Skip to main content

Mitochondrial transplantation as a novel therapeutic strategy for cardiovascular diseases

Abstract

Cardiovascular disease (CVD) is the leading cause of noncommunicable disease-related death worldwide, and effective therapeutic strategies against CVD are urgently needed. Mitochondria dysfunction involves in the onset and development of CVD. Nowadays, mitochondrial transplantation, an alternative treatment aimed at increasing mitochondrial number and improving mitochondrial function, has been emerged with great therapeutic potential. Substantial evidence indicates that mitochondrial transplantation improves cardiac function and outcomes in patients with CVD. Therefore, mitochondrial transplantation has profound implications in the prevention and treatment of CVD. Here, we review the mitochondrial abnormalities that occur in CVD and summarize the therapeutic strategies of mitochondrial transplantation for CVD.

Background

Cardiovascular diseases (CVD) are the leading cause of non-communicable disease-related deaths worldwide [1, 2]. With the gradually increased burden, CVD have become a major public health problem [2]. The diversity of risk factors, the complexity of pathological mechanisms, and the verity of comorbidities make the treatment of CVD even more challenging. Effective therapeutic strategies against CVD are urgently addressed.

Mitochondria play indispensable rolese in cardiovascular system. Mitochondrion is an energy center of the cardiomyocyte by constantly providing ATP. As one of the most complex and critical organelles in eukaryotic cells, mitochondria play an important role in cell signal transduction, redox balance, biotransformation of amino acids and lipids, calcium homeostasis, apoptosis and programmed cell death [3, 4] Intracellular energy balance is important for cardiomyocytes survival. Cardiomyocytes are one of the cell types with the highest content of mitochondria, and are highly dependent on mitochondrial oxidative phosphorylation to produce ATP. Mitochondria adapt quickly to changing environments to maintain metabolic homeostasis [5, 6]. Mitochondria dysfunction involves in the onset and development of CVD.

Nowadays, mitochondrial transplantation, an alternative treatment aimed at increasing mitochondrial number and improving mitochondrial function, has been emerged with great therapeutic potential [7]. Since protein components in mitochondria act as network hubs in multiple biological pathways are often affected simultaneously under pathological conditions, mitochondrial transplantation offers unique advantages over traditional pharmacological treatments targeting a single molecule [4]. Intracellular mitochondrial movement promotes the connection and formation of a dynamic mitochondrial network. In addition, the intercellular transfer of mitochondria was observed both in vitro and in vivo under physiological and pathophysiological conditions [3]. Since the dynamics and transferable capacity of mitochondria have been uncovered gradually, the therapeutic potential of mitochondrial transplantation for CVD as a rapidly developing field has attracted great attention. Substantial evidence indicates that mitochondrial transplantation improves cardiac function and outcomes in patients with CVD, and mitochondrial transplantation has profound implications in the prevention and treatment of CVD. Here we review the mitochondrial abnormalities that occur in CVD and summarize the therapeutic strategies of mitochondrial transplantation for CVD.

The role of mitochondria in cardiovascular system

Mitochondria, which are widely regarded as the “energy hub” of cells, play major roles in maintain metabolism and intracellular Ca2+ homeostasis, regulation of inflammatory reactions and molecular signaling.

Key signal pathways in mitochondria

AMP-activated protein kinase (AMPK) pathway is one of the mitochondria specific signaling pathways, which is activated when the adenosine monophosphate (AMP)/ATP ratio increases. AMPK has been implicated in the energy balance of key enzymes and regulatory nodes in various metabolic pathways, such as lipid and glucose metabolism, mitochondrial dynamics, autophagy, and protein synthesis [8]. In addition, the ratio of nicotinamide adenine dinucleotide (NAD) to NADH can be used as another indicator of mitochondrial metabolism, perceiving and transmitting the metabolic state of mitochondria. Act as cofactors for many metabolic reactions, NAD levels vary with metabolic activity. At the same time, it is also a family of proteases such as CD38, sirtuins protein (1–7) deacetylase and deacylase [9]. Reactive oxygen species (ROS) is a toxic by-product of mitochondrial dysfunction. The decrease of NAD level stably mediated by hypoxia inducible factor-1α is harmful to mitochondrial function in aged mice [10].

The main process of mitochondrial metabolism is that sugars, lipids, and proteins are oxidized through tricarboxylic acid cycle and oxidative phosphorylation, which involves a series of enzymatic reactions to provide energy for cardiomyocytes [11]. Under physiological conditions, compared with glycolysis, oxidative phosphorylation is more effective way to produce adenosine triphosphate (ATP). Intracellular calcium homeostasis is disrupted under stress, reducing oxidative phosphorylation efficiency and ATP production, producing excessive ROS, which further causes mitochondrial dysfunction and ultimately leads to the development of CVD (Fig. 1).

Fig. 1
figure 1

Key signal pathways of mitochondrial metabolism. A Major mitochondrial metabolites and cellular signaling pathways. B Main factors and pathways involved in mitochondrial metabolism under physiological conditions of cardiovascular system

Mitochondria dysfunction and CVD

Mitochondrial dysfunction leads to increased ROS, energy stress, and cell death, which are closely related to the onset and development of CVD [12].

Mitochondria abnormalities range from structure to function has been suggested in CVD [13]. Abnormalities in mitochondrial size and distribution, the most common manifestations of structural abnormalities, have been found in various CVD such as myocardial infraction, ischemia/reperfusion (I/R) injury, pulmonary hypertension, diabetes [14]. The two processes of fusion and fission occur simultaneously and continuously, and the number, shape, and distribution of mitochondria all influence the respiratory function of mitochondria [15, 16]. Ultrastructural analysis shows a significant increase in the number and size of mitochondria, resulting in the enlargement of cardiomyocytes and sarcomere disorders hinder contractile activity in mitochondrial heart disease [17].

The mitochondrial respiratory chain is also a major source of intracellular ROS while providing energy. Conversely, ROS can also affect the activities of respiratory chain complexes III and IV [15]. Mitochondrial ROS burst contributes to cardiomyocyte death, endothelial dysfunction and vascular occlusion in myocardial I/R injury [18, 19]. ROS causes mitochondrial Ca2+ overload, which in turn further increases ROS levels, this feedback loop maintains a Ca2+overload state with increased ROS production [20].

The nonspecific high conductance channels of mitochondrial permeability transition pore (mPTP) mediate the transition of mitochondrial permeability. Opening of mPTP leads to collapse of mitochondrial membrane potential, resulting in uncoupling of oxidative phosphorylation and ATP consumption [6, 21]. Related studies have shown that changes in mitochondrial permeability are associated with heart failure, hemorrhagic shock, hypertension, cardiomyopathy and cardiac I/R injury [14, 15, 22, 23]. In ischemic heart, Oxygen deprivation decrease oxidative phosphorylation in mitochondria, aerobic glycolysis becomes the primary mode of energy supply. Mitochondrial Ca2+ overload occurs along with increased ROS production and mPTP opening in ischemic cardiomyocytes [24].

Mitophagy is a defense behavior to restore mitochondrial components and selectively removes the accumulation of abnormal mitochondria, ensure energy supply and maintain cellular homeostasis [25]. Mitophagy abnormalities are closely related to a variety of diseases, especially CVD, including sepsis-induced myocardial dysfunction [21], myocardial I/R injury [15], cardiomyopathy [25], heart failure, atherosclerosis, myocardial infarction and hypertrophy and diabetic cardiomyopathy [26].

Mitochondrial transfer between cells and spontaneous mitochondrial transfer

Intercellular mitochondrial transfer is a form of intercellular interaction [27]. Mitochondrion derived proteins were initially discovered in exocrine proteomics, and subsequent studies further suggest that the secretion of damaged mitochondria by vesicles may be a mitochondrial quality control mechanism. Importantly, the transfer of healthy mitochondria from mesenchymal stem cells to target organs.

In 2006, Spees et al. demonstrated that wild mitochondria from bone marrow stem cells can be transferred to parenchymal cells displaying mitochondrial dysfunction to increase the aerobic respiration capacity of recipient mitochondria [28]. Although some studies have shown that exogenous mitochondria released from donor cells due to the delivery of autophagy may be degraded or integrated by recipient cells, the biological role of this interaction remains controversial [29, 30]. Accumulating evidence suggests that intercellular mitochondrial transfer occurs spontaneously in the cardiovascular system to maintain tissue homeostasis and development [7, 31]. In addition to bidirectional mitochondrial transfer between cardiomyocytes and fibroblasts [7], cardiomyocytes, vascular smooth muscle cells, and endothelial cells can act as donors or receptors during mitochondrial transfer [4].

Intercellular mitochondrial transfer has been demonstrated, but how exogenous mitochondria are internalized and integrated into the mitochondrial network of recipient cells is a key question in mitochondrial transplantation studies. Masuzawa et al. detected that mitochondria did not co-locate with any lysosomal or autophagosomal, indicating the internalization of exogenous mitochondria [32]. Later studies further revealed that mitochondrial internalization was actin-dependent in cardiomyocytes [33]. Many lines of evidence have demonstrated that the dynamic features of mitochondria are not restricted by cell boundaries, and the transfer of mitochondria between cells can integrate into the endogenous mitochondrial network of recipient cells to restore their biological functions [29, 34].

Strategy and mechanism of mitochondrial transfer in CVD

Methods for the treatment of CVD by mitochondrial transplantation include naked mitochondrial transplantation, cell-mediated approaches including tunnel nanotubes (TNTs), cell fusion, and extracellular vehicles (EVs) [3]. There are also gap junction channels-mediated synaptic complexes and dendritic networks that are used in other diseases but have not been reported in CVD [35, 36]. Since then, a number of studies have shown mitochondrial engraftment by TNTs and cell fusion, which are summarized in Table 1.

Table 1 Summary of mitochondrial transfer between MSCs and recipient cells of heart

Naked mitochondrial transplantation

Naked mitochondrial transplantation is a technique in which mitochondria isolated from healthy tissue or cells are injected directly into the myocardium or into coronary arteries or veins with the aid of microinjection techniques to improve cardiac function [37,38,39]. Interestingly, cell-free mitochondria were reported to be present in human blood, although they have no potential for oxidative phosphorylation and are unlikely to be functional in vivo [40]. This method has no risk of complications such as autoimmune reaction, microvascular occlusion, arrhythmia, and intramyocardial hematoma, but it is technically demanding and has low efficacy [4].

TNTs

Cell based mitochondrial transplantation can inject mesenchymal stem cells and progenitor cells into the myocardium. The main advantage of this technique is that mesenchymal stem cells can be derived from multiple tissues with high quality mitochondria [7]. TNTs is a transient filamentous membrane connected cell comprising cell membranes, F-actin, myosin, and tubulin, and studies have shown it to be a novel type of intercellular communication between neonatal rat cardiomyocytes and endothelial progenitor cells [41]. Mesenchymal stem cells (MSCs) rescued injured endothelial cells in an in vitro I/R model via tunnel structure mediated mitochondrial transfer [42]. In I/R injury and anthracycline induced cardiomyopathy, transplantation of MSCs mitochondria to endothelial cells enhances aerobic respiration to protect cardiomyocytes from oxidative stress, limit left ventricular dilatation and myocardial fibrosis [42,43,44]. As early as in 2005 it was shown that metabolic reprogramming can improve mitochondrial transfer in cardiac tissue.

The extension of TNTs required cell division control protein 42 homolog (CDC42), a cytoskeletal regulatory protein that controls the protrusion and growth [45]. M-Sec promoted nanotubes formation by enhancing the expression of epidermal growth factor receptor and activating the mammalian target of rapamycin (mTOR)/CDC42 signal transduction pathway, thereby increasing the expression of p53 [54]. In addition, mitochondrial Rho GTPase 1 (Miro1), a tail anchored outer mitochondrial membrane protein, mediated the movement of mitochondria along TNTs [51, 55]. Miro1 bound to the docking protein TRAK1/2 to recruit the motor protein kinesin and initiate microtubule based mitochondrial movement [56]. Ahmad et al. showed that Miro1 regulated the movement of mitochondria from MSCs to recipient cells, and its overexpression enhanced mitochondrial transfer and therapeutic effects [57]. An in vitro model of simulated I/R model, bone marrow MSCs rescued injured cardiomyocytes by TNTs mediated mitochondrial transfer, which increased mitochondrial membrane potential, enhanced mitochondrial function, and decreased cardiomyocytes apoptosis [52]. Mitochondria isolated from human MSCs via nasal administration in mice achieved rapid cellular internalization and restored brain structure and function [58]. However, nasal delivery has not been applied to CVD.

Cell fusion

Partial (temporary) or complete (permanent) cell fusion is the process of realizing the gradual sharing of organelle and cytoplasmic components by merging plasma membranes [59]. Acquistapace et al. studied the fusion of human bone marrow MSCs and human pluripotent adipose stem cells with partially mature mouse cardiomyocytes [49]. The co-culture of cardiomyocytes and human pluripotent adipose stem cells allowed the exchange of materials and mitochondria and facilitated cardiomyocytes reprogramming. Several studies have pointed out that MSCs rescued damaged cells through paracrine mechanisms, however, there are many safety concerns such as arrhythmia and microcirculation occlusion, which limited the application of this transplantation method [4].

EVs

EVs, including exocrine bodies, microvesicles and apoptotic bodies, are cell secreted nanoscale bilayer vesicles that stably reside in the extracellular fluid and participate as important messengers for cell communication, migration and angiogenesis [60]. Exocrine itself is related to the occurrence and development of CVD. Recent studies suggested that inhibition of inflammatory response and exocrine led to inhibition of apoptosis and oxidative stress in sepsis-induced cardiomyopathy. In mammalian cells microvesicle biogenesis was found to be controlled by cluster of differentiation 38 and cyclic adenosine diphosphate ribose signaling, while entry of exogenous mitochondria was regulated by integrin induced Src/Syk signaling [61]. Most importantly, EVs-mediated mitochondrial transfer is part of a fundamental cell biological process that occurs in multiple tissues, which makes the use of EVs for mitochondrial transfer as a new hotspot. Ibáñez and Villena-Gutierrez [62] proposed a novel strategy of mitochondrial transplantation, whereby EVs secreted the mitochondria from human-induced pluripotent stem cell-derived cardiomyocytes to damaged cardiomyocytes, attenuating myocardial infarction and I/R injury. Intracoronary or intravenous administration of mitochondria improved cardiac contractility and prevented left ventricular remodeling in I/R injury [63]. Brestoff et al. isolated mitochondria from adipocytes and transferred them into cardiomyocytes via small EVs to ameliorate I/R injury [64]. The complex composition of different EVs makes them unique for various diseases, and this method has high mitochondrial stability with no risk of microvascular occlusion and arrhythmias [4].

In addition, EVs are natural cell-derived drug carriers. EVs with a particle size larger than 200 nm, that is medium-to-large extracellular vesicles (m/lEVs) are naturally transported mitochondria during biogenesis to improve the survival rate of injured recipient tissues, and effectively protect mitochondrial integrity and activity, prolonging their life span in the blood, thus making EVs a promising carrier [65]. A recent study found that activation of PGC-1 α make m/lEVs carry a higher mitochondrial load, but it is often expensive and time-consuming [66]. Studies aimed at expanding the production capacity of EV and m/lEVs and increasing the mitochondrial load will expand the clinical application of mitochondrial therapeutics. We further compared the safety, effectiveness and flexibility of mitochondrial transplantation through co-incubation, microinjection, EVs mitochondrial delivery (Table 2).

Table 2 Different mitochondria transplantation methods in safety, efficacy and flexibility

Targeted mitochondrial transplantation is a challenging and promising task. For this reason, researchers are committed to finding ways to improve the efficiency of transplantation. Magneti or pressure driven techniques improved transplantation efficiency [66], and the application of optical tweezers achieved automated mitochondrial transplantation [67]. In addition, McCully and his group were granted a patent for an automated isolation of live mitochondrial device, which may be useful for the treatment of mitochondrial transplantation in CVD. Recently, a new delivery system was developed with the aid of a biocompatible polymer, that is, delivery by artificially encapsulating isolated mitochondria with a triphenylphosphine complex of dextran or a transactivates of transcribed glucan complexes [68, 69]. Mitochondria are almost completely contained in this delivery system, with high transfer efficiency and strong rescue ability.

Strategy and mechanism of mitochondrial transfer in CVD were summarized in Fig. 2. More feasible and possible strategic options will be available for mitochondrial transplantation in future studies.

Fig. 2
figure 2

Strategy and mechanism of mitochondrial transfer in CVD. A Naked mitochondria or mitochondria carrying MSCs. B Mitochondrial transplantation overview. CD Different delivery methods: mitochondria can either be transplanted through direct injection to the tissue of concern or through intravenous injection. E Cell-based mitochondrial transplantation. F Extracellular vehicles

Therapeutic practices of mitochondrial transplantation for CVD

Targeting important molecules involved in the mechanisms of mitochondrial dysfunction significantly improves cardiac function, implicating mitochondrial replacement therapy as an important approach for cardioprotection [70]. According to important breakthroughs in the study of mitochondrial transplantation (Fig. 3), more and more evidences prove that targeted mitochondrial transplantation is a promising strategy for the treatment of CVD.

Fig. 3
figure 3

Important breakthroughs in the study of mitochondrial transplantation

Myocardial I/R injury

The McCully group has been working on mitochondrial transplantation for the treatment of CVD. In their study, mitochondria isolated from the left ventricle of donor rabbit sham control or regional ischemia were injected directly into the ischemic site of the rabbit heart before reperfusion, it showed that mitochondria isolated from normal tissues significantly enhanced functional recovery and cell viability after ischemia [37]. Masuzawa et al. then successfully transplanted human mitochondria to a rabbit cardiac I/R model, achieving mitochondrial allotransplantation [32]. Their subsequent study transplanted exogenous mitochondria through the coronary vasculature and compared the outcomes of mitochondrial transplantation by two different routes of delivery. The concentration and distribution of mitochondria transported through the coronary arteries were higher than those achieved by direct mitochondrial injection in human fibroblasts [38]. Although both routes I/R injury, compared with safe, dense mitochondria injected in situ only near the administration site, vascular injection of mitochondria was less invasive and could be widely distributed over a short period of time. A study comparing the therapeutic effects of mitochondrial transplantation through coronary arteries before and during reperfusion, further demonstrated that coronary blood flow was temporarily restored and myocardial infarct size was reduced after mitochondrial transplantation [71]. Intracoronary implantation of mitochondria was further verified as a safe and effective method for the treatment of myocardial I/R injury, and ATP mediated vasodilation of inward potassium channels significantly increased coronary blood flow [72]. New implantation method improved the rate of mitochondrial uptake increasing the accuracy of mitochondrial distribution. However, serial multiple mitochondrial transplantation was not superior to single mitochondrial transplantation in a I/R injury model in Yorkshire swine, although both procedures significantly increased coronary blood flow and ejection fraction and reduced infarct size [73]. Mitochondria for transplantation can be derived from pectoralis major muscle cells as well as gastrocnemius myocytes. Isolation of allogeneic mitochondria from gastrocnemius muscles and injected them into coronary artery before harvesting donor heart showed prolonged cold ischemia time, enhanced graft function and reduced graft tissue injury [74]. These studies are summarized in Table 3.

Table 3 Research reports of mitochondrial transplantation for I/R injury

Cardiomyopathy

Diabetes cardiomyopathy is the most common cardiovascular complication of diabetes [23]. The insulin secretion of islet β cells is mainly related to the production of mitochondrial ATP stimulated by glucose. It has been found that human adipose MSCs transferred mitochondria to human islet β cells under symbiotic conditions to enhance the bioenergy and insulin secretion capacity of damaged β cells [75], implying mitochondrial transplantation as promising for the treatment of diabetes. Doulamis IP et al. also demonstrated that mitochondrial transplantation mitochondrial transplantation improves diabetic myocardial function [76]. These studies suggest that mitochondrial transplantation is a promising therapeutic strategy to reduce cardiac damage in type 2 diabetes.

It also showed that mitochondrial transplantation was more effective in treating sepsis-induced cardiomyopathy than simply improving mitochondrial function [22]. Recently, Mokhtari B et al. have demonstrated that mitochondrial transplantation improved the mitochondrial function, biogenesis and kinetics related to SIRT-1/PGC-1 α network to prevent myocardial dysfunction induced by sepsis [77]. Anthracycline induced cardiomyopathy is a dose-dependent progressive myocardial injury, after intramyocardial injection mitochondria derived from human induced pluripotent stem cell derived MSCs to neonatal mouse cardiomyopathy model induced by anthracycline, both mitochondrial respiration and cardiomyocyte viability were significantly enhanced [51]. These results provide evidence that mitochondrial transplantation improved anthracycline-induced cardiomyopathy.

Mitochondrial cardiomyopathies are structural or functional abnormalities of the myocardium caused by defects in nuclear DNA or mtDNA genes and are usually characterized by hypertrophic cardiomyopathy, dilated cardiomyopathy, and cardiac conduction defects [80]. Potential treatments are to transplant or deliver entirely new mitochondria from healthy cells to diseased cells to improve outcomes in patients with acquired or congenital mitochondrial defects due to mitochondrial mutations [44]. Recently, Park et al. proposed that mitochondrial transplantation was a valuable strategy for the treatment of a variety of mitochondrial diseases [81], but further studies are needed to prove its therapeutic efficacy.

Myocardial infarction and heart failure

Injection of human pluripotent adipocyte derived mitochondria into cardiomyocytes and endothelial cells surrounding the infarct zone increased heme oxygenase-1 expression and mitochondrial biogenesis [82]. MSCs-based mitochondrial transplantation opens a new avenue for the use of mitochondrial transplantation in the treatment of myocardial infarction.

A key pathophysiological mechanism of heart failure is defective myocardial mitochondrial function. In end-stage myocardial failure, the activity of citrate synthase of complex I of the mitochondrial respiratory chain was reduced by 28%, therefore, mitochondrial transplantation holds extensive promise in the treatment of heart failure. Studies showed that autologous mitochondrial transplantation from rat skeletal muscle cells to cardiomyocytes improved cellular respiration and energy production in a short time [44]. Injection autologous calf muscle mitochondria into the right ventricular free wall in a porcine model of right ventricular hypertrophy/failure prolonged the physiological adaptation of the right ventricle under pressure load and maintained contractile ability by reducing cardiomyocytes apoptosis [83]. In doxorubicin induced heart failure, transfer of M2-like macrophages reduced cardiac fibrosis and cardiomyocyte apoptosis to improve cardiac function, which may be related to mitochondrial transfer, moreover, co-culture of M2 macrophages in vitro translocated mitochondria to cardiomyocytes and promoted repair of myocardial injury [84]. Transplantation of mitochondria with varying metabolic status, showing that metabolically matched mitochondria restored mitochondrial membrane potential and protected against doxorubicin-induced heart failure [85].

Pulmonary hypertension

Pulmonary hypertension, a fatal progressive vascular disease, is caused by an increase in mean pulmonary artery pressure and right ventricular afterload due to pulmonary arteriolar occlusion, resulting in right ventricular hypertrophy and failure [86]. Two recent studies and other literature found that mitochondrial transplantation did not change the survival rate of animals [87, 88]. Mitochondrial transplantation in the experimental model of pulmonary hypertension provided beneficial effects in reducing pulmonary artery smooth muscle cell proliferation and pulmonary vasoconstriction, reducing pulmonary vascular remodeling, and improving right ventricular function [86,87,88]. Transplantation of mitochondria extracted from rat femoral artery smooth muscle cells into rat pulmonary artery smooth muscle cells by intravenous administration inhibits pulmonary vasoconstriction induced by acute hypoxia and attenuates pulmonary vascular remodeling induced by chronic hypoxia [87]. After mitochondria were isolated from immature rat soleus muscle and intravenously injected into pulmonary hypertensive rats, right ventricular mass and wall thickness returned to normal, and serum B-type natriuretic peptide levels and ventricular diameter were reduced, demonstrating that mitochondrial transplantation increased lung tissue ATP concentration, reversed pulmonary artery remodeling and improved right ventricular function [88].

Ischemic stroke

Ischemic stroke, a life-threatening disease caused by a sudden decrease in cerebral blood flow followed by a decrease in oxygen and glucose, is associated with mitochondrial dysfunction [89]. The transfer of mitochondria from endothelial progenitor cells to ischemic brain endothelial cells showed increased mitochondrial biogenesis and mitochondrial DNA copy number, restored ATP level after ischemic stroke [89]. Many studies have also shown that human MSCs reduce inflammation and promote vascular growth and neurite outgrowth as well as functional recovery [90]. Interestingly, MSCs based mitochondrial transplantation is a novel treatment for ischemic stroke by promoting the bioenergetic profile of neuron and neurite regeneration, enhancing angiogenesis, reducing infarct volume, and improving functional recovery in cerebral ischemic rats [90].

In view of the fact that (i) the source of mitochondria used in most studies is autologous mitochondria and (ii) mitochondrial haplotypes may have a great impact on genome expression, which is related to mitochondrial-nuclear incompatibility and the spread of harmful mutations, these technical challenges and ethical issues greatly limit the clinical use of mitochondrial transplantation [4, 44]. According to the current literature, mitochondrial transplantation has only studied the problems related to ischemic heart injury in clinical treatment.

In the first clinical research on mitochondrial transplantation, autologous mitochondria from non-ischemic skeletal muscle were isolated and pericardial injection was given to pediatric patients with I/R injury supported by extracorporeal membrane oxygenation (ECMO), it showed that mitochondrial therapy did not cause inflammation or rejection but instead improved ventricular function, while the conclusion was statistically significant due to the limited sample size [91]. In addition, mitochondrial transplantation resulted in significantly shorter ventricular strain and adverse cardiovascular events and fewer adverse cardiovascular events. This suggests the promise of mitochondrial transplantation in the treatment of cardiogenic shock in pediatric patients with I/R injury [38]. In 2021, the McCully team used autologous mitochondrial transplantation again in the treatment of refractory cardiogenic shock in children [92]. Autologous mitochondrial transplantation in ECMO patients with cardiogenic shock favored successful isolation of ECMO with enhanced ventricular strain, further reflecting the great potential of mitochondrial transplantation in clinical therapy.

Opportunities and challenges

The advantages of mitochondrial transplantation

Intercellular mitochondrial transfer plays an important role under both physiological and pathological conditions. It occurs spontaneously under physiological conditions and regulates the development of the cardiovascular system by promoting the proliferation and differentiation of stem cells into cardiomyocyte through progenitor cells reprogramming [4, 7]. Intercellular mitochondrial transfer contributes to the release of dysfunctional mitochondria under pathological conditions. Intercellular mitochondrial transmission can also be sensed by other cells, for example, the release of dysfunctional mitochondria in I/R injury was sensed by MSCs to enhance mitochondrial biogenesis as a negative feedback mechanism [53]. Intercellular mitochondrial transfer also rescued cellular injury by taking up functional mitochondria, thereby improving mitochondrial biogenesis [65].

A great advantage of mitochondrial transplantation over traditional mitochondria targeted drug delivery therapies lies in: (1) Achieve better therapeutic effects on multiple targets (2) Etiological treatments which are equivalent to gene level (3) More benefits from a single injection (4) The switch of cell-based therapy to cell-free therapy will greatly improve the convenience and cost of mitochondrial transplantation. At the same time, controversy still exists on mitochondrial transplantation technology, and there is still a long way to go to achieve its widespread clinical application.

Challenges and future prospective

In recent years, with the increasing popularity of mitochondrial transplantation, controversy over this treatment method has also emerged, mainly included in the following aspects.

Firstly, skeptics point out that McCully’s team did not rule out the possibility that mitochondria might be destroyed under injection conditions [93]. The concentration of calcium ions in the extracellular fluid is much higher than that in the intracellular fluid, whereas mitochondria are highly permeable to calcium ions, and the opening of the mPTP under high calcium conditions leads to mitochondrial inner membrane barrier disruption, mitochondrial permeability edema, inner membrane rupture, and even cell death. Although some earlier studies found mitochondria to be damaged in high calcium environments, this does not mean that they cannot survive at all. McCully team also discovered free mitochondria in the blood, however its activity may be normal or impaired [40, 94].

In addition, McCully’s group found that mitochondrial transplantation during reperfusion can improve cardiac function in as little as a few minutes [32], whereas mitochondria need to pass the endothelial barrier to integrate in the vicinity of cardiomyocytes, a process that takes several hours. And without a cytoplasmic environment, the conversion of glucose and fatty acids into pyruvate and fatty acyl coenzymes does not reveal how mitochondria produce ATP and complete cell contraction [93], this is also a very important aspect that needs to be clarified.

Furthermore, although McCully and colleagues found that mitochondrial transformation is feasible in cultured cells, whether mitochondria survive or not and are taken up by cardiomyocytes has not been clearly demonstrated, they entered cardiomyocytes but in low numbers [38], so the capacity to provide ATP might be limited. And studies showed that direct administration of exogenous ATP cannot significantly alleviate myocardial injury [37], in order to ensure the effect of mitochondrial transplantation, there must be sufficient mitochondria through the cell membrane to promote host cells to produce ATP. In fact, the immunogenicity of mitochondria is also closely related to transplantation efficiency. How to ensure that exogenous mitochondria are not rejected by individuals or phagocytosed by immune responses is also one of the very important issues.

It has become a consensus that mitochondria can be transferred between cells. Significant cellular endocytosis also appears from indirect or direct co incubation of isolated mitochondria with cells in in vitro experiments, but the mechanism of internalization after contact with tissue cells is not fully understood. What’s more, there is no definitive research evidence and conclusion as to whether there are differences in efficiency and efficacy when delivered in different ways.

Certainly, the timing and conditions of mitochondrial transplantation as a therapeutic approach can be further explored. The first step of mitochondrial transfer technically is the isolation of mitochondria, ensuring complete separation and good storage of mitochondria. Meanwhile, the core issue in mitochondrial transplantation is how to target and deliver mitochondria to specific tissues or organs. Therefore, future studies should focus on developing carriers for specific cellular delivery to improve the efficiency of mitochondrial internalization.

Finally, the effects of mitochondrial transplantation need to be further validated and evaluated in vivo. Delivery methods should be optimized according to the cell or tissue type and put into clinical practice.

Summary

Mitochondria participate in a series of important cellular processes and are the metabolic center and signal platform of cells. Mitochondrial dysfunction is one of the major pathological features of CVD. A number of preclinical trials have demonstrated significant advantages of mitochondrial transplantation over traditional approaches to improve mitochondrial function in CVD treatment. Although the study of mitochondrial transplantation is in its infancy and many issues remain to be unanswered, there is no doubt that this new technology will have a promising future.

Availability of data and materials

Not applicable.

Abbreviations

AMP:

Adenosine monophosphate

AMPK:

AMP-activated protein kinase

ATP:

Adenosine triphosphate

MSCs:

Mesenchymal stromal cells

CDC42:

Cell division control protein 42 homolog

CVD:

Cardiovascular disease

ECMO:

Extracorporeal membrane oxygenation

EVs:

Extracellular vesicles

hBM-MSCs:

Human bone marrow mesenchymal stromal cells

hMADS:

Human multipotent adipose-derived stem cells

iPSC-MSCs:

Pluripotent stem cell-derived mesenchymal stem cells

I/R:

Ischemia reperfusion

Miro1:

Mitochondrial Rho GTPase 1

mPTP:

Mitochondrial permeability transition pore

MSCs:

Mesenchymal stem cells

mtDNA:

Mitochondrial deoxyribonucleic acid

mTOR:

Mammalian target of rapamycin

NAD:

Nicotinamide adenine dinucleotide

ROS:

Reactive oxygen species

TNTs:

Tunnelling nanotubes

References

  1. Li Z, Wang L, Ren Y, Huang Y, Liu W, Lv Z, et al. Arginase: shedding light on the mechanisms and opportunities in cardiovascular diseases. Cell Death Discov. 2022. https://doi.org/10.1038/s41420-022-01200-4.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Tsao CW, Aday AW, Almarzooq ZI, Alonso A, Beaton AZ, Bittencourt MS, et al. Heart disease and stroke statistics-2022 update: a report from the American Heart Association. Circulation. 2022. https://doi.org/10.1161/cir.0000000000001052.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Liu Z, Sun Y, Qi Z, Cao L, Ding S. Mitochondrial transfer/transplantation: an emerging therapeutic approach for multiple diseases. Cell Biosci. 2022. https://doi.org/10.1186/s13578-022-00805-7.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Chen J, Zhong J, Wang LL, Chen YY. Mitochondrial transfer in cardiovascular disease: from mechanisms to therapeutic implications. Front Cardiovasc Med. 2021. https://doi.org/10.3389/fcvm.2021.771298.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Huang T, Zhang T, Gao J. Targeted mitochondrial delivery: a therapeutic new era for disease treatment. J Control Release. 2022. https://doi.org/10.1016/j.jconrel.2022.01.025.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Liu M, Lv J, Pan Z, Wang D, Zhao L, Guo X. Mitochondrial dysfunction in heart failure and its therapeutic implications. Front Cardiovasc Med. 2022. https://doi.org/10.3389/fcvm.2022.945142.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Tan YL, Eng SP, Hafez P, Abdul Karim N, Law JX, Ng MH. Mesenchymal stromal cell mitochondrial transfer as a cell rescue strategy in Regenerative Medicine: a review of evidence in preclinical models. Stem Cells Transl Med. 2022. https://doi.org/10.1093/stcltm/szac044.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Cai J, Chen X, Liu X, Li Z, Shi A, Tang X, et al. AMPK: the key to ischemia-reperfusion injury. J Cell Physiol. 2022. https://doi.org/10.1002/jcp.30875.

    Article  PubMed  Google Scholar 

  9. Chakraborty A, Minor KE, Nizami HL, Chiao YA, Lee CF. Harnessing NAD(+) metabolism as therapy for cardiometabolic diseases. Curr Heart Fail Rep. 2022. https://doi.org/10.1007/s11897-022-00550-5.

    Article  PubMed  Google Scholar 

  10. Osuru HP, Lavallee M, Thiele RH. Molecular and Cellular Response of the myocardium (H9C2 cells) towards hypoxia and HIF-1α inhibition. Front Cardiovasc Med. 2022. https://doi.org/10.3389/fcvm.2022.711421.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Huang X, Zeng Z, Li S, Xie Y, Tong X. The therapeutic strategies targeting mitochondrial metabolism in cardiovascular disease. Pharmaceutics. 2022. https://doi.org/10.3390/pharmaceutics14122760.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Sadoshima J, Kitsis RN, Editorial SS. Mitochondrial dysfunction and cardiovascular diseases. Front Cardiovasc Med. 2021. https://doi.org/10.3389/fcvm.2021.645986.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Pecoraro M, Pinto A, Popolo A. Mitochondria and cardiovascular disease: a brief account. Crit Rev Eukaryot Gene Expr. 2019. https://doi.org/10.1615/CritRevEukaryotGeneExpr.2019028579.

    Article  PubMed  Google Scholar 

  14. Manolis AS, Manolis AA, Manolis TA, Apostolaki NE, Apostolopoulos EJ, Melita H, et al. Mitochondrial dysfunction in cardiovascular disease: current status of translational research/clinical and therapeutic implications. Med Res Rev. 2021. https://doi.org/10.1002/med.21732.

    Article  PubMed  Google Scholar 

  15. Marin W, Marin D, Ao X, Liu Y. Mitochondria as a therapeutic target for cardiac ischemia–reperfusion injury (review). Int J Mol Med. 2021. https://doi.org/10.3892/ijmm.2020.4823.

    Article  PubMed  Google Scholar 

  16. Al Ojaimi M, Salah A, El-Hattab AW. Mitochondrial fission and fusion: molecular mechanisms, biological functions, and related disorders. Membranes. 2022. https://doi.org/10.3390/membranes12090893.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Stamerra CA, Di Giosia P, Giorgini P, Ferri C, Sukhorukov VN, Sahebkar A. Mitochondrial dysfunction and cardiovascular disease: pathophysiology and emerging therapies. Oxid Med Cell Longev. 2022. https://doi.org/10.1155/2022/9530007.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Kalogeris T, Baines CP, Krenz M, Korthuis RJ. Cell biology of ischemia/reperfusion injury. Int Rev Cell Mol Biol. 2012. https://doi.org/10.1016/b978-0-12-394309-5.00006-7.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Neri M, Riezzo I, Pascale N, Pomara C. Ischemia/reperfusion injury following acute myocardial infarction: a critical issue for clinicians and forensic pathologists. Mediators Inflamm. 2017. https://doi.org/10.1155/2017/7018393.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Penna C, Mancardi D, Rastaldo R, Pagliaro P. Cardioprotection: a radical view free radicals in pre and postconditioning. Biochim Biophys Acta. 2009. https://doi.org/10.1016/j.bbabio.2009.02.008.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Lin Y, Xu Y, Zhang Z. Sepsis-induced myocardial dysfunction (SIMD): the pathophysiological mechanisms and therapeutic strategies targeting Mitochondria. Inflammation. 2020. https://doi.org/10.1007/s10753-020-01233-w.

    Article  PubMed  Google Scholar 

  22. Mokhtari B, Yavari R, Badalzadeh R, Mahmoodpoor A. An overview on mitochondrial-based therapies in sepsis-related myocardial dysfunction: mitochondrial transplantation as a promising approach. Can J Infect Dis Med Microbiol. 2022. https://doi.org/10.1155/2022/3277274.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Dabravolski SA, Sadykhov NK, Kartuesov AG, Borisov EE, Sukhorukov VN, Orekhov AN. The role of mitochondrial abnormalities in diabetic cardiomyopathy. Int J Mol Sci. 2022. https://doi.org/10.3390/ijms23147863.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Shintani-Ishida K, Inui M, Yoshida K. Ischemia-reperfusion induces myocardial infarction through mitochondrial Ca2+ overload. J Mol Cell Cardiol. 2012. https://doi.org/10.1016/j.yjmcc.2012.05.012.

    Article  PubMed  Google Scholar 

  25. Sygitowicz G, Sitkiewicz D. Mitochondrial quality control: the role in cardiac injury. Front Biosci. 2022. https://doi.org/10.31083/j.fbl2703096.

    Article  Google Scholar 

  26. Li A, Gao M, Liu B, Qin Y, Chen L, Liu H, et al. Mitochondrial autophagy: molecular mechanisms and implications for cardiovascular disease. Cell Death Dis. 2022. https://doi.org/10.1038/s41419-022-04906-6.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Qin Y, Jiang X, Yang Q, Zhao J, Zhou Q, Zhou Y. The functions, methods, and mobility of mitochondrial transfer between cells. Front Oncol. 2021. https://doi.org/10.3389/fonc.2021.672781.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Spees JL, Olson SD, Whitney MJ, Prockop DJ. Mitochondrial transfer between cells can rescue aerobic respiration. Proc Natl Acad Sci USA. 2006. https://doi.org/10.1073/pnas.0510511103.

    Article  PubMed  PubMed Central  Google Scholar 

  29. D’Amato M, Morra F, Di Meo I. Mitochondrial transplantation in mitochondrial medicine: current challenges and future perspectives. Int J Mol Sci. 2023. https://doi.org/10.3390/ijms24031969.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Miliotis S, Nicolalde B, Ortega M, Yepez J, Caicedo A. Forms of extracellular mitochondria and their impact in health. Mitochondrion. 2019. https://doi.org/10.1016/j.mito.2019.02.002.

    Article  PubMed  Google Scholar 

  31. Liu D, Gao Y, Liu J, Huang Y, Yin J, Feng Y, et al. Intercellular mitochondrial transfer as a means of tissue revitalization. Signal Transduct Target Ther. 2021. https://doi.org/10.1038/s41392-020-00440-z.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Masuzawa A, Black KM, Pacak CA, Ericsson M, Barnett RJ, Drumm C, et al. Transplantation of autologously derived mitochondria protects the heart from ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol. 2013. https://doi.org/10.1152/ajpheart.00883.2012.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Pacak CA, Preble JM, Kondo H, Seibel P, Levitsky S, Del Nido PJ, et al. Actin-dependent mitochondrial internalization in cardiomyocytes: evidence for rescue of mitochondrial function. Biol Open. 2015. https://doi.org/10.1242/bio.201511478.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Valenti D, Vacca RA, Moro L. Mitochondria can cross cell boundaries: an overview of the biological relevance, pathophysiological implications and therapeutic perspectives of intercellular mitochondrial transfer. Int J Mol Sci. 2021. https://doi.org/10.3390/ijms22158312.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Picone P, Porcelli G, Bavisotto CC, Nuzzo D, Galizzi G, Biagio PLS, et al. Synaptosomes: new vesicles for neuronal mitochondrial transplantation. J Nanobiotechnol. 2021. https://doi.org/10.1186/s12951-020-00748-6.

    Article  Google Scholar 

  36. Gao J, Qin A, Liu D, Ruan R, Wang Q, Yuan J, et al. Endoplasmic reticulum mediates mitochondrial transfer within the osteocyte dendritic network. Sci Adv. 2019. https://doi.org/10.1126/sciadv.aaw7215.

    Article  PubMed  PubMed Central  Google Scholar 

  37. McCully JD, Cowan DB, Pacak CA, Toumpoulis IK, Dayalan H, Levitsky S. Injection of isolated mitochondria during early reperfusion for cardioprotection. Am J Physiol Heart Circ Physiol. 2009. https://doi.org/10.1152/ajpheart.00567.2008.

    Article  PubMed  Google Scholar 

  38. Cowan DB, Yao R, Akurathi V, Snay ER, Thedsanamoorthy JK, Zurakowski D, et al. Intracoronary delivery of mitochondria to the ischemic heart for cardioprotection. PLoS ONE. 2016. https://doi.org/10.1371/journal.pone.0160889.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Sun X, Gao R, Li W, Zhao Y, Yang H, Chen H, et al. Alda-1 treatment promotes the therapeutic effect of mitochondrial transplantation for myocardial ischemia-reperfusion injury. Bioact Mater. 2021. https://doi.org/10.1016/j.bioactmat.2020.12.024.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Stier A. Human blood contains circulating cell-free mitochondria, but are they really functional? Am J Physiol Endocrinol Metab. 2021. https://doi.org/10.1152/ajpendo.00054.2021.

    Article  PubMed  Google Scholar 

  41. Luchetti F, Carloni S, Nasoni MG, Reiter RJ, Balduini W. Melatonin, tunneling nanotubes, mesenchymal cells, and tissue regeneration. Neural Regen Res. 2023. https://doi.org/10.4103/1673-5374.353480.

    Article  PubMed  Google Scholar 

  42. Liu K, Ji K, Guo L, Wu W, Lu H, Shan P, et al. Mesenchymal stem cells rescue injured endothelial cells in an in vitro ischemia-reperfusion model via tunneling nanotube like structure-mediated mitochondrial transfer. Microvasc Res. 2014. https://doi.org/10.1016/j.mvr.2014.01.008.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Zhang L, Liu Q, Hu H, Zhao L, Zhu K. Progress in mesenchymal stem cell mitochondria transfer for the repair of tissue injury and treatment of disease. Biomed Pharmacother. 2022. https://doi.org/10.1016/j.biopha.2022.113482.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Ali Pour P, Hosseinian S, Kheradvar A. Mitochondrial transplantation in cardiomyocytes: foundation, methods, and outcomes. Am J Physiol Cell Physiol. 2021. https://doi.org/10.1152/ajpcell.00152.2021.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Walters HE, Cox LS. Intercellular transfer of Mitochondria between senescent cells through cytoskeleton-supported intercellular bridges requires mTOR and CDC42 signalling. Oxid Med Cell Longev. 2021. https://doi.org/10.1155/2021/6697861.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Koyanagi M, Brandes RP, Haendeler J, Zeiher AM, Dimmeler S. Cell-to-cell connection of endothelial progenitor cells with cardiac myocytes by nanotubes: a novel mechanism for cell fate changes? Circ Res. 2005. https://doi.org/10.1161/01.RES.0000168650.23479.0c.

    Article  PubMed  Google Scholar 

  47. Plotnikov EY, Khryapenkova TG, Vasileva AK, Marey MV, Galkina SI, Isaev NK, et al. Cell-to-cell cross-talk between mesenchymal stem cells and cardiomyocytes in co-culture. J Cell Mol Med. 2008. https://doi.org/10.1111/j.1582-4934.2007.00205.x.

    Article  PubMed  Google Scholar 

  48. Cselenyák A, Pankotai E, Horváth EM, Kiss L, 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. 2010. https://doi.org/10.1186/1471-2121-11-29.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Acquistapace A, Bru T, Lesault PF, Figeac F, Coudert AE, le Coz O, et al. Human mesenchymal stem cells reprogram adult cardiomyocytes toward a progenitor-like state through partial cell fusion and mitochondria transfer. Stem Cells. 2011. https://doi.org/10.1002/stem.632.

    Article  PubMed  Google Scholar 

  50. Vallabhaneni KC, Haller H, Dumler I. Vascular smooth muscle cells initiate proliferation of mesenchymal stem cells by mitochondrial transfer via tunneling nanotubes. Stem Cells Dev. 2012. https://doi.org/10.1089/scd.2011.0691.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Zhang Y, Yu Z, Jiang D, Liang X, Liao S, Zhang Z, et al. iPSC-MSCs with high intrinsic MIRO1 and sensitivity to TNF-α yield efficacious mitochondrial transfer to rescue anthracycline-induced cardiomyopathy. Stem Cell Rep. 2016. https://doi.org/10.1016/j.stemcr.2016.08.009.

    Article  PubMed  Google Scholar 

  52. Han H, Hu J, Yan Q, Zhu J, Zhu Z, Chen Y, et al. 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. 2016. https://doi.org/10.3892/mmr.2015.4726.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Mahrouf-Yorgov M, Augeul L, Da Silva CC, Jourdan M, Rigolet M, Manin S, et al. Mesenchymal stem cells sense mitochondria released from damaged cells as danger signals to activate their rescue properties. Cell Death Differ. 2017. https://doi.org/10.1038/cdd.2017.51.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Hase K, Kimura S, Takatsu H, Ohmae M, Kawano S, Kitamura H, et al. M-Sec promotes membrane nanotube formation by interacting with Ral and the exocyst complex. Nat Cell Biol. 2009. https://doi.org/10.1038/ncb1990.

    Article  PubMed  Google Scholar 

  55. Nahacka Z, Zobalova R, Dubisova M, Rohlena J, Neuzil J. Miro proteins connect mitochondrial function and intercellular transport. Crit Rev Biochem Mol Biol. 2021. https://doi.org/10.1080/10409238.2021.1925216.

    Article  PubMed  Google Scholar 

  56. Kassab S, Albalawi Z, Daghistani H, Kitmitto A. Mitochondrial arrest on the microtubule highway—a feature of heart failure and diabetic cardiomyopathy? Front Cardiovasc Med. 2021. https://doi.org/10.3389/fcvm.2021.689101.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Ahmad T, Mukherjee S, Pattnaik B, Kumar M, Singh S, Kumar M, et al. Miro1 regulates intercellular mitochondrial transport & enhances mesenchymal stem cell rescue efficacy. EMBO J. 2014. https://doi.org/10.1002/embj.201386030.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Alexander JF, Seua AV, Arroyo LD, Ray PR, Wangzhou A, Heiβ-Lückemann L, et al. Nasal administration of mitochondria reverses chemotherapy-induced cognitive deficits. Theranostics. 2021. https://doi.org/10.7150/thno.53474.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Gomzikova MO, James V, Rizvanov AA. Mitochondria donation by mesenchymal stem cells: current understanding and mitochondria transplantation strategies. Front Cell Dev Biol. 2021. https://doi.org/10.3389/fcell.2021.653322.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Zhang LJ, Chen F, Liang XR, Ponnusamy M, Qin H, Lin ZJ. Crosstalk among long non-coding RNA, tumor-associated macrophages and small extracellular vesicles in tumorigenesis and dissemination. Front Oncol. 2022. https://doi.org/10.3389/fonc.2022.1008856.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Hayakawa K, Esposito E, Wang X, Terasaki Y, Liu Y, Xing C, et al. Transfer of mitochondria from astrocytes to neurons after stroke. Nature. 2016. https://doi.org/10.1038/nature18928.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Ibáñez B, Villena-Gutierrez R. Cardiac mitochondrial transplantation: the force awakens. J Am Coll Cardiol. 2021. https://doi.org/10.1016/j.jacc.2021.01.017.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Ikeda G, Santoso MR, Tada Y, Li AM, Vaskova E, Jung JH, et al. Mitochondria-rich extracellular vesicles from autologous stem cell-derived cardiomyocytes restore energetics of ischemic myocardium. J Am Coll Cardiol. 2021. https://doi.org/10.1016/j.jacc.2020.12.060.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Brestoff JR, Wilen CB, Moley JR, Li Y, Zou W, Malvin NP, et al. Intercellular mitochondria transfer to macrophages regulates white adipose tissue homeostasis and is impaired in obesity. Cell Metab. 2021. https://doi.org/10.1016/j.cmet.2020.11.008.

    Article  PubMed  Google Scholar 

  65. Chen Y, Yang F, Chu Y, Yun Z, Yan Y, Jin J. Mitochondrial transplantation: opportunities and challenges in the treatment of obesity, diabetes, and nonalcoholic fatty liver disease. J Transl Med. 2022. https://doi.org/10.1186/s12967-022-03693-0.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Manickam DS. Delivery of mitochondria via extracellular vesicles—a new horizon in drug delivery. J Control Release. 2022. https://doi.org/10.1016/j.jconrel.2022.01.045.

    Article  PubMed  Google Scholar 

  67. Shakoor A, Wang B, Fan L, Kong L, Gao W, Sun J, et al. Automated optical tweezers manipulation to transfer mitochondria from fetal to adult MSCs to improve antiaging gene expressions. Small. 2021. https://doi.org/10.1002/smll.202103086.

    Article  PubMed  Google Scholar 

  68. Wu S, Zhang A, Li S, Chatterjee S, Qi R, Segura-Ibarra V, et al. Polymer functionalization of isolated mitochondria for cellular transplantation and metabolic phenotype alteration. Adv Sci. 2018. https://doi.org/10.1002/advs.201700530.

    Article  Google Scholar 

  69. Maeda H, Kami D, Maeda R, Murata Y, Jo JI, Kitani T, et al. TAT-dextran-mediated mitochondrial transfer enhances recovery from models of reperfusion injury in cultured cardiomyocytes. J Cell Mol Med. 2020. https://doi.org/10.1111/jcmm.15120.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Lee SE, Kang YC, Kim Y, Kim S, Yu SH, Park JH, et al. Preferred migration of mitochondria toward cells and tissues with mitochondrial damage. Int J Mol Sci. 2022. https://doi.org/10.3390/ijms232415734.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Kaza AK, Wamala I, Friehs I, Kuebler JD, Rathod RH, Berra I, et al. Myocardial rescue with autologous mitochondrial transplantation in a porcine model of ischemia/reperfusion. J Thorac Cardiovasc Surg. 2017. https://doi.org/10.1016/j.jtcvs.2016.10.077.

    Article  PubMed  Google Scholar 

  72. Shin B, Saeed MY, Esch JJ, Guariento A, Blitzer D, Moskowitzova K, et al. A novel biological strategy for myocardial protection by intracoronary delivery of mitochondria: safety and efficacy. JACC Basic Transl Sci. 2019. https://doi.org/10.1016/j.jacbts.2019.08.007.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Guariento A, Blitzer D, Doulamis I, Shin B, Moskowitzova K, Orfany A, et al. Preischemic autologous mitochondrial transplantation by intracoronary injection for myocardial protection. J Thorac Cardiovasc Surg. 2020. https://doi.org/10.1016/j.jtcvs.2019.06.111.

    Article  PubMed  Google Scholar 

  74. Moskowitzova K, Shin B, Liu K, Ramirez-Barbieri G, Guariento A, Blitzer D, et al. Mitochondrial transplantation prolongs cold ischemia time in murine heart transplantation. J Heart Lung Transplant. 2019. https://doi.org/10.1016/j.healun.2018.09.025.

    Article  PubMed  Google Scholar 

  75. Rackham CL, Hubber EL, Czajka A, Malik AN, King AJF, Jones PM. Optimizing beta cell function through mesenchymal stromal cell-mediated mitochondria transfer. Stem Cells. 2020. https://doi.org/10.1002/stem.3134.

    Article  PubMed  Google Scholar 

  76. Doulamis IP, Guariento A, Duignan T, Orfany A, Kido T, Zurakowski D, et al. Mitochondrial transplantation for myocardial protection in diabetic hearts. Eur J Cardiothorac Surg. 2020. https://doi.org/10.1093/ejcts/ezz326.

    Article  PubMed  Google Scholar 

  77. Mokhtari B, Hamidi M, Badalzadeh R, Mahmoodpoor A. Mitochondrial transplantation protects against sepsis-induced myocardial dysfunction by modulating mitochondrial biogenesis and fission/fusion and inflammatory response. Mol Biol Rep. 2023. https://doi.org/10.1007/s11033-022-08115-4.

    Article  PubMed  Google Scholar 

  78. Guariento A, Doulamis IP, Duignan T, Kido T, Regan WL, Saeed MY, et al. Mitochondrial transplantation for myocardial protection in ex-situ–perfused hearts donated after circulatory death. J Heart Lung Transplant. 2020. https://doi.org/10.1016/j.healun.2020.06.023.

    Article  PubMed  Google Scholar 

  79. Blitzer D, Guariento A, Doulamis IP, Shin B, Moskowitzova K, Barbieri GR, et al. Delayed transplantation of autologous mitochondria for cardioprotection in a porcine model. Ann Thorac Surg. 2020. https://doi.org/10.1016/j.athoracsur.2019.06.075.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Campbell T, Slone J, Huang T. Mitochondrial genome variants as a cause of mitochondrial cardiomyopathy. Cells. 2022. https://doi.org/10.3390/cells11182835.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Park A, Oh M, Lee SJ, Oh KJ, Lee EW, Lee SC, et al. Mitochondrial transplantation as a Novel Therapeutic Strategy for mitochondrial Diseases. Int J Mol Sci. 2021. https://doi.org/10.3390/ijms22094793.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Hu T, Wu Q, Yao Q, Jiang K, Yu J, Tang Q. Short-chain fatty acid metabolism and multiple effects on cardiovascular diseases. Ageing Res Rev. 2022. https://doi.org/10.1016/j.arr.2022.101706.

    Article  PubMed  Google Scholar 

  83. Weixler V, Lapusca R, Grangl G, Guariento A, Saeed MY, Cowan DB, et al. Autogenous mitochondria transplantation for treatment of right heart failure. J Thorac Cardiovasc Surg. 2021. https://doi.org/10.1016/j.jtcvs.2020.08.011.

    Article  PubMed  Google Scholar 

  84. Liu Y, Wu M, Zhong C, Xu B. M2-like macrophages transplantation protects against the doxorubicin-induced heart failure via mitochondrial transfer. Biomater Res. 2022. https://doi.org/10.1186/s40824-022-00260-y.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Zhang A, Liu Y, Pan J, Pontanari F, Chia-Hao Chang A, Wang H, et al. Delivery of mitochondria confers cardioprotection through mitochondria replenishment and metabolic compliance. Mol Ther. 2023. https://doi.org/10.1016/j.ymthe.2023.02.016.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Maarman GJ. Reviewing the suitability of mitochondrial transplantation as therapeutic approach for pulmonary hypertension in the era of personalized medicine. Am J Physiol Lung Cell Mol Physiol. 2022. https://doi.org/10.1152/ajplung.00484.2021.

    Article  PubMed  Google Scholar 

  87. Hsu CH, Roan JN, Fang SY, Chiu MH, Cheng TT, Huang CC, et al. Transplantation of viable mitochondria improves right ventricular performance and pulmonary artery remodeling in rats with pulmonary arterial hypertension. J Thorac Cardiovasc Surg. 2022. https://doi.org/10.1016/j.jtcvs.2020.08.014.

    Article  PubMed  Google Scholar 

  88. Zhu L, Zhang J, Zhou J, Lu Y, Huang S, Xiao R, et al. Mitochondrial transplantation attenuates hypoxic pulmonary hypertension. Oncotarget. 2016. https://doi.org/10.18632/oncotarget.10596.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Chen W, Huang J, Hu Y, Khoshnam SE, Sarkaki A. Mitochondrial transfer as a therapeutic strategy against ischemic stroke. Transl Stroke Res. 2020. https://doi.org/10.1007/s12975-020-00828-7.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Lu M, Guo J, Wu B, Zhou Y, Wu M, Farzaneh M, et al. Mesenchymal stem cell-mediated mitochondrial transfer: a therapeutic approach for ischemic stroke. Transl Stroke Res. 2021. https://doi.org/10.1007/s12975-020-00853-6.

    Article  PubMed  Google Scholar 

  91. Emani SM, Piekarski BL, Harrild D, Del Nido PJ, McCully JD. Autologous mitochondrial transplantation for dysfunction after ischemia-reperfusion injury. J Thorac Cardiovasc Surg. 2017. https://doi.org/10.1016/j.jtcvs.2017.02.018.

    Article  PubMed  Google Scholar 

  92. Guariento A, Piekarski BL, Doulamis IP, Blitzer D, Ferraro AM, Harrild DM, et al. Autologous mitochondrial transplantation for cardiogenic shock in pediatric patients following ischemia-reperfusion injury. J Thorac Cardiovasc Surg. 2021. https://doi.org/10.1016/j.jtcvs.2020.10.151.

    Article  PubMed  Google Scholar 

  93. Bertero E, Maack C, O’Rourke B. Mitochondrial transplantation in humans: “magical” cure or cause for concern? J Clin Invest. 2018. https://doi.org/10.1172/jci124944.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Al Amir Dache Z, Otandault A, Tanos R, Pastor B, Meddeb R, Sanchez C, et al. Blood contains circulating cell-free respiratory competent mitochondria. Faseb J. 2020. https://doi.org/10.1096/fj.201901917RR.

    Article  PubMed  Google Scholar 

Download references

Funding

This research was supported by funding from the National Natural Science Foundation of China (82070261, 82170251). National Key Research and Development Program of China (No.2021YFA1100501). The Youth Innovation Team of Shaanxi Universities. The Innovation Foundation for Graduate Students of Northwestern Polytechnical University (PF2023032). The Innovation Foundation for Doctor Dissertation of Northwestern Polytechnical University (CX2022068). The Seed Foundation of Innovation and Creation for Graduate Students in Northwestern Polytechnical University (CX202065).

Author information

Authors and Affiliations

Authors

Contributions

SMC drafted the original manuscript and revised the manuscript, JWH and MN contributed to specific sections, ZZH revised the manuscript, YL and MH conceptualized and provided overall supervision for the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Zihui Zhang, Lu Yu or Heng Ma.

Ethics declarations

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sun, M., Jiang, W., Mu, N. et al. Mitochondrial transplantation as a novel therapeutic strategy for cardiovascular diseases. J Transl Med 21, 347 (2023). https://doi.org/10.1186/s12967-023-04203-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12967-023-04203-6

Keywords