabet. 2021, 11, 33. [Google Scholar] [CrossRef]
  54. Li, Q.; Li, B.; Wei, S.; He, Z.; Huang, X.; Wang, L.; Xia, Y.; Xu, Z.; Li, Z.; Wang, W.; et al. Exosomal miR-21-5p derived from gastric cancer promotes peritoneal metastasis via mesothelial-to-mesenchymal transition. Cell Death Dis. 2018, 9, 854. [Google Scholar] [CrossRef]
  55. Li, Y.; Yin, Z.; Fan, J.; Zhang, S.; Yang, W. The roles of exosomal miRNAs and lncRNAs in lung diseases. Signal Transduct. Target. Ther. 2019, 4, 47. [Google Scholar] [CrossRef]
  56. Garcia, G.; Pinto, S.; Ferreira, S.; Lopes, D.; Serrador, M.J.; Fernandes, A.; Vaz, A.R.; de Mendonça, A.; Edenhofer, F.; Malm, T.; et al. Emerging Role of miR-21-5p in Neuron–Glia Dysregulation and Exosome Transfer Using Multiple Models of Alzheimer’s Disease. Cells 2022, 11, 3377. [Google Scholar] [CrossRef] [PubMed]
  57. Wang, W.; Hong, G.; Wang, S.; Gao, W.; Wang, P. Tumor-derived exosomal miRNA-141 promote angiogenesis and malignant pro-gression of lung cancer by targeting growth arrest-specific homeobox gene (GAX). Bioengineered 2021, 12, 821–831. [Google Scholar] [CrossRef]
  58. Ren, W.; Zhang, X.; Li, W.; Feng, Q.; Feng, H.; Tong, Y.; Rong, H.; Wang, W.; Zhang, D.; Zhang, Z.; et al. Exosomal miRNA-107 induces myeloid-derived suppressor cell expansion in gastric cancer. Cancer Manag. Res. 2019, 11, 4023–4040. [Google Scholar] [CrossRef]
  59. Mao, S.; Zheng, S.; Lu, Z.; Wang, X.; Wang, Y.; Zhang, G.; Xu, H.; Huang, J.; Lei, Y.; Liu, C.; et al. Exosomal miR-375-3p breaks vascular barrier and promotes small cell lung cancer metastasis by targeting claudin-1. Transl. Lung Cancer Res. 2021, 10, 3155–3172. [Google Scholar] [CrossRef]
  60. Yu, Y.; Bai, F.; Qin, N.; Liu, W.; Sun, Q.; Zhou, Y.; Yang, J. Non-proximal renal tubule-derived urinary exosomal miR-200b as a biomarker of renal fibrosis. Nephron 2018, 139, 269–282. [Google Scholar] [CrossRef]
  61. Lang, H.L.; Hu, G.W.; Chen, Y.; Liu, Y.; Tu, W.; Lu, Y.M.; Wu, L.; Xu, G.H. Glioma cells promote angiogenesis through the release of exosomes containing long non-coding RNA POU3F3. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 959–972. [Google Scholar]
  62. Sun, Y.; Hao, G.; Zhuang, M.; Lv, H.; Liu, C.; Su, K. MEG3 LncRNA from Exosomes Released from Cancer-Associated Fibroblasts Enhances Cisplatin Chemoresistance in SCLC via a MiR-15a-5p/CCNE1 Axis. Yonsei Med. J. 2022, 63, 229–240. [Google Scholar] [CrossRef]
  63. Zhang, P.; Zhou, H.; Lu, K.; Lu, Y.; Wang, Y.; Feng, T. Exosome-mediated delivery of MALAT1 induces cell proliferation in breast cancer. OncoTargets Ther. 2018, 11, 291–299. [Google Scholar] [CrossRef]
  64. Li, Z.; Yanfang, W.U.; Li, J.; Jiang, P.; Peng, T.; Chen, K.; Zhao, X.; Zhang, Y.; Zhen, P.; Zhu, J.; et al. Tumor-released exosomal circular RNA PDE8A promotes invasive growth via the miR-338/MACC1/MET pathway in pancreatic cancer. Cancer Lett. 2018, 432, 237–250. [Google Scholar] [CrossRef] [PubMed]
  65. Dai, X.; Chen, C.; Yang, Q.; Xue, J.; Chen, X.; Sun, B.; Luo, F.; Liu, X.; Xiao, T.; Xu, H.; et al. Exosomal circRNA_100284 from arsenite-transformed cells, via microRNA-217 regulation of EZH2, is involved in the malignant transformation of human hepatic cells by accelerating the cell cycle and promoting cell proliferation. Cell Death Dis. 2018, 9, 454. [Google Scholar] [CrossRef] [PubMed]
  66. Liu, J.; Wu, S.; Zheng, X.; Zheng, P.; Fu, Y.; Wu, C.; Lu, B.; Ju, J.; Jiang, J. Immune suppressed tumor microenvironment by exosomes derived from gastric cancer cells via modulating immune functions. Sci. Rep. 2020, 10, 14749. [Google Scholar] [CrossRef] [PubMed]
  67. Jafari, N.; Kolla, M.; Meshulam, T.; Shafran, J.S.; Qiu, Y.; Casey, A.N.; Pompa, I.R.; Ennis, C.S.; Mazzeo, C.S.; Rabhi, N.; et al. Adipo-cyte-derived exosomes may promote breast cancer progression in type 2 diabetes. Sci. Signal. 2021, 14, eabj2807. [Google Scholar] [CrossRef] [PubMed]
  68. Chen, G.; Huang, A.C.; Zhang, W.; Zhang, G.; Wu, M.; Xu, W.; Yu, Z.; Yang, J.; Wang, B.; Sun, H.; et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 2018, 560, 382–386. [Google Scholar] [CrossRef]
  69. Shi, Y.; Zhang, J.; Mao, Z.; Jiang, H.; Liu, W.; Shi, H.; Ji, R.; Xu, W.; Qian, H.; Zhang, X. Extracellular Vesicles from Gastric Cancer Cells Induce PD-L1 Expression on Neutrophils to Suppress T-Cell Immunity. Front. Oncol. 2020, 10, 629. [Google Scholar] [CrossRef]
  70. Kim, D.H.; Kim, H.; Choi, Y.J.; Kim, S.Y.; Lee, J.-E.; Sung, K.J.; Sung, Y.H.; Pack, C.-G.; Jung, M.-K.; Han, B.; et al. Exosomal PD-L1 promotes tumor growth through immune escape in non-small cell lung cancer. Exp. Mol. Med. 2019, 51, 1–13. [Google Scholar] [CrossRef]
  71. Poggio, M.; Hu, T.; Pai, C.-C.; Chu, B.; Belair, C.D.; Chang, A.; Montabana, E.; Lang, U.E.; Fu, Q.; Fong, L.; et al. Suppression of exosomal PD-L1 induces systemic anti-tumor immunity and memory. Cell 2019, 177, 414–427.e413. [Google Scholar] [CrossRef]
  72. Zhang, S.; Zhang, Y.; Qu, J.; Che, X.; Fan, Y.; Hou, K.; Guo, T.; Deng, G.; Song, N.; Li, C.; et al. Exosomes promote cetuximab resistance via the PTEN/Akt pathway in colon cancer cells. Braz. J. Med. Biol. Res. 2018, 51, e6472. [Google Scholar] [CrossRef] [PubMed]
  73. Du, W.W.; Li, X.; Ma, J.; Fang, L.; Wu, N.; Li, F.; Dhaliwal, P.; Yang, W.; Yee, A.J.; Yang, B.B. Promotion of tumor progression by exosome transmission of circular RNA circSKA3. Mol. Ther. Nucleic Acids 2022, 27, 276–292. [Google Scholar] [CrossRef] [PubMed]
  74. Dai, J.; Escara-Wilke, J.; Keller, J.M.; Jung, Y.; Taichman, R.S.; Pienta, K.J.; Keller, E.T. Primary prostate cancer educates bone stroma through exosomal pyruvate kinase M2 to promote bone metastasis. J. Exp. Med. 2019, 216, 2883–2899. [Google Scholar] [CrossRef]
  75. Yu, C.; Xue, B.; Li, J.; Zhang, Q. Tumor cell-derived exosome RNF126 affects the immune microenvironment and promotes naso-pharyngeal carcinoma progression by regulating PTEN ubiquitination. Apoptosis 2022, 27, 590–605. [Google Scholar] [CrossRef] [PubMed]
  76. Gyukity-Sebestyén, E.; Harmati, M.; Dobra, G.; Németh, I.B.; Mihály, J.; Zvara, Á.; Hunyadi-Gulyás, É.; Katona, R.; Nagy, I.; Horváth, P.; et al. Melanoma-derived exosomes induce PD-1 overexpression and tumor progression via mesenchymal stem cell onco-genic reprogramming. Front. Immunol. 2019, 10, 2459. [Google Scholar] [CrossRef]
  77. Zhao, K.; Wang, Z.; Hackert, T.; Pitzer, C.; Zöller, M. Tspan8 and Tspan8/CD151 knockout mice unravel the contribution of tumor and host exosomes to tumor progression. J. Exp. Clin. Cancer Res. 2018, 37, 312. [Google Scholar] [CrossRef]
  78. Wang, B.; Tan, Z.; Guan, F. Tumor-Derived Exosomes Mediate the Instability of Cadherins and Promote Tumor Progression. Int. J. Mol. Sci. 2019, 20, 3652. [Google Scholar] [CrossRef]
  79. Jeong, K.; Yu, Y.J.; You, J.Y.; Rhee, W.J.; Kim, J.A. Exosome-mediated microRNA-497 delivery for anti-cancer therapy in a microfluidic 3D lung cancer model. Lab Chip 2020, 20, 548–557. [Google Scholar] [CrossRef]
  80. Wu, X.; Jin, S.; Ding, C.; Wang, Y.; He, D.; Liu, Y. Mesenchymal Stem Cell-Derived Exosome Therapy of Microbial Diseases: From Bench to Bed. Front. Microbiol. 2022, 12, 4007. [Google Scholar] [CrossRef]
  81. Kaspi, H.; Semo, J.; Abramov, N.; Dekel, C.; Lindborg, S.; Kern, R.; Lebovits, C.; Aricha, R. MSC-NTF (NurOwn®) exosomes: A novel therapeutic modality in the mouse LPS-induced ARDS model. Stem Cell Res. Ther. 2021, 12, 72. [Google Scholar] [CrossRef]
  82. Wan, T.; Zhong, J.; Pan, Q.; Zhou, T.; Ping, Y.; Liu, X. Exosome-mediated delivery of Cas9 ribonucleoprotein complexes for tis-sue-specific gene therapy of liver diseases. Sci. Adv. 2022, 8, eabp9435. [Google Scholar] [CrossRef]
  83. Melzer, C.; Rehn, V.; Yang, Y.; Bähre, H.; von der Ohe, J.; Hass, R. Taxol-Loaded MSC-Derived Exosomes Provide a Therapeutic Vehicle to Target Metastatic Breast Cancer and Other Carcinoma Cells. Cancers 2019, 11, 798. [Google Scholar] [CrossRef] [PubMed]
  84. Zhuang, X.; Xiang, X.; Grizzle, W.; Sun, D.; Zhang, S.; Axtell, R.C.; Ju, S.; Mu, J.; Zhang, L.; Steinman, L.; et al. Treatment of Brain Inflammatory Diseases by Delivering Exosome Encapsulated Anti-inflammatory Drugs from the Nasal Region to the Brain. Mol. Ther. 2011, 19, 1769–1779. [Google Scholar] [CrossRef] [PubMed]
  85. Wu, H.; Zhou, J.; Zeng, C.; Wu, D.; Mu, Z.; Chen, B.; Xie, Y.; Ye, Y.; Liu, J. Curcumin increases exosomal TCF21 thus suppressing exo-some-induced lung cancer. Oncotarget 2016, 7, 87081. [Google Scholar] [CrossRef]
  86. Toffoli, G.; Hadla, M.; Corona, G.; Caligiuri, I.; Palazzolo, S.; Semeraro, S.; Gamini, A.; Canzonieri, V.; Rizzolio, F. Exosomal doxorubicin reduces the cardiac toxicity of doxorubicin. Nanomedicine 2015, 10, 2963–2971. [Google Scholar] [CrossRef] [PubMed]
  87. Wang, J.; Zheng, Y.; Zhao, M. Exosome-Based Cancer Therapy: Implication for Targeting Cancer Stem Cells. Front. Pharmacol. 2017, 7, 533. [Google Scholar] [CrossRef]
  88. Shen, L.-M.; Quan, L.; Liu, J. Tracking Exosomes in Vitro and in Vivo To Elucidate Their Physiological Functions: Implications for Diagnostic and Therapeutic Nanocarriers. ACS Appl. Nano Mater. 2018, 1, 2438–2448. [Google Scholar] [CrossRef]
  89. Hikita, T.; Miyata, M.; Watanabe, R.; Oneyama, C. In vivo imaging of long-term accumulation of cancer-derived exosomes using a BRET-based reporter. Sci. Rep. 2020, 10, 16616. [Google Scholar] [CrossRef]
  90. Ventimiglia, L.N.; Alonso, M.A. Biogenesis and function of T cell-derived exosomes. Front. Cell Dev. Biol. 2016, 4, 84. [Google Scholar] [CrossRef]
  91. Alonso, R.; Mazzeo, C.; Rodríguez, M.C.; Marsh, M.; Fraile-Ramos, A.; Calvo, V.; Avila-Flores, A.; Merida, I.; Izquierdo, M. Diacylglycerol kinase α regulates the formation and polarisation of mature multivesicular bodies involved in the secretion of Fas lig-and-containing exosomes in T lymphocytes. Cell Death Differ. 2011, 18, 1161–1173. [Google Scholar] [CrossRef] [PubMed]
  92. Gutierrez-Vazquez, C.; Villarroya-Beltri, C.; Mittelbrunn, M.; Sánchez-Madrid, F. Transfer of extracellular vesicles during immune cell-cell interactions. Immunol. Rev. 2012, 251, 125–142. [Google Scholar] [CrossRef] [PubMed]
  93. Clark, D.J.; McMillan, L.E.; Tan, S.L.; Bellomo, G.; Massoue, C.; Thompson, H.; Mykhaylechko, L.; Alibhai, D.; Ruan, X.; Singleton, K.L.; et al. Transient protein accumulation at the center of the T cell antigen-presenting cell interface drives efficient IL-2 secretion. Elife 2019, 8, e45789. [Google Scholar] [CrossRef]
  94. Abusamra, A.J.; Zhong, Z.; Zheng, X.; Li, M.; Ichim, T.E.; Chin, J.L.; Min, W.-P. Tumor exosomes expressing Fas ligand mediate CD8+ T-cell apoptosis. Blood Cells Mol. Dis. 2005, 35, 169–173. [Google Scholar] [CrossRef]
  95. Wahlgren, J.; Karlson, T.D.; Glader, P.; Telemo, E.; Valadi, H. Activated human T cells secrete exosomes that participate in IL-2 mediated immune response signaling. PLoS ONE 2012, 7, e49723. [Google Scholar] [CrossRef] [PubMed]
  96. Smyth, L.A.; Ratnasothy, K.; Tsang, J.Y.; Boardman, D.; Warley, A.; Lechler, R.; Lombardi, G. CD73 expression on extracellular vesicles derived from CD4+ CD25+ Foxp3+ T cells contributes to their regulatory function. Eur. J. Immunol. 2013, 43, 2430–2440. [Google Scholar] [CrossRef]
  97. Okoye, I.S.; Coomes, S.M.; Pelly, V.S.; Czieso, S.; Papayannopoulos, V.; Tolmachova, T.; Seabra, M.C.; Wilson, M.S. MicroRNA-containing T-regulatory-cell-derived exosomes suppress pathogenic T helper 1 cells. Immunity 2014, 41, 89–103. [Google Scholar] [CrossRef]
  98. Lin, C.; Guo, J.; Jia, R. Roles of Regulatory T Cell-Derived Extracellular Vesicles in Human Diseases. Int. J. Mol. Sci. 2022, 23, 11206. [Google Scholar] [CrossRef]
  99. Zhang, W.; Yan, Y.; Peng, J.; Thakur, A.; Bai, N.; Yang, K.; Xu, Z. Decoding Roles of Exosomal lncRNAs in Tumor-Immune Regulation and Therapeutic Potential. Cancers 2022, 15, 286. [Google Scholar] [CrossRef]
  100. Bettelli, E.; Korn, T.; Oukka, M.; Kuchroo, V.K. Induction and effector functions of TH17 cells. Nature 2008, 453, 1051–1057. [Google Scholar] [CrossRef]
  101. Zhu, J.; Yamane, H.; Paul, W.E. Differentiation of Effector CD4 T Cell Populations. Annu. Rev. Immunol. 2010, 28, 445–489. [Google Scholar] [CrossRef] [PubMed]
  102. Curtis, M.M.; Way, S.S. Interleukin-17 in host defence against bacterial, mycobacterial and fungal pathogens. Immunology 2009, 126, 177–185. [Google Scholar] [CrossRef] [PubMed]
  103. Bhattacharyya, N.; Feng, C.G. Regulation of T Helper Cell Fate by TCR Signal Strength. Front. Immunol. 2020, 11, 624. [Google Scholar] [CrossRef] [PubMed]
  104. Reppert, S.; Zinser, E.; Holzinger, C.; Sandrock, L.; Koch, S.; Finotto, S. NFATc1 deficiency in T cells protects mice from experimental autoimmune encephalomyelitis. Eur. J. Immunol. 2015, 45, 1426–1440. [Google Scholar] [CrossRef]
  105. Bhaumik, S.; Basu, R. Cellular and Molecular Dynamics of Th17 Differentiation and its Developmental Plasticity in the Intestinal Immune Response. Front. Immunol. 2017, 8, 254. [Google Scholar] [CrossRef]
  106. Revu, S.; Wu, J.; Henkel, M.; Rittenhouse, N.; Menk, A.; Delgoffe, G.M.; Poholek, A.C.; McGeachy, M.J. IL-23 and IL-1β Drive Human Th17 Cell Differentiation and Metabolic Reprogramming in Absence of CD28 Costimulation. Cell Rep. 2018, 22, 2642–2653. [Google Scholar] [CrossRef]
  107. Foley, J.F. STAT3 Regulates the Generation of Th17 Cells. Sci. STKE 2007, 2007, tw113. [Google Scholar] [CrossRef]
  108. Baricza, E.; Tamási, V.; Marton, N.; Buzás, E.I.; Nagy, G. The emerging role of aryl hydrocarbon receptor in the activation and differentiation of Th17 cells. Cell Mol. Life Sci. 2015, 73, 95–117. [Google Scholar] [CrossRef]
  109. Mudter, J.; Yu, J.; Zufferey, C.; Brüstle, A.; Wirtz, S.; Weigmann, B.; Hoffman, A.; Schenk, M.; Galle, P.R.; Lehr, H.A.; et al. IRF4 reg-ulates IL-17A promoter activity and controls RORγt-dependent Th17 colitis in vivo. Inflamm. Bowel Dis. 2011, 17, 1343–1358. [Google Scholar] [CrossRef]
  110. Ciofani, M.; Madar, A.; Galan, C.; Sellars, M.; Mace, K.; Pauli, F.; Agarwal, A.; Huang, W.; Parkurst, C.N.; Muratet, M.; et al. A Validated Regulatory Network for Th17 Cell Specification. Cell 2012, 151, 289–303. [Google Scholar] [CrossRef]
  111. Lin, J.; Tang, J.; Lin, J.; He, Y.; Yu, Z.; Jiang, R.; Yang, B.; Ou, Q. YY1 regulation by miR-124-3p promotes Th17 cell pathogenicity through interaction with T-bet in rheumatoid arthritis. J. Clin. Investig. 2021, 6, e149985. [Google Scholar] [CrossRef] [PubMed]
  112. Qian, Y.; Arellano, G.; Ifergan, I.; Lin, J.; Snowden, C.; Kim, T.; Thomas, J.J.; Law, C.; Guan, T.; Balabanov, R.D.; et al. ZEB1 promotes pathogenic Th1 and Th17 cell differentiation in multiple sclerosis. Cell Rep. 2021, 36, 109602. [Google Scholar] [CrossRef] [PubMed]
  113. Chen, J.; Martindale, J.L.; Abdelmohsen, K.; Kumar, G.; Fortina, P.M.; Gorospe, M.; Rostami, A.; Yu, S. RNA-binding protein HuR pro-motes Th17 cell differentiation and can be targeted to reduce autoimmune neuroinflammation. J. Immunol. 2020, 204, 2076–2087. [Google Scholar] [CrossRef] [PubMed]
  114. Bechara, R.; Amatya, N.; Majumder, S.; Zhou, C.; Li, Y.; Liu, Q.; McGeachy, M.J.; Gaffen, S.L. The RNA-binding protein IMP2 drives a stromal-Th17 cell circuit in autoimmune neuroinflammation. J. Clin. Investig. 2022, 7, e152766. [Google Scholar] [CrossRef]
  115. Avdeev, S.N.; Trushenko, N.V.; Tsareva, N.A.; Yaroshetskiy, A.I.; Merzhoeva, Z.M.; Nuralieva, G.S.; Nekludova, G.V.; Chikina, S.Y.; Gneusheva, T.Y.; Suvorova, O.A.; et al. Anti-IL-17 monoclonal antibodies in hospitalized patients with severe COVID-19: A pilot study. Cytokine 2021, 146, 155627. [Google Scholar] [CrossRef]
  116. Omokanye, A.; Ong, L.C.; Lebrero-Fernandez, C.; Bernasconi, V.; Schön, K.; Strömberg, A.; Bemark, M.; Saelens, X.; Czarnewski, P.; Lycke, N. Clonotypic analysis of protective influenza M2e-specific lung resident Th17 memory cells reveals extensive functional diversity. Mucosal Immunol. 2022, 15, 717–729. [Google Scholar] [CrossRef]
  117. Tan, Z.; Qian, X.; Jiang, R.; Liu, Q.; Wang, Y.; Chen, C.; Wang, X.; Ryffel, B.; Sun, B. IL-17A Plays a Critical Role in the Pathogenesis of Liver Fibrosis through Hepatic Stellate Cell Activation. J. Immunol. 2013, 191, 1835–1844. [Google Scholar] [CrossRef]
  118. Molesworth-Kenyon, S.J.; Yin, R.; Oakes, J.E.; Lausch, R.N. IL-17 receptor signaling influences virus-induced corneal inflammation. J. Leukoc. Biol. 2007, 83, 401–408. [Google Scholar] [CrossRef]
  119. Yuan, J.; Yu, M.; Lin, Q.W.; Cao, A.L.; Yu, X.; Dong, J.H.; Wang, J.P.; Zhang, J.H.; Wang, M.; Guo, H.P.; et al. Th17 cells contribute to viral replication in coxsackievirus B3-induced acute viral myocarditis. J. Immunol. 2010, 185, 4004–4010. [Google Scholar] [CrossRef]
  120. Li, B.; Huang, L.; Lv, P.; Li, X.; Liu, G.; Chen, Y.; Wang, Z.; Qian, X.; Shen, Y.; Li, Y.; et al. The role of Th17 cells in psoriasis. Immunol. Res. 2020, 68, 296–309. [Google Scholar] [CrossRef]
  121. Nishimoto, S.; Kotani, H.; Tsuruta, S.; Shimizu, N.; Ito, M.; Shichita, T.; Morita, R.; Takahashi, H.; Amagai, M.; Yoshimura, A. Th17 Cells Carrying TCR Recognizing Epidermal Autoantigen Induce Psoriasis-like Skin Inflammation. J. Immunol. 2013, 191, 3065–3072. [Google Scholar] [CrossRef] [PubMed]
  122. Furue, M.; Kadono, T. The contribution of IL-17 to the development of autoimmunity in psoriasis. J. Endotoxin Res. 2019, 25, 337–343. [Google Scholar] [CrossRef]
  123. Zhao, L.; Ghetie, D.; Jiang, Z.; Chu, C.Q. How Can We Manipulate the IL-23/IL-17 Axis? Curr. Treat. Options Rheumatol. 2015, 1, 182–196. [Google Scholar] [CrossRef]
  124. Balasa, R.; Barcutean, L.; Balasa, A.; Motataianu, A.; Roman-Filip, C.; Manu, D. The action of TH17 cells on blood brain barrier in multiple sclerosis and experimental autoimmune encephalomyelitis. Hum. Immunol. 2020, 81, 237–243. [Google Scholar] [CrossRef]
  125. Hu, W. Huntington’s disease is a TH17 related autoimmune disorder against mutant Huntingtin coded by multiple CAG triplets. Nat. Précéd. 2011, 219. [Google Scholar] [CrossRef]
  126. Da Silva, R.R.; Pereira, M.C.; Rêgo, M.J.; Hatzlhofer, B.L.; da Silva Araújo, A.; Bezerra, M.A.; da Rocha Pitta, I.; da Rocha Pitta, M.G. Evaluation of Th17 related cytokines associated with clinical and laboratorial parameters in sickle cell anemia patients with leg ulcers. Cytokine 2014, 65, 143–147. [Google Scholar] [CrossRef] [PubMed]
  127. Soleimani, A.; Adabavazeh, R.; Nikoueinejad, H.; Sharif, M.R.; Faraji, S.; Shahreza, B.O.; Akbari, H.; Einollahi, B. T helper 17 lymphocyte pathway in the diagnosis of autosomal dominant polycystic kidney disease. Iran. J. Kidney Dis. 2015, 9, 105–112. [Google Scholar] [PubMed]
  128. Zhang, M.; Yu, Q.; Tang, W.; Wu, Y.; Lv, J.; Sun, L.; Shi, G.; Wu, M.; Qu, J.; Di, C.; et al. Epithelial exosomal contactin-1 promotes monocyte-derived dendritic cell-dominant T-cell responses in asthma. J. Allergy Clin. Immunol. 2021, 148, 1458–1545. [Google Scholar] [CrossRef] [PubMed]
  129. Sun, J.; Jia, H.; Bao, X.; Wu, Y.; Zhu, T.; Li, R.; Zhao, H. Tumor exosome promotes Th17 cell differentiation by transmitting the lncRNA CRNDE-h in colorectal cancer. Cell Death Dis. 2021, 12, 123. [Google Scholar] [CrossRef] [PubMed]
  130. Li, Q.; Li, J.; Sun, L.; Sun, Y.; Zhao, F.; Liu, P.; Peng, X.; Xuan, X.; Li, Y.; Wang, P.; et al. Exosomes derived from LPS-stimulated human thymic mesenchymal stromal cells enhance inflammation via thrombospondin-1. Biosci. Rep. 2021, 41, BSR20203573. [Google Scholar] [CrossRef]
  131. Pourakbari, R.; Parhizkar, F.; Soltani-Zangbar, M.S.; Samadi, P.; Zamani, M.; Aghebati-Maleki, L.; Motavalli, R.; Mahmoodpoor, A.; Jadidi-Niaragh, F.; Yousefi, B.; et al. Preeclampsia-Derived Exosomes Imbalance the Activity of Th17 and Treg in PBMCs from Healthy Pregnant Women. Reprod. Sci. 2022, 1–2. [Google Scholar] [CrossRef]
  132. Liu, F.; Bu, Z.; Zhao, F.; Xiao, D. Increased T-helper 17 cell differentiation mediated by exosome-mediated micro RNA-451 redistribution in gastric cancer infiltrated T cells. Cancer Sci. 2018, 109, 65–73. [Google Scholar] [CrossRef] [PubMed]
  133. Zhu, D.; Tian, J.; Wu, X.; Li, M.; Tang, X.; Rui, K.; Guo, H.; Ma, J.; Xu, H.; Wang, S. G-MDSC-derived exosomes attenuate collagen-induced arthritis by impairing Th1 and Th17 cell responses. Biochim. Biophys. Acta Mol.-Basis Dis. 2019, 1865, 165540. [Google Scholar] [CrossRef] [PubMed]
  134. Xia, Y.; Cheng, T.; Zhou, M.; Kang, F.; Liao, C. hBMSC-Derived Exosomes Mitigate Th17/Treg Homeostasis in Periodontitis via miR-1246; Research Square: Durham, NC, USA, 2022. [Google Scholar]
  135. Yang, R.; Huang, H.; Cui, S.; Zhou, Y.; Zhang, T.; Zhou, Y. IFN-γ promoted exosomes from mesenchymal stem cells to attenuate colitis via miR-125a and miR-125b. Cell Death Dis. 2020, 11, 603. [Google Scholar] [CrossRef] [PubMed]
  136. Zhang, Y.; Chen, J.; Fu, H.; Kuang, S.; He, F.; Zhang, M.; Shen, Z.; Qin, W.; Lin, Z.; Huang, S. Exosomes derived from 3D-cultured MSCs improve therapeutic effects in periodontitis and experimental colitis and restore the Th17 cell/Treg balance in inflamed periodontium. Int. J. Oral Sci. 2021, 13, 43. [Google Scholar] [CrossRef] [PubMed]
  137. Tian, J.; Zhu, Q.; Zhang, Y.; Bian, Q.; Hong, Y.; Shen, Z.; Xu, H.; Rui, K.; Yin, K.; Wang, S. Olfactory ecto-mesenchymal stem cell-derived exosomes ameliorate experimental colitis via modulating Th1/Th17 and Treg cell responses. Front. Immunol. 2020, 11, 598322. [Google Scholar] [CrossRef]
  138. Mao, F.; Wu, Y.; Tang, X.; Kang, J.; Zhang, B.; Yan, Y.; Qian, H.; Zhang, X.; Xu, W. Exosomes derived from human umbilical cord mesenchymal stem cells relieve inflammatory bowel disease in mice. BioMed Res. Int. 2017, 2017, 5356760. [Google Scholar] [CrossRef]
  139. Tu, J.; Zheng, N.; Mao, C.; Liu, S.; Zhang, H.; Sun, L. UC-BSCs Exosomes Regulate Th17/Treg Balance in Patients with Systemic Lupus Erythematosus via miR-19b/KLF13. Cells 2022, 11, 4123. [Google Scholar] [CrossRef]
  140. Li, B.; Xing, Y.; Gan, Y.; He, J.; Hua, H. Labial gland-derived mesenchymal stem cells and their exosomes ameliorate murine Sjögren’s syndrome by modulating the balance of Treg and Th17 cells. Stem Cell Res. Ther. 2021, 12, 478. [Google Scholar] [CrossRef]
  141. Zheng, L.; Li, Z.; Ling, W.; Zhu, D.; Feng, Z.; Kong, L. Exosomes Derived from Dendritic Cells Attenuate Liver Injury by Modulating the Balance of Treg and Th17 Cells After Ischemia Reperfusion. Cell Physiol. Biochem. 2018, 46, 740–756. [Google Scholar] [CrossRef]
  142. Shi, Q.-Z.; Yu, H.-M.; Chen, H.-M.; Liu, M.; Cheng, X. Exosomes derived from mesenchymal stem cells regulate Treg/Th17 balance in aplastic anemia by transferring miR-23a-3p. Clin. Exp. Med. 2021, 21, 429–437. [Google Scholar] [CrossRef]
  143. Jia, Z.; Liu, J.; Li, B.; Yi, L.; Wu, Y.; Xing, J.; Wang, L.; Wang, J.; Guo, L. Exosomes with FOXP3 from gene-modified dendritic cells ameliorate the development of EAE by regulating the balance of Th/Treg. Int. J. Med. Sci. 2022, 19, 1265–1274. [Google Scholar] [CrossRef] [PubMed]
  144. Cai, Z.; Zhang, W.; Yang, F.; Yu, L.; Yu, Z.; Pan, J.; Wang, L.; Cao, X.; Wang, J. Immunosuppressive exosomes from TGF-β1 gene-modified dendritic cells attenuate Th17-mediated inflammatory autoimmune disease by inducing regulatory T cells. Cell Res. 2011, 22, 607–610. [Google Scholar] [CrossRef] [PubMed]
  145. Guo, D.; Chen, Y.; Wang, S.; Yu, L.; Shen, Y.; Zhong, H.; Yang, Y. Exosomes from heat-stressed tumour cells inhibit tumour growth by converting regulatory T cells to Th17 cells via IL-6. Immunology 2018, 154, 132–143. [Google Scholar] [CrossRef]
  146. Wang, H.; Li, Y.; Yue, Z.; Liu, Y.; Chen, Q.; Hu, D.; Han, J. Adipose-Derived Stem Cell Exosomes Inhibit Hypertrophic Scaring Formation by Regulating Th17/Treg Cell Balance. BioMed Res. Int. 2022, 2022, 9899135. [Google Scholar] [CrossRef] [PubMed]
  147. Xi, X.M.; Xia, S.J.; Lu, R. Drug loading techniques for exosome-based drug delivery systems. Pharm. Int. J. Pharm. Sci. 2021, 76, 61–67. [Google Scholar]
  148. Sterzenbach, U.; Putz, U.; Low, L.H.; Silke, J.; Tan, S.S.; Howitt, J. Engineered exosomes as vehicles for biologically active proteins. Mol. Ther. 2017, 25, 1269–1278. [Google Scholar] [CrossRef] [PubMed]
  149. Nakase, I.; Noguchi, K.; Aoki, A.; Takatani-Nakase, T.; Fujii, I.; Futaki, S. Arginine-rich cell-penetrating peptide-modified extracellular vesicles for active macropinocytosis induction and efficient intracellular delivery. Sci. Rep. 2017, 7, 1991. [Google Scholar] [CrossRef]
  150. Shimaoka, M.; Kawamoto, E.; Gaowa, A.; Okamoto, T.; Park, E. Connexins and Integrins in Exosomes. Cancers 2019, 11, 106. [Google Scholar] [CrossRef]
  151. Sercombe, L.; Veerati, T.; Moheimani, F.; Wu, S.Y.; Sood, A.K.; Hua, S. Advances and Challenges of Liposome Assisted Drug Delivery. Front. Pharmacol. 2015, 6, 286. [Google Scholar] [CrossRef]
  152. Elashiry, M.; Elsayed, R.; Cutler, C.W. Exogenous and endogenous dendritic cell-derived exosomes: Lessons learned for immu-notherapy and disease pathogenesis. Cells 2021, 11, 115. [Google Scholar] [CrossRef] [PubMed]
  153. Wei, W.; Ao, Q.; Wang, X.; Cao, Y.; Liu, Y.; Zheng, S.G.; Tian, X. Mesenchymal Stem Cell–Derived Exosomes: A Promising Biological Tool in Nanomedicine. Front. Pharmacol. 2021, 11, 590470. [Google Scholar] [CrossRef] [PubMed]
  154. Malla, R.R.; Pandrangi, S.; Kumari, S.; Gavara, M.M.; Badana, A.K. Exosomal tetraspanins as regulators of cancer progression and metastasis and novel diagnostic markers. Asia-Pacific J. Clin. Oncol. 2018, 14, 383–391. [Google Scholar] [CrossRef]
  155. Revenfeld, A.L.S.; Bæk, R.; Nielsen, M.H.; Stensballe, A.; Varming, K.; Jørgensen, M. Diagnostic and Prognostic Potential of Extracellular Vesicles in Peripheral Blood. Clin. Ther. 2014, 36, 830–846. [Google Scholar] [CrossRef] [PubMed]
  156. Huh, J.R.; Littman, D.R. Small molecule inhibitors of ROR γt: Targeting T h17 cells and other applications. Eur. J. Immunol. 2012, 42, 2232–2237. [Google Scholar] [CrossRef] [PubMed]
  157. Xue, X.; De Leon-Tabaldo, A.; Luna-Roman, R.; Castro, G.; Albers, M.; Schoetens, F.; DePrimo, S.; Devineni, D.; Wilde, T.; Goldberg, S.; et al. Preclinical and clinical characterization of the RORγt inhibitor JNJ-61803534. Sci. Rep. 2021, 11, 11066. [Google Scholar] [CrossRef] [PubMed]
  158. Cherney, R.J.; Cornelius, L.A.M.; Srivastava, A.; Weigelt, C.A.; Marcoux, D.; Duan, J.J.-W.; Shi, Q.; Batt, D.G.; Liu, Q.; Yip, S.; et al. Discovery of BMS-986251: A Clinically Viable, Potent, and Selective RORγt Inverse Agonist. ACS Med. Chem. Lett. 2020, 11, 1221–1227. [Google Scholar] [CrossRef]
  159. Cheung, K.; Lu, G.; Sharma, R.; Vincek, A.; Zhang, R.; Plotnikov, A.N.; Zhang, F.; Zhang, Q.; Ju, Y.; Hu, Y.; et al. BET N-terminal bromodomain inhibition selectively blocks Th17 cell differentiation and ameliorates colitis in mice. Proc. Nat. Acad. Sci. USA 2017, 114, 2952–2957. [Google Scholar] [CrossRef] [PubMed]
  160. Zou, Y.; Hu, X.; Schewitz-Bowers, L.P.; Stimpson, M.; Miao, L.; Ge, X.; Yang, L.; Li, Y.; Bible, P.W.; Wen, X.; et al. The DNA Methylation Inhibitor Zebularine Controls CD4+ T Cell Mediated Intraocular Inflammation. Front. Immunol. 2019, 10, 1950. [Google Scholar] [CrossRef]
  161. Hu, X.; Schewitz-Bowers, L.P.; Lait, P.J.P.; Copland, D.A.; Stimpson, M.L.; Li, J.J.; Liu, Y.; Dick, A.D.; Lee, R.W.J.; Wei, L. The Bromodomain and Extra-Terminal Protein Inhibitor OTX015 Suppresses T Helper Cell Proliferation and Differentiation. Curr. Mol. Med. 2019, 18, 594–601. [Google Scholar] [CrossRef]
  162. Aqel, S.I.; Kraus, E.E.; Jena, N.; Kumari, V.; Granitto, M.C.; Mao, L.; Yang, Y. Novel small molecule IL-6 inhibitor suppresses autoreactive Th17 development and promotes Treg development. Clin. Exp. Immunol. 2019, 196, 215–225. [Google Scholar] [CrossRef] [PubMed]
  163. Bissonnette, R.; Gold, L.S.; Rubenstein, D.S.; Tallman, A.M.; Armstrong, A. Tapinarof in the treatment of psoriasis: A review of the unique mechanism of action of a novel therapeutic aryl hydrocarbon receptor–modulating agent. J. Am. Acad. Dermatol. 2020, 84, 1059–1067. [Google Scholar] [CrossRef] [PubMed]
  164. Park, Y.H.; Kim, H.J.; Heo, T.H. A directly GP130-targeting small molecule ameliorates collagen-induced arthritis (CIA) by inhibiting IL-6/GP130 signalling and Th17 differentiation. Clin. Exp. Pharmacol. Physiol. 2020, 47, 628–639. [Google Scholar] [CrossRef] [PubMed]
  165. Chen, J.Y.; Yang, Y.J.; Ma, X.Q.; Cao, Q.; Wei, S.S.; Pan, R.R.; Nan, L.H.; Liu, Y.J.; Cao, Y.; Tian, X.Y.; et al. Neobaicalein Inhibits Th17 Cell Differentiation Resulting in Recovery of Th17/Treg Ratio through Blocking STAT3 Signaling Activation. Molecules 2022, 28, 18. [Google Scholar] [CrossRef]
  166. Hong, S.S.; Choi, J.H.; Lee, S.Y.; Park, Y.H.; Park, K.Y.; Lee, J.Y.; Kim, J.; Gajulapati, V.; Goo, J.I.; Singh, S.; et al. A novel small-molecule inhibitor targeting the IL-6 receptor β subunit, glycoprotein 130. J. Immunol. 2015, 195, 237–245. [Google Scholar] [CrossRef]
  167. Xiao, S.; Yosef, N.; Yang, J.; Wang, Y.; Zhou, L.; Zhu, C.; Wu, C.; Baloglu, E.; Schmidt, D.; Ramesh, R.; et al. Small-Molecule RORγt Antagonists Inhibit T Helper 17 Cell Transcriptional Network by Divergent Mechanisms. Immunity 2014, 40, 477–489. [Google Scholar] [CrossRef]
  168. Hu, X.; Zou, Y.; Copland, D.A.; Schewitz-Bowers, L.P.; Li, Y.; Lait, P.J.; Stimpson, M.; Zhang, Z.; Guo, S.; Liang, J.; et al. Epigenetic drug screen identified IOX1 as an inhibitor of Th17-mediated inflammation through targeting TET2. Ebiomedicine 2022, 86, 104333. [Google Scholar] [CrossRef]
  169. He, J.; Ren, W.; Wang, W.; Han, W.; Jiang, L.; Zhang, D.; Guo, M. Exosomal targeting and its potential clinical application. Drug Deliv. Transl. Res. 2022, 12, 2385–2402. [Google Scholar] [CrossRef]
  170. Kang, H.-K.; Chiang, M.-Y.; Liu, M.; Ecklund, D.; Datta, S.K. The Histone Peptide H471–94 Alone Is More Effective than a Cocktail of Peptide Epitopes in Controlling Lupus: Immunoregulatory Mechanisms. J. Clin. Immunol. 2011, 31, 379–394. [Google Scholar] [CrossRef]
  171. Niu, T.; Cheng, L.; Wang, H.; Zhu, S.; Yang, X.; Liu, K.; Jin, H.; Xu, X. KS23, a novel peptide derived from adiponectin, inhibits retinal inflammation and downregulates the proportions of Th1 and Th17 cells during experimental autoimmune uveitis. J. Neuroinflamm. 2019, 16, 278. [Google Scholar] [CrossRef]
  172. Zhang, Y.; Liang, R.; Xie, A.; Shi, W.; Huang, H.; Zhong, Y. Antagonistic peptides that specifically bind to the first and second ex-tracellular loops of CCR5 and anti-IL-23p19 antibody reduce airway inflammation by suppressing the IL-23/Th17 signaling pathway. Med. Inflamm. 2020, 2020, 1719467. [Google Scholar] [CrossRef]
  173. Min, H.K.; Choi, J.; Lee, S.Y.; Lee, A.R.; Min, B.M.; Cho, M.L.; Park, S.H. Vitronectin-derived bioactive peptide prevents spondyloarthritis by modulating Th17/Treg imbalance in mice with curdlan-induced spondyloarthritis. PLoS ONE 2022, 17, e0262183. [Google Scholar] [CrossRef] [PubMed]
  174. Park, Y.J.; Park, B.; Lee, M.; Jeong, Y.S.; Lee, H.Y.; Sohn, D.H.; Song, J.J.; Lee, J.H.; Hwang, J.S.; Bae, Y.-S. A novel antimicrobial peptide acting via formyl peptide receptor 2 shows therapeutic effects against rheumatoid arthritis. Sci. Rep. 2018, 8, 14664. [Google Scholar] [CrossRef]
  175. Franke, M.; Schröder, J.; Monhasery, N.; Ackfeld, T.; Hummel, T.M.; Rabe, B.; Garbers, C.; Becker-Pauly, C.; Floss, D.M.; Scheller, J. Human and Murine Interleukin 23 Receptors Are Novel Substrates for A Disintegrin and Metalloproteases ADAM10 and ADAM17. J. Biol. Chem. 2016, 291, 10551–10561. [Google Scholar] [CrossRef] [PubMed]
  176. Machado, M.V. New Developments in Celiac Disease Treatment. Int. J. Mol. Sci. 2023, 24, 945. [Google Scholar] [CrossRef] [PubMed]
  177. Wang, L.; Qiu, R.; Zhang, Z.; Han, Z.; Yao, C.; Hou, G.; Dai, D.; Jin, W.; Tang, Y.; Yu, X.; et al. The MicroRNA miR-22 Represses Th17 Cell Pathogenicity by Targeting PTEN-Regulated Pathways. Immunohorizons 2020, 4, 308–318. [Google Scholar] [CrossRef] [PubMed]
  178. Guan, Y.; Ma, Y.; Tang, Y.; Liu, X.; Zhao, Y.; An, L. MiRNA-221-5p suppressed the Th17/Treg ratio in asthma via RORγt/Foxp3 by targeting SOCS1. Allergy Asthma Clin. Immunol. 2021, 17, 123. [Google Scholar] [CrossRef] [PubMed]
  179. Bian, J.; Liu, R.; Fan, T.; Liao, L.; Wang, S.; Geng, W.; Wang, T.; Shi, W.; Ruan, Q. miR-340 Alleviates Psoriasis in Mice through Direct Targeting of IL-17A. J. Immunol. 2018, 201, 1412–1420. [Google Scholar] [CrossRef]
  180. Raveney, B.J.E.; Oki, S.; Yamamura, T. Nuclear Receptor NR4A2 Orchestrates Th17 Cell-Mediated Autoimmune Inflammation via IL-21 Signalling. PLoS ONE 2013, 8, e56595. [Google Scholar] [CrossRef]
  181. Søndergaard, J.N.; Geng, K.; Sommerauer, C.; Atanasoai, I.; Yin, X.; Kutter, C. Successful delivery of large-size CRISPR/Cas9 vectors in hard-to-transfect human cells using small plasmids. Commun. Biol. 2020, 3, 319. [Google Scholar] [CrossRef]
  182. Duan, L.; Ouyang, K.; Wang, J.; Xu, L.; Xu, X.; Wen, C.; Xie, Y.; Liang, Y.; Xia, J. Exosomes as Targeted Delivery Platform of CRISPR/Cas9 for Therapeutic Genome Editing. Chembiochem 2021, 22, 3360–3368. [Google Scholar] [CrossRef] [PubMed]
  183. Wu, L.; Hollinshead, K.E.; Hao, Y.; Au, C.; Kroehling, L.; Ng, C.; Lin, W.-Y.; Li, D.; Silva, H.M.; Shin, J.; et al. Niche-Selective Inhibition of Pathogenic Th17 Cells by Targeting Metabolic Redundancy. Cell 2020, 182, 641–654.e20. [Google Scholar] [CrossRef] [PubMed]
  184. Li, Z.; Zhou, X.; Wei, M.; Gao, X.; Zhao, L.; Shi, R.; Sun, W.; Duan, Y.; Yang, G.; Yuan, L. In Vitro and in Vivo RNA Inhibition by CD9-HuR Functionalized Exosomes Encapsulated with miRNA or CRISPR/dCas9. Nano Lett. 2018, 19, 19–28. [Google Scholar] [CrossRef] [PubMed]
  185. Smyth, T.; Petrova, K.; Payton, N.M.; Persaud, I.; Redzic, J.S.; Graner, M.W.; Smith-Jones, P.; Anchordoquy, T.J. Surface Functionalization of Exosomes Using Click Chemistry. Bioconj. Chem. 2014, 25, 1777–1784. [Google Scholar] [CrossRef] [PubMed]
  186. Song, H.; Chen, X.; Hao, Y.; Wang, J.; Xie, Q.; Wang, X. Nanoengineering facilitating the target mission: Targeted extracellular vesicles delivery systems design. J. Nanobiotechnol. 2022, 20, 1–23. [Google Scholar] [CrossRef]
  187. Shi, X.; Song, P.; Tao, S.; Zhang, X.; Chu, C.Q. silencing RoRγt in Human CD4+ T cells with CD30 aptamer-RORγt shRNA Chimera. Sci. Rep. 2019, 9, 10375. [Google Scholar] [CrossRef]
  188. Yang, Y.; Hong, Y.; Cho, E.; Kim, G.B.; Kim, I.-S. Extracellular vesicles as a platform for membrane-associated therapeutic protein delivery. J. Extracell. Vesicles 2018, 7, 1440131. [Google Scholar] [CrossRef]
  189. Choi, H.; Choi, Y.; Yim, H.Y.; Mirzaaghasi, A.; Yoo, J.-K.; Choi, C. Biodistribution of Exosomes and Engineering Strategies for Targeted Delivery of Therapeutic Exosomes. Tissue Eng. Regen. Med. 2021, 18, 499–511. [Google Scholar] [CrossRef]
Figure 1. Detailed schematic overview of exosome biogenesis and inhibitors that block the release of exosomes within the endosomal system. The formation of exosomes involves the inward budding and fusion of the limiting membrane of endocytic vesicles that engender intraluminal vesicles (ILVs). The maturation process of ILVs can be through endosomal complexes required for transport (ESCRT)-dependent and -independent mechanisms that process cargo sorting and formation of multivesicular bodies (MVBs). Components of MVBs can then be integrated into the membranes for release into the extracellular space or may be guided for lysosomal degradation. Exosomal contents released into the extracellular space can then be internalized by the target cell via membrane fusion, ligand-receptor conformation or endocytosis mechanism. This illustration further comprehends various chemical inhibitors, including SMase inhibitors, exosome release inhibitors and Rab inhibitors (depicted in black rectangular boxes) that prevent exosome biogenesis and secretion within the endosomal compartment. The sequential steps on exosome biogenesis follows: 1. Exosome formation; 2. Cargo sorting; 3. MVBs formation; 4. Exosome release; 5. Exosome-target cell interaction; 6. Cargo release into cytosol of target cells; 7. MVBs endosome recycling; and 8. Lysosome degradation of exosomes. Abbreviations: Rab; Ras associated binding protein, SNARE; SNAP receptor.
Figure 1. Detailed schematic overview of exosome biogenesis and inhibitors that block the release of exosomes within the endosomal system. The formation of exosomes involves the inward budding and fusion of the limiting membrane of endocytic vesicles that engender intraluminal vesicles (ILVs). The maturation process of ILVs can be through endosomal complexes required for transport (ESCRT)-dependent and -independent mechanisms that process cargo sorting and formation of multivesicular bodies (MVBs). Components of MVBs can then be integrated into the membranes for release into the extracellular space or may be guided for lysosomal degradation. Exosomal contents released into the extracellular space can then be internalized by the target cell via membrane fusion, ligand-receptor conformation or endocytosis mechanism. This illustration further comprehends various chemical inhibitors, including SMase inhibitors, exosome release inhibitors and Rab inhibitors (depicted in black rectangular boxes) that prevent exosome biogenesis and secretion within the endosomal compartment. The sequential steps on exosome biogenesis follows: 1. Exosome formation; 2. Cargo sorting; 3. MVBs formation; 4. Exosome release; 5. Exosome-target cell interaction; 6. Cargo release into cytosol of target cells; 7. MVBs endosome recycling; and 8. Lysosome degradation of exosomes. Abbreviations: Rab; Ras associated binding protein, SNARE; SNAP receptor.
Ijms 24 07647 g001
Figure 2. Pictorial representation of exosomal targets that participate during the T helper (Th)17 cell developmental stages and in promoting its effector function. Early activation of antigen-presenting cells (APCs) mounts an immune response that releases Th17 cell polarizing cytokines, such as IL-6, IL-23, IL-12, etc. These cytokines interact with the receptor on CD4+ T cells and induce its polarization to the Th17 cell phenotype via activation of intermediate kinases and transcription factors. Differentiated Th17 cells then migrate via the bloodstream to inflammatory sites and execute effector function. Furthermore, the diagram highlights multi-targets, including cytokines or its receptor, transcriptional factors or RNA-binding proteins that can be modulated via exosomal anti-Th17 therapies. Abbreviations: PAMPs; pathogen-associated molecular pattern molecules, STAT-3; signal transducer and activator of transcription 3, ZEB1; zinc finger E-box binding homeobox 1, AhR; aryl hydrocarbon receptor, YY1; yin yang 1, IMP2; IGF2 mRNA binding protein 2, TGF-β; transforming growth factor β, TCR; T cell receptor, CCL2; chemokine (C-C motif) ligand 2, REV-ERBα; nuclear receptor subfamily 1group D, ROR-γT; retinoic acid-related orphan receptor gamma t, HuR; human antigen R.
Figure 2. Pictorial representation of exosomal targets that participate during the T helper (Th)17 cell developmental stages and in promoting its effector function. Early activation of antigen-presenting cells (APCs) mounts an immune response that releases Th17 cell polarizing cytokines, such as IL-6, IL-23, IL-12, etc. These cytokines interact with the receptor on CD4+ T cells and induce its polarization to the Th17 cell phenotype via activation of intermediate kinases and transcription factors. Differentiated Th17 cells then migrate via the bloodstream to inflammatory sites and execute effector function. Furthermore, the diagram highlights multi-targets, including cytokines or its receptor, transcriptional factors or RNA-binding proteins that can be modulated via exosomal anti-Th17 therapies. Abbreviations: PAMPs; pathogen-associated molecular pattern molecules, STAT-3; signal transducer and activator of transcription 3, ZEB1; zinc finger E-box binding homeobox 1, AhR; aryl hydrocarbon receptor, YY1; yin yang 1, IMP2; IGF2 mRNA binding protein 2, TGF-β; transforming growth factor β, TCR; T cell receptor, CCL2; chemokine (C-C motif) ligand 2, REV-ERBα; nuclear receptor subfamily 1group D, ROR-γT; retinoic acid-related orphan receptor gamma t, HuR; human antigen R.
Ijms 24 07647 g002
Figure 3. Diagrammatic illustration of exosome engineering for loading molecules into the lumen or sorting them on their surface for specific targeting of Th17 cells. Exosomes are engineered to express CD30 or CD4 aptamer, peptide targeting αvβ3 and Vi polysaccharide (targets prohibitins) on their surface using different sorting modules that result in a sustained response against Th17 cells. Furthermore, the packaging substances including drugs, nucleic acid molecules, etc., can be loaded onto the engineered exosomes for targeting Th17 cells to inhibit its pathogenic mechanism in diseases. Abbreviations: PROCR; protein C receptor, MHC; major histocompatibility complex.
Figure 3. Diagrammatic illustration of exosome engineering for loading molecules into the lumen or sorting them on their surface for specific targeting of Th17 cells. Exosomes are engineered to express CD30 or CD4 aptamer, peptide targeting αvβ3 and Vi polysaccharide (targets prohibitins) on their surface using different sorting modules that result in a sustained response against Th17 cells. Furthermore, the packaging substances including drugs, nucleic acid molecules, etc., can be loaded onto the engineered exosomes for targeting Th17 cells to inhibit its pathogenic mechanism in diseases. Abbreviations: PROCR; protein C receptor, MHC; major histocompatibility complex.
Ijms 24 07647 g003
Table 1. List of exosomal RNAs in the occurrence of disease.
Table 1. List of exosomal RNAs in the occurrence of disease.
Exosome SourceExosome
Cargo
DiseaseDisease OutcomeReference
HCT116 and SerummiR-25, miR-130b and miR-425Colorectal CancerAggravates liver metastasis[52]
SerummiR-1247-3pLiver cancerPromotes lung metastasis[52]
A2780 CCMmiR-223Epithelial ovarian cancerPromotes chemoresistance[52]
VariablemiR-21Multiple cancersPromotes cancer[52]
SerummiR-7977Lung adenocarcinomaIncrease in proliferation, invasion and inhibits apoptosis[52]
Pan02 CCMmiR-155-5p and miR-221-5pPancreatic ductal adenocarcinomaPromotes metastasis[52]
HT-29/SW480miR-375-3pColon cancerInduces EMT[52]
MSCmiR-21-5pBreast cancerPromotes chemoresistance[52]
PlasmamiR-1-3pSepsisEndothelial cell dysfunction[52]
SerummiR-4449Diabetic kidneyPromotes pro-inflammation & oxidative stress[53]
MGC803, MKN45, HGC27, and SGC7901 CCMmiR-21-5pGastric cancerPromotes peritoneal metastasis[54]
Human bronchial epithelial cellsmiR-21 and miR-210COPDPromotes myofibroblast differentiation and hypoxia[55]
SerummiR-96, miR-222-3p, miR-499a-5pLung cancerPromote cell migration and invasion[55]
Induced pluripotent stem cells (IPSC)-derived astrocytes/microgliamiR-21-5pAdenocarcinomaInduce neurotoxic reactive astrocytosis, cognitive impairment[56]
TDEsmiR-141Lung cancerInduces angiogenesis and malignancy[57]
TDEsmiR-107Gastric CancerPromote immunosuppression[58]
SCLCmiR-375-3pLung CancerDisrupts vascular barrier[59]
UrinemiR-200bRenal fibrosisFibrosis progression[60]
Plasmalnc-MKRN2Parkinson DiseaseDevelops disease occurrence[52]
SerumlncRNA-UCA1Pancreatic CancerPromotes angiogenesis[52]
SerumHOXD-AS1Prostate CancerPromotes metastasis[52]
UrinelncBCYRN1, lncLNMAT2Bladder CancerPromotes lymphatic metastasis[52]
CCMLncPCGEM1Gastric cancerInduces metastasis and migration[52]
TGF-β A549lnc-MMP2-2Lung cancerPromotes invasion and metastasis[55]
A172 cellsPOU3F3GliomaPromotes angiogenesis[61]
CAFMEG3SCLCCisplatin resistance[62]
MCF7MALAT1Breast cancerPromotes proliferation[63]
Plasmacirc-RanGAP1Gastric cancerPromotes metastasis[52]
HCC CCMcirc-RNA-100338HCCPromotes angiogenesis and invasion[52]
SerumCirc-0006156Thyroid cancerPromotes tumorigenesis[52]
TDEsPDE8APancreatic cancerElevates invasive growth[64]
L-02 CCMcirc-100284HepatocarcinomaAccelerates cell cycle and proliferation[65]
Abbreviations: HCT, Human colorectal cancer cell line; CCM, Cell culture media; EMT, Epithelial to mesenchymal transition; MSC, Mesenchymal stem cell; COPD, Chronic obstructive pulmonary disease; TDEs, Tumor-derived exosomes; SCLC, Small cell lung cancer; CAF, Cancer-associated fibroblasts; MALAT1, Metastasis-associated lung adenocarcinoma transcript 1.
Table 2. Summary of exosomal cargo in pre-clinical and clinical trials as therapeutics.
Table 2. Summary of exosomal cargo in pre-clinical and clinical trials as therapeutics.
Exosome SourceExosome
Cargo
DiseaseClinical StatusReference
Plant (Grapes)CurcuminColon cancerNCT01294072
Phase I
-
Plant (Ginger)CurcuminInflammatory Bowel DiseaseNCT04879810
(Completed)
-
Dendritic cellDex2Non-small cell lung cancerNCT01159288
(Completed)
-
Plant (Grapes)LortabOral mucositisNCT01668849-
Mesenchymal stromal cellsKRASG12D siRNAPancreatic Ductal AdenocarcinomaNCT03608631
(Phase I)
-
BloodAnlotinibNon-small cell lung cancerNCT05218759-
BloodPembrolizumabHead and neck cancerNCT04453046
(Terminated)
-
Dendritic cell/macrophageChimeric exosomal tumor vaccinesBladder cancerNCT05559177
(Phase I)
-
Circulating lymphocytes and serumMerck 3475 PembrolizumabTriple-negative breast cancerNCT02977468
(Phase I)
-
Liquid biopsies18F-DCFPyL PET/CTProstatic neoplasmsNCT03824275
(Phase II/III)
-
MacrophageCDK-004Gastric cancer, colorectal cancerNCT05375604
(Phase I)
-
Human cellmiR-497Lung cancer-[79]
Dental pulp stem cellChitosan hydrogelExperimental periodontitis-[80]
MSC-NTF-COVID-19-induced ARDS-[81]
LX-2 cellsCas9 ribonucleoproteinLiver diseases-[82]
Table 3. List of drugs that can be exosomal cargo for specific targeting of Th17 cells.
Table 3. List of drugs that can be exosomal cargo for specific targeting of Th17 cells.
DrugStructureMolecular TargetReference
DigoxinIjms 24 07647 i001ROR-γT[156]
JNJ-61803534Ijms 24 07647 i002ROR-γT[157]
BMS-986251Ijms 24 07647 i003ROR-γT[158]
Izumerogant (IMU)-935-ROR-γTPhase I clinical trial
MS402Ijms 24 07647 i004BET N-terminal
bromodomain
[159]
ZebularineIjms 24 07647 i005IL-17[160]
Birabresib (OTX015)Ijms 24 07647 i006BET inhibitor[161]
MDL-101Ijms 24 07647 i007IL-6[162]
TapinarofIjms 24 07647 i008AhR[163]
LMT-28Ijms 24 07647 i009IL-6/GP130, STAT-3[164]
NeobaicaleinIjms 24 07647 i010STAT-3[165]
BazedoxifeneIjms 24 07647 i011IL-6/GP130[166]
2,20,40-TrihydroxychalconeIjms 24 07647 i012ROR-γT[167]
Ursolic AcidIjms 24 07647 i013ROR-γT[166]
IOX1Ijms 24 07647 i014TET2[168]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Samarpita, S.; Li, X. Leveraging Exosomes as the Next-Generation Bio-Shuttles: The Next Biggest Approach against Th17 Cell Catastrophe. Int. J. Mol. Sci. 2023, 24, 7647. https://doi.org/10.3390/ijms24087647

AMA Style

Samarpita S, Li X. Leveraging Exosomes as the Next-Generation Bio-Shuttles: The Next Biggest Approach against Th17 Cell Catastrophe. International Journal of Molecular Sciences. 2023; 24(8):7647. https://doi.org/10.3390/ijms24087647

Chicago/Turabian Style

Samarpita, Snigdha, and Xiaogang Li. 2023. "Leveraging Exosomes as the Next-Generation Bio-Shuttles: The Next Biggest Approach against Th17 Cell Catastrophe" International Journal of Molecular Sciences 24, no. 8: 7647. https://doi.org/10.3390/ijms24087647

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

Article Metrics

Back to TopTop