Skip to main content
Erschienen in: Journal of Translational Medicine 1/2023

Open Access 01.12.2023 | Review

Mitochondrial metabolic dysfunction and non-alcoholic fatty liver disease: new insights from pathogenic mechanisms to clinically targeted therapy

verfasst von: Youwei Zheng, Shiting Wang, Jialiang Wu, Yong Wang

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2023

Abstract

Metabolic dysfunction-associated fatty liver disease (MAFLD) is among the most widespread metabolic disease globally, and its associated complications including insulin resistance and diabetes have become threatening conditions for human health. Previous studies on non-alcoholic fatty liver disease (NAFLD) were focused on the liver’s lipid metabolism. However, growing evidence suggests that mitochondrial metabolism is involved in the pathogenesis of NAFLD to varying degrees in several ways, for instance in cellular division, oxidative stress, autophagy, and mitochondrial quality control. Ultimately, liver function gradually declines as a result of mitochondrial dysfunction. The liver is unable to transfer the excess lipid droplets outside the liver. Therefore, how to regulate hepatic mitochondrial function to treat NAFLD has become the focus of current research. This review provides details about the intrinsic link of NAFLD with mitochondrial metabolism and the mechanisms by which mitochondrial dysfunctions contribute to NAFLD progression. Given the crucial role of mitochondrial metabolism in NAFLD progression, the application potential of multiple mitochondrial function improvement modalities (including physical exercise, diabetic medications, small molecule agonists targeting Sirt3, and mitochondria-specific antioxidants) in the treatment of NAFLD was evaluated hoping to provide new insights into NAFLD treatment.
Hinweise
Youwei Zheng, Shiting Wang and Jialiang Wu contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
8-OH-DG
8-OH-deoxyguanosine
ACC
Acetyl-CoA carboxylase
AD
Alzheimer’s disease
AKAP1
A kinase anchoring protein 1
AP-1
Activator protein-1
BA
Bile acids
BCL-2
B-cell lymphoma-2
BNIP3
Bcl2/adenovirus E1B 19 kDa interacting protein 3
CAD
Coronary artery disease
cAMP
Cyclic adenosine monophosphate
CDCA
Chenodeoxycholic acid
circRNA
Circular RNA
CRMP
Controlled release mitochondrial plasmin
CVD
Cardiovascular disease
DCA
Deoxycholic acid
DNL
De novo lipogenesis
DRAK2
Death associated protein related apoptotic kinase 2
Drp1
Dynamin-related protein 1
EGR
Early growth response
eNOS
Endothelial nitric oxide synthase
ER
Endoplasmic reticulum
FADH2
Flavin adenine dinucleotide
FAs
Fatty acids
FFA
Free fatty acids
FGF
Fibroblast growth factor
FIS1
Fission protein 1
GFP
Green fluorescent protein
GLP-1
Glucagon-like peptide-1
GSH
Glutathione
GSSG
Glutathione disulfide
GST
Glutathione-S-transferase
GTP
Guanosine triphosphate
GWAS
Genome-wide association studies
H2O2
Hydrogen peroxide
HCC
Hepatocellular carcinoma
HD
Huntington’s chorea
HDL
High-density lipoprotein
HFD
High-fat diet
HNE
4-Hydroxy-2-nonenal
HO-1
Heme oxygenase-1
IL-18
Interleukin-18
IL-1β
Interleukin-1β
IMM
Inner mitochondrial membrane
JNK
c-Jun amino-terminal kinase
LCA
Lithocholic acid
LIR
LC3 interaction region
MAFLD
Metabolic dysfunction-associated fatty liver disease
MAMs
Mitochondria-associated endoplasmic reticulum membranes
MERS
Mitochondrial-endoplasmic reticulum contact
MFF
Mitochondrial fission factor
MFN1
Mitofusin 1
MFN2
Mitofusin 2
Mito-Q
Mito-quinone
MitoVit-E
MitoVitamin E
MQC
Mitochondrial quality control
Mst1
Macrophage-stimulated 1
mtDNA
Mitochondrial DNA
MT-ND6
Mitochondria-encoded NADH dehydrogenase 6 (MT-ND6)
MTP
Mitochondrial trifunctional protein complex
NADH
Nicotinamide adenine dinucleotide
NAFL
Non-alcoholic fatty liver
NAFLD
Non-alcoholic fatty liver disease
NASH
Nonalcoholic steatohepatitis
NFκB/Orai1
Nuclear factor kappa-B/calcium release-activated calcium modulator 1
Nix
Nip3-like protein X
NLRP3
NOD‑like receptors family pyrin domain containing 3
NO
Nitric oxide
NRF2
Nuclear factor E2-related factor 2
OLETF
Otsuka Long-Evans Tokushima Fatty
OMM
Outer mitochondrial membrane
OPA1
Optic atrophy 1
OXPHOS
Oxidative phosphorylation
PARL
Progerin-associated rhodopsin-like
PD
Parkinson’s disease
PINK
PTEN-induced putative kinase 1
PKA
Protein kinase A
PNPLA3
Patatin-like phospholipase domain-containing 3
PPAR-γ
Peroxisome proliferator-activated receptor gamma
PS1
Presenilin-1
ROS
Reactive oxygen species
S1R
Sigma-1 receptor
S637
Serine 637
SCD
Stearoyl-CoA desaturase
SIRT3
Recombinant Sirtuin 3
SREBP1c
Sterol regulatory element binding protein 1c
T2D
Type 2 diabetes
TC
Total cholesterol
TG
Triglycerides
TGS
Triglycerides
TM6SF2
Transmembrane 6 superfamily member 2
TNF-α
Tumor necrosis factor-α
TPP
Triphenylphosphine
TUDCA
Tauroursodeoxycholic acid
TZDs
Thiazolidinediones
UDCA
Ursodeoxycholic acid
VLDL
Very low density lipoproteins

Introduction

Over the past two decades, nonalcoholic fatty liver disease (NAFLD) has developed to rank among the most chronic and widespread hepatic diseases, and with the global obesity epidemic, its prevalence is increasing. It has become a serious threat to human health [1], because of the increased risk of comorbidities including cardiovascular disease, hepatocellular carcinoma (HCC), and type 2 diabetes (T2D) [24]. NAFLD is a metabolic stress liver injury closely related to insulin resistance (IR) and genetic susceptibility, with hepatic steatosis in the liver. However, other causes such as alcoholic liver disease, autoimmune hepatitis, drug damage and hypothyroidism are excluded. Genome-wide association studies (GWAS) have shown that changes in genes such as patatin-like phospholipase domain-containing 3 (PNPLA3) and transmembrane 6 superfamily member 2 (TM6SF2) have a differential impact on the development of NAFLD [5].The liver has an indispensable function in balancing lipid metabolism by modulating the production, oxidation, and transport of triglycerides (TGs), free fatty acids (FFA), cholesterol, and bile acids (BA) [6]. Excessive fat accumulation in the liver includes multiple conditions associated with the emergence and hepatic steatosis progression, these range from simple non-alcoholic fatty liver (NAFL) to complicated non-alcoholic steatohepatitis (NASH). NAFLD is not only closely related to liver cancer, but also involved in many extrahepatic diseases, such as bladder cancer and sarcopenia. Among them, the occurrence of bladder cancer is largely due to insulin resistance. A retrospective study shows that most patients with bladder cancer suffer from NAFLD, insulin resistance and T2D [7]. In addition, NAFLD is also closely related to sarcopenia, which we will focus on below. The excessive fat accumulation in Metabolic Dysfunction-Associated Fatty Liver Disease (MAFLD) patients, produces cytotoxic lipid oxidation byproducts that can cause NASH with a chronic necro-inflammatory state [8]. Notably, impaired mitochondrial metabolism is extensively associated with NAFLD by mediating the dysregulation of lipid metabolic homeostasis. With further research, the exclusive diagnosis of NAFLD can not meet the clinical needs, which is not conducive to early diagnosis and early intervention of diseases. Therefore, to reflect the hepatic manifestations of this metabolic disorder, the name NAFLD has been reconsidered and recently renamed MAFLD [911]. MAFLD is more likely to be obese and overweight.
The mechanism by which mitochondrial function regulates liver metabolism and therefore, interferes with the disease progression has been intensively investigated. Several studies have shown that factors such as mitophagy, oxidative stress, differentiation, and quality control, differentially influence mitochondrial function to promote liver fat accumulation and injury. These steps in part mediate impaired lipid metabolism to exacerbate the development of NAFLD. In patients with MAFLD, mitochondrial dysfunction-mediated disruption of lipid metabolism leads to excessive accumulation of TGs (> 5%) in hepatocytes and hepatic steatosis [12, 13]. MAFLD covers a range of diseases from NAFL with no obvious inflammatory manifestations to NASH, these hepatic steatoses are linked with lobular inflammation, pericyte fibrosis, and apoptosis, and cannot distinguish histologically from alcoholic steatohepatitis [14, 15]. However, the transition from NAFL to NASH is not only based on steatosis but is also driven by mitochondrial dysfunction characterized by dysregulated oxidative phosphorylation (OXPHOS) and reactive oxygen species (ROS) generation [16, 17]. Therefore, mitochondrial function regulation appears to be a potential strategy to stop the progression or even treat MAFLD.
Because of the underlying importance of mitochondrial metabolic disorders in NAFLD development, it is crucial to target its metabolic regulatory mechanisms for therapeutic strategies against NAFLD. The available literature suggests that physical exercise, antidiabetic drugs, and antioxidants may have the potential to reverse mitochondrial metabolic disorders and hold good promise for large-scale clinical application. Overall, this review focuses on the impact of mitochondrial metabolic dysregulation and lipid accumulation on the progression of NAFLD and discusses multiple potential therapeutic strategies to improve mitochondrial function, hoping to provide new insights for clinically targeted therapies based on NAFLD.
In reviewing the relevant literature, we drew on the approach of Baethge et al. to develop the following methodology for the literature search [18]. The first step is to retrieve the corresponding subject and free words in the corresponding database, using the logical operators “AND” and “OR”, the wildcard character “*”, etc. to connect the subject and free words. Also note the field identification of the corresponding phrase in the search box to limit the search and compose an advanced expression for advanced searching. The second step is to import the searched documents into the literature management software and subsequently read the title, abstract and, if necessary, the text in the literature management software to filter the required documents. If there is a large amount of literature, the relevant literature from the last 5 years was chosen.

Lipid accumulation promotes MAFLD progression

Recently, multiple theories have been put forth to elaborate on the occurrence and development of MAFLD. The “multiple-hit” hypothesis has replaced the “double-hit " hypothesis of NASH, the former takes into account the promotion of MAFLD by multiple impairments in genetically susceptible individuals, such as hormones secreted by adipose tissue, insulin resistance, nutritional factors, and gut microbiota [19, 20]. Adipose tissue stores excess food calories in the form of TGs, and the accumulation of TGs and FFA in the liver is the result of dysregulated fat metabolism [21]. Excessive caloric intake may induce MAFLD/NASH by mediating gut microbiota imbalance and increasing portal circulation of bacterial products, causing activation of the natural immune system [22, 23]. Due to the enhanced circulating plasma FFA, IR occurs in the muscles of the body, which is a key step in MAFLD development [24, 25]. Furthermore, IR increases hepatic De novo lipogenesis (DNL) and stimulates adipose tissue to secrete adipokine and inflammatory cytokine, ultimately leading to the dysregulation of adipose tissue’s lipolytic processes [26]. Interestingly, fat accumulation in the liver increases lipotoxicity, causing an increase in oxidative stress which can severely impair normal mitochondrial metabolic programs and cellular functions in the liver and provide the potential for FA accumulation [27]. Therefore, this positive feedback loop (FA accumulation- IR-impaired mitochondrial metabolism -FA accumulation) may support the continued progression of MAFLD/NASH and establishes the possibility for the development of certain malignant diseases such as HCC.
During MAFLD, FAs from different sources (such as dietary, DNL, and lipolysis) are utilized in hepatocytes to produce TGs, contributing to liver fat accumulation [28, 29]. Approximately 59% of the FAs that accumulate in the liver are sourced from adipose tissue lipolysis, 26% is from DNL and 15% is from the diet (Fig. 1) [28]. FFA can also be transformed into TGs and transported as very low-density lipoproteins (VLDL) [30]. This metabolic dysfunction mediates the accumulation of liver fat and causes the continued development of MAFLD. Interestingly, increased circulating levels of FFA and TGs and adipose tissue stored TGs also contribute to peripheral IR [31, 32], while persistent hyperglycemia and compensatory hyperinsulinemia contribute to the progression of T2D and fatty liver in obese patients. Indeed, FA accumulation mediates liver damage and fatty liver development in different dimensions. The ongoing fat metabolic breakdown related to IR causes elevated blood circulating FFA levels, which are readily transported to the liver and provide the conditions for fatty liver development [31]. Similarly, high carbohydrate consumption increases hepatic lipogenesis, thereby supporting fatty liver progression by increasing the synthesis and uptake of FFA [33, 34]. Notably, sustained accumulation of FFA may induce liver injury by activating immune-inflammatory pathways. FFA promotes nuclear factor-κB-dependent TNF-α expression by mediating lysosomal instability and stimulating increased hepatotoxicity to impair normal hepatic metabolic function [35].
It has been suggested that an abnormally high DNL could potentially contribute to the development of MAFLD [36]. A high carbohydrate diet supports NAFLD progression by inducing DNL to regulate the sterol regulatory element binding protein 1c (SREBP1c) expression [37, 38]. The development of NAFLD is accompanied by severe mitochondrial metabolic disruption and therefore, contributes to lipid accumulation to support NAFLD progression. For example, impaired mitochondrial metabolism in NAFLD impairs FA oxidation processes (e.g., mitochondrial beta-oxidation) and induces elevated DNL, which ultimately continues to drive NAFLD progression in the form of lipid accumulation [39, 40]. Moreover, when mitochondrial beta-oxidation is impaired, peroxisomal and cytochrome oxidation of FFA alternate, resulting in high ROS levels and dangerous oxidative by-products that support disease progression [41, 42]. Ma et al. [43] revealed that initially, the increased hepatic lipid availability in obese individuals with NAFLD enhances hepatic mitochondrial competence, however, it eventually stimulates enhanced hepatic oxidative stress and therefore, decreases mitochondrial activity, promoting the development of NAFLD into NASH [44]. Studies have also demonstrated that dysfunction in liver mitochondria precedes NAFLD and IR onset in obese rodent models [45]. Additionally, the endoplasmic reticulum (ER) is a crucial part of the stress response, it regulates a distinctive set of intracellular pathways, a phenomenon collectively referred to as the unfolded protein response [46, 47]. Interestingly, nuclearfactorkappa-B/calcium release-activated calcium modulator 1 (NFκB/Orai1) stimulates the regulation of the ER in intracellular pathways via oxidative stress to support the development of NAFLD [48]. Multiple NAFLD pathogenic mechanisms should not be viewed independently, as there may be ongoing crosstalk between different tissues or organs, where an intrinsic association of adipose tissue and the gut may contribute to NAFLD [49] because dysregulated intestinal metabolism contributes to fat accumulation and lipid deposition in the liver via the gut-adipose tissue-liver axis and mediates hepatic disease development [50]. Additionally, NAFLD pathogenesis is also influenced by genetic and epigenetic factors (including DNA methylation and histone modifications), with heritability estimates ranging from 20 to 70% [51, 52]. Specifically, activator protein-1 (AP-1) and early growth response (EGR) reprogram Kupffer cell properties and LXR functions required for survival to transform the associated macrophage phenotype and drive myeloid cell diversity in NASH [53]. As such, the known information suggests that epigenetic reprogramming may contribute to the phenotypic transformation of immune cells and favor the development of NAFLD by influencing their inflammatory environment [54, 55].
The elucidation of different mechanisms of disease progression facilitates the development of targeted therapies, while the intrinsic linkages in disease mechanisms are equally noteworthy, as their interactions pose serious challenges for clinical treatment. Therefore, understanding the interaction between different mechanisms is also beneficial for the clinical application of NAFLD-based targeted combination therapy.

Mitochondrial metabolic dysfunction and MAFLD

Mitochondrial division

The liver is one of the most mitochondria-rich organs, as the density of mitochondria depends mainly on the energy metabolic demand [56]. In hepatocytes, the mitochondrial activity includes metabolic pathways and signaling networks, which depend on mitochondrial DNA (mtDNA) integrity, affinity and antioxidant balance, membrane composition, transport of lipoprotein, and metabolic demand and supply [57]. For example, a case-control study assessed epigenetic changes in mtDNA, and mitochondria-encoded NADH dehydrogenase-6 (MT-ND6) hypermethylation was determined in liver biopsy tissue from MAFLD patients and correlated with the severity of histological disease [58]. These alterations are associated with ultrastructural changes in the mitochondria, manifested by inner mitochondrial membrane (IMM) loss, deep ridges folding, and mitochondrial granules loss, although these changes may be reversed by exercise [58]. The structural stability of mitochondria is essential for the maintenance of normal physiological activity. For example, exosomes released by Trem2-deficient macrophages can exacerbate NAFLD by damaging the mitochondrial structure of hepatocytes. The specific reason for this is because high levels of miR-106b-5p block Mitofusin 2 (MFN2) [59]. Similarly, the protein components of mitochondria-associated endoplasmic reticulum membranes (MAMs) are involved in the development of many diseases. Drp1 is a key factor in the control of mitochondrial division. In the striatum of Huntington’s chorea (HD) mice, overactive Drp1 activity is able to induce mitochondrial fragmentation, forcing mitochondria away from the endoplasmic reticulum and leading to the destruction of the MAM [60]. MAM proteins also include PTEN-induced putative kinase 1 (PINK), sigma-1 receptor (S1R), presenilin-1 (PS1) and so on. The above proteins are involved in Parkinson’s disease (PD) and Alzheimer’s disease (AD) respectively.In addition, genetic variants and metabolic stressors can stimulate mitochondrial dysfunction [61]. mtDNA mutations, particularly copy number variants and somatic point mutations, and mitochondrial metabolism abnormalities have been observed in HCC associated with NAFLD [62]. In this case, mitochondrial functions such as mitotic-nuclear communication and mitochondrial-ER contact (MERS) are impaired due to structural disruption, and this phenomenon may confer resistance to apoptosis and trigger the development of HCC [63, 64]. In addition, there is an association between impaired mitochondrial function and the development of sarcopenia. Mitochondrial dysfunction with impaired proteostatic mechanisms has been reported as an important factor in sarcopenia [65]. Interestingly, a reduction in muscle mass may contribute to the development of IR. Together with obesity, chronic inflammation and vitamin D deficiency, these factors are involved in the pathophysiological mechanisms of NAFLD [66]. Therefore, improving mitochondrial function can effectively alleviate sarcopenia and NAFLD. Maintaining the stability of mitochondrial structure and function and facilitating a complex balance between physical contacts between organelles and regulatory mechanisms at multiple levels, is also an essential basis for ensuring cellular homeostasis.
The length of the continuous mitochondrial renewal and degradation cycle determines the total number of mitochondria in the cell. During mtDNA replication, new mitochondria are produced from pre-existing ones when the transcription and translation processes of the nuclear and mitochondrial encoded genes are coordinated. Because mitochondria cannot be produced ab initio, cells use the alternating fusion-division steps in the mitochondrial pool to achieve its turnover [67, 68]. Mitochondrial fusion is usually associated with increased OXPHOS function and mitochondrial elongation, whereas, the main mitochondrial fusion proteins are Mitofusin 1 (MFN1)、MFN2 and optic atrophy 1 (OPA1), which are also required to maintain normal mitochondrial function. Specifically, MFN1 and MFN2 are widely distributed on the outer mitochondrial membrane (OMM), but they have different activities within the cell (Fig. 2). MFN1 is shown to have an 8-fold higher GTPase activity than MFN2 and mediates the fusion of the OMM in a guanosine triphosphate (GTP)-dependent reaction with a higher affinity for mitochondrial bridging [69]. OPA1 is crucial for the attachment of IMM with mitochondrial cristae formation. ATP-dependent Yme1L and Oma1 proteases regulate the cleavage of OPA1 into L-OPA1 or S-OPA1 [70, 71]. Depletion or abnormal activity of proteins mediating OMM fusion may lead to the emergence of defective mitochondria, which are then removed by autophagy. For example, uncontrolled Oma1-dependent cleavage prolongs the dissipation of mitochondrial potential, a phenomenon that leads to the complete inactivation of OPA1 and consequent disruption of mitochondrial structure [68].
Dynamin-related protein-1 (Drp1), which is mediated by mitochondrial fission protein 1 (FIS1), mitochondrial fission factor (Mff), and the mitochondrial dynamin 49/50 receptor, and is recruited and activated around OMMs, is the main trigger for the cytokinesis process (Fig. 3) [72]. Activation of Drp1 may drive cell division in different dimensions and impair normal mitochondrial metabolic functions. Interestingly, post-translational modifications, energy insufficiency, physical exercise, and enhanced cyclic adenosine monophosphate (cAMP) stimulate protein kinase-A (PKA), which phosphorylates serine 656 (S656) and S637 residues of Drp1, thereby inhibiting the activity of Drp1 on mitochondrial damage in cytokinesis and offers the possibility of delaying the progression of MAFLD [73]. Research suggests that high cAMP concentrations favor co-localization and anchoring of PKA and Drp1 on OMMs via the protein scaffold A-kinase anchoring protein 1 (AKAP1) and Mff respectively, while the PKA-AKAP1 complex inhibits Drp1 and the fission process [74, 75]. In contrast, calcium-regulated neurophosphatase, a calcium ion/calmodulin-dependent phosphatase, promotes the formation of a helical homopolymer complex of Drp1 to wrap the mitochondrial membrane by reversing the phosphorylation of S637 and Drp1, which gradually contracts and induce mitochondrial rupture [76]. In liver cells, redox status, energy stress, reduced ATP, and NADH may stimulate the division of mitochondrial via the Sirtuin1 (SIRT1)/cAMP-dependent AMP-activated protein kinase (AMPK) signaling pathway [77], which regulates Mff phosphorylation resulting in Drp1 functioning on OMMs. However, in the muscle and liver tissues, during autophagy, a cascade reaction via cAMP/PKA/AMPK can phosphorylate Drp1 and prevent mitochondria from dividing [78, 79], which may be attributed to autophagy against cell division. In general, Drp1 activity is closely related to mitochondrial function, and specific means to modulate Drp1 expression or activity may be a viable strategy to rescue mitochondrial dysfunction and treat MAFLD.

Mitophagy

The selective elimination of dysfunctional mitochondria by autophagic vesicles is called mitophagy [80]. Recognition of damaged mitochondria by autophagic vesicles is performed in a ubiquitin-independent and dependent pathway by LC3 adapters [81]. The PTEN-induced PINK1/Parkin pathway is the most researched mitochondrial autophagic pathway. In healthy mitochondria, progerin-associated rhodopsin-like (PARL) proteases and matrix processing peptidase (MPP) degrade PINK1 [82]. However, on the impaired mitochondria, PINK1 assembles and stimulates the elimination of these mitochondria in a docked manner [82]. The ubiquitin kinase PINK1 phosphorylates ubiquitin and activates the ubiquitin ligase Parkin [81]. GTPase mitofusin 2 on OMM is thought to regulate Parkin assembly in impaired mitochondria. PINK1 phosphorylates Mfn2 and promotes its parkin-mediated ubiquitination [83]. Parkin ubiquitinates proteins of OMM and facilitates their cross-talk with phagosomal adapters including NBR1, p62, and HDAC6 [81]. These junctions have LC3 interaction region (LIR) motifs, and LC3 can recognize and recruit labeled mitochondria into the autophagosome. In the mitophagy receptor pathway, receptors including Bcl2/Adenovirus E1B 19 kDa and Nip3-like protein X (Nix) and interacting Protein 3 (BNIP3) were found to bind directly to LC3 and promote mitochondrial phagocytosis by autophagic vesicles, thereby eliminating lysosome-induced mitochondrial destruction. Moreover, mice with Bnip3 gene deletion exhibited some degree of inflammation and higher levels of ROS [84]. Impaired mitophagy has been associated with a variety of human diseases (e.g. MAFLD and tumors) [85]. Diminished or impaired mitophagy disrupts the production of healthy mitochondria and causes impaired mitochondria aggregation. In addition, mitophagy also has a function in activating inflammatory vesicles [86]. Moore et al. linked mitochondrial damage to increased severity of NAFLD in obese patients [87]. He indicated a 40-50% alleviation in ß-oxidation in individuals with NASH, which was linked with elevated hepatic ROS and decreased indices of mitochondrial biosynthesis, mitosis, autophagy, fission, and fusion. The study of mitophagy and NAFLD goes far beyond this. Macrophage-stimulated 1 (Mst1) is a novel upstream regulator of mitophagy that affects apoptosis in cancer cells by inhibiting mitophagy. Mst1 was found to promote NAFLD by disrupting Parkin-associated mitophagy. The specific mechanism is that Mst1 regulates parkin expression through the AMPK pathway. AMPK blockade inhibits Parkin-associated mitophagy and thus affects hepatocyte mitochondrial apoptosis [88]. In addition to this, mitosis is regulated by hormones. Thyroid hormones also reportedly alleviate NAFLD by enhancing FA oxidation, mitosis, and mitochondrial biogenesis [89, 90], as these hormones can increase mRNA expression of NIX, BNIP3, p62, ULK1, and LC3 under certain conditions to mediate mitophagy [91]. Mitochondrial bioregulation alters and coordinates the mitochondrial pool’s amount and quality between mitoses, allowing cellular mitochondrial activity regulation in response to cellular stress, metabolic status, and other intracellular hormonal and environmental signals and to induce mitophagy to complete the repair of diseased mitochondria to slow the progression of NAFLD.

Oxidative stress

Mitochondria are organelles with complex structures and comprise OMM the IMM, and the mitochondrial matrix. Most mitochondria-associated proteins are produced in the cytoplasm and then transported to their site of action. The OMM is porous and allows the passage of ions and small uncharged molecules, while IMM consists of a complex of electron transport systems, transport proteins, and ATP synthases [92].
Energy is mainly produced in mitochondria in the form of ATP via pyruvate and FAs metabolism. To undergo β-oxidation, cytoplasmic FAs need mitochondrial entry [93], whereas, short- and medium-chain FAs can freely diffuse into the mitochondrial matrix. Long-chain Acyl-Coenzyme-A synthase in OMM activates long-chain FAs to Acyl-Coenzyme-A. On the outer side of the IMM, CPT1 transfers acyl groups from Acyl-Coenzyme-A to carnitine to form acyl-carnitine. Carrier protein carnosine-acylcarnitine translocase transports acyl-carnitine across the IMM. The mitochondrial matrix’s CPT2 converts acyl-carnitine to carnitine and Acyl-Coenzyme-A. Within the mitochondria, the β-oxidation cycle of four enzymatic steps degrades Acyl-Coenzyme-A [94]. In each cycle, Acyl-Coenzyme A shortens and two carboxy-terminal carbon atoms are released as acetyl coenzyme-A [95]. The first β-oxidation step is the dehydrogenation of acyl coenzyme-A to trans-2-enoyl-Coenzyme-A by Acyl-Coenzyme-A dehydrogenase. The last three steps are catalyzed by the mitochondrial trifunctional protein complex (MTP). In the second step, enoyl coenzyme-A hydratase catalyzes the hydration by producing (S)-3-hydroxy-Acyl-Coenzyme-A, which is then dehydrogenated by (S)-3-hydroxy-Acyl-Coenzyme-A dehydrogenase to generate 3-ketoacyl coenzyme A [39]. Lastly, thiolase cleaves 3-ketoacyl coenzyme-A to produce a two-carbon chain shortened Acyl-Coenzyme A and Acetyl-Coenzyme A. Aside from Acetyl-Coenzyme-A that enters ketogenesis and the TCA cycle, β-oxidation also generates nicotinamide adenine dinucleotide (NADH) and flavin adenine dinucleotide (FADH2). Electron Transport Chain (ETC) utilizes NADH and FADH2 to produce ATP. ATP is also generated by OXPHOS, which links the oxidation of NADH or FADH2 with the phosphorylation of ADP to form ATP [94].
Mitochondria produces 90% of cellular ROS [96, 97]. Fatty degeneration of the liver reduces the efficiency of the respiratory transport chain, generating ROS and ER stress (Fig. 4) [98]. Few electrons may escape ETC during ATP synthesis and react with oxygen to generate ROS. Under physiological conditions, only about 1–2% of mitochondrial oxygen loss produces ROS [99]. At this stage, electrons react with oxygen to form superoxide, which disrupts mitochondria by peroxidizing mtDNA [100], phospholipid Acyl chains, and respiratory transport chain enzymes [101]. In these conditions, ROS work as signaling molecules, and their production is countered by non-enzymatic and enzymatic antioxidant mechanisms. Additionally, increased lipid flow to hepatocytes dysregulates the de-phosphorylating activity of mitochondrial voltage-dependent anion channels and inner membrane permeability, causing mitochondrial depolarization, reduced ATP production, and loss of antioxidant activity [102104], enhanced production of ROS [105, 106], and lipid peroxidation products (malondialdehyde (MDA) and 4-Hydroxy-2-nonenal (HNE) [107]. These processes then subsequently promotes apoptosis, inflammation, and liver fibrosis. Saturated FAs disrupt the mitochondrial membrane composition, favoring NAFLD progression [108]. In NAFLD, sustained FFAs flow and chronic acetyl coenzyme-A production can separate the function of the TCA cycle from mitochondrial respiration, producing excessive ROS production [109]. Excessive production of ROS causes oxidative damage to hepatocytes and exacerbates NAFLD [109, 110]. The release of ROS outside of liver cells causes hepatic stellate cells (HSCs) activation and extracellular matrix deposition. The relationship between ROS and NAFLD is much more than that. By analysing mitochondrial circular RNA (circRNA) expression profiles in fibroblasts with NASH, the investigators found that circRNA was down-regulated in a significant proportion. However, the construction of mitochondria-targeted nanoparticles that target circRNA SCAR can alleviate high-fat diet-induced cirrhosis and insulin resistance [111]. Excess liver FFA can induce the storage of toxic lipid intermediates, including ceramides [112]. Research suggests that the use of antioxidants to eliminate ROS from the cytoplasmic lysates and mitochondrial matrix can prevent simple steatosis and NASH [113]. Distinctive GPX1 deletion in hepatocytes protects mice from diet-associated NASH. However, there are few investigations show conflicting results [114117]. The association of increased ROS production with enhanced detoxification and antioxidant activity of hepatic steatosis, but not with NASH, indicates that in NASH, the ROS overproduction mechanism may be inadequate [44]. As a continuous cycle sustained ROS release further damages hepatic tissue and dysregulates mitochondrial activity, thereby promoting fatty liver conversion to NASH in which mitochondrial adaptations are lost [118]. Indeed, ROS aggregation may activate c-Jun amino-terminal kinase (JNK) signaling and block the PPARα-FGF21 axis, thereby impeding mitochondrial β-oxidation and ketogenesis [119]. Chronic JNK stimulation is linked with apoptosis and chronic hepatic injury because it disrupts the function of the proto-oncogene non-receptor tyrosine kinase Src in the IMM, which is essential for the physiological activity of ETC [120, 121].
Decreases in the free hepatic radical scavengers, including mitochondrial MnSOD [45, 122], an increase in the oxidized form glutathione (GSH), and a reduction of glutathione disulfide (GSSG), also exacerbate oxidative stress. For instance, lower levels of MnSOD were similarly observed in humans, and in the liver of high-fat diet (HFD) fed rodents [44, 122]. In HFD-fed mice, accumulation of GSSG in the liver sensitizes hepatocytes to TNF-α-regulated cytotoxicity [123]. In addition, a study revealed that the level of both GSSG/GSH ratio and Glutathione-S-transferase (GST) were increased in the blood samples of 21 pediatric NASH children. Glutathione transferase is the second line of defense against oxidative stress. Simultaneously, increased electron donors from enhanced nutrient supply flood the mitochondria, for example, and create a high proton gradient at the IMM. In case the ATP synthase response to proton leakage fails to increase its activity, mitochondria may produce more ROS [124]. Compared to NAFLD and controls, obese NASH patients had the most enhanced levels of hydrogen peroxide (H2O2) and 8-OH-deoxyguanosine (8-OH-DG) in the liver, a marker of oxidative DNA damage. Thus, the imbalance between antioxidant defense and free radical growth creates a favorable environment for oxidative damage, inflammation, and fibrosis in hepatocytes, promoting lipid peroxidation of cell membranes, apoptosis, and ROS-mediated somatic mutations of nuclear and mtDNA.

Mitochondrial quality control

Excess lipids in hepatocytes stimulate the production of both mitochondrial ROS production and FA oxidation. In a vicious cycle, impaired mitochondria lose their activity, leading to aberrant OXPHOS and increased ROS generation. Increased accumulation and uncontrolled mitochondrial ROS generation can harm mitochondrial components, including proteins, membranes, and mtDNA, and induce mitochondrial quality control (MQC) [125127]. As an initial reaction to stress/ROS, MQC includes biogenesis, division, fusion, and mitosis, where mitochondria first try to retain their function and structure via DNA repair, antioxidants, protein folding, and degradation mechanisms. Mitochondrial biosynthesis, fusion, and division compensate for its activity. If the first response is ineffective, a more extensive MQC system is initiated [128, 129]. Impaired mitochondria can be improved by fusing with healthy mitochondria, however, severely damaged ones are isolated from healthy mitochondria by division and then targeted for degradation by mitophagy as described earlier [130, 131]. Endothelial nitric oxide synthase (eNOS) is a master regulator of mitochondrial quality control, and mice lacking eNOS are more susceptible to liver inflammation and fibrosis. A series of studies on liver mitochondria from eNOS knockout mice, among others, revealed that eNOS is an important regulator of liver mitochondrial content and function as well as NASH sensitivity [132]. MQC failure leads to loss of mitochondrial function and is among the potential causes of NAFLD progression [133].

Therapeutic potential of improved mitochondrial function for NAFLD

So far, several therapeutic strategies have emerged in this field, but with different goals, as stated below. More evidence is needed to validate these treatments before they can enter clinical trials.

Physical exercise

The current study shows that physical exercise stimulates the synthesis of mitochondria in the liver and transverse muscle [134]. Therefore, it can be hypothesized that physical activity can prevent and treat NAFLD by regulating mitochondrial activity and structure [135]. Studies have shown that an effective acute physical exercise session performed once has an impact on liver metabolism and redox status. However, whether there is permeability of protons in the mitochondrial membrane, 4th and enhanced 3rd state respirations, and stress response to mitochondrial permeability transition are affected needs further exploration [136, 137]. Prolonged physical activity for endurance training (or voluntary running) improves indices of the liver mitochondrial integrity and function and may promote a more stress-resistant or disease-resistant typical phenotype of the liver [137]. The restoration of mitochondrial function by physical exercise is not only seen in the liver. It was shown that in ovariectomised rats, exercise restored mitochondrial oxygen consumption and that even after ovariectomy, physical exercise compensated for the damage caused by ovariectomy by improving mitochondrial function [138].
The results of existing animal model studies have now presented an association between physical exercise and functional transformations of mitochondria. For example, Otsuka Long-Evans Tokushima Fatty (OLETF) rats developed obesity and T2D with multiple phenotypes of the metabolic syndrome [139, 140], the liver of OLEFT rats after daily voluntary running cycles for 16 or 36 weeks showed many changes including enhanced mitochondrial FA oxidation, oxidase function, and protein content. In addition, rat-based studies have also shown that levels of proteins associated with hepatic neolipogenesis are suppressed after physical exercise [141, 142]. Of course, this change parallels several indices of the mitochondrial OXPHOS apparatus, namely, increased citrate synthase, palmitate oxidation, β-hydroxyacyl-CoA dehydroacenaphthene, palmitoyl-CoA transferase 1 activity, cytochrome c, and ETC complex IV. Other effects include increased phosphorylated form of acetyl-CoA carboxylase (ACC) and decreased ACC, FA synthase, and activity of stearoyl-CoA desaturase (SCD) (inhibition markers of new hepatic lipogenesis) [143], further supporting the idea that physical exercise slows the progression of NAFLD by restoring normal mitochondrial biogenesis. In addition to this, exercise increases the oxidative capacity of liver mitochondria, which in turn improves the IR that drives hepatic steatosis. The mechanisms include enhanced FA oxidation and decreased synthesis of FA-derived ceramides and diacylglycerols associated with hepatic IR [144, 145]. Physical exercise and endurance training improve biogenesis and autophagy of mitochondria [146] and reduce mPTP opening in HFD-fed rats [147]. Daily physical activity is also linked with increased orthomorphic mitochondria in the liver, epigenetic modification of mtDNA (i.e., MT-ND6 hypermethylation), and improved severity of NAFLD [58]. In addition, physical exercise improves NAFLD by reducing intrahepatic fat content, increasing beta-oxidation of fatty acids, inducing hepatoprotective autophagy, overexpressing peroxisome proliferator-activated receptor gamma (PPAR-γ), and attenuating hepatocyte apoptosis and increasing insulin sensitivity. In conclusion, physical activity is closely related to the restoration of mitochondrial function and is essential for the treatment of NAFLD[148]. However, the low adherence to patients’ lifestyles has reflected the immediate medicine requirement [149].

Anti-diabetes drugs

Peroxisome proliferator-activated receptors (PPARs) belongs to the superfamily of nuclear receptor that binds to a variety of FAs and their derivatives, including binding to several FFAs and their derivatives, and transcriptionally modulates intracellular metabolic pathways [150]. Current studies suggest the existence of three types of PPARs, which are classified as PPAR-α, PPAR-δ (also called PPAR-β), and PPAR-γ based on the ligand and PPARs tissue distribution. PPAR-α is expressed in the adipose tissue, liver, skeletal muscle, heart, and kidney, and is responsible for the regulation of lipid transport, gluconeogenesis, and the hormone fibroblast growth factor (FGF)-21. PPAR-α activation switches liver metabolism toward FA transport and β-oxidation [151], improve plasma lipids by alleviating TGs and enhancing high-density lipoprotein (HDL) cholesterol [152]. In animal models PPAR-α deficiency showed a parallel trend with worsening hepatic steatosis, however, activation of the receptor by typical PPAR-α agonists (e.g. fibrates) did not show a significant effect on NAFLD, although serum TGs were reduced [153]. Liver, skeletal muscle, and macrophages expressed PPAR-δ improved insulin sensitivity, reduced liver glucose production [154], elevated FA oxidation, and reduced activation of Kupffer cells [155]. Liver PPAR-δ carried an anti-inflammatory role in macrophages [155]. Activation of PPAR-δ reduces steatosis, but overexpression of PPAR-δ may have an impact on maintaining glucose levels. For example, high doses of PPAR-δ agonists exhibit reduced fasting insulin concentrations in the treatment of Rhesus monkeys [156]. Fortunately, Elafibranor shows unique effects as a PPARα/δ agonist, not only increases FA β-oxidation (PPARα activity) but also improves IR and inflammation [154]. Therefore, its distinct and efficient effects on hepatic mitochondria demand further research. PPAR-γ is majorly found in adipose tissues and modulates glucose metabolism, adipogenesis, and adipose tissue differentiation. Thiazolidinediones (TZDs) are PPARγ agonists that act as insulin sensitizers and antidiabetic agents, and recent studies have found that this family of drugs may also have potential against NAFLD (Table 1) [157159]. For example, pioglitazone is effective in improving NASH to some extent [160, 161]. In addition to this, studies have shown that rosiglitazone also has the ability to improve NASH [162164]. Notably, there may be mitochondrial targets of thiazolidinediones (mTOT) in animals that are able to act as mitochondrial membrane complexes to participate in pyruvate transport. However, in a mouse model with NASH, pioglitazone appears to reverse this steatosis in the liver [165, 166]. The specific reason for this may be related to the ability of TZDs to inhibit the entry of pyruvate into the TCA cycle [167]. In addition, a new PPARγ agonist named MSDC-0602 K targets mitochondrial pyruvate carriers while minimizing direct binding to transcription factors [168]. Of course, further proof is required to determine the function of novel agents in the treatment of NAFLD, such as selective PPARα modulators (fibrates, K-877), and PPARγ agonists (INT-131), PPARα/γ (DSP-8658), and PPARδ (HPP-593) [151].
Table 1
Drugs under study or approved for the treatment of MAFLD
Category
Name
Function
References
PPARα/γ/δ agonists
Elafibranor
Increased fatty acid beta oxidation, improves insulin resistance and inflammation
[154]
 
Thiazolidinediones (TZD)
 
[157, 159]
 
Pioglitazone
Rosiglitazone
Improves mitochondrial function, protects pancreatic beta cells and increases tissue sensitivity to insulin
 
 
MSDC-0602K
 
[168]
Biguanide hypoglycemic drugs
Dimethylbiguanide
Phenelzine
Targeted mitochondrial pyruvate carrier
[224]
 
Buformin
Improved insulin sensitivity of liver and peripheral tissues
[225, 226]
GLP-1 receptor agonist
Liraglutide
Exenatide
Lisiratide
Enhancement of mitochondrial structure, attenuation of ROS production and promotion of autophagy
[227]
Mitochondria-targeted antioxidants
Ursodeoxycholic acid (UDCA)
Elamipretide
  
Decoupling agent
Aramchol
Affects mitochondrial electron transport
[196]
 
Mito-quinone (Mito-Q)
MitoVitamin-E (MitoVit-E)
Reduction of SCD1 and maintenance of cellular redox homeostasis
[198]
 
Silymarin
Protects cells from peroxide-induced oxidative damage and apoptosis
[228]
 
Controlled Release Mitochondrial Protonin (CRMP)
Regulation of thioredoxin and nitric oxide (NO) derivatives to reduce lipid peroxidation
[229, 230]
Intravenous injection of functional mitochondria
Mitotherapy
Liver mitochondrial uncoupling, improving liver fibrosis and liver protein synthesis
Reduced lipid content, improve cellular redox balance
[216, 217]
The classic T2D treatment drug, dimethyl-biguanide, improves insulin sensitivity in the liver and peripheral tissues. Loading oleic acid in cultured HepG2 cells induced steatosis, and metformin was able to reduce steatosis and improve hepatocyte function [169]. Among the mechanisms are a decrease in oxidative stress impairment, and modulation of protein levels linked with the mitochondrial apoptotic pathway and its inhibition. Metformin also stimulates AMPK, which activates mitochondrial synthesis and FA β-oxidation, which are important for maintaining and promoting mitochondrial function [170].
Liraglutide is an acylated glucagon-like peptide-1 (GLP-1) agonist. In cultured hepatic HepG2 cells, it ameliorated NASH by inhibiting nucleotide-binding oligomeric structural domains, NOD‑like receptors family pyrin domain containing 3 (NLRP3), and focalization via mitotic activation [171]. Additionally, liraglutide improved NAFLD in HFD-fed mice by increasing mitochondrial synthesis, decreasing ROS generation, and elevating autophagy via the SIRT1/SIRT3 pathway [172].

Targeted at SIRT3

Recombinant Sirtuin 3 (SIRT3) is a mitochondrial NAD+-dependent deacetylase that is important for regulating the activity of proteins related to cellular metabolism [173]. The SIRT3 gene encodes three isoforms, and the two long isoforms in mice are SIRT3 protein (M1 and M2) expressed in mitochondria. On contrary, the short-SIRT3 protein (M3) type is expressed on the cellular membrane and lacks N-terminal mitochondrial targeting signals. All isomers possess deacetylase activity, although they are distributed in different positions [174176]. During fasting, SIRT3 upregulates β-oxidation and ATP generation [177], inhibits ROS, and enhances mitochondrial biosynthesis by peroxisome proliferators-activated receptor γ coactivator lalpha (PGC-1α) activation [178]. In contrast, in mice lacking SIRT3, mitochondrial proteins are hyperacetylated and impair mitochondrial function [179]. SIRT3 is a greatly expressed sirtuin in the liver of mice, it improves mitochondrial activity and NAFLD by modulating ketogenesis, β-oxidation, mitogenesis, and antioxidant response systems [179]. In human and mouse NAFLD models, however, SIRT3 is downregulated [180]. SIRT3 and PGC-1α can be mutually regulated, and both decreased in HFD-fed mice, this downregulation of SIRT3 caused mitochondrial proteins hyperacetylation and enhances fat storage and oxidative stress in the liver [181]. Exposure of SIRT3-deficient mice to an HFD further elevated this acetylated state of hepatic proteins, reduced the activity of respiratory complex III and IV, and exacerbated oxidative stress [182, 183]. Palmitate-induced lipotoxicity increases ROS production and hepatocyte death in SIRT3-deficient primary hepatocytes [184]. SIRT3 overexpression altered the inhibition of ATP synthesis via palmitate treatment [185]. In addition, this overexpression also inhibited the production of ROS. HFD in SIRT3 deficient mice exacerbates obesity, IR, hyperlipidemia, hepatic steatosis, and inflammation, however, adenovirus overexpressing SIRT3 rescued this phenotype [183]. In addition to its mitochondrial effects, hepatic SIRT3 deficiency exacerbated hepatic steatosis in HFD mice by upregulating proteins associated with FA uptake, including CD36 and VLDL receptors [184].
SIRTs are a potent therapeutic NAFLD target because they provide protection to hepatocytes from the effects of lipotoxicity [186]. Small molecule sirtuin modulators have been developed, but a few compounds targeting human SIRTs are still in clinical development. The basic problem is to determine the isozyme specificity and site-specific delivery of SIRTs activators [187, 188]. Notably, SIRT4 may be associated with coronary artery disease (CAD) in obese patients with NAFLD and normal or slightly elevated liver enzymes [189]. Low levels of SIRT4 produce large amounts of ROS while regulating free fatty acids, which, together with the release of free fatty acids from adipose tissue breakdown, leads to endothelial cell dysfunction, resulting in the development of CAD.

Bile acids affect mitochondrial function

In bile acids (BA), the naturally present “tertiary” dihydroxy ursodeoxycholic acid (UDCA), exterior of chenodeoxycholic acid (CDCA), has shown multiple liver protecting effects and improved liver conditions in individuals with multiple chronic liver disorders [190]. On the basis of previous data, UDCA is also being examined in NASH individuals. Previously in an open-label pilot investigation, UDCA demonstrated some beneficial effects on hepatic enzymes and degree of steatosis (at biopsy) in NASH patients [191]. At the mitochondrial level, lipophilic BAs, such as deoxycholic acid (DCA), CDCA, and lithocholic acid (LCA), suppressed the ETC. The effect of high concentrations of BA (100 µmol/L) on the IMM of intact mitochondria was nonspecific, whereas the effect in broken or intact mitochondria propagated with low BA concentrations (10 µmol/L) was specific (damage to complex I and III) [192]. In the liver condition of biliary stasis with excessive BA retention, the antioxidant capacity of mitochondria is reduced [193]. In addition, cholestasis is associated with impaired mitochondrial function. At toxicologically suitable levels, most (but not all) BAs changes mitochondrial bioenergetics [194]. UDCA shows antioxidant and anti-inflammatory properties and prevents mitochondrial dysfunction in the progression of obesity-related diseases. In addition to this, it has been found that UDCA and Tauroursodeoxycholic Acid (TUDCA) as well as a lipophilic BA (CDCA and LCA) in mitochondria isolated from rat liver can have some effect on ETC function [195].
Aramchol (Arachidyl-amido cholanoic) also demonstrated potential effects in humans on hepatic steatosis, however, failed to improve glucose metabolism, hepatic enzymes, or insulin sensitivity [196, 197]. Aramchol in animal models ameliorates fibrosis and steatohepatitis by decreasing stearoyl coenzyme-A desaturase 1 (SCD1) and elevating fluxes that maintain cellular redox homeostasis through the transsulfur pathway [196]. Mice deficient in SCD1 have reduced lipid synthesis, increased mitochondrial FA β-oxidation, and insulin sensitivity in different tissues, including hepatic tissues. Therefore, SCD1 insufficiency has been associated with the prevention of hepatic steatosis in several nonalcoholic fatty liver mice models, such as high carbohydrate and HFD mice.

Mitochondria-targeted antioxidants

Several antioxidants targeting mitochondria exist, but their specific role in the clinical arena needs to be further explored. Mito-quinone (Mito-Q) and MitoVitamin E (MitoVit-E) are potential new antioxidant classes. Both molecules contain covalently linked lipophilic triphenylphosphine (TPP) cation molecules, which allow them to pass across and accumulate within the mitochondria [198200]. Mito-Q ameliorated the metabolic syndrome in 8 weeks of HFD rats [201] and revealed enhanced hepatic mitochondrial cardiolipin levels and central phospholipid synthase expression [202]. Mito-Q can prevent increased cholesterol, TG, glucose, and mtDNA damage by oxidation, and hepatic steatosis in atherosclerosis and metabolic syndrome experimental models [203, 204]. Low Mito-Q and MitoVit-E doses provide protection to cells against peroxide-stimulated apoptosis and oxidative damage, in contrast to low concentrations of non-targeted antioxidants such as ubiquinone and Vit-E [205, 206]. The protective efficiency of MitoVit-E and Mito-Q may be regulated by the inactivation of caspase-3 and cytochrome-c release. Additionally, they alleviate ROS-triggered transferrin receptor-induced iron uptake, lipid peroxidation and peroxidation-induced complex I inhibition, and aconitase in mitochondria [206]. A phase II investigation in chronic hepatitis-C individuals revealed that Mito-Q decreased circulating levels of transaminase; indicating a reduction in hepatic inflammation and necrosis in these individuals [207].
Silymarin is the main compound extracted from silymarin (Silybum marianum). Silymarin has few hepatoprotective activities [208] and may improve IR, hepatic injury, and hepatic enzymes in NAFLD individuals [209, 210]. Silymarin phospholipid complexes comprising vit-E alleviated hepatic steatosis in NAFLD patients and markedly reduced hepatic fat infiltration in HFD rats [211]. This phenomenon may be achieved by modulating thioredoxin and derivatives of nitric oxide (NO), as well as by substantially reducing lipid peroxidation. Silymarin also improved mitochondrial alterations in the respiratory complex and protected subunit CII-30 of complex II [212].
Inhibiting the production of mitochondrial ROS by uncoupling is an effective strategy and an antioxidant approach. 2,4-dinitrophenol (DNP) is an artificial uncoupling agent with a potential for toxicity [213], however, further verification is needed regarding whether it can alleviate NAFLD [214]. Controlled release mitochondrial plasmin (CRMP) is an oral DNP formulation that induces a mild uncoupling effect in liver mitochondria. In a rat model, it can alleviate increased TG, IR, hepatic steatosis, and T2D [215]. In addition, CRMP normalized plasma transaminase levels, alleviated liver fibrosis, and liver protein synthesis, and showed no toxicity at a systemic level in a methionine/choline-deficient NASH rat model [215].

Mitotherapy

Dispersion therapy is a procedure in which functional mitochondria from exogenous hepatocytes are administered intravenously. This process may be successful in ameliorating HFD-induced hepatic steatosis. Among the specific mechanisms include the reduction of lipid molecules and improvement of redox homeostasis in cells. With this approach, exogenous mitochondria can be labeled with a green fluorescent protein and reacquired in the lung, brain, liver, muscle, and kidney of mice [216, 217]. This approach reduces fat deposition, prevents cellular damage, and increases energy production while restoring liver cell activity. However, the mechanism by which intact mitochondria infiltrate various cells and resume cellular metabolic activities is still undetermined [218]. Currently, related studies are focused on mitochondrial metabolism, for instance, which metabolic and proteomic variations are present in mitochondria extracted from normal liver cells compared to those of non-tumor origin.

Conclusions and future perspectives

For the moment, there is some literature on the relationship between NAFLD and mitochondrial function. However, according to the search results, this literature is more about the link between NAFLD and a certain function of mitochondria, such as aspects of mitophagy [12, 80], oxidative stress [118], etc. Or a single presentation on how to treat NAFLD through mitochondria, such as mitochondrial transplants [32], herbal medicine [118], etc. We found no literature that integrates the above and the focus of this article is on the structure and function of mitochondria and the various current therapeutic modalities regarding mitochondria, respectively, to give the reader a better understanding of the close link between mitochondria and NAFLD. Metabolic dysfunction-related fatty liver or NAFL diseases are now among the most widespread chronic liver diseases globally, with MAFLD expected to overtake viral liver disease and put people at increased risk of terminal-stage liver disease, HCC, and CVD. Environment, genetics, and metabolic dysfunctions may be the cause of the pathophysiological development of NAFLD, such as alterations in liver lipid constituents, cellular impairment, and tumorigenesis [219]. Mitochondrial aberration and oxidative stress are hallmarks of NAFLD. Although animal models and human studies are contradictory, there is growing evidence that mitochondrial cycling can fall into imbalance during NAFLD. In terms of the mechanisms responsible for the regulation of mitochondrial morphology and dimensions, recent investigations suggest that mitochondrial metabolism alterations may begin in the initial NAFLD stages [220], speculating whether it could be considered a mitochondrial disorder. In addition, mitochondrial abnormalities persist during the disease course and may contribute to the advancement of MAFLD to HCC and NASH [221]. Therefore, it is crucial to prevent and treat NAFLD at any possible stage.
Some new potent drugs and molecular targets have been determined for improving treatment outcomes. However, recent clinical trial results suggest that understanding the pathophysiology of NASH is still limited and is far from achieving an optimal therapeutic strategy. In addition to affecting the genetic, metabolic, or environmental risk factors and stressors, therapeutic approaches may require several fundamentally important subcellular organelles as targets [222]. Shortly, one might consider using combination therapy. To date, no drug treatment has been approved against NAFLD, however, lifestyle alterations, physical exercise, and weight loss can regulate oxidative stress and the life cycle of mitochondria. The establishment of mitochondria-based therapies, some of which have been examined in humans (e.g., vit-E), has provided efficient benefits for NASH and has emerged as an anti-cancer adjuvant [223]. However, they depict only a small fragment of mitochondrial metabolism that may be regulated. Thus, a deep knowledge of the molecular structure of mitochondrial plasticity remains in its infancy, but it may furnish new doors for future physiological compound development having the potential for clinical NAFLD treatment. Ideally, factors that alter disease could simultaneously act on mitochondrial activity and energy production, rather than only on intracellular lipid metabolism regulators. Overall, we still need to be equipped with sufficient time and power to determine the prolonged efficacy and safety of each promising treatment option.

Acknowledgements

Not applicable.

Declarations

Not applicable.
All authors approved the version of the manuscript to be published.

Competing interests

The authors declare that they have no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Swinburn BA, Sacks G, Hall KD, McPherson K, Finegood DT, Moodie ML, Gortmaker SL. The global obesity pandemic: shaped by global drivers and local environments. Lancet. 2011;378:804–14.PubMedCrossRef Swinburn BA, Sacks G, Hall KD, McPherson K, Finegood DT, Moodie ML, Gortmaker SL. The global obesity pandemic: shaped by global drivers and local environments. Lancet. 2011;378:804–14.PubMedCrossRef
2.
Zurück zum Zitat Ballestri S, Zona S, Targher G, Romagnoli D, Baldelli E, Nascimbeni F, Roverato A, Guaraldi G, Lonardo A. Nonalcoholic fatty liver disease is associated with an almost twofold increased risk of incident type 2 diabetes and metabolic syndrome. Evidence from a systematic review and meta-analysis. J Gastroenterol Hepatol. 2016;31:936–44.PubMedCrossRef Ballestri S, Zona S, Targher G, Romagnoli D, Baldelli E, Nascimbeni F, Roverato A, Guaraldi G, Lonardo A. Nonalcoholic fatty liver disease is associated with an almost twofold increased risk of incident type 2 diabetes and metabolic syndrome. Evidence from a systematic review and meta-analysis. J Gastroenterol Hepatol. 2016;31:936–44.PubMedCrossRef
3.
Zurück zum Zitat Mittal S, El-Serag HB, Sada YH, Kanwal F, Duan Z, Temple S, May SB, Kramer JR, Richardson PA, Davila JA. Hepatocellular carcinoma in the absence of cirrhosis in United States veterans is associated with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol. 2016;14:124-131e121.PubMedCrossRef Mittal S, El-Serag HB, Sada YH, Kanwal F, Duan Z, Temple S, May SB, Kramer JR, Richardson PA, Davila JA. Hepatocellular carcinoma in the absence of cirrhosis in United States veterans is associated with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol. 2016;14:124-131e121.PubMedCrossRef
4.
Zurück zum Zitat Targher G, Day CP, Bonora E. Risk of cardiovascular disease in patients with nonalcoholic fatty liver disease. N Engl J Med. 2010;363:1341–50.PubMedCrossRef Targher G, Day CP, Bonora E. Risk of cardiovascular disease in patients with nonalcoholic fatty liver disease. N Engl J Med. 2010;363:1341–50.PubMedCrossRef
5.
Zurück zum Zitat Lohmueller KE, Pearce CL, Pike M, Lander ES, Hirschhorn JN. Meta-analysis of genetic association studies supports a contribution of common variants to susceptibility to common disease. Nat Genet. 2003;33:177–82.PubMedCrossRef Lohmueller KE, Pearce CL, Pike M, Lander ES, Hirschhorn JN. Meta-analysis of genetic association studies supports a contribution of common variants to susceptibility to common disease. Nat Genet. 2003;33:177–82.PubMedCrossRef
8.
Zurück zum Zitat Romero FA, Jones CT, Xu Y, Fenaux M, Halcomb RL. The race to bash NASH: emerging targets and drug development in a complex liver disease. J Med Chem. 2020;63:5031–73.PubMedCrossRef Romero FA, Jones CT, Xu Y, Fenaux M, Halcomb RL. The race to bash NASH: emerging targets and drug development in a complex liver disease. J Med Chem. 2020;63:5031–73.PubMedCrossRef
9.
Zurück zum Zitat Eslam M, George J. Reply to: correspondence regarding “A new definition for metabolic dysfunction-associated fatty liver disease: an international expert consensus statement”: bringing evidence to the NAFLD-MAFLD debate. J Hepatol. 2020;73:1575.PubMedCrossRef Eslam M, George J. Reply to: correspondence regarding “A new definition for metabolic dysfunction-associated fatty liver disease: an international expert consensus statement”: bringing evidence to the NAFLD-MAFLD debate. J Hepatol. 2020;73:1575.PubMedCrossRef
10.
Zurück zum Zitat Zheng KI, Fan JG, Shi JP, Wong VW, Eslam M, George J, Zheng MH. From NAFLD to MAFLD: a “redefining” moment for fatty liver disease. Chin Med J (Engl). 2020;133:2271–3.PubMedCrossRef Zheng KI, Fan JG, Shi JP, Wong VW, Eslam M, George J, Zheng MH. From NAFLD to MAFLD: a “redefining” moment for fatty liver disease. Chin Med J (Engl). 2020;133:2271–3.PubMedCrossRef
11.
Zurück zum Zitat Eslam M, Sanyal AJ, George J, International Consensus P. MAFLD: a consensus-driven proposed nomenclature for metabolic associated fatty liver disease. Gastroenterology. 2020;158:1999-2014 e1991.CrossRef Eslam M, Sanyal AJ, George J, International Consensus P. MAFLD: a consensus-driven proposed nomenclature for metabolic associated fatty liver disease. Gastroenterology. 2020;158:1999-2014 e1991.CrossRef
12.
Zurück zum Zitat Aryapour E, Kietzmann T. Mitochondria, mitophagy, and the role of deubiquitinases as novel therapeutic targets in liver pathology. J Cell Biochem. 2022;123:1634–46.PubMedPubMedCentralCrossRef Aryapour E, Kietzmann T. Mitochondria, mitophagy, and the role of deubiquitinases as novel therapeutic targets in liver pathology. J Cell Biochem. 2022;123:1634–46.PubMedPubMedCentralCrossRef
14.
15.
Zurück zum Zitat Ludwig J, Viggiano TR, McGill DB, Oh BJ. Nonalcoholic steatohepatitis: Mayo Clinic experiences with a hitherto unnamed disease. Mayo Clin Proc. 1980;55:434–8.PubMed Ludwig J, Viggiano TR, McGill DB, Oh BJ. Nonalcoholic steatohepatitis: Mayo Clinic experiences with a hitherto unnamed disease. Mayo Clin Proc. 1980;55:434–8.PubMed
16.
Zurück zum Zitat Fuchs M, Schnabl B. Introduction to the NAFLD and NASH special issue. Dig Dis Sci. 2016;61:1211–3.PubMedCrossRef Fuchs M, Schnabl B. Introduction to the NAFLD and NASH special issue. Dig Dis Sci. 2016;61:1211–3.PubMedCrossRef
18.
19.
Zurück zum Zitat Buzzetti E, Pinzani M, Tsochatzis EA. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism. 2016;65:1038–48.PubMedCrossRef Buzzetti E, Pinzani M, Tsochatzis EA. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism. 2016;65:1038–48.PubMedCrossRef
20.
Zurück zum Zitat Tilg H, Adolph TE, Moschen AR. Multiple parallel hits hypothesis in nonalcoholic fatty liver disease. Revisited after a decade. Hepatology. 2021;73:833–42.PubMedCrossRef Tilg H, Adolph TE, Moschen AR. Multiple parallel hits hypothesis in nonalcoholic fatty liver disease. Revisited after a decade. Hepatology. 2021;73:833–42.PubMedCrossRef
21.
Zurück zum Zitat Eccleston HB, Andringa KK, Betancourt AM, King AL, Mantena SK, Swain TM, Tinsley HN, Nolte RN, Nagy TR, Abrams GA, Bailey SM. Chronic exposure to a high-fat diet induces hepatic steatosis, impairs nitric oxide bioavailability, and modifies the mitochondrial proteome in mice. Antioxid Redox Signal. 2011;15:447–59.PubMedPubMedCentralCrossRef Eccleston HB, Andringa KK, Betancourt AM, King AL, Mantena SK, Swain TM, Tinsley HN, Nolte RN, Nagy TR, Abrams GA, Bailey SM. Chronic exposure to a high-fat diet induces hepatic steatosis, impairs nitric oxide bioavailability, and modifies the mitochondrial proteome in mice. Antioxid Redox Signal. 2011;15:447–59.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Hebbard L, George J. Animal models of nonalcoholic fatty liver disease. Nat Rev Gastroenterol Hepatol. 2011;8:35–44.PubMedCrossRef Hebbard L, George J. Animal models of nonalcoholic fatty liver disease. Nat Rev Gastroenterol Hepatol. 2011;8:35–44.PubMedCrossRef
25.
Zurück zum Zitat Utzschneider KM, Kahn SE. Review: the role of insulin resistance in nonalcoholic fatty liver disease. J Clin Endocrinol Metab. 2006;91:4753–61.PubMedCrossRef Utzschneider KM, Kahn SE. Review: the role of insulin resistance in nonalcoholic fatty liver disease. J Clin Endocrinol Metab. 2006;91:4753–61.PubMedCrossRef
26.
Zurück zum Zitat Guilherme A, Virbasius JV, Puri V, Czech MP. Adipocyte dysfunctions linking obesity to insulin resistance and type 2 diabetes. Nat Rev Mol Cell Biol. 2008;9:367–77.PubMedPubMedCentralCrossRef Guilherme A, Virbasius JV, Puri V, Czech MP. Adipocyte dysfunctions linking obesity to insulin resistance and type 2 diabetes. Nat Rev Mol Cell Biol. 2008;9:367–77.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Cusi K. Role of insulin resistance and lipotoxicity in non-alcoholic steatohepatitis. Clin Liver Dis. 2009;13:545–63.PubMedCrossRef Cusi K. Role of insulin resistance and lipotoxicity in non-alcoholic steatohepatitis. Clin Liver Dis. 2009;13:545–63.PubMedCrossRef
28.
Zurück zum Zitat Donnelly KL, Smith CI, Schwarzenberg SJ, Jessurun J, Boldt MD, Parks EJ. Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease. J Clin Invest. 2005;115:1343–51.PubMedPubMedCentralCrossRef Donnelly KL, Smith CI, Schwarzenberg SJ, Jessurun J, Boldt MD, Parks EJ. Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease. J Clin Invest. 2005;115:1343–51.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Bradbury MW. Lipid metabolism and liver inflammation. I. hepatic fatty acid uptake: possible role in steatosis. Am J Physiol Gastrointest Liver Physiol. 2006;290:G194-198.PubMedCrossRef Bradbury MW. Lipid metabolism and liver inflammation. I. hepatic fatty acid uptake: possible role in steatosis. Am J Physiol Gastrointest Liver Physiol. 2006;290:G194-198.PubMedCrossRef
31.
Zurück zum Zitat Tirosh A, Shai I, Bitzur R, Kochba I, Tekes-Manova D, Israeli E, Shochat T, Rudich A. Changes in triglyceride levels over time and risk of type 2 diabetes in young men. Diabetes Care. 2008;31:2032–7.PubMedPubMedCentralCrossRef Tirosh A, Shai I, Bitzur R, Kochba I, Tekes-Manova D, Israeli E, Shochat T, Rudich A. Changes in triglyceride levels over time and risk of type 2 diabetes in young men. Diabetes Care. 2008;31:2032–7.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Chen Y, Yang F, Chu Y, Yun Z, Yan Y, Jin J. Mitochondrial transplantation: opportunities and challenges in the treatment of obesity, diabetes, and nonalcoholic fatty liver disease. J Transl Med. 2022;20:483.PubMedPubMedCentralCrossRef Chen Y, Yang F, Chu Y, Yun Z, Yan Y, Jin J. Mitochondrial transplantation: opportunities and challenges in the treatment of obesity, diabetes, and nonalcoholic fatty liver disease. J Transl Med. 2022;20:483.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Morio B, Panthu B, Bassot A, Rieusset J. Role of mitochondria in liver metabolic health and diseases. Cell Calcium. 2021;94: 102336.PubMedCrossRef Morio B, Panthu B, Bassot A, Rieusset J. Role of mitochondria in liver metabolic health and diseases. Cell Calcium. 2021;94: 102336.PubMedCrossRef
34.
Zurück zum Zitat Fromenty B, Roden M. Mitochondrial alterations in fatty liver diseases. J Hepatol. 2023;78:415–29.PubMedCrossRef Fromenty B, Roden M. Mitochondrial alterations in fatty liver diseases. J Hepatol. 2023;78:415–29.PubMedCrossRef
35.
Zurück zum Zitat Feldstein AE, Werneburg NW, Canbay A, Guicciardi ME, Bronk SF, Rydzewski R, Burgart LJ, Gores GJ. Free fatty acids promote hepatic lipotoxicity by stimulating TNF-alpha expression via a lysosomal pathway. Hepatology. 2004;40:185–94.PubMedCrossRef Feldstein AE, Werneburg NW, Canbay A, Guicciardi ME, Bronk SF, Rydzewski R, Burgart LJ, Gores GJ. Free fatty acids promote hepatic lipotoxicity by stimulating TNF-alpha expression via a lysosomal pathway. Hepatology. 2004;40:185–94.PubMedCrossRef
36.
Zurück zum Zitat Lambert JE, Ramos-Roman MA, Browning JD, Parks EJ. Increased de novo lipogenesis is a distinct characteristic of individuals with nonalcoholic fatty liver disease. Gastroenterology. 2014;146:726–35.PubMedCrossRef Lambert JE, Ramos-Roman MA, Browning JD, Parks EJ. Increased de novo lipogenesis is a distinct characteristic of individuals with nonalcoholic fatty liver disease. Gastroenterology. 2014;146:726–35.PubMedCrossRef
37.
Zurück zum Zitat Sanders FW, Griffin JL. De novo lipogenesis in the liver in health and disease: more than just a shunting yard for glucose. Biol Rev Camb Philos Soc. 2016;91:452–68.PubMedCrossRef Sanders FW, Griffin JL. De novo lipogenesis in the liver in health and disease: more than just a shunting yard for glucose. Biol Rev Camb Philos Soc. 2016;91:452–68.PubMedCrossRef
38.
Zurück zum Zitat Ferre P, Foufelle F. Hepatic steatosis: a role for de novo lipogenesis and the transcription factor SREBP-1c. Diabetes Obes Metab. 2010;12(Suppl 2):83–92.PubMedCrossRef Ferre P, Foufelle F. Hepatic steatosis: a role for de novo lipogenesis and the transcription factor SREBP-1c. Diabetes Obes Metab. 2010;12(Suppl 2):83–92.PubMedCrossRef
40.
Zurück zum Zitat Li Z, Li Y, Zhang HX, Guo JR, Lam CWK, Wang CY, Zhang W. Mitochondria-mediated pathogenesis and therapeutics for non-alcoholic fatty liver disease. Mol Nutr Food Res. 2019;63:e1900043.PubMedCrossRef Li Z, Li Y, Zhang HX, Guo JR, Lam CWK, Wang CY, Zhang W. Mitochondria-mediated pathogenesis and therapeutics for non-alcoholic fatty liver disease. Mol Nutr Food Res. 2019;63:e1900043.PubMedCrossRef
41.
Zurück zum Zitat Del Rio LA, Lopez-Huertas E. ROS generation in peroxisomes and its role in cell signaling. Plant Cell Physiol. 2016;57:1364–76.PubMed Del Rio LA, Lopez-Huertas E. ROS generation in peroxisomes and its role in cell signaling. Plant Cell Physiol. 2016;57:1364–76.PubMed
42.
Zurück zum Zitat Abdelmegeed MA, Banerjee A, Yoo SH, Jang S, Gonzalez FJ, Song BJ. Critical role of cytochrome P450 2E1 (CYP2E1) in the development of high fat-induced non-alcoholic steatohepatitis. J Hepatol. 2012;57:860–6.PubMedPubMedCentralCrossRef Abdelmegeed MA, Banerjee A, Yoo SH, Jang S, Gonzalez FJ, Song BJ. Critical role of cytochrome P450 2E1 (CYP2E1) in the development of high fat-induced non-alcoholic steatohepatitis. J Hepatol. 2012;57:860–6.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Koliaki C, Szendroedi J, Kaul K, Jelenik T, Nowotny P, Jankowiak F, Herder C, Carstensen M, Krausch M, Knoefel WT, Schlensak M, Roden M. Adaptation of hepatic mitochondrial function in humans with non-alcoholic fatty liver is lost in steatohepatitis. Cell Metab. 2015;21:739–46.PubMedCrossRef Koliaki C, Szendroedi J, Kaul K, Jelenik T, Nowotny P, Jankowiak F, Herder C, Carstensen M, Krausch M, Knoefel WT, Schlensak M, Roden M. Adaptation of hepatic mitochondrial function in humans with non-alcoholic fatty liver is lost in steatohepatitis. Cell Metab. 2015;21:739–46.PubMedCrossRef
45.
Zurück zum Zitat Rector RS, Thyfault JP, Uptergrove GM, Morris EM, Naples SP, Borengasser SJ, Mikus CR, Laye MJ, Laughlin MH, Booth FW, Ibdah JA. Mitochondrial dysfunction precedes insulin resistance and hepatic steatosis and contributes to the natural history of non-alcoholic fatty liver disease in an obese rodent model. J Hepatol. 2010;52:727–36.PubMedPubMedCentralCrossRef Rector RS, Thyfault JP, Uptergrove GM, Morris EM, Naples SP, Borengasser SJ, Mikus CR, Laye MJ, Laughlin MH, Booth FW, Ibdah JA. Mitochondrial dysfunction precedes insulin resistance and hepatic steatosis and contributes to the natural history of non-alcoholic fatty liver disease in an obese rodent model. J Hepatol. 2010;52:727–36.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Dasgupta D, Nakao Y, Mauer AS, Thompson JM, Sehrawat TS, Liao CY, Krishnan A, Lucien F, Guo Q, Liu M, Xue F, Fukushima M, Katsumi T, Bansal A, Pandey MK, Maiers JL, DeGrado T, Ibrahim SH, Revzin A, Pavelko KD, Barry MA, Kaufman RJ, Malhi H. IRE1A stimulates hepatocyte-derived extracellular vesicles that promote inflammation in mice with steatohepatitis. Gastroenterology. 2020;159:1487-1503 e1417.PubMedCrossRef Dasgupta D, Nakao Y, Mauer AS, Thompson JM, Sehrawat TS, Liao CY, Krishnan A, Lucien F, Guo Q, Liu M, Xue F, Fukushima M, Katsumi T, Bansal A, Pandey MK, Maiers JL, DeGrado T, Ibrahim SH, Revzin A, Pavelko KD, Barry MA, Kaufman RJ, Malhi H. IRE1A stimulates hepatocyte-derived extracellular vesicles that promote inflammation in mice with steatohepatitis. Gastroenterology. 2020;159:1487-1503 e1417.PubMedCrossRef
47.
Zurück zum Zitat Zhang C, Wang G, Zheng Z, Maddipati KR, Zhang X, Dyson G, Williams P, Duncan SA, Kaufman RJ, Zhang K. Endoplasmic reticulum-tethered transcription factor cAMP responsive element-binding protein, hepatocyte specific, regulates hepatic lipogenesis, fatty acid oxidation, and lipolysis upon metabolic stress in mice. Hepatology. 2012;55:1070–82.PubMedCrossRef Zhang C, Wang G, Zheng Z, Maddipati KR, Zhang X, Dyson G, Williams P, Duncan SA, Kaufman RJ, Zhang K. Endoplasmic reticulum-tethered transcription factor cAMP responsive element-binding protein, hepatocyte specific, regulates hepatic lipogenesis, fatty acid oxidation, and lipolysis upon metabolic stress in mice. Hepatology. 2012;55:1070–82.PubMedCrossRef
48.
Zurück zum Zitat Zhang B, Li M, Zou Y, Guo H, Zhang B, Xia C, Zhang H, Yang W, Xu C. NFkappaB/Orai1 facilitates endoplasmic reticulum stress by oxidative stress in the pathogenesis of non-alcoholic fatty liver disease. Front Cell Dev Biol. 2019;7:202.PubMedPubMedCentralCrossRef Zhang B, Li M, Zou Y, Guo H, Zhang B, Xia C, Zhang H, Yang W, Xu C. NFkappaB/Orai1 facilitates endoplasmic reticulum stress by oxidative stress in the pathogenesis of non-alcoholic fatty liver disease. Front Cell Dev Biol. 2019;7:202.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Gregor MF, Hotamisligil GS. Thematic review series: adipocyte biology. Adipocyte stress: the endoplasmic reticulum and metabolic disease. J Lipid Res. 2007;48:1905–14.PubMedCrossRef Gregor MF, Hotamisligil GS. Thematic review series: adipocyte biology. Adipocyte stress: the endoplasmic reticulum and metabolic disease. J Lipid Res. 2007;48:1905–14.PubMedCrossRef
50.
Zurück zum Zitat Zhang S, Wong YT, Tang KY, Kwan HY, Su T. Chinese medicinal herbs targeting the gut-liver axis and adipose tissue-liver axis for non-alcoholic fatty liver disease treatments: the ancient wisdom and modern science. Front Endocrinol (Lausanne). 2020;11:572729.PubMedCrossRef Zhang S, Wong YT, Tang KY, Kwan HY, Su T. Chinese medicinal herbs targeting the gut-liver axis and adipose tissue-liver axis for non-alcoholic fatty liver disease treatments: the ancient wisdom and modern science. Front Endocrinol (Lausanne). 2020;11:572729.PubMedCrossRef
51.
Zurück zum Zitat Namjou B, Lingren T, Huang Y, Parameswaran S, Cobb BL, Stanaway IB, Connolly JJ, Mentch FD, Benoit B, Niu X, Wei WQ, Carroll RJ, Pacheco JA, Harley ITW, Divanovic S, Carrell DS, Larson EB, Carey DJ, Verma S, Ritchie MD, Gharavi AG, Murphy S, Williams MS, Crosslin DR, Jarvik GP, Kullo IJ, Hakonarson H, Li R, e MN, Xanthakos SA, Harley JB. GWAS and enrichment analyses of non-alcoholic fatty liver disease identify new trait-associated genes and pathways across eMERGE Network. BMC Med. 2019;17:135.PubMedPubMedCentralCrossRef Namjou B, Lingren T, Huang Y, Parameswaran S, Cobb BL, Stanaway IB, Connolly JJ, Mentch FD, Benoit B, Niu X, Wei WQ, Carroll RJ, Pacheco JA, Harley ITW, Divanovic S, Carrell DS, Larson EB, Carey DJ, Verma S, Ritchie MD, Gharavi AG, Murphy S, Williams MS, Crosslin DR, Jarvik GP, Kullo IJ, Hakonarson H, Li R, e MN, Xanthakos SA, Harley JB. GWAS and enrichment analyses of non-alcoholic fatty liver disease identify new trait-associated genes and pathways across eMERGE Network. BMC Med. 2019;17:135.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Schwimmer JB, Celedon MA, Lavine JE, Salem R, Campbell N, Schork NJ, Shiehmorteza M, Yokoo T, Chavez A, Middleton MS, Sirlin CB. Heritability of nonalcoholic fatty liver disease. Gastroenterology. 2009;136:1585–92.PubMedCrossRef Schwimmer JB, Celedon MA, Lavine JE, Salem R, Campbell N, Schork NJ, Shiehmorteza M, Yokoo T, Chavez A, Middleton MS, Sirlin CB. Heritability of nonalcoholic fatty liver disease. Gastroenterology. 2009;136:1585–92.PubMedCrossRef
53.
Zurück zum Zitat Seidman JS, Troutman TD, Sakai M, Gola A, Spann NJ, Bennett H, Bruni CM, Ouyang Z, Li RZ, Sun X, Vu BT, Pasillas MP, Ego KM, Gosselin D, Link VM, Chong LW, Evans RM, Thompson BM, McDonald JG, Hosseini M, Witztum JL, Germain RN, Glass CK. Niche-specific reprogramming of epigenetic landscapes drives myeloid cell diversity in nonalcoholic steatohepatitis. Immunity. 2020;52:1057-1074e1057.PubMedPubMedCentralCrossRef Seidman JS, Troutman TD, Sakai M, Gola A, Spann NJ, Bennett H, Bruni CM, Ouyang Z, Li RZ, Sun X, Vu BT, Pasillas MP, Ego KM, Gosselin D, Link VM, Chong LW, Evans RM, Thompson BM, McDonald JG, Hosseini M, Witztum JL, Germain RN, Glass CK. Niche-specific reprogramming of epigenetic landscapes drives myeloid cell diversity in nonalcoholic steatohepatitis. Immunity. 2020;52:1057-1074e1057.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat You X, Liu F, Binder M, Vedder A, Lasho T, Wen Z, Gao X, Flietner E, Rajagopalan A, Zhou Y, Finke C, Mangaonkar A, Liao R, Kong G, Ranheim EA, Droin N, Hunter AM, Nikolaev S, Balasis M, Abdel-Wahab O, Levine RL, Will B, Nadiminti KVG, Yang D, Geissler K, Solary E, Xu W, Padron E, Patnaik MM, Zhang J. Asxl1 loss cooperates with oncogenic Nras in mice to reprogram the immune microenvironment and drive leukemic transformation. Blood. 2022;139:1066–79.PubMedPubMedCentralCrossRef You X, Liu F, Binder M, Vedder A, Lasho T, Wen Z, Gao X, Flietner E, Rajagopalan A, Zhou Y, Finke C, Mangaonkar A, Liao R, Kong G, Ranheim EA, Droin N, Hunter AM, Nikolaev S, Balasis M, Abdel-Wahab O, Levine RL, Will B, Nadiminti KVG, Yang D, Geissler K, Solary E, Xu W, Padron E, Patnaik MM, Zhang J. Asxl1 loss cooperates with oncogenic Nras in mice to reprogram the immune microenvironment and drive leukemic transformation. Blood. 2022;139:1066–79.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Luo X, Hong L, Cheng C, Li N, Zhao X, Shi F, Liu J, Fan J, Zhou J, Bode AM, Cao Y. DNMT1 mediates metabolic reprogramming induced by Epstein-Barr virus latent membrane protein 1 and reversed by grifolin in nasopharyngeal carcinoma. Cell Death Dis. 2018;9:619.PubMedPubMedCentralCrossRef Luo X, Hong L, Cheng C, Li N, Zhao X, Shi F, Liu J, Fan J, Zhou J, Bode AM, Cao Y. DNMT1 mediates metabolic reprogramming induced by Epstein-Barr virus latent membrane protein 1 and reversed by grifolin in nasopharyngeal carcinoma. Cell Death Dis. 2018;9:619.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Degli Esposti D, Hamelin J, Bosselut N, Saffroy R, Sebagh M, Pommier A, Martel C, Lemoine A. Mitochondrial roles and cytoprotection in chronic liver injury. Biochem Res Int. 2012;2012: 387626.PubMedPubMedCentralCrossRef Degli Esposti D, Hamelin J, Bosselut N, Saffroy R, Sebagh M, Pommier A, Martel C, Lemoine A. Mitochondrial roles and cytoprotection in chronic liver injury. Biochem Res Int. 2012;2012: 387626.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Guerrieri F, Nicoletti C, Adorisio E, Caraccio G, Leonetti P, Zanotti F, Cantatore P. Correlation between decreased expression of mitochondrial F0F1-ATP synthase and low regenerating capability of the liver after partial hepatectomy in hypothyroid rats. J Bioenerg Biomembr. 2000;32:183–91.PubMedCrossRef Guerrieri F, Nicoletti C, Adorisio E, Caraccio G, Leonetti P, Zanotti F, Cantatore P. Correlation between decreased expression of mitochondrial F0F1-ATP synthase and low regenerating capability of the liver after partial hepatectomy in hypothyroid rats. J Bioenerg Biomembr. 2000;32:183–91.PubMedCrossRef
58.
Zurück zum Zitat Pirola CJ, Gianotti TF, Burgueno AL, Rey-Funes M, Loidl CF, Mallardi P, Martino JS, Castano GO, Sookoian S. Epigenetic modification of liver mitochondrial DNA is associated with histological severity of nonalcoholic fatty liver disease. Gut. 2013;62:1356–63.PubMedCrossRef Pirola CJ, Gianotti TF, Burgueno AL, Rey-Funes M, Loidl CF, Mallardi P, Martino JS, Castano GO, Sookoian S. Epigenetic modification of liver mitochondrial DNA is associated with histological severity of nonalcoholic fatty liver disease. Gut. 2013;62:1356–63.PubMedCrossRef
60.
Zurück zum Zitat Mao H, Chen W, Chen L, Li L. Potential role of mitochondria-associated endoplasmic reticulum membrane proteins in diseases. Biochem Pharmacol. 2022;199: 115011.PubMedCrossRef Mao H, Chen W, Chen L, Li L. Potential role of mitochondria-associated endoplasmic reticulum membrane proteins in diseases. Biochem Pharmacol. 2022;199: 115011.PubMedCrossRef
61.
Zurück zum Zitat Mansouri A, Gattolliat CH, Asselah T. Mitochondrial dysfunction and signaling in chronic liver diseases. Gastroenterology. 2018;155:629–47.PubMedCrossRef Mansouri A, Gattolliat CH, Asselah T. Mitochondrial dysfunction and signaling in chronic liver diseases. Gastroenterology. 2018;155:629–47.PubMedCrossRef
62.
Zurück zum Zitat Yin PH, Wu CC, Lin JC, Chi CW, Wei YH, Lee HC. Somatic mutations of mitochondrial genome in hepatocellular carcinoma. Mitochondrion. 2010;10:174–82.PubMedCrossRef Yin PH, Wu CC, Lin JC, Chi CW, Wei YH, Lee HC. Somatic mutations of mitochondrial genome in hepatocellular carcinoma. Mitochondrion. 2010;10:174–82.PubMedCrossRef
63.
Zurück zum Zitat Doghman-Bouguerra M, Lalli E. ER-mitochondria interactions: both strength and weakness within cancer cells. Biochim Biophys Acta Mol Cell Res. 2019;1866:650–62.PubMedCrossRef Doghman-Bouguerra M, Lalli E. ER-mitochondria interactions: both strength and weakness within cancer cells. Biochim Biophys Acta Mol Cell Res. 2019;1866:650–62.PubMedCrossRef
64.
Zurück zum Zitat Cox CS, McKay SE, Holmbeck MA, Christian BE, Scortea AC, Tsay AJ, Newman LE, Shadel GS. Mitohormesis in mice via sustained basal activation of mitochondrial and antioxidant signaling. Cell Metab. 2018;28:776-786 e775.PubMedPubMedCentralCrossRef Cox CS, McKay SE, Holmbeck MA, Christian BE, Scortea AC, Tsay AJ, Newman LE, Shadel GS. Mitohormesis in mice via sustained basal activation of mitochondrial and antioxidant signaling. Cell Metab. 2018;28:776-786 e775.PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Coen PM, Musci RV, Hinkley JM, Miller BF. Mitochondria as a target for mitigating sarcopenia. Front Physiol. 2018;9:1883.PubMedCrossRef Coen PM, Musci RV, Hinkley JM, Miller BF. Mitochondria as a target for mitigating sarcopenia. Front Physiol. 2018;9:1883.PubMedCrossRef
66.
69.
Zurück zum Zitat Ishihara N, Eura Y, Mihara K. Mitofusin 1 and 2 play distinct roles in mitochondrial fusion reactions via GTPase activity. J Cell Sci. 2004;117:6535–46.PubMedCrossRef Ishihara N, Eura Y, Mihara K. Mitofusin 1 and 2 play distinct roles in mitochondrial fusion reactions via GTPase activity. J Cell Sci. 2004;117:6535–46.PubMedCrossRef
70.
Zurück zum Zitat Sood A, Jeyaraju DV, Prudent J, Caron A, Lemieux P, McBride HM, Laplante M, Toth K, Pellegrini L. A Mitofusin-2-dependent inactivating cleavage of Opa1 links changes in mitochondria cristae and ER contacts in the postprandial liver. Proc Natl Acad Sci U S A. 2014;111:16017–22.PubMedPubMedCentralCrossRef Sood A, Jeyaraju DV, Prudent J, Caron A, Lemieux P, McBride HM, Laplante M, Toth K, Pellegrini L. A Mitofusin-2-dependent inactivating cleavage of Opa1 links changes in mitochondria cristae and ER contacts in the postprandial liver. Proc Natl Acad Sci U S A. 2014;111:16017–22.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Varanita T, Soriano ME, Romanello V, Zaglia T, Quintana-Cabrera R, Semenzato M, Menabo R, Costa V, Civiletto G, Pesce P, Viscomi C, Zeviani M, Di Lisa F, Mongillo M, Sandri M, Scorrano L. The OPA1-dependent mitochondrial cristae remodeling pathway controls atrophic, apoptotic, and ischemic tissue damage. Cell Metab. 2015;21:834–44.PubMedPubMedCentralCrossRef Varanita T, Soriano ME, Romanello V, Zaglia T, Quintana-Cabrera R, Semenzato M, Menabo R, Costa V, Civiletto G, Pesce P, Viscomi C, Zeviani M, Di Lisa F, Mongillo M, Sandri M, Scorrano L. The OPA1-dependent mitochondrial cristae remodeling pathway controls atrophic, apoptotic, and ischemic tissue damage. Cell Metab. 2015;21:834–44.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Kushnareva Y, Andreyev AY, Kuwana T, Newmeyer DD. Bax activation initiates the assembly of a multimeric catalyst that facilitates bax pore formation in mitochondrial outer membranes. PLoS Biol. 2012;10: e1001394.PubMedPubMedCentralCrossRef Kushnareva Y, Andreyev AY, Kuwana T, Newmeyer DD. Bax activation initiates the assembly of a multimeric catalyst that facilitates bax pore formation in mitochondrial outer membranes. PLoS Biol. 2012;10: e1001394.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Wang Z, Jiang H, Chen S, Du F, Wang X. The mitochondrial phosphatase PGAM5 functions at the convergence point of multiple necrotic death pathways. Cell. 2012;148:228–43.PubMedCrossRef Wang Z, Jiang H, Chen S, Du F, Wang X. The mitochondrial phosphatase PGAM5 functions at the convergence point of multiple necrotic death pathways. Cell. 2012;148:228–43.PubMedCrossRef
74.
Zurück zum Zitat Dickey AS, Strack S. PKA/AKAP1 and PP2A/Bbeta2 regulate neuronal morphogenesis via Drp1 phosphorylation and mitochondrial bioenergetics. J Neurosci. 2011;31:15716–26.PubMedPubMedCentralCrossRef Dickey AS, Strack S. PKA/AKAP1 and PP2A/Bbeta2 regulate neuronal morphogenesis via Drp1 phosphorylation and mitochondrial bioenergetics. J Neurosci. 2011;31:15716–26.PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Harborne SPD, Kunji ERS. Calcium-regulated mitochondrial ATP-Mg/P(i) carriers evolved from a fusion of an EF-hand regulatory domain with a mitochondrial ADP/ATP carrier-like domain. IUBMB Life. 2018;70:1222–32.PubMedPubMedCentralCrossRef Harborne SPD, Kunji ERS. Calcium-regulated mitochondrial ATP-Mg/P(i) carriers evolved from a fusion of an EF-hand regulatory domain with a mitochondrial ADP/ATP carrier-like domain. IUBMB Life. 2018;70:1222–32.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Park SJ, Ahmad F, Um JH, Brown AL, Xu X, Kang H, Ke H, Feng X, Ryall J, Philp A, Schenk S, Kim MK, Sartorelli V, Chung JH. Specific Sirt1 activator-mediated improvement in glucose homeostasis requires Sirt1-independent activation of AMPK. EBioMedicine. 2017;18:128–38.PubMedPubMedCentralCrossRef Park SJ, Ahmad F, Um JH, Brown AL, Xu X, Kang H, Ke H, Feng X, Ryall J, Philp A, Schenk S, Kim MK, Sartorelli V, Chung JH. Specific Sirt1 activator-mediated improvement in glucose homeostasis requires Sirt1-independent activation of AMPK. EBioMedicine. 2017;18:128–38.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Gomes LC, Di Benedetto G, Scorrano L. During autophagy mitochondria elongate, are spared from degradation and sustain cell viability. Nat Cell Biol. 2011;13:589–98.PubMedPubMedCentralCrossRef Gomes LC, Di Benedetto G, Scorrano L. During autophagy mitochondria elongate, are spared from degradation and sustain cell viability. Nat Cell Biol. 2011;13:589–98.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Rambold AS, Kostelecky B, Elia N, Lippincott-Schwartz J. Tubular network formation protects mitochondria from autophagosomal degradation during nutrient starvation. Proc Natl Acad Sci U S A. 2011;108:10190–5.PubMedPubMedCentralCrossRef Rambold AS, Kostelecky B, Elia N, Lippincott-Schwartz J. Tubular network formation protects mitochondria from autophagosomal degradation during nutrient starvation. Proc Natl Acad Sci U S A. 2011;108:10190–5.PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Zhu L, Wu X, Liao R. Mechanism and regulation of mitophagy in nonalcoholic fatty liver disease (NAFLD): a mini-review. Life Sci. 2023;312: 121162.PubMedCrossRef Zhu L, Wu X, Liao R. Mechanism and regulation of mitophagy in nonalcoholic fatty liver disease (NAFLD): a mini-review. Life Sci. 2023;312: 121162.PubMedCrossRef
81.
Zurück zum Zitat Lazarou M, Sliter DA, Kane LA, Sarraf SA, Wang C, Burman JL, Sideris DP, Fogel AI, Youle RJ. The ubiquitin kinase PINK1 recruits autophagy receptors to induce mitophagy. Nature. 2015;524:309–14.PubMedPubMedCentralCrossRef Lazarou M, Sliter DA, Kane LA, Sarraf SA, Wang C, Burman JL, Sideris DP, Fogel AI, Youle RJ. The ubiquitin kinase PINK1 recruits autophagy receptors to induce mitophagy. Nature. 2015;524:309–14.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Narendra DP, Jin SM, Tanaka A, Suen DF, Gautier CA, Shen J, Cookson MR, Youle RJ. PINK1 is selectively stabilized on impaired mitochondria to activate parkin. PLoS Biol. 2010;8: e1000298.PubMedPubMedCentralCrossRef Narendra DP, Jin SM, Tanaka A, Suen DF, Gautier CA, Shen J, Cookson MR, Youle RJ. PINK1 is selectively stabilized on impaired mitochondria to activate parkin. PLoS Biol. 2010;8: e1000298.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Vranas M, Lu Y, Rasool S, Croteau N, Krett JD, Sauve V, Gehring K, Fon EA, Durcan TM, Trempe JF. Selective localization of Mfn2 near PINK1 enables its preferential ubiquitination by parkin on mitochondria. Open Biol. 2022;12: 210255.PubMedPubMedCentralCrossRef Vranas M, Lu Y, Rasool S, Croteau N, Krett JD, Sauve V, Gehring K, Fon EA, Durcan TM, Trempe JF. Selective localization of Mfn2 near PINK1 enables its preferential ubiquitination by parkin on mitochondria. Open Biol. 2022;12: 210255.PubMedPubMedCentralCrossRef
84.
Zurück zum Zitat Glick D, Zhang W, Beaton M, Marsboom G, Gruber M, Simon MC, Hart J, Dorn GW 2, Brady MJ, Macleod KF. BNip3 regulates mitochondrial function and lipid metabolism in the liver. Mol Cell Biol. 2012;32:2570–84.PubMedPubMedCentralCrossRef Glick D, Zhang W, Beaton M, Marsboom G, Gruber M, Simon MC, Hart J, Dorn GW 2, Brady MJ, Macleod KF. BNip3 regulates mitochondrial function and lipid metabolism in the liver. Mol Cell Biol. 2012;32:2570–84.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Suomalainen A, Battersby BJ. Mitochondrial diseases: the contribution of organelle stress responses to pathology. Nat Rev Mol Cell Biol. 2018;19:77–92.PubMedCrossRef Suomalainen A, Battersby BJ. Mitochondrial diseases: the contribution of organelle stress responses to pathology. Nat Rev Mol Cell Biol. 2018;19:77–92.PubMedCrossRef
86.
Zurück zum Zitat Gurung P, Lukens JR, Kanneganti TD. Mitochondria: diversity in the regulation of the NLRP3 inflammasome. Trends Mol Med. 2015;21:193–201.PubMedCrossRef Gurung P, Lukens JR, Kanneganti TD. Mitochondria: diversity in the regulation of the NLRP3 inflammasome. Trends Mol Med. 2015;21:193–201.PubMedCrossRef
87.
Zurück zum Zitat Moore MP, Cunningham RP, Meers GM, Johnson SA, Wheeler AA, Ganga RR, Spencer NM, Pitt JB, Diaz-Arias A, Swi AIA, Hammoud GM, Ibdah JA, Parks EJ, Rector RS. Compromised hepatic mitochondrial fatty acid oxidation and reduced markers of mitochondrial turnover in human NAFLD. Hepatology. 2022;76:1452–65.PubMedCrossRef Moore MP, Cunningham RP, Meers GM, Johnson SA, Wheeler AA, Ganga RR, Spencer NM, Pitt JB, Diaz-Arias A, Swi AIA, Hammoud GM, Ibdah JA, Parks EJ, Rector RS. Compromised hepatic mitochondrial fatty acid oxidation and reduced markers of mitochondrial turnover in human NAFLD. Hepatology. 2022;76:1452–65.PubMedCrossRef
88.
Zurück zum Zitat Zhou T, Chang L, Luo Y, Zhou Y, Zhang J. Corrigendum to “Mst1 inhibition attenuates non-alcoholic fatty liver disease via reversing parkin-related mitophagy” [Redox Biol. 21 (2019 Feb) 101120]. Redox Biol. 2020;28: 101299.PubMedCrossRef Zhou T, Chang L, Luo Y, Zhou Y, Zhang J. Corrigendum to “Mst1 inhibition attenuates non-alcoholic fatty liver disease via reversing parkin-related mitophagy” [Redox Biol. 21 (2019 Feb) 101120]. Redox Biol. 2020;28: 101299.PubMedCrossRef
89.
Zurück zum Zitat Perra A, Simbula G, Simbula M, Pibiri M, Kowalik MA, Sulas P, Cocco MT, Ledda-Columbano GM, Columbano A. Thyroid hormone (T3) and TRbeta agonist GC-1 inhibit/reverse nonalcoholic fatty liver in rats. FASEB J. 2008;22:2981–9.PubMedCrossRef Perra A, Simbula G, Simbula M, Pibiri M, Kowalik MA, Sulas P, Cocco MT, Ledda-Columbano GM, Columbano A. Thyroid hormone (T3) and TRbeta agonist GC-1 inhibit/reverse nonalcoholic fatty liver in rats. FASEB J. 2008;22:2981–9.PubMedCrossRef
90.
Zurück zum Zitat Cable EE, Finn PD, Stebbins JW, Hou J, Ito BR, van Poelje PD, Linemeyer DL, Erion MD. Reduction of hepatic steatosis in rats and mice after treatment with a liver-targeted thyroid hormone receptor agonist. Hepatology. 2009;49:407–17.PubMedCrossRef Cable EE, Finn PD, Stebbins JW, Hou J, Ito BR, van Poelje PD, Linemeyer DL, Erion MD. Reduction of hepatic steatosis in rats and mice after treatment with a liver-targeted thyroid hormone receptor agonist. Hepatology. 2009;49:407–17.PubMedCrossRef
93.
Zurück zum Zitat Vanderperre B, Bender T, Kunji ER, Martinou JC. Mitochondrial pyruvate import and its effects on homeostasis. Curr Opin Cell Biol. 2015;33:35–41.PubMedCrossRef Vanderperre B, Bender T, Kunji ER, Martinou JC. Mitochondrial pyruvate import and its effects on homeostasis. Curr Opin Cell Biol. 2015;33:35–41.PubMedCrossRef
94.
Zurück zum Zitat Houten SM, Violante S, Ventura FV, Wanders RJ. The Biochemistry and Physiology of mitochondrial fatty acid beta-oxidation and its genetic Disorders. Annu Rev Physiol. 2016;78:23–44.PubMedCrossRef Houten SM, Violante S, Ventura FV, Wanders RJ. The Biochemistry and Physiology of mitochondrial fatty acid beta-oxidation and its genetic Disorders. Annu Rev Physiol. 2016;78:23–44.PubMedCrossRef
95.
Zurück zum Zitat Adeva-Andany MM, Carneiro-Freire N, Seco-Filgueira M, Fernandez-Fernandez C. Mourino-Bayolo, mitochondrial beta-oxidation of saturated fatty acids in humans. Mitochondrion. 2019;46:73–90.PubMedCrossRef Adeva-Andany MM, Carneiro-Freire N, Seco-Filgueira M, Fernandez-Fernandez C. Mourino-Bayolo, mitochondrial beta-oxidation of saturated fatty acids in humans. Mitochondrion. 2019;46:73–90.PubMedCrossRef
97.
Zurück zum Zitat Varela CD, Farhana A. Biochemistry, Superoxides. In: StatPearls, Treasure Island (FL): StatPearls Publishing; 2023. Varela CD, Farhana A. Biochemistry, Superoxides. In: StatPearls, Treasure Island (FL): StatPearls Publishing; 2023.
98.
Zurück zum Zitat Guha S, Sesili S, Mir IH, Thirunavukkarasu C. Epigenetics and mitochondrial dysfunction insights into the impact of the progression of non-alcoholic fatty liver disease. Cell Biochem Funct. 2023;41:4–19.PubMedCrossRef Guha S, Sesili S, Mir IH, Thirunavukkarasu C. Epigenetics and mitochondrial dysfunction insights into the impact of the progression of non-alcoholic fatty liver disease. Cell Biochem Funct. 2023;41:4–19.PubMedCrossRef
99.
Zurück zum Zitat Boveris A, Chance B. The mitochondrial generation of hydrogen peroxide. General properties and effect of hyperbaric oxygen. Biochem J. 1973;134:707–16.PubMedPubMedCentralCrossRef Boveris A, Chance B. The mitochondrial generation of hydrogen peroxide. General properties and effect of hyperbaric oxygen. Biochem J. 1973;134:707–16.PubMedPubMedCentralCrossRef
100.
Zurück zum Zitat Sanyal AJ, Campbell-Sargent C, Mirshahi F, Rizzo WB, Contos MJ, Sterling RK, Luketic VA, Shiffman ML, Clore JN. Nonalcoholic steatohepatitis: association of insulin resistance and mitochondrial abnormalities. Gastroenterology. 2001;120:1183–92.PubMedCrossRef Sanyal AJ, Campbell-Sargent C, Mirshahi F, Rizzo WB, Contos MJ, Sterling RK, Luketic VA, Shiffman ML, Clore JN. Nonalcoholic steatohepatitis: association of insulin resistance and mitochondrial abnormalities. Gastroenterology. 2001;120:1183–92.PubMedCrossRef
101.
Zurück zum Zitat Meex RCR, Watt MJ. Hepatokines: linking nonalcoholic fatty liver disease and insulin resistance. Nat Rev Endocrinol. 2017;13:509–20.PubMedCrossRef Meex RCR, Watt MJ. Hepatokines: linking nonalcoholic fatty liver disease and insulin resistance. Nat Rev Endocrinol. 2017;13:509–20.PubMedCrossRef
102.
Zurück zum Zitat Yin X, Zheng F, Pan Q, Zhang S, Yu D, Xu Z, Li H. Glucose fluctuation increased hepatocyte apoptosis under lipotoxicity and the involvement of mitochondrial permeability transition opening. J Mol Endocrinol. 2015;55:169–81.PubMedCrossRef Yin X, Zheng F, Pan Q, Zhang S, Yu D, Xu Z, Li H. Glucose fluctuation increased hepatocyte apoptosis under lipotoxicity and the involvement of mitochondrial permeability transition opening. J Mol Endocrinol. 2015;55:169–81.PubMedCrossRef
103.
Zurück zum Zitat Navarro CDC, Figueira TR, Francisco A, Dal’Bo GA, Ronchi JA, Rovani JC, Escanhoela CAF, Oliveira HCF, Castilho RF, Vercesi AE. Redox imbalance due to the loss of mitochondrial NAD(P)-transhydrogenase markedly aggravates high fat diet-induced fatty liver disease in mice. Free Radic Biol Med. 2017;113:190–202.PubMedCrossRef Navarro CDC, Figueira TR, Francisco A, Dal’Bo GA, Ronchi JA, Rovani JC, Escanhoela CAF, Oliveira HCF, Castilho RF, Vercesi AE. Redox imbalance due to the loss of mitochondrial NAD(P)-transhydrogenase markedly aggravates high fat diet-induced fatty liver disease in mice. Free Radic Biol Med. 2017;113:190–202.PubMedCrossRef
104.
Zurück zum Zitat King AL, Swain TM, Mao Z, Udoh US, Oliva CR, Betancourt AM, Griguer CE, Crowe DR, Lesort M, Bailey SM. Involvement of the mitochondrial permeability transition pore in chronic ethanol-mediated liver injury in mice. Am J Physiol Gastrointest Liver Physiol. 2014;306:G265-277.PubMedCrossRef King AL, Swain TM, Mao Z, Udoh US, Oliva CR, Betancourt AM, Griguer CE, Crowe DR, Lesort M, Bailey SM. Involvement of the mitochondrial permeability transition pore in chronic ethanol-mediated liver injury in mice. Am J Physiol Gastrointest Liver Physiol. 2014;306:G265-277.PubMedCrossRef
105.
Zurück zum Zitat Begriche K, Massart J, Robin MA, Borgne-Sanchez A, Fromenty B. Drug-induced toxicity on mitochondria and lipid metabolism: mechanistic diversity and deleterious consequences for the liver. J Hepatol. 2011;54:773–94.PubMedCrossRef Begriche K, Massart J, Robin MA, Borgne-Sanchez A, Fromenty B. Drug-induced toxicity on mitochondria and lipid metabolism: mechanistic diversity and deleterious consequences for the liver. J Hepatol. 2011;54:773–94.PubMedCrossRef
106.
Zurück zum Zitat Caldwell SH, Crespo DM. The spectrum expanded: cryptogenic cirrhosis and the natural history of non-alcoholic fatty liver disease. J Hepatol. 2004;40:578–84.PubMedCrossRef Caldwell SH, Crespo DM. The spectrum expanded: cryptogenic cirrhosis and the natural history of non-alcoholic fatty liver disease. J Hepatol. 2004;40:578–84.PubMedCrossRef
107.
Zurück zum Zitat Petrosillo G, Portincasa P, Grattagliano I, Casanova G, Matera M, Ruggiero FM, Ferri D, Paradies G. Mitochondrial dysfunction in rat with nonalcoholic fatty liver involvement of complex I, reactive oxygen species and cardiolipin. Biochim Biophys Acta. 2007;1767:1260–7.PubMedCrossRef Petrosillo G, Portincasa P, Grattagliano I, Casanova G, Matera M, Ruggiero FM, Ferri D, Paradies G. Mitochondrial dysfunction in rat with nonalcoholic fatty liver involvement of complex I, reactive oxygen species and cardiolipin. Biochim Biophys Acta. 2007;1767:1260–7.PubMedCrossRef
108.
Zurück zum Zitat Meex RCR, Blaak EE. Mitochondrial dysfunction is a key pathway that links saturated fat intake to the development and progression of NAFLD. Mol Nutr Food Res. 2021;65:e1900942.PubMedCrossRef Meex RCR, Blaak EE. Mitochondrial dysfunction is a key pathway that links saturated fat intake to the development and progression of NAFLD. Mol Nutr Food Res. 2021;65:e1900942.PubMedCrossRef
109.
Zurück zum Zitat Sunny NE, Bril F, Cusi K. Mitochondrial adaptation in nonalcoholic fatty liver disease: novel mechanisms and treatment strategies. Trends Endocrinol Metab. 2017;28:250–60.PubMedCrossRef Sunny NE, Bril F, Cusi K. Mitochondrial adaptation in nonalcoholic fatty liver disease: novel mechanisms and treatment strategies. Trends Endocrinol Metab. 2017;28:250–60.PubMedCrossRef
110.
Zurück zum Zitat Begriche K, Igoudjil A, Pessayre D, Fromenty B. Mitochondrial dysfunction in NASH: causes, consequences and possible means to prevent it. Mitochondrion. 2006;6:1–28.PubMedCrossRef Begriche K, Igoudjil A, Pessayre D, Fromenty B. Mitochondrial dysfunction in NASH: causes, consequences and possible means to prevent it. Mitochondrion. 2006;6:1–28.PubMedCrossRef
111.
Zurück zum Zitat Zhao Q, Liu J, Deng H, Ma R, Liao JY, Liang H, Hu J, Li J, Guo Z, Cai J, Xu X, Gao Z, Su S. Targeting Mitochondria-Located circRNA SCAR Alleviates NASH via Reducing mROS Output, Cell, 183 (2020) 76–9322.PubMedCrossRef Zhao Q, Liu J, Deng H, Ma R, Liao JY, Liang H, Hu J, Li J, Guo Z, Cai J, Xu X, Gao Z, Su S. Targeting Mitochondria-Located circRNA SCAR Alleviates NASH via Reducing mROS Output, Cell, 183 (2020) 76–9322.PubMedCrossRef
112.
Zurück zum Zitat Patterson RE, Kalavalapalli S, Williams CM, Nautiyal M, Mathew JT, Martinez J, Reinhard MK, McDougall DJ, Rocca JR, Yost RA, Cusi K, Garrett TJ, Sunny NE. Lipotoxicity in steatohepatitis occurs despite an increase in tricarboxylic acid cycle activity. Am J Physiol Endocrinol Metab. 2016;310:E484-494.PubMedPubMedCentralCrossRef Patterson RE, Kalavalapalli S, Williams CM, Nautiyal M, Mathew JT, Martinez J, Reinhard MK, McDougall DJ, Rocca JR, Yost RA, Cusi K, Garrett TJ, Sunny NE. Lipotoxicity in steatohepatitis occurs despite an increase in tricarboxylic acid cycle activity. Am J Physiol Endocrinol Metab. 2016;310:E484-494.PubMedPubMedCentralCrossRef
113.
Zurück zum Zitat Merry TL, Tran M, Dodd GT, Mangiafico SP, Wiede F, Kaur S, McLean CL, Andrikopoulos S, Tiganis T. Hepatocyte glutathione peroxidase-1 deficiency improves hepatic glucose metabolism and decreases steatohepatitis in mice. Diabetologia. 2016;59:2632–44.PubMedCrossRef Merry TL, Tran M, Dodd GT, Mangiafico SP, Wiede F, Kaur S, McLean CL, Andrikopoulos S, Tiganis T. Hepatocyte glutathione peroxidase-1 deficiency improves hepatic glucose metabolism and decreases steatohepatitis in mice. Diabetologia. 2016;59:2632–44.PubMedCrossRef
114.
Zurück zum Zitat Tsuchida T, Lee YA, Fujiwara N, Ybanez M, Allen B, Martins S, Fiel MI, Goossens N, Chou HI, Hoshida Y, Friedman SL. A simple diet- and chemical-induced murine NASH model with rapid progression of steatohepatitis, fibrosis and liver cancer. J Hepatol. 2018;69:385–95.PubMedPubMedCentralCrossRef Tsuchida T, Lee YA, Fujiwara N, Ybanez M, Allen B, Martins S, Fiel MI, Goossens N, Chou HI, Hoshida Y, Friedman SL. A simple diet- and chemical-induced murine NASH model with rapid progression of steatohepatitis, fibrosis and liver cancer. J Hepatol. 2018;69:385–95.PubMedPubMedCentralCrossRef
115.
Zurück zum Zitat Chachay VS, Macdonald GA, Martin JH, Whitehead JP, O’Moore-Sullivan TM, Lee P, Franklin M, Klein K, Taylor PJ, Ferguson M, Coombes JS, Thomas GP, Cowin GJ, Kirkpatrick CM, Prins JB, Hickman IJ. Resveratrol does not benefit patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol. 2014;12:2092-103 e2091-2096.PubMedCrossRef Chachay VS, Macdonald GA, Martin JH, Whitehead JP, O’Moore-Sullivan TM, Lee P, Franklin M, Klein K, Taylor PJ, Ferguson M, Coombes JS, Thomas GP, Cowin GJ, Kirkpatrick CM, Prins JB, Hickman IJ. Resveratrol does not benefit patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol. 2014;12:2092-103 e2091-2096.PubMedCrossRef
116.
Zurück zum Zitat Abenavoli L, Larussa T, Corea A, Procopio AC, Boccuto L, Dallio M, Federico A, Luzza F. Dietary polyphenols and non-alcoholic fatty liver disease. Nutrients. 2021;13. Abenavoli L, Larussa T, Corea A, Procopio AC, Boccuto L, Dallio M, Federico A, Luzza F. Dietary polyphenols and non-alcoholic fatty liver disease. Nutrients. 2021;13.
117.
Zurück zum Zitat Jakubczyk K, Skonieczna-Zydecka K, Kaldunska J, Stachowska E, Gutowska I, Janda K. Effects of resveratrol supplementation in patients with non-alcoholic fatty liver disease: a meta-analysis. Nutrients. 2020;12. Jakubczyk K, Skonieczna-Zydecka K, Kaldunska J, Stachowska E, Gutowska I, Janda K. Effects of resveratrol supplementation in patients with non-alcoholic fatty liver disease: a meta-analysis. Nutrients. 2020;12.
118.
Zurück zum Zitat Park MN, Rahman MA, Rahman MH, Kim JW, Choi M, Kim JW, Choi J, Moon M, Ahmed KR, Kim B. Potential therapeutic implication of herbal medicine in mitochondria-mediated oxidative stress-related liver diseases. Antioxid (Basel). 2022;11. Park MN, Rahman MA, Rahman MH, Kim JW, Choi M, Kim JW, Choi J, Moon M, Ahmed KR, Kim B. Potential therapeutic implication of herbal medicine in mitochondria-mediated oxidative stress-related liver diseases. Antioxid (Basel). 2022;11.
119.
Zurück zum Zitat Vernia S, Cavanagh-Kyros J, Garcia-Haro L, Sabio G, Barrett T, Jung DY, Kim JK, Xu J, Shulha HP, Garber M, Gao G, Davis RJ. The PPARalpha-FGF21 hormone axis contributes to metabolic regulation by the hepatic JNK signaling pathway. Cell Metab. 2014;20:512–25.PubMedPubMedCentralCrossRef Vernia S, Cavanagh-Kyros J, Garcia-Haro L, Sabio G, Barrett T, Jung DY, Kim JK, Xu J, Shulha HP, Garber M, Gao G, Davis RJ. The PPARalpha-FGF21 hormone axis contributes to metabolic regulation by the hepatic JNK signaling pathway. Cell Metab. 2014;20:512–25.PubMedPubMedCentralCrossRef
120.
Zurück zum Zitat Win S, Than TA, Min RW, Aghajan M, Kaplowitz N. c-Jun N-terminal kinase mediates mouse liver injury through a novel Sab (SH3BP5)-dependent pathway leading to inactivation of intramitochondrial Src. Hepatology. 2016;63:1987–2003.PubMedCrossRef Win S, Than TA, Min RW, Aghajan M, Kaplowitz N. c-Jun N-terminal kinase mediates mouse liver injury through a novel Sab (SH3BP5)-dependent pathway leading to inactivation of intramitochondrial Src. Hepatology. 2016;63:1987–2003.PubMedCrossRef
121.
Zurück zum Zitat Win S, Than TA, Zhang J, Oo C, Min RWM, Kaplowitz N. New insights into the role and mechanism of c-Jun-N-terminal kinase signaling in the pathobiology of liver diseases. Hepatology. 2018;67:2013–24.PubMedCrossRef Win S, Than TA, Zhang J, Oo C, Min RWM, Kaplowitz N. New insights into the role and mechanism of c-Jun-N-terminal kinase signaling in the pathobiology of liver diseases. Hepatology. 2018;67:2013–24.PubMedCrossRef
122.
Zurück zum Zitat Krautbauer S, Eisinger K, Lupke M, Wanninger J, Ruemmele P, Hader Y, Weiss TS, Buechler C. Manganese superoxide dismutase is reduced in the liver of male but not female humans and rodents with non-alcoholic fatty liver disease. Exp Mol Pathol. 2013;95:330–5.PubMedCrossRef Krautbauer S, Eisinger K, Lupke M, Wanninger J, Ruemmele P, Hader Y, Weiss TS, Buechler C. Manganese superoxide dismutase is reduced in the liver of male but not female humans and rodents with non-alcoholic fatty liver disease. Exp Mol Pathol. 2013;95:330–5.PubMedCrossRef
123.
Zurück zum Zitat Dou X, Li S, Hu L, Ding L, Ma Y, Ma W, Chai H, Song Z. Glutathione disulfide sensitizes hepatocytes to TNFalpha-mediated cytotoxicity via IKK-beta S-glutathionylation: a potential mechanism underlying non-alcoholic fatty liver disease. Exp Mol Med. 2018;50:1–16.PubMedCrossRef Dou X, Li S, Hu L, Ding L, Ma Y, Ma W, Chai H, Song Z. Glutathione disulfide sensitizes hepatocytes to TNFalpha-mediated cytotoxicity via IKK-beta S-glutathionylation: a potential mechanism underlying non-alcoholic fatty liver disease. Exp Mol Med. 2018;50:1–16.PubMedCrossRef
124.
Zurück zum Zitat Ploumi C, Daskalaki I, Tavernarakis N. Mitochondrial biogenesis and clearance: a balancing act. FEBS J. 2017;284:183–95.PubMedCrossRef Ploumi C, Daskalaki I, Tavernarakis N. Mitochondrial biogenesis and clearance: a balancing act. FEBS J. 2017;284:183–95.PubMedCrossRef
125.
126.
Zurück zum Zitat Picca A, Mankowski RT, Burman JL, Donisi L, Kim JS, Marzetti E, Leeuwenburgh C. Mitochondrial quality control mechanisms as molecular targets in cardiac ageing. Nat Rev Cardiol. 2018;15:543–54.PubMedPubMedCentralCrossRef Picca A, Mankowski RT, Burman JL, Donisi L, Kim JS, Marzetti E, Leeuwenburgh C. Mitochondrial quality control mechanisms as molecular targets in cardiac ageing. Nat Rev Cardiol. 2018;15:543–54.PubMedPubMedCentralCrossRef
127.
Zurück zum Zitat Cioffi F, Giacco A, Petito G, de Matteis R, Senese R, Lombardi A, de Lange P, Moreno M, Goglia F, Lanni A, Silvestri E. Altered mitochondrial quality control in rats with metabolic dysfunction-associated fatty liver disease (MAFLD) induced by high-fat feeding. Genes (Basel). 2022;13. Cioffi F, Giacco A, Petito G, de Matteis R, Senese R, Lombardi A, de Lange P, Moreno M, Goglia F, Lanni A, Silvestri E. Altered mitochondrial quality control in rats with metabolic dysfunction-associated fatty liver disease (MAFLD) induced by high-fat feeding. Genes (Basel). 2022;13.
128.
130.
Zurück zum Zitat Krishnasamy Y, Gooz M, Li L, Lemasters JJ, Zhong Z. Role of mitochondrial depolarization and disrupted mitochondrial homeostasis in non-alcoholic steatohepatitis and fibrosis in mice. Int J Physiol Pathophysiol Pharmacol. 2019;11:190–204.PubMedPubMedCentral Krishnasamy Y, Gooz M, Li L, Lemasters JJ, Zhong Z. Role of mitochondrial depolarization and disrupted mitochondrial homeostasis in non-alcoholic steatohepatitis and fibrosis in mice. Int J Physiol Pathophysiol Pharmacol. 2019;11:190–204.PubMedPubMedCentral
131.
Zurück zum Zitat Krause A, Brade V, Schoerner C, Solbach W, Kalden JR, Burmester GR. T cell proliferation induced by Borrelia burgdorferi in patients with Lyme borreliosis. Autologous serum required for optimum stimulation. Arthritis Rheum. 1991;34:393–402.PubMedCrossRef Krause A, Brade V, Schoerner C, Solbach W, Kalden JR, Burmester GR. T cell proliferation induced by Borrelia burgdorferi in patients with Lyme borreliosis. Autologous serum required for optimum stimulation. Arthritis Rheum. 1991;34:393–402.PubMedCrossRef
132.
Zurück zum Zitat Sheldon RD, Meers GM, Morris EM, Linden MA, Cunningham RP, Ibdah JA, Thyfault JP, Laughlin MH, Rector RS. eNOS deletion impairs mitochondrial quality control and exacerbates Western diet-induced NASH. Am J Physiol Endocrinol Metab. 2019;317:E605-16.PubMedPubMedCentralCrossRef Sheldon RD, Meers GM, Morris EM, Linden MA, Cunningham RP, Ibdah JA, Thyfault JP, Laughlin MH, Rector RS. eNOS deletion impairs mitochondrial quality control and exacerbates Western diet-induced NASH. Am J Physiol Endocrinol Metab. 2019;317:E605-16.PubMedPubMedCentralCrossRef
133.
Zurück zum Zitat Ordureau A, Heo JM, Duda DM, Paulo JA, Olszewski JL, Yanishevski D, Rinehart J, Schulman BA, Harper JW. Defining roles of PARKIN and ubiquitin phosphorylation by PINK1 in mitochondrial quality control using a ubiquitin replacement strategy. Proc Natl Acad Sci U S A. 2015;112:6637–42.PubMedPubMedCentralCrossRef Ordureau A, Heo JM, Duda DM, Paulo JA, Olszewski JL, Yanishevski D, Rinehart J, Schulman BA, Harper JW. Defining roles of PARKIN and ubiquitin phosphorylation by PINK1 in mitochondrial quality control using a ubiquitin replacement strategy. Proc Natl Acad Sci U S A. 2015;112:6637–42.PubMedPubMedCentralCrossRef
134.
Zurück zum Zitat Little JP, Safdar A, Benton CR, Wright DC. Skeletal muscle and beyond: the role of exercise as a mediator of systemic mitochondrial biogenesis. Appl Physiol Nutr Metab. 2011;36:598–607.PubMedCrossRef Little JP, Safdar A, Benton CR, Wright DC. Skeletal muscle and beyond: the role of exercise as a mediator of systemic mitochondrial biogenesis. Appl Physiol Nutr Metab. 2011;36:598–607.PubMedCrossRef
135.
Zurück zum Zitat Stevanovic J, Beleza J, Coxito P, Ascensao A, Magalhaes J. Physical exercise and liver “fitness”: role of mitochondrial function and epigenetics-related mechanisms in non-alcoholic fatty liver disease. Mol Metab. 2020;32:1–14.PubMedCrossRef Stevanovic J, Beleza J, Coxito P, Ascensao A, Magalhaes J. Physical exercise and liver “fitness”: role of mitochondrial function and epigenetics-related mechanisms in non-alcoholic fatty liver disease. Mol Metab. 2020;32:1–14.PubMedCrossRef
136.
Zurück zum Zitat Venditti P, Di Meo S. Antioxidants, tissue damage, and endurance in trained and untrained young male rats. Arch Biochem Biophys. 1996;331:63–8.PubMedCrossRef Venditti P, Di Meo S. Antioxidants, tissue damage, and endurance in trained and untrained young male rats. Arch Biochem Biophys. 1996;331:63–8.PubMedCrossRef
137.
Zurück zum Zitat Ascensao A, Martins MJ, Santos-Alves E, Goncalves IO, Portincasa P, Oliveira PJ, Magalhaes J. Modulation of hepatic redox status and mitochondrial metabolism by exercise: therapeutic strategy for liver diseases. Mitochondrion. 2013;13:862–70.PubMedCrossRef Ascensao A, Martins MJ, Santos-Alves E, Goncalves IO, Portincasa P, Oliveira PJ, Magalhaes J. Modulation of hepatic redox status and mitochondrial metabolism by exercise: therapeutic strategy for liver diseases. Mitochondrion. 2013;13:862–70.PubMedCrossRef
138.
Zurück zum Zitat Ignacio DL, Fortunato RS, Silvestre D, Matta L, de Vansconcelos AL, Carvalho DP, Galina A, Werneck-de-Castro JP. Cavalcanti-de-Albuquerque, physical exercise improves mitochondrial function in ovariectomized rats. J Endocrinol. 2022;254:77–90.PubMedCrossRef Ignacio DL, Fortunato RS, Silvestre D, Matta L, de Vansconcelos AL, Carvalho DP, Galina A, Werneck-de-Castro JP. Cavalcanti-de-Albuquerque, physical exercise improves mitochondrial function in ovariectomized rats. J Endocrinol. 2022;254:77–90.PubMedCrossRef
139.
Zurück zum Zitat Sheldon RD, Laughlin MH, Rector RS. Reduced hepatic eNOS phosphorylation is associated with NAFLD and type 2 diabetes progression and is prevented by daily exercise in hyperphagic OLETF rats. J Appl Physiol (1985). 2014;116:1156–64.PubMedCrossRef Sheldon RD, Laughlin MH, Rector RS. Reduced hepatic eNOS phosphorylation is associated with NAFLD and type 2 diabetes progression and is prevented by daily exercise in hyperphagic OLETF rats. J Appl Physiol (1985). 2014;116:1156–64.PubMedCrossRef
140.
Zurück zum Zitat Hung YH, Linden MA, Gordon A, Rector RS, Buhman KK. Endurance exercise training programs intestinal lipid metabolism in a rat model of obesity and type 2 diabetes. Physiol Rep. 2015;3. Hung YH, Linden MA, Gordon A, Rector RS, Buhman KK. Endurance exercise training programs intestinal lipid metabolism in a rat model of obesity and type 2 diabetes. Physiol Rep. 2015;3.
141.
Zurück zum Zitat Rector RS, Thyfault JP, Laye MJ, Morris RT, Borengasser SJ, Uptergrove GM, Chakravarthy MV, Booth FW, Ibdah JA. Cessation of daily exercise dramatically alters precursors of hepatic steatosis in Otsuka Long-Evans Tokushima fatty (OLETF) rats. J Physiol. 2008;586:4241–9.PubMedPubMedCentralCrossRef Rector RS, Thyfault JP, Laye MJ, Morris RT, Borengasser SJ, Uptergrove GM, Chakravarthy MV, Booth FW, Ibdah JA. Cessation of daily exercise dramatically alters precursors of hepatic steatosis in Otsuka Long-Evans Tokushima fatty (OLETF) rats. J Physiol. 2008;586:4241–9.PubMedPubMedCentralCrossRef
142.
Zurück zum Zitat Rector RS, Thyfault JP, Morris RT, Laye MJ, Borengasser SJ, Booth FW, Ibdah JA. Daily exercise increases hepatic fatty acid oxidation and prevents steatosis in Otsuka Long-Evans Tokushima fatty rats. Am J Physiol Gastrointest Liver Physiol. 2008;294:G619–626.PubMedCrossRef Rector RS, Thyfault JP, Morris RT, Laye MJ, Borengasser SJ, Booth FW, Ibdah JA. Daily exercise increases hepatic fatty acid oxidation and prevents steatosis in Otsuka Long-Evans Tokushima fatty rats. Am J Physiol Gastrointest Liver Physiol. 2008;294:G619–626.PubMedCrossRef
143.
Zurück zum Zitat Sun L, Shen W, Liu Z, Guan S, Liu J, Ding S. Endurance exercise causes mitochondrial and oxidative stress in rat liver: effects of a combination of mitochondrial targeting nutrients. Life Sci. 2010;86:39–44.PubMedCrossRef Sun L, Shen W, Liu Z, Guan S, Liu J, Ding S. Endurance exercise causes mitochondrial and oxidative stress in rat liver: effects of a combination of mitochondrial targeting nutrients. Life Sci. 2010;86:39–44.PubMedCrossRef
146.
Zurück zum Zitat Fernandes MSS, Silva L, Kubrusly MS, Lima T, Muller CR, Americo ALV, Fernandes MP, Cogliati B, Stefano JT, Lagranha CJ, Evangelista FS, Oliveira CP. Aerobic exercise training exerts beneficial effects upon oxidative metabolism and non-enzymatic antioxidant defense in the liver of Leptin Deficiency mice. Front Endocrinol (Lausanne). 2020;11:588502.PubMedCrossRef Fernandes MSS, Silva L, Kubrusly MS, Lima T, Muller CR, Americo ALV, Fernandes MP, Cogliati B, Stefano JT, Lagranha CJ, Evangelista FS, Oliveira CP. Aerobic exercise training exerts beneficial effects upon oxidative metabolism and non-enzymatic antioxidant defense in the liver of Leptin Deficiency mice. Front Endocrinol (Lausanne). 2020;11:588502.PubMedCrossRef
147.
Zurück zum Zitat Goncalves IO, Passos E, Diogo CV, Rocha-Rodrigues S, Santos-Alves E, Oliveira PJ, Ascensao A, Magalhaes J. Exercise mitigates mitochondrial permeability transition pore and quality control mechanisms alterations in nonalcoholic steatohepatitis. Appl Physiol Nutr Metab. 2016;41:298–306.PubMedCrossRef Goncalves IO, Passos E, Diogo CV, Rocha-Rodrigues S, Santos-Alves E, Oliveira PJ, Ascensao A, Magalhaes J. Exercise mitigates mitochondrial permeability transition pore and quality control mechanisms alterations in nonalcoholic steatohepatitis. Appl Physiol Nutr Metab. 2016;41:298–306.PubMedCrossRef
148.
Zurück zum Zitat Farzanegi P, Dana A, Ebrahimpoor Z, Asadi M, Azarbayjani MA. Mechanisms of beneficial effects of exercise training on non-alcoholic fatty liver disease (NAFLD): roles of oxidative stress and inflammation. Eur J Sport Sci. 2019;19:994–1003.PubMedCrossRef Farzanegi P, Dana A, Ebrahimpoor Z, Asadi M, Azarbayjani MA. Mechanisms of beneficial effects of exercise training on non-alcoholic fatty liver disease (NAFLD): roles of oxidative stress and inflammation. Eur J Sport Sci. 2019;19:994–1003.PubMedCrossRef
149.
150.
Zurück zum Zitat Gross B, Pawlak M, Lefebvre P, Staels B. PPARs in obesity-induced T2DM, dyslipidaemia and NAFLD. Nat Rev Endocrinol. 2017;13:36–49.PubMedCrossRef Gross B, Pawlak M, Lefebvre P, Staels B. PPARs in obesity-induced T2DM, dyslipidaemia and NAFLD. Nat Rev Endocrinol. 2017;13:36–49.PubMedCrossRef
151.
Zurück zum Zitat Rotman Y, Sanyal AJ. Current and upcoming pharmacotherapy for non-alcoholic fatty liver disease. Gut. 2017;66:180–90.PubMedCrossRef Rotman Y, Sanyal AJ. Current and upcoming pharmacotherapy for non-alcoholic fatty liver disease. Gut. 2017;66:180–90.PubMedCrossRef
152.
Zurück zum Zitat Pawlak M, Lefebvre P, Staels B. Molecular mechanism of PPARalpha action and its impact on lipid metabolism, inflammation and fibrosis in non-alcoholic fatty liver disease. J Hepatol. 2015;62:720–33.PubMedCrossRef Pawlak M, Lefebvre P, Staels B. Molecular mechanism of PPARalpha action and its impact on lipid metabolism, inflammation and fibrosis in non-alcoholic fatty liver disease. J Hepatol. 2015;62:720–33.PubMedCrossRef
153.
Zurück zum Zitat Musso G, Gambino R, Cassader M, Pagano G. A meta-analysis of randomized trials for the treatment of nonalcoholic fatty liver disease. Hepatology. 2010;52:79–104.PubMedCrossRef Musso G, Gambino R, Cassader M, Pagano G. A meta-analysis of randomized trials for the treatment of nonalcoholic fatty liver disease. Hepatology. 2010;52:79–104.PubMedCrossRef
154.
Zurück zum Zitat Bojic LA, Huff MW. Peroxisome proliferator-activated receptor delta: a multifaceted metabolic player. Curr Opin Lipidol. 2013;24:171–7.PubMedCrossRef Bojic LA, Huff MW. Peroxisome proliferator-activated receptor delta: a multifaceted metabolic player. Curr Opin Lipidol. 2013;24:171–7.PubMedCrossRef
155.
Zurück zum Zitat Odegaard JI, Ricardo-Gonzalez RR, Red Eagle A, Vats D, Morel CR, Goforth MH, Subramanian V, Mukundan L, Ferrante AW, Chawla A. Alternative M2 activation of Kupffer cells by PPARdelta ameliorates obesity-induced insulin resistance. Cell Metab. 2008;7:496–507.PubMedPubMedCentralCrossRef Odegaard JI, Ricardo-Gonzalez RR, Red Eagle A, Vats D, Morel CR, Goforth MH, Subramanian V, Mukundan L, Ferrante AW, Chawla A. Alternative M2 activation of Kupffer cells by PPARdelta ameliorates obesity-induced insulin resistance. Cell Metab. 2008;7:496–507.PubMedPubMedCentralCrossRef
156.
Zurück zum Zitat Riserus U, Sprecher D, Johnson T, Olson E, Hirschberg S, Liu A, Fang Z, Hegde P, Richards D, Sarov-Blat L, Strum JC, Basu S, Cheeseman J, Fielding BA, Humphreys SM, Danoff T, Moore NR, Murgatroyd P, O’Rahilly S, Sutton P, Willson T, Hassall D, Frayn KN, Karpe F. Activation of peroxisome proliferator-activated receptor (PPAR)delta promotes reversal of multiple metabolic abnormalities, reduces oxidative stress, and increases fatty acid oxidation in moderately obese men. Diabetes. 2008;57:332–9.PubMedCrossRef Riserus U, Sprecher D, Johnson T, Olson E, Hirschberg S, Liu A, Fang Z, Hegde P, Richards D, Sarov-Blat L, Strum JC, Basu S, Cheeseman J, Fielding BA, Humphreys SM, Danoff T, Moore NR, Murgatroyd P, O’Rahilly S, Sutton P, Willson T, Hassall D, Frayn KN, Karpe F. Activation of peroxisome proliferator-activated receptor (PPAR)delta promotes reversal of multiple metabolic abnormalities, reduces oxidative stress, and increases fatty acid oxidation in moderately obese men. Diabetes. 2008;57:332–9.PubMedCrossRef
157.
Zurück zum Zitat Sanyal AJ, Chalasani N, Kowdley KV, McCullough A, Diehl AM, Bass NM, Neuschwander-Tetri BA, Lavine JE, Tonascia J, Unalp A, Van Natta M, Clark J, Brunt EM, Kleiner DE, Hoofnagle JH, Robuck PR, Nash CRN. Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis. N Engl J Med. 2010;362:1675–85.PubMedPubMedCentralCrossRef Sanyal AJ, Chalasani N, Kowdley KV, McCullough A, Diehl AM, Bass NM, Neuschwander-Tetri BA, Lavine JE, Tonascia J, Unalp A, Van Natta M, Clark J, Brunt EM, Kleiner DE, Hoofnagle JH, Robuck PR, Nash CRN. Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis. N Engl J Med. 2010;362:1675–85.PubMedPubMedCentralCrossRef
158.
Zurück zum Zitat Belfort R, Harrison SA, Brown K, Darland C, Finch J, Hardies J, Balas B, Gastaldelli A, Tio F, Pulcini J, Berria R, Ma JZ, Dwivedi S, Havranek R, Fincke C, DeFronzo R, Bannayan GA, Schenker S, Cusi K. A placebo-controlled trial of pioglitazone in subjects with nonalcoholic steatohepatitis. N Engl J Med. 2006;355:2297–307.PubMedCrossRef Belfort R, Harrison SA, Brown K, Darland C, Finch J, Hardies J, Balas B, Gastaldelli A, Tio F, Pulcini J, Berria R, Ma JZ, Dwivedi S, Havranek R, Fincke C, DeFronzo R, Bannayan GA, Schenker S, Cusi K. A placebo-controlled trial of pioglitazone in subjects with nonalcoholic steatohepatitis. N Engl J Med. 2006;355:2297–307.PubMedCrossRef
159.
Zurück zum Zitat Promrat K, Lutchman G, Uwaifo GI, Freedman RJ, Soza A, Heller T, Doo E, Ghany M, Premkumar A, Park Y, Liang TJ, Yanovski JA, Kleiner DE, Hoofnagle JH. A pilot study of pioglitazone treatment for nonalcoholic steatohepatitis. Hepatology. 2004;39:188–96.PubMedCrossRef Promrat K, Lutchman G, Uwaifo GI, Freedman RJ, Soza A, Heller T, Doo E, Ghany M, Premkumar A, Park Y, Liang TJ, Yanovski JA, Kleiner DE, Hoofnagle JH. A pilot study of pioglitazone treatment for nonalcoholic steatohepatitis. Hepatology. 2004;39:188–96.PubMedCrossRef
160.
Zurück zum Zitat Aithal GP, Thomas JA, Kaye PV, Lawson A, Ryder SD, Spendlove I, Austin AS, Freeman JG, Morgan L, Webber J. Randomized, placebo-controlled trial of pioglitazone in nondiabetic subjects with nonalcoholic steatohepatitis. Gastroenterology. 2008;135:1176–84.PubMedCrossRef Aithal GP, Thomas JA, Kaye PV, Lawson A, Ryder SD, Spendlove I, Austin AS, Freeman JG, Morgan L, Webber J. Randomized, placebo-controlled trial of pioglitazone in nondiabetic subjects with nonalcoholic steatohepatitis. Gastroenterology. 2008;135:1176–84.PubMedCrossRef
161.
Zurück zum Zitat Cusi K, Orsak B, Bril F, Lomonaco R, Hecht J, Ortiz-Lopez C, Tio F, Hardies J, Darland C, Musi N, Webb A, Portillo-Sanchez P. Long-term pioglitazone treatment for patients with nonalcoholic steatohepatitis and prediabetes or type 2 diabetes Mellitus: a Randomized Trial. Ann Intern Med. 2016;165:305–15.PubMedCrossRef Cusi K, Orsak B, Bril F, Lomonaco R, Hecht J, Ortiz-Lopez C, Tio F, Hardies J, Darland C, Musi N, Webb A, Portillo-Sanchez P. Long-term pioglitazone treatment for patients with nonalcoholic steatohepatitis and prediabetes or type 2 diabetes Mellitus: a Randomized Trial. Ann Intern Med. 2016;165:305–15.PubMedCrossRef
162.
Zurück zum Zitat Neuschwander-Tetri BA, Brunt EM, Wehmeier KR, Oliver D, Bacon BR. Improved nonalcoholic steatohepatitis after 48 weeks of treatment with the PPAR-gamma ligand rosiglitazone. Hepatology. 2003;38:1008–17.PubMedCrossRef Neuschwander-Tetri BA, Brunt EM, Wehmeier KR, Oliver D, Bacon BR. Improved nonalcoholic steatohepatitis after 48 weeks of treatment with the PPAR-gamma ligand rosiglitazone. Hepatology. 2003;38:1008–17.PubMedCrossRef
163.
Zurück zum Zitat Ratziu V, Charlotte F, Bernhardt C, Giral P, Halbron M, Lenaour G, Hartmann-Heurtier A, Bruckert E, Poynard T, Group LS. Long-term efficacy of rosiglitazone in nonalcoholic steatohepatitis: results of the fatty liver improvement by rosiglitazone therapy (FLIRT 2) extension trial. Hepatology. 2010;51:445–53.PubMedCrossRef Ratziu V, Charlotte F, Bernhardt C, Giral P, Halbron M, Lenaour G, Hartmann-Heurtier A, Bruckert E, Poynard T, Group LS. Long-term efficacy of rosiglitazone in nonalcoholic steatohepatitis: results of the fatty liver improvement by rosiglitazone therapy (FLIRT 2) extension trial. Hepatology. 2010;51:445–53.PubMedCrossRef
164.
Zurück zum Zitat Ratziu V, Giral P, Jacqueminet S, Charlotte F, Hartemann-Heurtier A, Serfaty L, Podevin P, Lacorte JM, Bernhardt C, Bruckert E, Grimaldi A, Poynard T, Goroup LS. Rosiglitazone for nonalcoholic steatohepatitis: one-year results of the randomized placebo-controlled fatty liver improvement with Rosiglitazone Therapy (FLIRT) Trial. Gastroenterology; 2008. 135: 100–10.PubMedCrossRef Ratziu V, Giral P, Jacqueminet S, Charlotte F, Hartemann-Heurtier A, Serfaty L, Podevin P, Lacorte JM, Bernhardt C, Bruckert E, Grimaldi A, Poynard T, Goroup LS. Rosiglitazone for nonalcoholic steatohepatitis: one-year results of the randomized placebo-controlled fatty liver improvement with Rosiglitazone Therapy (FLIRT) Trial. Gastroenterology; 2008. 135: 100–10.PubMedCrossRef
165.
Zurück zum Zitat McCommis KS, Chen Z, Fu X, McDonald WG, Colca JR, Kletzien RF, Burgess SC, Finck BN. Loss of mitochondrial pyruvate carrier 2 in the liver leads to defects in gluconeogenesis and compensation via pyruvate-alanine Cycling. Cell Metab. 2015;22:682–94.PubMedPubMedCentralCrossRef McCommis KS, Chen Z, Fu X, McDonald WG, Colca JR, Kletzien RF, Burgess SC, Finck BN. Loss of mitochondrial pyruvate carrier 2 in the liver leads to defects in gluconeogenesis and compensation via pyruvate-alanine Cycling. Cell Metab. 2015;22:682–94.PubMedPubMedCentralCrossRef
166.
Zurück zum Zitat Shannon CE, Daniele G, Galindo C, Abdul-Ghani MA, DeFronzo RA, Norton L. Pioglitazone inhibits mitochondrial pyruvate metabolism and glucose production in hepatocytes. FEBS J. 2017;284:451–65.PubMedPubMedCentralCrossRef Shannon CE, Daniele G, Galindo C, Abdul-Ghani MA, DeFronzo RA, Norton L. Pioglitazone inhibits mitochondrial pyruvate metabolism and glucose production in hepatocytes. FEBS J. 2017;284:451–65.PubMedPubMedCentralCrossRef
167.
Zurück zum Zitat Colca JR, McDonald WG, Cavey GS, Cole SL, Holewa DD, Brightwell-Conrad AS, Wolfe CL, Wheeler JS, Coulter KR, Kilkuskie PM, Gracheva E, Korshunova Y, Trusgnich M, Karr R, Wiley SE, Divakaruni AS, Murphy AN, Vigueira PA, Finck BN, Kletzien RF. Identification of a mitochondrial target of thiazolidinedione insulin sensitizers (mTOT)--relationship to newly identified mitochondrial pyruvate carrier proteins. PLoS ONE. 2013;8:e61551.PubMedPubMedCentralCrossRef Colca JR, McDonald WG, Cavey GS, Cole SL, Holewa DD, Brightwell-Conrad AS, Wolfe CL, Wheeler JS, Coulter KR, Kilkuskie PM, Gracheva E, Korshunova Y, Trusgnich M, Karr R, Wiley SE, Divakaruni AS, Murphy AN, Vigueira PA, Finck BN, Kletzien RF. Identification of a mitochondrial target of thiazolidinedione insulin sensitizers (mTOT)--relationship to newly identified mitochondrial pyruvate carrier proteins. PLoS ONE. 2013;8:e61551.PubMedPubMedCentralCrossRef
168.
Zurück zum Zitat Harrison SA, Alkhouri N, Davison BA, Sanyal A, Edwards C, Colca JR, Lee BH, Loomba R, Cusi K, Kolterman O, Cotter G, Dittrich HC. Insulin sensitizer MSDC-0602K in non-alcoholic steatohepatitis: a randomized, double-blind, placebo-controlled phase IIb study. J Hepatol. 2020;72:613–26.PubMedCrossRef Harrison SA, Alkhouri N, Davison BA, Sanyal A, Edwards C, Colca JR, Lee BH, Loomba R, Cusi K, Kolterman O, Cotter G, Dittrich HC. Insulin sensitizer MSDC-0602K in non-alcoholic steatohepatitis: a randomized, double-blind, placebo-controlled phase IIb study. J Hepatol. 2020;72:613–26.PubMedCrossRef
169.
Zurück zum Zitat Wu PB, Song Q, Yu YJ, Yu HG, Luo HS, Tan SY. Effect of metformin on mitochondrial pathway of apoptosis and oxidative stress in cell model of nonalcoholic fatty liver disease. Zhonghua Gan Zang Bing Za Zhi. 2020;28:64–8.PubMed Wu PB, Song Q, Yu YJ, Yu HG, Luo HS, Tan SY. Effect of metformin on mitochondrial pathway of apoptosis and oxidative stress in cell model of nonalcoholic fatty liver disease. Zhonghua Gan Zang Bing Za Zhi. 2020;28:64–8.PubMed
170.
Zurück zum Zitat He L, Sabet A, Djedjos S, Miller R, Sun X, Hussain MA, Radovick S, Wondisford FE. Metformin and insulin suppress hepatic gluconeogenesis through phosphorylation of CREB binding protein. Cell. 2009;137:635–46.PubMedPubMedCentralCrossRef He L, Sabet A, Djedjos S, Miller R, Sun X, Hussain MA, Radovick S, Wondisford FE. Metformin and insulin suppress hepatic gluconeogenesis through phosphorylation of CREB binding protein. Cell. 2009;137:635–46.PubMedPubMedCentralCrossRef
171.
Zurück zum Zitat Yu X, Hao M, Liu Y, Ma X, Lin W, Xu Q, Zhou H, Shao N, Kuang H. Liraglutide ameliorates non-alcoholic steatohepatitis by inhibiting NLRP3 inflammasome and pyroptosis activation via mitophagy. Eur J Pharmacol. 2019;864: 172715.PubMedCrossRef Yu X, Hao M, Liu Y, Ma X, Lin W, Xu Q, Zhou H, Shao N, Kuang H. Liraglutide ameliorates non-alcoholic steatohepatitis by inhibiting NLRP3 inflammasome and pyroptosis activation via mitophagy. Eur J Pharmacol. 2019;864: 172715.PubMedCrossRef
172.
Zurück zum Zitat Tong W, Ju L, Qiu M, Xie Q, Chen Y, Shen W, Sun W, Wang W, Tian J. Liraglutide ameliorates non-alcoholic fatty liver disease by enhancing mitochondrial architecture and promoting autophagy through the SIRT1/SIRT3-FOXO3a pathway. Hepatol Res. 2016;46:933–43.PubMedCrossRef Tong W, Ju L, Qiu M, Xie Q, Chen Y, Shen W, Sun W, Wang W, Tian J. Liraglutide ameliorates non-alcoholic fatty liver disease by enhancing mitochondrial architecture and promoting autophagy through the SIRT1/SIRT3-FOXO3a pathway. Hepatol Res. 2016;46:933–43.PubMedCrossRef
173.
Zurück zum Zitat Zhang J, Xiang H, Liu J, Chen Y, He RR, Liu B. Mitochondrial sirtuin 3: new emerging biological function and therapeutic target. Theranostics. 2020;10:8315–42.PubMedPubMedCentralCrossRef Zhang J, Xiang H, Liu J, Chen Y, He RR, Liu B. Mitochondrial sirtuin 3: new emerging biological function and therapeutic target. Theranostics. 2020;10:8315–42.PubMedPubMedCentralCrossRef
174.
Zurück zum Zitat Jin L, Galonek H, Israelian K, Choy W, Morrison M, Xia Y, Wang X, Xu Y, Yang Y, Smith JJ, Hoffmann E, Carney DP, Perni RB, Jirousek MR, Bemis JE, Milne JC, Sinclair DA, Westphal CH. Biochemical characterization, localization, and tissue distribution of the longer form of mouse SIRT3. Protein Sci. 2009;18:514–25.PubMedPubMedCentralCrossRef Jin L, Galonek H, Israelian K, Choy W, Morrison M, Xia Y, Wang X, Xu Y, Yang Y, Smith JJ, Hoffmann E, Carney DP, Perni RB, Jirousek MR, Bemis JE, Milne JC, Sinclair DA, Westphal CH. Biochemical characterization, localization, and tissue distribution of the longer form of mouse SIRT3. Protein Sci. 2009;18:514–25.PubMedPubMedCentralCrossRef
175.
Zurück zum Zitat Yang Y, Hubbard BP, Sinclair DA, Tong Q. Characterization of murine SIRT3 transcript variants and corresponding protein products. J Cell Biochem. 2010;111:1051–8.PubMedPubMedCentralCrossRef Yang Y, Hubbard BP, Sinclair DA, Tong Q. Characterization of murine SIRT3 transcript variants and corresponding protein products. J Cell Biochem. 2010;111:1051–8.PubMedPubMedCentralCrossRef
177.
Zurück zum Zitat Ahn BH, Kim HS, Song S, Lee IH, Liu J, Vassilopoulos A, Deng CX, Finkel T. A role for the mitochondrial deacetylase Sirt3 in regulating energy homeostasis. Proc Natl Acad Sci U S A. 2008;105:14447–52.PubMedPubMedCentralCrossRef Ahn BH, Kim HS, Song S, Lee IH, Liu J, Vassilopoulos A, Deng CX, Finkel T. A role for the mitochondrial deacetylase Sirt3 in regulating energy homeostasis. Proc Natl Acad Sci U S A. 2008;105:14447–52.PubMedPubMedCentralCrossRef
178.
Zurück zum Zitat Kong X, Wang R, Xue Y, Liu X, Zhang H, Chen Y, Fang F, Chang Y. Sirtuin 3, a new target of PGC-1alpha, plays an important role in the suppression of ROS and mitochondrial biogenesis. PLoS ONE. 2010;5:e11707.PubMedPubMedCentralCrossRef Kong X, Wang R, Xue Y, Liu X, Zhang H, Chen Y, Fang F, Chang Y. Sirtuin 3, a new target of PGC-1alpha, plays an important role in the suppression of ROS and mitochondrial biogenesis. PLoS ONE. 2010;5:e11707.PubMedPubMedCentralCrossRef
179.
Zurück zum Zitat Nassir F, Arndt JJ, Johnson SA, Ibdah JA. Regulation of mitochondrial trifunctional protein modulates nonalcoholic fatty liver disease in mice. J Lipid Res. 2018;59:967–73.PubMedPubMedCentralCrossRef Nassir F, Arndt JJ, Johnson SA, Ibdah JA. Regulation of mitochondrial trifunctional protein modulates nonalcoholic fatty liver disease in mice. J Lipid Res. 2018;59:967–73.PubMedPubMedCentralCrossRef
180.
Zurück zum Zitat Wu T, Liu YH, Fu YC, Liu XM, Zhou XH. Direct evidence of sirtuin downregulation in the liver of non-alcoholic fatty liver disease patients. Ann Clin Lab Sci. 2014;44:410–8.PubMed Wu T, Liu YH, Fu YC, Liu XM, Zhou XH. Direct evidence of sirtuin downregulation in the liver of non-alcoholic fatty liver disease patients. Ann Clin Lab Sci. 2014;44:410–8.PubMed
181.
Zurück zum Zitat Bao J, Lu Z, Joseph JJ, Carabenciov D, Dimond CC, Pang L, Samsel L, McCoy JP Jr, Leclerc J, Nguyen P, Gius D, Sack MN. Characterization of the murine SIRT3 mitochondrial localization sequence and comparison of mitochondrial enrichment and deacetylase activity of long and short SIRT3 isoforms. J Cell Biochem. 2010;110:238–47.PubMedPubMedCentral Bao J, Lu Z, Joseph JJ, Carabenciov D, Dimond CC, Pang L, Samsel L, McCoy JP Jr, Leclerc J, Nguyen P, Gius D, Sack MN. Characterization of the murine SIRT3 mitochondrial localization sequence and comparison of mitochondrial enrichment and deacetylase activity of long and short SIRT3 isoforms. J Cell Biochem. 2010;110:238–47.PubMedPubMedCentral
182.
Zurück zum Zitat Hirschey MD, Shimazu T, Jing E, Grueter CA, Collins AM, Aouizerat B, Stancakova A, Goetzman E, Lam MM, Schwer B, Stevens RD, Muehlbauer MJ, Kakar S, Bass NM, Kuusisto J, Laakso M, Alt FW, Newgard CB, Farese RV Jr, Kahn CR, Verdin E. SIRT3 deficiency and mitochondrial protein hyperacetylation accelerate the development of the metabolic syndrome. Mol Cell. 2011;44:177–90.PubMedPubMedCentralCrossRef Hirschey MD, Shimazu T, Jing E, Grueter CA, Collins AM, Aouizerat B, Stancakova A, Goetzman E, Lam MM, Schwer B, Stevens RD, Muehlbauer MJ, Kakar S, Bass NM, Kuusisto J, Laakso M, Alt FW, Newgard CB, Farese RV Jr, Kahn CR, Verdin E. SIRT3 deficiency and mitochondrial protein hyperacetylation accelerate the development of the metabolic syndrome. Mol Cell. 2011;44:177–90.PubMedPubMedCentralCrossRef
183.
Zurück zum Zitat Kendrick AA, Choudhury M, Rahman SM, McCurdy CE, Friederich M, Van Hove JL, Watson PA, Birdsey N, Bao J, Gius D, Sack MN, Jing E, Kahn CR, Friedman JE, Jonscher KR. Fatty liver is associated with reduced SIRT3 activity and mitochondrial protein hyperacetylation. Biochem J. 2011;433:505–14.PubMedCrossRef Kendrick AA, Choudhury M, Rahman SM, McCurdy CE, Friederich M, Van Hove JL, Watson PA, Birdsey N, Bao J, Gius D, Sack MN, Jing E, Kahn CR, Friedman JE, Jonscher KR. Fatty liver is associated with reduced SIRT3 activity and mitochondrial protein hyperacetylation. Biochem J. 2011;433:505–14.PubMedCrossRef
184.
Zurück zum Zitat Barroso E, Rodriguez-Rodriguez R, Zarei M, Pizarro-Degado J, Planavila A, Palomer X, Villarroya F, Vazquez-Carrera M. SIRT3 deficiency exacerbates fatty liver by attenuating the HIF1alpha-LIPIN 1 pathway and increasing CD36 through Nrf2. Cell Commun Signal. 2020;18:147.PubMedPubMedCentralCrossRef Barroso E, Rodriguez-Rodriguez R, Zarei M, Pizarro-Degado J, Planavila A, Palomer X, Villarroya F, Vazquez-Carrera M. SIRT3 deficiency exacerbates fatty liver by attenuating the HIF1alpha-LIPIN 1 pathway and increasing CD36 through Nrf2. Cell Commun Signal. 2020;18:147.PubMedPubMedCentralCrossRef
185.
Zurück zum Zitat Li R, Xin T, Li D, Wang C, Zhu H, Zhou H. Therapeutic effect of Sirtuin 3 on ameliorating nonalcoholic fatty liver disease: the role of the ERK-CREB pathway and Bnip3-mediated mitophagy. Redox Biol. 2018;18:229–43.PubMedPubMedCentralCrossRef Li R, Xin T, Li D, Wang C, Zhu H, Zhou H. Therapeutic effect of Sirtuin 3 on ameliorating nonalcoholic fatty liver disease: the role of the ERK-CREB pathway and Bnip3-mediated mitophagy. Redox Biol. 2018;18:229–43.PubMedPubMedCentralCrossRef
186.
Zurück zum Zitat Zhang T, Liu J, Shen S, Tong Q, Ma X, Lin L. SIRT3 promotes lipophagy and chaperon-mediated autophagy to protect hepatocytes against lipotoxicity. Cell Death Differ. 2020;27:329–44.PubMedCrossRef Zhang T, Liu J, Shen S, Tong Q, Ma X, Lin L. SIRT3 promotes lipophagy and chaperon-mediated autophagy to protect hepatocytes against lipotoxicity. Cell Death Differ. 2020;27:329–44.PubMedCrossRef
187.
Zurück zum Zitat Dai H, Sinclair DA, Ellis JL, Steegborn C. Sirtuin activators and inhibitors: promises, achievements, and challenges. Pharmacol Ther. 2018;188:140–54.PubMedPubMedCentralCrossRef Dai H, Sinclair DA, Ellis JL, Steegborn C. Sirtuin activators and inhibitors: promises, achievements, and challenges. Pharmacol Ther. 2018;188:140–54.PubMedPubMedCentralCrossRef
189.
Zurück zum Zitat Tarantino G, Finelli C, Scopacasa F, Pasanisi F, Contaldo F, Capone D, Savastano S. Circulating levels of sirtuin 4, a potential marker of oxidative metabolism, related to coronary artery disease in obese patients suffering from NAFLD, with normal or slightly increased liver enzymes. Oxid Med Cell Longev. 2014;2014:920676.PubMedPubMedCentralCrossRef Tarantino G, Finelli C, Scopacasa F, Pasanisi F, Contaldo F, Capone D, Savastano S. Circulating levels of sirtuin 4, a potential marker of oxidative metabolism, related to coronary artery disease in obese patients suffering from NAFLD, with normal or slightly increased liver enzymes. Oxid Med Cell Longev. 2014;2014:920676.PubMedPubMedCentralCrossRef
190.
Zurück zum Zitat Lazaridis KN, Gores GJ, Lindor KD. Ursodeoxycholic acid ‘mechanisms of action and clinical use in hepatobiliary disorders.’ J Hepatol. 2001;35:134–46.PubMedCrossRef Lazaridis KN, Gores GJ, Lindor KD. Ursodeoxycholic acid ‘mechanisms of action and clinical use in hepatobiliary disorders.’ J Hepatol. 2001;35:134–46.PubMedCrossRef
191.
Zurück zum Zitat Laurin J, Lindor KD, Crippin JS, Gossard A, Gores GJ, Ludwig J, Rakela J, McGill DB. Ursodeoxycholic acid or clofibrate in the treatment of non-alcohol-induced steatohepatitis: a pilot study. Hepatology. 1996;23:1464–7.PubMedCrossRef Laurin J, Lindor KD, Crippin JS, Gossard A, Gores GJ, Ludwig J, Rakela J, McGill DB. Ursodeoxycholic acid or clofibrate in the treatment of non-alcohol-induced steatohepatitis: a pilot study. Hepatology. 1996;23:1464–7.PubMedCrossRef
192.
Zurück zum Zitat Krahenbuhl S, Talos C, Fischer S, Reichen J. Toxicity of bile acids on the electron transport chain of isolated rat liver mitochondria. Hepatology. 1994;19:471–9.PubMed Krahenbuhl S, Talos C, Fischer S, Reichen J. Toxicity of bile acids on the electron transport chain of isolated rat liver mitochondria. Hepatology. 1994;19:471–9.PubMed
193.
Zurück zum Zitat Krahenbuhl S, Talos C, Lauterburg BH, Reichen J. Reduced antioxidative capacity in liver mitochondria from bile duct ligated rats. Hepatology. 1995;22:607–12.PubMed Krahenbuhl S, Talos C, Lauterburg BH, Reichen J. Reduced antioxidative capacity in liver mitochondria from bile duct ligated rats. Hepatology. 1995;22:607–12.PubMed
194.
Zurück zum Zitat Rolo AP, Oliveira PJ, Moreno AJ, Palmeira CM. Bile acids affect liver mitochondrial bioenergetics: possible relevance for cholestasis therapy. Toxicol Sci. 2000;57:177–85.PubMedCrossRef Rolo AP, Oliveira PJ, Moreno AJ, Palmeira CM. Bile acids affect liver mitochondrial bioenergetics: possible relevance for cholestasis therapy. Toxicol Sci. 2000;57:177–85.PubMedCrossRef
195.
Zurück zum Zitat Krahenbuhl S, Fischer S, Talos C, Reichen J. Ursodeoxycholate protects oxidative mitochondrial metabolism from bile acid toxicity: dose-response study in isolated rat liver mitochondria. Hepatology. 1994;20:1595–601.PubMedCrossRef Krahenbuhl S, Fischer S, Talos C, Reichen J. Ursodeoxycholate protects oxidative mitochondrial metabolism from bile acid toxicity: dose-response study in isolated rat liver mitochondria. Hepatology. 1994;20:1595–601.PubMedCrossRef
196.
Zurück zum Zitat Iruarrizaga-Lejarreta M, Varela-Rey M, Fernandez-Ramos D, Martinez-Arranz I, Delgado TC, Simon J, Juan VG, delaCruz-Villar L, Azkargorta M, Lavin JL, Mayo R, Van Liempd SM, Aurrekoetxea I, Buque X, Cave DD, Pena A, Rodriguez-Cuesta J, Aransay AM, Elortza F, Falcon-Perez JM, Aspichueta P, Hayardeny L, Noureddin M, Sanyal AJ, Alonso C, Anguita J, Martinez-Chantar ML, Lu SC, Mato JM. Role of Aramchol in steatohepatitis and fibrosis in mice. Hepatol Commun. 2017;1:911–27.PubMedPubMedCentralCrossRef Iruarrizaga-Lejarreta M, Varela-Rey M, Fernandez-Ramos D, Martinez-Arranz I, Delgado TC, Simon J, Juan VG, delaCruz-Villar L, Azkargorta M, Lavin JL, Mayo R, Van Liempd SM, Aurrekoetxea I, Buque X, Cave DD, Pena A, Rodriguez-Cuesta J, Aransay AM, Elortza F, Falcon-Perez JM, Aspichueta P, Hayardeny L, Noureddin M, Sanyal AJ, Alonso C, Anguita J, Martinez-Chantar ML, Lu SC, Mato JM. Role of Aramchol in steatohepatitis and fibrosis in mice. Hepatol Commun. 2017;1:911–27.PubMedPubMedCentralCrossRef
197.
Zurück zum Zitat Safadi R, Konikoff FM, Mahamid M, Zelber-Sagi S, Halpern M, Gilat T, Oren R, Group F. The fatty acid-bile acid conjugate Aramchol reduces liver fat content in patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol. 2014;12:2085-2091 e2081.PubMedCrossRef Safadi R, Konikoff FM, Mahamid M, Zelber-Sagi S, Halpern M, Gilat T, Oren R, Group F. The fatty acid-bile acid conjugate Aramchol reduces liver fat content in patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol. 2014;12:2085-2091 e2081.PubMedCrossRef
198.
Zurück zum Zitat Asin-Cayuela J, Manas AR, James AM, Smith RA, Murphy MP. Fine-tuning the hydrophobicity of a mitochondria-targeted antioxidant. FEBS Lett. 2004;571:9–16.PubMedCrossRef Asin-Cayuela J, Manas AR, James AM, Smith RA, Murphy MP. Fine-tuning the hydrophobicity of a mitochondria-targeted antioxidant. FEBS Lett. 2004;571:9–16.PubMedCrossRef
199.
Zurück zum Zitat Rokitskaya TI, Klishin SS, Severina II, Skulachev VP, Antonenko YN. Kinetic analysis of permeation of mitochondria-targeted antioxidants across bilayer lipid membranes. J Membr Biol. 2008;224:9–19.PubMedCrossRef Rokitskaya TI, Klishin SS, Severina II, Skulachev VP, Antonenko YN. Kinetic analysis of permeation of mitochondria-targeted antioxidants across bilayer lipid membranes. J Membr Biol. 2008;224:9–19.PubMedCrossRef
200.
Zurück zum Zitat Smith RA, Porteous CM, Gane AM, Murphy MP. Delivery of bioactive molecules to mitochondria in vivo. Proc Natl Acad Sci U S A. 2003;100:5407–12.PubMedPubMedCentralCrossRef Smith RA, Porteous CM, Gane AM, Murphy MP. Delivery of bioactive molecules to mitochondria in vivo. Proc Natl Acad Sci U S A. 2003;100:5407–12.PubMedPubMedCentralCrossRef
201.
Zurück zum Zitat Feillet-Coudray C, Fouret G, Ebabe Elle R, Rieusset J, Bonafos B, Chabi B, Crouzier D, Zarkovic K, Zarkovic N, Ramos J, Badia E, Murphy MP, Cristol JP, Coudray C. The mitochondrial-targeted antioxidant MitoQ ameliorates metabolic syndrome features in obesogenic diet-fed rats better than apocynin or Allopurinol. Free Radic Res. 2014;48:1232–46.PubMedCrossRef Feillet-Coudray C, Fouret G, Ebabe Elle R, Rieusset J, Bonafos B, Chabi B, Crouzier D, Zarkovic K, Zarkovic N, Ramos J, Badia E, Murphy MP, Cristol JP, Coudray C. The mitochondrial-targeted antioxidant MitoQ ameliorates metabolic syndrome features in obesogenic diet-fed rats better than apocynin or Allopurinol. Free Radic Res. 2014;48:1232–46.PubMedCrossRef
202.
Zurück zum Zitat Fouret G, Tolika E, Lecomte J, Bonafos B, Aoun M, Murphy MP, Ferreri C, Chatgilialoglu C, Dubreucq E, Coudray C, Feillet-Coudray C. The mitochondrial-targeted antioxidant, MitoQ, increases liver mitochondrial cardiolipin content in obesogenic diet-fed rats. Biochim Biophys Acta. 2015;1847:1025–35.PubMedCrossRef Fouret G, Tolika E, Lecomte J, Bonafos B, Aoun M, Murphy MP, Ferreri C, Chatgilialoglu C, Dubreucq E, Coudray C, Feillet-Coudray C. The mitochondrial-targeted antioxidant, MitoQ, increases liver mitochondrial cardiolipin content in obesogenic diet-fed rats. Biochim Biophys Acta. 2015;1847:1025–35.PubMedCrossRef
203.
Zurück zum Zitat Mercer JR, Yu E, Figg N, Cheng KK, Prime TA, Griffin JL, Masoodi M, Vidal-Puig A, Murphy MP, Bennett MR. The mitochondria-targeted antioxidant MitoQ decreases features of the metabolic syndrome in ATM+/-/ApoE-/- mice. Free Radic Biol Med. 2012;52:841–9.PubMedCrossRef Mercer JR, Yu E, Figg N, Cheng KK, Prime TA, Griffin JL, Masoodi M, Vidal-Puig A, Murphy MP, Bennett MR. The mitochondria-targeted antioxidant MitoQ decreases features of the metabolic syndrome in ATM+/-/ApoE-/- mice. Free Radic Biol Med. 2012;52:841–9.PubMedCrossRef
204.
Zurück zum Zitat Xiao L, Xu X, Zhang F, Wang M, Xu Y, Tang D, Wang J, Qin Y, Liu Y, Tang C, He L, Greka A, Zhou Z, Liu F, Dong Z, Sun L. The mitochondria-targeted antioxidant MitoQ ameliorated tubular injury mediated by mitophagy in diabetic kidney disease via Nrf2/PINK1. Redox Biol. 2017;11:297–311.PubMedCrossRef Xiao L, Xu X, Zhang F, Wang M, Xu Y, Tang D, Wang J, Qin Y, Liu Y, Tang C, He L, Greka A, Zhou Z, Liu F, Dong Z, Sun L. The mitochondria-targeted antioxidant MitoQ ameliorated tubular injury mediated by mitophagy in diabetic kidney disease via Nrf2/PINK1. Redox Biol. 2017;11:297–311.PubMedCrossRef
205.
Zurück zum Zitat Babizhayev MA, Yegorov YE. Reactive oxygen species and the aging eye: specific role of metabolically active mitochondria in maintaining lens function and in the initiation of the oxidation-induced maturity onset cataract–a novel platform of mitochondria-targeted antioxidants with broad therapeutic potential for redox regulation and detoxification of oxidants in eye diseases. Am J Ther. 2016;23:e98-117.PubMedCrossRef Babizhayev MA, Yegorov YE. Reactive oxygen species and the aging eye: specific role of metabolically active mitochondria in maintaining lens function and in the initiation of the oxidation-induced maturity onset cataract–a novel platform of mitochondria-targeted antioxidants with broad therapeutic potential for redox regulation and detoxification of oxidants in eye diseases. Am J Ther. 2016;23:e98-117.PubMedCrossRef
206.
Zurück zum Zitat Dhanasekaran A, Kotamraju S, Kalivendi SV, Matsunaga T, Shang T, Keszler A, Joseph J, Kalyanaraman B. Supplementation of endothelial cells with mitochondria-targeted antioxidants inhibit peroxide-induced mitochondrial iron uptake, oxidative damage, and apoptosis. J Biol Chem. 2004;279:37575–87.PubMedCrossRef Dhanasekaran A, Kotamraju S, Kalivendi SV, Matsunaga T, Shang T, Keszler A, Joseph J, Kalyanaraman B. Supplementation of endothelial cells with mitochondria-targeted antioxidants inhibit peroxide-induced mitochondrial iron uptake, oxidative damage, and apoptosis. J Biol Chem. 2004;279:37575–87.PubMedCrossRef
207.
Zurück zum Zitat Gane EJ, Weilert F, Orr DW, Keogh GF, Gibson M, Lockhart MM, Frampton CM, Taylor KM, Smith RA, Murphy MP. The mitochondria-targeted anti-oxidant mitoquinone decreases liver damage in a phase II study of hepatitis C patients. Liver Int. 2010;30:1019–26.PubMedCrossRef Gane EJ, Weilert F, Orr DW, Keogh GF, Gibson M, Lockhart MM, Frampton CM, Taylor KM, Smith RA, Murphy MP. The mitochondria-targeted anti-oxidant mitoquinone decreases liver damage in a phase II study of hepatitis C patients. Liver Int. 2010;30:1019–26.PubMedCrossRef
208.
Zurück zum Zitat Saller R, Meier R, Brignoli R. The use of silymarin in the treatment of liver diseases. Drugs. 2001;61:2035–63.PubMedCrossRef Saller R, Meier R, Brignoli R. The use of silymarin in the treatment of liver diseases. Drugs. 2001;61:2035–63.PubMedCrossRef
209.
Zurück zum Zitat Trappoliere M, Caligiuri A, Schmid M, Bertolani C, Failli P, Vizzutti F, Novo E, di Manzano C, Marra F, Loguercio C, Pinzani M. Silybin, a component of sylimarin, exerts anti-inflammatory and anti-fibrogenic effects on human hepatic stellate cells. J Hepatol. 2009;50:1102–11.PubMedCrossRef Trappoliere M, Caligiuri A, Schmid M, Bertolani C, Failli P, Vizzutti F, Novo E, di Manzano C, Marra F, Loguercio C, Pinzani M. Silybin, a component of sylimarin, exerts anti-inflammatory and anti-fibrogenic effects on human hepatic stellate cells. J Hepatol. 2009;50:1102–11.PubMedCrossRef
210.
Zurück zum Zitat Solhi H, Ghahremani R, Kazemifar AM, Yazdi HZ. Silymarin in treatment of non-alcoholic steatohepatitis: a randomized clinical trial. Casp J Intern Med. 2014;5:9–12. Solhi H, Ghahremani R, Kazemifar AM, Yazdi HZ. Silymarin in treatment of non-alcoholic steatohepatitis: a randomized clinical trial. Casp J Intern Med. 2014;5:9–12.
211.
Zurück zum Zitat Loguercio C, Andreone P, Brisc C, Brisc MC, Bugianesi E, Chiaramonte M, Cursaro C, Danila M, de Sio I, Floreani A, Freni MA, Grieco A, Groppo M, Lazzari R, Lobello S, Lorefice E, Margotti M, Miele L, Milani S, Okolicsanyi L, Palasciano G, Portincasa P, Saltarelli P, Smedile A, Somalvico F, Spadaro A, Sporea I, Sorrentino P, Vecchione R, Tuccillo C, Del Vecchio Blanco C, Federico A. Silybin combined with phosphatidylcholine and vitamin E in patients with nonalcoholic fatty liver disease: a randomized controlled trial. Free Radic Biol Med. 2012;52:1658–65.PubMedCrossRef Loguercio C, Andreone P, Brisc C, Brisc MC, Bugianesi E, Chiaramonte M, Cursaro C, Danila M, de Sio I, Floreani A, Freni MA, Grieco A, Groppo M, Lazzari R, Lobello S, Lorefice E, Margotti M, Miele L, Milani S, Okolicsanyi L, Palasciano G, Portincasa P, Saltarelli P, Smedile A, Somalvico F, Spadaro A, Sporea I, Sorrentino P, Vecchione R, Tuccillo C, Del Vecchio Blanco C, Federico A. Silybin combined with phosphatidylcholine and vitamin E in patients with nonalcoholic fatty liver disease: a randomized controlled trial. Free Radic Biol Med. 2012;52:1658–65.PubMedCrossRef
212.
Zurück zum Zitat Vecchione G, Grasselli E, Voci A, Baldini F, Grattagliano I, Wang DQ, Portincasa P, Vergani L. Silybin counteracts lipid excess and oxidative stress in cultured steatotic hepatic cells. World J Gastroenterol. 2016;22:6016–26.PubMedPubMedCentralCrossRef Vecchione G, Grasselli E, Voci A, Baldini F, Grattagliano I, Wang DQ, Portincasa P, Vergani L. Silybin counteracts lipid excess and oxidative stress in cultured steatotic hepatic cells. World J Gastroenterol. 2016;22:6016–26.PubMedPubMedCentralCrossRef
213.
Zurück zum Zitat Colman E. Dinitrophenol and obesity: an early twentieth-century regulatory dilemma. Regul Toxicol Pharmacol. 2007;48:115–7.PubMedCrossRef Colman E. Dinitrophenol and obesity: an early twentieth-century regulatory dilemma. Regul Toxicol Pharmacol. 2007;48:115–7.PubMedCrossRef
214.
Zurück zum Zitat Zhang R, Chu K, Zhao N, Wu J, Ma L, Zhu C, Chen X, Wei G, Liao M. Corilagin alleviates nonalcoholic fatty liver disease in high-fat diet-induced C57BL/6 mice by ameliorating oxidative stress and restoring autophagic flux. Front Pharmacol. 2019;10:1693.PubMedCrossRef Zhang R, Chu K, Zhao N, Wu J, Ma L, Zhu C, Chen X, Wei G, Liao M. Corilagin alleviates nonalcoholic fatty liver disease in high-fat diet-induced C57BL/6 mice by ameliorating oxidative stress and restoring autophagic flux. Front Pharmacol. 2019;10:1693.PubMedCrossRef
215.
Zurück zum Zitat Perry RJ, Zhang D, Zhang XM, Boyer JL, Shulman GI. Controlled-release mitochondrial protonophore reverses diabetes and steatohepatitis in rats. Science. 2015;347:1253–6.PubMedPubMedCentralCrossRef Perry RJ, Zhang D, Zhang XM, Boyer JL, Shulman GI. Controlled-release mitochondrial protonophore reverses diabetes and steatohepatitis in rats. Science. 2015;347:1253–6.PubMedPubMedCentralCrossRef
216.
Zurück zum Zitat Fu A, Shi X, Zhang H, Fu B. Mitotherapy for fatty liver by intravenous administration of exogenous mitochondria in male mice. Front Pharmacol. 2017;8: 241.PubMedPubMedCentralCrossRef Fu A, Shi X, Zhang H, Fu B. Mitotherapy for fatty liver by intravenous administration of exogenous mitochondria in male mice. Front Pharmacol. 2017;8: 241.PubMedPubMedCentralCrossRef
217.
Zurück zum Zitat Ajith TA. Role of mitochondria and mitochondria-targeted agents in non-alcoholic fatty liver disease. Clin Exp Pharmacol Physiol. 2018;45:413–21.PubMedCrossRef Ajith TA. Role of mitochondria and mitochondria-targeted agents in non-alcoholic fatty liver disease. Clin Exp Pharmacol Physiol. 2018;45:413–21.PubMedCrossRef
218.
220.
Zurück zum Zitat Ajaz S, McPhail MJ, Gnudi L, Trovato FM, Mujib S, Napoli S, Carey I, Agarwal K. Mitochondrial dysfunction as a mechanistic biomarker in patients with non-alcoholic fatty liver disease (NAFLD). Mitochondrion. 2021;57:119–30.PubMedCrossRef Ajaz S, McPhail MJ, Gnudi L, Trovato FM, Mujib S, Napoli S, Carey I, Agarwal K. Mitochondrial dysfunction as a mechanistic biomarker in patients with non-alcoholic fatty liver disease (NAFLD). Mitochondrion. 2021;57:119–30.PubMedCrossRef
221.
Zurück zum Zitat Piccinin E, Villani G, Moschetta A. Metabolic aspects in NAFLD, NASH and hepatocellular carcinoma: the role of PGC1 coactivators. Nat Rev Gastroenterol Hepatol. 2019;16:160–74.PubMedCrossRef Piccinin E, Villani G, Moschetta A. Metabolic aspects in NAFLD, NASH and hepatocellular carcinoma: the role of PGC1 coactivators. Nat Rev Gastroenterol Hepatol. 2019;16:160–74.PubMedCrossRef
222.
Zurück zum Zitat Parafati M, Lascala A, Morittu VM, Trimboli F, Rizzuto A, Brunelli E, Coscarelli F, Costa N, Britti D, Ehrlich J, Isidoro C, Mollace V, Janda E. Bergamot polyphenol fraction prevents nonalcoholic fatty liver disease via stimulation of lipophagy in cafeteria diet-induced rat model of metabolic syndrome. J Nutr Biochem. 2015;26:938–48.PubMedCrossRef Parafati M, Lascala A, Morittu VM, Trimboli F, Rizzuto A, Brunelli E, Coscarelli F, Costa N, Britti D, Ehrlich J, Isidoro C, Mollace V, Janda E. Bergamot polyphenol fraction prevents nonalcoholic fatty liver disease via stimulation of lipophagy in cafeteria diet-induced rat model of metabolic syndrome. J Nutr Biochem. 2015;26:938–48.PubMedCrossRef
223.
Zurück zum Zitat Mehta K, Van Thiel DH, Shah N, Mobarhan S. Nonalcoholic fatty liver disease: pathogenesis and the role of antioxidants. Nutr Rev. 2002;60:289–93.PubMedCrossRef Mehta K, Van Thiel DH, Shah N, Mobarhan S. Nonalcoholic fatty liver disease: pathogenesis and the role of antioxidants. Nutr Rev. 2002;60:289–93.PubMedCrossRef
224.
Zurück zum Zitat Lavine JE, Schwimmer JB, Van Natta ML, Molleston JP, Murray KF, Rosenthal P, Abrams SH, Scheimann AO, Sanyal AJ, Chalasani N, Tonascia J, Unalp A, Clark JM, Brunt EM, Kleiner DE, Hoofnagle JH, Robuck PR. Nonalcoholic steatohepatitis clinical research, effect of vitamin E or metformin for treatment of nonalcoholic fatty liver disease in children and adolescents: the TONIC randomized controlled trial. JAMA. 2011;305:1659–68.PubMedPubMedCentralCrossRef Lavine JE, Schwimmer JB, Van Natta ML, Molleston JP, Murray KF, Rosenthal P, Abrams SH, Scheimann AO, Sanyal AJ, Chalasani N, Tonascia J, Unalp A, Clark JM, Brunt EM, Kleiner DE, Hoofnagle JH, Robuck PR. Nonalcoholic steatohepatitis clinical research, effect of vitamin E or metformin for treatment of nonalcoholic fatty liver disease in children and adolescents: the TONIC randomized controlled trial. JAMA. 2011;305:1659–68.PubMedPubMedCentralCrossRef
225.
Zurück zum Zitat Newsome PN, Buchholtz K, Cusi K, Linder M, Okanoue T, Ratziu V, Sanyal AJ, Sejling AS, Harrison SA, Investigators NN. A placebo-controlled trial of subcutaneous semaglutide in nonalcoholic steatohepatitis. N Engl J Med. 2021;384:1113–24.PubMedCrossRef Newsome PN, Buchholtz K, Cusi K, Linder M, Okanoue T, Ratziu V, Sanyal AJ, Sejling AS, Harrison SA, Investigators NN. A placebo-controlled trial of subcutaneous semaglutide in nonalcoholic steatohepatitis. N Engl J Med. 2021;384:1113–24.PubMedCrossRef
226.
Zurück zum Zitat Armstrong MJ, Hull D, Guo K, Barton D, Hazlehurst JM, Gathercole LL, Nasiri M, Yu J, Gough SC, Newsome PN, Tomlinson JW. Glucagon-like peptide 1 decreases lipotoxicity in non-alcoholic steatohepatitis. J Hepatol. 2016;64:399–408.PubMedPubMedCentralCrossRef Armstrong MJ, Hull D, Guo K, Barton D, Hazlehurst JM, Gathercole LL, Nasiri M, Yu J, Gough SC, Newsome PN, Tomlinson JW. Glucagon-like peptide 1 decreases lipotoxicity in non-alcoholic steatohepatitis. J Hepatol. 2016;64:399–408.PubMedPubMedCentralCrossRef
227.
Zurück zum Zitat Mueller M, Thorell A, Claudel T, Jha P, Koefeler H, Lackner C, Hoesel B, Fauler G, Stojakovic T, Einarsson C, Marschall HU, Trauner M. Ursodeoxycholic acid exerts farnesoid X receptor-antagonistic effects on bile acid and lipid metabolism in morbid obesity. J Hepatol. 2015;62:1398–404.PubMedPubMedCentralCrossRef Mueller M, Thorell A, Claudel T, Jha P, Koefeler H, Lackner C, Hoesel B, Fauler G, Stojakovic T, Einarsson C, Marschall HU, Trauner M. Ursodeoxycholic acid exerts farnesoid X receptor-antagonistic effects on bile acid and lipid metabolism in morbid obesity. J Hepatol. 2015;62:1398–404.PubMedPubMedCentralCrossRef
228.
Zurück zum Zitat Wah Kheong C, Nik Mustapha NR, Mahadeva S. A randomized trial of silymarin for the treatment of nonalcoholic steatohepatitis. Clin Gastroenterol Hepatol. 2017;15:1940–9 (e1948).PubMedCrossRef Wah Kheong C, Nik Mustapha NR, Mahadeva S. A randomized trial of silymarin for the treatment of nonalcoholic steatohepatitis. Clin Gastroenterol Hepatol. 2017;15:1940–9 (e1948).PubMedCrossRef
229.
Zurück zum Zitat Goedeke L, Murt KN, Di Francesco A, Camporez JP, Nasiri AR, Wang Y, Zhang XM, Cline GW, de Cabo R, Shulman GI. Sex- and strain-specific effects of mitochondrial uncoupling on age-related metabolic diseases in high-fat diet-fed mice. Aging Cell. 2022;21:e13539.PubMedPubMedCentralCrossRef Goedeke L, Murt KN, Di Francesco A, Camporez JP, Nasiri AR, Wang Y, Zhang XM, Cline GW, de Cabo R, Shulman GI. Sex- and strain-specific effects of mitochondrial uncoupling on age-related metabolic diseases in high-fat diet-fed mice. Aging Cell. 2022;21:e13539.PubMedPubMedCentralCrossRef
230.
Zurück zum Zitat Abulizi A, Vatner DF, Ye Z, Wang Y, Camporez JP, Zhang D, Kahn M, Lyu K, Sirwi A, Cline GW, Hussain MM, Aspichueta P, Samuel VT, Shulman GI. Membrane-bound sn-1,2-diacylglycerols explain the dissociation of hepatic insulin resistance from hepatic steatosis in MTTP knockout mice. J Lipid Res. 2020;61:1565–76.PubMedPubMedCentralCrossRef Abulizi A, Vatner DF, Ye Z, Wang Y, Camporez JP, Zhang D, Kahn M, Lyu K, Sirwi A, Cline GW, Hussain MM, Aspichueta P, Samuel VT, Shulman GI. Membrane-bound sn-1,2-diacylglycerols explain the dissociation of hepatic insulin resistance from hepatic steatosis in MTTP knockout mice. J Lipid Res. 2020;61:1565–76.PubMedPubMedCentralCrossRef
Metadaten
Titel
Mitochondrial metabolic dysfunction and non-alcoholic fatty liver disease: new insights from pathogenic mechanisms to clinically targeted therapy
verfasst von
Youwei Zheng
Shiting Wang
Jialiang Wu
Yong Wang
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2023
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-023-04367-1

Weitere Artikel der Ausgabe 1/2023

Journal of Translational Medicine 1/2023 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.