Next Article in Journal
Differential Responses of Antioxidant Enzymes and Lignin Metabolism in Susceptible and Resistant Sweetpotato Cultivars during Root-Knot Nematode Infection
Next Article in Special Issue
From Obesity-Induced Low-Grade Inflammation to Lipotoxicity and Mitochondrial Dysfunction: Altered Multi-Crosstalk between Adipose Tissue and Metabolically Active Organs
Previous Article in Journal
Application of a Cold-Pressing Treatment to Improve Virgin Olive Oil Production and the Antioxidant Phenolic Profile of Its by-Products
Previous Article in Special Issue
PGC-1α Is a Master Regulator of Mitochondrial Lifecycle and ROS Stress Response
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mitochondrial Fission as a Therapeutic Target for Metabolic Diseases: Insights into Antioxidant Strategies

1
Consortium for Health and Military Performance, Department of Military and Emergency Medicine, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD 20814, USA
2
Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
3
Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
4
Department of Pathology, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD 20814, USA
5
Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
*
Author to whom correspondence should be addressed.
Antioxidants 2023, 12(6), 1163; https://doi.org/10.3390/antiox12061163
Submission received: 5 May 2023 / Revised: 22 May 2023 / Accepted: 25 May 2023 / Published: 27 May 2023

Abstract

:
Mitochondrial fission is a crucial process in maintaining metabolic homeostasis in normal physiology and under conditions of stress. Its dysregulation has been associated with several metabolic diseases, including, but not limited to, obesity, type 2 diabetes (T2DM), and cardiovascular diseases. Reactive oxygen species (ROS) serve a vital role in the genesis of these conditions, and mitochondria are both the main sites of ROS production and the primary targets of ROS. In this review, we explore the physiological and pathological roles of mitochondrial fission, its regulation by dynamin-related protein 1 (Drp1), and the interplay between ROS and mitochondria in health and metabolic diseases. We also discuss the potential therapeutic strategies of targeting mitochondrial fission through antioxidant treatments for ROS-induced conditions, including the effects of lifestyle interventions, dietary supplements, and chemicals, such as mitochondrial division inhibitor-1 (Mdivi-1) and other mitochondrial fission inhibitors, as well as certain commonly used drugs for metabolic diseases. This review highlights the importance of understanding the role of mitochondrial fission in health and metabolic diseases, and the potential of targeting mitochondrial fission as a therapeutic approach to protecting against these conditions.

1. Introduction

Metabolic diseases are a significant public health concern worldwide due to their high prevalence and impact on individual and societal health [1,2]. Obesity, type 2 diabetes (T2DM), and cardiovascular diseases, among the most common metabolic disorders, are known to increase the risk of other chronic illnesses such as cancer, stroke, and hypertension [3,4,5]. These conditions arise from complex interactions among genetic, environmental, and lifestyle factors, which makes their prevention and treatment challenging [6,7]. The rising global burden of metabolic diseases underscores the need for effective interventions to improve metabolic health and reduce the incidence of related complications. Strategies such as lifestyle modifications, pharmacological interventions, and public health policies are vital in mitigating the impact of these diseases on individuals and populations worldwide.
One of the underlying mechanisms of metabolic diseases is oxidative stress, which is characterized by an imbalance between the production of reactive oxygen species (ROS) and the antioxidant defense system [8,9,10]. Mitochondria are critical in regulating oxidative stress, as mitochondrial electron transport chain (ETC) complexes are both the main sites of ROS production and the primary targets of ROS, and their dynamic nature plays an essential role in metabolic health [10,11,12,13,14]. Mitochondrial fission is a critical process that regulates the size, number, and function of mitochondria [15,16,17]. In this review, we will explore the physiological and pathological roles of mitochondrial fission and how it can be targeted for antioxidants in metabolic diseases.

2. Reactive Oxygen Species: Physiology, Pathology, and Therapeutic Potential

Reactive oxygen species (ROS) are a group of highly reactive molecules, including superoxide anions (O2•–), hydrogen peroxide (H2O2), and hydroxyl radicals (OH). In mammals, they are generated as natural byproducts of cellular metabolism [9,11,13,18]. ROS serve critical roles in cellular signaling and homeostasis, but excessive ROS production can lead to oxidative stress and cellular damage, which contribute to the pathogenesis of several metabolic diseases [9,10,11,19]. ROS can also be induced by exposure to environmental toxins [20], ultraviolet radiation [21], and chemical stressors [22]. Understanding the complex interplay between ROS and cellular homeostasis is essential for developing novel therapeutic strategies for metabolic diseases.

2.1. ROS Generation and Metabolism

ROS are generated by various cellular sources, including mitochondria, peroxisomes, and reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidases. Mitochondrial ETC complexes are the primary source of cellular ROS, with superoxide anion produced as a byproduct of oxidative phosphorylation (Figure 1) [11,12,13,14]. Peroxisomes are involved in both the production and breakdown of ROS. They contain various types of oxidases, which catalyze the transfer of electrons to molecular oxygen, leading to the production of ROS such as H2O2 and O2•– [23]. NADPH oxidases are membrane-bound enzymes, which produce ROS as defense response to various stimuli, including cytokines, growth factors, microbial pathogens, and metabolic stress [24,25,26]. Of note, NADPH oxidase 4 may locate mitochondria and mediate cross-talk between NADPH oxidase and mitochondria [25,27].
ROS contain unpaired electrons derived from partially reduced molecular oxygen (O2) and have a short lifespan. These molecules are continually produced, transformed, and eliminated in various cellular processes. Several defensive mechanisms and mediators of redox signaling in cells help manage ROS at physiological levels. These include: (1) superoxide dismutases (SODs), which convert O2•– into less reactive H2O2 and O2 [28]; (2) catalase, which further reduces H2O2 to water and O2 [29]; and (3) glutathione peroxidases, which eliminate H2O2 through the reducing power derived from glutathione [30]. In addition, other molecules serve a role in redox signaling and act as mediators of defense against ROS. These include peroxiredoxin, thioredoxin (TRX), and the glutathione/glutaredoxin systems [31,32,33]. Together, these defense mechanisms work to regulate the levels of ROS and prevent damage to cells.

2.2. Physiological ROS Signaling

ROS play a role in many physiological processes, including proliferation, differentiation, apoptosis, and immune defense [9]. In particular, ROS have been shown to be important in the regulation of cellular metabolism, including insulin signaling and glucose uptake [34,35]. In physiological conditions, low levels of ROS can activate several transcription factors, including nuclear factor kappa B (NF-κB), activator protein-1 (AP-1), and hypoxia-inducible factor-1α (HIF-1α), which regulate the expression of genes involved in inflammation, angiogenesis, and metabolism [19,36].
ROS also serve a role in the regulation of mitochondrial biogenesis through the cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) signaling pathway [37], and mitophagy, the process by which damaged mitochondria are removed from the cell [38]. In addition, ROS can modulate ion channels and transporters [39], which affect neuronal excitability and synaptic transmission [40], and regulate glucose-dependent insulin secretion by the β-cells [39].

2.3. The Role of ROS in Metabolic Diseases

ROS can promote oxidative stress, which may lead to deoxyribonucleic acid (DNA) damage, protein oxidation, and lipid peroxidation, and contribute to cellular dysfunction and disease progression (Figure 1) [9]. Excessive ROS production has been linked to the pathogenesis of several metabolic diseases, including obesity, T2DM, and cardiovascular disease [8,12,35]. The dysregulation of ROS production and homeostasis can contribute to the pathogenesis of metabolic diseases through multiple pathways, including insulin resistance, adipose tissue dysfunction (dyslipidemia), and endothelial dysfunction (Figure 1) [34,35,41,42]. The association of oxidative stress with mitochondrial dysfunction and their implications in metabolic diseases are discussed in the next section.
Insulin resistance, a hallmark of T2DM, is associated with increased ROS production and decreased antioxidant defense. ROS can impair insulin signaling by altering the activity of key enzymes involved in glucose metabolism, such as glycogen synthase and phosphatidylinositol-3-kinase (PI3K), and by inactivating key components of the insulin signaling pathway, including insulin receptor substrate (IRS) and Akt [43,44]. ROS can also activate proinflammatory kinases, such as c-Jun N-terminal kinase (JNK), pro-inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6), and inhibitor of kappa B kinase (IKK), which inhibit insulin signaling by phosphorylating IRS and impairing insulin sensitivity [45,46].
Several studies have shown that ROS also play a critical role in the regulation of adipogenesis, the process by which precursor cells differentiate into adipocytes (fat cells) [5,47,48]. ROS can promote adipogenesis by activating the key transcription factor peroxisome proliferator-activated receptor gamma (PPARγ), which regulates the expression of genes involved in lipid metabolism [49]. Increased adipogenesis can lead to the development of obesity, a major risk factor for T2DM and other metabolic diseases.
Increased ROS production can also cause damage to endothelial cells and impair endothelial function [8]. One of the mechanisms by which ROS contribute to endothelial dysfunction in metabolic disease is through activation of the renin–angiotensin system (RAS). Angiotensin II (Ang II), a key component of the RAS, can increase ROS production and promote inflammation in the endothelium. This leads to impaired nitric oxide (NO)-mediated vasodilation and increased vascular permeability, which contributes to endothelial dysfunction [50]. In addition, Ang II can promote the formation of advanced glycation end-products (AGEs), which are known to contribute to endothelial dysfunction in metabolic disease [51,52].

2.4. Therapeutic Potential of ROS

ROS perform critical roles in cellular signaling and homeostasis, but the dysregulation of ROS production can lead to oxidative stress and cellular damage, which may then contribute to the pathogenesis of several metabolic diseases. Understanding the complex interplay between ROS and cellular homeostasis is essential for the development of novel therapeutic strategies for metabolic diseases.
Several compounds have been studied for their potential therapeutic effects in ROS-related diseases. For example, the antioxidants vitamin C, vitamin E, and resveratrol have been shown to reduce oxidative stress and improve cellular function in various disease models [53,54]. Similarly, inhibitors of NAPDH oxidase (NOX) enzymes, such as apocynin and diphenyleneiodonium (DPI), have been shown to reduce ROS production and improve cellular function in several diseases, including cardiovascular diseases and neurodegenerative diseases [55]. Moreover, compounds that modulate ROS signaling, such as angiotensin-converting enzyme (ACE) inhibitors, have been shown to reduce inflammation and improve cellular function in several diseases [48,52]. The following section will examine the targeting of mitochondrial fission as a potential antioxidant treatment for metabolic diseases.

3. Mitochondrial Fission Mechanism and Implications for Metabolic Diseases

Mitochondrial fission is a highly regulated process by which mitochondria tubules divide into smaller fragments. This process is essential for mitochondrial quality control, as it allows for the removal of damaged or dysfunctional mitochondria via mitophagy [56,57,58]. Mitochondrial fission also serves critical roles in energy metabolism, calcium signaling, and apoptosis [13,17,56,57]. The dysregulation of mitochondrial fission has been implicated in the pathogenesis of several metabolic diseases, including obesity, diabetes, and cardiovascular disease [12,13,59]. Understanding the complex interplay between mitochondrial fission and metabolic homeostasis is essential for the development of novel therapeutic strategies for metabolic diseases.

3.1. Overview of Mitochondrial Membrane Structure and Dynamics

Mitochondria are double-membrane-bound organelles, with the inner mitochondrial membrane (IMM) containing four large complexes of the ETC, which pump protons across the membrane to generate an electrochemical gradient that is ultimately used by ATP synthase (also called Complex V) to produce ATP through oxidative phosphorylation (Figure 1) [12,13]. The overall morphology of mitochondrial networks is determined by the balance between fusion and fission events (collectively termed mitochondrial dynamics), which maintain their functional and structural integrity [12,13].
Mitochondrial fusion is facilitated by the outer mitochondrial membrane (OMM) proteins mitofusin 1 and 2 (MFN1 and 2) and the IMM protein optic atrophy 1 (OPA1) [60,61]. Mitochondrial fusion contributes to the distribution of genetic material and the maintenance of a functional mitochondrial network [62]. The molecular mechanisms of mitochondrial fusion have been reviewed extensively elsewhere [56,60,63,64]. Mitochondrial fission is regulated by the cytosolic dynamin-related protein 1 (Drp1) [15,65,66]. In this review, the authors focus on the mitochondrial fission mechanism and how it can be targeted for antioxidants in metabolic diseases.

3.2. Mitochondrial Fission Mediator Drp1 and Its Regulation

Drp1 is a 78 kDa cytosolic protein that belongs to the dynamin family of large GTPases (Figure 2A). This protein is composed of several domains, each of which has a specific function [12,67]. The N-terminal domain of Drp1 contains a GTPase domain, which is responsible for the GTP-binding and GTPase activity of the protein. The middle domain of Drp1 is the membrane-binding domain that interacts with OMM and is responsible for recruiting Drp1 to the site of fission. The GTPase effector domain (GED) of Drp1 is located at the C-terminus and is involved in the regulation of Drp1 activity. The GED domain interacts with the GTPase domain to regulate the hydrolysis of GTP, which is required for Drp1 function in mitochondrial fission. The variable domain (VD) is located between the middle and GED domains. This domain is not well-understood, but has been proposed to be involved in regulating Drp1 function through interactions with other proteins (Figure 2A).
Upon activation, Drp1 translocates to the OMM and assembles into oligomeric complexes that constrict the membrane to initiate fission (Figure 2B) [15,67,68]. Drp1 is recruited to the OMM by a variety of adaptor proteins, including mitochondrial fission 1 protein (Fis1), mitochondrial fission factor (Mff), and mitochondrial dynamics protein of 49 kDa (MiD49) and of 51 kDa (MiD51) [69,70]. These adaptor proteins contain domains that interact with Drp1 and help to localize it to the mitochondrial surface.
Ganglioside-induced differentiation-associated protein 1 (GDAP1) may promote the assembly and activation of Drp1 on the OMM [71]. Once recruited to the OMM, Drp1 assembles into higher-order structures that wrap around the mitochondrion, constricting it through a GTP hydrolysis-dependent mechanism until it eventually divides into two daughter mitochondria (Figure 2B) [15,68]. This process requires the coordinated action of other proteins, including the GTPase dynamin-2 and various accessory proteins that facilitate the membrane remodeling events required for fission to occur [66].
Drp1 is regulated by several post-translational modifications, including phosphorylation, ubiquitination, SUMOylation, and S-nitrosylation. Drp1 phosphorylation occurs mainly at two serine residues: Ser616 and Ser637. The phosphorylation of Drp1 at Ser616 by cyclin-dependent kinase CDK1 and PKA enhances its GTPase activity, whereas the phosphorylation of Drp1 at Ser637 by calcium/calmodulin-dependent protein kinase I (CaMKI) inhibits its GTPase activity and mitochondrial translocation, resulting in decreased mitochondrial fission. [72,73,74,75].
The ubiquitination of Drp1 by an E3 ubiquitin ligase such as Parkin promotes its proteasomal degradation, and hence, inhibits mitochondrial fission [76]. The deubiquitination of Drp1 by ubiquitin-specific protease 30 stabilizes Drp1 and enhances mitochondrial fission [77]. The SUMOylation of Drp1 in the GTPase domain and B domain of Drp1 involves the covalent attachment of a member of the SUMO (small ubiquitin-like modifier) family of proteins to lysine residues to increase its activity, whereas the deSUMOylation of Drp1 by the SUMO-specific peptidases SENP5 and SENP3 has been reported to decrease Drp1 activity and inhibit mitochondrial fission [78]. The S-nitrosylation of Drp1 at cysteine residues by NO inhibits Drp1 activity and mitochondrial fission [79,80].

3.3. Pathological Role of Mitochondrial Fission in Metabolic Diseases—The Interplay between Mitochondria and ROS

Mitochondria are dynamic organelles that constantly change their shape and size in response to energy demand and nutrient availability. During exercise or periods of starvation, mitochondria become elongated and more interconnected in skeletal muscle and other tissues, which allows them to be more efficient in producing ATP. This elongation is regulated by Drp1 Ser637 dephosphorylation and a reduction in the fission proteins Drp1 and Fis1, as well as activation of the key mitochondrial biogenesis inductors PGC-1α and upregulation of the levels of the fusion proteins MFN1/2 and OPA1 [81,82,83,84]. Conversely, nutrient excess, such as high glucose/lipid levels, excess dietary fatty acids, or high fructose, can lead to increased mitochondrial ROS production and mitochondrial fragmentation in many cell types, including hepatocytes and skeletal muscle cells (Figure 3) [41,85,86,87,88].
The activation of mitochondrial fission is regulated by several proteins, including Drp1 and its activators, such as receptor-interacting protein kinase 1 (RIPK1) and protein kinase Cδ (PKCδ). Under conditions of hyperglycemia and/or hyperlipidemia, Drp1 is phosphorylated by Rho-associated protein kinase 1 (ROCK1) and extracellular signal-regulated kinase 1/2 (ERK1/2), which leads to its translocation to the OMM and the initiation of fission [75,89]. This process is facilitated by an increase in intracellular calcium levels and a decrease in mitochondrial membrane potential, which activate PKCδ and RIPK1, respectively [75,89]. Increased mitochondrial fragmentation has been reported in various tissues in animal and human models of diabetes, obesity, and cardiovascular disease, including liver, muscle, kidney, blood vessel, and adipose tissue [41,90,91,92,93,94].
Mitochondrial fragmentation has been linked to an increase in ROS production (Figure 3), which leads to oxidative stress and cellular damage and contributes to metabolic disease progression (Figure 1) [12,13]. Mitochondria produce ROS as a byproduct of the ETC during oxidative phosphorylation, which is tightly regulated by several factors, including ETC complex activity and mitochondrial membrane potential (Figure 1) [13]. One proposed mechanism is that excessive mitochondrial fragmentation can induce disruption of the IMM and cristae structure, thus causing deformities in ETC complexes, which disrupt electron transfer and proton transport [95,96,97]. As a result, electron and proton leak occur, which leads to decreased ATP production and increased ROS production [12,13]. Additionally, ROS can activate Drp1, a protein responsible for mitochondrial fission, and lead to further mitochondrial fission and ROS production, thus creating a positive feedback loop [98,99]. Finally, the dysregulation of mitochondrial fission can lead to impaired mitophagy and the accumulation of damaged mitochondria, which can then contribute to oxidative stress and inflammation in metabolic diseases [17,56,57,58,64].
Mitochondria are the main target of ROS. They are more susceptible to ROS-induced damage compared to other organelles within cells, and this is due to several factors: First, the ETC, located on the IMM, is a major source of ROS production, which exposes mitochondria to high levels of ROS and makes them more susceptible to damage [9,11,12,13,99]. Second, mitochondria have limited antioxidant defenses compared to other organelles. Although they do contain some antioxidant enzymes (e.g., superoxide dismutase and glutathione peroxidase), these defenses are not as robust as those found in other cellular compartments [38,100]. Third, mitochondria contain their own DNA (mtDNA) that encodes 13 subunits of the ETC complex. mtDNA is more susceptible to oxidative damage than nuclear DNA because it lacks protective histones. As a result, mtDNA damage may lead to mutations and deletions that can impair mitochondrial function and contribute to oxidative stress and metabolic diseases [100,101]. Fourth, the lipids, particularly cardiolipin that is present almost exclusively in the IMM, are susceptible to oxidative damage, which can lead to lipid peroxidation and the disruption of mitochondrial membrane structure and function [100,102,103,104]. Finally, ROS can trigger the mitochondrial permeability transition pore, the release of apoptogenic factors, and the activation of cell death pathways, such as apoptosis, necrosis, or autophagy-mediated cell death [12,17,105].
Several studies have demonstrated the importance of mitochondrial fission in the pathogenesis of metabolic disorders. ROCK1 knockout mice have shown less mitochondrial fragmentation, reduced levels of ROS, and decreased apoptosis in kidney glomeruli under diabetic conditions [89]. Similarly, inhibiting mitochondrial fission by using the dominant-negative fission mutant Drp1-K38A has been shown to reduce ROS levels and oxidative stress, and improve liver function, in diabetic mice [92]. In summary, hyperglycemia and/or high-fat diets promote mitochondrial fission and increase ROS production, which leads to a range of metabolic complications. The mechanism of this process involves the activation of Drp1 by various kinases, intracellular calcium levels, and mitochondrial membrane potential. Numerous recent review articles have emphasized the connections among specific metabolic diseases, mitochondrial fission/fusion, and ROS signaling. We recommend that readers interested in exploring this topic further refer to these articles [106,107,108,109,110,111].

4. Target Mitochondrial Fission for Antioxidants in Metabolic Diseases

As discussed above, Drp1-mediated mitochondrial fission serves a critical role in regulating mitochondrial function and ROS production, and the dysregulation of mitochondrial fission has been linked to the pathogenesis of several metabolic diseases. Lifestyle factors, such as exercise and diet, as well as some dietary supplement ingredients, have been shown to improve mitochondrial function and reduce ROS production by modulating mitochondrial fission. Moreover, chemicals targeting mitochondrial fission, such as mitochondrial division inhibitor-1 (Mdivi-1) and elamipretide (SS-31), have emerged as potential therapeutic agents for metabolic diseases. In this section, we discuss the potential benefits of targeting mitochondrial fission using various approaches, including lifestyle modifications, dietary supplements, and chemicals (Figure 4). We also discuss the interplay between several widely used metabolic disease drugs and mitochondrial dynamics, particularly mitochondrial fission.

4.1. Effect of Lifestyle on Mitochondrial Fission and ROS

Exercise is a well-established intervention for the prevention and treatment of metabolic diseases. Exercise has beneficial effects on mitochondrial function and fission. Regular exercise increases mitochondrial biogenesis through the activation of the AMPK/ PGC-1α and CaMK/Mitogen-activated protein kinase signaling pathways [112,113], decreases ROS production by upregulating various antioxidant enzymes [114], and enhances mitochondrial quality control (MCQ) mechanisms by activating autophagy [115,116]. Recent studies also show that exercise can modulate mitochondrial fission [117]. High-intensity interval training, for example, has been shown to increase mitochondrial network volume and decrease mitochondrial fragmentation through NOX2 signaling in skeletal muscle [118]. Swimming as a form of exercise can help alleviate endothelial mitochondrial fragmentation by inhibiting Drp1, which contributes to the positive effects of exercise on vascular function and blood pressure in individuals with hypertension [119]. In the nematode Caenorhabditis elegans, a single exercise session triggers a process of mitochondrial fragmentation, followed by fusion after a recovery period in the body-wall muscle [120]. In addition, regular daily exercise sessions can delay mitochondrial fragmentation and the decline of physical fitness [120]. Chronic exercise induces mitochondrial fusion, which leads to improved mitochondrial dynamics and insulin sensitivity in adipose tissue [121].
Various dietary patterns, such as high-fat diets and sugar-sweetened beverages, can increase mitochondrial fission and impair mitochondrial function, and thereby contribute to the progression of metabolic diseases [12,13]. On the other hand, calorie restriction has been shown to decrease mitochondrial fission and ROS production [122]. Low-fat diets enriched in complex carbohydrates, fiber, and plant-based proteins have been shown to reduce mitochondrial fission and promote mitochondrial fusion, leading to improved energy metabolism and mitochondrial function [123,124]. This is due in part to the fact that carbohydrates stimulate insulin secretion, which activates the PI3K/Akt pathway, and lead to an increase in mitochondrial biogenesis and function [125]. A diet rich in polyunsaturated fatty acids, such as those found in fish, nuts, and seeds, has been associated with improved mitochondrial function and decreased fission in metabolic diseases [126]. In partiular, omega-3 fatty acids may reduce mitochondrial fission by enhancing membrane fluidity and reducing oxidative stress in human skeletal muscle [127].
Chronic stress has been shown to increase mitochondrial fission and ROS production, which can contribute to the development of cardiovascular disease, diabetes, and obesity [128]. This may reflect activation of the hypothalamic–pituitary–adrenal axis and sympathetic nervous system. Chronic activation of these pathways leads to the release of stress hormones such as cortisol and adrenaline, which can affect mitochondrial dynamics and function [128,129,130]. For example, chronic exposure to elevated levels of cortisol or dexamethasone has been shown to increase mitochondrial fission and decrease fusion, which may lead to mitochondrial dysfunction [131,132]. Another mechanism involves the activation of inflammatory pathways in response to chronic stress. Inflammation can lead to the production of ROS and oxidative stress, which can, in turn, affect mitochondrial function and dynamics [133]. The inflammatory cytokine TNF-α can increase mitochondrial fission and ROS production [134]. The pro-inflammatory cytokine IL-6 has also been shown to increase mitochondrial fission and decrease fusion, which leads to mitochondrial fragmentation and dysfunction [135,136]. Thus, stress management techniques, such as meditation and yoga, can help to improve mitochondrial integrity and function. and prevent the development of metabolic diseases.

4.2. Effect of Dietary Supplements on Mitochondrial Fission and ROS

Some ingredients found in dietary supplements may have potential as adjunct therapies for metabolic diseases, particularly when combined with lifestyle interventions such as exercise and dietary changes as discussed above. In this section, we consider several ingredients that have been shown to improve mitochondrial function and reduce oxidative stress in metabolic diseases. Of note, further research is needed to fully elucidate the mechanisms by which these ingredients improve mitochondrial integrity and function, as well as to determine the optimal amounts and combinations for mitigating metabolic diseases.
Coenzyme Q10 (CoQ10), a key component of the electron transport chain, is required for ATP production. CoQ10 exhibits antioxidant properties by providing electrons to unstable molecules such as free radicals, thereby neutralizing them. CoQ10 also aids in the regeneration of other antioxidants such as Vitamin E, and has been shown to improve mitochondrial function and reduce oxidative stress in various animal and human models [137,138]. Several recent studies found that CoQ10 supplementation can reduce mitochondrial fission and increase mitochondrial fusion, to result in improved mitochondrial function and reduced ROS production [139,140,141]. In a mouse model of obesity and T2DM, CoQ10 supplementation improved lipid metabolism and mitigated obesity, possibly through CaMKII-mediated phosphodiesterase-4 inhibition [142]. Clinical studies have shown that CoQ10 supplementation can improve insulin resistance, glycemic control, and lipid metabolism in diabetic patients, to potentially reduce the risk of diabetic complications [143]. In obesity, CoQ10 supplementation has been shown to improve mitochondrial function and reduce inflammation, which is a hallmark of obesity-induced insulin resistance [144]. These findings suggest that CoQ10 may have potential as a therapeutic agent for the management of metabolic diseases.
Resveratrol is a polyphenol found in grapes, berries, and other plants. Recent studies have shown that resveratrol has antioxidant properties and can improve mitochondrial function in metabolic diseases. Resveratrol can activate sirtuin 1, a key regulator of mitochondrial biogenesis and function [145,146,147,148]. Resveratrol has been shown to promote mitochondrial fusion and decrease the expression of the fission proteins Drp1 and Fis1, which leads to a decrease in mitochondrial fission [145,146,149]. This decrease in mitochondrial fission results in an improvement in mitochondrial function, including increased ATP production, reduced oxidative stress, and improved insulin sensitivity [147,148,150,151]. Resveratrol also reduces oxidative stress by increasing the activity of antioxidant enzymes [152]. Clinical studies have suggested that resveratrol supplementation may have beneficial effects on metabolic diseases in both animal models and humans [153,154].
Astaxanthin is a carotenoid pigment with potent antioxidant and anti-inflammatory properties. Several studies have investigated the effects of astaxanthin on mitochondrial fission and ROS production in the context of metabolic diseases. In mouse skeletal muscle and hypothalamus, astaxanthin improved mitochondrial biogenesis, inhibited mitochondrial fragmentation, and reduced oxidative damage [155,156]. Astaxanthin can activate the mammalian target of rapamycin (mTOR) pathway, which results in elevated mitochondrial fusion and reduced fission [157]. In the blood vessels, astaxanthin prevented Drp1 Ser616 phosphorylation and the excessive generation of mitochondrial ROS [158]. In rats, astaxanthin reduced Drp1 protein expression and delayed the pathogenesis of diabetic nephropathy [159]. The activation of the AMPK pathway by astaxanthin may contribute to the stimulation of mitochondrial biogenesis and the improvement of insulin resistance [160]. In human subjects, astaxanthin exhibits some advantages in enhancing mitochondrial function and halting the progression of metabolic diseases [161,162]. However, more research is needed to fully understand the potential benefits and mechanisms of astaxanthin in metabolic disease.
Curcumin, a polyphenol found in turmeric, has been shown to inhibit mitochondrial fission and improve mitochondrial function in obesity and diabetes. Curcumin regulates ROS hormesis, which can serve to inhibit mitochondrial fission and promote fusion/elongation and biogenesis, and improve function, in the skeletal muscle of rodents, possibly through NADPH oxidase-dependent redox signaling [26,163]. Curcumin treatment in mice resulted in upregulation of the expression of the transcriptional coactivator PGC1α protein, which likely restored mitochondrial fusion and improved mitochondrial function [164]. In human neuroblastoma cells, curcumin was found to increase mitochondrial fusion activity, decrease fission machinery, and enhance biogenesis [165]. The results of clinical studies have been reviewed recently, and suggest that curcumin may be beneficial in patients with metabolic diseases [166,167].
L-citrulline is a non-essential amino acid found in watermelon and other foods. L-citrulline is a precursor to L-arginine, a key substrate for the production of nitric oxide (NO) in the body. Excessive NO has been shown to stimulate the S-nitrosylation of Drp1 and promote mitochondrial fission [79]. L-NAME, a non-selective NO synthase inhibitor, abolished the S-nitrosylation and phosphorylation of Drp1 at Ser616 in neurons, which led to elongation of the mitochondria [168]. L-citrulline caused an increase in NO levels in skeletal muscle, which resulted in the reduced phosphorylation of Drp1 at Ser616 and increased phosphorylation at Ser637. As a result, mitochondrial fission was inhibited, and oxidative stress and its associated damage were alleviated. [169]. Animal and human studies suggest that L-citrulline may have potential therapeutic benefits in various metabolic diseases by improving mitochondrial function and reducing oxidative stress, due to the ability of L-citrulline to increase NO production [170,171]. Additional research is necessary to confirm whether supplementation with L-citrulline could be an effective therapeutic approach to preventing and treating metabolic disorders.

4.3. Drp1 Chemical Inhibitors

Drp1 inhibitors are a type of chemical compound that can block the activity of Drp1 and prevent it from functioning properly. These inhibitors work by either directly targeting the GTPase activity of Drp1 or disrupting the interactions between Drp1 and its binding proteins. Several Drp1 inhibitors have been developed and studied for their potential use in treating a variety of metabolic disorders, including obesity, T2DM, and cardiovascular diseases.
Mitochondrial division inhibitor-1 (Mdivi-1) was first identified as a selective inhibitor of Drp1 in a high-throughput screen of small molecules [172]. It inhibits Drp1 GTPase activity and disrupts the assembly of the Drp1 complex, which then leads to reduced mitochondrial fission [172]. Mdivi-1 has been shown to be effective in various animal models of diseases characterized by dysregulated mitochondrial fission. Mdivi-1 treatment improved mitochondrial function and reduced oxidative injury in the skeletal muscle of heat-stressed mice [173]. It reduced adipose tissue inflammation and improved glucose metabolism in obese mice [174]. Mdivi-1 treatment also improved insulin sensitivity and glucose uptake in adipose tissue and skeletal muscle [174,175,176]. In studies on high-fat diet-induced obesity in mice and obese insulin-resistance in humans, Mdivi-1 was found to reduce body weight, ROS production, and insulin resistance. It improved glucose tolerance and decreased fasting blood glucose levels and reduced oxidative stress and inflammation in the liver and skeletal muscle [174,175,176]. Treatment with Mdivi-1 also improved mitochondrial function and reduced oxidative stress in the hearts of diabetic mice, which led to improvements in cardiac function [177]. In addition to its effects on mitochondrial dynamics, Mdivi-1 has been found to have anti-inflammatory and anti-oxidative properties [178,179,180], and it inhibits autophagy [178,181], which may contribute to its therapeutic effects in metabolic diseases.
P110 is a peptide inhibitor that disrupts the interaction between Drp1 and Fis1, and inhibits Drp1 enzyme activity [182]. By inhibiting Drp1, P110 prevents excessive mitochondrial fragmentation and protects against mitochondrial dysfunction and cell death [182]. P110 and Mdivi-1 have been shown to reduce the translocation of Drp1 to the mitochondria and ROS production, which led to improved mitochondrial and heart function [183,184,185]. P110 reduced oxidative stress in neurons and improved mitochondrial function, motor performance, and survival [186]. This effect may be mediated through TNF-α-induced inflammation, H2O2-induced oxidation, and/or the RIPK1/RIPK3 pathway [187]. P110 may also abolish the association of p53 with the mitochondria and reduce brain infarction in rats subjected to brain ischemia/reperfusion injury [188]. P110 was able to decrease Drp1 Thr595 phosphorylation and reduce mitochondrial dysfunction [189]. Overall, these studies suggest that Drp1 inhibitors such as P110 have the potential to improve metabolic function and may be useful in the treatment of metabolic diseases [190,191].
Dynasore is a small molecule that can inhibit Drp1 activity by blocking the activity of dynamin, a protein involved in the formation of membrane vesicles [192]. Dynamin is involved in the regulation of glucose transporter 4 trafficking, which is essential for insulin-mediated glucose uptake [193]. One of the main mechanisms by which Dynasore may exert its effects on metabolic diseases is through the inhibition of glucose uptake. Glucose uptake is a critical step in the development of metabolic diseases, as increased glucose uptake by cells leads to insulin resistance and impaired glucose homeostasis [194]. Dynasore has been shown to improve insulin resistance and glucose metabolism in human liver cells [195]. The dysregulation of lipid metabolism is a hallmark of metabolic diseases, and the inhibition of endocytosis by Dynasore has been shown to inhibit lipolysis and improve lipid metabolism in adipocytes [196]. In human HeLa cells and macrophages, treatment with Dynasore resulted in a decrease in the levels of intracellular cholesterol and impaired the trafficking of cholesterol from the plasma membrane to the endoplasmic reticulum [197]. In addition, Dynasore directly inhibited the mitochondrial fission protein Drp1 and reduced oxidative stress in cardiomyocytes under ischemic/reperfusion conditions, which led to increased cardiomyocyte survival and improved overall cardiac function [198].

4.4. Effect of Metabolic Disease Drugs on Mitochondrial Fission and ROS

Metformin, a commonly prescribed medication for T2DM, has been extensively studied for its effects on mitochondrial function. Metformin has been shown to inhibit mitochondrial fission and promote mitochondrial fusion [199,200]. It exerts its effects by activating AMPK, which, in turn, inhibits Drp1-mediated mitochondrial fission [199,200]. Interestingly, several recent studies have shown that metformin improves mitochondrial respiration by increasing mitochondrial fission through AMPK-Mff signaling [201], or by phosphorylating AMPK/Drp1 Ser616 [202]. Nevertheless, in patients with T2DM, metformin restored mitochondrial fission/fusion balance and improved mitochondrial function [203]. Additionally, metformin has antioxidant properties and can reduce ROS production, leading to decreased oxidative stress [204]. These effects contribute to the potential benefits of metformin in mitigating mitochondrial dysfunction and oxidative damage associated with metabolic diseases.
Sodium–Glucose Cotransporter-2 inhibitors (SGLT-2i) are a class of medications used to lower blood glucose levels in individuals with T2DM. A recent review article has summarized that SGLT-2 inhibitors may have direct effects on mitochondrial function [205]. SGLT-2i treatment has been shown to increase mitochondrial biogenesis and improve mitochondrial function in various tissues, including the heart and kidney [205]. While the exact impact on mitochondrial fission is not yet fully understood, the beneficial effects of SGLT-2 inhibitors on mitochondrial function may directly or indirectly influence mitochondrial fission and fusion proteins [206,207,208,209]. Furthermore, SGLT-2 inhibitors have been reported to reduce oxidative stress and ROS production, potentially through their glucose-lowering and hemodynamic effects, which can contribute to their overall metabolic benefits [210].
Statins are widely used cholesterol-lowering drugs that inhibit the enzyme HMG-CoA reductase, a key enzyme in cholesterol synthesis [211]. Statins have been reported to affect mitochondrial function [212,213], but their effects on mitochondrial fission are not well-defined. Some studies have reported that statin treatment can reduce the expression and activity of the mitochondrial fission proteins Drp1 and Fis1 [214,215,216]. However, the results from different studies are conflicting, with some reporting an increase in fission-related proteins/mitochondrial dysfunction and others showing no significant effect of statin treatment [213,217,218,219]. It is likely that the effects of statins on mitochondrial dynamics vary depending on the specific statin, its dosage, the duration of treatment, and the experimental model or tissue studied. Further research is needed to elucidate the precise mechanisms and determine the consistent effects of statins on mitochondrial fission and fusion.
ATP-citrate lyase (ACLY) inhibitors are emerging as potential therapeutics for metabolic diseases and cancer, targeting key enzymes involved in de novo lipogenesis [220]. ACLY plays a role in providing acetyl-CoA, which is known to modulate the post-translational modifications of proteins, including lysine acetylation [220,221]. The acetylation of Drp1 has been shown to increase its activity and mitochondrial localization, thereby activating mitochondrial fission [222]. By inhibiting ACLY and reducing the availability of acetyl-CoA, mitochondrial fission could be indirectly affected. However, the specific consequences would depend on the context and the overall metabolic state of the cell. Further research is needed to fully understand the relationship between ACLY inhibition and mitochondrial fission and to elucidate the molecular mechanisms involved.

5. Conclusions and Perspectives

In conclusion, targeting mitochondrial fission represents a promising strategy for mitigating mitochondrial dysfunction and oxidative stress in metabolic diseases. The balance between ROS production and scavenging is tightly regulated, and mitochondrial fission serves a critical role in this process. By inhibiting mitochondrial fission or enhancing antioxidant capacity, it may be possible to restore mitochondrial homeostasis and improve metabolic health (Figure 4).
However, there are limitations and challenges to consider in targeting mitochondrial fission for metabolic disease treatment. For example, the role of mitochondrial fission in metabolic diseases is complex and involves multiple factors, and the optimal duration and intensity of mitochondrial fission inhibition are not yet clear. In addition, the effects of long-term mitochondrial fission inhibition on other physiological processes, such as cellular respiration and autophagy, need to be further investigated. Furthermore, effective and safe mitochondrial fission inhibitors with clinical relevance are lacking.
While mitochondrial fission has been extensively discussed in this review, it is important to acknowledge the role of mitochondrial fusion in maintaining mitochondrial function and metabolic health. Studies have shown that abnormalities in mitochondrial fusion can also lead to metabolic dysfunction and contribute to the development of metabolic diseases such as obesity, insulin resistance, and T2DM [12,106,223]. In particular, impaired mitochondrial fusion has been associated with reduced oxidative capacity and impaired glucose uptake in skeletal muscle and adipose tissue, which are important tissues for whole-body glucose homeostasis [106,224].
Therefore, future studies should focus on identifying the specific mechanisms underlying mitochondrial fission and ROS production in metabolic diseases, as well as on developing novel and effective mitochondrial fission inhibitors with minimal side effects. Moreover, it is important to explore the potential synergistic effects of targeting mitochondrial fission with other antioxidant therapies, such as lifestyle modifications and dietary supplements. A better understanding of the complex relationships and interactions among mitochondrial fission/fusion, ROS, and metabolic diseases will ultimately lead to the development of new and effective therapeutic strategies for these conditions.

Author Contributions

Conceptualization, T.Y.; Writing—Original Draft Preparation, T.Y.; Writing—Review and Editing, T.Y., L.W., L.Z. and P.A.D. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the U.S. Department of Defense (DoD)’s Defense Health Program (HU00011920047).

Acknowledgments

We thank Beth McNally for her critical reading of the manuscript.

Conflicts of Interest

The opinions and assertions expressed herein are those of the authors and do not reflect the official policy or position of the Uniformed Services University or the Department of Defense. The contents of this publication are the sole responsibility of the authors and do not necessarily reflect the views, opinions or policies of The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. Mention of trade names, commercial products, or organizations does not imply endorsement by the U.S. Government. The authors have no financial interests or relationships to disclose.

References

  1. Saklayen, M.G. The Global Epidemic of the Metabolic Syndrome. Curr. Hypertens. Rep. 2018, 20, 12. [Google Scholar] [CrossRef] [PubMed]
  2. Okunogbe, A.; Nugent, R.; Spencer, G.; Ralston, J.; Wilding, J. Economic impacts of overweight and obesity: Current and future estimates for eight countries. BMJ Glob. Health 2021, 6, e006351. [Google Scholar] [CrossRef] [PubMed]
  3. Powell-Wiley, T.M.; Poirier, P.; Burke, L.E.; Despres, J.P.; Gordon-Larsen, P.; Lavie, C.J.; Lear, S.A.; Ndumele, C.E.; Neeland, I.J.; Sanders, P.; et al. Obesity and Cardiovascular Disease: A Scientific Statement From the American Heart Association. Circulation 2021, 143, e984–e1010. [Google Scholar] [CrossRef] [PubMed]
  4. Ginsberg, H.N.; MacCallum, P.R. The obesity, metabolic syndrome, and type 2 diabetes mellitus pandemic: Part I. Increased cardiovascular disease risk and the importance of atherogenic dyslipidemia in persons with the metabolic syndrome and type 2 diabetes mellitus. J. Cardiometab. Syndr. 2009, 4, 113–119. [Google Scholar] [CrossRef]
  5. Ghaben, A.L.; Scherer, P.E. Adipogenesis and metabolic health. Nat. Rev. Mol. Cell Biol. 2019, 20, 242–258. [Google Scholar] [CrossRef]
  6. Barroso, I.; McCarthy, M.I. The Genetic Basis of Metabolic Disease. Cell 2019, 177, 146–161. [Google Scholar] [CrossRef]
  7. Ordovas, J.M.; Shen, J. Gene-environment interactions and susceptibility to metabolic syndrome and other chronic diseases. J. Periodontol. 2008, 79, 1508–1513. [Google Scholar] [CrossRef]
  8. Incalza, M.A.; D’Oria, R.; Natalicchio, A.; Perrini, S.; Laviola, L.; Giorgino, F. Oxidative stress and reactive oxygen species in endothelial dysfunction associated with cardiovascular and metabolic diseases. Vascul. Pharmacol. 2018, 100, 1–19. [Google Scholar] [CrossRef]
  9. Sies, H.; Belousov, V.V.; Chandel, N.S.; Davies, M.J.; Jones, D.P.; Mann, G.E.; Murphy, M.P.; Yamamoto, M.; Winterbourn, C. Defining roles of specific reactive oxygen species (ROS) in cell biology and physiology. Nat. Rev. Mol. Cell Biol. 2022, 23, 499–515. [Google Scholar] [CrossRef]
  10. Mailloux, R.J. An Update on Mitochondrial Reactive Oxygen Species Production. Antioxidants 2020, 9, 472. [Google Scholar] [CrossRef]
  11. Hernansanz-Agustin, P.; Enriquez, J.A. Generation of Reactive Oxygen Species by Mitochondria. Antioxidants 2021, 10, 415. [Google Scholar] [CrossRef] [PubMed]
  12. Yoon, Y.; Galloway, C.A.; Jhun, B.S.; Yu, T. Mitochondrial dynamics in diabetes. Antioxid. Redox Signal. 2011, 14, 439–457. [Google Scholar] [CrossRef] [PubMed]
  13. Yu, T.; Wang, L.; Yoon, Y. Morphological control of mitochondrial bioenergetics. Front. Biosci. (Landmark Ed) 2015, 20, 229–246. [Google Scholar] [PubMed]
  14. Jezek, J.; Cooper, K.F.; Strich, R. Reactive Oxygen Species and Mitochondrial Dynamics: The Yin and Yang of Mitochondrial Dysfunction and Cancer Progression. Antioxidants 2018, 7, 13. [Google Scholar] [CrossRef]
  15. Kraus, F.; Roy, K.; Pucadyil, T.J.; Ryan, M.T. Function and regulation of the divisome for mitochondrial fission. Nature 2021, 590, 57–66. [Google Scholar] [CrossRef] [PubMed]
  16. Ihenacho, U.K.; Meacham, K.A.; Harwig, M.C.; Widlansky, M.E.; Hill, R.B. Mitochondrial Fission Protein 1: Emerging Roles in Organellar Form and Function in Health and Disease. Front. Endocrinol. 2021, 12, 660095. [Google Scholar] [CrossRef]
  17. Youle, R.J.; van der Bliek, A.M. Mitochondrial fission, fusion, and stress. Science 2012, 337, 1062–1065. [Google Scholar] [CrossRef]
  18. Weidinger, A.; Kozlov, A.V. Biological Activities of Reactive Oxygen and Nitrogen Species: Oxidative Stress versus Signal Transduction. Biomolecules 2015, 5, 472–484. [Google Scholar] [CrossRef]
  19. Ray, P.D.; Huang, B.W.; Tsuji, Y. Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell. Signal. 2012, 24, 981–990. [Google Scholar] [CrossRef]
  20. Caito, S.W.; Aschner, M. Mitochondrial Redox Dysfunction and Environmental Exposures. Antioxid. Redox Signal. 2015, 23, 578–595. [Google Scholar] [CrossRef]
  21. Ichihashi, M.; Ueda, M.; Budiyanto, A.; Bito, T.; Oka, M.; Fukunaga, M.; Tsuru, K.; Horikawa, T. UV-induced skin damage. Toxicology 2003, 189, 21–39. [Google Scholar] [CrossRef] [PubMed]
  22. Sarmiento-Salinas, F.L.; Perez-Gonzalez, A.; Acosta-Casique, A.; Ix-Ballote, A.; Diaz, A.; Trevino, S.; Rosas-Murrieta, N.H.; Millan-Perez-Pena, L.; Maycotte, P. Reactive oxygen species: Role in carcinogenesis, cancer cell signaling and tumor progression. Life Sci. 2021, 284, 119942. [Google Scholar] [CrossRef] [PubMed]
  23. Fransen, M.; Nordgren, M.; Wang, B.; Apanasets, O. Role of peroxisomes in ROS/RNS-metabolism: Implications for human disease. Biochim. Biophys. Acta 2012, 1822, 1363–1373. [Google Scholar] [CrossRef] [PubMed]
  24. Magnani, F.; Mattevi, A. Structure and mechanisms of ROS generation by NADPH oxidases. Curr. Opin. Struct. Biol. 2019, 59, 91–97. [Google Scholar] [CrossRef]
  25. Vermot, A.; Petit-Hartlein, I.; Smith, S.M.E.; Fieschi, F. NADPH Oxidases (NOX): An Overview from Discovery, Molecular Mechanisms to Physiology and Pathology. Antioxidants 2021, 10, 890. [Google Scholar] [CrossRef]
  26. Yu, T.; Dohl, J.; Wang, L.; Chen, Y.; Gasier, H.G.; Deuster, P.A. Curcumin Ameliorates Heat-Induced Injury through NADPH Oxidase-Dependent Redox Signaling and Mitochondrial Preservation in C2C12 Myoblasts and Mouse Skeletal Muscle. J. Nutr. 2020, 150, 2257–2267. [Google Scholar] [CrossRef]
  27. Matsushima, S.; Sadoshima, J. Yin and Yang of NADPH Oxidases in Myocardial Ischemia-Reperfusion. Antioxidants 2022, 11, 1069. [Google Scholar] [CrossRef]
  28. Islam, M.N.; Rauf, A.; Fahad, F.I.; Emran, T.B.; Mitra, S.; Olatunde, A.; Shariati, M.A.; Rebezov, M.; Rengasamy, K.R.R.; Mubarak, M.S. Superoxide dismutase: An updated review on its health benefits and industrial applications. Crit. Rev. Food Sci. Nutr. 2022, 62, 7282–7300. [Google Scholar] [CrossRef]
  29. Alfonso-Prieto, M.; Biarnes, X.; Vidossich, P.; Rovira, C. The molecular mechanism of the catalase reaction. J. Am. Chem. Soc. 2009, 131, 11751–11761. [Google Scholar] [CrossRef]
  30. Brigelius-Flohe, R.; Maiorino, M. Glutathione peroxidases. Biochim. Biophys. Acta 2013, 1830, 3289–3303. [Google Scholar] [CrossRef]
  31. Lu, J.; Holmgren, A. The thioredoxin antioxidant system. Free Radic. Biol Med 2014, 66, 75–87. [Google Scholar] [CrossRef] [PubMed]
  32. Rhee, S.G. Overview on Peroxiredoxin. Mol. Cells 2016, 39, 1–5. [Google Scholar] [CrossRef] [PubMed]
  33. Holmgren, A. Thioredoxin and glutaredoxin systems. J. Biol. Chem. 1989, 264, 13963–13966. [Google Scholar] [CrossRef] [PubMed]
  34. Rains, J.L.; Jain, S.K. Oxidative stress, insulin signaling, and diabetes. Free Radic. Biol. Med. 2011, 50, 567–575. [Google Scholar] [CrossRef]
  35. Yaribeygi, H.; Sathyapalan, T.; Atkin, S.L.; Sahebkar, A. Molecular Mechanisms Linking Oxidative Stress and Diabetes Mellitus. Oxidative Med. Cell. Longev. 2020, 2020, 8609213. [Google Scholar] [CrossRef]
  36. Kohlgruber, S.; Upadhye, A.; Dyballa-Rukes, N.; McNamara, C.A.; Altschmied, J. Regulation of Transcription Factors by Reactive Oxygen Species and Nitric Oxide in Vascular Physiology and Pathology. Antioxid. Redox Signal. 2017, 26, 679–699. [Google Scholar] [CrossRef]
  37. Bouchez, C.; Devin, A. Mitochondrial Biogenesis and Mitochondrial Reactive Oxygen Species (ROS): A Complex Relationship Regulated by the cAMP/PKA Signaling Pathway. Cells 2019, 8, 287. [Google Scholar] [CrossRef]
  38. Schofield, J.H.; Schafer, Z.T. Mitochondrial Reactive Oxygen Species and Mitophagy: A Complex and Nuanced Relationship. Antioxid. Redox Signal. 2021, 34, 517–530. [Google Scholar] [CrossRef]
  39. Kiselyov, K.; Muallem, S. ROS and intracellular ion channels. Cell Calcium 2016, 60, 108–114. [Google Scholar] [CrossRef]
  40. Massaad, C.A.; Klann, E. Reactive oxygen species in the regulation of synaptic plasticity and memory. Antioxid. Redox Signal. 2011, 14, 2013–2054. [Google Scholar] [CrossRef]
  41. Gasier, H.G.; Dohl, J.; Suliman, H.B.; Piantadosi, C.A.; Yu, T. Skeletal muscle mitochondrial fragmentation and impaired bioenergetics from nutrient overload are prevented by carbon monoxide. Am. J. Physiol. Cell Physiol. 2020, 319, C746–C756. [Google Scholar] [CrossRef] [PubMed]
  42. Palomer, X.; Pizarro-Delgado, J.; Barroso, E.; Vazquez-Carrera, M. Palmitic and Oleic Acid: The Yin and Yang of Fatty Acids in Type 2 Diabetes Mellitus. Trends Endocrinol. Metab. 2018, 29, 178–190. [Google Scholar] [CrossRef] [PubMed]
  43. Tan, Y.; Miao, L.; Xiao, J.; Cheang, W.S. 3,3′,4,5′-Tetramethoxy-trans-stilbene Improves Insulin Resistance by Activating the IRS/PI3K/Akt Pathway and Inhibiting Oxidative Stress. Curr. Issues Mol. Biol. 2022, 44, 2175–2185. [Google Scholar] [CrossRef] [PubMed]
  44. Zaulkffali, A.S.; Md Razip, N.N.; Syed Alwi, S.S.; Abd Jalil, A.; Abd Mutalib, M.S.; Gopalsamy, B.; Chang, S.K.; Zainal, Z.; Ibrahim, N.N.; Zakaria, Z.A.; et al. Vitamins D and E Stimulate the PI3K-AKT Signalling Pathway in Insulin-Resistant SK-N-SH Neuronal Cells. Nutrients 2019, 11, 2525. [Google Scholar] [CrossRef] [PubMed]
  45. Li, H.; Yu, X. Emerging role of JNK in insulin resistance. Curr. Diabetes Rev. 2013, 9, 422–428. [Google Scholar] [CrossRef] [PubMed]
  46. Kim, J.J.; Sears, D.D. TLR4 and Insulin Resistance. Gastroenterol. Res. Pract. 2010, 2010, 212563. [Google Scholar] [CrossRef]
  47. Tormos, K.V.; Anso, E.; Hamanaka, R.B.; Eisenbart, J.; Joseph, J.; Kalyanaraman, B.; Chandel, N.S. Mitochondrial complex III ROS regulate adipocyte differentiation. Cell Metab. 2011, 14, 537–544. [Google Scholar] [CrossRef]
  48. Atashi, F.; Modarressi, A.; Pepper, M.S. The role of reactive oxygen species in mesenchymal stem cell adipogenic and osteogenic differentiation: A review. Stem Cells Dev. 2015, 24, 1150–1163. [Google Scholar] [CrossRef]
  49. Lee, H.; Lee, Y.J.; Choi, H.; Ko, E.H.; Kim, J.W. Reactive oxygen species facilitate adipocyte differentiation by accelerating mitotic clonal expansion. J. Biol. Chem. 2009, 284, 10601–10609. [Google Scholar] [CrossRef]
  50. Oak, J.H.; Cai, H. Attenuation of angiotensin II signaling recouples eNOS and inhibits nonendothelial NOX activity in diabetic mice. Diabetes 2007, 56, 118–126. [Google Scholar] [CrossRef]
  51. Miyata, T.; van Ypersele de Strihou, C.; Ueda, Y.; Ichimori, K.; Inagi, R.; Onogi, H.; Ishikawa, N.; Nangaku, M.; Kurokawa, K. Angiotensin II receptor antagonists and angiotensin-converting enzyme inhibitors lower in vitro the formation of advanced glycation end products: Biochemical mechanisms. J. Am. Soc. Nephrol. 2002, 13, 2478–2487. [Google Scholar] [CrossRef] [PubMed]
  52. Yamagishi, S.; Maeda, S.; Matsui, T.; Ueda, S.; Fukami, K.; Okuda, S. Role of advanced glycation end products (AGEs) and oxidative stress in vascular complications in diabetes. Biochim. Biophys. Acta 2012, 1820, 663–671. [Google Scholar] [CrossRef]
  53. Carrizzo, A.; Forte, M.; Damato, A.; Trimarco, V.; Salzano, F.; Bartolo, M.; Maciag, A.; Puca, A.A.; Vecchione, C. Antioxidant effects of resveratrol in cardiovascular, cerebral and metabolic diseases. Food Chem. Toxicol. 2013, 61, 215–226. [Google Scholar] [CrossRef] [PubMed]
  54. Higgins, M.R.; Izadi, A.; Kaviani, M. Antioxidants and Exercise Performance: With a Focus on Vitamin E and C Supplementation. Int. J. Environ. Res. Public Health 2020, 17, 8452. [Google Scholar] [CrossRef] [PubMed]
  55. Kouki, A.; Ferjani, W.; Ghanem-Boughanmi, N.; Ben-Attia, M.; Dang, P.M.; Souli, A.; El-Benna, J. The NADPH Oxidase Inhibitors Apocynin and Diphenyleneiodonium Protect Rats from LPS-Induced Pulmonary Inflammation. Antioxidants 2023, 12, 770. [Google Scholar] [CrossRef]
  56. Chan, D.C. Mitochondrial Dynamics and Its Involvement in Disease. Annu. Rev. Pathol. 2020, 15, 235–259. [Google Scholar] [CrossRef]
  57. Zorov, D.B.; Vorobjev, I.A.; Popkov, V.A.; Babenko, V.A.; Zorova, L.D.; Pevzner, I.B.; Silachev, D.N.; Zorov, S.D.; Andrianova, N.V.; Plotnikov, E.Y. Lessons from the Discovery of Mitochondrial Fragmentation (Fission): A Review and Update. Cells 2019, 8, 175. [Google Scholar] [CrossRef]
  58. Suliman, H.B.; Piantadosi, C.A. Mitochondrial Quality Control as a Therapeutic Target. Pharmacol. Rev. 2016, 68, 20–48. [Google Scholar] [CrossRef]
  59. Ong, S.B.; Hall, A.R.; Hausenloy, D.J. Mitochondrial dynamics in cardiovascular health and disease. Antioxid. Redox Signal. 2013, 19, 400–414. [Google Scholar] [CrossRef]
  60. Noone, J.; O’Gorman, D.J.; Kenny, H.C. OPA1 regulation of mitochondrial dynamics in skeletal and cardiac muscle. Trends Endocrinol. Metab. 2022, 33, 710–721. [Google Scholar] [CrossRef]
  61. Chen, H.; Detmer, S.A.; Ewald, A.J.; Griffin, E.E.; Fraser, S.E.; Chan, D.C. Mitofusins Mfn1 and Mfn2 coordinately regulate mitochondrial fusion and are essential for embryonic development. J. Cell Biol. 2003, 160, 189–200. [Google Scholar] [CrossRef] [PubMed]
  62. Chen, H.; Vermulst, M.; Wang, Y.E.; Chomyn, A.; Prolla, T.A.; McCaffery, J.M.; Chan, D.C. Mitochondrial fusion is required for mtDNA stability in skeletal muscle and tolerance of mtDNA mutations. Cell 2010, 141, 280–289. [Google Scholar] [CrossRef] [PubMed]
  63. Gao, S.; Hu, J. Mitochondrial Fusion: The Machineries In and Out. Trends Cell Biol. 2021, 31, 62–74. [Google Scholar] [CrossRef]
  64. Adebayo, M.; Singh, S.; Singh, A.P.; Dasgupta, S. Mitochondrial fusion and fission: The fine-tune balance for cellular homeostasis. FASEB J. 2021, 35, e21620. [Google Scholar] [CrossRef] [PubMed]
  65. Yu, T.; Deuster, P.; Chen, Y. Role of dynamin-related protein 1-mediated mitochondrial fission in resistance of mouse C2C12 myoblasts to heat injury. J. Physiol. 2016, 594, 7419–7433. [Google Scholar] [CrossRef] [PubMed]
  66. Lee, J.E.; Westrate, L.M.; Wu, H.; Page, C.; Voeltz, G.K. Multiple dynamin family members collaborate to drive mitochondrial division. Nature 2016, 540, 139–143. [Google Scholar] [CrossRef] [PubMed]
  67. Kalia, R.; Wang, R.Y.; Yusuf, A.; Thomas, P.V.; Agard, D.A.; Shaw, J.M.; Frost, A. Structural basis of mitochondrial receptor binding and constriction by DRP1. Nature 2018, 558, 401–405. [Google Scholar] [CrossRef] [PubMed]
  68. Kamerkar, S.C.; Kraus, F.; Sharpe, A.J.; Pucadyil, T.J.; Ryan, M.T. Dynamin-related protein 1 has membrane constricting and severing abilities sufficient for mitochondrial and peroxisomal fission. Nat. Commun. 2018, 9, 5239. [Google Scholar] [CrossRef]
  69. Yu, T.; Fox, R.J.; Burwell, L.S.; Yoon, Y. Regulation of mitochondrial fission and apoptosis by the mitochondrial outer membrane protein hFis1. J. Cell Sci. 2005, 118, 4141–4151. [Google Scholar] [CrossRef]
  70. Loson, O.C.; Song, Z.; Chen, H.; Chan, D.C. Fis1, Mff, MiD49, and MiD51 mediate Drp1 recruitment in mitochondrial fission. Mol. Biol. Cell 2013, 24, 659–667. [Google Scholar] [CrossRef]
  71. Huber, N.; Guimaraes, S.; Schrader, M.; Suter, U.; Niemann, A. Charcot-Marie-Tooth disease-associated mutants of GDAP1 dissociate its roles in peroxisomal and mitochondrial fission. EMBO Rep. 2013, 14, 545–552. [Google Scholar] [CrossRef] [PubMed]
  72. Cribbs, J.T.; Strack, S. Reversible phosphorylation of Drp1 by cyclic AMP-dependent protein kinase and calcineurin regulates mitochondrial fission and cell death. EMBO Rep. 2007, 8, 939–944. [Google Scholar] [CrossRef]
  73. Toyama, E.Q.; Herzig, S.; Courchet, J.; Lewis, T.L., Jr.; Loson, O.C.; Hellberg, K.; Young, N.P.; Chen, H.; Polleux, F.; Chan, D.C.; et al. AMP-activated protein kinase mediates mitochondrial fission in response to energy stress. Science 2016, 351, 275–281. [Google Scholar] [CrossRef] [PubMed]
  74. Gomes, L.C.; Di Benedetto, G.; Scorrano, L. During autophagy mitochondria elongate, are spared from degradation and sustain cell viability. Nat. Cell Biol. 2011, 13, 589–598. [Google Scholar] [CrossRef] [PubMed]
  75. Yu, T.; Jhun, B.S.; Yoon, Y. High-glucose stimulation increases reactive oxygen species production through the calcium and mitogen-activated protein kinase-mediated activation of mitochondrial fission. Antioxid. Redox Signal. 2011, 14, 425–437. [Google Scholar] [CrossRef]
  76. Wang, H.; Song, P.; Du, L.; Tian, W.; Yue, W.; Liu, M.; Li, D.; Wang, B.; Zhu, Y.; Cao, C.; et al. Parkin ubiquitinates Drp1 for proteasome-dependent degradation: Implication of dysregulated mitochondrial dynamics in Parkinson disease. J. Biol. Chem. 2011, 286, 11649–11658. [Google Scholar] [CrossRef]
  77. Gu, L.; Zhu, Y.; Lin, X.; Li, Y.; Cui, K.; Prochownik, E.V.; Li, Y. Amplification of Glyceronephosphate O-Acyltransferase and Recruitment of USP30 Stabilize DRP1 to Promote Hepatocarcinogenesis. Cancer Res. 2018, 78, 5808–5819. [Google Scholar] [CrossRef]
  78. Figueroa-Romero, C.; Iniguez-Lluhi, J.A.; Stadler, J.; Chang, C.R.; Arnoult, D.; Keller, P.J.; Hong, Y.; Blackstone, C.; Feldman, E.L. SUMOylation of the mitochondrial fission protein Drp1 occurs at multiple nonconsensus sites within the B domain and is linked to its activity cycle. FASEB J. 2009, 23, 3917–3927. [Google Scholar] [CrossRef]
  79. Cho, D.H.; Nakamura, T.; Fang, J.; Cieplak, P.; Godzik, A.; Gu, Z.; Lipton, S.A. S-nitrosylation of Drp1 mediates beta-amyloid-related mitochondrial fission and neuronal injury. Science 2009, 324, 102–105. [Google Scholar] [CrossRef]
  80. De Palma, C.; Falcone, S.; Pisoni, S.; Cipolat, S.; Panzeri, C.; Pambianco, S.; Pisconti, A.; Allevi, R.; Bassi, M.T.; Cossu, G.; et al. Nitric oxide inhibition of Drp1-mediated mitochondrial fission is critical for myogenic differentiation. Cell Death Differ. 2010, 17, 1684–1696. [Google Scholar] [CrossRef]
  81. Pfluger, P.T.; Kabra, D.G.; Aichler, M.; Schriever, S.C.; Pfuhlmann, K.; Garcia, V.C.; Lehti, M.; Weber, J.; Kutschke, M.; Rozman, J.; et al. Calcineurin Links Mitochondrial Elongation with Energy Metabolism. Cell Metab. 2015, 22, 838–850. [Google Scholar] [CrossRef] [PubMed]
  82. Axelrod, C.L.; Fealy, C.E.; Mulya, A.; Kirwan, J.P. Exercise training remodels human skeletal muscle mitochondrial fission and fusion machinery towards a pro-elongation phenotype. Acta Physiol. 2019, 225, e13216. [Google Scholar] [CrossRef] [PubMed]
  83. Patten, D.A.; Wong, J.; Khacho, M.; Soubannier, V.; Mailloux, R.J.; Pilon-Larose, K.; MacLaurin, J.G.; Park, D.S.; McBride, H.M.; Trinkle-Mulcahy, L.; et al. OPA1-dependent cristae modulation is essential for cellular adaptation to metabolic demand. EMBO J. 2014, 33, 2676–2691. [Google Scholar] [CrossRef] [PubMed]
  84. Cartoni, R.; Leger, B.; Hock, M.B.; Praz, M.; Crettenand, A.; Pich, S.; Ziltener, J.L.; Luthi, F.; Deriaz, O.; Zorzano, A.; et al. Mitofusins 1/2 and ERRalpha expression are increased in human skeletal muscle after physical exercise. J. Physiol. 2005, 567, 349–358. [Google Scholar] [CrossRef] [PubMed]
  85. Dohl, J.; Foldi, J.; Heller, J.; Gasier, H.G.; Deuster, P.A.; Yu, T. Acclimation of C2C12 myoblasts to physiological glucose concentrations for in vitro diabetes research. Life Sci. 2018, 211, 238–244. [Google Scholar] [CrossRef]
  86. Yu, T.; Robotham, J.L.; Yoon, Y. Increased production of reactive oxygen species in hyperglycemic conditions requires dynamic change of mitochondrial morphology. Proc. Natl. Acad. Sci. USA 2006, 103, 2653–2658. [Google Scholar] [CrossRef] [PubMed]
  87. Cioffi, F.; Giacco, A.; Petito, G.; de Matteis, R.; Senese, R.; Lombardi, A.; de Lange, P.; Moreno, M.; Goglia, F.; Lanni, A.; et al. Altered Mitochondrial Quality Control in Rats with Metabolic Dysfunction-Associated Fatty Liver Disease (MAFLD) Induced by High-Fat Feeding. Genes 2022, 13, 315. [Google Scholar] [CrossRef]
  88. Petito, G.; Giacco, A.; Cioffi, F.; Mazzoli, A.; Magnacca, N.; Iossa, S.; Goglia, F.; Senese, R.; Lanni, A. Short-term fructose feeding alters tissue metabolic pathways by modulating microRNAs expression both in young and adult rats. Front. Cell Dev. Biol. 2023, 11, 1101844. [Google Scholar] [CrossRef]
  89. Wang, W.; Wang, Y.; Long, J.; Wang, J.; Haudek, S.B.; Overbeek, P.; Chang, B.H.; Schumacker, P.T.; Danesh, F.R. Mitochondrial fission triggered by hyperglycemia is mediated by ROCK1 activation in podocytes and endothelial cells. Cell Metab. 2012, 15, 186–200. [Google Scholar] [CrossRef]
  90. Wang, L.; Yu, T.; Lee, H.; O’Brien, D.K.; Sesaki, H.; Yoon, Y. Decreasing mitochondrial fission diminishes vascular smooth muscle cell migration and ameliorates intimal hyperplasia. Cardiovasc. Res. 2015, 106, 272–283. [Google Scholar] [CrossRef]
  91. Yu, T.; Wang, L.; Lee, H.; O’Brien, D.K.; Bronk, S.F.; Gores, G.J.; Yoon, Y. Decreasing mitochondrial fission prevents cholestatic liver injury. J. Biol. Chem. 2014, 289, 34074–34088. [Google Scholar] [CrossRef] [PubMed]
  92. Galloway, C.A.; Lee, H.; Nejjar, S.; Jhun, B.S.; Yu, T.; Hsu, W.; Yoon, Y. Transgenic control of mitochondrial fission induces mitochondrial uncoupling and relieves diabetic oxidative stress. Diabetes 2012, 61, 2093–2104. [Google Scholar] [CrossRef]
  93. Makino, A.; Scott, B.T.; Dillmann, W.H. Mitochondrial fragmentation and superoxide anion production in coronary endothelial cells from a mouse model of type 1 diabetes. Diabetologia 2010, 53, 1783–1794. [Google Scholar] [CrossRef] [PubMed]
  94. Kelley, D.E.; He, J.; Menshikova, E.V.; Ritov, V.B. Dysfunction of mitochondria in human skeletal muscle in type 2 diabetes. Diabetes 2002, 51, 2944–2950. [Google Scholar] [CrossRef] [PubMed]
  95. Lee, H.; Smith, S.B.; Yoon, Y. The short variant of the mitochondrial dynamin OPA1 maintains mitochondrial energetics and cristae structure. J. Biol. Chem. 2017, 292, 7115–7130. [Google Scholar] [CrossRef]
  96. Sessions, D.T.; Kim, K.B.; Kashatus, J.A.; Churchill, N.; Park, K.S.; Mayo, M.W.; Sesaki, H.; Kashatus, D.F. Opa1 and Drp1 reciprocally regulate cristae morphology, ETC function, and NAD+ regeneration in KRas-mutant lung adenocarcinoma. Cell Rep. 2022, 41, 111818. [Google Scholar] [CrossRef]
  97. Frezza, C.; Cipolat, S.; Martins de Brito, O.; Micaroni, M.; Beznoussenko, G.V.; Rudka, T.; Bartoli, D.; Polishuck, R.S.; Danial, N.N.; De Strooper, B.; et al. OPA1 controls apoptotic cristae remodeling independently from mitochondrial fusion. Cell 2006, 126, 177–189. [Google Scholar] [CrossRef]
  98. Hu, J.; Zhang, Y.; Jiang, X.; Zhang, H.; Gao, Z.; Li, Y.; Fu, R.; Li, L.; Li, J.; Cui, H.; et al. ROS-mediated activation and mitochondrial translocation of CaMKII contributes to Drp1-dependent mitochondrial fission and apoptosis in triple-negative breast cancer cells by isorhamnetin and chloroquine. J. Exp. Clin. Cancer Res. 2019, 38, 225. [Google Scholar] [CrossRef]
  99. Zorov, D.B.; Juhaszova, M.; Sollott, S.J. Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release. Physiol. Rev. 2014, 94, 909–950. [Google Scholar] [CrossRef]
  100. Apostolova, N.; Victor, V.M. Molecular strategies for targeting antioxidants to mitochondria: Therapeutic implications. Antioxid. Redox Signal. 2015, 22, 686–729. [Google Scholar] [CrossRef]
  101. Bogenhagen, D.F. Mitochondrial DNA nucleoid structure. Biochim. Biophys. Acta 2012, 1819, 914–920. [Google Scholar] [CrossRef]
  102. Ademowo, O.S.; Dias, H.K.I.; Burton, D.G.A.; Griffiths, H.R. Lipid (per) oxidation in mitochondria: An emerging target in the ageing process? Biogerontology 2017, 18, 859–879. [Google Scholar] [CrossRef]
  103. Paradies, G.; Paradies, V.; Ruggiero, F.M.; Petrosillo, G. Oxidative stress, cardiolipin and mitochondrial dysfunction in nonalcoholic fatty liver disease. World J. Gastroenterol. 2014, 20, 14205–14218. [Google Scholar] [CrossRef] [PubMed]
  104. Panov, A.V.; Dikalov, S.I. Cardiolipin, Perhydroxyl Radicals, and Lipid Peroxidation in Mitochondrial Dysfunctions and Aging. Oxidative Med. Cell. Longev. 2020, 2020, 1323028. [Google Scholar] [CrossRef] [PubMed]
  105. Yu, T.; Sheu, S.S.; Robotham, J.L.; Yoon, Y. Mitochondrial fission mediates high glucose-induced cell death through elevated production of reactive oxygen species. Cardiovasc. Res. 2008, 79, 341–351. [Google Scholar] [CrossRef] [PubMed]
  106. Dai, W.; Jiang, L. Dysregulated Mitochondrial Dynamics and Metabolism in Obesity, Diabetes, and Cancer. Front. Endocrinol. 2019, 10, 570. [Google Scholar] [CrossRef]
  107. Poznyak, A.V.; Kirichenko, T.V.; Borisov, E.E.; Shakhpazyan, N.K.; Kartuesov, A.G.; Orekhov, A.N. Mitochondrial Implications in Cardiovascular Aging and Diseases: The Specific Role of Mitochondrial Dynamics and Shifts. Int. J. Mol. Sci. 2022, 23, 2951. [Google Scholar] [CrossRef] [PubMed]
  108. Jin, J.Y.; Wei, X.X.; Zhi, X.L.; Wang, X.H.; Meng, D. Drp1-dependent mitochondrial fission in cardiovascular disease. Acta Pharmacol. Sin. 2021, 42, 655–664. [Google Scholar] [CrossRef] [PubMed]
  109. Forte, M.; Schirone, L.; Ameri, P.; Basso, C.; Catalucci, D.; Modica, J.; Chimenti, C.; Crotti, L.; Frati, G.; Rubattu, S.; et al. The role of mitochondrial dynamics in cardiovascular diseases. Br. J. Pharmacol. 2021, 178, 2060–2076. [Google Scholar] [CrossRef]
  110. Williams, M.; Caino, M.C. Mitochondrial Dynamics in Type 2 Diabetes and Cancer. Front. Endocrinol. 2018, 9, 211. [Google Scholar] [CrossRef]
  111. Kyriazis, I.D.; Vassi, E.; Alvanou, M.; Angelakis, C.; Skaperda, Z.; Tekos, F.; Garikipati, V.N.S.; Spandidos, D.A.; Kouretas, D. The impact of diet upon mitochondrial physiology (Review). Int. J. Mol. Med. 2022, 50, 135. [Google Scholar] [CrossRef] [PubMed]
  112. Palacios, O.M.; Carmona, J.J.; Michan, S.; Chen, K.Y.; Manabe, Y.; Ward, J.L., 3rd; Goodyear, L.J.; Tong, Q. Diet and exercise signals regulate SIRT3 and activate AMPK and PGC-1alpha in skeletal muscle. Aging 2009, 1, 771–783. [Google Scholar] [CrossRef] [PubMed]
  113. Gurd, B.J.; Menezes, E.S.; Arhen, B.B.; Islam, H. Impacts of altered exercise volume, intensity, and duration on the activation of AMPK and CaMKII and increases in PGC-1alpha mRNA. Semin. Cell Dev. Biol. 2023, 143, 17–27. [Google Scholar] [CrossRef]
  114. Gomez-Cabrera, M.C.; Domenech, E.; Vina, J. Moderate exercise is an antioxidant: Upregulation of antioxidant genes by training. Free Radic. Biol. Med. 2008, 44, 126–131. [Google Scholar] [CrossRef] [PubMed]
  115. Mooren, F.C.; Kruger, K. Exercise, Autophagy, and Apoptosis. Prog. Mol. Biol. Transl. Sci. 2015, 135, 407–422. [Google Scholar] [CrossRef] [PubMed]
  116. Wang, L.; Wang, J.; Cretoiu, D.; Li, G.; Xiao, J. Exercise-mediated regulation of autophagy in the cardiovascular system. J. Sport Health Sci. 2020, 9, 203–210. [Google Scholar] [CrossRef]
  117. Trewin, A.J.; Berry, B.J.; Wojtovich, A.P. Exercise and Mitochondrial Dynamics: Keeping in Shape with ROS and AMPK. Antioxidants 2018, 7, 7. [Google Scholar] [CrossRef]
  118. Henriquez-Olguin, C.; Renani, L.B.; Arab-Ceschia, L.; Raun, S.H.; Bhatia, A.; Li, Z.; Knudsen, J.R.; Holmdahl, R.; Jensen, T.E. Adaptations to high-intensity interval training in skeletal muscle require NADPH oxidase 2. Redox Biol. 2019, 24, 101188. [Google Scholar] [CrossRef]
  119. Li, G.; Xu, K.; Xing, W.; Yang, H.; Li, Y.; Wang, X.; Zhou, J.; An, J.; Dong, L.; Zhang, X.; et al. Swimming Exercise Alleviates Endothelial Mitochondrial Fragmentation via Inhibiting Dynamin-Related Protein-1 to Improve Vascular Function in Hypertension. Hypertension 2022, 79, e116–e128. [Google Scholar] [CrossRef]
  120. Campos, J.C.; Marchesi Bozi, L.H.; Krum, B.; Grassmann Bechara, L.R.; Ferreira, N.D.; Arini, G.S.; Albuquerque, R.P.; Traa, A.; Ogawa, T.; van der Bliek, A.M.; et al. Exercise preserves physical fitness during aging through AMPK and mitochondrial dynamics. Proc. Natl. Acad. Sci. USA 2023, 120, e2204750120. [Google Scholar] [CrossRef]
  121. de Las Heras, N.; Klett-Mingo, M.; Ballesteros, S.; Martin-Fernandez, B.; Escribano, O.; Blanco-Rivero, J.; Balfagon, G.; Hribal, M.L.; Benito, M.; Lahera, V.; et al. Chronic Exercise Improves Mitochondrial Function and Insulin Sensitivity in Brown Adipose Tissue. Front. Physiol. 2018, 9, 1122. [Google Scholar] [CrossRef] [PubMed]
  122. Savencu, C.E.; Linta, A.; Farcas, G.; Bina, A.M.; Cretu, O.M.; Malita, D.C.; Muntean, D.M.; Sturza, A. Impact of Dietary Restriction Regimens on Mitochondria, Heart, and Endothelial Function: A Brief Overview. Front. Physiol. 2021, 12, 768383. [Google Scholar] [CrossRef] [PubMed]
  123. Serrano, J.C.E.; Cassanye, A.; Martin-Gari, M.; Granado-Serrano, A.B.; Portero-Otin, M. Effect of Dietary Bioactive Compounds on Mitochondrial and Metabolic Flexibility. Diseases 2016, 4, 14. [Google Scholar] [CrossRef]
  124. Khalil, M.; Shanmugam, H.; Abdallah, H.; John Britto, J.S.; Galerati, I.; Gomez-Ambrosi, J.; Fruhbeck, G.; Portincasa, P. The Potential of the Mediterranean Diet to Improve Mitochondrial Function in Experimental Models of Obesity and Metabolic Syndrome. Nutrients 2022, 14, 3112. [Google Scholar] [CrossRef]
  125. Huang, X.; Liu, G.; Guo, J.; Su, Z. The PI3K/AKT pathway in obesity and type 2 diabetes. Int. J. Biol. Sci. 2018, 14, 1483–1496. [Google Scholar] [CrossRef]
  126. Lepretti, M.; Martucciello, S.; Burgos Aceves, M.A.; Putti, R.; Lionetti, L. Omega-3 Fatty Acids and Insulin Resistance: Focus on the Regulation of Mitochondria and Endoplasmic Reticulum Stress. Nutrients 2018, 10, 350. [Google Scholar] [CrossRef] [PubMed]
  127. Herbst, E.A.; Paglialunga, S.; Gerling, C.; Whitfield, J.; Mukai, K.; Chabowski, A.; Heigenhauser, G.J.; Spriet, L.L.; Holloway, G.P. Omega-3 supplementation alters mitochondrial membrane composition and respiration kinetics in human skeletal muscle. J. Physiol. 2014, 592, 1341–1352. [Google Scholar] [CrossRef]
  128. Picard, M.; McEwen, B.S. Psychological Stress and Mitochondria: A Conceptual Framework. Psychosom. Med. 2018, 80, 126–140. [Google Scholar] [CrossRef]
  129. Daniels, T.E.; Olsen, E.M.; Tyrka, A.R. Stress and Psychiatric Disorders: The Role of Mitochondria. Annu. Rev. Clin. Psychol. 2020, 16, 165–186. [Google Scholar] [CrossRef]
  130. Picard, M.; McEwen, B.S. Psychological Stress and Mitochondria: A Systematic Review. Psychosom. Med. 2018, 80, 141–153. [Google Scholar] [CrossRef]
  131. Du, J.; Wang, Y.; Hunter, R.; Wei, Y.; Blumenthal, R.; Falke, C.; Khairova, R.; Zhou, R.; Yuan, P.; Machado-Vieira, R.; et al. Dynamic regulation of mitochondrial function by glucocorticoids. Proc. Natl. Acad. Sci. USA 2009, 106, 3543–3548. [Google Scholar] [CrossRef]
  132. Hernandez-Alvarez, M.I.; Paz, J.C.; Sebastian, D.; Munoz, J.P.; Liesa, M.; Segales, J.; Palacin, M.; Zorzano, A. Glucocorticoid modulation of mitochondrial function in hepatoma cells requires the mitochondrial fission protein Drp1. Antioxid. Redox Signal. 2013, 19, 366–378. [Google Scholar] [CrossRef] [PubMed]
  133. Fernandez-Sanchez, A.; Madrigal-Santillan, E.; Bautista, M.; Esquivel-Soto, J.; Morales-Gonzalez, A.; Esquivel-Chirino, C.; Durante-Montiel, I.; Sanchez-Rivera, G.; Valadez-Vega, C.; Morales-Gonzalez, J.A. Inflammation, oxidative stress, and obesity. Int. J. Mol. Sci. 2011, 12, 3117–3132. [Google Scholar] [CrossRef] [PubMed]
  134. Delmotte, P.; Marin Mathieu, N.; Sieck, G.C. TNFalpha induces mitochondrial fragmentation and biogenesis in human airway smooth muscle. Am. J. Physiol. Lung Cell. Mol. Physiol. 2021, 320, L137–L151. [Google Scholar] [CrossRef] [PubMed]
  135. White, J.P.; Puppa, M.J.; Sato, S.; Gao, S.; Price, R.L.; Baynes, J.W.; Kostek, M.C.; Matesic, L.E.; Carson, J.A. IL-6 regulation on skeletal muscle mitochondrial remodeling during cancer cachexia in the ApcMin/+ mouse. Skelet. Muscle 2012, 2, 14. [Google Scholar] [CrossRef]
  136. Abid, H.; Ryan, Z.C.; Delmotte, P.; Sieck, G.C.; Lanza, I.R. Extramyocellular interleukin-6 influences skeletal muscle mitochondrial physiology through canonical JAK/STAT signaling pathways. FASEB J. 2020, 34, 14458–14472. [Google Scholar] [CrossRef]
  137. Hernandez-Camacho, J.D.; Bernier, M.; Lopez-Lluch, G.; Navas, P. Coenzyme Q10 Supplementation in Aging and Disease. Front. Physiol. 2018, 9, 44. [Google Scholar] [CrossRef]
  138. Cirilli, I.; Damiani, E.; Dludla, P.V.; Hargreaves, I.; Marcheggiani, F.; Millichap, L.E.; Orlando, P.; Silvestri, S.; Tiano, L. Role of Coenzyme Q10 in Health and Disease: An Update on the Last 10 Years (2010–2020). Antioxidants 2021, 10, 1325. [Google Scholar] [CrossRef]
  139. Xie, T.; Wang, C.; Jin, Y.; Meng, Q.; Liu, Q.; Wu, J.; Sun, H. CoenzymeQ10-Induced Activation of AMPK-YAP-OPA1 Pathway Alleviates Atherosclerosis by Improving Mitochondrial Function, Inhibiting Oxidative Stress and Promoting Energy Metabolism. Front. Pharmacol. 2020, 11, 1034. [Google Scholar] [CrossRef]
  140. Li, H.N.; Zimmerman, M.; Milledge, G.Z.; Hou, X.L.; Cheng, J.; Wang, Z.H.; Li, P.A. Water-Soluble Coenzyme Q10 Reduces Rotenone-Induced Mitochondrial Fission. Neurochem. Res. 2017, 42, 1096–1103. [Google Scholar] [CrossRef]
  141. Nakazawa, H.; Ikeda, K.; Shinozaki, S.; Yasuhara, S.; Yu, Y.M.; Martyn, J.A.J.; Tompkins, R.G.; Yorozu, T.; Inoue, S.; Kaneki, M. Coenzyme Q10 protects against burn-induced mitochondrial dysfunction and impaired insulin signaling in mouse skeletal muscle. FEBS Open Bio 2019, 9, 348–363. [Google Scholar] [CrossRef] [PubMed]
  142. Xu, Z.; Huo, J.; Ding, X.; Yang, M.; Li, L.; Dai, J.; Hosoe, K.; Kubo, H.; Mori, M.; Higuchi, K.; et al. Coenzyme Q10 Improves Lipid Metabolism and Ameliorates Obesity by Regulating CaMKII-Mediated PDE4 Inhibition. Sci. Rep. 2017, 7, 8253. [Google Scholar] [CrossRef] [PubMed]
  143. Shen, Q.; Pierce, J.D. Supplementation of Coenzyme Q10 among Patients with Type 2 Diabetes Mellitus. Healthcare 2015, 3, 296–309. [Google Scholar] [CrossRef] [PubMed]
  144. Alam, M.A.; Rahman, M.M. Mitochondrial dysfunction in obesity: Potential benefit and mechanism of Co-enzyme Q10 supplementation in metabolic syndrome. J. Diabetes Metab. Disord. 2014, 13, 60. [Google Scholar] [CrossRef] [PubMed]
  145. Zheng, M.; Bai, Y.; Sun, X.; Fu, R.; Liu, L.; Liu, M.; Li, Z.; Huang, X. Resveratrol Reestablishes Mitochondrial Quality Control in Myocardial Ischemia/Reperfusion Injury through Sirt1/Sirt3-Mfn2-Parkin-PGC-1alpha Pathway. Molecules 2022, 27, 5545. [Google Scholar] [CrossRef]
  146. Peng, K.; Tao, Y.; Zhang, J.; Wang, J.; Ye, F.; Dan, G.; Zhao, Y.; Cai, Y.; Zhao, J.; Wu, Q.; et al. Resveratrol Regulates Mitochondrial Biogenesis and Fission/Fusion to Attenuate Rotenone-Induced Neurotoxicity. Oxid. Med. Cell. Longev. 2016, 2016, 6705621. [Google Scholar] [CrossRef]
  147. Iside, C.; Scafuro, M.; Nebbioso, A.; Altucci, L. SIRT1 Activation by Natural Phytochemicals: An Overview. Front. Pharmacol. 2020, 11, 1225. [Google Scholar] [CrossRef]
  148. Lagouge, M.; Argmann, C.; Gerhart-Hines, Z.; Meziane, H.; Lerin, C.; Daussin, F.; Messadeq, N.; Milne, J.; Lambert, P.; Elliott, P.; et al. Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1alpha. Cell 2006, 127, 1109–1122. [Google Scholar] [CrossRef]
  149. Robb, E.L.; Moradi, F.; Maddalena, L.A.; Valente, A.J.F.; Fonseca, J.; Stuart, J.A. Resveratrol stimulates mitochondrial fusion by a mechanism requiring mitofusin-2. Biochem. Biophys. Res. Commun. 2017, 485, 249–254. [Google Scholar] [CrossRef]
  150. Hou, C.Y.; Tain, Y.L.; Yu, H.R.; Huang, L.T. The Effects of Resveratrol in the Treatment of Metabolic Syndrome. Int J Mol Sci 2019, 20, 535. [Google Scholar] [CrossRef]
  151. Zheng, J.; Chen, L.L.; Zhang, H.H.; Hu, X.; Kong, W.; Hu, D. Resveratrol improves insulin resistance of catch-up growth by increasing mitochondrial complexes and antioxidant function in skeletal muscle. Metabolism 2012, 61, 954–965. [Google Scholar] [CrossRef] [PubMed]
  152. Xia, N.; Daiber, A.; Forstermann, U.; Li, H. Antioxidant effects of resveratrol in the cardiovascular system. Br. J. Pharmacol. 2017, 174, 1633–1646. [Google Scholar] [CrossRef] [PubMed]
  153. Mongioi, L.M.; La Vignera, S.; Cannarella, R.; Cimino, L.; Compagnone, M.; Condorelli, R.A.; Calogero, A.E. The Role of Resveratrol Administration in Human Obesity. Int. J. Mol. Sci. 2021, 22, 4362. [Google Scholar] [CrossRef] [PubMed]
  154. Su, M.; Zhao, W.; Xu, S.; Weng, J. Resveratrol in Treating Diabetes and Its Cardiovascular Complications: A Review of Its Mechanisms of Action. Antioxidants 2022, 11, 1085. [Google Scholar] [CrossRef] [PubMed]
  155. Yu, T.; Dohl, J.; Chen, Y.; Gasier, H.G.; Deuster, P.A. Astaxanthin but not quercetin preserves mitochondrial integrity and function, ameliorates oxidative stress, and reduces heat-induced skeletal muscle injury. J. Cell. Physiol. 2019, 234, 13292–13302. [Google Scholar] [CrossRef]
  156. Chen, Y.; Yu, T.; Deuster, P. Astaxanthin Protects Against Heat-induced Mitochondrial Alterations in Mouse Hypothalamus. Neuroscience 2021, 476, 12–20. [Google Scholar] [CrossRef]
  157. Huang, C.; Wen, C.; Yang, M.; Li, A.; Fan, C.; Gan, D.; Li, Q.; Zhao, J.; Zhu, L.; Lu, D. Astaxanthin Improved the Cognitive Deficits in APP/PS1 Transgenic Mice Via Selective Activation of mTOR. J. Neuroimmune Pharmacol. 2021, 16, 609–619. [Google Scholar] [CrossRef]
  158. Chen, Y.; Li, S.; Guo, Y.; Yu, H.; Bao, Y.; Xin, X.; Yang, H.; Ni, X.; Wu, N.; Jia, D. Astaxanthin Attenuates Hypertensive Vascular Remodeling by Protecting Vascular Smooth Muscle Cells from Oxidative Stress-Induced Mitochondrial Dysfunction. Oxidative Med. Cell. Longev. 2020, 2020, 4629189. [Google Scholar] [CrossRef]
  159. Ma, L.; Liu, S.; Zhen, X.; Qiao, W.; Ma, L.; Zhang, X. Astaxanthin delayed the pathogenesis of diabetic nephropathy in type 1 diabetic rats. BIOCELL 2022, 46, 1911–1916. [Google Scholar] [CrossRef]
  160. Nishida, Y.; Nawaz, A.; Kado, T.; Takikawa, A.; Igarashi, Y.; Onogi, Y.; Wada, T.; Sasaoka, T.; Yamamoto, S.; Sasahara, M.; et al. Astaxanthin stimulates mitochondrial biogenesis in insulin resistant muscle via activation of AMPK pathway. J. Cachexia Sarcopenia Muscle 2020, 11, 241–258. [Google Scholar] [CrossRef]
  161. Nishida, Y.; Nawaz, A.; Hecht, K.; Tobe, K. Astaxanthin as a Novel Mitochondrial Regulator: A New Aspect of Carotenoids, beyond Antioxidants. Nutrients 2021, 14, 107. [Google Scholar] [CrossRef] [PubMed]
  162. Leung, L.Y.; Chan, S.M.; Tam, H.L.; Wong, E.S. Astaxanthin Influence on Health Outcomes of Adults at Risk of Metabolic Syndrome: A Systematic Review and Meta-Analysis. Nutrients 2022, 14, 2050. [Google Scholar] [CrossRef] [PubMed]
  163. Yu, T.; Dohl, J.; Elenberg, F.; Chen, Y.; Deuster, P. Curcumin induces concentration-dependent alterations in mitochondrial function through ROS in C2C12 mouse myoblasts. J. Cell. Physiol. 2019, 234, 6371–6381. [Google Scholar] [CrossRef] [PubMed]
  164. Eckert, G.P.; Schiborr, C.; Hagl, S.; Abdel-Kader, R.; Muller, W.E.; Rimbach, G.; Frank, J. Curcumin prevents mitochondrial dysfunction in the brain of the senescence-accelerated mouse-prone 8. Neurochem. Int. 2013, 62, 595–602. [Google Scholar] [CrossRef] [PubMed]
  165. Reddy, P.H.; Manczak, M.; Yin, X.; Grady, M.C.; Mitchell, A.; Kandimalla, R.; Kuruva, C.S. Protective effects of a natural product, curcumin, against amyloid beta induced mitochondrial and synaptic toxicities in Alzheimer’s disease. J. Investig. Med. 2016, 64, 1220–1234. [Google Scholar] [CrossRef] [PubMed]
  166. Jabczyk, M.; Nowak, J.; Hudzik, B.; Zubelewicz-Szkodzinska, B. Curcumin in Metabolic Health and Disease. Nutrients 2021, 13, 4440. [Google Scholar] [CrossRef] [PubMed]
  167. Nurcahyanti, A.D.R.; Cokro, F.; Wulanjati, M.P.; Mahmoud, M.F.; Wink, M.; Sobeh, M. Curcuminoids for Metabolic Syndrome: Meta-Analysis Evidences Toward Personalized Prevention and Treatment Management. Front. Nutr. 2022, 9, 891339. [Google Scholar] [CrossRef]
  168. Lee, D.S.; Kim, J.E. PDI-mediated S-nitrosylation of DRP1 facilitates DRP1-S616 phosphorylation and mitochondrial fission in CA1 neurons. Cell Death Dis. 2018, 9, 869. [Google Scholar] [CrossRef]
  169. Yu, T.; Park, Y.M.; Wang, L.; Deuster, P.A. L-citrulline prevents heat-induced mitochondrial dysfunction and cell injury through nitric oxide-mediated Drp1 inhibition in mouse C2C12 myoblasts. Br. J. Nutr. 2022, 129, 936–946. [Google Scholar] [CrossRef]
  170. Allerton, T.D.; Proctor, D.N.; Stephens, J.M.; Dugas, T.R.; Spielmann, G.; Irving, B.A. l-Citrulline Supplementation: Impact on Cardiometabolic Health. Nutrients 2018, 10, 921. [Google Scholar] [CrossRef]
  171. Eshreif, A.; Al Batran, R.; Jamieson, K.L.; Darwesh, A.M.; Gopal, K.; Greenwell, A.A.; Zlobine, I.; Aburasayn, H.; Eaton, F.; Mulvihill, E.E.; et al. l-Citrulline supplementation improves glucose and exercise tolerance in obese male mice. Exp. Physiol. 2020, 105, 270–281. [Google Scholar] [CrossRef] [PubMed]
  172. Cassidy-Stone, A.; Chipuk, J.E.; Ingerman, E.; Song, C.; Yoo, C.; Kuwana, T.; Kurth, M.J.; Shaw, J.T.; Hinshaw, J.E.; Green, D.R.; et al. Chemical inhibition of the mitochondrial division dynamin reveals its role in Bax/Bak-dependent mitochondrial outer membrane permeabilization. Dev. Cell 2008, 14, 193–204. [Google Scholar] [CrossRef] [PubMed]
  173. Yu, T.; Ferdjallah, I.; Elenberg, F.; Chen, S.K.; Deuster, P.; Chen, Y. Mitochondrial fission contributes to heat-induced oxidative stress in skeletal muscle but not hyperthermia in mice. Life Sci. 2018, 200, 6–14. [Google Scholar] [CrossRef] [PubMed]
  174. Finocchietto, P.; Perez, H.; Blanco, G.; Miksztowicz, V.; Marotte, C.; Morales, C.; Peralta, J.; Berg, G.; Poderoso, C.; Poderoso, J.J.; et al. Inhibition of mitochondrial fission by Drp-1 blockade by short-term leptin and Mdivi-1 treatment improves white adipose tissue abnormalities in obesity and diabetes. Pharmacol. Res. 2022, 178, 106028. [Google Scholar] [CrossRef]
  175. Kugler, B.A.; Deng, W.; Duguay, A.L.; Garcia, J.P.; Anderson, M.C.; Nguyen, P.D.; Houmard, J.A.; Zou, K. Pharmacological inhibition of dynamin-related protein 1 attenuates skeletal muscle insulin resistance in obesity. Physiol. Rep. 2021, 9, e14808. [Google Scholar] [CrossRef]
  176. Jheng, H.F.; Tsai, P.J.; Guo, S.M.; Kuo, L.H.; Chang, C.S.; Su, I.J.; Chang, C.R.; Tsai, Y.S. Mitochondrial fission contributes to mitochondrial dysfunction and insulin resistance in skeletal muscle. Mol. Cell. Biol. 2012, 32, 309–319. [Google Scholar] [CrossRef]
  177. Ding, M.; Dong, Q.; Liu, Z.; Liu, Z.; Qu, Y.; Li, X.; Huo, C.; Jia, X.; Fu, F.; Wang, X. Inhibition of dynamin-related protein 1 protects against myocardial ischemia-reperfusion injury in diabetic mice. Cardiovasc. Diabetol. 2017, 16, 19. [Google Scholar] [CrossRef]
  178. Liu, X.; Zhang, X.; Niu, X.; Zhang, P.; Wang, Q.; Xue, X.; Song, G.; Yu, J.; Xi, G.; Song, L.; et al. Mdivi-1 Modulates Macrophage/Microglial Polarization in Mice with EAE via the Inhibition of the TLR2/4-GSK3beta-NF-kappaB Inflammatory Signaling Axis. Mol. Neurobiol. 2022, 59, 1–16. [Google Scholar] [CrossRef]
  179. Li, Y.H.; Xu, F.; Thome, R.; Guo, M.F.; Sun, M.L.; Song, G.B.; Li, R.L.; Chai, Z.; Ciric, B.; Rostami, A.M.; et al. Mdivi-1, a mitochondrial fission inhibitor, modulates T helper cells and suppresses the development of experimental autoimmune encephalomyelitis. J. Neuroinflamm. 2019, 16, 149. [Google Scholar] [CrossRef]
  180. Duan, C.; Wang, L.; Zhang, J.; Xiang, X.; Wu, Y.; Zhang, Z.; Li, Q.; Tian, K.; Xue, M.; Liu, L.; et al. Mdivi-1 attenuates oxidative stress and exerts vascular protection in ischemic/hypoxic injury by a mechanism independent of Drp1 GTPase activity. Redox Biol. 2020, 37, 101706. [Google Scholar] [CrossRef]
  181. Aishwarya, R.; Alam, S.; Abdullah, C.S.; Morshed, M.; Nitu, S.S.; Panchatcharam, M.; Miriyala, S.; Kevil, C.G.; Bhuiyan, M.S. Pleiotropic effects of mdivi-1 in altering mitochondrial dynamics, respiration, and autophagy in cardiomyocytes. Redox Biol. 2020, 36, 101660. [Google Scholar] [CrossRef] [PubMed]
  182. Qi, X.; Qvit, N.; Su, Y.C.; Mochly-Rosen, D. A novel Drp1 inhibitor diminishes aberrant mitochondrial fission and neurotoxicity. J. Cell Sci. 2013, 126, 789–802. [Google Scholar] [CrossRef] [PubMed]
  183. Tian, L.; Neuber-Hess, M.; Mewburn, J.; Dasgupta, A.; Dunham-Snary, K.; Wu, D.; Chen, K.H.; Hong, Z.; Sharp, W.W.; Kutty, S.; et al. Ischemia-induced Drp1 and Fis1-mediated mitochondrial fission and right ventricular dysfunction in pulmonary hypertension. J. Mol. Med. 2017, 95, 381–393. [Google Scholar] [CrossRef] [PubMed]
  184. Haileselassie, B.; Mukherjee, R.; Joshi, A.U.; Napier, B.A.; Massis, L.M.; Ostberg, N.P.; Queliconi, B.B.; Monack, D.; Bernstein, D.; Mochly-Rosen, D. Drp1/Fis1 interaction mediates mitochondrial dysfunction in septic cardiomyopathy. J. Mol. Cell. Cardiol. 2019, 130, 160–169. [Google Scholar] [CrossRef]
  185. Joshi, A.U.; Ebert, A.E.; Haileselassie, B.; Mochly-Rosen, D. Drp1/Fis1-mediated mitochondrial fragmentation leads to lysosomal dysfunction in cardiac models of Huntington’s disease. J. Mol. Cell. Cardiol. 2019, 127, 125–133. [Google Scholar] [CrossRef]
  186. Joshi, A.U.; Saw, N.L.; Vogel, H.; Cunnigham, A.D.; Shamloo, M.; Mochly-Rosen, D. Inhibition of Drp1/Fis1 interaction slows progression of amyotrophic lateral sclerosis. EMBO Mol. Med. 2018, 10, e8166. [Google Scholar] [CrossRef]
  187. Luo, F.; Herrup, K.; Qi, X.; Yang, Y. Inhibition of Drp1 hyper-activation is protective in animal models of experimental multiple sclerosis. Exp. Neurol. 2017, 292, 21–34. [Google Scholar] [CrossRef]
  188. Guo, X.; Sesaki, H.; Qi, X. Drp1 stabilizes p53 on the mitochondria to trigger necrosis under oxidative stress conditions in vitro and in vivo. Biochem. J. 2014, 461, 137–146. [Google Scholar] [CrossRef]
  189. Su, Y.C.; Qi, X. Inhibition of excessive mitochondrial fission reduced aberrant autophagy and neuronal damage caused by LRRK2 G2019S mutation. Hum. Mol. Genet. 2013, 22, 4545–4561. [Google Scholar] [CrossRef]
  190. Rovira-Llopis, S.; Banuls, C.; Diaz-Morales, N.; Hernandez-Mijares, A.; Rocha, M.; Victor, V.M. Mitochondrial dynamics in type 2 diabetes: Pathophysiological implications. Redox Biol. 2017, 11, 637–645. [Google Scholar] [CrossRef]
  191. Bhatti, J.S.; Bhatti, G.K.; Reddy, P.H. Mitochondrial dysfunction and oxidative stress in metabolic disorders—A step towards mitochondria based therapeutic strategies. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2017, 1863, 1066–1077. [Google Scholar] [CrossRef] [PubMed]
  192. Macia, E.; Ehrlich, M.; Massol, R.; Boucrot, E.; Brunner, C.; Kirchhausen, T. Dynasore, a cell-permeable inhibitor of dynamin. Dev. Cell 2006, 10, 839–850. [Google Scholar] [CrossRef] [PubMed]
  193. Ishiki, M.; Klip, A. Minireview: Recent developments in the regulation of glucose transporter-4 traffic: New signals, locations, and partners. Endocrinology 2005, 146, 5071–5078. [Google Scholar] [CrossRef] [PubMed]
  194. Merz, K.E.; Thurmond, D.C. Role of Skeletal Muscle in Insulin Resistance and Glucose Uptake. Compr. Physiol. 2020, 10, 785–809. [Google Scholar] [CrossRef]
  195. Hall, C.; Yu, H.; Choi, E. Insulin receptor endocytosis in the pathophysiology of insulin resistance. Exp. Mol. Med. 2020, 52, 911–920. [Google Scholar] [CrossRef]
  196. Liu, Y.; Zhou, D.; Abumrad, N.A.; Su, X. ADP-ribosylation factor 6 modulates adrenergic stimulated lipolysis in adipocytes. Am. J. Physiol. Cell Physiol. 2010, 298, C921–C928. [Google Scholar] [CrossRef]
  197. Girard, E.; Paul, J.L.; Fournier, N.; Beaune, P.; Johannes, L.; Lamaze, C.; Vedie, B. The dynamin chemical inhibitor dynasore impairs cholesterol trafficking and sterol-sensitive genes transcription in human HeLa cells and macrophages. PLoS ONE 2011, 6, e29042. [Google Scholar] [CrossRef]
  198. Gao, D.; Zhang, L.; Dhillon, R.; Hong, T.T.; Shaw, R.M.; Zhu, J. Dynasore protects mitochondria and improves cardiac lusitropy in Langendorff perfused mouse heart. PLoS ONE 2013, 8, e60967. [Google Scholar] [CrossRef]
  199. Wang, Q.; Zhang, M.; Torres, G.; Wu, S.; Ouyang, C.; Xie, Z.; Zou, M.H. Metformin Suppresses Diabetes-Accelerated Atherosclerosis via the Inhibition of Drp1-Mediated Mitochondrial Fission. Diabetes 2017, 66, 193–205. [Google Scholar] [CrossRef]
  200. Li, A.; Zhang, S.; Li, J.; Liu, K.; Huang, F.; Liu, B. Metformin and resveratrol inhibit Drp1-mediated mitochondrial fission and prevent ER stress-associated NLRP3 inflammasome activation in the adipose tissue of diabetic mice. Mol. Cell. Endocrinol. 2016, 434, 36–47. [Google Scholar] [CrossRef]
  201. Wang, Y.; An, H.; Liu, T.; Qin, C.; Sesaki, H.; Guo, S.; Radovick, S.; Hussain, M.; Maheshwari, A.; Wondisford, F.E.; et al. Metformin Improves Mitochondrial Respiratory Activity through Activation of AMPK. Cell Rep. 2019, 29, 1511–1523.e5. [Google Scholar] [CrossRef] [PubMed]
  202. Su, Y.; Hou, C.; Wang, M.; Ren, K.; Zhou, D.; Liu, X.; Zhao, S.; Liu, X. Metformin induces mitochondrial fission and reduces energy metabolism by targeting respiratory chain complex I in hepatic stellate cells to reverse liver fibrosis. Int. J. Biochem. Cell Biol. 2023, 157, 106375. [Google Scholar] [CrossRef]
  203. de Maranon, A.M.; Canet, F.; Abad-Jimenez, Z.; Jover, A.; Morillas, C.; Rocha, M.; Victor, V.M. Does Metformin Modulate Mitochondrial Dynamics and Function in Type 2 Diabetic Patients? Antioxid. Redox Signal. 2021, 35, 377–385. [Google Scholar] [CrossRef]
  204. Nasri, H.; Rafieian-Kopaei, M. Metformin: Current knowledge. J. Res. Med. Sci. 2014, 19, 658–664. [Google Scholar] [PubMed]
  205. Dabravolski, S.A.; Zhuravlev, A.D.; Kartuesov, A.G.; Borisov, E.E.; Sukhorukov, V.N.; Orekhov, A.N. Mitochondria-Mediated Cardiovascular Benefits of Sodium-Glucose Co-Transporter 2 Inhibitors. Int. J. Mol. Sci. 2022, 23, 5371. [Google Scholar] [CrossRef] [PubMed]
  206. Zhou, H.; Wang, S.; Zhu, P.; Hu, S.; Chen, Y.; Ren, J. Empagliflozin rescues diabetic myocardial microvascular injury via AMPK-mediated inhibition of mitochondrial fission. Redox Biol. 2018, 15, 335–346. [Google Scholar] [CrossRef]
  207. Lee, Y.H.; Kim, S.H.; Kang, J.M.; Heo, J.H.; Kim, D.J.; Park, S.H.; Sung, M.; Kim, J.; Oh, J.; Yang, D.H.; et al. Empagliflozin attenuates diabetic tubulopathy by improving mitochondrial fragmentation and autophagy. Am. J. Physiol. Renal Physiol. 2019, 317, F767–F780. [Google Scholar] [CrossRef]
  208. Takagi, S.; Li, J.; Takagaki, Y.; Kitada, M.; Nitta, K.; Takasu, T.; Kanasaki, K.; Koya, D. Ipragliflozin improves mitochondrial abnormalities in renal tubules induced by a high-fat diet. J. Diabetes Investig. 2018, 9, 1025–1032. [Google Scholar] [CrossRef]
  209. Liu, X.; Xu, C.; Xu, L.; Li, X.; Sun, H.; Xue, M.; Li, T.; Yu, X.; Sun, B.; Chen, L. Empagliflozin improves diabetic renal tubular injury by alleviating mitochondrial fission via AMPK/SP1/PGAM5 pathway. Metabolism 2020, 111, 154334. [Google Scholar] [CrossRef]
  210. Tsai, K.F.; Chen, Y.L.; Chiou, T.T.; Chu, T.H.; Li, L.C.; Ng, H.Y.; Lee, W.C.; Lee, C.T. Emergence of SGLT2 Inhibitors as Powerful Antioxidants in Human Diseases. Antioxidants 2021, 10, 1166. [Google Scholar] [CrossRef]
  211. Luo, J.; Xu, L.; Yu, T.; Li, J. Effect of statins therapy prior to percutaneous coronary intervention. J. Interv. Cardiol. 2012, 25, 156–162. [Google Scholar] [CrossRef] [PubMed]
  212. Mollazadeh, H.; Tavana, E.; Fanni, G.; Bo, S.; Banach, M.; Pirro, M.; von Haehling, S.; Jamialahmadi, T.; Sahebkar, A. Effects of statins on mitochondrial pathways. J. Cachexia Sarcopenia Muscle 2021, 12, 237–251. [Google Scholar] [CrossRef] [PubMed]
  213. Ramachandran, R.; Wierzbicki, A.S. Statins, Muscle Disease and Mitochondria. J. Clin. Med. 2017, 6, 75. [Google Scholar] [CrossRef] [PubMed]
  214. Ghavami, S.; Mutawe, M.M.; Hauff, K.; Stelmack, G.L.; Schaafsma, D.; Sharma, P.; McNeill, K.D.; Hynes, T.S.; Kung, S.K.; Unruh, H.; et al. Statin-triggered cell death in primary human lung mesenchymal cells involves p53-PUMA and release of Smac and Omi but not cytochrome c. Biochim. Biophys. Acta 2010, 1803, 452–467. [Google Scholar] [CrossRef] [PubMed]
  215. Thonusin, C.; Pantiya, P.; Jaiwongkam, T.; Kerdphoo, S.; Arunsak, B.; Amput, P.; Palee, S.; Pratchayasakul, W.; Chattipakorn, N.; Chattipakorn, S.C. A proprotein convertase subtilisin/kexin type 9 inhibitor provides comparable efficacy with lower detriment than statins on mitochondria of oxidative muscle of obese estrogen-deprived rats. Menopause 2020, 27, 1155–1166. [Google Scholar] [CrossRef] [PubMed]
  216. Southern, W.M.; Nichenko, A.S.; Shill, D.D.; Spencer, C.C.; Jenkins, N.T.; McCully, K.K.; Call, J.A. Skeletal muscle metabolic adaptations to endurance exercise training are attainable in mice with simvastatin treatment. PLoS ONE 2017, 12, e0172551. [Google Scholar] [CrossRef]
  217. Ali, A.U.; Abd-Elkareem, M.; Kamel, A.A.; Abou Khalil, N.S.; Hamad, D.; Nasr, N.E.H.; Hassan, M.A.; El Faham, T.H. Impact of porous microsponges in minimizing myotoxic side effects of simvastatin. Sci. Rep. 2023, 13, 5790. [Google Scholar] [CrossRef]
  218. Andres, A.M.; Hernandez, G.; Lee, P.; Huang, C.; Ratliff, E.P.; Sin, J.; Thornton, C.A.; Damasco, M.V.; Gottlieb, R.A. Mitophagy is required for acute cardioprotection by simvastatin. Antioxid. Redox Signal. 2014, 21, 1960–1973. [Google Scholar] [CrossRef]
  219. Apostolopoulou, M.; Corsini, A.; Roden, M. The role of mitochondria in statin-induced myopathy. Eur. J. Clin. Investig. 2015, 45, 745–754. [Google Scholar] [CrossRef]
  220. Batchuluun, B.; Pinkosky, S.L.; Steinberg, G.R. Lipogenesis inhibitors: Therapeutic opportunities and challenges. Nat. Rev. Drug Discov. 2022, 21, 283–305. [Google Scholar] [CrossRef]
  221. Christensen, D.G.; Xie, X.; Basisty, N.; Byrnes, J.; McSweeney, S.; Schilling, B.; Wolfe, A.J. Post-translational Protein Acetylation: An Elegant Mechanism for Bacteria to Dynamically Regulate Metabolic Functions. Front. Microbiol. 2019, 10, 1604. [Google Scholar] [CrossRef] [PubMed]
  222. Hu, Q.; Zhang, H.; Gutierrez Cortes, N.; Wu, D.; Wang, P.; Zhang, J.; Mattison, J.A.; Smith, E.; Bettcher, L.F.; Wang, M.; et al. Increased Drp1 Acetylation by Lipid Overload Induces Cardiomyocyte Death and Heart Dysfunction. Circ. Res. 2020, 126, 456–470. [Google Scholar] [CrossRef] [PubMed]
  223. Abel, E.D. Mitochondrial Dynamics and Metabolic Regulation in Cardiac and Skeletal Muscle. Trans. Am. Clin. Climatol. Assoc. 2018, 129, 266–278. [Google Scholar] [PubMed]
  224. Sergi, D.; Naumovski, N.; Heilbronn, L.K.; Abeywardena, M.; O’Callaghan, N.; Lionetti, L.; Luscombe-Marsh, N. Mitochondrial (Dys)function and Insulin Resistance: From Pathophysiological Molecular Mechanisms to the Impact of Diet. Front. Physiol. 2019, 10, 532. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Mitochondrial reactive oxygen species (ROS) production by the electron transport chain and its implications in metabolic disease. Illustration shows ROS production by the electron transport chain (ETC) in the inner mitochondrial membrane (IMM). Electron carriers NADH and FADH2 feed electrons (e) into the ETC at complex I and complex II, respectively. Electrons traverse through the ETC and release energy to pump H+ into the intermembrane space, generating a proton gradient and a membrane potential. ATP synthase (Complex V) uses the energy of the proton gradient to generate adenosine triphosphate (ATP) from adenosine diphosphate (ADP) and inorganic phosphate (Pi). Complex I and complex III, where electron leakage can occur, lead to the formation of superoxide radicals (O2•−) and other ROS. Excessive ROS production can lead to oxidative stress and contribute to the pathogenesis of metabolic diseases. Note: ubiquinone—Q; cytochrome c—C.
Figure 1. Mitochondrial reactive oxygen species (ROS) production by the electron transport chain and its implications in metabolic disease. Illustration shows ROS production by the electron transport chain (ETC) in the inner mitochondrial membrane (IMM). Electron carriers NADH and FADH2 feed electrons (e) into the ETC at complex I and complex II, respectively. Electrons traverse through the ETC and release energy to pump H+ into the intermembrane space, generating a proton gradient and a membrane potential. ATP synthase (Complex V) uses the energy of the proton gradient to generate adenosine triphosphate (ATP) from adenosine diphosphate (ADP) and inorganic phosphate (Pi). Complex I and complex III, where electron leakage can occur, lead to the formation of superoxide radicals (O2•−) and other ROS. Excessive ROS production can lead to oxidative stress and contribute to the pathogenesis of metabolic diseases. Note: ubiquinone—Q; cytochrome c—C.
Antioxidants 12 01163 g001
Figure 2. Mitochondrial fission mechanism and its role in physiology and pathology. (A) Schematic illustration of dynamin-related protein 1 (Drp1) structure. From the N-terminal to C-terminal, Drp1 is composed of a GTPase domain, middle domain, variable domain (VD), and GTPase effector domain (GED). (B) Drp1 is recruited to the outer mitochondrial membrane by the adaptor proteins (i.e., Fis1 and Mff), where Drp1 assembles into a ring-like structure around the mitochondrial tubules and constricts them, leading to their division into two daughter mitochondria. Drp1-mediated fission has broader physiological functions, whereas the dysregulation of Drp1 has been linked to a variety of pathological conditions.
Figure 2. Mitochondrial fission mechanism and its role in physiology and pathology. (A) Schematic illustration of dynamin-related protein 1 (Drp1) structure. From the N-terminal to C-terminal, Drp1 is composed of a GTPase domain, middle domain, variable domain (VD), and GTPase effector domain (GED). (B) Drp1 is recruited to the outer mitochondrial membrane by the adaptor proteins (i.e., Fis1 and Mff), where Drp1 assembles into a ring-like structure around the mitochondrial tubules and constricts them, leading to their division into two daughter mitochondria. Drp1-mediated fission has broader physiological functions, whereas the dysregulation of Drp1 has been linked to a variety of pathological conditions.
Antioxidants 12 01163 g002
Figure 3. Mitochondrial fragmentation is associated with ROS overproduction in diabetes and obesity. (A,B) Mouse C2C12 cell line myoblasts maintained in medium containing 5.6 mM glucose, or treated for 2 h with 15 mM glucose (HG) and 0.25 mM palmitate (HF). (C,D) Flexor digitorum brevis myofibers isolated from lean and obese rats. Mitochondria were labeled with MitoTrackerTM Red (A,C), and ROS levels were detected using the superoxide indicator dihydroethidium (B,D); scale bar: 10 μm. For methods and quantified results, please refer to our previous publications [40,83].
Figure 3. Mitochondrial fragmentation is associated with ROS overproduction in diabetes and obesity. (A,B) Mouse C2C12 cell line myoblasts maintained in medium containing 5.6 mM glucose, or treated for 2 h with 15 mM glucose (HG) and 0.25 mM palmitate (HF). (C,D) Flexor digitorum brevis myofibers isolated from lean and obese rats. Mitochondria were labeled with MitoTrackerTM Red (A,C), and ROS levels were detected using the superoxide indicator dihydroethidium (B,D); scale bar: 10 μm. For methods and quantified results, please refer to our previous publications [40,83].
Antioxidants 12 01163 g003
Figure 4. Targeting mitochondrial fission for antioxidants in metabolic diseases. Illustration shows that targeting mitochondrial fission represents a promising strategy for treating metabolic diseases by reducing ROS production and preventing mitochondrial damage. The relationship between ROS and mitochondria in metabolic diseases is complex and bidirectional, with mitochondria being both the primary sites of ROS production and targets of their damaging effects. Approaches such as lifestyle modifications (exercise and diet), dietary supplements, and Drp1 chemical inhibitors have been explored to target mitochondrial fission, and some have shown promising results in reducing ROS production and improving metabolic outcomes. Note: oxygen—O2; water—H2O; oxidative phosphorylation—oxphos.
Figure 4. Targeting mitochondrial fission for antioxidants in metabolic diseases. Illustration shows that targeting mitochondrial fission represents a promising strategy for treating metabolic diseases by reducing ROS production and preventing mitochondrial damage. The relationship between ROS and mitochondria in metabolic diseases is complex and bidirectional, with mitochondria being both the primary sites of ROS production and targets of their damaging effects. Approaches such as lifestyle modifications (exercise and diet), dietary supplements, and Drp1 chemical inhibitors have been explored to target mitochondrial fission, and some have shown promising results in reducing ROS production and improving metabolic outcomes. Note: oxygen—O2; water—H2O; oxidative phosphorylation—oxphos.
Antioxidants 12 01163 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yu, T.; Wang, L.; Zhang, L.; Deuster, P.A. Mitochondrial Fission as a Therapeutic Target for Metabolic Diseases: Insights into Antioxidant Strategies. Antioxidants 2023, 12, 1163. https://doi.org/10.3390/antiox12061163

AMA Style

Yu T, Wang L, Zhang L, Deuster PA. Mitochondrial Fission as a Therapeutic Target for Metabolic Diseases: Insights into Antioxidant Strategies. Antioxidants. 2023; 12(6):1163. https://doi.org/10.3390/antiox12061163

Chicago/Turabian Style

Yu, Tianzheng, Li Wang, Lei Zhang, and Patricia A. Deuster. 2023. "Mitochondrial Fission as a Therapeutic Target for Metabolic Diseases: Insights into Antioxidant Strategies" Antioxidants 12, no. 6: 1163. https://doi.org/10.3390/antiox12061163

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop