Get 20M+ Full-Text Papers For Less Than $1.50/day. Start a 14-Day Trial for You or Your Team.

Learn More →

The potential therapeutic role of camel milk exosomes – A review

The potential therapeutic role of camel milk exosomes – A review Ann. Anim. Sci., Vol. 23, No. 2 (2023) 353–362 DOI: 10.2478/aoas-2022-0072 1 2♦ 3 4 5 Norah A. Althobaiti , Sayed Haidar Abbas Raza , Mona N. BinMowyna , Reem D. Aldawsari , Sameh A. Abdelnour , 6 2 7 8 2 Mahmoud Abdel-Hamid , Dwi Wijayanti , Afaf Kamal-Eldin , Atif Khurshid Wani , Linsen Zan Biology Department, College of Science and Humanities, Al Quwaiiyah, Shaqra University, Al Quwaiiyah 19257, Saudi Arabia College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China College of Applied Medical Sciences, Shaqra University, Shaqra, Saudi Arabia Biology Department, College of Science and Humanities-Al Quwaiiyah, ‎Shaqra University, Al Quwaiiyah 19257, Saudi Arabia Department of Animal Production, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt Dairy Science Department, Faculty of Agriculture, Cairo University, Giza 12613, Egypt Department of Food Science, College of Food and Agriculture, United Arab Emirates University (UAEU), Al Ain, United Arab Emirates School of Bioengineering and Biosciences, Lovely Professional University, Punjab (144411), India Corresponding author: dr.haiderabbasraza@gmail.com abstract exosomes (exos) are naturally occurring nano-sized vesicles released into the extracellular environment by exocytosis. exos contribute to intercellular communications by regulating Dna, mrna, and protein levels. exos are considered good vehicles for delivering thera - peutic agents in multiple diseases. camel milk (cm) is a valuable food with a rich source of exos. camel milk exosomes (cmexos) have unique ingredients compared to other animal milks. it is reported that cmexos reduce the growth of cancer cells by inducing apoptosis, and inhibiting oxidative stress and inflammation. CMEXOs can block stress signaling pathways including inflammation and apoptosis which could be resulted in adverse effects if medication levels exceed the therapeutic range. moreover, cmexos improve the antioxidant capability in both in vitro and in vivo experiments. on the other hand, the anti-angiogenesis property of cmexos has been reported via decrease in expression of the angiogenesis-related gene of vascular endothelial growth factor (VEGF). it is predicted that exosomal lactoferrin (lf) and kappa casein (kc) mrnas are crucial parts of cmexos mediating their anticancer effects. The immunomodulatory effect of cmexos may be attributed to their high contents of lf and kc. according to previous works, cmexos are promising alternatives in developing new therapeutic models for multipile diseases. This review aims to provide an overview of the isolation, characterization and biological activities of the exosomes derived from camel milk for addressing their possible use in thera- peutics. Key words: camel milk exosomes, anticancer activity, anti-inflammatory activity, immuno-modulatory activity, antioxidant activity In the Middle East, the daily drinking of camel milk The EXOs are located in different tissues and biologi- (CM) is usually supposed to improve immune capacity and cal fluids including blood plasma, milk, saliva, oviductal, decrease the risk of many diseases. Several beneficial prop- urine, cerebrospinal fluid, ascites, amniotic fluid and erties of CM were reported including immunostimulatory, others (Colombo et al., 2014). Among them milk is the anti-microbial, anti-viral, anti-parasitic, anti-inflammatory, only biological fluid comprising high amounts of EXOs antioxidant, and anti-allergic effects (Badawy et al., 2018; (Adriano et al., 2021). Camel milk (CM) is one of the El-Kattawy et al., 2021; Khan et al., 2021). The main health- wealthy sources of beta-caseins and whey proteins, com- encouraging bioactive components in CM are lactoferrin prising lactoperoxidase, lysozyme, alpha-lactalbumin, (LF), kappa casein (KC), and some other molecules having lactoferrin, and immunoglobulins. The antioxidant, anti- the mentioned properties (Iigo et al., 2009; Shariatikia et al., bacterial, anti-diabetic, anti-viral, anti-cancer, and anti- 2017). Exosomes (EXOs) are common extracellular nan- inflammatory properties of CM might be attributed to the ovesicles that contribute to intercellular communications by high contents of those biological components (Khan et transporting, supporting and chaperoning biologically ki- al., 2021; Swelum et al., 2020; Mostafa et al., 2022). netic elements such as DNA, mRNA, microRNA, proteins, Among all types of exosomes in different milk sourc- etc. (Colombo et al., 2014; Cintio et al., 2020). Because of es, camel milk exosomes (CMEXOs) have received their nano size (40–100 nm diameter), EXOs are auspicious much awareness in the last few years, owing to their di- delivery elements in individual therapy (Bunggulawa et agnostic, theranostic, and therapeutic uses. al., 2018). EXOs that are isolated from milk, have superior Several authors reported that EXOs of CM have more significance impacts due to their capability in transferring stability in low pH and high and low temperature (Yassin genetic material from mother to infants (Mincheva-Nilsson et al., 2016) and it can be easily absorbed through the and Baranov, 2010). gastrointestinal barrier to circulation (Baier et al., 2014). 354 N.A. Althobaiti et al. Furthermore, EXOs of cow or human milks also have of doxorubicin-encapsulated goat milk exosomes caused anti-cancer properties and improve immune response significant inhibition of cell viability in several types against different tumor cells (Anand, 2010; Abels and of cancer cells including A549, HCT-116, and HepG2 Breakefield, 2016). cells (Ahmed et al., 2022). Santos-Coquillat et al. (2022) The anti-cancer properties of CMEXOs have been reported that the concentration of pure goat’s milk ex- described via suppression of inflammation, metastasis, osomes was 2.29±0.25 mg/mL as quantified by Coomas- oxidative stress (OS), and augmentation of tumor cell sie-Bradford test. While Yassin et al. (2016) found that apoptosis (Abels and Breakefield, 2016). Generally, the levels of phosphatidylserine, and phosphatidylcho- CMEXOs have greater biocompatibility, low adverse line ranged between ~10–15 µg/mL and ~20–25 µg/mL, immune effects and no systemic toxicity, and inflam- respectively, in dromedary milk exosomes which were matory responses (Badawy et al., 2018; Bunggulawa et steady throughout various lactation stages. Several sci- al., 2018). In the last years, it was reported that EXOs entific papers suggested that the beneficial health effects are promising candidates as anti-cancer drugs by induc- of CM might be attributed to its high contents of phos- tion of apoptosis and suppression of angiogenesis and pholipids, such as those reported in CMEXOs (Ali et al., inflammation pathways (El-Kattawy et al., 2021). The 2017; Zheng et al., 2022). After extensive investigations immunomodulatory and antioxidant effects of CMEXOs of milk exosomes, it should be clear that no standard are due to the high contents of caseins and lactoferrin amounts of EXOs have been recorded. From this point of (Ibrahim et al., 2019). However, the use of milk EXOs view, extensive clarification should be done in this scien- in therapeutic sciences still needs to be further explored. tific point to develop the method of characterization and Therefore, there is an indispensable necessity to dis- identification of milk EXOs. cover more operative, secure, and selective additives to produce anti-cancer treatments to enhance their thera- isolation of milk exosomes peutic plausibility and reduce their secondary impacts. Different centrifugation techniques are adopted to In this paper, we overviewed the isolation of CMEXOs isolate milk EXOs as a result of variability in milk con- and their possible therapeutic use as anti-cancer, anti- tent of animals (Yassin et al., 2016; Cintio et al., 2020; inflammatory, anti-angiogenesis, and antioxidant inter - El-Kattawy et al., 2021). Successive centrifugations are mediates. applied to detach the EXOs. First, a low speed centrifu- gation (600–1000× g) is operated to defat the milk and 3 3 milk exosomes precipitate the cells, then 10×10 – 16×10 g is used to According to the procedures of the International So- precipitate milk proteins, and finally, one or numerous 3 3 ciety for Extracellular Vesicles, various immunological, multi-ultracentrifugations at 100×10 – 200×10 g are uti- physical, and physicochemical methods are used to iso- lized to separate the EXOs (Yassin et al., 2016; Badawy et late milk EXOs (Lötvall et al., 2014). EXOs are often al., 2018; El-Kattawy et al., 2021). Commercially, sever- produced by multivesicular bodies which are released via al supply materials are used, including volume-excluding the fusion process with the plasmatic membrane of a cell polymers for isolation of milk EXOs such as ExoQuick (Adriano et al., 2021). Based on the size of secretion fac- (System Biosciences, Palo Alto, CA, USA), containing tors, these released vesicles were named as “exosomes”, PEG 8000 kDa; Total Exosome Isolation (Thermo Fisher which have a size range between 40 and 100 nm, while Scientific, Waltham, MA, USA), comprising polyvi- the other secretions called microvesicles have a 1 μm nyl, polyethylene glycol, and dextrans with a molecular range size (Adriano et al., 2021). weight above 1000 kDa, and their mixtures with other The literature supported that the EXOs contain sev- approaches (Jiao et al., 2020). Additionally, the immuno- eral fragments from the parent cells, comprising differ- magnetic approach is applied to separate EXOs through ent types of RNAs (microRNA, long non-coding RNA, a semipermeable membrane (Bunggulawa et al., 2018). and tRNA), DNA nucleotides, proteins and lipids (Ngu EXOs of high quality could be obtained by ultracentrifu- et al., 2022). For quantification and characterization of gation from different body fluids (milk, seminal plasma, EXOs in milk or other fluids, the most common method oviductal, uterine, blood plasma, urine, lacrymal). In ad- for visualization is via the transmission electron micro- dition, cell culture fluid generally has “nonvesicles” with scopic images (Lázaro-Ibáñez et al., 2014; El-Kattawy 20–100 nm in diameter. These “nonvesicles” are morpho- et al., 2021). A further full clarification of the biogenesis logically accredited to intermediate and very-low-density and structure of exosomes, and other types of extracellu- (40–100 nm) and low-density lipoproteins (20–40 nm) lar vesicles (components) are mentioned in recent papers (Grigor’eva et al., 2017). Transmission electron micros- (Akers et al., 2013). copy can detect the ultrastructure of the cell discriminate EXOs, therefore EXOs were categorized by transmission exosomes in milk of different mammalian species electron microscopy (Badawy et al., 2018; El-Kattawy Many publications have revealed that the goat milk et al., 2021). The previously method detected specific exosomes had the highest loading capacity compared to protein markers such as TSG101 (a truncated 35 KDa other milk exosomes derived from cow and buffaloes protein), which was discovered explicitly for CMEXOs (Ahmed et al., 2022; Yan et al., 2020). Moreover, the use rather than the previously described 43 KDa mammalian The potential therapeutic role of camel milk exosomes 355 one (Yassin et al., 2016). This may be attributed to the many proteins that contribute to various biological func- specific protein in each species and the unique proper - tions in cellular development. ties of CMEXOs. Decisively, several commercial kits are Furthermore, CMEXOs have many specific types of also offered for rapid isolation of EXOs. Among them, proteins that regulate their biological roles, specifically, ThermoFisher, Sigma, FUJIFILM Wako Pure Chemical conveyance of microRNA and adhesion to the target cells Corporation, Nano View Biosciences, Qiagen, Takara including tetraspanins, CD81, CD63, and CD9 as previ- Bio, Biocompare, System Biosciences, IZON, BioCat ously reviewed by Khan et al. (2021). More specifically, GmbH, Miltenyi Biotec, BioVision Inc., etc are well- both lactoferrins and kappa casein mRNAs were noticed known companies. Nonetheless, such kits are used for in camel milk exosomes (Ibrahim et al., 2019). El-Kat- the isolation of EXOs from the cell culture medium and/ tawy et al. (2021) found a significantly higher transcript or human biological fluid. Consequently, in this part, the of kappa casein and lactoferrins genes in colostrum- generally applied isolation approaches are discussed for derived EXOs than in other lactation stage-derivative milk EXOs (Yassin et al., 2016; Badawy et al., 2018; El- EXOs. The higher contents of EXOs in colostrum show Kattawy et al., 2021). There is no one standard technique a greater immune-stimulatory function, which can be for the isolation of EXOs from milk and other biological a part of the innate immunity of infants (Gu et al., 2012). fluids. Successful isolation of milk-derived EXOs is un- On the other hand, no substantial alteration was detected der investigation and there is an unmet need to develop in expression amounts of both kappa casein and lacto- a reliable and reproducible EXOs separation technique. ferrins in late- and mid-lactation milk of camel. The recognition of kappa casein and lactoferrins mRNAs in Bioactive compounds of milk exosomes CMEXOs suggests that their mRNAs are protected from CM comprises the important nutrients as well as degradation by milk RNase (Keller et al., 2011; Yassin et potentially health-beneficial compounds with antihy - al., 2016; Khan et al., 2021). The antioxidant capacity of pertensive, anti-viral, anti-inflammatory, antidiabetic, CM has also been credited to the structural configuration anticarcinogenic, and antioxidant properties (Al-Majali of high amount of beta-caseins content. Upon hydroly- et al., 2007; Izadi et al., 2019; Ghazzawi, 2020). It has sis, the CM beta-caseins are converted to some bioac- been used for preventing or treating some diseases such tive peptides showing helpful antioxidant actions. The as allergies, diabetes, and autism. Also, CM could de- caseins and whey proteins of CM have antioxidant and crease blood cholesterol levels and improve metabolism cytotoxic activities alongside the MCF7 cells. Moreover, because CM is a rich source of the unique combination casein of CM presented plausible apoptotic and anti- of biological compounds such as protein, nucleic acids, HCV activity in HeLa cell lines and human hepatoma and EXOs (Jilo and Tegegne, 2016). Additionally, CM is (Almahdy et al., 2011). EXO RNAs are one of the most full of beta-caseins and many of whey proteins, compris- common components in camel milk, however, there is no ing lactoperoxidase, immunoglobulin, lactoferrin, alpha- published data regarding the characterization and global lactalbumin, and lysozymes (Jilo and Tegegne, 2016). transcriptomic microRNA in this species. Therefore, CM Lactoferrin is the second main protein after casein, it has can be a good environment to explore the profile of EXO numerous biological roles, regulating the iron metabo- microRNAs for using in different biomedical and scien- lism in the body, boosting the immune response, and has tific components. In the other species, various microRNA a critical role in the cellular defense against pathogenic of milk and its EXOs generally have various targets and bacteria through its bactericidal and/or bacteriostatic ac- have a significant role in cell proliferation, pregnancy, tivities (Al-Majali et al., 2007; Jilo and Tegegne, 2016). fetal development, immune system improvement, insulin CMEXOs have received more attention in the last years resistance, sugar metabolism and inflammation (Sedykh due to their health-promoting properties. The biologi- et al., 2020). In addition, natural lipid compounds of cal compounds of milk EXOs can meaningfully affect milk EXOs are more well-matched and less toxic than biomolecules delivery. Therefore, a comprehensive scru- synthetic liposomes for uses as drug delivery vehicles tiny of the content of milk EXOs that are also termed (Elsharkasy et al., 2020). According to the literature, pro- “exosomics” (by analogy with genomics, transcriptome, teins of milk EXOs have significant roles in formation of proteomics, and other -omics technologies), is compul- cytoskeleton protein, homeostasis, cell development, cel- sory (Yassin et al., 2016). Al-Majali et al. (2007) sug- lular defense, and signal transduction etc., but this kind gested that camel milk EXOs proteins such as TSG101 of roles for CM proteins have been not illustrated and (a 35 KDa of molecular weight) could be accredited with need further exploration. various biological roles in cell development and regula- In a recent study, He et al. (2021) performed the prot- tion pattern. To date, the biological function of TSG101 eomic analysis on Bactrian camel milk and found higher is ambiguous and needs further explorations to demon- contents of lactoferrin, lactoperoxidase isoform 1 prepro- strate the molecular purpose. protein, and FABP domain-containing protein as well as Studies revealed that the biological properties of κ-casein, alpha-1-acid glycoprotein and peptidoglycan- CM proteins might be attributed to their high content of recognition protein. It has been reported that EXOs de- exosomes (extracellular nanovesicles) (Gu et al., 2012; pending on their source can be divided into virtuous and Khan et al., 2021). Generally, the milk exosomes have malicious exosomes. Exosomes derived from CMEXOs 356 N.A. Althobaiti et al. are an example of virtuous EXOs which exhibited a su- breast tumor development. The cancer therapy might be perior anticancer effect (inhibition of the growth and associated with higher apoptosis pathways; eminent at- metastatic potential of MCF7 cells) to that of CM (Bad- tenuation in angiogenesis, inflammation, and metastasis; awy et al., 2018). as well as notable reduction of oxidative stress synthe- The phospholipid membrane encircling EXOs de- sis, and greater immune response (Badawy et al., 2018). fends exosomal substances (DNA, RNA, protein) from Furthermore, Almahdy et al. (2011) and Korashy et al. degradation by digestive enzymes and acidic conditions (2012) showed that the administration of whole CM (Record et al., 2014). This gastric constancy may eluci- led to high cytotoxic influence on MCF7 tumor cells in date anti-cancer property for EXOs when it is adminis- a dose-dependent manner. Earlier investigations showed tered orally compared to CM used alone. Besides, the that CM triggered apoptosis of tumor cell line (type absorbed EXOs will take a few hours to arrive at the cel- MCF7) (Korashy et al., 2012) and other cell lines com- lular target and may undergo dilution, by circulation all prising BT-474, Hela, and HepG2, via the same molecu- over the body, before contacting the tissue tumors. This lar pathway (Almahdy et al., 2011; Hasson et al., 2015). shows that local injection of EXOs in tumor tissues has This stimulatory machinery of CM could be connected better consequences than oral administration. However, to high contents of immunoproteins such as lactoferrin, regarding the biological properties of EXO, it is urgent α-lactalbumin, and other bioactive compounds (for ex- to do more explorations, especially in CMEXOs to detect ample exosomes) which have been unclarified (Almahdy different EXOs of this species which could be used in et al., 2011). Badawy et al. (2018) reported that the apop- several biomedical fields. tosis indices such as both caspase 3, and Bax transcripts were significantly increased and decreased Bcl2 gene ex- camel milk exosomes as: pression, as well as high DNA fragmentation was record- Anti-cancer agent ed in EXOs treatment compared to CM treatment, indi- The potential therapeutic role of CMRXOs in cancer cating that CM could induce apoptosis, at least in part, remains an emerging scope of research. A huge number interceded by exosomes. Totally, the inhibitory upshot of of reports have illustrated that CMEXOs have capabil- CM and its EXOs on tumor cells is owing to the antioxi- ity to prevent the progression of tumor cancer in several dative properties and induction of the apoptosis process. in vivo or in vitro experiments (Badawy et al., 2018; El- And the anti-cancer activity of CMEXOs is related to the Kattawy et al., 2021). Studies have indicated that cancer attendance of defensive proteins such as lysozyme, lacto- cells survive via different molecular mechanisms such as ferrin and lactoperoxidase and other immunological in- restraining of apoptosis, immunosuppression, and stimu- dices such as IgG, and IgA (Jilo and Tegegne, 2016). As lation of inflammation (Wong, 2011; Diakos et al., 2014; previously clarified, the high levels of lactoferrin in CM Badawy et al., 2018; El-Kattawy et al., 2021). It is iden- could prevent the propagation of colorectal cancer cells tified that a conceivable part of CMEXOs inhibits the and have antioxidant and DNA injury-repressing charac- growth and proliferation of MCF7 cells and diminishes teristics in cancerous cells (Habib et al., 2013). Figure 1. The method for isolation and purification of camel milk-derived exosomes using ultra-centrifugation method The potential therapeutic role of camel milk exosomes 357 Figure 2. The biological components of camel milk documented in the present article Lately, Badawy et al. (2021) showed a significantly Anti-inflammatory activity superior DNA impairment catalogue in tumor tissues In addition to anti-cancer property, CMEXOs also such as HepaRG cells after treating with CMEXOs. In exhibited high capability of anti-inflammatory activity. this case, CMEXOs triggered HepaRG cells dispersion Augmented amounts of pro-inflammatory cytokines like via apoptosis pathways that can be associated with the TNFα, TGFβ1, IL1β, and NFkB, are indispensable for potentiality of exosomes-treated cells to exhibit a sig- tumor initiation, metastasis and attack (Han et al., 2014; nificant increment in the expression of caspase 3 and Elgazar et al., 2018). In a study on rats, Arab et al. (2018) Bax and a significantly lower expression of Bcl2 than reported that CM could restore the inflammatory environ- the control group (Badawy et al., 2021). The apoptosis ments connected with diabetes and 5-fluorouracil-incited induction approach is one of the main innovative tools renal impairment in rats via restraining the pro-inflam- for cancer therapy. Tumor cells sustain their viability via matory cytokines statement. Additionally, Badawy et al. induction of inflammation, and suppression of apoptosis (2018) described that the anti-inflammatory property of and angiogenesis in the tumor microenvironment (Dia- CMEXOs exhibited superior impact to CM, as docu- kos et al., 2014). Likewise, CMEXOs showed a potent mented by downregulation of the inflammation-related apoptotic property on HepaRG as discovered by a sub- genes, interleukin 1 beta (IL1β), and nuclear factor-kappa stantial upregulation of Bax and caspase3 and a notewor- B (NFκB) in cancer tissues. In parallel with the previ- thy downregulation of Bcl2, with noteworthy fluctuations ous results, the anti-inflammatory effect of camel urine in colostrum-treated cells. Parallel apoptotic alterations was evidenced on the tumor cell such MCF7 cells in rats were observed in the subsequent treatment of HepG2 and (Romli et al., 2017). Linking with the biomolecules of MCF7 (Korashy et al., 2012; Badawy et al., 2018). In camel urine and milk, it is conceivable that EXOs are in vivo and in vitro studies, TR35, which was separated the main players in this achievement as they are formed from Xinjiang Bactrian camel milk, could meaningfully by almost all cell subgroups and plentifully isolated or constrain Eca109 cell propagation and persuade its ap- derived from the urine or/and milk (Romli et al., 2017). optosis (showed by caspase-3 activity, MTT assessment, Ibrahim et al. (2019) clarified that the CMEXOs dis- and Annexin V-FITC) (Yang et al., 2019). Moreover, play anti-inflammatory activities against tumors through TR35 could constrain the development and progres- a substantial augment in the expression of TNF-α and sion of xenografted tumors in nude mice without loss in IL-6 as well as a noteworthy reduction in the expression body weight (Yang et al., 2019). The tiny proteins iso- abundance of IFN-γ after administration with cyclophos- lated from CM could be used as a potential therapeutic phamide as related to the control rats (Ibrahim et al., ingredient according to its historical anti-cancer activity. 2019). NF-κB is one of the vital transcription features Although several investigations have explored the use that helps the expression of various genes of immune and of the whole CM in different types of cancer therapies, inflammatory responses such as IL-6 and TNF-α (Mauriz further explorations are needed to discover the effect of et al., 2013). A previous description documented the anti- nano biomolecules isolated from CM that can be used in inflammatory property for CMEXOs via the downregula- modern therapy. tion of NF-κB and IL-1β in rat’s mammary tumor tissues 358 N.A. Althobaiti et al. (Badawy et al., 2018). Certain in vivo studies revealed that synthesis of IFN-γ in the attendance of allogeneic human camel undenatured whey proteins restricted the extended T-lymphocytes (Actor et al., 2009), and it can trigger the inflammation in diabetic rats via the decrease in the con- synthesis of IL-12 by antigen offering cells, which in turn centrations of serum IL-1β, IL-6 and TNF-α (Ebaid et improves IFN-γ synthesis and initiates Th1 cell growth al., 2013). Moreover, CM repressed renal inflammation (Actor et al., 2009; Daneshmandi et al., 2017). persuaded by 5-fluorouracil via suppression of IL-1β, Casein-loaded dendritic cells could somewhat pro- and TNF-α as well as repressed NF-κB stimulation in voke the Th1 cell response via the improvement of Wistar rats (Arab et al., 2018). Numerous bioactive pep- lymphocyte propagation and inducement of IFN-γ syn- tides are showing various health-promoting purposeful thesis (Daneshmandi et al., 2017). Recently, CMEXOs properties in milk proteins. Consequently, camel unde- showed a significant modulation in the inflammation natured whey protein can boost the immune response via process of cancer cells through significant downregula- the augmentation of mRNA expression of both IFN-γ and tion of inflammatory-related genes such as TNFα, NFkB, IL-2 in rats after four months of streptozotocin-induced TGFβ1, and Cox2 in HepaRG cells (Badawy et al., diabetes (Ebaid, 2014). Among the chronic hepatitis B 2021). In rats bearing breast cancer, it has been reported patients, it was reported that the levels of serum IFN-γ in that the CMEXOs have a potential role in repressing the camel milk-drinking patients were meaningfully greater pro-inflammatory cytokines (Badawy et al., 2018). Col- than those in patients who did not receive CM (Saltanat lectively, previous evidence clarified the pulsative role et al., 2009). This might be showing the potential anti- of CMEXOs in reduction of the pro-inflammatory cy- viral activity of CM or its bioactive peptides. Moreover, tokines response in different in vitro and in vivo models, lactoferrin supports the development of dendritic cells by however, further explorations are essential to elucidate augmenting their capability to induce propagation and more molecular mechanisms behind their effects. Figure 3. Therapeutic potential of camel milk exosomes with potent anticancer, antioxidant, anti-inflammatory, and anti-angiogenesis effects Figure 4. The mechanistic roles of exosomes isolated from camel milk The potential therapeutic role of camel milk exosomes 359 Anti-angiogenesis activity reveals a potent anti-angiogenetic activity of EXOs for the Generation of new development of the vascular net- incorporated therapy compared to a single treatment. work is vital since the metastatic spread, proliferation of cancer cells be contingent on a satisfactory supply of Immuno-modulatory activity nutrients and oxygen and the elimination of waste prod- The immune system plays an essential function in dif- ucts (Nishida et al., 2006). Angiogenesis is controlled by ferent cellular development and growth in living organ- both inhibitor and activator molecules. The detection of isms as well as is responsible for induction of different angiogenic inhibitors or anti-angiogenesis can support diseases. This feature is primarily intermediated by sev- a decrease in both mortality and morbidity from carcino- eral immune cells: T cells, including CD4+ helper, CD8+ mas. Milk EXOs are hopeful candidates in emerging new cytotoxic, and natural killer (NK) cells (Grivennikov et therapeutic tactics to exhibit different anti-angiogenesis al., 2010). CM proteins exhibited better immune respons- activities. Various diseases have been reported to have es connected with the enhancement of infectious diseas- higher activity of vascular endothelial growth factor es. In this regard, Badawy et al. (2018) clarified that ad- (VEGF) (Nishida et al., 2006). A substantial correlation ministration of CM and its EXOs significantly enhanced between the expression of VEGF and prognosis has been the immune criteria such as NK+, CD8+, and CD4+ T described in several types of cancers and other diseases cells in the spleen of rats. Higher T cell differential de- (Badawy et al., 2018, 2021). Administration of CMEXOs tected in animals could reflect better immunity and lesser led to substantial downregulation of angiogenesis-related tumor sizes. Likewise, various articles documented that gene (VEGF) expression in tumor tissues as compared to cancer patients with high levels of T cells such as NK+, the tumor group (Badawy et al., 2018, 2021). This fea- CD8+, and CD4+ cells have superior prognosis and sur- ture could suggest the inhibitory property of CMEXOs vival rates (Malmberg et al., 2008; Laghi et al., 2009). on tumor environment angiogenesis. Besides, CMEXOs Improving the levels of the CD4+ cell could constrain also exhibited an antimetastatic activity on MCF7 cells the development and progress of tumors via suppression as shown by an in vitro wound healing test, which dis- of inflammation interrelated cytokines (Chen et al., 2021; covered inferior cell migration percentage with superior Lei et al., 2022; Hou et al., 2022). On the other hand, effect when EXOs were employed (Badawy et al., 2021). CD8+ cells can reduce cancer cells directly or indirectly To further authenticate the antimetastatic probability of by augmenting the synthesis of other cytotoxic cytokines CM and its EXOs, the metastasis-related genes (ICAM1 (Grivennikov et al., 2010). and MMP9) were examined. As predicted, the applica- Surprisingly, administration of CM caused superior tion of CM and its EXOs meaningfully downregulated percentages of NK1, CD8+, and CD4+ compared to co- the transcript of MMP9 and ICAM1 in tumor tissues with administration of EXOs. This might be explained by the the greatest enhancement by EXOs administration (Bad- fact that the CM has other immune-stimulant compounds awy et al., 2021). than those existing in their EXOs. The immune-stimula- Similarly, studies have indicated that the inferior ex- tory activity of CM could be attributed to its content of pression of the metastasis-related gene such as ICAM1 immunoglobulins, casein, lysozyme, lactoferrin, and lac- was observed in tumor tissues of mice subsequently toperoxidase (El Agamy et al., 1992). Furthermore, CM treated with camel urine (Romli et al., 2017) or CM or its comprises unique nanobodies with variable heavy chain, EXOs (Badawy et al., 2021). Furthermore, the upregula- and has no typical light chains like normal antibodies. tion of ICAM1 was related to the invasive and metastatic These very tiny nanobodies presented in CM IgGs can capability of several cancer cells (breast cancer and other easily penetrate the cell membrane of any cells (includ- carcinoma cells). As previously indicated, chemokine in ing cancer cells) and localize intracellularly to achieve the tumor microenvironment shows a significant function different immune-stimulatory activities (Korashy et al., in the invasion and migration of carcinoma cells (Fernan- 2012; Sedykh et al., 2020; Khan et al., 2021). Conse- dis et al., 2004). Badawy et al. (2018) confirmed that CM quently, it was supposed that CMEXOs can transport or and its EXOs are new disruptors for chemotaxis-based be a ligand for these immune-stimulant components. This migration of MCF7 cells (Badawy et al., 2018). HepaRG mechanistic insight needs further explorations to be clari- cells treated with EXOs displayed significant downregu- fied. Ibrahim et al. (2019) reported that CM and its EXOs lation of VEGF, which indicates the aptitude of EXOs to moderately enhanced lymphocyte profile of rats treated constrain the angiogenesis process in the tumor tissues. with cyclophosphamide. The same authors indicated that A comparable anti-angiogenesis property was described the rats co-treated with CMEXOs and cyclophosphamide for EXOs derived from mesenchymal stem cells in the exhibited significant increases in both CD8+, CD4+ HCC microenvironment, unlike those originating from cells and T-lymphocyte relative to the cyclophospha- cancer stem cells which presented angiogenic activ- mide group. Authors suggested that the immunomodu- ity (Alzahrani et al., 2018). Mice co-administered with latory effects of CM and its EXO may be accredited to CMEXOs and some phytochemical displayed inferior ex- their contents of caseins and lactoferrin. In addition, pression of both VEGF and Bcl2 and superior expression of they confirmed fruitful delivery of CMEXO kappa Cas3 and Bax genes in MCF7 xenograft compared to those casein and lactoferrin mRNAs into the rat tissues via administered with each alone (Badawy et al., 2021). This separation and sequencing of camel kappa casein and 360 N.A. Althobaiti et al. lactoferrin from the spleen of the treated rats. Danesh- activity but also boost antioxidant status that decreases mandi et al. (2017) showed that casein-loaded dendrit- OS impairment triggered by tamoxifen and/or cancer ic cells could improve the Th1 immune status in mice. problem (Badawy et al., 2021). The activation of NF-κB, lactoferrin and MAP kinase can stimulate the proliferation, maturation, differen- conclusions and future perspective tiation, activation of different immune cells (Actor et An increasing body of evidence has discovered the al., 2009). It has been revealed that lactoferrin shows potential role of CMEXOs in anti-cancer, anti-inflam - a beneficial role in the reconstitution of the cellular matory, antioxidant and anti-microbial properties which immune response as it promotes cellularity and en- could be used in treating several ailments, enhancing hancement of CD4+, CD3+ lymphocytes in the spleen health, as well as contributing as a new therapeutic win- and reinstates the host T cell compartment (Artym et dow. Investigations exhibited that EXOs derived from al., 2003; Bhattacharyya et al., 2014) Besides, lacto- CM have more stable form under adverse environments, ferrin infusion produced a strong Th1- response and such as freezing-thawing, low pH, and heating, and can encouraged the activation of T-lymphocytes such as be easily absorbed through the intestinal barriers to the CD8+ and CD4+ in tumor-bearing models. circulation system. In this paper, the potential roles of CMEXOs including anti-cancer, anti-inflammatory, an- Antioxidant activity tioxidant were reviewed. Moreover, the CMEXOs can Oxidative stress (OS) can be defined as a dispar - also serve as oral delivery vehicles for chemotherapeu- ity between the free radical production and the body’s tic mediators and further amendment of the EXOs with capability to combat against the destructive impacts of a tumor-targeting ligand allows the targeted delivery of antioxidants. The subsequent difference in homeostasis drugs to the tumor sites. Furthermore, the significance on the explanation of excessive reactive oxygen species of these novel achievements of CMEXOs is constructed (ROS) induces cell impairment and is one of the leading through their advantages over prevailing therapeutic reasons for emerging ailments such as gastric/colorectal opportunities. Developments in technical isolation of cancer, inflammatory bowel disease, and gastroduode- EXOs and a greater understanding of the mechanistic nal ulcers (Bhattacharyya et al., 2014). Badawy et al. details will support the medical community to extend (2018) indicated that CM and its EXO significantly en- us a safe and effective tool for resolving public health hanced the synthesis of antioxidant enzymes (catalase problems on a broader scale. In addition, it would be (CAT), superoxide dismutase (SOD), and glutathione significant to create a standardized and cost-effective peroxidase (GPX)), and reduced the content of lipid technique to isolate, purify, and employ EXOs from peroxidation indicator malondialdehyde (MDA), and CM to safeguard the quality of the EXOs for industrial the transcript of the OS indicator iNOS in tumor mat- and clinical uses. Future reports are essential to vali- ters as relative to the tumor group. This suggests that date how individual bioactive compounds within EXOs OS/antioxidant pathway may be complicated with the employ biological roles, including antitumor, antidia- repressive role of CM and its EXOs on tumor devel- betic, anti-obesity, and antihypertensive properties to opment. It was revealed that co-treatments of cyclo- elucidate any plausible undesirable effects upon EXO phosphamide with CM or CMEXOs meaningfully de- remediation. creased the lipid peroxidation signal MDA amount and increased the antioxidant enzymes GPX, CAT, and SOD funding levels as relative to the cyclophosphamide -treated rats This work was funded by the National Key Re- (Ibrahim et al., 2019). CM enhanced the antioxidant re- search and Development Program of China (No. sponse in renal tissues and repressed the OS triggered 2018YFD0501700), National Modern Agricultural In- via 5-fluorouracil induction in Wistar rats (Arab et al., dustry Special Program (No. CARS-37), the National 2018). Furthermore, a prior investigation verified the Natural Science Foundation of China (31972994), the antioxidant capabilities of various peptides primarily Agricultural Science and Technology Innovation and attained from caseins of the CM (Ibrahim et al., 2019). Transformation Project of Shaanxi Province (NYKJ- Synergistic effect of EXOs and some phytochemical 2018-LY09) and the Key Research and Development showed significantly lower MDA, and higher CAT and Program of Ningxia (Grant No. 2019BEF02004). GPx activities in hepatic tissues than in control mice (Badawy et al., 2021). This potential ability of EXOs to acknowledgments bring the apoptosis to avoid angiogenesis and invasion We thank Shaqra University and the Scientific Re- in MCF7 cells could be a beneficial approach in treat- search Deanship of Shaqra University, KSA for their ing some diseases. Remarkably, the combined therapy kind support to Althobaiti N.A., BinMowyna M.N., and using phytochemical (tamoxifen, and hesperidin) and Aldawsari R.D. EXOs also restored the disrupted OS/antioxidant status to normal levels related to that of the standard control. competing interests Inclusively, tamoxifen, hesperidin and EXOs mixture The authors declare that they have no competing inte- treatment can not only potentiate tamoxifen anti-cancer rests. The potential therapeutic role of camel milk exosomes 361 refernces Diakos C.I., Charles K.A., McMillan D.C., Clarke S.J. (2014). Cancer- related inflammation and treatment effectiveness. Lancet. Oncol., Abels E.R., Breakefield X.O. (2016). Introduction to extracellular 15: e493–e503. vesicles: Biogenesis, RNA cargo selection, content, release, and Ebaid H. (2014). Promotion of immune and glycaemic functions in uptake. Cell. Mol. Neurobiol., 36: 301–312. streptozotocin-induced diabetic rats treated with un-denatured Actor J.K., Hwang S.A., Kruzel M.L. (2009). Lactoferrin as a natural camel milk whey proteins. Nutr. Metab., 11: 1–13. immune modulator. Curr. Pharm. Design, 15: 1956–1973. Ebaid H., Ahmed O.M., Mahmoud A.M., Ahmed R.R. (2013). Limit- Adriano B., Cotto N.M., Chauhan N., Jaggi M., Chauhan S.C., Yallapu ing prolonged inflammation during proliferation and remodeling M.M. (2021). Milk exosomes: Nature's abundant nanoplatform phases of wound healing in streptozotocin-induced diabetic rats for theranostic applications. Bioactive Mat., 6: 2479–2490. supplemented with camel undenatured whey protein. BMC Im- Ahmed F., Tamma M., Pathigadapa U., Reddanna P., Yenuganti V.R. munol., 14: 31–31. (2022). Drug loading and functional efficacy of cow, buffalo, and El Agamy E.I., Ruppanner R., Ismail A., Champagne C.P., Assaf R. goat milk-derived exosomes: a comparative study. Mol. Pharmac., (1992). Antibacterial and antiviral activity of camel milk protec- 19: 763–774. tive proteins. J. Dairy Res., 59: 169–175. Akers J.C., Gonda D., Kim R., Carter B.S., Chen C.C. (2013). Biogen- Elgazar A.A., Selim N.M., Abdel-Hamid N.M., El-Magd M.A., El esis of extracellular vesicles (EV): exosomes, microvesicles, retro- Hefnawy H.M. (2018). Isolates from Alpinia officinarum Hance virus-like vesicles, and apoptotic bodies. J. Neurooncol., 113: 1–11. attenuate LPS-induced inflammation in HepG2: Evidence from in Ali M.Z., Qureshi A.S., Usman M., Kausar R., Ateeq M.K. (2017). silico and in vitro studies. Phyto. Res., 32: 1273–1288. Comparative effect of camel milk and black seed oil in induced El-Kattawy A.M., Algezawy O., Alfaifi, M.Y., Noseer E.A., Hawsawi diabetic female albino rats. Pak. Vet. J., 37: 293–298. Y.M., Alzahrani O.R., Algarni A., Kahilo K.A., El-Magd M.A. Al-Majali A.M., Ismail Z.B., Al-Hami Y., Nour A.Y. (2007). Lactofer- (2021). Therapeutic potential of camel milk exosomes against rin concentration in milk from camels (Camelus dromedarius) with HepaRG cells with potent apoptotic, anti-inflammatory, and anti- and without subclinical mastitis. Int. J. App. Res. Vet. Med., 5: 120. angiogenesis effects for colostrum exosomes. Biomed. Pharma., Almahdy O., El-Fakharany E.M., El-Dabaa E., Redwan E.M. (2011). 143: 112220. Examination of the activity of camel milk casein against hepatitis Elsharkasy O.M., Nordin J.Z., Hagey D.W., de Jong O.G., Schiffelers C virus (genotype-4a) and its apoptotic potential in hepatoma and R.M., Andaloussi S.E., Vader P. (2020). Extracellular vesicles as hela cell lines. Hepat Mon., 11: 724–730. drug delivery systems: Why and how? Advan. Drug Del. Rev., Alzahrani F.A., El-Magd M.A., Abdelfattah-Hassan A., Saleh A.A., 159: 332–343. Saadeldin I.M., El-Shetry E.S., Badawy A.A., Alkarim S. (2018). Fernandis A.Z., Prasad A., Band H., Klösel R., Ganju R.K. (2004). Potential effect of exosomes derived from cancer stem cells and Regulation of CXCR4-mediated chemotaxis and chemoinvasion MSCs on progression of DEN-induced HCC in rats. St. Cell. Int., of breast cancer cells. Oncogene, 23: 157–167. 2018: 8058979. Ghazzawi H. (2020). Health-improving and disease-preventing po- Anand P.K. (2010). Exosomal membrane molecules are potent im- tential of camel milk against chronic diseases and autism: cam- mune response modulators. Commun. Integr. Biol., 3: 405–408. el milk and chronic diseases. Handbook of Research on Health Arab H.H., Salama S.A., Maghrabi I.A. (2018). Camel milk amelio- and Environmental Benefits of Camel Products. IGI Global, rates 5-fluorouracil-induced renal injury in rats: targeting MAPKs, pp. 155–184. NF-κB and PI3K/Akt/eNOS pathways. Cell. Physiol. Biochem., Grigor’eva A., Dyrkheeva N., Bryzgunova O., Tamkovich S., Ch- 46: 1628–1642. elobanov B., Ryabchikova E. (2017). Contamination of exosome Artym J., Zimecki M., Kruzel M.L. (2003). Reconstitution of the cel- preparations, isolated from biological fluids. Biochemistry (Mos - lular immune response by lactoferrin in cyclophosphamide-treat- cow), Suppl. Series Biomed. Chem., 11: 265–271. ed mice is correlated with renewal of T cell compartment. Immu- Grivennikov S.I., Greten F.R., Karin M. (2010). Immunity, inflamma - nobiology, 207: 197–205. tion, and cancer. Cell, 140: 883–899. Badawy A.A., El-Magd M.A., AlSadrah S.A. (2018). Therapeutic ef- Gu Y., Li M., Wang T., Liang Y., Zhong Z., Wang X., Zhou Q., Chen fect of camel milk and its exosomes on MCF7 cells in vitro and in L., Lang Q., He Z., Chen X., Gong J., Gao X., Li X., Lv X. (2012). vivo. Integr. Cancer Ther., 17: 1235–1246. Lactation-related microRNA expression profiles of porcine breast Badawy A.A., Othman R.Q.A., El-Magd M.A. (2021). Effect of com- milk exosomes. PLoS One, 7: e43691–e43691. bined therapy with camel milk-derived exosomes, tamoxifen, and Habib H.M., Ibrahim W.H., Schneider-Stock R., Hassan H.M. (2013). hesperidin on breast cancer. Mol. Cell. Toxicol, 1–10. Camel milk lactoferrin reduces the proliferation of colorectal can- Baier S.R., Nguyen C., Xie F., Wood J.R., Zempleni J. (2014). Mi- cer cells and exerts antioxidant and DNA damage inhibitory ac- croRNAs are absorbed in biologically meaningful amounts from tivities. Food Chem., 141: 148–152. nutritionally relevant doses of cow milk and affect gene expres- Han J., Bae S.Y., Oh S.J., Lee J., Lee J.H., Lee H.c., Lee S.K., Kil sion in peripheral blood mononuclear cells, HEK-293 kidney cell W.H., Kim S.W., Nam S.J. (2014). Zerumbone suppresses IL-1β- cultures, and mouse livers. J. Nutr., 144: 1495–1500. induced cell migration and invasion by inhibiting IL-8 and MMP- Bhattacharyya A., Chattopadhyay R., Mitra S., Crowe S.E. (2014). 3 expression in human triple-negative breast cancer cells. Phyto. Oxidative stress: an essential factor in the pathogenesis of gastro- Res., 28: 1654–1660. intestinal mucosal diseases. Physiol. Rev., 94: 329–354. Hasson S.S., Al-Busaidi J.Z., Al-Qarni Z.A., Rajapakse S., Al-Bahlani Bunggulawa E.J., Wang W., Yin T., Wang N., Durkan C., Wang Y., S., Idris M.A., Sallam T.A. (2015). In vitro apoptosis triggering in Wang G. (2018). Recent advancements in the use of exosomes as the BT-474 human breast cancer cell line by lyophilised camel’s drug delivery systems. J. Nanobiotech., 16: 1–13. milk. Asian. Pac. J. Cancer Prev., 16: 6651–6661. Chen X., Kang R., Kroemer G., Tang D. (2021). Broadening hori- He J., Chen Q., Yi L., Ming L., Ji R. (2021). Proteomics and micro- zons: the role of ferroptosis in cancer. Nat. Rev. Clin. Oncol., 18: structure profiling of Bactrian camel milk protein after homogeni - 280–296. zation. LWT, 152: 112287. Cintio M., Polacchini G., Scarsella E., Montanari T., Stefanon B., Hou C.X., Sun N.N., Han W., Meng Y., Wang C.X., Zhu Q.H., Tang Colitti M. (2020). MicroRNA milk exosomes: From cellular regu- Y.T., Ye J.H. (2022). Exosomal microRNA-23b-3p promotes tu- lator to genomic marker. Animals, 10: 1126. mor angiogenesis and metastasis by targeting PTEN in salivary Colombo M., Raposo G., Théry C. (2014). Biogenesis, secretion, and adenoid cystic carcinoma. Carcinogenesis, 43: 682–692. intercellular interactions of exosomes and other extracellular ves- Ibrahim H.M., Mohammed-Geba K., Tawfic A.A., El-Magd M.A. icles. Ann. Rev. Cell Dev. Biol., 30: 255–289. (2019). Camel milk exosomes modulate cyclophosphamide-in- Daneshmandi S., Nourizadeh M., Pourpak Z., Pourfathollah A.A. duced oxidative stress and immuno-toxicity in rats. Food Func., (2017). Eliciting Th1 immune response using casein (alpha s1)- 10: 7523–7532. loaded dendritic cells. Iran. J. Aller. Asth. Immunol., 159–168. Iigo M., Alexander D.B., Long N., Xu J., Fukamachi K., Futakuchi M., 362 N.A. Althobaiti et al. Takase M., Tsuda H. (2009). Anticarcinogenesis pathways acti- of camel milk in children with autism: its impact on serum levels vated by bovine lactoferrin in the murine small intestine. Biochi- of vasoactive intestinal peptide. Int. J. Med. Sci. Clin. Invent., 8: mie, 91: 86–101. 5698–5707. Izadi A., Khedmat L., Mojtahedi S.Y. (2019). Nutritional and thera- Ngu A., Wang S., Wang H., Khanam A., Zempleni J. (2022). Milk peutic perspectives of camel milk and its protein hydrolysates: exosomes in nutrition and drug delivery. Am. J. Physiol.-Cell A review on versatile biofunctional properties. J. Func. Foods, 60: Physiol., 322: C865–C874. 103441. Nishida N., Yano H., Nishida T., Kamura T., Kojiro M. (2006). Angio- Jiao R., Sun S., Gao X., Cui R., Cao G., Wei H., Wang S., Zhang Z., genesis in cancer. Vasc. Heal. Risk Manag., 2: 213–219. Bai H. (2020). A polyethylene glycol-based method for enrich- Record M., Carayon K., Poirot M., Silvente-Poirot S. (2014). Exo- ment of extracellular vesicles from culture supernatant of human somes as new vesicular lipid transporters involved in cell–cell ovarian cancer cell line A2780 and body fluids of high-grade se - communication and various pathophysiologies. Biochim. Bio- rous carcinoma patients. Canc. Manag. Res., 12: 6291. phys. Acta -Mol. Cell Biol. Lip., 1841: 108–120. Jilo K., Tegegne D. (2016). Chemical composition and medicinal val- Romli F., Abu N., Khorshid F.A., Syed Najmuddin S.U.F., Keong ues of camel milk. Int. J. Res. Stud. Biosci., 4: 13–25. Y.S., Mohamad N.E., Hamid M., Alitheen N.B., Nik Abd Rahman Keller S., Ridinger J., Rupp A.-K., Janssen J.W., Altevogt P. (2011). N.M.A. (2017). The growth inhibitory potential and antimetastatic Body fluid derived exosomes as a novel template for clinical diag - effect of camel urine on breast cancer cells in vitro and in vivo. nostics. J. Transl. Med., 9: 1–9. Integr. Cancer Ther., 16: 540–555. Khan M.Z., Xiao J., Ma Y., Ma J., Liu S., Khan A., Khan J.M., Cao Z. Saltanat H., Li H., Xu Y., Wang J., Liu F., Geng X.H. (2009). The influ - (2021). Research development on anti-microbial and antioxidant ences of camel milk on the immune response of chronic hepatitis properties of camel milk and its role as an anti-cancer and anti- B patients. Ch. J. Cell. Mol. Immunol., 25: 431–433. hepatitis agent. Antioxidants, 10: 788. Santos-Coquillat A., González M.I., Clemente-Moragón A., González- Korashy H.M., Maayah Z.H., Abd-Allah A.R., El-Kadi A.O., Alhaider Arjona M., Albaladejo-García V., Peinado H., Muñoz J., Embún A.A. (2012). Camel milk triggers apoptotic signaling pathways P.X., Ibañez B., Oliver E., Desco M., Salinas B. (2022). Goat milk in human hepatoma HepG2 and breast cancer MCF7 cell lines exosomes as natural nanoparticles for detecting inflammatory pro - through transcriptional mechanism. J. Biomed. Biotechnol., 2012: cesses by optical imaging. Small, 18: 2105421. 593195. Sedykh S., Kuleshova A., Nevinsky G. (2020). Milk exosomes: Perspec- Laghi L., Bianchi P., Miranda E., Balladore E., Pacetti V., Grizzi F., Al- tive agents for anticancer drug delivery. Int. J. Mol. Sci., 21: 6646. lavena P., Torri V., Repici A., Santoro A., Mantovani A., Roncalli Shariatikia M., Behbahani M., Mohabatkar H. (2017). Anticancer ac- M., Malesci A. (2009). CD3+ cells at the invasive margin of deep- tivity of cow, sheep, goat, mare, donkey and camel milks and their ly invading (pT3–T4) colorectal cancer and risk of post-surgical caseins and whey proteins and in silico comparison of the caseins. metastasis: a longitudinal study. Lancet. Oncol., 10: 877–884. Mol. Biol. Res. Commun., 6: 57–64. Lázaro-Ibáñez E., Sanz-Garcia A., Visakorpi T., Escobedo-Lucea C., Swelum A.A.A., Hashem N.M., Abo-Ahmed A.I., Abd El-Hack M.E., Siljander P., Ayuso-Sacido Á., Yliperttula M. (2014). Different Abdo M. (2020). The role of heat shock proteins in reproductive gDNA content in the subpopulations of prostate cancer extracel- functions. In: Heat shock proteins, Asea A.A.A., Kaur P. (eds). lular vesicles: apoptotic bodies, microvesicles, and exosomes. Springer Nature Switzerland, pp. 407–427. Prostate, 74: 1379–1390. Wong R.S. (2011). Apoptosis in cancer: from pathogenesis to treat- Lei G., Zhuang L., Gan B. (2022). Targeting ferroptosis as a vulner- ment. J. Exper. Clin. Cancer Res., 30: 1–14. ability in cancer. Nat. Rev. Cancer, 22: 381–396. Yan F., Zhong Z., Wang Y., Feng Y., Mei Z., Li H., Chen X., Cai L., Li Lötvall J., Hill A.F., Hochberg F., Buzás E.I., Di Vizio D., Gardiner C. (2020). Exosome-based biomimetic nanoparticles targeted to C., Gho Y.S., Kurochkin I.V., Mathivanan S., Quesenberry P., Sa- inflamed joints for enhanced treatment of rheumatoid arthritis. J. hoo S., Tahara H., Wauben M.H., Witwer K.W., Théry C. (2014). Nanobiotechnol., 18: 1–15. Minimal experimental requirements for definition of extracellular Yang J., Dou Z., Peng X., Wang H., Shen T., Liu J., Li G., Gao Y. vesicles and their functions: a position statement from the Inter- (2019). Transcriptomics and proteomics analyses of anti-cancer national Society for Extracellular Vesicles. J. Extracell. Vesicles, mechanisms of TR35 – An active fraction from Xinjiang Bac- 3: 26913. trian camel milk in esophageal carcinoma cell. Clin. Nutr., 38: Malmberg K.-J., Bryceson Y.T., Carlsten M., Andersson S., Björklund 2349–2359. A., Björkström N.K., Baumann B.C., Fauriat C., Alici E., Dilber Yassin A.M., Abdel Hamid M.I., Farid O.A., Amer H., Warda M. M.S., Ljunggren H.-G. (2008). NK cell-mediated targeting of hu- (2016). Dromedary milk exosomes as mammary transcriptome man cancer and possibilities for new means of immunotherapy. nano-vehicle: Their isolation, vesicular and phospholipidomic Cancer Immunol. Immunother., 57: 1541–1552. characterizations. J. Adv. Res., 7: 749–756. Mauriz J.L., Collado P.S., Veneroso C., Reiter R.J., González-Gallego Zheng N., Min L., Li D., Tan S., Gao Y., Wang J. (2022). Occurrence J. (2013). A review of the molecular aspects of melatonin’s anti- of aflatoxin M1 in cow, goat, buffalo, camel, and yak milk in Chi - inflammatory actions: recent insights and new perspectives. J. Pi - na in 2016. Toxins, 14: 870. neal Res., 54: 1–14. Mincheva-Nilsson L., Baranov V. (2010). The role of placental exo- somes in reproduction. Amer. J. Rep. Immunol., 63: 520–533. Received: 26 I 2022 Mostafa G.A., Bjørklund G., Al-Ayadhi L. (2021). Therapeutic effect Accepted: 16 IX 2022 http://www.deepdyve.com/assets/images/DeepDyve-Logo-lg.png Annals of Animal Science de Gruyter

Loading next page...
 
/lp/de-gruyter/the-potential-therapeutic-role-of-camel-milk-exosomes-a-review-0nX15k0dhP

References

References for this paper are not available at this time. We will be adding them shortly, thank you for your patience.

Publisher
de Gruyter
Copyright
© 2023 Norah A. Althobaiti et al., published by Sciendo
ISSN
1642-3402
eISSN
2300-8733
DOI
10.2478/aoas-2022-0072
Publisher site
See Article on Publisher Site

Abstract

Ann. Anim. Sci., Vol. 23, No. 2 (2023) 353–362 DOI: 10.2478/aoas-2022-0072 1 2♦ 3 4 5 Norah A. Althobaiti , Sayed Haidar Abbas Raza , Mona N. BinMowyna , Reem D. Aldawsari , Sameh A. Abdelnour , 6 2 7 8 2 Mahmoud Abdel-Hamid , Dwi Wijayanti , Afaf Kamal-Eldin , Atif Khurshid Wani , Linsen Zan Biology Department, College of Science and Humanities, Al Quwaiiyah, Shaqra University, Al Quwaiiyah 19257, Saudi Arabia College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China College of Applied Medical Sciences, Shaqra University, Shaqra, Saudi Arabia Biology Department, College of Science and Humanities-Al Quwaiiyah, ‎Shaqra University, Al Quwaiiyah 19257, Saudi Arabia Department of Animal Production, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt Dairy Science Department, Faculty of Agriculture, Cairo University, Giza 12613, Egypt Department of Food Science, College of Food and Agriculture, United Arab Emirates University (UAEU), Al Ain, United Arab Emirates School of Bioengineering and Biosciences, Lovely Professional University, Punjab (144411), India Corresponding author: dr.haiderabbasraza@gmail.com abstract exosomes (exos) are naturally occurring nano-sized vesicles released into the extracellular environment by exocytosis. exos contribute to intercellular communications by regulating Dna, mrna, and protein levels. exos are considered good vehicles for delivering thera - peutic agents in multiple diseases. camel milk (cm) is a valuable food with a rich source of exos. camel milk exosomes (cmexos) have unique ingredients compared to other animal milks. it is reported that cmexos reduce the growth of cancer cells by inducing apoptosis, and inhibiting oxidative stress and inflammation. CMEXOs can block stress signaling pathways including inflammation and apoptosis which could be resulted in adverse effects if medication levels exceed the therapeutic range. moreover, cmexos improve the antioxidant capability in both in vitro and in vivo experiments. on the other hand, the anti-angiogenesis property of cmexos has been reported via decrease in expression of the angiogenesis-related gene of vascular endothelial growth factor (VEGF). it is predicted that exosomal lactoferrin (lf) and kappa casein (kc) mrnas are crucial parts of cmexos mediating their anticancer effects. The immunomodulatory effect of cmexos may be attributed to their high contents of lf and kc. according to previous works, cmexos are promising alternatives in developing new therapeutic models for multipile diseases. This review aims to provide an overview of the isolation, characterization and biological activities of the exosomes derived from camel milk for addressing their possible use in thera- peutics. Key words: camel milk exosomes, anticancer activity, anti-inflammatory activity, immuno-modulatory activity, antioxidant activity In the Middle East, the daily drinking of camel milk The EXOs are located in different tissues and biologi- (CM) is usually supposed to improve immune capacity and cal fluids including blood plasma, milk, saliva, oviductal, decrease the risk of many diseases. Several beneficial prop- urine, cerebrospinal fluid, ascites, amniotic fluid and erties of CM were reported including immunostimulatory, others (Colombo et al., 2014). Among them milk is the anti-microbial, anti-viral, anti-parasitic, anti-inflammatory, only biological fluid comprising high amounts of EXOs antioxidant, and anti-allergic effects (Badawy et al., 2018; (Adriano et al., 2021). Camel milk (CM) is one of the El-Kattawy et al., 2021; Khan et al., 2021). The main health- wealthy sources of beta-caseins and whey proteins, com- encouraging bioactive components in CM are lactoferrin prising lactoperoxidase, lysozyme, alpha-lactalbumin, (LF), kappa casein (KC), and some other molecules having lactoferrin, and immunoglobulins. The antioxidant, anti- the mentioned properties (Iigo et al., 2009; Shariatikia et al., bacterial, anti-diabetic, anti-viral, anti-cancer, and anti- 2017). Exosomes (EXOs) are common extracellular nan- inflammatory properties of CM might be attributed to the ovesicles that contribute to intercellular communications by high contents of those biological components (Khan et transporting, supporting and chaperoning biologically ki- al., 2021; Swelum et al., 2020; Mostafa et al., 2022). netic elements such as DNA, mRNA, microRNA, proteins, Among all types of exosomes in different milk sourc- etc. (Colombo et al., 2014; Cintio et al., 2020). Because of es, camel milk exosomes (CMEXOs) have received their nano size (40–100 nm diameter), EXOs are auspicious much awareness in the last few years, owing to their di- delivery elements in individual therapy (Bunggulawa et agnostic, theranostic, and therapeutic uses. al., 2018). EXOs that are isolated from milk, have superior Several authors reported that EXOs of CM have more significance impacts due to their capability in transferring stability in low pH and high and low temperature (Yassin genetic material from mother to infants (Mincheva-Nilsson et al., 2016) and it can be easily absorbed through the and Baranov, 2010). gastrointestinal barrier to circulation (Baier et al., 2014). 354 N.A. Althobaiti et al. Furthermore, EXOs of cow or human milks also have of doxorubicin-encapsulated goat milk exosomes caused anti-cancer properties and improve immune response significant inhibition of cell viability in several types against different tumor cells (Anand, 2010; Abels and of cancer cells including A549, HCT-116, and HepG2 Breakefield, 2016). cells (Ahmed et al., 2022). Santos-Coquillat et al. (2022) The anti-cancer properties of CMEXOs have been reported that the concentration of pure goat’s milk ex- described via suppression of inflammation, metastasis, osomes was 2.29±0.25 mg/mL as quantified by Coomas- oxidative stress (OS), and augmentation of tumor cell sie-Bradford test. While Yassin et al. (2016) found that apoptosis (Abels and Breakefield, 2016). Generally, the levels of phosphatidylserine, and phosphatidylcho- CMEXOs have greater biocompatibility, low adverse line ranged between ~10–15 µg/mL and ~20–25 µg/mL, immune effects and no systemic toxicity, and inflam- respectively, in dromedary milk exosomes which were matory responses (Badawy et al., 2018; Bunggulawa et steady throughout various lactation stages. Several sci- al., 2018). In the last years, it was reported that EXOs entific papers suggested that the beneficial health effects are promising candidates as anti-cancer drugs by induc- of CM might be attributed to its high contents of phos- tion of apoptosis and suppression of angiogenesis and pholipids, such as those reported in CMEXOs (Ali et al., inflammation pathways (El-Kattawy et al., 2021). The 2017; Zheng et al., 2022). After extensive investigations immunomodulatory and antioxidant effects of CMEXOs of milk exosomes, it should be clear that no standard are due to the high contents of caseins and lactoferrin amounts of EXOs have been recorded. From this point of (Ibrahim et al., 2019). However, the use of milk EXOs view, extensive clarification should be done in this scien- in therapeutic sciences still needs to be further explored. tific point to develop the method of characterization and Therefore, there is an indispensable necessity to dis- identification of milk EXOs. cover more operative, secure, and selective additives to produce anti-cancer treatments to enhance their thera- isolation of milk exosomes peutic plausibility and reduce their secondary impacts. Different centrifugation techniques are adopted to In this paper, we overviewed the isolation of CMEXOs isolate milk EXOs as a result of variability in milk con- and their possible therapeutic use as anti-cancer, anti- tent of animals (Yassin et al., 2016; Cintio et al., 2020; inflammatory, anti-angiogenesis, and antioxidant inter - El-Kattawy et al., 2021). Successive centrifugations are mediates. applied to detach the EXOs. First, a low speed centrifu- gation (600–1000× g) is operated to defat the milk and 3 3 milk exosomes precipitate the cells, then 10×10 – 16×10 g is used to According to the procedures of the International So- precipitate milk proteins, and finally, one or numerous 3 3 ciety for Extracellular Vesicles, various immunological, multi-ultracentrifugations at 100×10 – 200×10 g are uti- physical, and physicochemical methods are used to iso- lized to separate the EXOs (Yassin et al., 2016; Badawy et late milk EXOs (Lötvall et al., 2014). EXOs are often al., 2018; El-Kattawy et al., 2021). Commercially, sever- produced by multivesicular bodies which are released via al supply materials are used, including volume-excluding the fusion process with the plasmatic membrane of a cell polymers for isolation of milk EXOs such as ExoQuick (Adriano et al., 2021). Based on the size of secretion fac- (System Biosciences, Palo Alto, CA, USA), containing tors, these released vesicles were named as “exosomes”, PEG 8000 kDa; Total Exosome Isolation (Thermo Fisher which have a size range between 40 and 100 nm, while Scientific, Waltham, MA, USA), comprising polyvi- the other secretions called microvesicles have a 1 μm nyl, polyethylene glycol, and dextrans with a molecular range size (Adriano et al., 2021). weight above 1000 kDa, and their mixtures with other The literature supported that the EXOs contain sev- approaches (Jiao et al., 2020). Additionally, the immuno- eral fragments from the parent cells, comprising differ- magnetic approach is applied to separate EXOs through ent types of RNAs (microRNA, long non-coding RNA, a semipermeable membrane (Bunggulawa et al., 2018). and tRNA), DNA nucleotides, proteins and lipids (Ngu EXOs of high quality could be obtained by ultracentrifu- et al., 2022). For quantification and characterization of gation from different body fluids (milk, seminal plasma, EXOs in milk or other fluids, the most common method oviductal, uterine, blood plasma, urine, lacrymal). In ad- for visualization is via the transmission electron micro- dition, cell culture fluid generally has “nonvesicles” with scopic images (Lázaro-Ibáñez et al., 2014; El-Kattawy 20–100 nm in diameter. These “nonvesicles” are morpho- et al., 2021). A further full clarification of the biogenesis logically accredited to intermediate and very-low-density and structure of exosomes, and other types of extracellu- (40–100 nm) and low-density lipoproteins (20–40 nm) lar vesicles (components) are mentioned in recent papers (Grigor’eva et al., 2017). Transmission electron micros- (Akers et al., 2013). copy can detect the ultrastructure of the cell discriminate EXOs, therefore EXOs were categorized by transmission exosomes in milk of different mammalian species electron microscopy (Badawy et al., 2018; El-Kattawy Many publications have revealed that the goat milk et al., 2021). The previously method detected specific exosomes had the highest loading capacity compared to protein markers such as TSG101 (a truncated 35 KDa other milk exosomes derived from cow and buffaloes protein), which was discovered explicitly for CMEXOs (Ahmed et al., 2022; Yan et al., 2020). Moreover, the use rather than the previously described 43 KDa mammalian The potential therapeutic role of camel milk exosomes 355 one (Yassin et al., 2016). This may be attributed to the many proteins that contribute to various biological func- specific protein in each species and the unique proper - tions in cellular development. ties of CMEXOs. Decisively, several commercial kits are Furthermore, CMEXOs have many specific types of also offered for rapid isolation of EXOs. Among them, proteins that regulate their biological roles, specifically, ThermoFisher, Sigma, FUJIFILM Wako Pure Chemical conveyance of microRNA and adhesion to the target cells Corporation, Nano View Biosciences, Qiagen, Takara including tetraspanins, CD81, CD63, and CD9 as previ- Bio, Biocompare, System Biosciences, IZON, BioCat ously reviewed by Khan et al. (2021). More specifically, GmbH, Miltenyi Biotec, BioVision Inc., etc are well- both lactoferrins and kappa casein mRNAs were noticed known companies. Nonetheless, such kits are used for in camel milk exosomes (Ibrahim et al., 2019). El-Kat- the isolation of EXOs from the cell culture medium and/ tawy et al. (2021) found a significantly higher transcript or human biological fluid. Consequently, in this part, the of kappa casein and lactoferrins genes in colostrum- generally applied isolation approaches are discussed for derived EXOs than in other lactation stage-derivative milk EXOs (Yassin et al., 2016; Badawy et al., 2018; El- EXOs. The higher contents of EXOs in colostrum show Kattawy et al., 2021). There is no one standard technique a greater immune-stimulatory function, which can be for the isolation of EXOs from milk and other biological a part of the innate immunity of infants (Gu et al., 2012). fluids. Successful isolation of milk-derived EXOs is un- On the other hand, no substantial alteration was detected der investigation and there is an unmet need to develop in expression amounts of both kappa casein and lacto- a reliable and reproducible EXOs separation technique. ferrins in late- and mid-lactation milk of camel. The recognition of kappa casein and lactoferrins mRNAs in Bioactive compounds of milk exosomes CMEXOs suggests that their mRNAs are protected from CM comprises the important nutrients as well as degradation by milk RNase (Keller et al., 2011; Yassin et potentially health-beneficial compounds with antihy - al., 2016; Khan et al., 2021). The antioxidant capacity of pertensive, anti-viral, anti-inflammatory, antidiabetic, CM has also been credited to the structural configuration anticarcinogenic, and antioxidant properties (Al-Majali of high amount of beta-caseins content. Upon hydroly- et al., 2007; Izadi et al., 2019; Ghazzawi, 2020). It has sis, the CM beta-caseins are converted to some bioac- been used for preventing or treating some diseases such tive peptides showing helpful antioxidant actions. The as allergies, diabetes, and autism. Also, CM could de- caseins and whey proteins of CM have antioxidant and crease blood cholesterol levels and improve metabolism cytotoxic activities alongside the MCF7 cells. Moreover, because CM is a rich source of the unique combination casein of CM presented plausible apoptotic and anti- of biological compounds such as protein, nucleic acids, HCV activity in HeLa cell lines and human hepatoma and EXOs (Jilo and Tegegne, 2016). Additionally, CM is (Almahdy et al., 2011). EXO RNAs are one of the most full of beta-caseins and many of whey proteins, compris- common components in camel milk, however, there is no ing lactoperoxidase, immunoglobulin, lactoferrin, alpha- published data regarding the characterization and global lactalbumin, and lysozymes (Jilo and Tegegne, 2016). transcriptomic microRNA in this species. Therefore, CM Lactoferrin is the second main protein after casein, it has can be a good environment to explore the profile of EXO numerous biological roles, regulating the iron metabo- microRNAs for using in different biomedical and scien- lism in the body, boosting the immune response, and has tific components. In the other species, various microRNA a critical role in the cellular defense against pathogenic of milk and its EXOs generally have various targets and bacteria through its bactericidal and/or bacteriostatic ac- have a significant role in cell proliferation, pregnancy, tivities (Al-Majali et al., 2007; Jilo and Tegegne, 2016). fetal development, immune system improvement, insulin CMEXOs have received more attention in the last years resistance, sugar metabolism and inflammation (Sedykh due to their health-promoting properties. The biologi- et al., 2020). In addition, natural lipid compounds of cal compounds of milk EXOs can meaningfully affect milk EXOs are more well-matched and less toxic than biomolecules delivery. Therefore, a comprehensive scru- synthetic liposomes for uses as drug delivery vehicles tiny of the content of milk EXOs that are also termed (Elsharkasy et al., 2020). According to the literature, pro- “exosomics” (by analogy with genomics, transcriptome, teins of milk EXOs have significant roles in formation of proteomics, and other -omics technologies), is compul- cytoskeleton protein, homeostasis, cell development, cel- sory (Yassin et al., 2016). Al-Majali et al. (2007) sug- lular defense, and signal transduction etc., but this kind gested that camel milk EXOs proteins such as TSG101 of roles for CM proteins have been not illustrated and (a 35 KDa of molecular weight) could be accredited with need further exploration. various biological roles in cell development and regula- In a recent study, He et al. (2021) performed the prot- tion pattern. To date, the biological function of TSG101 eomic analysis on Bactrian camel milk and found higher is ambiguous and needs further explorations to demon- contents of lactoferrin, lactoperoxidase isoform 1 prepro- strate the molecular purpose. protein, and FABP domain-containing protein as well as Studies revealed that the biological properties of κ-casein, alpha-1-acid glycoprotein and peptidoglycan- CM proteins might be attributed to their high content of recognition protein. It has been reported that EXOs de- exosomes (extracellular nanovesicles) (Gu et al., 2012; pending on their source can be divided into virtuous and Khan et al., 2021). Generally, the milk exosomes have malicious exosomes. Exosomes derived from CMEXOs 356 N.A. Althobaiti et al. are an example of virtuous EXOs which exhibited a su- breast tumor development. The cancer therapy might be perior anticancer effect (inhibition of the growth and associated with higher apoptosis pathways; eminent at- metastatic potential of MCF7 cells) to that of CM (Bad- tenuation in angiogenesis, inflammation, and metastasis; awy et al., 2018). as well as notable reduction of oxidative stress synthe- The phospholipid membrane encircling EXOs de- sis, and greater immune response (Badawy et al., 2018). fends exosomal substances (DNA, RNA, protein) from Furthermore, Almahdy et al. (2011) and Korashy et al. degradation by digestive enzymes and acidic conditions (2012) showed that the administration of whole CM (Record et al., 2014). This gastric constancy may eluci- led to high cytotoxic influence on MCF7 tumor cells in date anti-cancer property for EXOs when it is adminis- a dose-dependent manner. Earlier investigations showed tered orally compared to CM used alone. Besides, the that CM triggered apoptosis of tumor cell line (type absorbed EXOs will take a few hours to arrive at the cel- MCF7) (Korashy et al., 2012) and other cell lines com- lular target and may undergo dilution, by circulation all prising BT-474, Hela, and HepG2, via the same molecu- over the body, before contacting the tissue tumors. This lar pathway (Almahdy et al., 2011; Hasson et al., 2015). shows that local injection of EXOs in tumor tissues has This stimulatory machinery of CM could be connected better consequences than oral administration. However, to high contents of immunoproteins such as lactoferrin, regarding the biological properties of EXO, it is urgent α-lactalbumin, and other bioactive compounds (for ex- to do more explorations, especially in CMEXOs to detect ample exosomes) which have been unclarified (Almahdy different EXOs of this species which could be used in et al., 2011). Badawy et al. (2018) reported that the apop- several biomedical fields. tosis indices such as both caspase 3, and Bax transcripts were significantly increased and decreased Bcl2 gene ex- camel milk exosomes as: pression, as well as high DNA fragmentation was record- Anti-cancer agent ed in EXOs treatment compared to CM treatment, indi- The potential therapeutic role of CMRXOs in cancer cating that CM could induce apoptosis, at least in part, remains an emerging scope of research. A huge number interceded by exosomes. Totally, the inhibitory upshot of of reports have illustrated that CMEXOs have capabil- CM and its EXOs on tumor cells is owing to the antioxi- ity to prevent the progression of tumor cancer in several dative properties and induction of the apoptosis process. in vivo or in vitro experiments (Badawy et al., 2018; El- And the anti-cancer activity of CMEXOs is related to the Kattawy et al., 2021). Studies have indicated that cancer attendance of defensive proteins such as lysozyme, lacto- cells survive via different molecular mechanisms such as ferrin and lactoperoxidase and other immunological in- restraining of apoptosis, immunosuppression, and stimu- dices such as IgG, and IgA (Jilo and Tegegne, 2016). As lation of inflammation (Wong, 2011; Diakos et al., 2014; previously clarified, the high levels of lactoferrin in CM Badawy et al., 2018; El-Kattawy et al., 2021). It is iden- could prevent the propagation of colorectal cancer cells tified that a conceivable part of CMEXOs inhibits the and have antioxidant and DNA injury-repressing charac- growth and proliferation of MCF7 cells and diminishes teristics in cancerous cells (Habib et al., 2013). Figure 1. The method for isolation and purification of camel milk-derived exosomes using ultra-centrifugation method The potential therapeutic role of camel milk exosomes 357 Figure 2. The biological components of camel milk documented in the present article Lately, Badawy et al. (2021) showed a significantly Anti-inflammatory activity superior DNA impairment catalogue in tumor tissues In addition to anti-cancer property, CMEXOs also such as HepaRG cells after treating with CMEXOs. In exhibited high capability of anti-inflammatory activity. this case, CMEXOs triggered HepaRG cells dispersion Augmented amounts of pro-inflammatory cytokines like via apoptosis pathways that can be associated with the TNFα, TGFβ1, IL1β, and NFkB, are indispensable for potentiality of exosomes-treated cells to exhibit a sig- tumor initiation, metastasis and attack (Han et al., 2014; nificant increment in the expression of caspase 3 and Elgazar et al., 2018). In a study on rats, Arab et al. (2018) Bax and a significantly lower expression of Bcl2 than reported that CM could restore the inflammatory environ- the control group (Badawy et al., 2021). The apoptosis ments connected with diabetes and 5-fluorouracil-incited induction approach is one of the main innovative tools renal impairment in rats via restraining the pro-inflam- for cancer therapy. Tumor cells sustain their viability via matory cytokines statement. Additionally, Badawy et al. induction of inflammation, and suppression of apoptosis (2018) described that the anti-inflammatory property of and angiogenesis in the tumor microenvironment (Dia- CMEXOs exhibited superior impact to CM, as docu- kos et al., 2014). Likewise, CMEXOs showed a potent mented by downregulation of the inflammation-related apoptotic property on HepaRG as discovered by a sub- genes, interleukin 1 beta (IL1β), and nuclear factor-kappa stantial upregulation of Bax and caspase3 and a notewor- B (NFκB) in cancer tissues. In parallel with the previ- thy downregulation of Bcl2, with noteworthy fluctuations ous results, the anti-inflammatory effect of camel urine in colostrum-treated cells. Parallel apoptotic alterations was evidenced on the tumor cell such MCF7 cells in rats were observed in the subsequent treatment of HepG2 and (Romli et al., 2017). Linking with the biomolecules of MCF7 (Korashy et al., 2012; Badawy et al., 2018). In camel urine and milk, it is conceivable that EXOs are in vivo and in vitro studies, TR35, which was separated the main players in this achievement as they are formed from Xinjiang Bactrian camel milk, could meaningfully by almost all cell subgroups and plentifully isolated or constrain Eca109 cell propagation and persuade its ap- derived from the urine or/and milk (Romli et al., 2017). optosis (showed by caspase-3 activity, MTT assessment, Ibrahim et al. (2019) clarified that the CMEXOs dis- and Annexin V-FITC) (Yang et al., 2019). Moreover, play anti-inflammatory activities against tumors through TR35 could constrain the development and progres- a substantial augment in the expression of TNF-α and sion of xenografted tumors in nude mice without loss in IL-6 as well as a noteworthy reduction in the expression body weight (Yang et al., 2019). The tiny proteins iso- abundance of IFN-γ after administration with cyclophos- lated from CM could be used as a potential therapeutic phamide as related to the control rats (Ibrahim et al., ingredient according to its historical anti-cancer activity. 2019). NF-κB is one of the vital transcription features Although several investigations have explored the use that helps the expression of various genes of immune and of the whole CM in different types of cancer therapies, inflammatory responses such as IL-6 and TNF-α (Mauriz further explorations are needed to discover the effect of et al., 2013). A previous description documented the anti- nano biomolecules isolated from CM that can be used in inflammatory property for CMEXOs via the downregula- modern therapy. tion of NF-κB and IL-1β in rat’s mammary tumor tissues 358 N.A. Althobaiti et al. (Badawy et al., 2018). Certain in vivo studies revealed that synthesis of IFN-γ in the attendance of allogeneic human camel undenatured whey proteins restricted the extended T-lymphocytes (Actor et al., 2009), and it can trigger the inflammation in diabetic rats via the decrease in the con- synthesis of IL-12 by antigen offering cells, which in turn centrations of serum IL-1β, IL-6 and TNF-α (Ebaid et improves IFN-γ synthesis and initiates Th1 cell growth al., 2013). Moreover, CM repressed renal inflammation (Actor et al., 2009; Daneshmandi et al., 2017). persuaded by 5-fluorouracil via suppression of IL-1β, Casein-loaded dendritic cells could somewhat pro- and TNF-α as well as repressed NF-κB stimulation in voke the Th1 cell response via the improvement of Wistar rats (Arab et al., 2018). Numerous bioactive pep- lymphocyte propagation and inducement of IFN-γ syn- tides are showing various health-promoting purposeful thesis (Daneshmandi et al., 2017). Recently, CMEXOs properties in milk proteins. Consequently, camel unde- showed a significant modulation in the inflammation natured whey protein can boost the immune response via process of cancer cells through significant downregula- the augmentation of mRNA expression of both IFN-γ and tion of inflammatory-related genes such as TNFα, NFkB, IL-2 in rats after four months of streptozotocin-induced TGFβ1, and Cox2 in HepaRG cells (Badawy et al., diabetes (Ebaid, 2014). Among the chronic hepatitis B 2021). In rats bearing breast cancer, it has been reported patients, it was reported that the levels of serum IFN-γ in that the CMEXOs have a potential role in repressing the camel milk-drinking patients were meaningfully greater pro-inflammatory cytokines (Badawy et al., 2018). Col- than those in patients who did not receive CM (Saltanat lectively, previous evidence clarified the pulsative role et al., 2009). This might be showing the potential anti- of CMEXOs in reduction of the pro-inflammatory cy- viral activity of CM or its bioactive peptides. Moreover, tokines response in different in vitro and in vivo models, lactoferrin supports the development of dendritic cells by however, further explorations are essential to elucidate augmenting their capability to induce propagation and more molecular mechanisms behind their effects. Figure 3. Therapeutic potential of camel milk exosomes with potent anticancer, antioxidant, anti-inflammatory, and anti-angiogenesis effects Figure 4. The mechanistic roles of exosomes isolated from camel milk The potential therapeutic role of camel milk exosomes 359 Anti-angiogenesis activity reveals a potent anti-angiogenetic activity of EXOs for the Generation of new development of the vascular net- incorporated therapy compared to a single treatment. work is vital since the metastatic spread, proliferation of cancer cells be contingent on a satisfactory supply of Immuno-modulatory activity nutrients and oxygen and the elimination of waste prod- The immune system plays an essential function in dif- ucts (Nishida et al., 2006). Angiogenesis is controlled by ferent cellular development and growth in living organ- both inhibitor and activator molecules. The detection of isms as well as is responsible for induction of different angiogenic inhibitors or anti-angiogenesis can support diseases. This feature is primarily intermediated by sev- a decrease in both mortality and morbidity from carcino- eral immune cells: T cells, including CD4+ helper, CD8+ mas. Milk EXOs are hopeful candidates in emerging new cytotoxic, and natural killer (NK) cells (Grivennikov et therapeutic tactics to exhibit different anti-angiogenesis al., 2010). CM proteins exhibited better immune respons- activities. Various diseases have been reported to have es connected with the enhancement of infectious diseas- higher activity of vascular endothelial growth factor es. In this regard, Badawy et al. (2018) clarified that ad- (VEGF) (Nishida et al., 2006). A substantial correlation ministration of CM and its EXOs significantly enhanced between the expression of VEGF and prognosis has been the immune criteria such as NK+, CD8+, and CD4+ T described in several types of cancers and other diseases cells in the spleen of rats. Higher T cell differential de- (Badawy et al., 2018, 2021). Administration of CMEXOs tected in animals could reflect better immunity and lesser led to substantial downregulation of angiogenesis-related tumor sizes. Likewise, various articles documented that gene (VEGF) expression in tumor tissues as compared to cancer patients with high levels of T cells such as NK+, the tumor group (Badawy et al., 2018, 2021). This fea- CD8+, and CD4+ cells have superior prognosis and sur- ture could suggest the inhibitory property of CMEXOs vival rates (Malmberg et al., 2008; Laghi et al., 2009). on tumor environment angiogenesis. Besides, CMEXOs Improving the levels of the CD4+ cell could constrain also exhibited an antimetastatic activity on MCF7 cells the development and progress of tumors via suppression as shown by an in vitro wound healing test, which dis- of inflammation interrelated cytokines (Chen et al., 2021; covered inferior cell migration percentage with superior Lei et al., 2022; Hou et al., 2022). On the other hand, effect when EXOs were employed (Badawy et al., 2021). CD8+ cells can reduce cancer cells directly or indirectly To further authenticate the antimetastatic probability of by augmenting the synthesis of other cytotoxic cytokines CM and its EXOs, the metastasis-related genes (ICAM1 (Grivennikov et al., 2010). and MMP9) were examined. As predicted, the applica- Surprisingly, administration of CM caused superior tion of CM and its EXOs meaningfully downregulated percentages of NK1, CD8+, and CD4+ compared to co- the transcript of MMP9 and ICAM1 in tumor tissues with administration of EXOs. This might be explained by the the greatest enhancement by EXOs administration (Bad- fact that the CM has other immune-stimulant compounds awy et al., 2021). than those existing in their EXOs. The immune-stimula- Similarly, studies have indicated that the inferior ex- tory activity of CM could be attributed to its content of pression of the metastasis-related gene such as ICAM1 immunoglobulins, casein, lysozyme, lactoferrin, and lac- was observed in tumor tissues of mice subsequently toperoxidase (El Agamy et al., 1992). Furthermore, CM treated with camel urine (Romli et al., 2017) or CM or its comprises unique nanobodies with variable heavy chain, EXOs (Badawy et al., 2021). Furthermore, the upregula- and has no typical light chains like normal antibodies. tion of ICAM1 was related to the invasive and metastatic These very tiny nanobodies presented in CM IgGs can capability of several cancer cells (breast cancer and other easily penetrate the cell membrane of any cells (includ- carcinoma cells). As previously indicated, chemokine in ing cancer cells) and localize intracellularly to achieve the tumor microenvironment shows a significant function different immune-stimulatory activities (Korashy et al., in the invasion and migration of carcinoma cells (Fernan- 2012; Sedykh et al., 2020; Khan et al., 2021). Conse- dis et al., 2004). Badawy et al. (2018) confirmed that CM quently, it was supposed that CMEXOs can transport or and its EXOs are new disruptors for chemotaxis-based be a ligand for these immune-stimulant components. This migration of MCF7 cells (Badawy et al., 2018). HepaRG mechanistic insight needs further explorations to be clari- cells treated with EXOs displayed significant downregu- fied. Ibrahim et al. (2019) reported that CM and its EXOs lation of VEGF, which indicates the aptitude of EXOs to moderately enhanced lymphocyte profile of rats treated constrain the angiogenesis process in the tumor tissues. with cyclophosphamide. The same authors indicated that A comparable anti-angiogenesis property was described the rats co-treated with CMEXOs and cyclophosphamide for EXOs derived from mesenchymal stem cells in the exhibited significant increases in both CD8+, CD4+ HCC microenvironment, unlike those originating from cells and T-lymphocyte relative to the cyclophospha- cancer stem cells which presented angiogenic activ- mide group. Authors suggested that the immunomodu- ity (Alzahrani et al., 2018). Mice co-administered with latory effects of CM and its EXO may be accredited to CMEXOs and some phytochemical displayed inferior ex- their contents of caseins and lactoferrin. In addition, pression of both VEGF and Bcl2 and superior expression of they confirmed fruitful delivery of CMEXO kappa Cas3 and Bax genes in MCF7 xenograft compared to those casein and lactoferrin mRNAs into the rat tissues via administered with each alone (Badawy et al., 2021). This separation and sequencing of camel kappa casein and 360 N.A. Althobaiti et al. lactoferrin from the spleen of the treated rats. Danesh- activity but also boost antioxidant status that decreases mandi et al. (2017) showed that casein-loaded dendrit- OS impairment triggered by tamoxifen and/or cancer ic cells could improve the Th1 immune status in mice. problem (Badawy et al., 2021). The activation of NF-κB, lactoferrin and MAP kinase can stimulate the proliferation, maturation, differen- conclusions and future perspective tiation, activation of different immune cells (Actor et An increasing body of evidence has discovered the al., 2009). It has been revealed that lactoferrin shows potential role of CMEXOs in anti-cancer, anti-inflam - a beneficial role in the reconstitution of the cellular matory, antioxidant and anti-microbial properties which immune response as it promotes cellularity and en- could be used in treating several ailments, enhancing hancement of CD4+, CD3+ lymphocytes in the spleen health, as well as contributing as a new therapeutic win- and reinstates the host T cell compartment (Artym et dow. Investigations exhibited that EXOs derived from al., 2003; Bhattacharyya et al., 2014) Besides, lacto- CM have more stable form under adverse environments, ferrin infusion produced a strong Th1- response and such as freezing-thawing, low pH, and heating, and can encouraged the activation of T-lymphocytes such as be easily absorbed through the intestinal barriers to the CD8+ and CD4+ in tumor-bearing models. circulation system. In this paper, the potential roles of CMEXOs including anti-cancer, anti-inflammatory, an- Antioxidant activity tioxidant were reviewed. Moreover, the CMEXOs can Oxidative stress (OS) can be defined as a dispar - also serve as oral delivery vehicles for chemotherapeu- ity between the free radical production and the body’s tic mediators and further amendment of the EXOs with capability to combat against the destructive impacts of a tumor-targeting ligand allows the targeted delivery of antioxidants. The subsequent difference in homeostasis drugs to the tumor sites. Furthermore, the significance on the explanation of excessive reactive oxygen species of these novel achievements of CMEXOs is constructed (ROS) induces cell impairment and is one of the leading through their advantages over prevailing therapeutic reasons for emerging ailments such as gastric/colorectal opportunities. Developments in technical isolation of cancer, inflammatory bowel disease, and gastroduode- EXOs and a greater understanding of the mechanistic nal ulcers (Bhattacharyya et al., 2014). Badawy et al. details will support the medical community to extend (2018) indicated that CM and its EXO significantly en- us a safe and effective tool for resolving public health hanced the synthesis of antioxidant enzymes (catalase problems on a broader scale. In addition, it would be (CAT), superoxide dismutase (SOD), and glutathione significant to create a standardized and cost-effective peroxidase (GPX)), and reduced the content of lipid technique to isolate, purify, and employ EXOs from peroxidation indicator malondialdehyde (MDA), and CM to safeguard the quality of the EXOs for industrial the transcript of the OS indicator iNOS in tumor mat- and clinical uses. Future reports are essential to vali- ters as relative to the tumor group. This suggests that date how individual bioactive compounds within EXOs OS/antioxidant pathway may be complicated with the employ biological roles, including antitumor, antidia- repressive role of CM and its EXOs on tumor devel- betic, anti-obesity, and antihypertensive properties to opment. It was revealed that co-treatments of cyclo- elucidate any plausible undesirable effects upon EXO phosphamide with CM or CMEXOs meaningfully de- remediation. creased the lipid peroxidation signal MDA amount and increased the antioxidant enzymes GPX, CAT, and SOD funding levels as relative to the cyclophosphamide -treated rats This work was funded by the National Key Re- (Ibrahim et al., 2019). CM enhanced the antioxidant re- search and Development Program of China (No. sponse in renal tissues and repressed the OS triggered 2018YFD0501700), National Modern Agricultural In- via 5-fluorouracil induction in Wistar rats (Arab et al., dustry Special Program (No. CARS-37), the National 2018). Furthermore, a prior investigation verified the Natural Science Foundation of China (31972994), the antioxidant capabilities of various peptides primarily Agricultural Science and Technology Innovation and attained from caseins of the CM (Ibrahim et al., 2019). Transformation Project of Shaanxi Province (NYKJ- Synergistic effect of EXOs and some phytochemical 2018-LY09) and the Key Research and Development showed significantly lower MDA, and higher CAT and Program of Ningxia (Grant No. 2019BEF02004). GPx activities in hepatic tissues than in control mice (Badawy et al., 2021). This potential ability of EXOs to acknowledgments bring the apoptosis to avoid angiogenesis and invasion We thank Shaqra University and the Scientific Re- in MCF7 cells could be a beneficial approach in treat- search Deanship of Shaqra University, KSA for their ing some diseases. Remarkably, the combined therapy kind support to Althobaiti N.A., BinMowyna M.N., and using phytochemical (tamoxifen, and hesperidin) and Aldawsari R.D. EXOs also restored the disrupted OS/antioxidant status to normal levels related to that of the standard control. competing interests Inclusively, tamoxifen, hesperidin and EXOs mixture The authors declare that they have no competing inte- treatment can not only potentiate tamoxifen anti-cancer rests. The potential therapeutic role of camel milk exosomes 361 refernces Diakos C.I., Charles K.A., McMillan D.C., Clarke S.J. (2014). Cancer- related inflammation and treatment effectiveness. Lancet. Oncol., Abels E.R., Breakefield X.O. (2016). Introduction to extracellular 15: e493–e503. vesicles: Biogenesis, RNA cargo selection, content, release, and Ebaid H. (2014). Promotion of immune and glycaemic functions in uptake. Cell. Mol. Neurobiol., 36: 301–312. streptozotocin-induced diabetic rats treated with un-denatured Actor J.K., Hwang S.A., Kruzel M.L. (2009). Lactoferrin as a natural camel milk whey proteins. Nutr. Metab., 11: 1–13. immune modulator. Curr. Pharm. Design, 15: 1956–1973. Ebaid H., Ahmed O.M., Mahmoud A.M., Ahmed R.R. (2013). Limit- Adriano B., Cotto N.M., Chauhan N., Jaggi M., Chauhan S.C., Yallapu ing prolonged inflammation during proliferation and remodeling M.M. (2021). Milk exosomes: Nature's abundant nanoplatform phases of wound healing in streptozotocin-induced diabetic rats for theranostic applications. Bioactive Mat., 6: 2479–2490. supplemented with camel undenatured whey protein. BMC Im- Ahmed F., Tamma M., Pathigadapa U., Reddanna P., Yenuganti V.R. munol., 14: 31–31. (2022). Drug loading and functional efficacy of cow, buffalo, and El Agamy E.I., Ruppanner R., Ismail A., Champagne C.P., Assaf R. goat milk-derived exosomes: a comparative study. Mol. Pharmac., (1992). Antibacterial and antiviral activity of camel milk protec- 19: 763–774. tive proteins. J. Dairy Res., 59: 169–175. Akers J.C., Gonda D., Kim R., Carter B.S., Chen C.C. (2013). Biogen- Elgazar A.A., Selim N.M., Abdel-Hamid N.M., El-Magd M.A., El esis of extracellular vesicles (EV): exosomes, microvesicles, retro- Hefnawy H.M. (2018). Isolates from Alpinia officinarum Hance virus-like vesicles, and apoptotic bodies. J. Neurooncol., 113: 1–11. attenuate LPS-induced inflammation in HepG2: Evidence from in Ali M.Z., Qureshi A.S., Usman M., Kausar R., Ateeq M.K. (2017). silico and in vitro studies. Phyto. Res., 32: 1273–1288. Comparative effect of camel milk and black seed oil in induced El-Kattawy A.M., Algezawy O., Alfaifi, M.Y., Noseer E.A., Hawsawi diabetic female albino rats. Pak. Vet. J., 37: 293–298. Y.M., Alzahrani O.R., Algarni A., Kahilo K.A., El-Magd M.A. Al-Majali A.M., Ismail Z.B., Al-Hami Y., Nour A.Y. (2007). Lactofer- (2021). Therapeutic potential of camel milk exosomes against rin concentration in milk from camels (Camelus dromedarius) with HepaRG cells with potent apoptotic, anti-inflammatory, and anti- and without subclinical mastitis. Int. J. App. Res. Vet. Med., 5: 120. angiogenesis effects for colostrum exosomes. Biomed. Pharma., Almahdy O., El-Fakharany E.M., El-Dabaa E., Redwan E.M. (2011). 143: 112220. Examination of the activity of camel milk casein against hepatitis Elsharkasy O.M., Nordin J.Z., Hagey D.W., de Jong O.G., Schiffelers C virus (genotype-4a) and its apoptotic potential in hepatoma and R.M., Andaloussi S.E., Vader P. (2020). Extracellular vesicles as hela cell lines. Hepat Mon., 11: 724–730. drug delivery systems: Why and how? Advan. Drug Del. Rev., Alzahrani F.A., El-Magd M.A., Abdelfattah-Hassan A., Saleh A.A., 159: 332–343. Saadeldin I.M., El-Shetry E.S., Badawy A.A., Alkarim S. (2018). Fernandis A.Z., Prasad A., Band H., Klösel R., Ganju R.K. (2004). Potential effect of exosomes derived from cancer stem cells and Regulation of CXCR4-mediated chemotaxis and chemoinvasion MSCs on progression of DEN-induced HCC in rats. St. Cell. Int., of breast cancer cells. Oncogene, 23: 157–167. 2018: 8058979. Ghazzawi H. (2020). Health-improving and disease-preventing po- Anand P.K. (2010). Exosomal membrane molecules are potent im- tential of camel milk against chronic diseases and autism: cam- mune response modulators. Commun. Integr. Biol., 3: 405–408. el milk and chronic diseases. Handbook of Research on Health Arab H.H., Salama S.A., Maghrabi I.A. (2018). Camel milk amelio- and Environmental Benefits of Camel Products. IGI Global, rates 5-fluorouracil-induced renal injury in rats: targeting MAPKs, pp. 155–184. NF-κB and PI3K/Akt/eNOS pathways. Cell. Physiol. Biochem., Grigor’eva A., Dyrkheeva N., Bryzgunova O., Tamkovich S., Ch- 46: 1628–1642. elobanov B., Ryabchikova E. (2017). Contamination of exosome Artym J., Zimecki M., Kruzel M.L. (2003). Reconstitution of the cel- preparations, isolated from biological fluids. Biochemistry (Mos - lular immune response by lactoferrin in cyclophosphamide-treat- cow), Suppl. Series Biomed. Chem., 11: 265–271. ed mice is correlated with renewal of T cell compartment. Immu- Grivennikov S.I., Greten F.R., Karin M. (2010). Immunity, inflamma - nobiology, 207: 197–205. tion, and cancer. Cell, 140: 883–899. Badawy A.A., El-Magd M.A., AlSadrah S.A. (2018). Therapeutic ef- Gu Y., Li M., Wang T., Liang Y., Zhong Z., Wang X., Zhou Q., Chen fect of camel milk and its exosomes on MCF7 cells in vitro and in L., Lang Q., He Z., Chen X., Gong J., Gao X., Li X., Lv X. (2012). vivo. Integr. Cancer Ther., 17: 1235–1246. Lactation-related microRNA expression profiles of porcine breast Badawy A.A., Othman R.Q.A., El-Magd M.A. (2021). Effect of com- milk exosomes. PLoS One, 7: e43691–e43691. bined therapy with camel milk-derived exosomes, tamoxifen, and Habib H.M., Ibrahim W.H., Schneider-Stock R., Hassan H.M. (2013). hesperidin on breast cancer. Mol. Cell. Toxicol, 1–10. Camel milk lactoferrin reduces the proliferation of colorectal can- Baier S.R., Nguyen C., Xie F., Wood J.R., Zempleni J. (2014). Mi- cer cells and exerts antioxidant and DNA damage inhibitory ac- croRNAs are absorbed in biologically meaningful amounts from tivities. Food Chem., 141: 148–152. nutritionally relevant doses of cow milk and affect gene expres- Han J., Bae S.Y., Oh S.J., Lee J., Lee J.H., Lee H.c., Lee S.K., Kil sion in peripheral blood mononuclear cells, HEK-293 kidney cell W.H., Kim S.W., Nam S.J. (2014). Zerumbone suppresses IL-1β- cultures, and mouse livers. J. Nutr., 144: 1495–1500. induced cell migration and invasion by inhibiting IL-8 and MMP- Bhattacharyya A., Chattopadhyay R., Mitra S., Crowe S.E. (2014). 3 expression in human triple-negative breast cancer cells. Phyto. Oxidative stress: an essential factor in the pathogenesis of gastro- Res., 28: 1654–1660. intestinal mucosal diseases. Physiol. Rev., 94: 329–354. Hasson S.S., Al-Busaidi J.Z., Al-Qarni Z.A., Rajapakse S., Al-Bahlani Bunggulawa E.J., Wang W., Yin T., Wang N., Durkan C., Wang Y., S., Idris M.A., Sallam T.A. (2015). In vitro apoptosis triggering in Wang G. (2018). Recent advancements in the use of exosomes as the BT-474 human breast cancer cell line by lyophilised camel’s drug delivery systems. J. Nanobiotech., 16: 1–13. milk. Asian. Pac. J. Cancer Prev., 16: 6651–6661. Chen X., Kang R., Kroemer G., Tang D. (2021). Broadening hori- He J., Chen Q., Yi L., Ming L., Ji R. (2021). Proteomics and micro- zons: the role of ferroptosis in cancer. Nat. Rev. Clin. Oncol., 18: structure profiling of Bactrian camel milk protein after homogeni - 280–296. zation. LWT, 152: 112287. Cintio M., Polacchini G., Scarsella E., Montanari T., Stefanon B., Hou C.X., Sun N.N., Han W., Meng Y., Wang C.X., Zhu Q.H., Tang Colitti M. (2020). MicroRNA milk exosomes: From cellular regu- Y.T., Ye J.H. (2022). Exosomal microRNA-23b-3p promotes tu- lator to genomic marker. Animals, 10: 1126. mor angiogenesis and metastasis by targeting PTEN in salivary Colombo M., Raposo G., Théry C. (2014). Biogenesis, secretion, and adenoid cystic carcinoma. Carcinogenesis, 43: 682–692. intercellular interactions of exosomes and other extracellular ves- Ibrahim H.M., Mohammed-Geba K., Tawfic A.A., El-Magd M.A. icles. Ann. Rev. Cell Dev. Biol., 30: 255–289. (2019). Camel milk exosomes modulate cyclophosphamide-in- Daneshmandi S., Nourizadeh M., Pourpak Z., Pourfathollah A.A. duced oxidative stress and immuno-toxicity in rats. Food Func., (2017). Eliciting Th1 immune response using casein (alpha s1)- 10: 7523–7532. loaded dendritic cells. Iran. J. Aller. Asth. Immunol., 159–168. Iigo M., Alexander D.B., Long N., Xu J., Fukamachi K., Futakuchi M., 362 N.A. Althobaiti et al. Takase M., Tsuda H. (2009). Anticarcinogenesis pathways acti- of camel milk in children with autism: its impact on serum levels vated by bovine lactoferrin in the murine small intestine. Biochi- of vasoactive intestinal peptide. Int. J. Med. Sci. Clin. Invent., 8: mie, 91: 86–101. 5698–5707. Izadi A., Khedmat L., Mojtahedi S.Y. (2019). Nutritional and thera- Ngu A., Wang S., Wang H., Khanam A., Zempleni J. (2022). Milk peutic perspectives of camel milk and its protein hydrolysates: exosomes in nutrition and drug delivery. Am. J. Physiol.-Cell A review on versatile biofunctional properties. J. Func. Foods, 60: Physiol., 322: C865–C874. 103441. Nishida N., Yano H., Nishida T., Kamura T., Kojiro M. (2006). Angio- Jiao R., Sun S., Gao X., Cui R., Cao G., Wei H., Wang S., Zhang Z., genesis in cancer. Vasc. Heal. Risk Manag., 2: 213–219. Bai H. (2020). A polyethylene glycol-based method for enrich- Record M., Carayon K., Poirot M., Silvente-Poirot S. (2014). Exo- ment of extracellular vesicles from culture supernatant of human somes as new vesicular lipid transporters involved in cell–cell ovarian cancer cell line A2780 and body fluids of high-grade se - communication and various pathophysiologies. Biochim. Bio- rous carcinoma patients. Canc. Manag. Res., 12: 6291. phys. Acta -Mol. Cell Biol. Lip., 1841: 108–120. Jilo K., Tegegne D. (2016). Chemical composition and medicinal val- Romli F., Abu N., Khorshid F.A., Syed Najmuddin S.U.F., Keong ues of camel milk. Int. J. Res. Stud. Biosci., 4: 13–25. Y.S., Mohamad N.E., Hamid M., Alitheen N.B., Nik Abd Rahman Keller S., Ridinger J., Rupp A.-K., Janssen J.W., Altevogt P. (2011). N.M.A. (2017). The growth inhibitory potential and antimetastatic Body fluid derived exosomes as a novel template for clinical diag - effect of camel urine on breast cancer cells in vitro and in vivo. nostics. J. Transl. Med., 9: 1–9. Integr. Cancer Ther., 16: 540–555. Khan M.Z., Xiao J., Ma Y., Ma J., Liu S., Khan A., Khan J.M., Cao Z. Saltanat H., Li H., Xu Y., Wang J., Liu F., Geng X.H. (2009). The influ - (2021). Research development on anti-microbial and antioxidant ences of camel milk on the immune response of chronic hepatitis properties of camel milk and its role as an anti-cancer and anti- B patients. Ch. J. Cell. Mol. Immunol., 25: 431–433. hepatitis agent. Antioxidants, 10: 788. Santos-Coquillat A., González M.I., Clemente-Moragón A., González- Korashy H.M., Maayah Z.H., Abd-Allah A.R., El-Kadi A.O., Alhaider Arjona M., Albaladejo-García V., Peinado H., Muñoz J., Embún A.A. (2012). Camel milk triggers apoptotic signaling pathways P.X., Ibañez B., Oliver E., Desco M., Salinas B. (2022). Goat milk in human hepatoma HepG2 and breast cancer MCF7 cell lines exosomes as natural nanoparticles for detecting inflammatory pro - through transcriptional mechanism. J. Biomed. Biotechnol., 2012: cesses by optical imaging. Small, 18: 2105421. 593195. Sedykh S., Kuleshova A., Nevinsky G. (2020). Milk exosomes: Perspec- Laghi L., Bianchi P., Miranda E., Balladore E., Pacetti V., Grizzi F., Al- tive agents for anticancer drug delivery. Int. J. Mol. Sci., 21: 6646. lavena P., Torri V., Repici A., Santoro A., Mantovani A., Roncalli Shariatikia M., Behbahani M., Mohabatkar H. (2017). Anticancer ac- M., Malesci A. (2009). CD3+ cells at the invasive margin of deep- tivity of cow, sheep, goat, mare, donkey and camel milks and their ly invading (pT3–T4) colorectal cancer and risk of post-surgical caseins and whey proteins and in silico comparison of the caseins. metastasis: a longitudinal study. Lancet. Oncol., 10: 877–884. Mol. Biol. Res. Commun., 6: 57–64. Lázaro-Ibáñez E., Sanz-Garcia A., Visakorpi T., Escobedo-Lucea C., Swelum A.A.A., Hashem N.M., Abo-Ahmed A.I., Abd El-Hack M.E., Siljander P., Ayuso-Sacido Á., Yliperttula M. (2014). Different Abdo M. (2020). The role of heat shock proteins in reproductive gDNA content in the subpopulations of prostate cancer extracel- functions. In: Heat shock proteins, Asea A.A.A., Kaur P. (eds). lular vesicles: apoptotic bodies, microvesicles, and exosomes. Springer Nature Switzerland, pp. 407–427. Prostate, 74: 1379–1390. Wong R.S. (2011). Apoptosis in cancer: from pathogenesis to treat- Lei G., Zhuang L., Gan B. (2022). Targeting ferroptosis as a vulner- ment. J. Exper. Clin. Cancer Res., 30: 1–14. ability in cancer. Nat. Rev. Cancer, 22: 381–396. Yan F., Zhong Z., Wang Y., Feng Y., Mei Z., Li H., Chen X., Cai L., Li Lötvall J., Hill A.F., Hochberg F., Buzás E.I., Di Vizio D., Gardiner C. (2020). Exosome-based biomimetic nanoparticles targeted to C., Gho Y.S., Kurochkin I.V., Mathivanan S., Quesenberry P., Sa- inflamed joints for enhanced treatment of rheumatoid arthritis. J. hoo S., Tahara H., Wauben M.H., Witwer K.W., Théry C. (2014). Nanobiotechnol., 18: 1–15. Minimal experimental requirements for definition of extracellular Yang J., Dou Z., Peng X., Wang H., Shen T., Liu J., Li G., Gao Y. vesicles and their functions: a position statement from the Inter- (2019). Transcriptomics and proteomics analyses of anti-cancer national Society for Extracellular Vesicles. J. Extracell. Vesicles, mechanisms of TR35 – An active fraction from Xinjiang Bac- 3: 26913. trian camel milk in esophageal carcinoma cell. Clin. Nutr., 38: Malmberg K.-J., Bryceson Y.T., Carlsten M., Andersson S., Björklund 2349–2359. A., Björkström N.K., Baumann B.C., Fauriat C., Alici E., Dilber Yassin A.M., Abdel Hamid M.I., Farid O.A., Amer H., Warda M. M.S., Ljunggren H.-G. (2008). NK cell-mediated targeting of hu- (2016). Dromedary milk exosomes as mammary transcriptome man cancer and possibilities for new means of immunotherapy. nano-vehicle: Their isolation, vesicular and phospholipidomic Cancer Immunol. Immunother., 57: 1541–1552. characterizations. J. Adv. Res., 7: 749–756. Mauriz J.L., Collado P.S., Veneroso C., Reiter R.J., González-Gallego Zheng N., Min L., Li D., Tan S., Gao Y., Wang J. (2022). Occurrence J. (2013). A review of the molecular aspects of melatonin’s anti- of aflatoxin M1 in cow, goat, buffalo, camel, and yak milk in Chi - inflammatory actions: recent insights and new perspectives. J. Pi - na in 2016. Toxins, 14: 870. neal Res., 54: 1–14. Mincheva-Nilsson L., Baranov V. (2010). The role of placental exo- somes in reproduction. Amer. J. Rep. Immunol., 63: 520–533. Received: 26 I 2022 Mostafa G.A., Bjørklund G., Al-Ayadhi L. (2021). Therapeutic effect Accepted: 16 IX 2022

Journal

Annals of Animal Sciencede Gruyter

Published: Apr 1, 2023

Keywords: camel milk exosomes; anticancer activity; anti-inflammatory activity; immuno-modulatory activity; antioxidant activity

There are no references for this article.