Production and quality checking of nanoalgosomes
Our recent work showed that nanoalgosome is a new type of EVs with biophysical and biochemical properties as outlined in the MISEV guidelines, specifically applied to nanoalgosomes15. Further, the isolation efficiency of differential ultracentrifugation and tangential flow filtration (dUC and TFF) has been tested15. dUC was elected as the small-scale isolation method for microalgal EVs15,16. For larger scale processes (up to 7 L), TFF was more feasible16 and then applied in the present study. Here, physicochemical features have been analyzed in-depth to evaluate nanoalgosome quality and yield, in each nanoalgosome preparations (Figure 1, upper panel). Multiple orthogonal physical and molecular techniques have been applied, including Nanoparticle Tracking Analyses (NTA), Dynamic Light Scattering (DLS), Atomic Force Microscopy (AFM), microBCA, SDS-PAGE and Western blot, also, membrane-labelling using specific fluorescent dyes and validations with fluorescent-nanoparticle tracking analysis (F-NTA) was performed (Figure 1, lower panel). Our results confirmed the presence of sEVs in the microalgal-conditioned media, with expected average diameter (e.g., 100+10 nm measured by NTA and DLS), morphology, EV markers (e.g., Alix, H+-ATPase and enolase positivity) and total EV protein/particle number ratio (e.g., 1 µg of total EV protein corresponds to a range of 5-10x109 in all nanoalgosome batches, n=6), showing that the variability of the input material is minimized. All these features grant high batch-to-batch consistency and quality, and repeatedly yielding vesicles (~1012 nanoalgosomes/L of microalgal conditioned-medium, corresponding to ~109 nanoalgosomes/mg of dried microalgal biomass) with stable biophysical and biochemical properties.
Our previous study on nanoalgosomes showed that they are highly stable in human blood plasma, non-cytotoxic in vitro and can be taken up by various cellular systems15,24. Here, to fully characterize the compatibility, distribution, and bioactivity of nanoalgosomes, we conducted a comprehensive analysis in vivo, using Caenorhabditis elegans and mice, ex vivo with human basophils from whole blood, and in vitro with human cell lines.
Nanoalgosome biocompatibility in vivo, ex vivo and in vitro
In vivo biocompatibility of nanoalgosomes in C. elegans
For in vivo analyses of nanoalgosome biocompatibility, we firstly used the model system C. elegans which is highly recommended for toxicological studies since it raises fewer ethical concerns, is less expensive, and faster to use than higher level model organisms, thus reducing the number of vertebrate animals used. C. elegans offers a comprehensive set of experiments that allow the evaluation, in a whole living animal, of the putative toxic effect of nanoparticles on animal survival, growth, lifespan, fertility, lethality, and neuron viability, among others. After quality checks on the nanoalgosome batches (Figure 1), we treated wild-type animals through in liquido culturing for 72 hours with increasing concentrations of nanoalgosomes (1, 20, 64, 128 µg/mL), first evaluating animal viability and capability of reaching adulthood. These concentrations were chosen because we had previously demonstrated that nanoalgosomes are successfully taken up by C. elegans at 20 µg/mL24. After treatment for 4 days in liquido, we did not observe any toxic effect of nanoalgosomes, as all the animals were alive and reached adulthood, similarly to the animals treated with mock (Figure 2a-b). Thereafter, we decided to use the concentration of 20 µg/mL to further evaluate nanoalgosome effects on animal fitness25. C. elegans' development and growth are finely regulated in four larval stages followed by a fertile adult stage, which can be altered when the animals are exposed to toxic agents26. To evaluate animal growth, we treated animals chronically with nanoalgosomes and tested the percentage of animals at each developmental stage every 24 hours, without observing any alterations or delays compared to animals treated with PBS as mock (Figure 2c). We also treated animals over their entire lifespan and, also in this case, nanoalgosomes had no effect on their survival curve (Figure 2d).
Another aspect used to assess the biocompatibility of nanoparticles is to evaluate their effects on animal fertility and offspring survival. Therefore, after chronic treatment of animals from eggs to L4 larval stage, we assessed the animals’ egg-laying ability and egg hatching (Figure 2e-f), without observing any effects of nanoalgosome treatment compared to mock. This result confirms that nanoalgosomes do not affect animal fertility, offspring release, or embryonic survival. Moreover, we decided to evaluate the putative neurotoxicity of nanoalgosomes. C. elegans’ transparency makes it possible to observe specific cell types, such as neurons, in living animals by using fluorescent proteins that are only expressed in the cells of interest. We thus evaluated the effects of nanoalgosomes on three different neuronal classes: GABAergic motoneurons, dopaminergic sensory neurons, and glutamatergic mechanosensory neurons. Interestingly, the invariant cell lineage in C. elegans determines a fixed number of neurons in adults, with 19 GABAergic motoneurons in the ventral cord, 6 dopaminergic neurons in the head, and 6 mechanosensory neurons along the body. Chronic treatment with nanoalgosomes did not cause any degeneration of the neurons analyzed (Figure 2g-i; Supplementary Figure 1a-c) as the number of visible neurons was not affected by the treatment. Moreover, all these neurons exhibited normal morphology. Taken together, our results demonstrate that nanoalgosomes are very well tolerated in vivo by C. elegans, and no toxicity was observed after chronic exposure at these concentrations.
In vivo biocompatibility of nanoalgosomes in wild-type mice
Furthermore, biochemical and hematological studies were conducted on wild-type mice to thoroughly evaluate the in vivo biocompatibility of nanoalgosomes in immune-competent mice, after a single intravenous (I.V.) administration of nanoalgosomes. Wild-type BALB/c mice (n=4, 2 male and 2 female) were injected with nanoalgosomes and a negative control (i.e., PBS), via the intra-ocular vein. Blood samples were collected from two mice (1 male, 1 female) at 4 time points (3, 6, 24, 48 h) to analyze hematocrit, creatinine, blood urea nitrogen (BUN), liver transaminase enzymes (Serum Aspartate Transaminase, AST, and Serum Alanine Transaminase, ALT), and lymphocyte numbers. The starting nanoalgosome doses were based on literature; in different pre-clinical studies, the dose of EVs per kg of body weight ranged from 0.10 to 100 mg of total EV proteins, with an average of 2.75 mg/kg27-29. Based on the estimation that the body weight of 6-week-old BALB/c mouse is approximately 20 grams, the average I.V. dose of EVs in mice corresponds to 55 µg/mouse30,31. Thus, in our pre-clinical studies we used the following doses: Dose 1 (low dose) = 10 µg/mouse, corresponding to 4x1010 EVs/mouse; Dose 2 (high dose) = 50 µg/mouse, corresponding to 2x1011 EVs/mouse. Table 1 shows no changes in white and red blood cell counts, hemoglobin, or hematocrit compared to control mice (i.e., injected with PBS).
Table 1. Hematological and biochemical analyses of peripheral blood from BALB/c mice after ocular I.V. injection of nanoalgosomes after 48h.
Parameters
|
Dose 1
|
Dose 2
|
Control
|
Hematological
|
|
|
|
Red blood cells (x106/μL)
|
10.1 ± 0.0
|
9.9 ± 1.0
|
10.1 ± 0.1
|
Hemoglobin (g/dL)
|
15.2 ± 0.0
|
15.0 ± 1.4
|
15.5 ± 0.5
|
MCV (fL)
|
47.0 ± 0.2
|
45.5 ± 0.5
|
47.5 ± 0.5
|
Hematocrit (%)
|
47.0 ± 0.2
|
46.0 ± 5.0
|
48.0 ± 1.0
|
Reticulocytes (%)
|
3.4 ± 0.1
|
2.1 ± 0.0
|
2.2 ± 0.3
|
MCH (pg)
|
15.0 ± 0.0
|
15.0 ± 0.0
|
15.5 ± 0.5
|
Leukocytes (x103/μL)
|
3.1 ± 0.0
|
3.1 ± 2.0
|
4.8 ± 0.9
|
Lymphocytes (%)
|
72.0 ± 0.3
|
67.0 ± 2.0
|
78.0 ± 2.0
|
Neutrophils (%)
|
20.0 ± 0.1
|
25.0 ± 1.0
|
16.0 ± 2.0
|
Monocytes (%)
|
2.0 ± 0.0
|
3.0 ± 1.0
|
1.5 ± 0.5
|
Eosinophils (%)
|
3.0 ± 0.0
|
3.4 ± 0.5
|
2.5 ± 0.5
|
Basophils (%)
|
0.0 ± 0.0
|
0.5 ± 0.0
|
0.5 ± 0.0
|
Biochemical
|
|
|
|
Creatinine (mg/L)
|
4.0 ± 0.0
|
4.0 ± 0.8
|
4.2 ± 0.4
|
BUN (mg/dL)
|
23.0 ± 5.7
|
20.6 ± 6.6
|
27.0 ± 2.2
|
Urea (g/L)
|
0.5 ± 0.1
|
0.4 ± 0.1
|
0.5 ± 0.0
|
ALT (x103 UI/L)
|
2.8 ± 1.5
|
2.6 ± 0.4
|
2.1 ± 0.4
|
AST (x103UI/L)
|
2.5 ± 0.6
|
4.0 ± 0.6
|
3.4 ± 0.3
|
Further, creatinine, urea and BUN, as well as AST and ALT values were similar to the control group, indicating normal kidney and liver functions. These results suggest that nanoalgosomes did not exhibit toxic effects, blood parameter alterations after a single acute I.V. administration. The effects of nanoalgosomes in mice were also evaluated through clinical signs, body weight, and visual observations. Daily clinical examinations were performed to check behavior and signs of suffering, such as cachexia, weakness, and difficulty moving or feeding, and compound toxicity, like hunching and convulsions. Supplementary Table 1 shows that there was not significant body weight loss, indicating that nanoalgosome administration was well-tolerated by the animals. Our results demonstrate that a single dose of up to 2x1011 nanoalgosomes per mouse did not elicit any noticeable local and systemic toxicity in immune-competent BALB/c mice, allowing for dose escalation or repeated administrations. This is in line with our C. elegans results and with previously published data on I.V. administration in mice of xenogenic milk-, human- and plant-derived EVs4,32,33. However, additional safety evaluations are necessary, since it is important to further expand in vivo studies on nanoalgosome tolerability with repeated administration in mice and larger animals, along with bio-distribution information to fully understand the potential and future applications29.
Immune-compatibility assessment ex vivo
To verify the absence of potential immune-reaction to nanoalgosomes, we performed an ex vivo basophil activation test using whole blood from healthy subjects (n=3). A specific monoclonal antibody (anti-FcɛRI), binding to the high affinity IgE binding receptor, was used as positive control. Flow cytometry characterization showed no basophil activation following nanoalgosome treatments at different doses (0.25-2 µg/mL), with a percentage of basophil activation similar to the negative control (Figure 3; Supplementary Table 2).
Genotoxicity assessment in vitro and in vivo
To evaluate nanoalgosomes’ potential as therapeutic effectors, a comprehensive preclinical study delving even deeper into the molecular level was crucial. Our group recently demonstrated that nanoalgosomes did not elicit cytotoxicity, hepatotoxicity or genotoxicity in different cell lines15,16. Here, we have examined whether nanoalgosome treatment could trigger the activation of DNA-damage pathways by conducting gene expression analysis in 1-7 HB2 normal mammary epithelial cells. Typically, DNA damage leads to cell cycle arrest, regulation of DNA replication, and activation of the repair pathway. DNA damage triggers the activation of DNA-damage sensors such as ATR (ATM and Rad3-related serine/threonine kinase) and their recruitment to DNA damage sites34,35. In addition, checkpoint kinase 1 (CHEK1) is a key downstream molecule of DNA-damage response signaling; CHEK1 phosphorylates various intracellular substrate proteins, including the RAD51 recombinase which is central to the homologous recombination pathway, and binds single-stranded DNA at damage sites, forming filaments observed microscopically as nuclear foci. Therefore, these genes are considered to be involved in the response to DNA damage maintaining genome integrity, with ATR initiating a signaling cascade that activates CHEK1 and RAD5136-38. Gene expression analysis of 1-7 HB2 cells treated with 2 µg/mL of nanoalgosomes for 24 hours showed not significant changes in the expression level of these selected DNA-damage sensors (ATR, CHECK1, RAD51), thus suggesting that the outcomes related to nanoalgosomes are negligible on the activation of the DNA damage signaling pathway (Figure 4a).
The C. elegans germline can be used as a tool to study genotoxicity in vivo39. In fact, genotoxic agents, such as doxorubicin, are capable of increasing physiological germline apoptosis40. An average of three apoptotic corpses per gonadal arm can be visualized using SYTO12, an apoptotic-DNA fluorescent marker, in the wild-type41,42. After chronic treatment of animals with nanoalgosomes at 20 µg/mL we did not observe any increase in the number of apoptotic corpses in the germline compared to mock (Figure 4b). Differently, animals treated with doxorubicin showed a higher number of apoptotic corpses compared to animals treated with mock, thus confirming that nanoalgosomes have no genotoxic effect at the concentration tested neither in vitro nor in vivo.
Biodistribution of nanoalgosomes in nude mice
Due to their lipophilic nature, EVs can be labeled with fluorescent lipid dyes and their biodistribution as well as pharmacokinetics has been evaluated in pre-clinical studies mainly using mouse models and recently using larger animals, including the pig-tailed macaque (Macaca nemestrina)29,43,44. To assess the biodistribution of nanoalgosomes systemically delivered in mice, the near-infrared dye DiR was used for nanoalgosome labeling. DiR is a lipophilic dye that fluoresces intensely only when inserted into a lipid membrane. The fluorescence spectrum of this dye (emission peak of 790 nm) allows for efficient penetration through bones and tissues with low autofluorescence, making it ideal for imaging in living animals. Nanoalgosomes were labeled with the DiR probe, while the same amount of DiR diluted in PBS was used as a free dye control. Both samples underwent NTA analysis after the removal of the free dye by ultracentrifugation and extensive washing steps. The NTA data showed a typical nanoalgosome size distribution with a mode size of 100 nm in diameter, after DiR labeling (Supplementary Figure 2a). The presence of DiR-fluorescence in the nanoalgosome-labeled samples was verified by IR measurements using an Odyssey scanner, while for the free dye control, no fluorescence signal was detected (Supplementary Figure 2b). Athymic nude mice (n=6; 3 male, 3 female) were injected with aforementioned DiR-labeled nanoalgosomes at two doses (or the same volume of the free dye control) via the intra-ocular vein. The biodistribution of the DiR-labeled nanoalgosomes was examined in live animals using IVIS Spectrum Imaging. Prone and supine mice (prone male in Figure 5a-c; prone female and supine male in Supplementary Figure 3a-b) were imaged at 3, 6, 24 and 48 hours post-IV injection. At 3 hours post-injection, the DiR-nanoalgosome fluorescent signals were accumulating mainly in the liver. Fluorescent signals increased in a time- and dose-dependent manner, as shown in the total radiant efficiency plot (Figure 5d). In contrast, lower and more constant fluorescent background signals were detected for the free dye control-treated mice after each imaging, highlighting the specificity of the DiR-nanoalgosome signal. The fluorescence levels of DiR-nanoalgosome signals in two target areas (liver and femur) were quantified at each time point using IVIS software. Interestingly, as shown in Figure 5e and f, nanoalgosomes were mainly localized in the liver 3 hours post-injection (as commonly observed for other EVs) and their concentration significantly decreased over time. Meanwhile, their concentration increased and significantly accumulated in bones (femur) by 48 hours. This organotropism is peculiar for nanoalgosomes, as most mouse studies with MSC-derived EVs have reported that EVs accumulate to the liver, spleen, and sometimes kidney and lung, with rapid clearance from blood circulation after 24h systemic injection27,28,43,44.
Nanoalgosome internalization mechanism and intracellular localization
In our previous study, we demonstrated that nanoalgosomes, when labeled with different fluorescent dyes (i.e., Di-8-ANEPPS, PKH26, or DiR), are taken up and localized in vitro in the cytoplasm of cells and in vivo in the cytoplasm of C. elegans intestinal cells15,24. Therefore, we investigated the molecular mechanisms involved in nanoalgosome internalization in vitro and in vivo. Since we proved that nanoalgosomes are internalized within human cells in a dose- and time-dependent manner through an energy-dependent mechanism, we hypothesized active endocytic pathways, including micropinocytosis, clathrin- and caveolae-mediated endocytosis, as possible mechanisms of nanoalgosome internalization that were reported for other EVs45. To test among these alternative hypotheses, we used three specific blocking agents in 1-7 HB2 cells: dynasore to inhibit clathrin-mediated endocytosis, nystatin to interfere with caveolae-dependent uptake, and EIPA to inhibit micropinocytosis45-49. We monitored intracellular nanoalgosome uptake in cells by measuring the fluorescence intensity of Di-8-ANEPPS-labeled nanoalgosomes after 2 and 3 hours of incubation with nanoalgosomes, with or without inhibitors (Figure 6a, Supplementary Figure 4). The results showed that cells treated with dynasore (60µM) had a nanoalgosome internalization trend similar to the negative control (i.e., cells incubated at 4°C, which are inhibited for all energy-dependent processes), thus indicating that dynasore inhibited nanoalgosome cellular uptake. In contrast, no significant endocytosis inhibition was observed in cells treated with EIPA (10µM) or nystatin (50µM), which showed a nanoalgosome internalization level similar to the positive control (i.e., cells incubated at 37°C). Cell are viable for each treatment, demonstrating that none of the inhibitors used, at any concentration, were toxic to the cells (Supplementary Figure 4a). These results indicate that clathrin-dependent endocytosis plays a role in the cellular uptake of nanoalgosomes. To confirm the results obtained in vitro, we took advantage of the ease in C. elegans to use genetic mutants and dissect molecular pathways. In particular, when the clathrin heavy chain gene is mutated in C. elegans, the uptake of nanoparticles is impaired50.
Thus, we used a KO mutant in the clathrin heavy chain, chc-1(ok2369), and since chc-1 is an essential gene and its depletion causes animal lethality, we analyzed heterozygous balanced animals (chc-1 KO/+). We treated for 24 hours animals with nanoalgosome fluorescently labelled with Di-8-Anepps and observed, in wild-type animals, a fluorescent signal in the intestinal cells (Figure 6b, left panel). On the contrary, in all chc-1 KO/+ animals analyzed, the fluorescent signal in the intestinal cells was strongly reduced (Figure 6b, right panel). Our results demonstrate that nanoalgosome are actively internalized in vitro and in vivo by clathrin-mediated endocytosis.
We further investigated the subcellular fate of labeled nanoalgosomes once internalized, both in vitro and in vivo. The subcellular localization of PKH26-labeled nanoalgosomes in MDA-MB 231 cells cell line was determined using immunofluorescence and confocal microscopy (Figure 7a). Three distinct subcellular compartments were evaluated for in vitro study: the endosomal compartment, the lysosomal system, and the endoplasmic reticulum (ER), using established biomarkers (CD63, LAMP1, and Calnexin, respectively).
After 24 hours of incubation, we found a co-localization of nanoalgosomes (red signal) with the endosomal protein CD63 (green signal). Fluorescence images (Supplementary Figure 5) showed this co-localization in a large portion of the cells, suggesting involvement in endosomal compartments. Confocal images (Figure 7a) further confirmed this direct relation with the endosomal system in the area where the fluorescence appears yellow.
Furthermore, the localization of PKH26-labeled nanoalgosomes was evaluated in relation to two other intracellular markers, LAMP-1 and calnexin (Figure 7a). Both fluorescence and confocal images showed that intracellular LAMP-1- and calnexin-positive compartments (green) did not co-localize with internalized PKH26-labeled vesicles, suggesting that lysosomes and ER were not involved in their intracellular trafficking.
To confirm these observations in vivo, we took advantage of C. elegans’ transparency and transgenic expression of fluorescent proteins in the Golgi (GFP fused to AMAN-2 protein) or late endosomes (GFP fused to RAB-7 protein). AMAN-2 is a membrane protein of the Golgi, and we did not observe any co-localization with Di-8-ANEPPS-labeled nanoalgosomes, as shown in Figure 7b (upper panels). On the other hand, when we used a marker for late endosome membranes (RAB-7), we observed the nanoalgosome fluorescent signal inside the endosomes (Figure 7b, lower panels). Taken together, our results suggest that after nanoalgosomes are internalized by clathrin-mediated endocytosis, the fluorescent signal of nanoalgosome membrane co-localizes only with the endosome and not with the lysosomes, thus suggesting that nanoalgosomes are not destined to lysosomal degradation. These data also demonstrate that nanoalgosome internalization mechanisms are conserved across species.
Nanoalgosome bioactivity in vitro and in vivo
Antioxidant activity of nanoalgosomes
Microalgae are sources of antioxidant compounds51,52, so we sought to determine the potential antioxidant role of microalgae-derived EVs. Nanoalgosome antioxidant activity was evaluated in two different cell lines: tumoral (MDA-MB 231) and normal (1-7 HB2), using the cell-permeable DCF-DA fluorescent probe that emits fluorescence proportionally to intracellular ROS content. Cell viability analyses showed that none of the concentrations of the oxidant agents used (i.e., H2O2 and tert-butylhydroperoxide, TBH) were toxic to the cells (Supplementary Figure 6a-c). Figure 8a shows the percentage of ROS levels in cells treated with different nanoalgosome concentrations (0.5, 1, and 2 μg/mL) for 24 hours, with and without oxidative stress induction. As shown, treatments with nanoalgosomes per se did not induce oxidative stress, and the percentage of ROS levels was comparable to the negative control (untreated cells). After treatment with oxidant agents, ROS levels significantly increased in both cell lines. However, these increases in ROS levels were significantly lower in stressed cells which had been pre-treated with nanoalgosomes for 24 hours, suggesting that nanoalgosomes significantly reduced ROS levels and rebalanced the physiological ROS levels in both cell lines. To further investigate the antioxidant abilities of nanoalgosomes, we analyzed whether they could counteract ROS, directly or indirectly modulating the expression of oxidative stress responsive genes. We selected a panel of genes that play important roles in regulating oxidative stress and maintaining cellular homeostasis. For instance, AKR1C2 (aldo-keto reductase family 1 member C2) plays a role in detoxifying lipid peroxidation products, which can contribute to the production of ROS. FTH1 (ferritin heavy chain1) helps to regulate iron levels, which is important for ROS regulation since iron can catalyze the production of free radicals. Alox12 (arachidonate 12-Lipoxygenase) can contribute to oxidative stress by producing leukotrienes, which are inflammatory mediators that can increase ROS production. NOS2 (nitric oxide synthase) produces nitric oxide, which can have harmful effects on oxidative stress. Further, to counteract the effects of ROS, CAT, GPX1 and GSR (catalase, glutathione peroxidase and reductase, respectively) work to neutralize ROS by converting them into less harmful products and to maintain the redox balance by reducing glutathione disulfide, which can be formed as a result of ROS exposure53. Thus, these proteins are interconnected and work together to regulate ROS levels in cells and maintain cellular homeostasis. After incubating 1-7 HB2 cells with 2 µg/mL of nanoalgosomes for 24 hours, with and without oxidant agent treatment, real-time PCR analyses were carried out to evaluate the mRNA expression levels of the selected genes involved in the oxidative stress cellular signaling (Figure 8b-d). The results showed that the expression of oxidative stress-related genes in cells treated with nanoalgosomes for 24 hours was similar to that of untreated cells, confirming that nanoalgosomes did not induce expression alterations of genes related to oxidative stress. As expected, after oxidative stress induction, these genes were upregulated, while interestingly the expression levels of most of the genes analyzed were significantly re-established or lowered in stressed cells pre-treated for 24 hours with nanoalgosomes. These results suggested that nanoalgosomes have potent antioxidant abilities, likely due to their antioxidant cargo and ability to neutralize free radicals, promoting protective mechanisms inside cells. To validate the nanoalgosome antioxidant effect in vivo, we evaluated the response of treated C. elegans animals to exogenous oxidative stress. Acute treatment of C. elegans with H2O2 can induce a nearly complete loss of mobility. In fact, 2 hours of treatment with H2O2 caused a strong decrease in movement (Figure 8e, mock), in contrast to the untreated animals. Interestingly, nanoalgosome treatment counteracted oxidative stress by increasing movement (Figure 8e). This rescue was similar to one obtained with a positive control, N-AcetylCysteine (NAC) (Figure 8e), a widely used antioxidant agent54. C. elegans copes with changes in ROS levels by expressing detoxifying genes, such as glutathione S-transferase gst-455.
To determine whether nanoalgosomes could also directly affect detoxification gene expression, we used a transgenic strain expressing GFP under the control of the gst-4 promoter56. After chronic treatment with nanoalgosomes, we observed a significant decrease in gst-4 expression levels, suggesting that nanoalgosomes can modulate detoxification gene expression (Figure 8f and Supplementary Figure 7). Moreover, antioxidant molecules are capable of counteracting aging and motility decline57. In fact, the animal movement is influenced by aging and starting from three days from hatching (young adult stage) it slowly declines58.
Therefore, we decided to assess nanoalgosome antioxidant effects by performing an acute treatment at low concentration (10 mg/mL for 16 h), and we analyzed the locomotion of young adult animals (right after the treatment) and older animals (10 days from hatching, 7 days after treatment). As expected, we observed a physiological decline in locomotion between young and old animals (Figure 8g). Interestingly, nanoalgosomes prevented aging effects by preserving the movement capability in older animals and keeping it similar to the one observed in younger animals (Figure 8g). Taken together, our results indicate that nanoalgosomes are bioactive with a clear antioxidant effect both in vitro and in vivo, supporting their potential role in counteracting aging at molecular and functional level, having the potential to be used as natural and innovative antioxidant effectors.
Anti-inflammatory activity of nanoalgosomes
The bioactivity of nanoalgosomes was investigated also in immune-responsive macrophage cells. Initially, we checked the viability of THP-1 cells treated with different concentrations of nanoalgosomes for up to 72 hours, confirming that nanoalgosomes (up to 2 µg/mL) did not induce any cell toxicity (Supplementary Figure 8). Subsequently, THP-1 cells were pre-treated with 0.5 µg/mL of nanoalgosomes for 4 hours and afterwards exposed to lipopolysaccharide (LPS)-induced inflammation for 20 hours. We first excluded a cytotoxic effect induced by these experimental conditions (figure 9a), then we monitored interleukin-6 (IL-6), which is a marker of inflammation, using qRT-PCR and ELISA tests. The results in Figure 9b-c show no significant differences in IL-6 induction following 24 hours of nanoalgosome treatment compared to untreated cells; this result is in line with the in vivo data, previously shown, and is indicative of the immune-compatibility of nanoalgosomes. Further, figure 9b-c shows that nanoalgosomes significantly reduced IL-6 induction in LPS-treated THP-1 cells, leading to a 4.5-fold reduction in mRNA level and a 7-fold reduction in IL-6 production, and indicating their anti-inflammatory activity in vitro (Figure 9b-c).
In conclusion, this study demonstrates that nanoalgosomes are a new type of EVs endowed with unparalleled biocompatibility and unique tropism and bioactivities, including attributes making them suitable as innate antioxidant and anti-inflammatory effectors. Indeed, nanoalgosomes reduced the levels of reactive oxygen species and prevented oxidative stress in both tumoral and normal cell lines and in whole organisms, suggesting that they are enriched with antioxidant compounds. In addition, nanoalgosomes are immune-tolerated and exhibit an anti-inflammatory activity in vitro. Furthermore, they come with the capacity to be produced at mass scale, with a renewable and sustainable bioprocess from an edible source. This places this technology in an ideal situation to tap into the enormous and still underexploited potential of EVs as novel biological therapeutics, and eventually for further bioengineering to act as delivery vehicles for therapeutic agents