Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
World Academy of Sciences Journal
Join Editorial Board Propose a Special Issue
Print ISSN: 2632-2900 Online ISSN: 2632-2919
Journal Cover
January-February 2026 Volume 8 Issue 1

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
January-February 2026 Volume 8 Issue 1

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review Open Access

Mesenchymal stem cells and the rescue of mitochondrial damage in different disease models (Review)

  • Authors:
    • Canh Minh Tran
    • Van T.t. Nguyen
  • View Affiliations / Copyright

    Affiliations: VNUHCM‑US Stem Cell Institute, University of Science, Vietnam National University, Ho Chi Minh City 70000, Vietnam
    Copyright: © Tran et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].
  • Article Number: 4
    |
    Published online on: November 24, 2025
       https://doi.org/10.3892/wasj.2025.419
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Mitochondrial transplantation is based on the fundamental principle of mitochondrial transfer, through which recipient cells take up exogenous mitochondria, thereby restoring cellular function. Several studies have demonstrated that isolated mitochondria can effectively improve cellular function in various diseases characterized by mitochondrial dysfunction as a key factor in pathological progression. The success of mitochondrial therapy depends on the source of donor cells, the quality and quantity of mitochondria, and their competency and compatibility with recipient cells. Mesenchymal stem cells (MSCs) are highly proliferative and possess the potential to differentiate into various cell types, a process requiring competent mitochondria to supply energy and essential building blocks for cellular activities. Consequently, MSC‑derived mitochondria exhibit high plasticity and functional capability, rendering them promising candidates for mitochondrial transplantation therapy. The present review discusses the role of mitochondria in health and disease progression. The present review also summarizes current evidence from the literature, highlighting the emerging role of MSCs as a valuable source for mitochondrial therapy in both in vitro and in vivo pathological models. Given the diverse sources available for MSC isolation, mitochondrial therapy, particularly when combined with stem cell technology, represents a potential avenue for future research in therapeutic and pharmacological development.

1. Introduction

Mitochondria are the powerhouses of mammalian cells, providing the energy and materials required for cellular function. Mitochondria utilize pyruvate, products from glycolysis, amino acid metabolism and fatty acid oxidation to transfer electrons through the electron transport chain, synthesizing energy in the form of adenosine triphosphate (ATP). Mitochondrial dysfunction causes bioenergetic imbalance and increases oxidative stress, which paves the way for pathogenesis to occur. Multiple diseases arise from mitochondrial damage or are a consequence of impaired mitochondria due to tissue injury. Aging and degenerative diseases are common examples of conditions involving mitochondrial dysfunction. While genetic modifications and the reduction of telomere length are definite hallmarks of aging, impaired mitochondrial function and the accumulation of mitochondrial DNA (mtDNA) mutations critically determine healthy aging outcomes and contribute to longevity (1). Degenerative diseases, such as brain, muscle and macular degeneration are consequences of mitochondrial dysfunction. Modifications in mitochondrial membrane potential, reduced calcium homeostasis and decreased ATP production are associated with mitochondrial dynamic dysregulation and increased levels of reactive oxygen species (ROS). This leads to protein degradation, the loss of intercellular interactions and eventually, apoptosis (2-5). For instance, in the immune system, reduced mitochondrial respiration and metabolism are associated with immune paralysis following septic shock (6,7). In metabolic syndromes, such as type 2 diabetes, mitochondrial oxidative stress promotes glucose intolerance and excessive lipid accumulation in adipocytes, and drives systemic inflammation, which further accelerates insulin resistance (8-10). Mitochondria have also been an unrecognized factor in the progression of skin diseases (11). In pulmonary diseases derived from smoking, cigarette smoke directly blocks complexes I and II, causes mitochondrial depolarisation through altered mitophagy, and induces mitochondrial ROS generation. This consequently results in the apoptosis and necrosis of pulmonary endothelial cells, contributing to the development of chronic and fibrotic conditions (12-14). In addition, mitochondria exhibiting a low oxygen consumption rate, high levels of mtDNA damage and ROS production are observed in the vascular endothelial cells of preterm infants with bronchopulmonary dysplasia and adverse outcomes (15). Mitochondria are also a key site for calcium homeostasis, as they buffer cytoplasmic calcium ions through interactions with the endoplasmic reticulum (16). The disruption of calcium balance may interfere with mitochondrial respiration and ATP production, leading to the development of pathological conditions and diseases, such as neuron degeneration, cancers, diabetes, and muscular and cardiac dystrophy (17-19). Thus, the ability of cells to maintain competent mitochondrial function and capacity is key to preventing disease progression and negative outcomes.

Mitochondrial transfer refers to the process through which cells exchange mitochondria, serving as a form of cell-cell communication. This transfer may involve the sharing of damaged mitochondria to signal neighboring cells about oxidative damage, while surrounding cells offer functional mitochondria to stressed cells. Receiving mitochondria supports cells in recovering from oxidative stress, improving cellular respiration and ATP production, and consequently, reversing the effects of cellular dysfunction and tissue damage (20-22). The mechanisms of intercellular mitochondrial transfer have been well established and are illustrated in Fig. 1A. The application of mitochondrial transfer has been transformed into mitochondrial transplantation therapy (Fig. 1B), in which exogenous mitochondria are isolated and artificially transferred into cells or tissues. Mitochondrial therapy, which encompasses both mitochondrial transfer and transplantation, is effective in improving cell metabolism, restoring cells from stress and preventing tissue degeneration (23-25). Early clinical trials of mitochondrial therapy have been conducted in ischemic diseases, demonstrating that the injection of exogenous mitochondria effectively improves the conditions of patients and reduces the recovery time (26,27). These data indicate that mitochondrial therapy holds potential for future applications. However, the autologous transfer of mitochondria may be invasive and non-accessible in a number of cases; thus, a source of qualified mitochondrial donors is critical.

Mechanisms of mitochondrial transfer
in the (A) microenvironment and (B) artificial mitochondrial
transplantation. (A) Mitochondrial transfer in the microenvironment
is facilitated via multiple mechanisms. TNTs form a channel for
mitochondria trafficking between cells at certain distances. Cells
in close contact can exchange mitochondria via gap junctions on the
cell membrane or even fuse their membranes (cell fusion) to allow
mitochondrial transfer. Mitochondria can be released via
extracellular vesicles or dispersed freely in the microenvironment
and taken up by other cells. (B) Mitochondrial transplantation
technologies include forced centrifugation, in which centrifugation
force increases the mitochondria-membrane contact and induces
penetration through the membrane. Magnetomitotransfer enhances
penetration by magnetic force. Penetrating peptides wrapping around
mitochondria allow mitochondrial transition through the cell
membrane. Droplet microfluidics creates isolated spaces, allowing
close contact between cells and isolated mitochondria, thereby
enhancing the chances of mitochondrial intake. TNT, tunneling
nanotube.

Figure 1

Mechanisms of mitochondrial transfer in the (A) microenvironment and (B) artificial mitochondrial transplantation. (A) Mitochondrial transfer in the microenvironment is facilitated via multiple mechanisms. TNTs form a channel for mitochondria trafficking between cells at certain distances. Cells in close contact can exchange mitochondria via gap junctions on the cell membrane or even fuse their membranes (cell fusion) to allow mitochondrial transfer. Mitochondria can be released via extracellular vesicles or dispersed freely in the microenvironment and taken up by other cells. (B) Mitochondrial transplantation technologies include forced centrifugation, in which centrifugation force increases the mitochondria-membrane contact and induces penetration through the membrane. Magnetomitotransfer enhances penetration by magnetic force. Penetrating peptides wrapping around mitochondria allow mitochondrial transition through the cell membrane. Droplet microfluidics creates isolated spaces, allowing close contact between cells and isolated mitochondria, thereby enhancing the chances of mitochondrial intake. TNT, tunneling nanotube.

Mesenchymal stem cells (MSCs) feature high plasticity and proliferation, which demonstrates their inherent mitochondrial capacity. The ability to acquire, handle and expand MSCs in large quantities in culture, along with a stable and reliable assessment of mitochondrial quality, may further facilitate access to mitochondrial transplantation in various clinical settings, rendering MSCs promising for such therapeutic purposes. The use of MSCs as a mitochondrial source has been shown in in vitro and in vivo models of different diseases, indicating that mitochondria isolated from MSCs are efficient and safe for translational studies (Fig. 2) (28-30). The present review summarizes data from numerous studies in the literature on mitochondrial transfer using MSCs as mitochondrial donors. Detailed descriptions of the mechanisms underlying mitochondrial therapy are also included. The present review also provides an update on the broad application of mitochondrial donation in various pathological conditions and diseases, indicating that MSCs from different origins are valuable sources for mitochondrial collection and transplantation technology.

MSC sources for mitochondrial therapy
and the associated recipient cells/tissues. Mitochondria were
isolated from selected sources of MSCs and transferred/transplanted
into different cell types and tissues. The effectiveness of
mitochondrial therapy has been examined in various pathological
models. MSCs, mesenchymal stem cells.

Figure 2

MSC sources for mitochondrial therapy and the associated recipient cells/tissues. Mitochondria were isolated from selected sources of MSCs and transferred/transplanted into different cell types and tissues. The effectiveness of mitochondrial therapy has been examined in various pathological models. MSCs, mesenchymal stem cells.

2. Mitochondrial transfer from mesenchymal stem cells into different disease models

In pulmonary tissues and lung diseases

Asthma is a chronic condition involving a persistent inflammatory disorder in the airways (31). Mitochondrial dysfunction causes oxidative stress and reduced ATP synthase activity, leading to apoptosis and damage to airway epithelial cells, which results in allergic responses and inflammation (32,33). Through tunneling nanotubes (TNTs), mitochondria transferred from MSCs have been shown to protect the human bronchial epithelium cell line (BEAS-2B) against mitochondrial dysfunction and apoptosis induced by cobalt(II) chloride (CoCl2), which has been used as a hypoxia-mimicking agent (34). Connexin43 (Cx43), a transmembrane protein encoded by the GJA1 gene, is essential for TNT formation and induced pluripotent stem cell (iPSC)-MSC-mediated mitochondrial transfer. This transfer alleviates the symptoms of allergic airway inflammation by recovering mitochondrial function in the bronchi and inhibiting programmed cell death in the lungs (34). The positive effects of mitochondrial transfer have also been reported when inducing airway inflammation by ovalbumin, specifically via the restoration of mitochondrial membrane potential (MMP) and the inhibition of caspase-3 and -9(34).

The dysfunction of mitochondria, which is associated with changes in mitochondrial morphology and homeostasis and the disruption of MMP in lung cells, is one of the main causes of pulmonary fibrosis (PF) (35). ROS overproduction in the mitochondria is associated with mitochondrial dysfunction, leading to lung damage and inflammatory responses, which contributes to the pathogenesis of PF (36). Increasing mitochondrial biogenesis within human MSCs (hMSCs) using pioglitazone and iron oxide nanoparticles has been shown to enhance mitochondrial transfer to mouse alveolar epithelial cells (TC-1) damaged by bleomycin (BLM), a treatment that induces mtDNA strand breaks and mitochondrial respiratory chain dysfunction (37). TC-1 cells damaged by BLM exhibit restored ATP levels, reduced intracellular ROS levels and recover MMP when they receive functional mitochondria from hMSCs (37).

In the model of chronic obstructive pulmonary disease (COPD), mitochondrial dysfunction due to oxidative stress contributes to tissue inflammation and remodeling (38). Oxidative stress accelerates the inflammatory process by stimulating neutrophils and macrophages (39), leading to apoptosis, autophagy and cellular senescence in the airways (38). Smoking is a well-known contributing factor to the development of COPD and airway structure remodeling. Cigarette smoke medium (CSM) has been shown to alter mitochondrial metabolic regulation, leading to glycolysis and ROS production (40,41). It was previously demonstrated that the transfer of mitochondria from iPSC-MSCs reduced intracellular ROS levels and prevented MMP impairment in airway smooth muscle cells induced by CSM, significantly decreasing apoptosis by 50% (42). Antoehr study demonstrated that BEAS-2B cells treated with 2% CSM for 24 h induced significant inflammatory responses and oxidative stress without leading to cell death, and also received mitochondria from bone marrow-derived MSCs (BM-MSCs) (43). Mitochondrial transfer from BM-MSCs to BEAS-2B cells treated with CSM showed recovery in intracellular ATP levels (43). This evidence indicated that transferring mitochondria from MSCs can restore mitochondrial dysfunction in the respiratory system by preventing oxidative stress, reducing epithelial cell death, and airway inflammation, and is a potential supportive method in pulmonary diseases.

In damaged cardiac tissue

In cardiac tissue, damage to cardiac muscles can initiate numerous cardiac issues. For example, progressive myocardial damage can be caused by the use of anthracyclines, including doxorubicin, which are commonly used as drugs for the treatment of cancer. Mitochondrial biogenesis inhibition has been shown to contribute to this damage (44). The overproduction of ROS caused by doxorubicin induces mitochondrial dysfunction through the impairment of MMP, the opening of mitochondrial permeability transition pore (mPTP), and the alteration of mitochondrial morphology in human vascular endothelial cells (45). Mitochondrial dysfunction caused by doxorubicin has a significant effect on cardiomyocytes as mitochondria comprise 40% of the volume of each cardiomyocyte, and the majority of energy in these cells is generated through mitochondrial respiration (46). In this context, the supplementation of fresh mitochondria could be beneficial. Evidence has indicated that the injection of autologous mitochondria isolated from skeletal muscles into the ischemic hearts of pediatric patients effectively helped them separate from extracorporeal membrane oxygenation (27). However, isolating mitochondria from skeletal muscles may be invasive and impossible in some cases. Thus, a readily available source of healthy mitochondria for transplantation is expected to advance current treatment. As such, MSCs appear as a potential source for mitochondria isolation due to their rapid proliferation. Several studies have demonstrated that MSCs may be a safe source for mitochondrial transfer in cardiovascular conditions (Table I). A previous study demonstrated that functional mitochondrial transfer by iPSC-MSCs protected against doxorubicin-induced damage in cardiomyocytes from neonatal mice, resulting in a marked recovery of the mitochondrial oxygen consumption rate (OCR), ATP production, and optimal mitochondrial respiration in cardiomyocytes (47). It was noted that this effect was distinct from the paracrine activity of MSCs and involved mitochondrial Rho GTPase 1 (Miro1), an outer mitochondrial membrane protein, in the formation of TNTs (47). In another study, the transplantation of mitochondria derived from MSCs reduced apoptosis and ROS levels, while increasing ATP production in cardiomyocytes treated with doxorubicin (48).

Table I

Summary of recipient cell types receiving mitochondrial transfer from MSCs.

Table I

Summary of recipient cell types receiving mitochondrial transfer from MSCs.

System/tissueTarget diseaseDefectsDonor cellsRecipient cellsEffects(Refs.)
PulmonaryAsthmaCoCl2, OVAMSCs, iPSC-MSCsBEAS-2B, hyperplastic goblet cells- Restoration of mitochondrial function(34)
     - Inhibition of apoptosis. 
 Pulmonary fibrosisBLMhMSCs derived from the placentaTC-1 cells- Restoration of mitochondrial function, reduction of ROS.(37)
 Chronic obstructive pulmonaryCSMiPSC-MSCsASMCs- Restoration of mitochondrial function.(42,43)
     - Inhibition of apoptosis. 
     - Reduction of inflammatory response and oxidative stress. 
CardiacMyocardia damagedDoxorubiciniPSC-MSCsNMCs, HL-1 cells- Restoration of mitochondrial function.(47)
     - Inhibition of apoptosis. 
     - Regulation of autophagy. 
 IschemicPhosphatidylserine, hypoxia; H2O2BM-MSCs, ADSCs; hADSCsH9c2 cells, rCMs, RL4 cells, HUVECs- Restoration of mitochondrial function.(53-55)
     - Reduction of collagen accumulation. 
     - Regulation of autophagy. 
     - Regulation of the HO-1 expression. 
MusculoskeletalOsteoarthritisOligomycin, rotenoneBM-MSCs, MSCsChondrocytes- Restoration of mitochondrial function.(61,63)
     - Enhancement of collagen and proteo- glycan production. 
 Tendon injuries H2O2, TNF-αUC-MSCs, BM-MSCsTenocytes- Restoration of mitochondrial function.(70,71)
     - Inhibition of apoptosis. 
     - Enhancement of collagen production. 
     - Reduction of inflammation responses. 
 Skeletal muscle atrophyDexamethasoneUC-MSCs, MuSCsMuscle cells- Restoration of mitochondrial function.(81,79)
     - Restoration of muscle bioenergetics. 
BrainBrain ischemic, strokeOGD, H2O2MMSCs, BM-MSCsAstrocytes, VSC4.1 cells, neuron-like p0 PC12 pheochromocytoma cells- Restoration of mitochondrial function.(88,89,91)
     - Inhibition of apoptosis. 
     - Reduction of inflammation responses. 
 Spinal cord injuryRsl3UC-MSCsHt22- Restoration of mitochondrial function.(98)
     - Reduction of the total intracellular iron and free Fe2+ level. 
 Parkinson's diseaseMPP+, 6-OHDA, rotenoneUC-MSCs, iPSC- MSCsDA neurons- Reduced cell death.(104,106)
     - Decreased expression of pro-inflammatory cytokines. 
     - Restored intracellular ATP levels. 
 Side-effects of chemothera-peuticsCisplatinMSCsNSCs- Restoration of mitochondrial function.(109)
     - Promotion of the survival of NSCs. 
EyeOcular diseaseRotenone. Compensates for mitochondrial deficiencyMSCs; iPSC-MSCshCECs; neuronal cells in the retinal ganglion cell layer.- Restoration of mitochondrial function.(111,114,119)
     - Regulation of metabolic processes. 
     - Regulation of Müller cells activation. 
     - Reduction of inflammation responses 
 Mitochondrial mutation diseasemtDNA mutationWJMSCs, RECsMFHi, MELAS neurons, MERRF cybrids- Restoration of mitochondrial function.(155,156,158)
     - Regulation of metabolic processes. 
     - Inhibition of apoptosis. 
     - Promote cell proliferation. 
Metabolic diseasesDiabetic nephropathySTZBM-MSCsProximal tubular endothelial cells (PTECs)- Inhibition of apoptosis.(29)
     - Reduction of oxidative stress. 
     - Regulation of SOD2 protein family expression 
     - Regulation of glucose transporter protein family expression. 
 NASHHFDBM-MSCsMouse hepatocytes- Restoration of mitochondrial function.(141,140)
     - Regulation of MDA expression. 
 SAPHypoxia (5% O2), sodium taurocholateUC-MSCsRat pancreatic acinar cells- Restoration of mitochondrial function.(146)
     - Reversal of the reduction of glycerol 3-phosphate, 3-phosphoglyceric acid, and phosphoenolpyruvate and the accumulation of citric. 
     - Increase glycolytic flux. 
  Hypoxia (1% O2)ADSCsIslet cells- Restoration of mitochondrial function.(151)
     - Control of insulin production. 
Engraftment function of stem cells  H2O2, under serum starvationhMSCs, ADSCs, BM-MSCs.hMSCs, ADSCs, BM-MSCs.- Restoration of mitochondrial function.(169,167,170)
     - Promote cell proliferation. 
     - Enhance stress resistance and anti-aging. 
     - Promote cell cycle progression into the S and G2/M phases. 
ImmuneAutoimmune UC-MSCs, BM-MSCs, MSCs, ASCsT CD3+ cells, Th17 cells, T CD4+ cells, Tregs- Differentiate into Tregs.(173,174,178,176)
     - Control autoimmunity. 
     - Regulate the activity of Th17 cells. 
     - Regulate the activity of T CD4+ cells. 
     - Maintain mitochondrial function in Treg cells. 
     - Enhance Treg function. 
 Diabetes mellitushigh glucoseMSCsRAW264.7 cells- Restoration of mitochondrial function.(185)
     - Regulation of metabolic processes. 
 Acute and chronic inflammatory responses in the testiculartesticular torsionSLCsmacrophages- Reduction of oxidative stress.(249)
     - Enhance testosterone production. 
     - Enhance sperm fertilization capability. 
 Acute and chronic inflammatory responses in the lungLPS, E. coliBM-MSCs, ADSCsAMs, MDMs, macrophages- Enhancement of mitochondrial function.(187-189,190,192)
     - Enhance the phagocytic ability of macrophages. 
     - Induce differentiation into the M2 phenotype of macrophages. 
 Acute and chronic inflammatory responsesCholesterolMSCs7-ketocholesterol- load macrophages- Enhance the phagocytic ability of macrophages.(250)
     - Reduction of lipid content. 
     - Enhance energy in macrophages. 

[i] CoCl2, Cobalt(II) chloride; OVA, ovalbumin; MSCs, mesenchymal stem cells; iPSC, induced pluripotent stem cell; BLM, bleomycin; CSM, cigarette smoke medium; ROS, reactive oxygen species; ASMCs, airway smooth muscle cells; NMCs, neonatal mouse cardiomyocytes; BM-MSCs, bone marrow-derived MSCs; rCMs, rat cardiomyocytes; H2O2, hydrogen peroxide; HUVECs, human umbilical vein endothelial cells; UC-MSCs, umbilical cord-MSCs; MuSCs, muscle stem cells; MMP+,1-methyl-4-phenylpyridinium; 6-OHDA, 6-hydroxydopamine; mtDNA, mitochondrial DNA; NSCs, neural stem cells; hCECs, Human corneal endothelial cells; WJMSCs, Wharton's Jelly-derived MSCs; MELAS, myopathy, encephalomyopathy with lactic acidosis, and stroke-like episodes; MERRF, myoclonus epilepsy with ragged red fiber syndrome; STZ, streptozotocin; NASH, non-alcoholic steatohepatitis; HFD, high-fat diet; SAP, severe acute pancreatitis; ASCs, adipose-derived stem cells; SLCs, stem Leydig cells.

Mitochondrial dysfunction is also a key event in simulated ischemia/reperfusion injury (IRI) (49). Abnormal mitochondrial morphology and irregularities in mitochondrial quality control have been identified as cellular mechanisms leading to IRI (50). Mitochondrial ROS production is a primary target of damage caused by hypoxia (51). In an ischemic model, MSCs were shown to reduce the death of H9c2 cardiomyocytes when co-cultured under hypoxic conditions for 150 min via the transfer of healthy mitochondria (52). In another study, BM-MSCs delivered functional mitochondria to injured H9c2 rat ventricular cells, which were cultured in a medium deprived of serum and glucose and incubated under hypoxic conditions for 12 h (53). Co-culturing injured H9c2 cells with BM-MSCs showed significant downregulation of Bax expression and an upregulation of B-cell lymphoma (Bcl-2) expression (53). Furthermore, BM-MSCs also decreased the caspase-3 levels induced by SI/R in H9c2 cells, as this enzyme is an executor of the apoptosis process (53). However, when BM-MSCs were treated with latrunculin A (LatA), which is known to inhibit TNT formation, apoptosis increased (53), suggesting that TNT formation is critical. In another study, when co-culturing human adipose-derived stem cells (hADSCs) with rat cardiomyocytes for 24 h under hypoxic conditions, functional ADSC-derived mitochondria enhanced the OCR of rat cardiomyocytes. This effect was shown to be independent of the paracrine effects from ADSCs (54). On the other hand, mitochondria released by damaged somatic cells, such as hydrogen peroxide (H2O2)-treated RL14 cardiomyocytes or human umbilical vein endothelial cells (HUVECs), activate MSCs to transfer healthy mitochondria toward dying cells (55). Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme metabolism and exerts a wide range of anti-inflammatory, anti-apoptotic, and immunoregulatory effects in various diseases (56). Co-culturing MSCs with RL4 cells damaged by H2O2 and HUVECs leads to the increased expression of HO-1 and enhances HO-1 enzyme activity in MSCs (55). These data indicate that MSCs are promising sources of mitochondria that can be used for mitochondrial transplantation technologies.

In disorders of the musculoskeletal system

Osteoarthritis (OA) is a common musculoskeletal disorder related to the aging process, which is characterized by cartilage degradation, joint pain, and impaired function (57). Chondrocytes play a functional role in the synthesis, deposition and formation of the extracellular matrix of cartilage tissue (57). The mitochondria of chondrocytes in the OA model exhibit a reduced activity of mitochondrial respiratory chain (MRC) enzymes (complex I, II, III, IV and ATP synthase), along with decreased MMP (58). Furthermore, mitochondria exhibiting a swollen morphology and loss of cristae regularity have also been observed in chondrocytes in models of OA (58). Mitochondrial dysfunction and energy metabolism disruption markedly contribute to the pathogenesis of OA, including cartilage degeneration, increased synovial inflammation and the calcification of the cartilage matrix (59). The abnormal alteration in the metabolic process of cartilage cells is a response to inflammation and is correlated with the process of cartilage degeneration (60). In a previous study, the bilateral anterior cruciate ligament and medial meniscus were resected in 8-week-old rats to induce OA pathogenesis (61). Functional mitochondria from BM-MSCs rescued OA chondrocytes from mitochondrial dysfunction by restoring the normal morphology of mitochondria, MMP, MRCI, II, and III activity, citrate synthase, and ATP content (61). Another study indicated that mitochondrial dysfunction in chondrocytes in OA is characterized by a reduced type II collagen secretion (58). It should be noted that type II collagen and proteoglycans are key components of cartilage tissue (62). OA chondrocytes co-cultured with MSCs showed a significant increase in the concentration of type II collagen and proteoglycan compared to the OA chondrocytes group (61). Another study indicated that inhibiting mitochondria in chondrocytes with agents such as rotenone or oligomycin promoted MSCs to transfer functional mitochondria to chondrocytes with mitochondrial dysfunction (63). Although chondrocytes adapt to low oxygen conditions, a number of their functions are still oxygen-dependent (64). Thus, culture conditions, such as hyperoxia and hyperglycemia, affected the mitochondrial transfer from MSCs to chondrocytes because chondrocyte homeostasis responds to the low oxygen levels and nutrient-deficient conditions in vivo (63).

Tendon injuries often occur in the severely hypoxic zone, characterized by mitochondrial dysfunction and increased oxidative stress (65). Mitochondrial dysfunction is characterized by an increase in ROS, decreased superoxide dismutase activity, and a reduced number of mitochondria, all of which contribute to tendon pathology (66). In a previous study on a murine model of supraspinatus tendinopathy, mitochondrial dysfunction led to the altered expression of genes related to morphology and abnormal mitochondrial quantity associated with the development of tendinitis (67). Another study indicated that H2O2 activated the apoptotic pathway by inducing oxidative stress and the depolarization of MMP in mouse models of Achilles tendinopathy (68). Tenomodulin (TNMD) is a marker specific to tendons and contributes to the durability and aging of tendon tissue at the tissue level (69). TNMD is associated with the structural and functional properties of collagen 1 (COL1) (69). A reduction in the levels of tenomodulin is a sign of pathogenesis. Mitochondrial transfer from BM-MSCs to H2O2-damaged tenocytes triggered anti-apoptotic mechanisms and restored mitochondrial function by recovering MMP and ATP levels (70). Functional mitochondria derived from MSCs enhanced the expression of TNMD and collagen 1 and reduced the expression of matrix metalloproteinase 1 in tenocytes treated with tumor necrosis factor-α (TNF-α) (71). Moreover, functional mitochondria from MSCs also reduced levels of pro-inflammatory cytokines, such as interleukin (IL)-1β and IL-6(70), demonstrating an anti-inflammatory effect.

Mitochondria contribute to several key functions for the survival of muscle cells, such as ATP production, substrate biosynthesis, metabolism and participation in the regulation of apoptosis (72). Skeletal muscle atrophy is characterized by a reduction in muscle mass and strength and is associated with mitochondrial dysfunction as mitochondria occupy a significant portion of muscle cell volume (72,73). The reduction in mitochondrial mass, MMP, ATP production and mitochondrial glucose metabolism activity are characteristic features of muscle atrophy (74). ROS overproduction related to mitochondrial dysfunction is the main mechanism associated with muscle atrophy (75-77). Moreover, mitochondrial dysfunction has also been implicated in the pathology of muscular dystrophy (78). The restoration of mitochondrial bioenergetics has been shown to ameliorate the progression of muscular dystrophy in mice (79). Dexamethasone is an agent that induces muscular atrophy through a process of pyroptosis, a form of programmed cell death involving inflammation as the initial process (80). A previous study demonstrated that mitochondrial transplantation derived from umbilical cord-derived MSCs (UC-MSCs) promoted myofiber hypertrophy and reduced intracellular lactate concentration in dexamethasone-induced atrophied muscles (81). This also contributed to enhancing the expression of muscle-specific markers, such as desmin and activated adenosine monophosphate-activated protein kinase, which aided the functional recovery of atrophied muscles (81).

These data indicate that the use of mitochondria isolated from MSCs may be beneficial in different types of pathology in the musculoskeletal system. The addition of MSC mitochondria aids in the restoration of various cell types, the construction of different functional parts, and the reduction of tissue destruction caused by immune responses to stress.

In conditions of the brain

Ischemic stroke is associated with a reduction in cerebral blood flow, leading to severe oxygen and glucose deprivation (OGD) in neurons (82-84). OGD has been shown to cause neuronal mitochondrial dysfunction, such as reduced mitochondrial complex I activity, MMP collapse, decreased ATP synthesis and increased oxidative stress (85). Transient focal cerebral ischemia is characterized by irreversible neuronal loss, leading to persistent neurological impairment in individuals (86). The in vitro model of brain ischemia is a state of OGD closely related to oxidative stress due to the overproduction of ROS (87). In a previous study, mitochondrial damage in astrocytes caused by OGD, which resulted in fragmented mitochondria, stimulated the transfer of functional mitochondria from multipotent MSCs (MMSCs) to astrocytes (88). The functional mitochondria from MMSCs not only restored bioenergetics, but also normalized the proliferation of neuron-like ρ0 PC12 pheochromocytoma cells, which carried damaged mitochondrial DNA and generated most of their energy through anaerobic glycolysis (88). Injured astrocytes received more functional mitochondria from MMSCs when MMSCs overexpressed Miro1(88). Furthermore, BM-MSCs also transferred functional mitochondria to VSC4.1 motor neurons injured by OGD (89). BM-MSCs exhibited an increased CD157 expression when co-cultured with VSC4.1 motor neurons injured by OGD, an effect that enhanced the mitochondrial transfer capacity of BM-MSCs (89). Mitochondria derived from BM-MSCs could alleviate apoptosis and inflammation in damaged VSC4.1 motor neurons and promoted the outgrowth of VSC4.1 motor neurons, but only affected their length without changing their number (89). The mitochondrial transfer from iPSC-MSCs increased intracellular ATP levels in PC12 cells following CoCl2-induced hypoxic damage (90). Furthermore, the mitochondrial transfer from MSCs improved mitochondrial respiration, ATP production, and the basal metabolic rate in neurons after H2O2-induced injury (91).

Spinal cord injury (SCI) is a severe type of central nervous system injury and is associated with oxidative stress caused by the excessive production of ROS (92). Mitochondrial dysfunction related to mitochondrial homeostasis leads to the inactivation of ATP-dependent ion pumps and is closely associated with the activation of cell death (93). Mitochondrial quality control and the mitochondria-related process of ferroptosis play a crucial in SCI (94). Ferroptosis is an iron-dependent cell death process that occurs due to the accumulation of damaged phospholipids on the cell membrane and is associated with SCI (95-97). In a previous study, UC-MSCs were shown to transfer mitochondria to HT22 neuronal cells under the stimulation of the ferroptosis inducer Rsl3 when co-cultured (98). UC-MSCs helped to restore mitochondrial mass, promoted mitochondrial fusion, restored MMP, and balanced intracellular ROS and ATP levels in ferroptosis neurons (98). Furthermore, mitochondrial transfer from UC-MSCs reduced ferroptosis by decreasing the total intracellular iron content and free Fe2+ levels in neurons undergoing ferroptosis (98). Inhibiting ferroptosis using dihydroorotate dehydrogenase in injured neurons has been shown to normalize mitochondrial morphology and MMP (96).

Parkinson's disease (PD) is a condition related to the degeneration of the nervous system of the brain, and damaged mitochondria are one of the causes of PD (99,100). Mitochondrial dysfunction has been identified as a major factor contributing to the death of dopaminergic (DA) neurons (101). The dysfunction of mitochondrial respiratory chain complex I is a major cause of PD pathogenesis (102). The impairment of mitophagy and oxidative stress are hallmarks of PD (103). Mitochondria derived from UC-MSCs reduce DA cell death caused by 1-methyl-4-phenylpyridinium, 6-hydroxydopamine and rotenone (104). Furthermore, mitochondria derived from UC-MSCs also reduce the mRNA expression of TNF-α and nitric oxide synthase, and decrease the expression of IL-6 and IL-8 induced by lipopolysaccharide (LPS) in murine microglia BV2 cells (104). The reduced expression of pro-inflammatory cytokines has been shown to alleviate oxidative stress, inflammation and normalize mitochondrial dysfunction in a mouse model of PD (105). In a previous study, astrocytes derived from iPSC-MSCs provided functional mitochondria to DA neurons damaged by rotenone, a compound that inhibits complex I (106). Notably, the neuroprotective effect of astrocytes was not due to paracrine activity, but primarily to the transfer of mitochondria to DA neurons (106). The transfer of mitochondria from iPSC-MSCs-derived astrocytes restored ATP levels impaired by rotenone in DA neurons (106).

In addition, cisplatin is a chemotherapeutic agent used to treat cancer; however, it is known to cause morphological changes and mitochondrial dysfunction in mouse neurons (107). Cisplatin has been shown to cause excessive ROS production and alter the mitochondrial content of cells (108). The donation of MSCs was shown to normalize the membrane potential of neural stem cells (NSCs) treated with cisplatin, which had shown a marked decrease in basal respiratory capacity, ATP levels, and MMP (109). Furthermore, excessive expression of Miro1 in MSCs increased mitochondrial transfer and promoted the survival of NSCs (109). In summary, these findings indicate that the transfer of functional mitochondria from MSCs can improve mitochondrial dysfunction and neuronal survival under various pathological conditions in brain tissue (106).

In conditions of the eyes

Mitochondrial damage and abnormalities in mitochondrial homeostasis are among the causes of ocular diseases (110,111). The risk of mitochondrial damage increases when the cornea is exposed to ultraviolet rays (112). Corneal endothelial cells (CECs) play a crucial role in maintaining the transparency of the cornea; therefore, a decline in their function will negatively impact vision (110). Metabolic activity and mitochondrial respiration, such as the tricarboxylic acid cycle and acetyl coenzyme A-related enzymes, are closely related to the survival and function of CECs (113). Therefore, the addition of functional mitochondria is expected to support the recovery of eye disorders where impaired mitochondria have been identified to play a role. In a model of human corneal endothelial cells (hCECs) damaged by rotenone, the inhibition of mitochondrial complex I and increased ROS production were shown to promote the uptake of MSC mitochondria through TNT, whereas healthy hCECs could not accelerate similar effects (111,114). Rotenone-induced ROS promote NF-κB, which enhances TNT formation via the upregulation of TNF-α-induced protein 2 (TNFαip2) (114). The underlying mechanism has been previously reported (115). The contribution of functional mitochondria from MSCs improves basal respiration, ATP production, and the expression of mitochondrial component proteins, such as complex I, mitochondrial matrix components and the mitochondrial membrane (111). The transfer of functional mitochondria from MSCs is the primary mechanism for protecting CECs from the effects of rotenone, although the paracrine effects of MSCs are also a contributing factor (114). However, the transferred mitochondria are either digested by lysosomes in the host cell or excreted after 8 days (111), suggesting that the therapeutic effect of exogenous mitochondria may be limited to the short term in CECs.

Degeneration of retinal ganglion cells (RGCs) is the cause of optic atrophy (116). The dysfunction of mitochondrial NADH dehydrogenase and complex I in the retina is related to an innate immune and inflammatory response and is associated with neuropathies, which has been shown to cause RGC degeneration in mice (116-118). It was previously demonstrated that neuronal cells in the RGC layer directly received mitochondria from the injected iPSC-MSCs to compensate for mitochondrial dysfunction in the retinas of mice deficient in NADH dehydrogenase Fe-S protein 4(119). On the other hand, Müller cells, the main glial cells of the retina, play a crucial role in maintaining neurovascular homeostasis in the retina (120). The abnormal activation of Müller cells, which may also lead to excessive ATP release, has been shown to contribute to the apoptosis of RGCs and is a pathogenic factor in glaucoma (121,122). In addition, inflammation should be accounted for in ocular and macular diseases. IL-17 has been shown to increase the production of glial fibrillary acidic proteins, vascular endothelial growth factor, and glutamate, leading to abnormal activation and dysfunction of Müller cells (123). IL-1β contributes to the recruitment of macrophages in the retina of in vivo models (124). Mitochondrial transfer from iPSC-MSCs reduced the abnormal activation of Müller cells in mouse retinal and downregulated pro-inflammatory cytokines such as IL-17 and IL-1β (119). These data demonstrated that mitochondrial transfer could alleviate ocular pathological conditions both by restoring ocular cell function and reducing inflammatory effects.

In models of metabolic disorders

Mitochondria play a crucial role in the functional activity of renal tubular epithelial cells (TECs) (125,126), and the disruption in mitochondrial activities has been shown to cause the aging of renal TECs, leading to renal complications and pathogenesis (127-129). Hyperglycemia causes mitochondrial dysfunction, characterized by a reduction in intracellular ATP and MMP, changes in mitochondrial morphology, and the excessive production of ROS in renal TECs, which contribute to diabetic nephropathy (130). A recent study revealed that high glucose levels increased oxidatively modified proteins in mitochondria and intracellular ROS in TECs (131). Streptozotocin (STZ) is an agent that causes alterations in MMP and mitochondrial NAD and NADH enzyme activity, leading to the inhibition of ATP synthesis, and thereby inducing oxidative stress and apoptosis in pancreatic β-cells (132). The use of STZ to model diabetes has been shown to lead to cellular metabolic changes and mitochondrial dysfunction in renal cells (133). In that context, mitochondrial transfer from BM-MSCs was previously been shown to inhibit apoptosis and ROS production in damaged proximal TECs induced by STZ (29). This was achieved by promoting the expression of mitochondrial superoxide dismutase 2 and Bcl-2, which are involved in regulating the process of apoptosis of proximal TECs (29). Furthermore, BM-MSCs-derived functional mitochondria improved the expression of megalin, a surface receptor protein in TECs that participates in the reabsorption of proteins from urine (134), as well as sodium-glucose cotransporter-2 (SGLT2), a member of the glucose transporter family in proximal TECs (29). Since high glucose levels have been shown to reduce the expression of megalin (which is also associated with glucose transport via SGLT) (135), the recovery of megalin and SGLT2 is a sign of improved TEC function.

Non-alcoholic steatohepatitis (NASH) has been shown to be a result of dysfunctional mitochondria, which includes alterations of enzymatic activity, changes in mitochondrial shape and quantity, and disrupted calcium regulation (136,137). In the NASH disease model, impaired mitochondria facilitate hepatic lipid accumulation by promoting ROS production and lipid peroxidation processes (138). High-fat diets over a long period of time have been shown to cause mitochondrial deformation and alter the levels of proteins involved in mitochondrial dynamics (139). It was previously shown that when fatty hepatocytes were co-cultured with BM-MSCs, mitochondrial transfer was observed through cell-to-cell contact, which promoted lipolytic activities via enhancing oxidative capacity and mitochondrial biogenesis (140,141). Mitochondria from BM-MSCs restored several mitochondrial abnormalities in the hepatocytes of mice fed a high-fat diet, including MMP, mtDNA copy number, OCR and ATP levels (140). Functional mitochondrial-derived BM-MSCs also restored impaired calcium activity in steatotic cells (140), an effect which is important for glucose production and lipogenesis (142).

Severe acute pancreatitis (SAP) is an inflammation of the pancreas that can lead to mortality. SAP is often linked to smoking, gallstone disease, alcohol consumption, obesity, diabetes, and hyperlipidemia (143). The excessive accumulation of intracellular ROS is associated with pyroptosis in pancreatic cells during acute inflammation (144). The dysregulation of the mPTP caused by excessive intracellular calcium levels results in changes in MMP, which is associated with the progression of SAP (145). The transfer of mitochondria via extracellular vesicles (EVs) from hypoxia (5% O2)-treated hUC-MSCs (hypo-EVs) was previously shown to restore ATP levels and OCR, and to maintain relatively stable MMP in sodium taurocholate-induced damaged rat pancreatic acinar cells (PACs) (146). EVs carrying mitochondria also promoted the normalization of glycolytic activity in PACs (146).

Allogeneic islet transplantation provides a promising opportunity for a limited group of patients with type I diabetes (147). Allogeneic transplantation of human islets requires short-term cultivation to ensure safety assessments (148). However, islet functions are influenced by various environmental conditions during this period, which may induce oxidative stress and inflammation (148). The insulin-secreting ability of pancreatic islet β-cells is associated with mitochondrial mass and respiratory capacity (149,150). It is also primarily regulated by mitochondrial ATP production in response to extracellular glucose levels (150). ADSCs have been shown to transfer mitochondria through extracellular vesicles to mouse islet cells exposed to hypoxia (1% oxygen), resulting in enhanced OCR and insulin secretion of islet cells when stimulated with 20 mM glucose (151). These data suggest that mitochondrial transfer can also be used as a supportive technique in optimizing islet pre-transplantation conditions.

In cells with mtDNA mutation or damage

Mitochondrial myopathy, encephalomyopathy with lactic acidosis, and stroke-like episodes (MELAS) is the acronym for a clinical subgroup of mtDNA diseases affecting several body systems (152). MELAS is often a result of point mutations in the mtDNA and is thus inheritable from the affected mother (152). The transition from adenine to guanine at nucleotide 3243 in the mtDNA of the MT-LT1 gene encoding tRNAleu (UUR), is among the most frequently observed pathogenic mutations in individuals with MELAS (153). This alteration leads to the impaired translation and synthesis of ETC protein subunits, resulting in reduced mitochondrial energy production (154). Patients with MELAS have fibroblasts (MELAS fibroblasts) with a high mutation burden, featuring mitochondrial dysfunction and increased ROS production (155). It was previously shown that Wharton's jelly-derived MSCs (WJMSCs) transferred mitochondria via TNTs into MELAS fibroblasts pre-treated with rotenone, resulting in an increased protein expression of the respiratory complexes, which suppressed ROS production and enhanced MMP (155). Furthermore, this also enhanced the proliferation of fibroblasts and reduced the expression of cleaved caspase-3 and TUNEL, thereby suppressing apoptosis (155). The results of another study suggested that highly purified MSCs have a greater ability to transfer mitochondria into MELAS neurons compared to traditional MSCs (156). Furthermore, co-culturing with RECs or MSCs significantly enhanced the viability of MELAS neurons (156).

Myoclonus epilepsy with ragged red fiber syndrome (MERRF) is a result of a transition from adenine to guanine at nucleotide 8344 (mt8344A>G) within the mitochondrial tRNALys coding gene (157). This mutation has been shown to increase ROS production and oxidative stress, along with compromised mitochondrial bioenergetics (157). MERRF cybrids, which were created by fusing ρ0 cells lacking mtDNA with human platelets in a medium devoid of pyruvate and uridine, were unable to survive in a galactose-added and glucose-free medium. This suggests that the mt8344A>G mutation leads to mitochondrial dysfunction (158). The MERRF mutation impairs the translation of 13 proteins encoded by mtDNA and produces abnormal translation products (159). The transfer of mitochondria from WJMSCs into MERRF cybrids decreases intracellular ROS, re-establishes MMP, and enables the growth of MERRF cells in glucose-free environments (158).

In clinical applications, hematopoietic stem cells (HSCs) were utilized for mitochondrial transplantation into the HSCs of patients with large-scale mtDNA deletion, such as Pearson or Kearns-Sayre syndromes (160,161). Recipient HSCs were then infused back into the patients, improving their quality of life (160,161). This evidence suggests a novel therapeutic application of mitochondrial transplantation for currently incurable diseases. While MSCs have not yet been used in such clinical trials, they can be collected from multiple sources and easily expanded. Their mitochondria vary in both morphology and functional capacity, and can be selected to suit HSC biology, making them a potential source for mitochondrial transplantation into HSCs for similar cases. Although replacing or supplementing defective mitochondria in all body systems appears challenging and warrants further research, mitochondrial transplantation holds therapeutic potential in mitochondrial pathogenesis.

In enhancing the function of cell engraftment therapy

Stem cell therapies have often been ineffective due to low cell survival and engraftment (162,163). The excessive production of ROS in damaged tissues usually decreases the viability of transplanted MSCs (164). Additionally, MSC functional characteristics may be compromised with prolonged isolation and in vitro cultivation, or due to the age of the donor or disease conditions (165,166). In this context, it has been shown that healthy hMSCs could donate mitochondria to those pre-treated with H2O2 through TNT formation (167). The treatment of hMSCs with mesenchymal stem cell adjuvant has been shown to enhance their mitochondrial transfer into other H2O2-induced damaged hMSCs, resulting in a significant reduction in intracellular ROS levels, maintenance of MMP, and an increase in TNT length and formation (167). Mitochondrial Drp 1 phosphorylation (Drp1 S616) is involved in mitochondrial fission and fragmentation (168). It was previously demonstrated that functional mitochondria derived from healthy hMSCs reduced mitochondrial fragmentation and Drp1 S616 phosphorylation in damaged hMSCs (167). The mitochondria derived from ADSCs supplied ATP to fulfill metabolic needs, thereby enhancing the overall metabolic activity of the recipient ADSCs through glycolysis (169). Moreover, ADSCs that received donated mitochondria showed enhanced proliferation and mobility in serum starvation conditions, demonstrating enhanced stress tolerance, anti-aging capabilities and multidirectional differentiation potential in ADSCs treated with doxorubicin (169). It should be noted that mitochondria can be artificially introduced into BM-MSCs in vitro, and within certain limits, the addition of extracellular mitochondria could enhance the capacity of the recipient cells to take up more mitochondria (170). Receiving mitochondria also increases BM-MSC proliferation and migration, promoting cells to enter the S and G2/M phases, and resisting the replicative senescence of BM-MSCs (170). In addition, mitochondrial transfer also enhances the osteogenic differentiation potential and mitochondrial functions, including OCR and ATP production in BM-MSCs (170).

A recent study demonstrated that the transfer of mitochondria from MSCs to endothelial cells, either by direct cell-to-cell contact or artificial transplantation, significantly enhanced the engraftment ability of the cells in the in vivo environment (171). The stimulation of these effects was shown to be related to mitophagy; however, the precise mechanism remained unknown. These data suggested that boosting mitochondrial activity by mitochondrial transfer and donation can also support cell survival in an in vivo environment, proposing a potential application for mitochondrial therapy in cell transplantation technology.

In conditions of the immune system

MSCs possess the capacity to modulate immune responses, particularly those of T-cells, indicating their therapeutic potential in treating autoimmune disorders, preventing graft rejection and managing graft vs. host disease (GVHD) (172). One mechanism of immunoregulation by MSCs may be attributed to their ability to transfer mitochondria to immune cells, regulating cell function and consequent immune outcomes. It was previously demonstrated that CD3+ T-cells receiving mitochondria from UC-MSCs increased the expression of CD25 (a factor for T-cell activation) and FoxP3 [a marker of T-regulatory cell (Treg) differentiation], compared to CD3+ T-cells not receiving mitochondria from UC-MSCs (173). Since Tregs participate in maintaining tolerance and preventing autoimmunity, they are involved in various pathological conditions, including GVHD (174). Treg cell differentiation induced by mitochondria transferred from UC-MSCs enhanced the ability to inhibit the proliferation of peripheral blood mononuclear cells (173). As previously demonstrated, mitochondrial transfer also markedly improved the survival rate of mice with GVHD, while decreasing tissue inflammation (173). The key strategy employed by Tregs to preserve immune homeostasis is the inhibition of conventional T-cell (Tconv) proliferation (175). A previous study demonstrated that ADSCs transferred mitochondria to Tregs during co-culture, 85% of which remained functional, allowing Tregs to efficiently inhibit the proliferation of Tconvs (176). The insufficient presence of HLA class II antigens on ASCs restricted the uptake of mitochondria by >80% (176). Thus, mitochondrial transfer from MSCs supports T-cells in regulating uncontrolled immunity.

In addition, it should be noted that both CD4+ and CD8+ T-cells participate in immune responses in GVHD, autoimmune disorders and tissue rejection (177). It was previously demonstrated that mitochondria donated by BM-MSCs suppressed T-helper 17 (Th17) cells, a class of CD4+ T-cells involved in the progression of autoimmune conditions, while promoting the formation of Treg cells (178). In a co-culture system, BM-MSCs transferred mitochondria to pathogenic Th17 cells, leading to increased OCR and a decrease in IL-17 expression in Th17 cells (178). The inhibition of IL-17 production was related to the interconversion into Tregs (178). In another study, mitochondria transferred from MSCs also helped to inhibit the response of CD4+ T-cells and reduced the production of interferon-γ (IFNγ), a cytokine that activates M1 macrophages, and promoted CD8+ T-cell function (172). The differentiation of IFNγ-producing CD4+ Th1 cells relies on the key transcription factor, T-bet, the expression of which is increased upon encountering antigens to promote gene expression, particularly IFNγ, thus determining the differentiation of the effector subset Th1(179). In a previous study, co-culturing with MSCs that were primed with TNFα and IFNγ for 48 h inhibited T-bet expression following CD4+ T-cell activation and suppressed Th1 differentiation (172).

Diabetes mellitus is chiefly characterized by increased macrophage infiltration in the kidneys. Implementing strategies to prevent the pro-inflammatory (M1) phenotype or to shift them to an anti-inflammatory (M2) state has been proposed to help reduce kidney injury in diabetic mice (180). Metabolic and physiological changes in mitochondria are essential for regulating macrophage polarization, proliferation and survival (181). While M1 macrophages utilize glycolysis for energy, M2 macrophages depend on mitochondrial oxidative phosphorylation (182,183). Mitochondrial dysfunction in macrophages derived from diabetic mice has been shown to lead to M1 polarization and resulted in subsequent inflammation (184). In that context, in another study, mitochondria transferred from MSCs to RAW264.7 macrophages pre-treated with high glucose helped the cells to reduce ROS production and restore MMP and ATP levels by regulating gene expression of the glycolytic and citric acid cycles (185). RAW264.7 macrophages induced by high glucose that took up mitochondria exhibited lower levels of cytokine secretion, such as IL-1β and TNF-α, two cytokines which are critical for mitigating inflammation (185).

Alveolar macrophages (AMs), found in the lung lumen adjacent to the epithelium, function to phagocytose debris and help resolve inflammation (186). Dysfunctional AMs with an irregular mitochondrial function frequently contribute to severe inflammation in the lungs (186). It has been shown that MSCs transferring mitochondria to macrophages can enhance their phagocytosis, which may uncover a mechanism behind the antimicrobial effects of BM-MSCs (187). Furthermore, mitochondrial transfer from MSCs can boost the anti-inflammatory and antibacterial functions of macrophages by promoting their differentiation into the M2 phenotype (188). As previously demonstrated, when MSCs and primary human monocyte-derived macrophages (MDMs) were co-cultured for 4 h, the mitochondrial transfer from BM-MSCs through TNTs enhanced the ability of macrophages to kill up to 80% of extracellular Escherichia coli bacteria (187). Notably, macrophages co-cultured with MSCs exhibited a substantial and consistent enhancement of both OCR and ATP production (187). This was consistent with a report that macrophages receiving mitochondria from exosomes secreted by MSCs in the co-culture settings significantly increased OCR and reduced proton leak (189). This functioned as a survival strategy in response to oxidative stress, where donated mitochondria were re-utilized through a fusion process to enhance the bioenergetics of macrophages (189). Exosomes derived from ADSCs could transfer mitochondrial components to murine alveolar macrophages, which then fused with the macrophage mitochondria (190). Murine alveolar macrophages in acute lung injury exhibited an increased number of mitochondria, improved mitochondrial morphology, and increased membrane potential after receiving mitochondria from ADSCs (190). Moreover, mitochondria played a critical role in the metabolic programming of immune cells and in reshaping cellular phenotypes and function (191). The transfer of mitochondria via ADSC-derived exosomes caused LPS-stimulated macrophages to transition from a pro-inflammatory M1 phenotype to an anti-inflammatory M2 phenotype (190). Furthermore, mitochondria have been shown to be transferred from MSCs to macrophages via EVs, which boosted oxidative phosphorylation (OXPHOS), leading to enhanced phagocytosis of macrophages (192). These data demonstrate that MSCs employ multiple mechanisms to transfer mitochondria to immune cells, which often have high mobility in circulation. Thus, questions regarding the driving factors for mitochondrial transfer from MSCs to immune cells warrant future research, potentially opening a potential opportunity for therapeutic application.

In conditions of the reproductive system

The reproductive system consists of various organs and structures that are distinct in males and females. Different cell compartments in the reproductive system are responsible for specific functions that support the formation and production of offspring. The two key cell types directly responsible for fertilization are sperm and eggs. While mitochondria are crucial for sperm maturation, survival, and mobility, oocyte mitochondria are critical for fertilization and embryonic development, which are also majorly inherited by the fetus (193). Thus, both mtDNA content and mitochondrial function are essential for successful fertilization, pregnancy and live births. Mitochondria in oocytes produce energy mainly via OXPHOS, as glycolysis is only unblocked at the early stage of blastocyst formation. ATP production is associated with egg quality, as aged and poor-quality eggs have significantly lower mtDNA content and produce much less ATP compared to normal and healthy oocytes (194). This could potentially result in low or no fertilization (195,196). Aging is one of the major contributing factors to reduced female fertility, as it directly affects mtDNA replication and mutation, and changes mitochondrial metabolism (197).

Mitochondria can be isolated from healthy donors and directly injected into the eggs or embryos of unhealthy individuals who may carry mutations interfering with mitochondrial function. The technology is effective in supplementing oocytes with healthy mitochondria to improve oocyte quality and fertilization rate, and to increase chances of live births and offspring with normal phenotypes, providing hope to infertile women and those with inherited diseases (198). Multiple studies have been conducted on both animal and human models, aiming to examine the applicability of mitochondria isolated from various cell types to identify the most suitable source of mitochondria specifically for oocyte improvement; a number of these sources were from MSCs of different origins. Even though the current technology focuses on the autologous grafting of mitochondria due to concerns about mtDNA and nuclear DNA mismatch (199), the therapy has yielded positive results in assisting cases of aged and low-quality oocytes. For example, in animal models, for instance, the mitochondria isolated from MSCs derived from UC-MSCs were directly injected into metaphase I oocytes of aged mice, which consequently resulted in improved mitochondrial function and development of early embryos (200). On the other hand, mitochondria derived from ADSCs were shown to be effective in improving oocyte competence, fertilization rate, and blastocyst development in both aged-MII and cryopreserved oocytes (201,202). MSCs (EnMSCs) were also a source of cells that have been tested in mice. The increase in oocyte quality and maturation associated with an enhanced birth rate and embryonic development suggested that mitochondrial transfer from EnMSCs was effective in rescuing infertility due to the poor quality of germinal vesicle oocytes (203). Additionally, mitochondria isolated from iPSCs were also shown to be a good source for mitochondrial transfer due to their compatibility with the mitochondrial morphology in oocytes and embryos (204). Furthermore, advancements have been made to bring the technology of mitochondrial therapy to humans. Particularly, mitochondria from urine-derived MSCs and oogonial stem cells were isolated for use in aged human oocytes. The results revealed an improvement in pregnancy rates, embryonic development and live birth rates (205-209). These data highlight the promising potential of mitochondrial transplantation technology in supporting the reproductive system, particularly in addressing female infertility. Future research is also anticipated to advance mitochondrial therapy in the male reproductive system.

3. Mechanisms of mitochondrial donation from MSCs

Previous in vitro studies have demonstrated the role of MSCs in rescuing damaged cells by transferring their healthy mitochondria (53,210). In cells exposed to mitochondrial toxicity stimulators, mitophagy is activated to eliminate dysfunctional mitochondria. These dysfunctional mitochondria, which can be composed of mtDNA and other mitochondrial components, help maintain mitochondrial homeostasis (211). These mitochondrial components are released into the microenvironment and function as stress signals, which can prompt nearby MSCs to provide support (47,55,212) (Fig. 3). Following internalization by MSCs, damaged mitochondria or their parts undergo autophagy, thereby triggering MSCs for mitochondrial transfer to rescue injured cells (55,171). The activation of autophagy enhances the formation of TNTs in MSCs, which is essential for the mitochondrial transfer process (212). Autophagy also activates the expression of HO-1, a cytoprotective enzyme, and increases mitochondrial biogenesis in MSCs through the regulation of peroxisome proliferator-activated receptor gamma coactivator-1-α and mitochondrial transcription factor A expression (55). These data suggest that the degradation of external mitochondria within MSCs by autophagy is a key trigger of mitochondrial transfer. Doxorubicin-induced injured cardiomyocytes also exhibit an increased expression of TNF-α, which stimulates the formation of TNTs in MSCs (47). Similar to doxorubicin, cells treated with hydrogen peroxide (H2O2) were also shown to be rescued by MSCs (55). In fact, the overproduction of ROS in mitochondria has been demonstrated as a severe signal of cellular stress (88,213,214). OGD/reoxygenation causes ROS overproduction in injured cells, which stimulates the uptake of healthy mitochondria from MSCs (213,215). Additionally, high levels of abnormal ROS from oxidative stress can cause mitochondrial fragmentation, leading to mitochondrial transfer from MSCs (88). A previous in vitro study demonstrated that the inhibition of OXPHOS upregulated related adhesion promoters, a mechanism central to mitochondrial transfer from MSCs to acute myeloid leukemia (AML) cells (216). Specifically, under the pressure of chemotherapeutic OXPHOS inhibition, ROS levels increase, and the conversion of LC3-I to LC3-II occurs as a sign of autophagy. This process promotes MSCs to transfer mitochondria to OCI-AML3 cells (171,216). However, in the event that N-acetyl-L-cysteine (NAC) is used to decrease ROS production, the mitochondrial uptake in damaged cells is impaired (214,215). These data indicate that various stress signals are crucial for the mitochondrial transfer rescue effect mediated by MSCs.

Mechanisms of MSC mitochondrial
transfer rescue effects. Injured cells release damaged mitochondria
or their components into the surrounding microenvironment as a
distress signal. MSCs respond to these stress signals by activating
the mitochondrial transfer mechanism via the upregulation of PGC1-α
and the expression of HO-1. The expression of Miro1, TNFαip2 and
Cx43 is also upregulated in MSCs to facilitate the transfer of
healthy mitochondria to rescue stressed cells from damage. MSCs,
mesenchymal stem cells; ROS, reactive oxygen species; PGC1-α,
proliferator-activated receptor gamma coactivator-1-α; HO-1, heme
oxygenase 1; Miro1, mitochondrial Rho GTPase 1; TNFαip2,
TNF-α-induced protein 2; Cx43, connexin43.

Figure 3

Mechanisms of MSC mitochondrial transfer rescue effects. Injured cells release damaged mitochondria or their components into the surrounding microenvironment as a distress signal. MSCs respond to these stress signals by activating the mitochondrial transfer mechanism via the upregulation of PGC1-α and the expression of HO-1. The expression of Miro1, TNFαip2 and Cx43 is also upregulated in MSCs to facilitate the transfer of healthy mitochondria to rescue stressed cells from damage. MSCs, mesenchymal stem cells; ROS, reactive oxygen species; PGC1-α, proliferator-activated receptor gamma coactivator-1-α; HO-1, heme oxygenase 1; Miro1, mitochondrial Rho GTPase 1; TNFαip2, TNF-α-induced protein 2; Cx43, connexin43.

The molecular mechanism of TNT formation involves Miro1, an outer mitochondrial membrane protein essential for mitochondrial transport and donation (217,218). Following the increased levels of Miro1 induced by oxidative stress, MSCs enhance their capacity for mitochondrial transfer through TNTs (47,55,88,219). Miro1 deletion has been proven to inhibit MSC mitochondrial transfer and impair the microtubule movement of donated mitochondria (217,218). It should be noted that the expression of Miro1 and TNFαip2 are interrelated in mitochondrial transfer (219). TNFαip2 is a marker of TNT formation that regulates intercellular interactions (114). TNF-α-induced oxidative stress causes significantly high TNFαip2 expression (220). When TNFαip2 is deleted in MSCs, the overexpression of Miro1 also decreases mitochondrial transfer, and the control of ROS levels is impaired both in vitro and in vivo (217). NF-κB and TNFαip2 overexpression increase ROS levels, which subsequently leads to TNTs formation, and NAC reverses this process by reducing ROS (47,114,221). Furthermore, there is an association between the phosphorylation of NF-κB and TNT formation inhibition (114,221,222). The expression of TNFαip2 is directly related to mitochondrial transfer capacity between cells through TNTs (219,220). The N- and C-terminal region of the TNFaip2 protein have different contributions to the remodeling of the MSC's plasma membrane to form TNTs (223).

Additionally, after receiving stimulation from injured cell signals, MSCs also promote cell-to-cell interactions and mitochondrial transfer capacity. Connexin 43 (Cx43) has been shown to be related to the transfer of mitochondria between cells (63,224). ROS overproduction caused by LPS in injured cells promotes the mitochondrial transfer from MSCs through Cx43 regulation (214). Cx43 knockdown in MSCs interrupts the mitochondrial transfer from MSCs to injured chondrocytes (63,225). In the event that MSCs upregulate Cx43 expression by iron oxide nanoparticles, the mitochondrial transfer ability of MSCs will be enhanced (226). On the other hand, various mechanisms can be utilized by other cells, particularly cancer cells, to steal mitochondria from their neighboring cells in the microenvironment (227-231); however, these mechanisms have not been implicated in the MSC-mediated mitochondrial rescue pathway. Questions remain as to whether these mechanisms exist between MSCs and other cells.

4. Fates of donated mitochondria in host cells

There are two separate fates for donated exogenous mitochondria once internalized inside recipient cells: Integration into the host mitochondrial network or degradation through mitophagy and autophagy processes. Within the host cells, exogenous mitochondria can fuse with the host mitochondrial network, and this fusion can occur via different mechanisms across various types of cells (232-234). This process is associated with the expression of mitofusin (MNF)1, MNF2 and optic atrophy 1 (OPA1) (235). Under oxidative stress conditions, cells downregulate the expression of these mitochondrial fusion proteins, particularly MFN1 and OPA1(232). In the majority of cases, this downregulation is affected by the presence of exogenous mitochondria. A previous study demonstrated that following exogenous mitochondrial transplantation, the recipient cells contained nearly 12% mtDNA from donor cells after 24 h (171). Notably, this exogenous mtDNA cannot be detected on day 7. Lysosomes of host cells degrade parts of exogenous mitochondria (232,233,235). On the other hand, it has been shown that at ~6-12 h following transplantation, exogenous mitochondria enter the autophagy process in host cells (171). There is evidence to indicate that donated mitochondria from MSCs inside endothelial cells undergo mitophagy mediated by the PINK1-Parkin pathway, which facilitates biogenesis in the recipient cells and enhances their engraftment in vivo (171). Future research is warranted to elucidate the precise mechanisms through which host cells take up and regulate the use of exogenous mitochondria within their cytoplasm.

5. Conclusion and future perspectives

Mitochondrial transfer and transplantation appear to be an innovative and promising tool for targeting mitochondria-related pathological conditions (25,236-238). However, identifying a qualified and ready-to-use source of mitochondria has been a challenge for clinical translation. Difficulties in obtaining mitochondrial sources can limit the reproducibility of the approach, increase the need for invasive procedures in patients, and hinder the quality control of the transplanted mitochondria. In clinical trials, ready-to-use mitochondria may help to improve the feasibility and efficacy of their application, ensuring timely treatment (239). A recent study demonstrated that storable human mitochondria, as a ready source of exogenous mitochondria, are effective in a mouse model of Leigh syndrome (240). Thus, a stable and quality source of mitochondria is key to the success of mitochondrial transplantation. In this context, MSCs have emerged as potential donors of mitochondria in mitochondrial therapy, and preclinical data indicate that their use is effective across a range of cell types, diseases and conditions. A summary of recipient cell types receiving mitochondrial transfer from MSCs is presented in Table I. However, although the exact mechanisms underlying tissue regeneration through mitochondrial transfer are not yet fully understood, it is proposed to involve several rescuing effects (Fig. 4).

Regenerative mechanism in
mitochondrial therapy using MSCs. Cells or tissues receiving
mitochondria from MSCs exhibit improvement and restoration in
several aspects. The supplementation with healthy mitochondria from
MSCs can directly replace defective mtDNA and improve metabolism
and fitness, supporting ATP generation and ROS reduction. As
mitochondrial biogenesis improves in recipient cells, cell survival
is significantly enhanced, particularly in vivo, thereby
restoring cell and tissue function. Cells that received donated
mitochondria from MSCs showed enhanced differentiation and
proliferation, simultaneously reducing apoptosis. Mitochondria
supplementation also helps to regulate the immune response and
cytokine secretion, resulting in positive outcomes. The degradation
of internalized mitochondria further activates mitophagy and
autophagy in recipient cells, thereby indirectly regulating
cellular homeostasis and improving cell function. MSCs, mesenchymal
stem cells; mtDNA, mitochondrial DNA; ROS, reactive oxygen
species.

Figure 4

Regenerative mechanism in mitochondrial therapy using MSCs. Cells or tissues receiving mitochondria from MSCs exhibit improvement and restoration in several aspects. The supplementation with healthy mitochondria from MSCs can directly replace defective mtDNA and improve metabolism and fitness, supporting ATP generation and ROS reduction. As mitochondrial biogenesis improves in recipient cells, cell survival is significantly enhanced, particularly in vivo, thereby restoring cell and tissue function. Cells that received donated mitochondria from MSCs showed enhanced differentiation and proliferation, simultaneously reducing apoptosis. Mitochondria supplementation also helps to regulate the immune response and cytokine secretion, resulting in positive outcomes. The degradation of internalized mitochondria further activates mitophagy and autophagy in recipient cells, thereby indirectly regulating cellular homeostasis and improving cell function. MSCs, mesenchymal stem cells; mtDNA, mitochondrial DNA; ROS, reactive oxygen species.

Despite this promising application, it should be noted that mitochondrial transfer and transplantation may also pose significant health risks in the case of misuse. Mitochondrial therapy, whether utilizing mitochondria from MSCs or other cell types (both autologous or allogenic), may encounter various challenges, including the acceleration of the immune response and the promotion of tumor metastasis (241-243). For instance, allogenic mtDNA has been shown to accelerate the innate immune response in mice (244), and even in cases of autologous transplantation, an mtDNA mutation in the cells of the donor could elicit an immune response that is dependent on host major histocompatibility complex (245). Although allogenic mitochondrial transplantation has been shown in several studies, it is important to understand the mechanism and effects of the treatment, and screening for mtDNA is crucial to ensure safety in clinical applications. In addition, several ethical questions have also been raised, primarily in the field of assisted reproductive technology, concerning the retention of exogenous mitochondria and the potential for mtDNA/nuclear DNA mismatch within host cells (243,246,247). Given the relevance of these issues in other clinical applications, further research is required to clarify the mechanisms of exogenous mitochondrial interaction with the host cells and determine the effective dosages for different cases. Furthermore, optimizing the procedures and routes of application for specific pathological conditions is crucial to enhance the accessibility of this technique in diverse clinical settings.

Although challenges persist, progress has been achieved to improve the safety and broad application of mitochondrial transfer and transplantation. Firstly, research has focused on optimizing the cellular uptake of exogenous mitochondria through various techniques of artificial transplantation (Fig. 1B). These technologies focus on improving mitochondrial permeabilization across the cell membranes by utilizing both mechanical forces and penetrating peptides. While none of these methods are currently suitable for clinical application, ongoing efforts are focused on refining existing techniques and developing new materials and technologies. Current approaches, however, have successfully achieved mitochondrial transplantation into isolated cells. This success holds promise for improving cell function in therapeutic areas, such as stem cell and immune cell therapies. Furthermore, research is being conducted to understand the mechanism of intercellular mitochondrial transfer, aiming to enhance the rescue of damaged cells in in vivo settings (248). For instance, stimulating mitochondrial transfer from MSCs, such as enhancing the formation of TNT, increasing the secretion of EVs, and promoting cell fusion, could be advantageous for multiple applications. Most importantly, future research is required to verify the effectiveness of different stem cell types and their culture conditions, while clarifying and defining appropriate mitochondrial quality measures. With the current progress, it is expected that MSCs will provide a cell source for mitochondrial transplantation, offering a new approach to regenerative medicine and adding an essential element to stem cell therapy.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

CMT was involved in the conceptualization of the study, in the preparation of the figures, and in the writing of the original draft of the manuscript. VTTN was involved in the conceptualization of the study, in the preparation of the figures visualization, in the writing of the original draft of the manuscript, and in the writing, reviewing and editing of the manuscript. Both authors have read and confirmed the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Use of artificial intelligence tools

During the preparation of this work, AI tools were used to improve the readability and language of the manuscript or to generate images, and subsequently, the authors revised and edited the content produced by the AI tools as necessary, taking full responsibility for the ultimate content of the present manuscript.

References

1 

Amorim JA, Coppotelli G, Rolo AP, Palmeira CM, Ross JM and Sinclair DA: Mitochondrial and metabolic dysfunction in ageing and age-related diseases. Nat Rev Endocrinol. 18:243–258. 2022.PubMed/NCBI View Article : Google Scholar

2 

Hyatt H, Deminice R, Yoshihara T and Powers SK: Mitochondrial dysfunction induces muscle atrophy during prolonged inactivity: A review of the causes and effects. Arch Biochem Biophys. 662:49–60. 2019.PubMed/NCBI View Article : Google Scholar

3 

Dagda RK: Role of mitochondrial dysfunction in degenerative brain diseases, an overview. Brain Sci. 8(178)2018.PubMed/NCBI View Article : Google Scholar

4 

Kaarniranta K, Uusitalo H, Blasiak J, Felszeghy S, Kannan R, Kauppinen A, Salminen A, Sinha D and Ferrington D: Mechanisms of mitochondrial dysfunction and their impact on age-related macular degeneration. Prog Retin Eye Res. 79(100858)2020.PubMed/NCBI View Article : Google Scholar

5 

Zilio E, Piano V and Wirth B: Mitochondrial dysfunction in spinal muscular atrophy. Int J Mol Sci. 23(10878)2022.PubMed/NCBI View Article : Google Scholar

6 

Weiss SL, Zhang D, Bush J, Graham K, Starr J, Murray J, Tuluc F, Henrickson S, Deutschman CS, Becker L, et al: Mitochondrial dysfunction is associated with an immune paralysis phenotype in pediatric sepsis. Shock. 54:285–293. 2020.PubMed/NCBI View Article : Google Scholar

7 

McBride MA, Owen AM, Stothers CL, Hernandez A, Luan L, Burelbach KR, Patil TK, Bohannon JK, Sherwood ER and Patil NK: The metabolic basis of immune dysfunction following sepsis and trauma. Front Immunol. 11(1043)2020.PubMed/NCBI View Article : Google Scholar

8 

Prasun P: Mitochondrial dysfunction in metabolic syndrome. Biochim Biophys Acta Mol Basis Dis. 1866(165838)2020.PubMed/NCBI View Article : Google Scholar

9 

Bhatti JS, Bhatti GK and Reddy PH: Mitochondrial dysfunction and oxidative stress in metabolic disorders-a step towards mitochondria based therapeutic strategies. Biochim Biophys Acta Mol Basis Dis. 1863:1066–1077. 2017.PubMed/NCBI View Article : Google Scholar

10 

Montgomery MK: Mitochondrial dysfunction and diabetes: Is mitochondrial transfer a friend or foe? Biology (Basel). 8(33)2019.PubMed/NCBI View Article : Google Scholar

11 

Feichtinger RG, Sperl W, Bauer JW and Kofler B: Mitochondrial dysfunction: A neglected component of skin diseases. Exp Dermatol. 23:607–614. 2014.PubMed/NCBI View Article : Google Scholar

12 

Mizumura K, Cloonan SM, Nakahira K, Bhashyam AR, Cervo M, Kitada T, Glass K, Owen CA, Mahmood A, Washko GR, et al: Mitophagy-dependent necroptosis contributes to the pathogenesis of COPD. J Clin Invest. 124:3987–4003. 2014.PubMed/NCBI View Article : Google Scholar

13 

Li X, Zhang W, Cao Q, Wang Z, Zhao M, Xu L and Zhuang Q: Mitochondrial dysfunction in fibrotic diseases. Cell Death Discov. 6(80)2020.PubMed/NCBI View Article : Google Scholar

14 

Ryter SW, Rosas IO, Owen CA, Martinez FJ, Choi ME, Lee CG, Elias JA and Choi AMK: Mitochondrial dysfunction as a pathogenic mediator of chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis. Ann Am Thorac Soc. 15 (Suppl 4):S266–S272. 2018.PubMed/NCBI View Article : Google Scholar

15 

Kandasamy J, Olave N, Ballinger SW and Ambalavanan N: Vascular endothelial mitochondrial function predicts death or pulmonary outcomes in preterm infants. Am J Respir Crit Care Med. 196:1040–1049. 2017.PubMed/NCBI View Article : Google Scholar

16 

Rizzuto R, De Stefani D, Raffaello A and Mammucari C: Mitochondria as sensors and regulators of calcium signalling. Nat Rev Mol Cell Biol. 13:566–578. 2012.PubMed/NCBI View Article : Google Scholar

17 

Matuz-Mares D, González-Andrade M, Araiza-Villanueva MG, Vilchis-Landeros MM and Vázquez-Meza H: Mitochondrial calcium: Effects of its imbalance in disease. Antioxidants (Basel). 11(801)2022.PubMed/NCBI View Article : Google Scholar

18 

Belosludtsev KN, Talanov EY, Starinets VS, Agafonov AV, Dubinin MV and Belosludtseva NV: Transport of Ca2+ and Ca2+-dependent permeability transition in rat liver mitochondria under the streptozotocin-induced type I diabetes. Cells. 8(1014)2019.PubMed/NCBI View Article : Google Scholar

19 

Sundaramoorthy P, Sim JJ, Jang YS, Mishra SK, Jeong KY, Mander P, Chul OB, Shim WS, Oh SH, Nam KY and Kim HM: Modulation of intracellular calcium levels by calcium lactate affects colon cancer cell motility through calcium-dependent calpain. PLoS One. 10(e0116984)2015.PubMed/NCBI View Article : Google Scholar

20 

Spees JL, Olson SD, Whitney MJ and Prockop DJ: Mitochondrial transfer between cells can rescue aerobic respiration. Proc Natl Acad Sci USA. 103:1283–1288. 2006.PubMed/NCBI View Article : Google Scholar

21 

Rodriguez AM, Nakhle J, Griessinger E and Vignais ML: Intercellular mitochondria trafficking highlighting the dual role of mesenchymal stem cells as both sensors and rescuers of tissue injury. Cell Cycle. 17:712–721. 2018.PubMed/NCBI View Article : Google Scholar

22 

Plotnikov EY, Babenko VA, Silachev DN, Zorova LD, Khryapenkova TG, Savchenko ES, Pevzner IB and Zorov DB: Intercellular transfer of mitochondria. Biochemistry (Mosc). 80:542–548. 2015.PubMed/NCBI View Article : Google Scholar

23 

Hayashida K, Takegawa R, Shoaib M, Aoki T, Choudhary RC, Kuschner CE, Nishikimi M, Miyara SJ, Rolston DM, Guevara S, et al: Mitochondrial transplantation therapy for ischemia reperfusion injury: A systematic review of animal and human studies. J Transl Med. 19(214)2021.PubMed/NCBI View Article : Google Scholar

24 

Yang C, Yokomori R, Chua LH, Tan SH, Tan DQ, Miharada K, Sanda T and Suda T: Mitochondria transfer mediates stress erythropoiesis by altering the bioenergetic profiles of early erythroblasts through CD47. J Exp Med. 219(e20220685)2022.PubMed/NCBI View Article : Google Scholar

25 

Liu D, Gao Y, Liu J, Huang Y, Yin J, Feng Y, Shi L, Meloni BP, Zhang C, Zheng M and Gao J: Intercellular mitochondrial transfer as a means of tissue revitalization. Signal Transduct Target Ther. 6(65)2021.PubMed/NCBI View Article : Google Scholar

26 

Emani SM, Piekarski BL, Harrild D, Del Nido PJ and McCully JD: Autologous mitochondrial transplantation for dysfunction after ischemia-reperfusion injury. J Thorac Cardiovasc Surg. 154:286–289. 2017.PubMed/NCBI View Article : Google Scholar

27 

Guariento A, Piekarski BL, Doulamis IP, Blitzer D, Ferraro AM, Harrild DM, Zurakowski D, Del Nido PJ, McCully JD and Emani SM: Autologous mitochondrial transplantation for cardiogenic shock in pediatric patients following ischemia-reperfusion injury. J Thorac Cardiovasc Surg. 162:992–1001. 2021.PubMed/NCBI View Article : Google Scholar

28 

Michaeloudes C, Li X, Mak JCW and Bhavsar PK: Study of mesenchymal stem cell-mediated mitochondrial transfer in in vitro models of oxidant-mediated airway epithelial and smooth muscle cell injury. Methods Mol Biol. 2269:93–105. 2021.PubMed/NCBI View Article : Google Scholar

29 

Konari N, Nagaishi K, Kikuchi S and Fujimiya M: Mitochondria transfer from mesenchymal stem cells structurally and functionally repairs renal proximal tubular epithelial cells in diabetic nephropathy in vivo. Sci Rep. 9(5184)2019.PubMed/NCBI View Article : Google Scholar

30 

Moschoi R, Imbert V, Nebout M, Chiche J, Mary D, Prebet T, Saland E, Castellano R, Pouyet L, Collette Y, et al: Protective mitochondrial transfer from bone marrow stromal cells to acute myeloid leukemic cells during chemotherapy. Blood. 128:253–264. 2016.PubMed/NCBI View Article : Google Scholar

31 

Lambrecht BN and Hammad H: The immunology of asthma. Nat Immunol. 16:45–56. 2015.PubMed/NCBI View Article : Google Scholar

32 

Chan TK, Tan WSD, Peh HY and Wong WSF: Aeroallergens induce reactive oxygen species production and DNA damage and dampen antioxidant responses in bronchial epithelial cells. J Immunol. 199:39–47. 2017.PubMed/NCBI View Article : Google Scholar

33 

Zhao L, Gao J, Chen G, Huang C, Kong W, Feng Y and Zhen G: Mitochondria dysfunction in airway epithelial cells is associated with type 2-low asthma. Front Genet. 14(1186317)2023.PubMed/NCBI View Article : Google Scholar

34 

Yao Y, Fan XL, Jiang D, Zhang Y, Li X, Xu ZB, Fang SB, Chiu S, Tse HF, Lian Q and Fu QL: Connexin 43-mediated mitochondrial transfer of iPSC-MSCs alleviates asthma inflammation. Stem Cell Reports. 11:1120–1135. 2018.PubMed/NCBI View Article : Google Scholar

35 

Malsin ES and Kamp DW: The mitochondria in lung fibrosis: Friend or foe? Transl Res. 202:1–23. 2018.PubMed/NCBI View Article : Google Scholar

36 

Takahashi M, Mizumura K, Gon Y, Shimizu T, Kozu Y, Shikano S, Iida Y, Hikichi M, Okamoto S, Tsuya K, et al: Iron-dependent mitochondrial dysfunction contributes to the pathogenesis of pulmonary fibrosis. Front Pharmacol. 12(643980)2022.PubMed/NCBI View Article : Google Scholar

37 

Huang T, Lin R, Su Y, Sun H, Zheng X, Zhang J, Lu X, Zhao B, Jiang X, Huang L, et al: Efficient intervention for pulmonary fibrosis via mitochondrial transfer promoted by mitochondrial biogenesis. Nat Commun. 14(5781)2023.PubMed/NCBI View Article : Google Scholar

38 

Li CL, Liu JF and Liu SF: Mitochondrial dysfunction in chronic obstructive pulmonary disease: Unraveling the molecular nexus. Biomedicines. 12(814)2024.PubMed/NCBI View Article : Google Scholar

39 

Canton M, Sánchez-Rodríguez R, Spera I, Venegas FC, Favia M, Viola A and Castegna A: Reactive oxygen species in macrophages: Sources and targets. Front Immunol. 12(734229)2021.PubMed/NCBI View Article : Google Scholar

40 

Aridgides DS, Mellinger DL, Armstrong DA, Hazlett HF, Dessaint JA, Hampton TH, Atkins GT, Carroll JL and Ashare A: Functional and metabolic impairment in cigarette smoke-exposed macrophages is tied to oxidative stress. Sci Rep. 9(9624)2019.PubMed/NCBI View Article : Google Scholar

41 

Tulen CBM, Wang Y, Beentjes D, Jessen PJJ, Ninaber DK, Reynaert NL, van Schooten FJ, Opperhuizen A, Hiemstra PS and Remels AHV: Dysregulated mitochondrial metabolism upon cigarette smoke exposure in various human bronchial epithelial cell models. Dis Model Mech. 15(dmm049247)2022.PubMed/NCBI View Article : Google Scholar

42 

Li X, Michaeloudes C, Zhang Y, Wiegman CH, Adcock IM, Lian Q, Mak JCW, Bhavsar PK and Chung KF: Mesenchymal stem cells alleviate oxidative stress-induced mitochondrial dysfunction in the airways. J Allergy Clin Immunol. 141:1634–1645.e5. 2018.PubMed/NCBI View Article : Google Scholar

43 

Li X, Zhang Y, Yeung SC, Liang Y, Liang X, Ding Y, Ip MS, Tse HF, Mak JC and Lian Q: Mitochondrial transfer of induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial cells attenuates cigarette smoke-induced damage. Am J Respir Cell Mol Biol. 51:455–465. 2014.PubMed/NCBI View Article : Google Scholar

44 

Wu BB, Leung KT and Poon EN: Mitochondrial-targeted therapy for doxorubicin-induced cardiotoxicity. Int J Mol Sci. 23(1912)2022.PubMed/NCBI View Article : Google Scholar

45 

He H, Wang L, Qiao Y, Zhou Q, Li H, Chen S, Yin D, Huang Q and He M: Doxorubicin induces endotheliotoxicity and mitochondrial dysfunction via ROS/eNOS/NO pathway. Front Pharmacol. 10(1531)2020.PubMed/NCBI View Article : Google Scholar

46 

Tang H, Tao A, Song J, Liu Q, Wang H and Rui T: Doxorubicin-induced cardiomyocyte apoptosis: Role of mitofusin 2. Int J Biochem Cell Biol. 88:55–59. 2017.PubMed/NCBI View Article : Google Scholar

47 

Zhang Y, Yu Z, Jiang D, Liang X, Liao S, Zhang Z, Yue W, Li X, Chiu SM, Chai YH, et al: iPSC-MSCs with high intrinsic MIRO1 and sensitivity to TNF-α yield efficacious mitochondrial transfer to rescue anthracycline-induced cardiomyopathy. Stem Cell Reports. 7:749–763. 2016.PubMed/NCBI View Article : Google Scholar

48 

Jin N, Zhang M, Zhou L, Jin S, Cheng H, Li X, Shi Y, Xiang T, Zhang Z, Liu Z, et al: Mitochondria transplantation alleviates cardiomyocytes apoptosis through inhibiting AMPKα-mTOR mediated excessive autophagy. FASEB J. 38(e23655)2024.PubMed/NCBI View Article : Google Scholar

49 

Tanaka-Esposito C, Chen Q and Lesnefsky EJ: Blockade of electron transport before ischemia protects mitochondria and decreases myocardial injury during reperfusion in aged rat hearts. Transl Res. 160:207–216. 2012.PubMed/NCBI View Article : Google Scholar

50 

Huang J, Li R and Wang C: The role of mitochondrial quality control in cardiac ischemia/reperfusion injury. Oxid Med Cell Longev. 2021(5543452)2021.PubMed/NCBI View Article : Google Scholar

51 

Bouhamida E, Morciano G, Perrone M, Kahsay AE, Della Sala M, Wieckowski MR, Fiorica F, Pinton P, Giorgi C and Patergnani S: The interplay of hypoxia signaling on mitochondrial dysfunction and inflammation in cardiovascular diseases and cancer: From molecular mechanisms to therapeutic approaches. Biology (Basel). 11(300)2022.PubMed/NCBI View Article : Google Scholar

52 

Cselenyák A, Pankotai E, Horváth EM, Kiss L and Lacza Z: Mesenchymal stem cells rescue cardiomyoblasts from cell death in an in vitro ischemia model via direct cell-to-cell connections. BMC Cell Biol. 11(29)2010.PubMed/NCBI View Article : Google Scholar

53 

Han H, Hu J, Yan Q, Zhu J, Zhu Z, Chen Y, Sun J and Zhang R: Bone marrow-derived mesenchymal stem cells rescue injured H9c2 cells via transferring intact mitochondria through tunneling nanotubes in an in vitro simulated ischemia/reperfusion model. Mol Med Rep. 13:1517–1524. 2016.PubMed/NCBI View Article : Google Scholar

54 

Mori D, Miyagawa S, Kawamura T, Yoshioka D, Hata H, Ueno T, oda K, Kuratani T, Oota M, Kawai K, et al: Mitochondrial transfer induced by adipose-derived mesenchymal stem cell transplantation improves cardiac function in rat models of ischemic cardiomyopathy. Cell Transplant. 32(9636897221148457)2023.PubMed/NCBI View Article : Google Scholar

55 

Mahrouf-Yorgov M, Augeul L, Da Silva CC, Jourdan M, Rigolet M, Manin S, Ferrera R, Ovize M, Henry A, Guguin A, et al: Mesenchymal stem cells sense mitochondria released from damaged cells as danger signals to activate their rescue properties. Cell Death Differ. 24:1224–1238. 2017.PubMed/NCBI View Article : Google Scholar

56 

Zhang ZH, Zhu W, Ren HZ, Zhao X, Wang S, Ma HC and Shi XL: Mesenchymal stem cells increase expression of heme oxygenase-1 leading to anti-inflammatory activity in treatment of acute liver failure. Stem Cell Res Ther. 8(70)2017.PubMed/NCBI View Article : Google Scholar

57 

Tang Q, Zheng G, Feng Z, Chen Y, Lou Y, Wang C, Zhang X, Zhang Y, Xu H, Shang P and Liu H: Trehalose ameliorates oxidative stress-mediated mitochondrial dysfunction and ER stress via selective autophagy stimulation and autophagic flux restoration in osteoarthritis development. Cell Death Dis. 8(e3081)2017.PubMed/NCBI View Article : Google Scholar

58 

Liu H, Li Z, Cao Y, Cui Y, Yang X, Meng Z and Wang R: Effect of chondrocyte mitochondrial dysfunction on cartilage degeneration: A possible pathway for osteoarthritis pathology at the subcellular level. Mol Med Rep. 20:3308–3316. 2019.PubMed/NCBI View Article : Google Scholar

59 

Sanchez-Lopez E, Coras R, Torres A, Lane NE and Guma M: Synovial inflammation in osteoarthritis progression. Nat Rev Rheumatol. 18:258–275. 2022.PubMed/NCBI View Article : Google Scholar

60 

Zheng L, Zhang Z, Sheng P and Mobasheri A: The role of metabolism in chondrocyte dysfunction and the progression of osteoarthritis. Ageing Res Rev. 66(101249)2021.PubMed/NCBI View Article : Google Scholar

61 

Wang R, Maimaitijuma T, Ma YY, Jiao Y and Cao YP: Mitochondrial transfer from bone-marrow-derived mesenchymal stromal cells to chondrocytes protects against cartilage degenerative mitochondrial dysfunction in rats chondrocytes. Chin Med J (Engl). 134:212–218. 2020.PubMed/NCBI View Article : Google Scholar

62 

Wu Z, Korntner SH, Mullen AM and Zeugolis DI: Collagen type II: From biosynthesis to advanced biomaterials for cartilage engineering. Biomater Biosyst. 4(100030)2021.PubMed/NCBI View Article : Google Scholar

63 

Fahey M, Bennett M, Thomas M, Montney K, Vivancos-Koopman I, Pugliese B, Browning L, Bonassar LJ and Delco M: Mesenchymal stromal cells donate mitochondria to articular chondrocytes exposed to mitochondrial, environmental, and mechanical stress. Sci Rep. 12(21525)2022.PubMed/NCBI View Article : Google Scholar

64 

Michelacci YM, Baccarin RYA and Rodrigues NNP: Chondrocyte homeostasis and differentiation: Transcriptional control and signaling in healthy and osteoarthritic conditions. Life (Basel). 13(1460)2023.PubMed/NCBI View Article : Google Scholar

65 

Fearon U, Canavan M, Biniecka M and Veale DJ: Hypoxia, mitochondrial dysfunction and synovial invasiveness in rheumatoid arthritis. Nat Rev Rheumatol. 12:385–397. 2016.PubMed/NCBI View Article : Google Scholar

66 

Zhang X, Eliasberg CD and Rodeo SA: Mitochondrial dysfunction and potential mitochondrial protectant treatments in tendinopathy. Ann N Y Acad Sci. 1490:29–41. 2021.PubMed/NCBI View Article : Google Scholar

67 

Zhang X, Wada S, Zhang Y, Chen D, Deng XH and Rodeo SA: Assessment of mitochondrial dysfunction in a murine model of supraspinatus tendinopathy. J Bone Joint Surg Am. 103:174–183. 2021.PubMed/NCBI View Article : Google Scholar

68 

Liu YC, Wang HL, Huang YZ, Weng YH, Chen RS, Tsai WC, Yeh TH, Lu CS, Chen YL, Lin YW, et al: Alda-1, an activator of ALDH2, ameliorates Achilles tendinopathy in cellular and mouse models. Biochem Pharmacol. 175(113919)2020.PubMed/NCBI View Article : Google Scholar

69 

Dex S, Alberton P, Willkomm L, Söllradl T, Bago S, Milz S, Shakibaei M, Ignatius A, Bloch W, Clausen-Schaumann H, et al: Tenomodulin is required for tendon endurance running and collagen I fibril adaptation to mechanical load. EBioMedicine. 20:240–254. 2017.PubMed/NCBI View Article : Google Scholar

70 

Wei B, Ji M, Lin Y, Wang S, Liu Y, Geng R, Hu X, Xu L, Li Z, Zhang W and Lu J: Mitochondrial transfer from bone mesenchymal stem cells protects against tendinopathy both in vitro and in vivo. Stem Cell Res Ther. 14(104)2023.PubMed/NCBI View Article : Google Scholar

71 

Lee JM, Hwang JW, Kim MJ, Jung SY, Kim KS, Ahn EH, Min K and Choi YS: Mitochondrial transplantation modulates inflammation and apoptosis, alleviating tendinopathy both in vivo and in vitro. Antioxidants (Basel). 10(696)2021.PubMed/NCBI View Article : Google Scholar

72 

Kubat GB, Bouhamida E, Ulger O, Turkel I, Pedriali G, Ramaccini D, Ekinci O, Ozerklig B, Atalay O, Patergnani S, et al: Mitochondrial dysfunction and skeletal muscle atrophy: Causes, mechanisms, and treatment strategies. Mitochondrion. 72:33–58. 2023.PubMed/NCBI View Article : Google Scholar

73 

Hyatt HW and Powers SK: Mitochondrial dysfunction is a common denominator linking skeletal muscle wasting due to disease, aging, and prolonged inactivity. Antioxidants (Basel). 10(588)2021.PubMed/NCBI View Article : Google Scholar

74 

Matsumoto C, Sekine H, Nahata M, Mogami S, Ohbuchi K, Fujitsuka N and Takeda H: Role of mitochondrial dysfunction in the pathogenesis of cisplatin-induced myotube atrophy. Biol Pharm Bull. 45:780–792. 2022.PubMed/NCBI View Article : Google Scholar

75 

Shen S, Liao Q, Liu J, Pan R, Lee SMY and Lin L: Myricanol rescues dexamethasone-induced muscle dysfunction via a sirtuin 1-dependent mechanism. J Cachexia Sarcopenia Muscle. 10:429–444. 2019.PubMed/NCBI View Article : Google Scholar

76 

Yang B, Yang X, Sun X, Shi J, Shen Y and Chen R: IL-6 deficiency attenuates skeletal muscle atrophy by inhibiting mitochondrial ROS production through the upregulation of PGC-1 α in septic mice. Oxid Med Cell Longev. 2022(9148246)2022.PubMed/NCBI View Article : Google Scholar

77 

Xiao Y, Karam C, Yi J, Zhang L, Li X, Yoon D, Wang H, Dhakal K, Ramlow P, Yu T, et al: ROS-related mitochondrial dysfunction in skeletal muscle of an ALS mouse model during the disease progression. Pharmacol Res. 138:25–36. 2018.PubMed/NCBI View Article : Google Scholar

78 

Moore TM, Lin AJ, Strumwasser AR, Cory K, Whitney K, Ho T, Ho T, Lee JL, Rucker DH, Nguyen CQ, et al: Mitochondrial dysfunction is an early consequence of partial or complete dystrophin loss in mdx mice. Front Physiol. 11(690)2020.PubMed/NCBI View Article : Google Scholar

79 

Mohiuddin M, Choi JJ, Lee NH, Jeong H, Anderson SE, Han WM, Aliya B, Peykova TZ, Verma S, García AJ, et al: Transplantation of muscle stem cell mitochondria rejuvenates the bioenergetic function of dystrophic muscle. bioRxiv: 2020.04.17.017822, 2020.

80 

Wang L, Jiao XF, Wu C, Li XQ, Sun HX, Shen XY, Zhang KZ, Zhao C, Liu L, Wang M, et al: Trimetazidine attenuates dexamethasone-induced muscle atrophy via inhibiting NLRP3/GSDMD pathway-mediated pyroptosis. Cell Death Discov. 7(251)2021.PubMed/NCBI View Article : Google Scholar

81 

Kim MJ, Lee JM, Min K and Choi YS: Xenogeneic transplantation of mitochondria induces muscle regeneration in an in vivo rat model of dexamethasone-induced atrophy. J Muscle Res Cell Motil. 45:53–68. 2024.PubMed/NCBI View Article : Google Scholar

82 

Xu S, Li Y, Chen JP, Li DZ, Jiang Q, Wu T and Zhou XZ: Oxygen glucose deprivation/re-oxygenation-induced neuronal cell death is associated with Lnc-D63785 m6A methylation and miR-422a accumulation. Cell Death Dis. 11(816)2020.PubMed/NCBI View Article : Google Scholar

83 

Juntunen M, Hagman S, Moisan A, Narkilahti S and Miettinen S: In vitro oxygen-glucose deprivation-induced stroke models with human neuroblastoma cell- and induced pluripotent stem cell-derived neurons. Stem Cells Int. 2020(8841026)2020.PubMed/NCBI View Article : Google Scholar

84 

Liu F, Lu J, Manaenko A, Tang J and Hu Q: Mitochondria in ischemic stroke: New insight and implications. Aging Dis. 9:924–937. 2018.PubMed/NCBI View Article : Google Scholar

85 

Norat P, Soldozy S, Sokolowski JD, Gorick CM, Kumar JS, Chae Y, Yağmurlu K, Prada F, Walker M, Levitt MR, et al: Mitochondrial dysfunction in neurological disorders: Exploring mitochondrial transplantation. NPJ Regen Med. 5(22)2020.PubMed/NCBI View Article : Google Scholar

86 

Babenko VA, Silachev DN, Zorova LD, Pevzner IB, Khutornenko AA, Plotnikov EY, Sukhikh GT and Zorov DB: Improving the post-stroke therapeutic potency of mesenchymal multipotent stromal cells by cocultivation with cortical neurons: The role of crosstalk between cells. Stem Cells Transl Med. 4:1011–1020. 2015.PubMed/NCBI View Article : Google Scholar

87 

Jian Z, Ding S, Deng H, Wang J, Yi W, Wang L, Zhu S, Gu L and Xiong X: Probenecid protects against oxygen-glucose deprivation injury in primary astrocytes by regulating inflammasome activity. Brain Res. 1643:123–129. 2016.PubMed/NCBI View Article : Google Scholar

88 

Babenko VA, Silachev DN, Popkov VA, Zorova LD, Pevzner IB, Plotnikov EY, Sukhikh GT and Zorov DB: Miro1 enhances mitochondria transfer from multipotent mesenchymal stem cells (MMSC) to neural cells and improves the efficacy of cell recovery. Molecules. 23(687)2018.PubMed/NCBI View Article : Google Scholar

89 

Li J, Li H, Cai S, Bai S, Cai H and Zhang X: CD157 in bone marrow mesenchymal stem cells mediates mitochondrial production and transfer to improve neuronal apoptosis and functional recovery after spinal cord injury. Stem Cell Res Ther. 12(289)2021.PubMed/NCBI View Article : Google Scholar

90 

Yang Y, Ye G, Zhang YL, He HW, Yu BQ, Hong YM, You W and Li X: Transfer of mitochondria from mesenchymal stem cells derived from induced pluripotent stem cells attenuates hypoxia-ischemia-induced mitochondrial dysfunction in PC12 cells. Neural Regen Res. 15:464–472. 2020.PubMed/NCBI View Article : Google Scholar

91 

Tseng N, Lambie SC, Huynh CQ, Sanford B, Patel M, Herson PS and Ormond DR: Mitochondrial transfer from mesenchymal stem cells improves neuronal metabolism after oxidant injury in vitro: The role of Miro1. J Cereb Blood Flow Metab. 41:761–770. 2021.PubMed/NCBI View Article : Google Scholar

92 

Alizadeh A, Dyck SM and Karimi-Abdolrezaee S: Traumatic spinal cord injury: An overview of pathophysiology, models and acute injury mechanisms. Front Neurol. 10(282)2019.PubMed/NCBI View Article : Google Scholar

93 

Schmidt J and Quintá HR: Mitochondrial dysfunction as a target in spinal cord injury: Intimate correlation between pathological processes and therapeutic approaches. Neural Regen Res. 18:2161–2166. 2023.PubMed/NCBI View Article : Google Scholar

94 

Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al: Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell. 149:1060–1072. 2012.PubMed/NCBI View Article : Google Scholar

95 

Rong Y, Fan J, Ji C, Wang Z, Ge X, Wang J, Ye W, Yin G, Cai W and Liu W: USP11 regulates autophagy-dependent ferroptosis after spinal cord ischemia-reperfusion injury by deubiquitinating beclin 1. Cell Death Differ. 29:1164–1175. 2022.PubMed/NCBI View Article : Google Scholar

96 

Li D, Lu X, Xu G, Liu S, Gong Z, Lu F, Xia X, Jiang J, Wang H, Zou F and Ma X: Dihydroorotate dehydrogenase regulates ferroptosis in neurons after spinal cord injury via the P53-ALOX15 signaling pathway. CNS Neurosci Ther. 29:1923–1939. 2023.PubMed/NCBI View Article : Google Scholar

97 

Stockwell BR: Ferroptosis turns 10: Emerging mechanisms, physiological functions, and therapeutic applications. Cell. 185:2401–2421. 2022.PubMed/NCBI View Article : Google Scholar

98 

Yao S, Pang M, Wang Y, Wang X, Lin Y, Lv Y, Xie Z, Hou J, Du C, Qiu Y, et al: Mesenchymal stem cell attenuates spinal cord injury by inhibiting mitochondrial quality control-associated neuronal ferroptosis. Redox Biol. 67(102871)2023.PubMed/NCBI View Article : Google Scholar

99 

Jain R, Begum N, Tryphena KP, Singh SB, Srivastava S, Rai SN, Vamanu E and Khatri DK: Inter and intracellular mitochondrial transfer: Future of mitochondrial transplant therapy in Parkinson's disease. Biomed Pharmacother. 159(114268)2023.PubMed/NCBI View Article : Google Scholar

100 

Poewe W, Seppi K, Tanner CM, Halliday GM, Brundin P, Volkmann J, Schrag AE and Lang AE: Parkinson disease. Nat Rev Dis Primers. 3(17013)2017.PubMed/NCBI View Article : Google Scholar

101 

van der Bliek AM, Sedensky MM and Morgan PG: Cell biology of the mitochondrion. Genetics. 207:843–871. 2017.PubMed/NCBI View Article : Google Scholar

102 

Ahn EH, Lei K, Kang SS, Wang ZH, Liu X, Hong W, Wang YT, Edgington-Mitchell LE, Jin L and Ye K: Mitochondrial dysfunction triggers the pathogenesis of Parkinson's disease in neuronal C/EBPβ transgenic mice. Mol Psychiatry. 26:7838–7850. 2021.PubMed/NCBI View Article : Google Scholar

103 

Xiao B, Kuruvilla J and Tan EK: Mitophagy and reactive oxygen species interplay in Parkinson's disease. NPJ Parkinsons Dis. 8(135)2022.PubMed/NCBI View Article : Google Scholar

104 

Eo H, Yu SH, Choi Y, Kim Y, Kang YC, Lee H, Kim JH, Han K, Lee HK, Chang MY, et al: Mitochondrial transplantation exhibits neuroprotective effects and improves behavioral deficits in an animal model of Parkinson's disease. Neurotherapeutics. 21(e00355)2024.PubMed/NCBI View Article : Google Scholar

105 

Thakur P and Nehru B: Inhibition of neuroinflammation and mitochondrial dysfunctions by carbenoxolone in the rotenone model of Parkinson's disease. Mol Neurobiol. 51:209–219. 2015.PubMed/NCBI View Article : Google Scholar

106 

Cheng XY, Biswas S, Li J, Mao CJ, Chechneva O, Chen J, Li K, Li J, Zhang JR, Liu CF and Deng WB: Human iPSCs derived astrocytes rescue rotenone-induced mitochondrial dysfunction and dopaminergic neurodegeneration in vitro by donating functional mitochondria. Transl Neurodegener. 9(13)2020.PubMed/NCBI View Article : Google Scholar

107 

Chiu GS, Maj MA, Rizvi S, Dantzer R, Vichaya EG, Laumet G, Kavelaars A and Heijnen CJ: Pifithrin-μ prevents cisplatin-induced chemobrain by preserving neuronal mitochondrial function. Cancer Res. 77:742–752. 2017.PubMed/NCBI View Article : Google Scholar

108 

Kleih M, Böpple K, Dong M, Gaißler A, Heine S, Olayioye MA, Aulitzky WE and Essmann F: Direct impact of cisplatin on mitochondria induces ROS production that dictates cell fate of ovarian cancer cells. Cell Death Dis. 10(851)2019.PubMed/NCBI View Article : Google Scholar

109 

Boukelmoune N, Chiu GS, Kavelaars A and Heijnen CJ: Mitochondrial transfer from mesenchymal stem cells to neural stem cells protects against the neurotoxic effects of cisplatin. Acta Neuropathol Commun. 6(139)2018.PubMed/NCBI View Article : Google Scholar

110 

Méthot S, Proulx S, Brunette I and Rochette PJ: Rescuing cellular function in Fuchs endothelial corneal dystrophy by healthy exogenous mitochondrial internalization. Sci Rep. 13(3380)2023.PubMed/NCBI View Article : Google Scholar

111 

Jiang D, Chen FX, Zhou H, Lu YY, Tan H, Yu SJ, Yuan J, Liu H, Meng W and Jin ZB: Bioenergetic crosstalk between mesenchymal stem cells and various ocular cells through the intercellular trafficking of mitochondria. Theranostics. 10:7260–7272. 2020.PubMed/NCBI View Article : Google Scholar

112 

Vallabh NA, Romano V and Willoughby CE: Mitochondrial dysfunction and oxidative stress in corneal disease. Mitochondrion. 36:103–113. 2017.PubMed/NCBI View Article : Google Scholar

113 

Numa K, Ueno M, Fujita T, Ueda K, Hiramoto N, Mukai A, Tokuda Y, Nakano M, Sotozono C, Kinoshita S and Hamuro J: Mitochondria as a Platform for dictating the cell fate of cultured human corneal endothelial cells. Invest Ophthalmol Vis Sci. 61(10)2020.PubMed/NCBI View Article : Google Scholar

114 

Jiang D, Gao F, Zhang Y, Wong DS, Li Q, Tse HF, Xu G, Yu Z and Lian Q: Mitochondrial transfer of mesenchymal stem cells effectively protects corneal epithelial cells from mitochondrial damage. Cell Death Dis. 7(e2467)2016.PubMed/NCBI View Article : Google Scholar

115 

Hanna SJ, McCoy-Simandle K, Leung E, Genna A, Condeelis J and Cox D: Tunneling nanotubes, a novel mode of tumor cell-macrophage communication in tumor cell invasion. J Cell Sci. 132(jcs223321)2019.PubMed/NCBI View Article : Google Scholar

116 

Wang L, Klingeborn M, Travis AM, Hao Y, Arshavsky VY and Gospe SM III: Progressive optic atrophy in a retinal ganglion cell-specific mouse model of complex I deficiency. Sci Rep. 10(16326)2020.PubMed/NCBI View Article : Google Scholar

117 

Yu AK, Song L, Murray KD, van der List D, Sun C, Shen Y, Xia Z and Cortopassi GA: Mitochondrial complex I deficiency leads to inflammation and retinal ganglion cell death in the Ndufs4 mouse. Hum Mol Genet. 24:2848–2860. 2015.PubMed/NCBI View Article : Google Scholar

118 

Warwick AM, Bomze HM, Wang L, Hao Y, Stinnett SS and Gospe SM III: Hypoxia-mediated rescue of retinal ganglion cells deficient in mitochondrial complex I is independent of the hypoxia-inducible factor pathway. Sci Rep. 14(24114)2024.PubMed/NCBI View Article : Google Scholar

119 

Jiang D, Xiong G, Feng H, Zhang Z, Chen P, Yan B, Chen L, Gandhervin K, Ma C, Li C, et al: Donation of mitochondria by iPSC-derived mesenchymal stem cells protects retinal ganglion cells against mitochondrial complex I defect-induced degeneration. Theranostics. 9:2395–2410. 2019.PubMed/NCBI View Article : Google Scholar

120 

Navneet S, Wilson K and Rohrer B: Muller glial cells in the macula: Their activation and cell-cell interactions in age-related macular degeneration. Invest Ophthalmol Vis Sci. 65(42)2024.PubMed/NCBI View Article : Google Scholar

121 

Xue B, Xie Y, Xue Y, Hu N, Zhang G, Guan H and Ji M: Involvement of P2X7 receptors in retinal ganglion cell apoptosis induced by activated Müller cells. Exp Eye Res. 153:42–50. 2016.PubMed/NCBI View Article : Google Scholar

122 

Hu X, Zhao GL, Xu MX, Zhou H, Li F, Miao Y, Lei B, Yang XL and Wang Z: Interplay between Muller cells and microglia aggravates retinal inflammatory response in experimental glaucoma. J Neuroinflammation. 18(303)2021.PubMed/NCBI View Article : Google Scholar

123 

Qiu AW, Bian Z, Mao PA and Liu QH: IL-17A exacerbates diabetic retinopathy by impairing Müller cell function via Act1 signaling. Exp Mol Med. 48(e280)2016.PubMed/NCBI View Article : Google Scholar

124 

Natoli R, Fernando N, Madigan M, Chu-Tan JA, Valter K, Provis J and Rutar M: Microglia-derived IL-1β promotes chemokine expression by Müller cells and RPE in focal retinal degeneration. Mol Neurodegener. 12(31)2017.PubMed/NCBI View Article : Google Scholar

125 

Marques E, Alves Teixeira M, Nguyen C, Terzi F and Gallazzini M: Lipocalin-2 induces mitochondrial dysfunction in renal tubular cells via mTOR pathway activation. Cell Rep. 42(113032)2023.PubMed/NCBI View Article : Google Scholar

126 

Chen Y, Yang Y, Liu Z and He L: Adiponectin promotes repair of renal tubular epithelial cells by regulating mitochondrial biogenesis and function. Metabolism. 128(154959)2022.PubMed/NCBI View Article : Google Scholar

127 

Ji X, Yang X, Gu X, Chu L, Sun S, Sun J, Song P, Mu Q, Wang Y, Sun X, et al: CUL3 induces mitochondrial dysfunction via MRPL12 ubiquitination in renal tubular epithelial cells. FEBS J. 290:5340–5352. 2023.PubMed/NCBI View Article : Google Scholar

128 

Cai F, Li D, Xie Y, Wang X, Ma H, Xu H, Cheng J, Zhuang H and Hua ZC: Sulfide:quinone oxidoreductase alleviates ferroptosis in acute kidney injury via ameliorating mitochondrial dysfunction of renal tubular epithelial cells. Redox Biol. 69(102973)2024.PubMed/NCBI View Article : Google Scholar

129 

Xiong YB, Huang WY, Ling X, Zhou S, Wang XX, Li XL and Zhou LL: Mitochondrial calcium uniporter promotes kidney aging in mice through inducing mitochondrial calcium-mediated renal tubular cell senescence. Acta Pharmacol Sin. 45:2149–2162. 2024.PubMed/NCBI View Article : Google Scholar

130 

Wang J, Yue X, Meng C, Wang Z, Jin X, Cui X, Yang J, Shan C, Gao Z, Yang Y, et al: Acute hyperglycemia may induce renal tubular injury through mitophagy inhibition. Front Endocrinol (Lausanne). 11(536213)2020.PubMed/NCBI View Article : Google Scholar

131 

Aluksanasuwan S, Plumworasawat S, Malaitad T, Chaiyarit S and Thongboonkerd V: High glucose induces phosphorylation and oxidation of mitochondrial proteins in renal tubular cells: A proteomics approach. Sci Rep. 10(5843)2020.PubMed/NCBI View Article : Google Scholar

132 

Nahdi AMTA, John A and Raza H: Elucidation of molecular mechanisms of streptozotocin-induced oxidative stress, apoptosis, and mitochondrial dysfunction in Rin-5F pancreatic β-cells. Oxid Med Cell Longev. 2017(7054272)2017.PubMed/NCBI View Article : Google Scholar

133 

Bhargava P and Schnellmann RG: Mitochondrial energetics in the kidney. Nat Rev Nephrol. 13:629–646. 2017.PubMed/NCBI View Article : Google Scholar

134 

Kapetanaki S, Kumawat AK, Persson K and Demirel I: TMAO suppresses megalin expression and albumin uptake in human proximal tubular cells via PI3K and ERK signaling. Int J Mol Sci. 23(8856)2022.PubMed/NCBI View Article : Google Scholar

135 

Peruchetti DB, Silva-Aguiar RP, Siqueira GM, Dias WB and Caruso-Neves C: High glucose reduces megalin-mediated albumin endocytosis in renal proximal tubule cells through protein kinase B O-GlcNAcylation. J Biol Chem. 293:11388–11400. 2018.PubMed/NCBI View Article : Google Scholar

136 

Kupriyanova Y, Zaharia OP, Bobrov P, Karusheva Y, Burkart V, Szendroedi J, Hwang JH and Roden M: GDS group. Early changes in hepatic energy metabolism and lipid content in recent-onset type 1 and 2 diabetes mellitus. J Hepatol. 74:1028–1037. 2021.PubMed/NCBI View Article : Google Scholar

137 

Tilg H, Moschen AR and Roden M: NAFLD and diabetes mellitus. Nat Rev Gastroenterol Hepatol. 14:32–42. 2017.PubMed/NCBI View Article : Google Scholar

138 

Nassir F and Ibdah JA: Role of mitochondria in nonalcoholic fatty liver disease. Int J Mol Sci. 15:8713–8742. 2014.PubMed/NCBI View Article : Google Scholar

139 

Zheng P, Ma W, Gu Y, Wu H, Bian Z, Liu N, Yang D and Chen X: High-fat diet causes mitochondrial damage and downregulation of mitofusin-2 and optic atrophy-1 in multiple organs. J Clin Biochem Nutr. 73:61–76. 2023.PubMed/NCBI View Article : Google Scholar

140 

Bi Y, Guo X, Zhang M, Zhu K, Shi C, Fan B, Wu Y, Yang Z and Ji G: Bone marrow derived-mesenchymal stem cell improves diabetes-associated fatty liver via mitochondria transformation in mice. Stem Cell Res Ther. 12(602)2021.PubMed/NCBI View Article : Google Scholar

141 

Hsu MJ, Karkossa I, Schäfer I, Christ M, Kühne H, Schubert K, Rolle-Kampczyk UE, Kalkhof S, Nickel S, Seibel P, et al: Mitochondrial transfer by human mesenchymal stromal cells ameliorates hepatocyte lipid load in a mouse model of NASH. Biomedicines. 8(350)2020.PubMed/NCBI View Article : Google Scholar

142 

Gaspers LD, Pierobon N and Thomas AP: Intercellular calcium waves integrate hormonal control of glucose output in the intact liver. J Physiol. 597:2867–2885. 2019.PubMed/NCBI View Article : Google Scholar

143 

Barreto SG, Habtezion A, Gukovskaya A, Lugea A, Jeon C, Yadav D, Hegyi P, Venglovecz V, Sutton R and Pandol SJ: Critical thresholds: key to unlocking the door to the prevention and specific treatments for acute pancreatitis. Gut. 70:194–203. 2021.PubMed/NCBI View Article : Google Scholar

144 

Liu W, Ren Y, Wang T, Wang M, Xu Y, Zhang J, Bi J, Wu Z, Zhang Y and Wu R: Blocking CIRP protects against acute pancreatitis by improving mitochondrial function and suppressing pyroptosis in acinar cells. Cell Death Discov. 10(156)2024.PubMed/NCBI View Article : Google Scholar

145 

Mukherjee R, Mareninova OA, Odinokova IV, Huang W, Murphy J, Chvanov M, Javed MA, Wen L, Booth DM, Cane MC, et al: Mechanism of mitochondrial permeability transition pore induction and damage in the pancreas: Inhibition prevents acute pancreatitis by protecting production of ATP. Gut. 65:1333–1346. 2016.PubMed/NCBI View Article : Google Scholar

146 

Hu Z, Wang D, Gong J, Li Y, Ma Z, Luo T, Jia X, Shi Y and Song Z: MSCs deliver hypoxia-treated mitochondria reprogramming acinar metabolism to alleviate severe acute pancreatitis injury. Adv Sci (Weinh). 10(e2207691)2023.PubMed/NCBI View Article : Google Scholar

147 

Shapiro AM, Pokrywczynska M and Ricordi C: Clinical pancreatic islet transplantation. Nat Rev Endocrinol. 13:268–277. 2017.PubMed/NCBI View Article : Google Scholar

148 

Kin T, Senior P, O'Gorman D, Richer B, Salam A and Shapiro AMJ: Risk factors for islet loss during culture prior to transplantation. Transpl Int. 21:1029–1035. 2008.PubMed/NCBI View Article : Google Scholar

149 

Jimenez-Sánchez C, Brun T and Maechler P: Mitochondrial carriers regulating insulin secretion profiled in human islets upon metabolic stress. Biomolecules. 10(1543)2020.PubMed/NCBI View Article : Google Scholar

150 

Zhang K, Bao R, Huang F, Yang K, Ding Y, Lauterboeck L, Yoshida M, Long Q and Yang Q: ATP synthase inhibitory factor subunit 1 regulates islet β-cell function via repression of mitochondrial homeostasis. Lab Invest. 102:69–79. 2022.PubMed/NCBI View Article : Google Scholar

151 

Rackham CL, Hubber EL, Czajka A, Malik AN, King AJF and Jones PM: Optimizing beta cell function through mesenchymal stromal cell-mediated mitochondria transfer. Stem Cells. 38:574–584. 2020.PubMed/NCBI View Article : Google Scholar

152 

Shoop WK, Lape J, Trum M, Powell A, Sevigny E, Mischler A, Bacman SR, Fontanesi F, Smith J, Jantz D, et al: Efficient elimination of MELAS-associated m.3243G mutant mitochondrial DNA by an engineered mitoARCUS nuclease. Nat Metab. 5:2169–2183. 2023.PubMed/NCBI View Article : Google Scholar

153 

Ikeda T, Osaka H, Shimbo H, Tajika M, Yamazaki M, Ueda A, Murayama K and Yamagata T: Mitochondrial DNA 3243A>T mutation in a patient with MELAS syndrome. Hum Genome Var. 5(25)2018.PubMed/NCBI View Article : Google Scholar

154 

El-Hattab AW, Adesina AM, Jones J and Scaglia F: MELAS syndrome: Clinical manifestations, pathogenesis, and treatment options. Mol Genet Metab. 116:4–12. 2015.PubMed/NCBI View Article : Google Scholar

155 

Lin TK, Chen SD, Chuang YC, Lan MY, Chuang JH, Wang PW, Hsu TY, Wang FS, Tsai MH, Huang ST, et al: Mitochondrial transfer of wharton's jelly mesenchymal stem cells eliminates mutation burden and rescues mitochondrial bioenergetics in rotenone-stressed MELAS fibroblasts. Oxid Med Cell Longev. 2019(9537504)2019.PubMed/NCBI View Article : Google Scholar

156 

Liu L, Yang J, Otani Y, Shiga T, Yamaguchi A, Oda Y, Hattori M, Goto T, Ishibashi S, Kawashima-Sonoyama Y, et al: MELAS-derived neurons functionally improve by mitochondrial transfer from highly purified mesenchymal stem cells (REC). Int J Mol Sci. 24(17186)2023.PubMed/NCBI View Article : Google Scholar

157 

Tuppen HA, Blakely EL, Turnbull DM and Taylor RW: Mitochondrial DNA mutations and human disease. Biochim Biophys Acta. 1797:113–128. 2010.PubMed/NCBI View Article : Google Scholar

158 

Chuang YC, Liou CW, Chen SD, Wang PW, Chuang JH, Tiao MM, Hsu TY, Lin HY and Lin TK: Mitochondrial transfer from wharton's jelly mesenchymal stem cell to MERRF cybrid reduces oxidative stress and improves mitochondrial bioenergetics. Oxid Med Cell Longev. 2017(5691215)2017.PubMed/NCBI View Article : Google Scholar

159 

Capristo M, Del Dotto V, Tropeano CV, Fiorini C, Caporali L, La Morgia C, Valentino ML, Montopoli M, Carelli V and Maresca A: Rapamycin rescues mitochondrial dysfunction in cells carrying the m.8344A > G mutation in the mitochondrial tRNALys. Mol Med. 28(90)2022.PubMed/NCBI View Article : Google Scholar

160 

Jacoby E, Blumkin M, Anikster Y, Varda-Bloom N, Pansheen J, Bar Yoseph O, Gruber N, Lahav E, Besser MJ, Schachter J, et al: First-in-human mitochondrial augmentation of hematopoietic stem cells in pearson syndrome. Blood. 132(1024)2018.

161 

Jacoby E, Bar-Yosef O, Gruber N, Lahav E, Varda-Bloom N, Bolkier Y, Bar D, Blumkin MB, Barak S, Eisenstein E, et al: Mitochondrial augmentation of hematopoietic stem cells in children with single large-scale mitochondrial DNA deletion syndromes. Sci Transl Med. 14(eabo3724)2022.PubMed/NCBI View Article : Google Scholar

162 

Fu Y, Ni J and Chen J, Ma G, Zhao M, Zhu S, Shi T, Zhu J, Huang Z, Zhang J and Chen J: Dual-functionalized MSCs that express CX3CR1 and IL-25 exhibit enhanced therapeutic effects on inflammatory bowel disease. Mol Ther. 28:1214–1228. 2020.PubMed/NCBI View Article : Google Scholar

163 

Van Nguyen TT, Vu NB and Van Pham P: Mesenchymal stem cell transplantation for ischemic diseases: Mechanisms and challenges. Tissue Eng Regen Med. 18:587–611. 2021.PubMed/NCBI View Article : Google Scholar

164 

Zhu XY, Lerman A and Lerman LO: Concise review: Mesenchymal stem cell treatment for ischemic kidney disease. Stem Cells. 31:1731–1736. 2013.PubMed/NCBI View Article : Google Scholar

165 

Baldari S, Di Rocco G, Piccoli M, Pozzobon M, Muraca M and Toietta G: Challenges and strategies for improving the regenerative effects of mesenchymal stromal cell-based therapies. Int J Mol Sci. 18(2087)2017.PubMed/NCBI View Article : Google Scholar

166 

Hu C and Li L: Preconditioning influences mesenchymal stem cell properties in vitro and in vivo. J Cell Mol Med. 22:1428–1442. 2018.PubMed/NCBI View Article : Google Scholar

167 

Li CJ, Chen PK, Sun LY and Pang CY: Enhancement of mitochondrial transfer by antioxidants in human mesenchymal stem cells. Oxid Med Cell Longev. 2017(8510805)2017.PubMed/NCBI View Article : Google Scholar

168 

Lee DS and Kim JE: PDI-mediated S-nitrosylation of DRP1 facilitates DRP1-S616 phosphorylation and mitochondrial fission in CA1 neurons. Cell Death Dis. 9(869)2018.PubMed/NCBI View Article : Google Scholar

169 

Yao X, Ma Y, Zhou W, Liao Y, Jiang Z, Lin J, He Q, Wu H, Wei W, Wang X, et al: In-cytoplasm mitochondrial transplantation for mesenchymal stem cells engineering and tissue regeneration. Bioeng Transl Med. 7(e10250)2021.PubMed/NCBI View Article : Google Scholar

170 

Guo Y, Chi X, Wang Y, Heng BC, Wei Y, Zhang X, Zhao H, Yin Y and Deng X: Mitochondria transfer enhances proliferation, migration, and osteogenic differentiation of bone marrow mesenchymal stem cell and promotes bone defect healing. Stem Cell Res Ther. 11(245)2020.PubMed/NCBI View Article : Google Scholar

171 

Lin RZ, Im GB, Luo AC, Zhu Y, Hong X, Neumeyer J, Tang HW, Perrimon N and Melero-Martin JM: Mitochondrial transfer mediates endothelial cell engraftment through mitophagy. Nature. 629:660–668. 2024.PubMed/NCBI View Article : Google Scholar

172 

Akhter W, Nakhle J, Vaillant L, Garcin G, Le Saout C, Simon M, Crozet C, Djouad F, Jorgensen C, Vignais ML and Hernandez J: Transfer of mesenchymal stem cell mitochondria to CD4+ T cells contributes to repress Th1 differentiation by downregulating T-bet expression. Stem Cell Res Ther. 14(12)2023.PubMed/NCBI View Article : Google Scholar

173 

Court AC, Le-Gatt A, Luz-Crawford P, Parra E, Aliaga-Tobar V, Bátiz LF, Contreras RA, Ortúzar MI, Kurte M, Elizondo-Vega R, et al: Mitochondrial transfer from MSCs to T cells induces Treg differentiation and restricts inflammatory response. EMBO Rep. 21(e48052)2020.PubMed/NCBI View Article : Google Scholar

174 

Romano M, Tung SL, Smyth LA and Lombardi G: Treg therapy in transplantation: A general overview. Transpl Int. 30:745–753. 2017.PubMed/NCBI View Article : Google Scholar

175 

Li LZ, Zhang Z and Bhoj VG: Conventional T cell therapies pave the way for novel Treg therapeutics. Cell Immunol. 359(104234)2021.PubMed/NCBI View Article : Google Scholar

176 

Piekarska K, Urban-Wójciuk Z, Kurkowiak M, Pelikant-Małecka I, Schumacher A, Sakowska J, Spodnik JH, Arcimowicz Ł, Zielińska H, Tymoniuk B, et al: Mesenchymal stem cells transfer mitochondria to allogeneic Tregs in an HLA-dependent manner improving their immunosuppressive activity. Nat Commun. 13(856)2022.PubMed/NCBI View Article : Google Scholar

177 

Espinosa-Carrasco G, Le Saout C, Fontanaud P, Stratmann T, Mollard P, Schaeffer M and Hernandez J: CD4+ T helper cells play a key role in maintaining diabetogenic CD8+ T cell function in the pancreas. Front Immunol. 8(2001)2018.PubMed/NCBI View Article : Google Scholar

178 

Luz-Crawford P, Hernandez J, Djouad F, Luque-Campos N, Caicedo A, Carrère-Kremer S, Brondello JM, Vignais ML, Pène J and Jorgensen C: Mesenchymal stem cell repression of Th17 cells is triggered by mitochondrial transfer. Stem Cell Res Ther. 10(232)2019.PubMed/NCBI View Article : Google Scholar

179 

Lazarevic V, Glimcher LH and Lord GM: T-bet: A bridge between innate and adaptive immunity. Nat Rev Immunol. 13:777–789. 2013.PubMed/NCBI View Article : Google Scholar : Ji L, Chen Y, Wang H, Zhang W, He L, Wu J and Liu Y: Overexpression of Sirt6 promotes M2 macrophage transformation, alleviating renal injury in diabetic nephropathy. Int J Oncol 55: 103-115, 2019.

180 

Xu L, Yan X, Zhao Y, Wang J, Liu B, Yu S, Fu J, Liu Y and Su J: Macrophage polarization mediated by mitochondrial dysfunction induces adipose tissue inflammation in obesity. Int J Mol Sci. 23(9252)2022.PubMed/NCBI View Article : Google Scholar

181 

Ran L, Zhang S, Wang G, Zhao P, Sun J, Zhou J, Gan H, Jeon R, Li Q, Herrmann J and Wang F: Mitochondrial pyruvate carrier-mediated metabolism is dispensable for the classical activation of macrophages. Nat Metab. 5:804–820. 2023.PubMed/NCBI View Article : Google Scholar

182 

De Santa F, Vitiello L, Torcinaro A and Ferraro E: The role of metabolic remodeling in macrophage polarization and its effect on skeletal muscle regeneration. Antioxid Redox Signal. 30:1553–1598. 2019.PubMed/NCBI View Article : Google Scholar

183 

Yuan Y, Chen Y, Peng T, Li L, Zhu W, Liu F, Luo R, Cheng J, Liu J and Lu Y: Mitochondrial ROS-induced lysosomal dysfunction impairs autophagic flux and contributes to M1 macrophage polarization in a diabetic condition. Clin Sci (Lond). 133:1759–1777. 2019.PubMed/NCBI View Article : Google Scholar

184 

Yuan Y, Yuan L, Li L, Liu F, Liu J, Chen Y, Cheng J and Lu Y: Mitochondrial transfer from mesenchymal stem cells to macrophages restricts inflammation and alleviates kidney injury in diabetic nephropathy mice via PGC-1α activation. Stem Cells. 39:913–928. 2021.PubMed/NCBI View Article : Google Scholar

185 

Hussell T and Bell TJ: Alveolar macrophages: Plasticity in a tissue-specific context. Nat Rev Immunol. 14:81–93. 2014.PubMed/NCBI View Article : Google Scholar

186 

Jackson MV, Morrison TJ, Doherty DF, McAuley DF, Matthay MA, Kissenpfennig A, O'Kane CM and Krasnodembskaya AD: Mitochondrial transfer via tunneling nanotubes is an important mechanism by which mesenchymal stem cells enhance macrophage phagocytosis in the in vitro and in vivo models of ARDS. Stem Cells. 34:2210–2223. 2016.PubMed/NCBI View Article : Google Scholar

187 

Jackson MV and Krasnodembskaya AD: Analysis of mitochondrial transfer in direct Co-cultures of human monocyte-derived macrophages (MDM) and mesenchymal stem cells (MSC). Bio Protoc. 7(e2255)2017.PubMed/NCBI View Article : Google Scholar

188 

Phinney DG, Di Giuseppe M, Njah J, Sala E, Shiva S, St Croix CM, Stolz DB, Watkins SC, Di YP, Leikauf GD, et al: Mesenchymal stem cells use extracellular vesicles to outsource mitophagy and shuttle microRNAs. Nat Commun. 6(8472)2015.PubMed/NCBI View Article : Google Scholar

189 

Xia L, Zhang C, Lv N, Liang Z, Ma T, Cheng H, Xia Y and Shi L: AdMSC-derived exosomes alleviate acute lung injury via transferring mitochondrial component to improve homeostasis of alveolar macrophages. Theranostics. 12:2928–2947. 2022.PubMed/NCBI View Article : Google Scholar

190 

West AP, Khoury-Hanold W, Staron M, Tal MC, Pineda CM, Lang SM, Bestwick M, Duguay BA, Raimundo N, MacDuff DA, et al: Mitochondrial DNA stress primes the antiviral innate immune response. Nature. 520:553–557. 2015.PubMed/NCBI View Article : Google Scholar

191 

Morrison TJ, Jackson MV, Cunningham EK, Kissenpfennig A, McAuley DF, O'Kane CM and Krasnodembskaya AD: Mesenchymal stromal cells modulate macrophages in clinically relevant lung injury models by extracellular vesicle mitochondrial transfer. Am J Respir Crit Care Med. 196:1275–1286. 2017.PubMed/NCBI View Article : Google Scholar

192 

Kang MH, Kim YJ and Lee JH: Mitochondria in reproduction. Clin Exp Reprod Med. 50:1–11. 2023.PubMed/NCBI View Article : Google Scholar

193 

Dadarwal D, Pfeifer L, Cervantes M, Adams GP and Singh J: Effect of maternal age on ATP content and distribution of mitochondria in bovine oocytes. PLoS One. 19(e0302444)2024.PubMed/NCBI View Article : Google Scholar

194 

Zhao J and Li Y: Adenosine triphosphate content in human unfertilized oocytes, undivided zygotes and embryos unsuitable for transfer or cryopreservation. J Int Med Res. 40:734–739. 2012.PubMed/NCBI View Article : Google Scholar

195 

May-Panloup P, Chrétien MF, Jacques C, Vasseur C, Malthièry Y and Reynier P: Low oocyte mitochondrial DNA content in ovarian insufficiency. Hum Reprod. 20:593–597. 2005.PubMed/NCBI View Article : Google Scholar

196 

Zhang D, Keilty D, Zhang ZF and Chian RC: Mitochondria in oocyte aging: Current understanding. Facts Views Vis Obgyn. 9:29–38. 2017.PubMed/NCBI

197 

Cozzolino M, Marin D and Sisti G: New frontiers in IVF: mtDNA and autologous germline mitochondrial energy transfer. Reprod Biol Endocrinol. 17(55)2019.PubMed/NCBI View Article : Google Scholar

198 

Takeda K: Functional consequences of mitochondrial mismatch in reconstituted embryos and offspring. J Reprod Dev. 65:485–489. 2019.PubMed/NCBI View Article : Google Scholar

199 

Tang S, Yang N, Yu M, Wang S, Hu X, Ni H and Cai W: Noninvasive autologous mitochondria transport improves the quality and developmental potential of oocytes from aged mice. F S Sci. 3:310–321. 2022.PubMed/NCBI View Article : Google Scholar

200 

Wang ZB, Hao JX, Meng TG, Guo L, Dong MZ, Fan LH, Ouyang YC, Wang G, Sun QY, Ou XH and Yao YQ: Transfer of autologous mitochondria from adipose tissue-derived stem cells rescues oocyte quality and infertility in aged mice. Aging (Albany NY). 9:2480–2488. 2017.PubMed/NCBI View Article : Google Scholar

201 

Kankanam Gamage US, Hashimoto S, Miyamoto Y, Nakano T, Yamanaka M, Koike A, Satoh M and Morimoto Y: Mitochondria transfer from adipose stem cells improves the developmental potential of cryopreserved oocytes. Biomolecules. 12(1008)2022.PubMed/NCBI View Article : Google Scholar

202 

Zhang Q, Hao JX, Liu BW, Ouyang YC, Guo JN, Dong MZ, Wang ZB, Gao F and Yao YQ: Supplementation of mitochondria from endometrial mesenchymal stem cells improves oocyte quality in aged mice. Cell Prolif. 56(e13372)2023.PubMed/NCBI View Article : Google Scholar

203 

Zhang C, Tao L, Yue Y, Ren L, Zhang Z, Wang X, Tian J and An L: Mitochondrial transfer from induced pluripotent stem cells rescues developmental potential of in vitro fertilized embryos from aging females†. Biol Reprod. 104:1114–1125. 2021.PubMed/NCBI View Article : Google Scholar

204 

Tilly J and Woods D: Compositions and methods for autologous germline mitochondrial energy transfer. United States Patent Number. 8,642,329, 2014.

205 

Fakih MH, Shmoury ME, Szeptycki J, Cruz DBD, Lux C, Verjee S, Burgess CM, Cohn GM and Casper RF: The AUGMENTSM treatment: Physician reported outcomes of the initial global patient experience. JFIV Reprod Med Genet. 3:1–7. 2015.

206 

Oktay K, Baltaci V, Sonmezer M, Turan V, Unsal E, Baltaci A, Aktuna S and Moy F: Oogonial precursor cell-derived autologous mitochondria injection to improve outcomes in women with multiple IVF failures due to low oocyte quality: A clinical translation. Reprod Sci. 22:1612–1617. 2015.PubMed/NCBI View Article : Google Scholar

207 

Jiang Z, Shi C, Han H, Fu M, Zhu H, Han T, Fei J, Huang Y, Jin Z, He J, et al: Autologous non-invasively derived stem cells mitochondria transfer shows therapeutic advantages in human embryo quality rescue. Biol Res. 56(60)2023.PubMed/NCBI View Article : Google Scholar

208 

Morimoto Y, Gamage USK, Yamochi T, Saeki N, Morimoto N, Yamanaka M, Koike A, Miyamoto Y, Tanaka K, Fukuda A, et al: Mitochondrial transfer into human oocytes improved embryo quality and clinical outcomes in recurrent pregnancy failure cases. Int J Mol Sci. 24(2738)2023.PubMed/NCBI View Article : Google Scholar

209 

Acquistapace A, Bru T, Lesault PF, Figeac F, Coudert AE, le Coz O, Christov C, Baudin X, Auber F, Yiou R, et al: Human mesenchymal stem cells reprogram adult cardiomyocytes toward a progenitor-like state through partial cell fusion and mitochondria transfer. Stem Cells. 29:812–824. 2011.PubMed/NCBI View Article : Google Scholar

210 

Jiao H, Jiang D, Hu X, Du W, Ji L, Yang Y, Li X, Sho T, Wang X, Li Y, et al: Mitocytosis, a migrasome-mediated mitochondrial quality-control process. Cell. 184:2896–2910.e13. 2021.PubMed/NCBI View Article : Google Scholar

211 

Sadeghsoltani F, Avci ÇB, Hassanpour P, Haiaty S, Rahmati M, Mota A, Rahbarghazi R, Nemati M, Mahdipour M, Talebi M, et al: Autophagy modulation effect on homotypic transfer of intracellular components via tunneling nanotubes in mesenchymal stem cells. Stem Cell Res Ther. 15(189)2024.PubMed/NCBI View Article : Google Scholar

212 

Dai L, Wu Z, Yin L, Cheng L, Zhou Q and Ding F: Exogenous functional mitochondria derived from bone mesenchymal stem cells that respond to ROS can rescue neural cells following ischemic stroke. J Inflamm Res. 17:3383–3395. 2024.PubMed/NCBI View Article : Google Scholar

213 

Mistry JJ, Marlein CR, Moore JA, Hellmich C, Wojtowicz EE, Smith JGW, Macaulay I, Sun Y, Morfakis A, Patterson A, et al: ROS-mediated PI3K activation drives mitochondrial transfer from stromal cells to hematopoietic stem cells in response to infection. Proc Natl Acad Sci USA. 116:24610–24619. 2019.PubMed/NCBI View Article : Google Scholar

214 

Li Y, Wang Y, Yang W, Wu Z, Ma D, Sun J, Tao H, Ye Q, Liu J, Ma Z, et al: ROS-responsive exogenous functional mitochondria can rescue neural cells post-ischemic stroke. Front Cell Dev Biol. 11(1207748)2023.PubMed/NCBI View Article : Google Scholar

215 

Saito K, Zhang Q, Yang H, Yamatani K, Ai T, Ruvolo V, Baran N, Cai T, Ma H, Jacamo R, et al: Exogenous mitochondrial transfer and endogenous mitochondrial fission facilitate AML resistance to OxPhos inhibition. Blood Adv. 5:4233–4255. 2021.PubMed/NCBI View Article : Google Scholar

216 

Ahmad T, Mukherjee S, Pattnaik B, Kumar M, Singh S, Kumar M, Rehman R, Tiwari BK, Jha KA, Barhanpurkar AP, et al: Miro1 regulates intercellular mitochondrial transport & enhances mesenchymal stem cell rescue efficacy. EMBO J. 33:994–1010. 2014.PubMed/NCBI View Article : Google Scholar

217 

Novak J, Nahacka Z, Oliveira GL, Brisudova P, Dubisova M, Dvorakova S, Miklovicova S, Dalecka M, Puttrich V, Grycova L, et al: The adaptor protein Miro1 modulates horizontal transfer of mitochondria in mouse melanoma models. Cell Rep. 44(115154)2025.PubMed/NCBI View Article : Google Scholar

218 

Barutta F, Corbetta B, Bellini S, Gambino R, Bruno S, Kimura S, Hase K, Ohno H and Gruden G: Protective effect of mesenchymal stromal cells in diabetic nephropathy: The In vitro and In vivo role of the M-Sec-tunneling nanotubes. Clin Sci (Lond). 138:1537–1559. 2024.PubMed/NCBI View Article : Google Scholar

219 

Barutta F, Kimura S, Hase K, Bellini S, Corbetta B, Corbelli A, Fiordaliso F, Barreca A, Papotti MG, Ghiggeri GM, et al: Protective role of the M-Sec-tunneling nanotube system in podocytes. J Am Soc Nephrol. 32:1114–1130. 2021.PubMed/NCBI View Article : Google Scholar

220 

Kastl L, Sauer SW, Ruppert T, Beissbarth T, Becker MS, Süss D, Krammer PH and Gülow K: TNF-α mediates mitochondrial uncoupling and enhances ROS-dependent cell migration via NF-κB activation in liver cells. FEBS Lett. 588:175–183. 2014.PubMed/NCBI View Article : Google Scholar

221 

Wang K, Zhou L, Mao H, Liu J, Chen Z and Zhang L: Intercellular mitochondrial transfer alleviates pyroptosis in dental pulp damage. Cell Prolif. 56(e13442)2023.PubMed/NCBI View Article : Google Scholar

222 

Kimura S, Yamashita M, Yamakami-Kimura M, Sato Y, Yamagata A, Kobashigawa Y, Inagaki F, Amada T, Hase K, Iwanaga T, et al: Distinct roles for the N- and C-terminal regions of M-Sec in plasma membrane deformation during tunneling nanotube formation. Sci Rep. 6(33548)2016.PubMed/NCBI View Article : Google Scholar

223 

Gao C, Dai Y, Spezza PA, Boasiako P, Tang A, Rasquinha G, Zhong H, Shao B, Liu Y, Shi PA, et al: Megakaryocytes transfer mitochondria to bone marrow mesenchymal stromal cells to lower platelet activation. J Clin Invest. 135(e189801)2025.PubMed/NCBI View Article : Google Scholar

224 

Irwin RM, Thomas MA, Fahey MJ, Mayán MD, Smyth JW and Delco ML: Connexin 43 regulates intercellular mitochondrial transfer from human mesenchymal stromal cells to chondrocytes. Stem Cell Res Ther. 15(359)2024.PubMed/NCBI View Article : Google Scholar

225 

Huang T, Zhang T, Jiang X, Li A, Su Y, Bian Q, Wu H, Lin R, Li N, Cao H, et al: Iron oxide nanoparticles augment the intercellular mitochondrial transfer-mediated therapy. Sci Adv. 7(eabj0534)2021.PubMed/NCBI View Article : Google Scholar

226 

Qiao X, Huang N, Meng W, Liu Y, Li J, Li C, Wang W, Lai Y, Zhao Y, Ma Z, et al: Beyond mitochondrial transfer, cell fusion rescues metabolic dysfunction and boosts malignancy in adenoid cystic carcinoma. Cell Rep. 43(114652)2024.PubMed/NCBI View Article : Google Scholar

227 

Powell AE, Anderson EC, Davies PS, Silk AD, Pelz C, Impey S and Wong MH: Fusion between intestinal epithelial cells and macrophages in a cancer context results in nuclear reprogramming. Cancer Res. 71:1497–1505. 2011.PubMed/NCBI View Article : Google Scholar

228 

Nahacka Z, Novak J, Zobalova R and Neuzil J: Miro proteins and their role in mitochondrial transfer in cancer and beyond. Front Cell Dev Biol. 10(937753)2022.PubMed/NCBI View Article : Google Scholar

229 

Zhang H, Yu X, Ye J, Li H, Hu J, Tan Y, Fang Y, Akbay E, Yu F, Weng C, et al: Systematic investigation of mitochondrial transfer between cancer cells and T cells at single-cell resolution. Cancer Cell. 41:1788–1802.e10. 2023.PubMed/NCBI View Article : Google Scholar

230 

Goliwas KF, Libring S, Berestesky E, Gholizadeh S, Schwager SC, Frost AR, Gaborski TR, Zhang J and Reinhart-King CA: Mitochondrial transfer from cancer-associated fibroblasts increases migration in aggressive breast cancer. J Cell Sci. 136(jcs260419)2023.PubMed/NCBI View Article : Google Scholar

231 

Xie Q, Zeng J, Zheng Y, Li T, Ren J, Chen K, Zhang Q, Xie R, Xu F and Zhu J: Mitochondrial transplantation attenuates cerebral ischemia-reperfusion injury: Possible involvement of mitochondrial component separation. Oxid Med Cell Longev. 2021(1006636)2021.PubMed/NCBI View Article : Google Scholar

232 

Peruzzotti-Jametti L, Bernstock JD, Willis CM, Manferrari G, Rogall R, Fernandez-Vizarra E, Williamson JC, Braga A, van den Bosch A, Leonardi T, et al: Neural stem cells traffic functional mitochondria via extracellular vesicles. PLoS Biol. 19(e3001166)2021.PubMed/NCBI View Article : Google Scholar

233 

Zheng D, Zhou H, Wang H, Zhu Y, Wu Y, Li Q, Li T and Liu L: Mesenchymal stem cell-derived microvesicles improve intestinal barrier function by restoring mitochondrial dynamic balance in sepsis rats. Stem Cell Res Ther. 12(299)2021.PubMed/NCBI View Article : Google Scholar

234 

Cowan DB, Yao R, Thedsanamoorthy JK, Zurakowski D, del Nido PJ and McCully JD: Transit and integration of extracellular mitochondria in human heart cells. Sci Rep. 7(17450)2017.PubMed/NCBI View Article : Google Scholar

235 

Fu A: Mitotherapy as a novel therapeutic strategy for mitochondrial diseases. Curr Mol Pharmacol. 13:41–49. 2020.PubMed/NCBI View Article : Google Scholar

236 

Chen J, Zhong J, Wang LL and Chen YY: Mitochondrial transfer in cardiovascular disease: From mechanisms to therapeutic implications. Front Cardiovasc Med. 8(771298)2021.PubMed/NCBI View Article : Google Scholar

237 

Wang ZH, Chen L, Li W, Chen L and Wang YP: Mitochondria transfer and transplantation in human health and diseases. Mitochondrion. 65:80–87. 2022.PubMed/NCBI View Article : Google Scholar

238 

Walker M, Levitt MR, Federico EM, Miralles FJ, Levy SH, Lynne Prijoles K, Winter A, Swicord JK and Sancak Y: Autologous mitochondrial transplant for acute cerebral ischemia: Phase 1 trial results and review. J Cereb Blood Flow Metab: Dec 4, 2024 (Epub ahead of print).

239 

Nakai R, Varnum S, Field RL, Shi H, Giwa R, Jia W, Krysa SJ, Cohen EF, Borcherding N, Saneto RP, et al: Mitochondria transfer-based therapies reduce the morbidity and mortality of Leigh syndrome. Nat Metab. 6:1886–1896. 2024.PubMed/NCBI View Article : Google Scholar

240 

Li C, Cheung MKH, Han S, Zhang Z, Chen L, Chen J, Zeng H and Qiu J: Mesenchymal stem cells and their mitochondrial transfer: A double-edged sword. Biosci Rep. 39(BSR20182417)2019.PubMed/NCBI View Article : Google Scholar

241 

Hosseinian S, Ali Pour P and Kheradvar A: Prospects of mitochondrial transplantation in clinical medicine: Aspirations and challenges. Mitochondrion. 65:33–44. 2022.PubMed/NCBI View Article : Google Scholar

242 

Caicedo A, Aponte PM, Cabrera F, Hidalgo C and Khoury M: Artificial mitochondria transfer: Current challenges, advances, and future applications. Stem Cells Int. 2017(7610414)2017.PubMed/NCBI View Article : Google Scholar

243 

Ishikawa K, Toyama-Sorimachi N, Nakada K, Morimoto M, Imanishi H, Yoshizaki M, Sasawatari S, Niikura M, Takenaga K, Yonekawa H and Hayashi J: The innate immune system in host mice targets cells with allogenic mitochondrial DNA. J Exp Med. 207:2297–2305. 2010.PubMed/NCBI View Article : Google Scholar

244 

Deuse T, Hu X, Agbor-Enoh S, Koch M, Spitzer MH, Gravina A, Alawi M, Marishta A, Peters B, Kosaloglu-Yalcin Z, et al: De novo mutations in mitochondrial DNA of iPSCs produce immunogenic neoepitopes in mice and humans. Nat Biotechnol. 37:1137–1144. 2019.PubMed/NCBI View Article : Google Scholar

245 

Klopstock T, Klopstock B and Prokisch H: Mitochondrial replacement approaches: Challenges for clinical implementation. Genome Med. 8(126)2016.PubMed/NCBI View Article : Google Scholar

246 

Yamada Y, Ito M, Arai M, Hibino M, Tsujioka T and Harashima H: Challenges in promoting mitochondrial transplantation therapy. Int J Mol Sci. 21(6365)2020.PubMed/NCBI View Article : Google Scholar

247 

Yuan J, Chen F, Jiang D, Xu Z, Zhang H and Jin ZB: ROCK inhibitor enhances mitochondrial transfer via tunneling nanotubes in retinal pigment epithelium. Theranostics. 14:5762–5777. 2024.PubMed/NCBI View Article : Google Scholar

248 

Chi A, Yang B, Dai H, Li X, Mo J, Gao Y, Chen Z, Feng X, Ma M, Li Y, et al: Stem Leydig cells support macrophage immunological homeostasis through mitochondrial transfer in mice. Nat Commun. 15(2120)2024.PubMed/NCBI View Article : Google Scholar

249 

Játiva S, Calle P, Torrico S, Muñoz Á, García M, Martinez I, Sola A and Hotter G: Mitochondrial transplantation enhances phagocytic function and decreases lipid accumulation in foam cell macrophages. Biomedicines. 10(329)2022.PubMed/NCBI View Article : Google Scholar

Related Articles

  • Abstract
  • View
  • Download
Copy and paste a formatted citation
Spandidos Publications style
Tran CM and Nguyen VT: Mesenchymal stem cells and the rescue of mitochondrial damage in different disease models (Review). World Acad Sci J 8: 4, 2026.
APA
Tran, C.M., & Nguyen, V.T. (2026). Mesenchymal stem cells and the rescue of mitochondrial damage in different disease models (Review). World Academy of Sciences Journal, 8, 4. https://doi.org/10.3892/wasj.2025.419
MLA
Tran, C. M., Nguyen, V. T."Mesenchymal stem cells and the rescue of mitochondrial damage in different disease models (Review)". World Academy of Sciences Journal 8.1 (2026): 4.
Chicago
Tran, C. M., Nguyen, V. T."Mesenchymal stem cells and the rescue of mitochondrial damage in different disease models (Review)". World Academy of Sciences Journal 8, no. 1 (2026): 4. https://doi.org/10.3892/wasj.2025.419
Copy and paste a formatted citation
x
Spandidos Publications style
Tran CM and Nguyen VT: Mesenchymal stem cells and the rescue of mitochondrial damage in different disease models (Review). World Acad Sci J 8: 4, 2026.
APA
Tran, C.M., & Nguyen, V.T. (2026). Mesenchymal stem cells and the rescue of mitochondrial damage in different disease models (Review). World Academy of Sciences Journal, 8, 4. https://doi.org/10.3892/wasj.2025.419
MLA
Tran, C. M., Nguyen, V. T."Mesenchymal stem cells and the rescue of mitochondrial damage in different disease models (Review)". World Academy of Sciences Journal 8.1 (2026): 4.
Chicago
Tran, C. M., Nguyen, V. T."Mesenchymal stem cells and the rescue of mitochondrial damage in different disease models (Review)". World Academy of Sciences Journal 8, no. 1 (2026): 4. https://doi.org/10.3892/wasj.2025.419
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team