Research Paper Engineered Extracellular Vesicle-based Sonotheranostics .

1y ago
2 Views
1 Downloads
3.27 MB
20 Pages
Last View : 3d ago
Last Download : 3m ago
Upload by : Lucca Devoe
Transcription

Theranostics 2022, Vol. 12, Issue 3IvyspringInternational PublisherResearch Paper1247Theranostics2022; 12(3): 1247-1266. doi: 10.7150/thno.65516Engineered extracellular vesicle-based sonotheranosticsfor dual stimuli-sensitive drug release and photoacousticimaging-guided chemo-sonodynamic cancer therapyThuy Giang Nguyen Cao1*, Ji Hee Kang2*, Wangyu Kim3*, Junha Lim4*, Su Jin Kang1, Jae Young You1, QuanTruong Hoang1, Won Jong Kim4, Won Jong Rhee1,5 , Chulhong Kim3 , Young Tag Ko2 , Min Suk Shim1 1.2.3.4.5.Division of Bioengineering, Incheon National University, Incheon 22012, Republic of Korea.College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea.Department of Electrical Engineering, Convergence IT Engineering, and Mechanical Engineering, Medical Device Innovation Center, Pohang University of Science andTechnology (POSTECH), Pohang 37673, Republic of Korea.Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea.Research Center for Bio Materials & Process Development, Incheon National University, 119 Academy-ro, Yeonsu-gu, Incheon 22012, Republic of Korea.*These authors contributed equally to this work. Corresponding authors: Dr. Won Jong Rhee, Tel: 82-32-835-8299; E-mail address: wjrhee@inu.ac.kr; Dr. Chulhong Kim, Tel: 82-54-279-8805; E-mail address:chulhong@postech.edu; Dr. Young Tag Ko, Tel: 82-32-820-4923; E-mail address: youngtakko@gachon.ac.kr; Dr. Min Suk Shim, Tel: 82-32-835-8268; E-mail address:msshim@inu.ac.kr. The author(s). This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/).See http://ivyspring.com/terms for full terms and conditions.Received: 2021.07.30; Accepted: 2021.12.07; Published: 2022.01.01AbstractSonodynamic therapy has shown promise as an effective alternative to conventional photodynamic therapyowing to its ability to treat deep-seated tumors. However, the development of stimuli-responsivesonosensitizers with high biocompatibility faces a significant challenge.Methods: In this study, we developed dual stimuli-responsive sonosensitizers with desirable biosafety usingextracellular vesicles (EVs), a class of naturally occurring nanoparticles. Indocyanine green (ICG), whichfunctions as both a sonosensitizer and photoacoustic (PA) imaging agent, was loaded into EVs, together withpaclitaxel (PTX) and sodium bicarbonate (SBC), to achieve pH-responsive PA imaging-guidedchemo-sonodynamic combination therapy.Results: The EVs significantly improved the cellular uptake of ICG, thus triggering enhanced sonodynamiceffects in breast cancer cells. SBC-, ICG-, and PTX-loaded EV [SBC-EV(ICG/PTX)] efficiently released the PTXin response to acidic pH in the endo/lysosomes because CO2 bubbles generated from the SBC caused the EVmembranes to burst. The drug release was further facilitated by ultrasound (US) treatment, demonstrating dualpH/US-responsive drug release. The ICG- and PTX-loaded EVs exhibited efficient anticancer activity againstbreast tumor cells owing to the combination of chemo-sonodynamic therapy. High-resolution PA imagingvisualized the preferential tumor accumulation of SBC-EV(ICG/PTX) in tumor-bearing mice. Notably, a singleintravenous injection of SBC-EV(ICG/PTX) with US irradiation significantly suppressed tumor growth in micewithout systemic toxicity.Conclusions: Our findings demonstrate that dual stimuli-responsive SBC-EV(ICG/PTX) are promisingsonotheranostic nanoplatforms for safe and efficient chemo-sonodynamic combination cancer therapy andphotoacoustic imaging.Key words: extracellular vesicle, sonodynamic cancer therapy, sonotheranostics, pH-sensitive release, photoacoustic imagingIntroductionOxidative stress-inducing anticancer therapiesby increasing the concentration of reactive oxygenspecies (ROS) have recently emerged as an attractiveoption for efficient cancer treatment [1-3]. Based onthis approach, photodynamic therapy (PDT), in whichthe activation of photosensitizers with light generatescytotoxic ROS via energy transfer to neighboringoxygen molecules, has garnered significant attentionfor targeted and effective cancer therapy [4].Although PDT has shown selective tumor eradicationhttps://www.thno.org

Theranostics 2022, Vol. 12, Issue 3and low systemic toxicity, its clinical application hasbeen challenged by its limited light penetration intissues [5]. Sonodynamic therapy (SDT), which adoptssonosensitizers to generate ROS under ultrasound(US), enables the treatment of various kinds ofdeep-seated tumors owing to the deep tissuepenetration of US. Therefore, SDT has emerged as apromising alternative [6, 7].Although the outcome of SDT is greatly affectedby the potency of sonosensitizers, the development ofeffective and safe sonosensitizers, considering thequantum yields of ROS production, biocompatibility,cancer-selectivity, and long-term stability, remains asignificant challenge [8]. The emergence and rapiddevelopment of nanomaterials together withsonosensitizers has gained considerable attentionbecause they can significantly improve thephysicochemical properties and anticancer activitiesof sonosensitizers by addressing their criticallimitations [9, 10]. It has been reported that nanocarriers, including micelles, liposomes, and intracellular delivery, cancer-specificity, and in vivopharmacokinetic properties of sonosensitizers [9 11].Recently, several studies have demonstrated thatcombination cancer therapy can realize synergisticeffects in cancer treatment and reduce the therapeuticdoses of anticancer drugs, thereby resulting inminimal side effects [12 15]. Combination ofchemotherapy with SDT has shown great promise toachieve synergistic therapeutic outcomes for cancertreatment [16]. SDT has also been demonstrated toovercome drug resistance of chemotherapy byimproving the cellular internalization of drugs andreducing the expression levels of ATP-bindingcassette transporters such as P-glycoprotein [16]. Toobtain effective chemo-sonodynamic combinedtherapeutic efficacy, the use of nanocarriers that cansimultaneously and preferentially deliver bothsonosensitizers and anticancer drugs into cancer cellsis desirable [16]. In addition, engineering nanocarriersthat can facilitate the release of anticancer drugs inresponse to specific stimuli in tumors is crucial tomaximize anticancer efficacy while avoidingunwanted drug release in surrounding normaltissues. Despite the enormous potential of stimuliresponsive nanocarriers for chemo-sonodynamiccombination therapy, their inherent toxicity andimmunogenicity have limited their widespread use inclinical practice [9]. Therefore, to facilitate thetranslation of chemo-sonodynamic combinationtherapy in clinical practice, the development of highlybiocompatible stimuli-responsive nanocarriers iscrucial.In recent years, extracellular vesicles (EVs),1248nanoscale cell-derived membrane vesicles, haveemerged as safe and versatile nanoplatforms for drugdelivery [17, 18]. These endogenous nanoplatformsensure exceptional biocompatibility, prolonged bloodcirculation, and minimal or no immunogenicity [19,20]. In addition, nanoscale EVs can preferentiallyaccumulate into tumors owing to enhancedpermeability and retention (EPR) effects [21]. Thus,EVs could be effective nanoplatforms for the selectivedelivery of sonosensitizers and anticancer drugs intotumor legions, ensuring a synergistic and tumorspecific combination anticancer therapy.Optical imaging for the early diagnosis ofdiseases and treatment evaluation is crucial forpersonalized medicine [22, 23]. Theranosticnanocarriers that integrate therapy and diagnostics ina single platform have exhibited the substantialpotential to play pivotal roles in personalized cancertherapy, including early diagnosis, personalizedtreatment, and treatment monitoring [24, 25].Photoacoustic (PA) imaging, which obtains imagesvia conversion of photon energy into acousticpressure waves, has rapidly emerged as an attractiveimaging modality for clinical cancer diagnosis [26-28]owing to the imaging of deep tissues at high spatialresolution [29-32]. Therefore, the development of atheranostic agent that can achieve PA imaging-guidedcancer therapy is crucial because it can enhance y.In this study, we developed sonosensitizer andanticancer drug-loaded EVs to achieve simultaneousPA imaging and combination chemo-SDT. A nearinfrared (NIR)-absorbing indocyanine green (ICG), abiocompatible theranostic dye approved by the Foodand Drug Administration (FDA) [33, 34], was loadedinto the EVs to function as both a sonosensitizer andPA imaging agent. The anticancer drug, paclitaxel(PTX), was also encapsulated into the EVs to realizecombined chemo-SDT. In addition, to renderpH-sensitive EVs, NaHCO3 (sodium bicarbonate,SBC) was incorporated into the EVs together with ICGand PTX. In acidic environments, the entrapped SBCin the EVs decomposes spontaneously, rapidlygenerating CO2 bubbles [35], which can cause EVmembranes to burst and readily release the payloads.Such pH-responsive EVs could be effective for theintracellular release of drugs once they enter theacidic endo/lysosomes after endocytosis into cancercells.These pH-sensitive EVs encapsulating ICG andPTX exhibited several salient features. Theencapsulation of ICG into EVs can address theintrinsic drawbacks of ICG, such as low aqueousstability, rapid systemic clearance, and lack ofhttps://www.thno.org

Theranostics 2022, Vol. 12, Issue 3cancer-targeted delivery [36, 37]. Notably, EV-baseddrug delivery systems have better safety profiles thanother synthetic nanocarriers because they solelycomprise biocompatible materials [that is, EVs andcargo (i.e., ICG)]. These pH-sensitive EVs can facilitatethe endo/lysosomal escape and cytoplasmic release ofdrugs via the acid-destabilization of the EVs inresponse to the acidic pH in the endo/lysosomalcompartments, thereby enabling enhanced anticancereffects. Finally, nanoscale ICG-loaded EVs canfunction as cancer-targeted theranostic nanocarriersto achieve PA imaging-guided SDT against cancer.Figure 1 presents a schematic of the PAimaging-guided combination chemo-SDT usingpH-sensitive SBC-loaded EVs comprising ICG andPTX [SBC-EV(ICG/PTX)]. Once the EVs enter thecancer cells via endocytosis, the acidic pH in theendo/lysosomes facilitates the release of ICG and PTXby the generation of CO2 bubbles from the SBC. Afterthe cytoplasmic release of ICG and PTX, USirradiation activates the ICG to generate ROS in breastcancer cells, which subsequently triggers apoptosis.PTX also induces apoptosis, resulting in synergisticanticancer effects via a combination of chemotherapyand SDT. The in vivo biodistribution of pH-sensitiveEVs can be monitored by PA imaging. To the best of1249our knowledge, this is the first study to utilize dualpH/US-sensitive EVs for PA imaging-guided,cancer-targeted combination chemo-SDT. Dualstimuli-sensitive EVs have great potential for clinicalapplications owing to their high biocompatibility andanticancer efficacies.Results and DiscussionDesign, preparation, and characterization ofdrug-loaded EVsA significant limitation to anticancer drugdelivery is the selective delivery of therapeutic drugsto tumor areas [38]. It is well known that nanoscaledrug carriers can selectively accumulate into tumorlesions via EPR effects [38-40]. However, artificialnanocarriers face certain critical challenges, such assystemic toxicity and nonspecific uptake by thereticuloendothelial system [41]. In contrast tosynthetic nanocarriers, EVs are naturally occurringnanoparticles with excellent biocompatibility andlong-term blood circulatory capability [42]. In thisstudy, EVs were engineered as sonotheranosticnanocarriers for safe and efficient delivery ofsonosensitizers (i.e., ICG) and chemo drugs (i.e., PTX)into tumors.Figure 1. Schematic illustration of the sonotheranostic action of dual pH/US-responsive EVs co-encapsulating ICG, PTX, and SBC. Once the EVs enter thecancer cells via endocytosis, the acidic pH in the endo/lysosomes facilitates the release of ICG and PTX owing to bursting effects triggered by the generation of CO2 bubbles fromthe SBC. After the cytoplasmic release of ICG and PTX, US irradiation triggers sonodynamic effects by generating intracellular ROS, subsequently triggering the apoptosis ofbreast cancer cells. The ICG-loaded EVs generate PA imaging signals, thus enabling PA imaging-guided chemo-sonodynamic combination cancer therapy.https://www.thno.org

Theranostics 2022, Vol. 12, Issue 31250Figure 2. (A) Absorption spectra of blank EV, free ICG, free PTX, EV(ICG), EV(ICG/PTX), and SBC-EV(ICG/PTX) samples. (B) TEM images of blank EV and SBC-EV(ICG/PTX).Scale bars indicate 50 nm. (C) Western blot of typical exosomal markers (CD81, CD63, and syntenin) from HEK-293T cells and various EV samples. Calnexin and GAPDH,housekeeping genes, were used as a control. (D) Changes in fluorescence intensity of free ICG, EV(ICG), SBC-EV(ICG), and SBC-EV(ICG/PTX) after incubation in PBS (4 C) for14 days. (E) PA amplitude of free ICG, EV(ICG/PTX), SBC-EV(ICG/PTX) at different ICG concentrations. (F) Change in the size of SBC-EV(ICG/PTX) under physiological (i.e.,pH 7.4) and acidic (i.e., pH 6.0) conditions, determined by DLS. After 48 h of incubation in pH 6.0 buffers, SBC-EV(ICG/PTX) were destabilized and swollen, and the sizedistribution increased drastically. (G) TEM images of SBC-EV(ICG/PTX) after incubation in pH 7.4 and 6.0 buffers for 48 h. Scale bars are 50 nm. (H) PTX release profiles fromSBC-EV(ICG/PTX) at different pH and US (1 min of irradiation) conditions. (I) PTX release profiles of EV(ICG/PTX) and SBC-EV(ICG/PTX) at different pH conditions after 8 hof incubation (*p 0.05, **p 0.01, n 3).To prepare ICG- and PTX-loaded EVs, EVs wereobtained from human embryonic kidney HEK-293Tcells. It was reported that EVs obtained fromHEK-293T cells are benign in terms of in vivoimmunogenicity and toxicity [43]. HEK-293Tcell-derived EVs are known to contain low levels ofproteins and RNAs associated with the disease- orcancer-related pathways [44]. HEK-293T cell-derivedEVs in phosphate-buffered saline (PBS) were solelymixed with an ICG solution or together with a PTXsolution in dimethyl sulfoxide (DMSO). DMSO wasused as a permeability enhancer by increasing thesolubility of drugs and their permeability across thelipid membrane of EVs [45, 46]. It has been reportedthat DMSO can induce water pores in lipid bilayersand thus enhance the membrane permeability of bothhydrophilic and hydrophobic molecules [46]. Theloading capacities of ICG and PTX into EVs wereproportional to the concentrations of DMSO (Table 1).4% (v/v) DMSO in PBS was used for drugencapsulation because higher DMSO concentrationssignificantly destroyed EVs.The loading of ICG and PTX into EVs wasconfirmed by UV-Vis spectroscopy. The UV-Visabsorption spectra of blank EV, ICG, PTX, ICG-loadedEV [EV(ICG)], ICG- and PTX-loaded EV[EV(ICG/PTX)],andSBC-EV(ICG/PTX)areillustrated in Figure 2A. Both EV(ICG) andhttps://www.thno.org

Theranostics 2022, Vol. 12, Issue 3EV(ICG/PTX) indicated the distinct absorption peakof ICG at 786 nm (Figure 2A). The encapsulationefficiencies of ICG and PTX for SBC-EV(ICG/PTX)were 42.9% and 39.9%, respectively (Table 1). Theloading capacities of ICG and PTX forSBC-EV(ICG/PTX) were 4.76 10-10 and 1.92 10-10μg/EVs, respectively. The loading capacities of ICGand PTX in EV(ICG/PTX) were 4.77 10-10 and 1.90 10-10 μg/EVs. This indicates that SBC does not affectthe drug loading of ICG and PTX into EVs. BecausePTX is a hydrophobic compound, it might beincorporated into the hydrophobic lipid bilayers ofEVs, as reported in a previous study [47]. Relativelyhydrophilic ICG might be entrapped within theaqueous core or at the bilayer interface [48].Table 1. Drug encapsulation efficiency of ICG inSBC-EV(ICG/PTX) with different DMSO composition ratios (v/v)in PBS for the preparationDMSO composition ratio (v/v)ICG encapsulation efficiency (%)PTX encapsulation efficiency (%)0%1%2%3%4%23.4 1.8 36.8 0.8 38.2 0.2 41.7 0.8 42.9 3.5N/A5.7 3.6 19.6 0.5 32.4 1.3 39.9 1.2Because EVs are surrounded by a lipid ctures, they can carry various hydrophobic andhydrophilic drugs. To demonstrate the capability ofEVs to encapsulate versatile drugs, hydrophobicmodel drug, piperlongumine, and hydrophilic modeldrug, doxorubicin hydrochloride (DOX·HCl), wereencapsulated into EVs in the same manner used forICG and PTX. Both piperlongumine and DOX·HClwere successfully encapsulated into EVs with highencapsulationefficiencies(36.3 2.3%forpiperlongumine and 41.1 3.2% for DOX·HCl),demonstrating that EVs are universal drug carriers.The encapsulation efficiency and loadingcapacity of SBC in EVs were measured by quantifyingof sodium ions via inductively coupled plasma opticalemission spectrometry (ICP-OES). The encapsulationefficiency of SBC for SBC-EV(ICG/PTX) was 26.4%.The loading capacity of SBC for SBC-EV(ICG/PTX)was 1.09 10-10 μg/EVs. It was determined thatapproximately 91% of the initial amount of SBC in theSBC-EV(ICG/PTX) remained stable for about 48 hupon incubation in pH 7.4 buffers.The sizes and surface charges of variousEV-derived samples were measured. As indicated inTable 2, the sizes of blank EV, EV(ICG), SBC- andICG-loaded EV [SBC-EV(ICG)], EV(ICG/PTX), andSBC-EV(ICG/PTX)were117.1 2.0,129.6 3.1,132.7 6.8, 149.9 5.2 and 150.8 4.2 nm, respectively.The size of the EVs increased slightly when they wereloaded with ICG and PTX. The zeta potentials ofblank EV, EV(ICG), SBC-EV(ICG), EV(ICG/PTX), and1251SBC-EV(ICG/PTX) were 19.6 1.2, 20.1 0.3, 23.6 2.5, 22.7 1.2, and 26.4 2.4 mV, respectively(Table 2). The morphologies of the blank EV andSBC-EV(ICG/PTX) were analyzed by transmissionelectron microscopy (TEM). They exhibited sphericalmorphologies with diameters of 50–90 nm (Figure 2B).Table 2. Size and surface charges of various EV-derived samplesSamplesBlank X)Size (nm)117.1 2.0121.1 3.2129.6 3.1132.7 6.8149.9 5.2150.8 4.2Zeta potential (mV) 19.6 1.2 20.2 0.5 20.1 0.3 23.6 2.5 22.7 1.2 26.4 2.4The expressions of three typical EV markerproteins, CD81, CD63, and syntenin, were visualizedby western blotting to verify whether the ICG- andPTX-encapsulation processes affect the proteincontent of EVs. Blank EV, EV(ICG/PTX), andSBC-EV(ICG/PTX) showed clear bands representingCD81, CD63, and syntenin (Figure 2C). This resultindicates that the original protein content of the EVswas not affected by the drug-loading processes.High colloidal stability and storage stability ofengineered EVsThe interaction of nanoparticles with proteins inthe blood is one of the crucial factors influencing theirphysicochemical properties, colloidal stability, anddelivery efficacy in vivo [49]. To investigate X) against serum proteins, they weresuspended in PBS containing 10% fetal bovine serum(FBS), followed by incubation at 37 C for 96 h. Thesizes of the EV(ICG/PTX) and SBC-EV(ICG/PTX)were monitored to determine whether aggregationoccurred during the incubation. As illustrated inFigure S1, both EV(ICG/PTX) and SBC-EV(ICG/PTX)did not exhibit significant size changes up to 96 h,regardless of incubation conditions, which indicatestheir high stability against serum proteins. The highcolloidal stability of SBC-EV(ICG/PTX) ensuresprolonged blood circulation for efficient chemo-SDTin vivo.Enhanced aqueous stability of ICG viaencapsulation into EVsICG suffers from irreversible degradation byions and radicals in aqueous solutions, reducing itsabsorption and fluorescence simultaneously [50, 51].EVs contain biocompatible phospholipid bilayers toprotect the encapsulated cargos from extracellularenvironments. To investigate whether EVs can protecthttps://www.thno.org

Theranostics 2022, Vol. 12, Issue 3ICG from degradation in aqueous solutions, wequantified the fluorescence intensities of free ICG,EV(ICG), SBC-EV(ICG), and SBC-EV(ICG/PTX)during incubation in PBS at 4 C. As represented inFigure 2D, all ICG-loaded EVs retained initialfluorescence intensities of ICG higher than 84% after14 days of incubation. In contrast, the free ICGsolution substantially lost its fluorescence after 14days of incubation (49% of its initial value), verifyingits poor aqueous stability. The findings imply thatEVs can efficiently protect the entrapped ICG fromdestructive species in buffer solutions, confirmed byprevious studies [52, 53]. In addition to the highcolloidal stability against serum proteins (Figure S1),the improved aqueous stability of ICG-loaded EVsindicates that EV-mediated ICG delivery exhibitssubstantial potential for clinical applications.In vitro PA imagingTo assess the PA properties of ICG-encapsulatedEVs, the PA signals of free ICG, EV(ICG/PTX), andSBC-EV(ICG/PTX) were measured at different ICGconcentrations (Figure 2E). The PA signals from allthe samples at 780 nm linearly increased as theconcentration increased, thus validating theirexcellent PA contrast.Acid-triggered CO2 generation by SBC-loadedEVsThe generation of CO2 from SBC-loaded EVsunder acidic conditions was determined by anacid-base titration method using a commerciallyavailable CO2 quantification kit. CO2 generated fromSBC-loaded EVs dissolves in aqueous solutions andforms carbonic acid (H2CO3). The carbonic n as the indicator. The relative levels ofCO2 generation from SBC-loaded EVs and blank EVsat different pH conditions was determined by theamount of NaOH used to neutralize the solution ofcarbonic acid. As illustrated in Figure S2, blank EVexhibited no significant increase in CO2 generationcompared to untreated groups, regardless of pHconditions. In contrast, SBC-EV showed a significantincrease in CO2 generation at pH 6.0, demonstratingthe acid-triggered CO2 generation by SBC inside EVs.pH-responsiveness of SBC-loaded EVsTo demonstrate the pH-responsiveness ofSBC-loaded EVs, changes in size under acidicconditions were measured using dynamic lightscattering (DLS). The average size and sizedistribution of SBC-EV(ICG/PTX) dramaticallyincreased after 48 h of incubation in pH 6.0 buffers(Figure 2F). This result indicates that SBC-EV(ICG/PTX) are destabilized under acidic conditions,1252resulting from the CO2-triggered bursting of the EVmembranes. The morphology change of the SBCEV(ICG/PTX) under acidic conditions was observedby TEM. The TEM image clearly showed noticeablemembrane destruction with pores when theSBC-EV(ICG/PTX) were incubated in pH 6.0 buffers(Figure 2G). This result demonstrates the acidtriggered destabilization of the SBC-EV(ICG/PTX).Therefore, it is expected that SBC-EV(ICG/PTX)would facilitate drug release under acidic conditions.Dual pH- and US-responsive drug release bySBC-loaded EVsTo validate the ability of SBC-loaded EVs torealize pH-responsive drug release, the releaseprofiles of PTX and ICG were determined at differentpH conditions via the dialysis method. In addition, toevaluate whether US could facilitate pH-sensitivedrug release from the SBC-loaded EVs, the release ofPTX was determined at different pH conditions (e.g.,endo/lysosomal pH of 6.0 and tumor microenvironmental pH of 6.6) in the presence or absence ofUS irradiation. As represented in Figure 2H, thecumulative release of PTX at pH 5.0 and 6.0 wasmarkedly higher than that at pH 7.4, therebydemonstrating that pH-sensitive SBC-EV(ICG/PTX)can rapidly release PTX under mildly acidicconditions, similar to that of endo/lysosomalcompartments. Interestingly, regardless of pHconditions, US treatment further facilitated the releaseof PTX, demonstrating the dual pH- and USresponsive drug release by SBC-loaded EVs (Figure2H). The US-triggered drug release by SBC-loadedEVs can be ascribed to the US-induced cavitationeffect (collapse of vapor-filled bubbles in liquids). Thiscollapse creates shock waves that can destabilizeSBC-EV(ICG/PTX) and release the encapsulateddrugs [54]. In addition, US irradiation-triggered ROSgeneration from ICG may lead to lipid peroxidationand consequent destabilization of EVs, facilitating therelease of ICG and PTX [55]. This physical andchemical disruption of EVs through US irradiationwould facilitate the release of PTX entrapped in thehydrophobic lipid bilayers of EVs.The cumulative release of PTX fromSBC-EV(ICG/PTX) was the highest when incubatedat pH 5.0, in the presence of US (Figure 2H). To verifythat SBC is an actual contributor in triggering thepH-sensitive drug release from SBC-loaded EVs, wecompared the cumulative PTX release fromEV(ICG/PTX) and SBC-EV(ICG/PTX) at different pHconditions. The amount of PTX released fromSBC-EV(ICG/PTX) was significantly higher than thatfrom EV(ICG/PTX) when they were incubated at pH5.0 (Figure 2I). In contrast, the amount of PTX releasedhttps://www.thno.org

Theranostics 2022, Vol. 12, Issue 3from SBC-EV(ICG/PTX) was similar to that releasedfrom EV(ICG/PTX) when incubated at pH 7.4,thereby implying that SBC primarily contributes tothe pH-sensitive drug release of the SBC-EV(ICG/PTX). The dual pH/US-dependent ICG release fromSBC-EV(ICG/PTX) was also demonstrated (FigureS3). The highest cumulative release of ICG wasobserved at pH 5.0, regardless of US irradiation andincubation time (Figure S3). Similar to what wasobserved with the PTX release, the release of ICGfrom SBC-EV(ICG/PTX) was further facilitated by UStreatment. Drug release profiles of SBC-EV(ICG/PTX)were also evaluated at pH 6.6 that mimics mildlyacidic tumor microenvironment conditions. As shownin Figure S4, the amounts of PTX and ICG releasedfrom SBC-EV(ICG/PTX) were significantly higherthan those from EV(ICG/PTX) when they wereincubated at pH 6.6. The dual pH- and US-responsiveSBC-loaded EVs would regulate the release of drugsmore precisely at the target tumor region. Theenhanced release of ICG and PTX by dualpH/US-responsive EVs would ensure highanticancer effects of chemo-SDT in target cancercells.Efficient cellular uptake of ICG-loaded EVsTo investigate whether EVs can enhanceintracellular delivery of their therapeutic cargos, freeICG, EV(ICG), and SBC-EV(ICG) at equivalentconcentrations of ICG were added to MCF-7 humanbreast cancer cells. The cellular uptake of ICG forEV(ICG) and SBC-EV(ICG) was significantlyincreased compared to free ICG (Figure 3A). Theenhanced cellular uptake of EV(ICG) andSBC-EV(ICG) can be explained by the favorableinteraction of EVs with the cell membranes viareceptor-ligand binding or direct fusion [56]. It iswell-documented that EVs efficiently enter the cellsvia numerous molecular mechanisms, includingclathrin-dependent receptor-mediated endocytosis,micropinocytosis, membrane fusion, etc [57]. Theroutes of cellular uptake depend on the presence ofspecific surface proteins on both the EVs and therecipient cells. Although it is still unclear whichuptake mechanism is preferred, EVs can fuse withendosomal/lysosomal membranes and release theircargos into the cytoplasm [58].To investigate whether SBC in EVs affects theircellular uptake, fluorescein isothiocyanate (FITC)labeled EVs were used to prepare EV(ICG) andSBC-EV(ICG). The cellular uptake of SBC-EV(ICG)and EV(ICG) was compared by measuring thefluorescence intensity of FITC. The result showed thatthere was no significant difference in cellular uptakebetween FITC-EV(ICG) and FITC-SBC-EV(ICG)1253(Figure S5).Enhanced cytoplasmic release of ICG bySBC-loaded EVsTo investigate whether SBC-loaded EVs canfacilitate the cytoplasmic release of their cargos,intracellular localization and endosomal escape ofSBC-EV(ICG) were evaluated by confocal laserscanning microscopy. To track the intracellulardistribution of EVs, rhodamine B (RB)-labeled EVswere used to encapsulate SBC and ICG. The resultingRB-labeled SBC-EV(ICG) was incubated with liveMCF-7 cells. RB-labeled EV(ICG) was also used as anon-pH-responsive counterpart. MCF-7 cells werestained with LysoTracker, which selectivelyaccumulates into endo/lysosomal regions, toinvestigate whether EVs can be internalized by thecells via endocytosis. As shown in Figure 3B, a largeamount of RB-labeled EV(ICG) and RB-labeledSBC-EV(ICG) (red dots) was colocalized withLysoTracker (green). This result confirms thatICG-loaded EVs were internalized by the cells viaendocytosis, which is consistent with previous studies[59].After endocytosis of EVs, a prerequisite forefficient cytoplasmic delivery of their cargos is theirescape from endo/lysosomal compartments. Toinvestigate whether ICG-laded EVs can escape theendo/lysosomal compartments, the degree ofcolocalization between RB-labeled EVs andLysotracker was quantified at different incubationtimes. The quantitative result (Figure 3C) revealedthat colocalization of pH-responsive RB-labeledSBC-EV(ICG) with endo/lysosomes significantlydecreased with time. In contrast, colocalization omes was slightly reduced. The moreefficient endosomal escape of the SBC-EV(ICG) mightbe implicated in the destabilization of endo/lysosomal membranes via bursting effects of theSBC-EV(ICG) in response to acidic endo/lysosomalpH. However, the detailed mechanism of endosomalescape of the SBC-loaded EVs still remainedunexplored. A recent study proposed that directfusion of EVs with endo/lysosomal membranes is onethe main mechanisms for endo/lysosomal escape ofEVs [59]. This study demonstrated that a fraction ofinternalized EVs undergo fusion with endo/lysosomal membranes, resulting in the release of theircargos [59]. The fusion of EVs with the endo/lysosomal membranes is facilitated by the acidic pHin the endo/lysosomes that may induce changes inmembrane fluidity and lipid compositions of EVs [60].https://www.thno.org

Theranostics 2022,

functions as both a sonosensitizer and photoacoustic (PA) imaging agent ,was loaded into EVs, together with paclitaxel (PTX) and sodium bicarbonate (SBC), to achieve pH-responsive PA imagingguided - chemo-sonodynamic combination therapy. Results: The EVs significantly improved the cellular uptake of ICG, thus triggering enhanced sonodynamic

Related Documents:

RESEARCH ARTICLE Open Access Salivary extracellular vesicle-associated miRNAs as potential biomarkers in oral squamous cell carcinoma Chiara Gai1, Francesco Camussi2, Roberto Broccoletti2, Alessio Gambino2, Marco Cabras2, Luca Molinaro1, Stefano Carossa2, Giovanni

2. Extracellular matrices contribute to the viscoelastic properties of connective tissues including the dermis The viscoelastic properties of human tissues are principally governed by the nature of the extracellular matrix (ECM). The ECM comprises secreted proteins that are deposited into the extracellular space.

shallow. A transverse ventral cervical groove presents (Fig. 5) the anterior end usually with a cuticular inflation or vesicle limited behind, about the level of the excretory pore, by a transverse ventral groove (Fig. 3). There is a ventral cervical groove near the anterior to which the cuticle is dilated to form a cephalic vesicle .This groove

Ca2 ‐triggered patch model for membrane repair Membrane disruption Ca2 entry through the injury site Ca2 ‐triggering vesicle exocytosis Vesicle fusion Patch formation; membrane resealing Nature Reviews Molecular Cell Biology 6, 499-505 (June 2005)

Simpson Engineered Connectors ENGINEERED WOOD PRODUCTS 6th Edition 10/16 Job-site delivery available in most areas Full scanning & printing capabilities Covered storage of engineered products Rapid turnaround of plans Wide range of engineered products Commercial joists available In-house

CAPE Management of Business Specimen Papers: Unit 1 Paper 01 60 Unit 1 Paper 02 68 Unit 1 Paper 03/2 74 Unit 2 Paper 01 78 Unit 2 Paper 02 86 Unit 2 Paper 03/2 90 CAPE Management of Business Mark Schemes: Unit 1 Paper 01 93 Unit 1 Paper 02 95 Unit 1 Paper 03/2 110 Unit 2 Paper 01 117 Unit 2 Paper 02 119 Unit 2 Paper 03/2 134

Capture of large extracellular particles in vesicles. how many single-celled organisms feed (e.g., amoeba) how cells of the immune system destroy invaders PINOCYTOSIS Plasma membrane Vesicle 0.5 µm Pinocytosis vesicles forming (arrows) in a cell lining a small blood vessel (TEM) Pinocytosis ("cell drinking")

Anatomy and Physiology for Sports Massage 11. LEVEL: 3: Term: Definition: Visuals: Cytoplasm Within cells, the cytoplasm is made up of a jelly-like fluid (called the cytosol) and other : structures that surround the nucleus. Cytoskeleton The cytoskeleton is a network of long fibres that make up the cell’s structural framework. The cytoskeleton has several critical functions, including .