Logo-ija
Int J Aging.1 :e22. doi: 10.34172/ija.2023.e22

Review Article

Potential Roles of Exosomes in Aging and Age-Related Diseases

Leila Hosseini 1, * ORCID logo, Nasrin Abolhasanpour 2 ORCID logo

Author information:
1Research Center of Psychiatry and Behavioral Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
2Research Center for Evidence Based Medicine, Tabriz University of Medical Sciences, Tabriz, Iran

*Corresponding Author: Leila Hosseini, Email: leilahosseini337@gmail.com

Abstract

Objectives:

To review exosomes in aging and age-associated diseases.

Design:

A review study.

Participants:

Aged animals.

Interventions:

Exosome treatment.

Outcome measures:

The occurrence of age-associated diseases, aging skin, cognition, and cardiac dysfunction.

Results:

Exosomes are secreted by various cell types and comprise proteins, lipids, functional messenger RNAs, cytokines, growth factors, different noncoding RNA, micro-RNAs, and other bioactive substances. These nanoparticles are implicated in several physiological processes, including intercellular communication, cell migration, angiogenesis, and anti-tumor immunity, and have gained major interest in regenerative medicine. Furthermore, several studies have demonstrated the potential roles of exosomes in age-associated diseases such as aging skin, cognition, Alzheimer’s disease (AD), Parkinson’s disease (PD), and osteoarthritis (OA).

Conclusions:

We summarized various mechanisms of exosomes in the treatment of age-related diseases, including OA, PD, AD, and aged skin. These vesicles can be of efficient medicinal value for aged-associated disease therapy in preclinical trials. Further clinical trials are needed, but the majority of the literature suggests research directions that may provide new treatment approaches and strategies for clinical application.

Keywords: Aging, Exosome, Neurodegenerative diseases, Brain, Skin, Osteoarthritis

Copyright and License Information

© 2023 The Author(s).
This is an open-access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Introduction

As the world population is aging, it is a fascinating topic for researchers around the world. Aging is related to a progressive diminution in the effectiveness of mechanisms that preserve homeostasis of the body and tissues.1 It is a major risk factor for several diseases such as cardiovascular disease, aging skin, cognition dysfunction, and the like as well as the reduction in the quality of life among elderly people.2 It is well known that oxidative stress and inflammation are important contributors to aging.3 At present, there is no effective therapeutic intervention that can lessen age-related diseases and also slow down the aging process. The development of therapies that postpone aging and the progression of age-associated diseases will be the main implication for the betterment of public health.

Exosomes are nanometer-sized membrane-bound extracellular vesicles secreted by different cell types for intercellular communication and can be detected in biological fluids in pathological and physiological contexts.4 They are found in numerous biological fluids, including serum, saliva, breast milk, serum, cerebrospinal fluid, and the like. Exosomes carry cargo molecules from their cell of origin, including proteins, mRNAs, microRNA, and lipids,5-7 and are delivered to the surrounding cells or carried to the distal cells. Owing to their active cargo content, they reprogram the recipient cells. Lately, due to their excellent characteristics, they have also been regarded as a suitable delivery vehicle for medicinal products. Furthermore, exosomes have a lipidic bimolecular structure similar to the cell membrane, thereby enabling them to be loaded efficiently with hydrophobic and hydrophilic drugs.8

A large body of studies has demonstrated that exosomes play an important role in the treatment of a wide range of diseases including, nerve injury,9 stroke,10 neurodegenerative diseases,4 aging,11,12 and heart diseases.13 In addition, evidence has shown that after intravenous injection, exosomes are mostly distributed in vascular-rich organs and organs related to the reticuloendothelial system such as the kidneys, spleen, lungs, and liver.14 Because of the dysregulation of exosomes in diseases and their role in delivering drugs to target cells, they are of therapeutic interest. This review summarizes the application and possible mechanism and function of exosomes in aging and some age-related diseases such as Alzheimer’s disease (AD), Parkinson’s disease (PD), aged skin, and osteoarthritis (OA) and reveals the current and the latest research progress.


Methods

PubMed, Google Scholar, and Scopus databases were searched to recognize publications from peer-reviewed journals using several keywords and their MeSH terms, including, ‘aging’, ‘exosome’, ‘diseases’, ‘Alzheimer’s disease’, ‘Parkinson’s disease’, and ‘elderly’. The search was conducted on April 1, 2023, and there was no search filter on publication type. In addition, studies written in English were selected, and reference lists of all relevant publications were manually selected to identify advanced-qualified studies.


Results

Molecular Mechanisms of Aging and Age-Related Diseases

As fertility reduces and life expectancy rises, the proportion of individuals aged 60 and over increases. According to UNESAS, about 900 million people are aged 60 and over worldwide, and this number will reach 21. 5% of the world population by 2050, and the prevention of aging is a common problem.15 Aging is known as the most important driving factor for various diseases and finally death. With the dramatic increase in life expectancy in recent decades, age-related diseases such as AD, frontotemporal dementia, cardiovascular diseases, skin diseases, PD, and other diseases have become one of the most serious global public health problems. High generation of reactive oxygen species (ROS), mitochondrial damage, neuroinflammation, telomere shortening, and abnormal homocysteine metabolism are crucial for the aging process and age-associated diseases. According to growing evidence, oxidative stress rises with age due to ROS accumulation and is accompanied by lipids, nucleic acids, proteins DNA, carbohydrates damage, and a decrease in cell repair mechanisms, eventually leading to the impairment of the epigenetic state of the cell 16. In addition,mitochondrial dysfunction can lead to respiratory chain deficiencies, increased production of ROS, decreased adenosine triphosphate levels, and promoted apoptosis and inflammation, resulting in numerous age-related diseases.17 With advanced age, the expression of autophagy genes such as ATG5, ATG7, and BECN1 declines, and the stimulation of autophagy increases the healthy lifespan in various model organisms such as rodents and primates.18 It was found that neurodegenerative diseases are correlated to defects in autophagy and mitochondria. Autophagy is a necessary process to remove abnormal protein aggregates in cells. It also maintains protein balance. Some studies found that autophagy problems happen in the early stages of AD. Autophagy is complicated in the generation and metabolism of β-amyloid (Aβ) and also affects the phosphorylation status and clearance of tau. Therefore, its malfunction causes the progress of AD.19 Moreover, the overexpression of α-synuclein protein in PD has been thought to impair autophagy.20

Exosomes

Exosomes are tiny extracellular vesicles that are 30 to 150 nanometers in diameter and are released from most cells, including mast cells, lymphocytes, neurons, and dendritic, epithelial, and endothelial cells during physiological and pathological conditions.21,22 Exosomes spread throughout the body and are found in blood, saliva, urine, and breast milk. These vesicles have a fluid lipid bilayer membrane and contain protein, nucleic acid, and lipids and are key regulators of many biological settings.23 These extracellular vesicles have different sizes. Large exosomes are 90 to 120 nm, and small exosomes are 60 to 80 nm in diameter. In addition, in recent years, a group of non-membrane nanoparticles called exomers has been discovered with a diameter of less than 35 nm.24 Exosomes in the plasma membrane are secreted into the extracellular environment after binding with multivesicular bodies.25 Consequently, exosomes join neighboring cells by endocytosis or are degraded via lysosomes.26,27 Exosomes are involved in multiple biological processes, including cell-to-cell communication, autophagy, lysosomal exocytosis,25 crosstalk between organs, intercellular signaling, inhibition of apoptosis,28 cleansing of cell waste products, maintaining cell homeostasis in an optimal level, the modulation of the immune and inflammatory systems, and angiogenesis; moreover, exosomes have the potential to diagnose and prognosis a wide range of diseases.25,28,29 The use of exosomes in clinical trials has advantages such as stability for a long time, easy internalization, and operation in recipient cells.28 Exosomes can be stored, they are unlikely to be rejected by the immune system, they have a low risk of forming tumors and clots in blood vessels, and they have no potential for toxicity. Furthermore, it was identified that microRNAs (miRNAs) carried by exosomes activate restorative and protective pathways in recipient cells by inducing genetic instructions30-32 (Figure 1).

ija-1-e22-g001
Figure 1.

Schematic Figure of Exosome. Note. MVBs: Multivesicular bodies. These vesicles are released from MVBs in the vesicle trafficking process



Discussion

Effects of Exosomes on Aging and Age-associated Diseases

Aging

Delayed neurocognitive recovery (dNCR) is a rampant complexity of the central nervous system in aged patients after surgery, leading to cognitive impairment, memory and inattentiveness disturbances, and also increased morbidity and mortality in sufferers in 30 days following operation.33 Liu et al revealed that exogenous mesenchymal stem cells (MSCs)-exosome downregulates the levels of ROS, malondialdehyde, Fe2+, and P53, whereas it rises glutathione, GPX4, and SLC7A11 levels in dNCR-aged mice.34 In addition, they found that treatment with exosomes derived from MSCs ameliorates cognitive dysfunction by inhibiting ferroptosis in the hippocampus of dNCR-aged animals by activating the Sirt1/Nrf2/HO-1 signaling pathway. The mentioned effects of MSCs-exosome on dNCR-aged animals were prevented by the selective Sirt1 inhibitor.34

The previous reports suggested that long non-coding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript (MALAT1) regulates cell cycle and inflammation, and its expression is reduced with age.35 Furthermore, umbilical cord MSC-derived exosomes attenuate age-induced heart dysfunction via an exosome/lncRNA MALAT1/NF-κB/TNF-α signaling pathway.36

It was reported that levels of mmu-miR-126-5p and mmu-miR-466c-5p in lungs, liver, and exosomes are downregulated, while levels of mmu-miR-184-3p and mmu-miR-200b-5p raised in exosomes in aged mice compared to young animals. The administration of young exosomes could reverse the mmu-miR-126-5p level in the lungs and liver in old animals.37 Additionally, treatment with young exosomes could result in a decrease in the level of aging-associated biomarkers such as p16Ink4A, MTOR, and IGF1R in the lungs and also in the liver of aged mice. Furthermore, telomerase-related genes (Men1, Mre11a, Tep1, Terf2, Tert, and Tnks ) were increased in the liver of aged animals following the injection of young exosomes.37 It has been shown that telomeres are shortened when telomerase activity in human somatic cells declines with aging, and it was found that the transfection of a telomerase gene to aged animals delays aging and increases longevity.38

The intrabursal injection of exosomes derived from human umbilical cord MSCs into old mice revealed their rescuing effects on the age-related reduction in fertility by increasing oocyte generation and improving oocyte quality.39 Several studies have found that exosomes secreted by MSCs promote neurogenesis in the subventricular zone of the lateral ventricles and the subgranular zone of the hippocampal dentate gyrus and reduce cognitive impairment correlated with stroke, traumatic brain injury, and PD.40,41 Zhang et al reported that exosomes from young MSCs translocate exosomal miR-136 and reduce apoptotic peptidase activators, thereby enhancing the activity of aged MSCs and increasing myocardial repair function.42

Aged Skin

The number and proliferation of dermal fibroblasts alter with age which diminishes collagen synthesis and repair, and the existing skin matrix degradation by matrix-degrading enzymes accelerates, decreasing the regenerative capacity of skin.43 Aging results in slowed wound healing,44 which is a dynamic biological process that requires the interaction of different cell types and cellular activities (e.g., differentiation, migration, and proliferation) and the synthesis of extracellular matrix proteins.45,46 Age-associated defects in the repair wound are related to decreased myofibroblasts and extracellular matrix deposition dysfunction.46 MSC exosomes have numerous therapeutic effects on the skin. A study demonstrated that exosomes transfer miR-125b from young into aged fibroblasts and promote the migration and transition of the fibroblast to thwart aging via the inhibition of sirtuin 7.47 Oh et al mentioned that exosomes derived from human iPSCs improve skin aging caused by UVB irradiation by accelerating the proliferation and migration of human dermal fibroblasts (HDFs), the inhibition of overexpression matrix metalloproteinases -1/3, and also the restoration of expression of the collagen type I in old HDFs.48 Chernoff conducted a pilot study on 40 patients with aging skin. This study has demonstrated that human placental mesenchymal-derived exosomes could improve the tone, quality, and clarity of skin and also reduce wrinkles, pores, pigment, and oiliness compared to the control group.49 Moreover, there were no allergic and hypersensitivity reactions or adverse events in all groups.49

Alzheimer’s Disease

AD is characterized by cognitive impairments, the loss of memory, and personality changes taking place with advancing age.50 AD is neuropathologically defined by the accumulation of extracellular senile Aβ plaques and intracellular neurofibrillary tangles, consisting of hyperphosphorylated tau proteins.51 Several studies showed that exosomes derived from stem cells have therapeutic effects in AD. It was found that exosomes derived from human umbilical cord-derived MSCs attenuate neuroinflammation and boost the degradation of Aβ.52 Moreover, the intravenous injection of MSC-exosomes with neprilysin and insulin-degrading enzyme (zinc metallopeptidase) activity decreased the deposition of Aβ plaques in AD transgenic mice.53 Exosomes isolated from adipose-derived stem cells exerted significant neuroprotective effects on AD mice through decreasing the Aβ1-42/1-40 ratio, Aβ levels, and neuronal cell apoptosis, as well as increasing neurite outgrowth.54 Furthermore, combined curcumin and exosomes inhibited tau phosphorylation and activated the GSK-3/AKT signaling pathway, to avoid neuronal death and relieve symptoms.55

Parkinson’s Disease

PD is a chronic neurodegenerative age-related disorder affecting the motor system in the population aged over 70 years. PD cases broadly fall into two categories: sporadic and familial with the same pathological hallmarks such as the dopaminergic neurons loss in the substantia nigra pars compacta and the inclusions of Lewy bodies and neurites of surviving neurons in the midbrain.56 A study has shown that exosomes from stem cells derived from the dental pulp of human exfoliated deciduous teeth (SHEDs) inhibit 6-hydroxy dopamine-induced apoptosis in dopaminergic neurons.57 In addition, the intranasal delivery of exosomes from SHEDS could ameliorate dyskinesia and the loss of dopaminergic neurons and normalize tyrosine hydroxylase expression in the striatum and substantia nigra in PD rats.58 Furthermore, the intracerebroventricular injection of exosome-mediated delivery of antisense oligonucleotides-4 (one antisense oligonucleotides targeting human α-syn sequence) to the brain of PD mice significantly diminished the expression of α-syn and aggregation, improved locomotor functions, and also ameliorated dopaminergic neuron degeneration.59

Osteoarthritis

OA is a common and debilitating age-related joint disease. The pathology of OA is the result of synovial inflammation, cartilage degradation, subchondral bone sclerosis, and osteophyte formation, which are common with human aging. While there are many ways to treat OA, no single treatment has been successful in reversing its progression.60 Zhang et al proved the ability of MSC exosomes to repair OA. They found that MSC exosomes promoted temporomandibular joint repair and regeneration, suppressed pain, attenuated inflammation, and restored the matrix and overall joint homeostasis.61 Exosomes derived from bone marrow MSCs can be endocytosed by chondrocytes. These exosomes have been shown to restore chondrocyte proliferation, promote extracellular matrix synthesis, and reduce knee OA pain.62 Moreover, exosomes from healthy chondrocytes exhibited high biological activity in eliminating mitochondrial dysfunction and restoring the immune response by regulating M2 macrophage infiltration, thereby slowing the progression of the OA.63 Exosome-derived miRNAs exert potent therapeutic effects on in vitro and in vivo models of OA by promoting proliferation and inhibiting apoptosis.64 Furthermore, treatment with exosomes attenuated the senescence-related β-galactosidase activity in OA osteoblasts, oxidative stress, and the accumulation of γ H2AX foci, possibly due to the protective effects on mitochondria.65 The effects of exosomes on age-associated diseases are summarized in Table 1.


Table 1. Exosomes as Therapy Agents in the Treatment of Aged-rated Diseases
Source Disease Model Main Findings References
BM-MSCs dNCR in aged mice Amelioration of cognitive dysfunction via suppression ferroptosis and activating SIRT1/Nrf2/HO-1
signaling pathway in dNCR-aged animals
34
UMSCs Aged Prevention of aging-induced cardiac dysfunction by releasing novel lncRNA MALAT1 and inhibition of the NF-κB/TNF-αsignaling pathway 36
Young fibroblast Skin wound Increase abundance of myofibroblasts and improvement of wound healing in old animals 47
iPSCs Skin wound Reduction of SA-Gal and MMP-1/3 expression and restoration of expression of collagen type I in old HDFs 48
HPMSC Aging skin Decrease wrinkles, pores, pigment, oiliness, and improve evenness of skin and vascularity 49
Blood Aged mice Upregulation of telomerase-related genes in the liver and downregulation of p16Ink4A, MTOR, and IGF1R in liver or lung of aged mice 37
HucMSC-exos Aged mice Activation of the oocyte PI3K/mTOR signaling pathway 39
HucMSC-exos Aged mice Cardiac function improvement, reduction of fibrosis, and increase of angiogenesis. 42
hMSC AD mice Decreases amyloid plaque deposition. 52
hucMSCs AD mice Reduced Aβ deposition through modulating microglial activation. 66
ADSC AD mice Decreased β-amyloid pathology and apoptosis of neuronal cells. 54
Endothelial cell AD mice Reduced phosphorylation of the Tau protein by the activation the AKT/GSK-3β signaling pathway. 55
Exosomes derived from SHEDs PD Rat Suppressed apoptosis in dopaminergic neurons. 57
Evs-derived SHEDs PD Rat Improved motor symptoms and normalized expression of tyrosine hydroxylase 58
Exosome-mediated delivery of ASO4 PD mice Exosomes-ASO4 reduced the degeneration of dopaminergic neurons and α-syn pathology and improved motor function in PD animals. 59
ADSC OA osteoblast Downregulated inflammation and oxidative stress 65
BM-MSCs Rat model of OA Decreased IL-1β, increased COL2A1 protein, and reduced MMP13 protein in the cartilage tissue. 62

Note. BM-MSCs: Bone marrow mesenchymal stem cells; dNCR: Delayed neurocognitive recovery; UMSCs: Umbilical mesenchymal stem cell; iPSCs: Induced pluripotent stem cells; HDFs: Human dermal fibroblasts; HPMSC: Human placental mesenchymal stem cell; HucMSC-exos: Exosomes derived from human umbilical cord mesenchymal stem cells; Extracellular vesicles derived from human exfoliated deciduous teeth stem cells; ASO4: antisense oligonucleotide-4; ADSC: Adipose-derived stem cells; OA: Osteoarthritis; SHEDs: Stem cells derived from the dental pulp of human exfoliated deciduous teeth; AD: Alzheimer’s disease; PD: Parkinson’s disease.


Conclusions

Studies suggested the therapeutic potential of exosomes in aging and some of age-related diseases through various molecular mechanisms and pathways. Exosomes play essential roles in intercellular communication between donor and recipient cells by delivering proteins and RNAs. These small extracellular vesiclesemerge as a therapeutic agent for regenerative medicine because of their roles in anti-inflammatory outcomes, wound healing, and anti-aging properties. Although there is a growing body of evidence on exosomes in aging and age-related diseases, principally focusing on pathophysiological mechanisms and treatment, most of the studies that yielded such results have been conducted using animal models not human models. Applying exosomes to technical and therapeutic safety issues is a main challenge. Cell culture conditions and storage methods can significantly impact exosome content and function and require the standardization of exosome extraction and storage. Moreover, the content, function, and activity of exosomes depend on the origin of the generating cells. Therefore, it is essential to optimize the source of exosome cells, including comorbidities, age, and other factors associated with exosome-producing cells. On the other hand, researchers mainly focus on exosome functional aspect, and negative effects are seldom studied. Hence, more investigations into exosome functions will help develop novel strategies for protection against aging and age-related diseases.


Acknowledgments

This study was supported by the Aging Research Institute and the Faculty of Medicine at the Tabriz University of Medical Sciences, Iran.


Funding

None.


Data availability statement

Not applicable.


Ethical approval

Not applicable.


Conflict of interests

The authors declare no conflict of interests.


References

  1. da Costa JP, Vitorino R, Silva GM, Vogel C, Duarte AC, Rocha-Santos T. A synopsis on aging-theories, mechanisms and future prospects. Ageing Res Rev 2016; 29:90-112. doi: 10.1016/j.arr.2016.06.005 [Crossref] [ Google Scholar]
  2. Safiri S, Motlagh Asghari K, Sullman MJM. The global burden of diseases and injuries among older adults. Int J Aging 2023; 1:e16. doi: 10.34172/ija.2023.e16 [Crossref] [ Google Scholar]
  3. Wu M, Luo Q, Nie R, Yang X, Tang Z, Chen H. Potential implications of polyphenols on aging considering oxidative stress, inflammation, autophagy, and gut microbiota. Crit Rev Food Sci Nutr 2021; 61(13):2175-93. doi: 10.1080/10408398.2020.1773390 [Crossref] [ Google Scholar]
  4. D’Anca M, Fenoglio C, Serpente M, Arosio B, Cesari M, Scarpini EA. Exosome determinants of physiological aging and age-related neurodegenerative diseases. Front Aging Neurosci 2019; 11:232. doi: 10.3389/fnagi.2019.00232 [Crossref] [ Google Scholar]
  5. Kanninen KM, Bister N, Koistinaho J, Malm T. Exosomes as new diagnostic tools in CNS diseases. Biochim Biophys Acta 2016; 1862(3):403-10. doi: 10.1016/j.bbadis.2015.09.020 [Crossref] [ Google Scholar]
  6. Piper RC, Katzmann DJ. Biogenesis and function of multivesicular bodies. Annu Rev Cell Dev Biol 2007; 23:519-47. doi: 10.1146/annurev.cellbio.23.090506.123319 [Crossref] [ Google Scholar]
  7. Mazini L, Rochette L, Hamdan Y, Malka G. Skin immunomodulation during regeneration: emerging new targets. J Pers Med 2021; 11(2):85. doi: 10.3390/jpm11020085 [Crossref] [ Google Scholar]
  8. Kim MS, Haney MJ, Zhao Y, Yuan D, Deygen I, Klyachko NL. Engineering macrophage-derived exosomes for targeted paclitaxel delivery to pulmonary metastases: in vitro and in vivo evaluations. Nanomedicine 2018; 14(1):195-204. doi: 10.1016/j.nano.2017.09.011 [Crossref] [ Google Scholar]
  9. Yu T, Xu Y, Ahmad MA, Javed R, Hagiwara H, Tian X. Exosomes as a promising therapeutic strategy for peripheral nerve injury. Curr Neuropharmacol 2021; 19(12):2141-51. doi: 10.2174/1570159x19666210203161559 [Crossref] [ Google Scholar]
  10. Hong SB, Yang H, Manaenko A, Lu J, Mei Q, Hu Q. Potential of exosomes for the treatment of stroke. Cell Transplant 2019; 28(6):662-70. doi: 10.1177/0963689718816990 [Crossref] [ Google Scholar]
  11. Ha DH, Kim HK, Lee J, Kwon HH, Park GH, Yang SH. Mesenchymal stem/stromal cell-derived exosomes for immunomodulatory therapeutics and skin regeneration. Cells 2020; 9(5):1157. doi: 10.3390/cells9051157 [Crossref] [ Google Scholar]
  12. Wang AL, Lukas TJ, Yuan M, Du N, Tso MO, Neufeld AH. Autophagy and exosomes in the aged retinal pigment epithelium: possible relevance to drusen formation and age-related macular degeneration. PLoS One 2009; 4(1):e4160. doi: 10.1371/journal.pone.0004160 [Crossref] [ Google Scholar]
  13. Raissi Dehkordi N, Raissi Dehkordi N, Farjoo MH. Therapeutic properties of stem cell-derived exosomes in ischemic heart disease. Eur J Pharmacol 2022; 920:174839. doi: 10.1016/j.ejphar.2022.174839 [Crossref] [ Google Scholar]
  14. Aimaletdinov AM, Gomzikova MO. Tracking of extracellular vesicles’ biodistribution: new methods and approaches. Int J Mol Sci 2022; 23(19):11312. doi: 10.3390/ijms231911312 [Crossref] [ Google Scholar]
  15. Liu Y, Weng W, Gao R, Liu Y. New insights for cellular and molecular mechanisms of aging and aging-related diseases: herbal medicine as potential therapeutic approach. Oxid Med Cell Longev 2019; 2019:4598167. doi: 10.1155/2019/4598167 [Crossref] [ Google Scholar]
  16. Maynard S, Schurman SH, Harboe C, de Souza-Pinto NC, Bohr VA. Base excision repair of oxidative DNA damage and association with cancer and aging. Carcinogenesis 2009; 30(1):2-10. doi: 10.1093/carcin/bgn250 [Crossref] [ Google Scholar]
  17. Green DR, Galluzzi L, Kroemer G. Mitochondria and the autophagy-inflammation-cell death axis in organismal aging. Science 2011; 333(6046):1109-12. doi: 10.1126/science.1201940 [Crossref] [ Google Scholar]
  18. Aman Y, Schmauck-Medina T, Hansen M, Morimoto RI, Simon AK, Bjedov I. Autophagy in healthy aging and disease. Nat Aging 2021; 1(8):634-50. doi: 10.1038/s43587-021-00098-4 [Crossref] [ Google Scholar]
  19. Li Q, Liu Y, Sun M. Autophagy and Alzheimer’s disease. Cell Mol Neurobiol 2017; 37(3):377-88. doi: 10.1007/s10571-016-0386-8 [Crossref] [ Google Scholar]
  20. Winslow AR, Rubinsztein DC. The Parkinson disease protein α-synuclein inhibits autophagy. Autophagy 2011; 7(4):429-31. doi: 10.4161/auto.7.4.14393 [Crossref] [ Google Scholar]
  21. Zhang J, Zhang X. Ischaemic preconditioning-induced serum exosomes protect against myocardial ischaemia/reperfusion injury in rats by activating the PI3K/AKT signalling pathway. Cell Biochem Funct 2021; 39(2):287-95. doi: 10.1002/cbf.3578 [Crossref] [ Google Scholar]
  22. Yang D, Zhang W, Zhang H, Zhang F, Chen L, Ma L. Progress, opportunity, and perspective on exosome isolation - efforts for efficient exosome-based theranostics. Theranostics 2020; 10(8):3684-707. doi: 10.7150/thno.41580 [Crossref] [ Google Scholar]
  23. Ailawadi S, Wang X, Gu H, Fan GC. Pathologic function and therapeutic potential of exosomes in cardiovascular disease. Biochim Biophys Acta 2015; 1852(1):1-11. doi: 10.1016/j.bbadis.2014.10.008 [Crossref] [ Google Scholar]
  24. Zhang H, Freitas D, Kim HS, Fabijanic K, Li Z, Chen H. Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation. Nat Cell Biol 2018; 20(3):332-43. doi: 10.1038/s41556-018-0040-4 [Crossref] [ Google Scholar]
  25. Hessvik NP, Llorente A. Current knowledge on exosome biogenesis and release. Cell Mol Life Sci 2018; 75(2):193-208. doi: 10.1007/s00018-017-2595-9 [Crossref] [ Google Scholar]
  26. Mulcahy LA, Pink RC, Carter DR. Routes and mechanisms of extracellular vesicle uptake. J Extracell Vesicles 2014; 3(1):24641. doi: 10.3402/jev.v3.24641 [Crossref] [ Google Scholar]
  27. Tian T, Zhu YL, Hu FH, Wang YY, Huang NP, Xiao ZD. Dynamics of exosome internalization and trafficking. J Cell Physiol 2013; 228(7):1487-95. doi: 10.1002/jcp.24304 [Crossref] [ Google Scholar]
  28. Zhu W, Sun L, Zhao P, Liu Y, Zhang J, Zhang Y, et al. MIF facilitates the therapeutic efficacy of mesenchymal stem cells derived exosomes in acute myocardial infarction through up-regulating miR-133a-3p. Res Sq [Preprint]. December 7, 2020. Available from: https://europepmc.org/article/ppr/ppr250344.
  29. Venkat P, Chen J, Chopp M. Exosome-mediated amplification of endogenous brain repair mechanisms and brain and systemic organ interaction in modulating neurological outcome after stroke. J Cereb Blood Flow Metab 2018; 38(12):2165-78. doi: 10.1177/0271678x18782789 [Crossref] [ Google Scholar]
  30. Xin H, Katakowski M, Wang F, Qian JY, Liu XS, Ali MM. MicroRNA cluster miR-17-92 cluster in exosomes enhance neuroplasticity and functional recovery after stroke in rats. Stroke 2017; 48(3):747-53. doi: 10.1161/strokeaha.116.015204 [Crossref] [ Google Scholar]
  31. Xin H, Wang F, Li Y, Lu QE, Cheung WL, Zhang Y. Secondary release of exosomes from astrocytes contributes to the increase in neural plasticity and improvement of functional recovery after stroke in rats treated with exosomes harvested from microRNA 133b-overexpressing multipotent mesenchymal stromal cells. Cell Transplant 2017; 26(2):243-57. doi: 10.3727/096368916x693031 [Crossref] [ Google Scholar]
  32. Fu DL, Jiang H, Li CY, Gao T, Liu MR, Li HW. MicroRNA-338 in MSCs-derived exosomes inhibits cardiomyocyte apoptosis in myocardial infarction. Eur Rev Med Pharmacol Sci 2020; 24(19):10107-17. doi: 10.26355/eurrev_202010_23230 [Crossref] [ Google Scholar]
  33. Li Y, Liu T, Li Y, Han D, Hong J, Yang N. Baicalin ameliorates cognitive impairment and protects microglia from LPS-induced neuroinflammation via the SIRT1/HMGB1 pathway. Oxid Med Cell Longev 2020; 2020:4751349. doi: 10.1155/2020/4751349 [Crossref] [ Google Scholar]
  34. Liu J, Huang J, Zhang Z, Zhang R, Sun Q, Zhang Z. Mesenchymal stem cell-derived exosomes ameliorate delayed neurocognitive recovery in aged mice by inhibiting hippocampus ferroptosis via activating SIRT1/Nrf2/HO-1 signaling pathway. Oxid Med Cell Longev 2022; 2022:3593294. doi: 10.1155/2022/3593294 [Crossref] [ Google Scholar]
  35. Rapicavoli NA, Qu K, Zhang J, Mikhail M, Laberge RM, Chang HY. A mammalian pseudogene lncRNA at the interface of inflammation and anti-inflammatory therapeutics. Elife 2013; 2:e00762. doi: 10.7554/eLife.00762 [Crossref] [ Google Scholar]
  36. Zhu B, Zhang L, Liang C, Liu B, Pan X, Wang Y. Stem cell-derived exosomes prevent aging-induced cardiac dysfunction through a novel exosome/lncRNA MALAT1/NF-κB/TNF-α signaling pathway. Oxid Med Cell Longev 2019; 2019:9739258. doi: 10.1155/2019/9739258 [Crossref] [ Google Scholar]
  37. Lee BR, Kim JH, Choi ES, Cho JH, Kim E. Effect of young exosomes injected in aged mice. Int J Nanomedicine 2018; 13:5335-45. doi: 10.2147/ijn.s170680 [Crossref] [ Google Scholar]
  38. Hornsby PJ. Telomerase and the aging process. Exp Gerontol 2007; 42(7):575-81. doi: 10.1016/j.exger.2007.03.007 [Crossref] [ Google Scholar]
  39. Yang W, Zhang J, Xu B, He Y, Liu W, Li J. HucMSC-derived exosomes mitigate the age-related retardation of fertility in female mice. Mol Ther 2020; 28(4):1200-13. doi: 10.1016/j.ymthe.2020.02.003 [Crossref] [ Google Scholar]
  40. Yang J, Zhang X, Chen X, Wang L, Yang G. Exosome mediated delivery of miR-124 promotes neurogenesis after ischemia. Mol Ther Nucleic Acids 2017; 7:278-87. doi: 10.1016/j.omtn.2017.04.010 [Crossref] [ Google Scholar]
  41. Zhang Y, Chopp M, Zhang ZG, Katakowski M, Xin H, Qu C. Systemic administration of cell-free exosomes generated by human bone marrow derived mesenchymal stem cells cultured under 2D and 3D conditions improves functional recovery in rats after traumatic brain injury. Neurochem Int 2017; 111:69-81. doi: 10.1016/j.neuint.2016.08.003 [Crossref] [ Google Scholar]
  42. Zhang N, Zhu J, Ma Q, Zhao Y, Wang Y, Hu X. Exosomes derived from human umbilical cord MSCs rejuvenate aged MSCs and enhance their functions for myocardial repair. Stem Cell Res Ther 2020; 11(1):273. doi: 10.1186/s13287-020-01782-9 [Crossref] [ Google Scholar]
  43. Tigges J, Krutmann J, Fritsche E, Haendeler J, Schaal H, Fischer JW. The hallmarks of fibroblast ageing. Mech Ageing Dev 2014; 138:26-44. doi: 10.1016/j.mad.2014.03.004 [Crossref] [ Google Scholar]
  44. Keyes BE, Segal JP, Heller E, Lien WH, Chang CY, Guo X. Nfatc1 orchestrates aging in hair follicle stem cells. Proc Natl Acad Sci U S A 2013; 110(51):E4950-9. doi: 10.1073/pnas.1320301110 [Crossref] [ Google Scholar]
  45. Keyes BE, Liu S, Asare A, Naik S, Levorse J, Polak L, et al. Impaired epidermal to dendritic T cell signaling slows wound repair in aged skin. Cell 2016;167(5):1323-38.e14. 10.1016/j.cell.2016.10.052.
  46. Mascharak S, desJardins-Park HE, Davitt MF, Griffin M, Borrelli MR, Moore AL. Preventing Engrailed-1 activation in fibroblasts yields wound regeneration without scarring. Science 2021; 372(6540):eaba2374. doi: 10.1126/science.aba2374 [Crossref] [ Google Scholar]
  47. Xia W, Li M, Jiang X, Huang X, Gu S, Ye J. Young fibroblast-derived exosomal microRNA-125b transfers beneficial effects on aged cutaneous wound healing. J Nanobiotechnology 2022; 20(1):144. doi: 10.1186/s12951-022-01348-2 [Crossref] [ Google Scholar]
  48. Oh M, Lee J, Kim YJ, Rhee WJ, Park JH. Exosomes derived from human induced pluripotent stem cells ameliorate the aging of skin fibroblasts. Int J Mol Sci 2018; 19(6):1715. doi: 10.3390/ijms19061715 [Crossref] [ Google Scholar]
  49. Chernoff G. The utilization of human placental mesenchymal stem cell derived exosomes in aging skin: an investigational pilot study. J Surg 2021; 6:1388. doi: 10.29011/2575-9760.001388 [Crossref] [ Google Scholar]
  50. Safiri S, Noori M, Nejadghaderi SA, Mousavi SE, Sullman MJM, Araj-Khodaei M. The burden of Alzheimer’s disease and other types of dementia in the Middle East and North Africa region, 1990-2019. Age Ageing 2023; 52(3):afad042. doi: 10.1093/ageing/afad042 [Crossref] [ Google Scholar]
  51. Hosseini L, Mahmoudi J, Pashazadeh F, Salehi-Pourmehr H, Sadigh-Eteghad S. Protective effects of nicotinamide adenine dinucleotide and related precursors in Alzheimer’s disease: a systematic review of preclinical studies. J Mol Neurosci 2021; 71(7):1425-35. doi: 10.1007/s12031-021-01842-6 [Crossref] [ Google Scholar]
  52. Cone AS, Yuan X, Sun L, Duke LC, Vreones MP, Carrier AN. Mesenchymal stem cell-derived extracellular vesicles ameliorate Alzheimer’s disease-like phenotypes in a preclinical mouse model. Theranostics 2021; 11(17):8129-42. doi: 10.7150/thno.62069 [Crossref] [ Google Scholar]
  53. Ding M, Shen Y, Wang P, Xie Z, Xu S, Zhu Z. Exosomes isolated from human umbilical cord mesenchymal stem cells alleviate neuroinflammation and reduce amyloid-beta deposition by modulating microglial activation in Alzheimer’s disease. Neurochem Res 2018; 43(11):2165-77. doi: 10.1007/s11064-018-2641-5 [Crossref] [ Google Scholar]
  54. Lee M, Ban JJ, Yang S, Im W, Kim M. The exosome of adipose-derived stem cells reduces β-amyloid pathology and apoptosis of neuronal cells derived from the transgenic mouse model of Alzheimer’s disease. Brain Res 2018; 1691:87-93. doi: 10.1016/j.brainres.2018.03.034 [Crossref] [ Google Scholar]
  55. Wang H, Sui H, Zheng Y, Jiang Y, Shi Y, Liang J. Curcumin-primed exosomes potently ameliorate cognitive function in AD mice by inhibiting hyperphosphorylation of the Tau protein through the AKT/GSK-3β pathway. Nanoscale 2019; 11(15):7481-96. doi: 10.1039/c9nr01255a [Crossref] [ Google Scholar]
  56. Nila IS, Sumsuzzman DM, Khan ZA, Jung JH, Kazema AS, Kim SJ. Identification of exosomal biomarkers and its optimal isolation and detection method for the diagnosis of Parkinson’s disease: a systematic review and meta-analysis. Ageing Res Rev 2022; 82:101764. doi: 10.1016/j.arr.2022.101764 [Crossref] [ Google Scholar]
  57. Jarmalavičiūtė A, Tunaitis V, Pivoraitė U, Venalis A, Pivoriūnas A. Exosomes from dental pulp stem cells rescue human dopaminergic neurons from 6-hydroxy-dopamine-induced apoptosis. Cytotherapy 2015; 17(7):932-9. doi: 10.1016/j.jcyt.2014.07.013 [Crossref] [ Google Scholar]
  58. Narbute K, Piļipenko V, Pupure J, Dzirkale Z, Jonavičė U, Tunaitis V. Intranasal administration of extracellular vesicles derived from human teeth stem cells improves motor symptoms and normalizes tyrosine hydroxylase expression in the substantia nigra and striatum of the 6-hydroxydopamine-treated rats. Stem Cells Transl Med 2019; 8(5):490-9. doi: 10.1002/sctm.18-0162 [Crossref] [ Google Scholar]
  59. Yang J, Luo S, Zhang J, Yu T, Fu Z, Zheng Y. Exosome-mediated delivery of antisense oligonucleotides targeting α-synuclein ameliorates the pathology in a mouse model of Parkinson’s disease. Neurobiol Dis 2021; 148:105218. doi: 10.1016/j.nbd.2020.105218 [Crossref] [ Google Scholar]
  60. de Lange-Brokaar BJ, Ioan-Facsinay A, van Osch GJ, Zuurmond AM, Schoones J, Toes RE. Synovial inflammation, immune cells and their cytokines in osteoarthritis: a review. Osteoarthritis Cartilage 2012; 20(12):1484-99. doi: 10.1016/j.joca.2012.08.027 [Crossref] [ Google Scholar]
  61. Zhang S, Teo KYW, Chuah SJ, Lai RC, Lim SK, Toh WS. MSC exosomes alleviate temporomandibular joint osteoarthritis by attenuating inflammation and restoring matrix homeostasis. Biomaterials 2019; 200:35-47. doi: 10.1016/j.biomaterials.2019.02.006 [Crossref] [ Google Scholar]
  62. He L, He T, Xing J, Zhou Q, Fan L, Liu C. Bone marrow mesenchymal stem cell-derived exosomes protect cartilage damage and relieve knee osteoarthritis pain in a rat model of osteoarthritis. Stem Cell Res Ther 2020; 11(1):276. doi: 10.1186/s13287-020-01781-w [Crossref] [ Google Scholar]
  63. Zheng L, Wang Y, Qiu P, Xia C, Fang Y, Mei S. Primary chondrocyte exosomes mediate osteoarthritis progression by regulating mitochondrion and immune reactivity. Nanomedicine (Lond) 2019; 14(24):3193-212. doi: 10.2217/nnm-2018-0498 [Crossref] [ Google Scholar]
  64. Mihanfar A, Shakouri SK, Khadem-Ansari MH, Fattahi A, Latifi Z, Nejabati HR. Exosomal miRNAs in osteoarthritis. Mol Biol Rep 2020; 47(6):4737-48. doi: 10.1007/s11033-020-05443-1 [Crossref] [ Google Scholar]
  65. Tofiño-Vian M, Guillén MI, Pérez Del Caz MD, Castejón MA, Alcaraz MJ. Extracellular vesicles from adipose-derived mesenchymal stem cells downregulate senescence features in osteoarthritic osteoblasts. Oxid Med Cell Longev 2017; 2017:7197598. doi: 10.1155/2017/7197598 [Crossref] [ Google Scholar]
  66. Ding M, Shen Y, Wang P, Xie Z, Xu S, Zhu Z. Exosomes isolated from human umbilical cord mesenchymal stem cells alleviate neuroinflammation and reduce amyloid-beta deposition by modulating microglial activation in Alzheimer’s disease. Neurochem Res 2018; 43(11):2165-77. doi: 10.1007/s11064-018-2641-5 [Crossref] [ Google Scholar]
Submitted: 05 Jul 2023
Revised: 22 Sep 2023
Accepted: 27 Sep 2023
First published online: 30 Oct 2023
EndNote EndNote

(Enw Format - Win & Mac)

BibTeX BibTeX

(Bib Format - Win & Mac)

Bookends Bookends

(Ris Format - Mac only)

EasyBib EasyBib

(Ris Format - Win & Mac)

Medlars Medlars

(Txt Format - Win & Mac)

Mendeley Web Mendeley Web
Mendeley Mendeley

(Ris Format - Win & Mac)

Papers Papers

(Ris Format - Win & Mac)

ProCite ProCite

(Ris Format - Win & Mac)

Reference Manager Reference Manager

(Ris Format - Win only)

Refworks Refworks

(Refworks Format - Win & Mac)

Zotero Zotero

(Ris Format - FireFox Plugin)

Abstract View: 982
PDF Download: 797
Full Text View: 23