• Open Access
    Review

    Impact of mitochondrial lipid alterations on liver disease mechanisms and progression

    Laura Fàbrega 1,2,3
    José C. Fernández-Checa 1,2,3,4
    Laura Conde de la Rosa 1,2,3*
    Carmen Garcia-Ruiz 1,2,3,4*

    Explor Dig Dis. 2024;3:382–413 DOI: https://doi.org/10.37349/edd.2024.00057

    Received: April 06, 2024 Accepted: July 01, 2024 Published: September 10, 2024

    Academic Editor: Marco Falasca, Curtin University, Australia

    This article belongs to the special issue Mitochondria and Lipid Signalling in Liver Diseases

    Abstract

    Lipids are intricate biomolecules responsible for the building up of biological membranes. Besides this structural function, they also display crucial roles in signaling, acting as second messengers that activate specific pathways. Mitochondria are fundamental for cells as they participate in several pivotal functions, such as ATP synthesis, cell survival, metabolic pathways, and calcium homeostasis. Thus, the lipid composition of mitochondrial membranes can affect specific proteins and impact vital functions of mitochondria, such as oxidative phosphorylation and dynamics. The liver possesses a critical function in lipid homeostasis, involving the generation, oxidation, and trafficking of free fatty acids (FFA), triglycerides (TG), cholesterol, and bile acids (BAs). Mitochondria play a key role in lipid storage regulation in hepatocytes, which can control liver function. Their diverse tasks are affected by the lipid composition of mitochondrial membranes, characterized by low cholesterol content and enrichment of specific lipids such as cardiolipin. As mitochondria determine the bioenergetic status of cells and are key regulators of cell viability, alterations of mitochondrial lipid composition can contribute to the induction and progression of chronic diseases, including alcohol-related liver disease (ARLD) and metabolic dysfunction-associated steatotic liver disease (MASLD), two of the most common forms of liver diseases characterized by steatosis, necroinflammation, and fibrosis, which can progress to hepatocellular carcinoma (HCC). Thus, the disruption of lipid metabolism and membrane composition of mitochondria are characteristic features of cancer cells, and altered mitochondrial lipid composition may be a critical player in the progression of chronic liver diseases toward HCC. This review will address the mechanisms whereby alterations of mitochondrial lipid composition lead to the onset and progression of chronic liver diseases. Thus, a better characterization of the alterations of lipid composition in mitochondria may be a crucial step to design strategies and novel therapeutic opportunities for the treatment of MASLD and ARLD.

    Keywords

    Mitochondria, cholesterol, glutathione, ARLD, MASLD, MASH

    Introduction

    Mitochondria are intracellular double membrane-bound organelles that supply energy for the intracellular metabolism in eukaryotic cells. Mitochondria are crucial for the assembly of iron-sulfur clusters, calcium homeostasis, and metabolism of carbohydrates, lipids, and proteins. Mitochondria are also pivotal in cellular survival by regulating strategic pathways involved in the intrinsic or death receptor-mediated cell death. Their structure and membrane composition are unique (Figure 1). The existence of double encircling membranes, the outer (OMM) and the inner (IMM) membrane, is the manifestation of the endosymbiotic foundation of mitochondria about two million years ago [1, 2]. The presence of the OMM and IMM defines two different spatial regions: the intermembrane space (IMS) and the mitochondrial matrix, where mitochondrial DNA (mtDNA) and ribosomes are present and participate in the transcription and translation of proteins encoded by mitochondria [3, 4]. The IMS, localized between OMM and IMM, controls important functions, such as protein sorting, redox balance with glutathione (GSH) reduction and oxidation, cytochrome c (Cyt C) release, and apoptotic cascade activation [4, 5]. Distinctive characteristics and particular functions distinguish mitochondrial membranes from other membrane bilayers. Even at the specific contact sites between the OMM and IMM, there exists a different protein and lipid composition [6, 7].

    Schematic illustration of mitochondrial architecture and function. Mitochondria are conformed by two bilayers separated by the intermembrane space: the outer mitochondrial membrane (OMM) and the inner mitochondrial membrane (IMM) [13]. The IMM surrounds the mitochondrial matrix, where mitochondrial DNA (mtDNA) and ribosomes are located [4], and it includes the inner boundary membrane (IBM) and mitochondrial cristae. Each bilayer has a specific lipid composition and IMM is specially enriched in proteins [7]. Concretely, mitochondrial cristae contain the respiratory chain complexes and the F1F0-ATP synthase, harboring oxidative phosphorylation to obtain energy. The overflow of electrons between each complex generates reactive oxygen species (ROS) that are reduced by mitochondrial antioxidants, such as glutathione (GSH) [8]. ROS can impair mitochondrial constituents, such as mtDNA, phospholipids, and proteins, leading to cytochrome c (Cyt c) detachment from IMM and releasing it into the cytosol via Bax/Bak oligomerization [4, 5, 9]. Furthermore, ROS production can be enhanced by an altered calcium (Ca2+) homeostasis. Mitochondrial dynamics depend on fusion/fission processes. Fusion relies on mitofusin 1/2 (MFN1/2) of the OMM, and optic atrophy protein 1 (OPA1), placed at IMM. In contrast, fission is regulated by dynamin-related protein 1 (Drp-1), which interacts with fission protein 1 (FIS1) [10, 11]. Mitochondria also have a protein machinery to translocate proteins into the matrix. This machinery is composed of the translocase of the outer membrane (TOM) channel the sorting and assembly machinery (SAM) in the OMM and the translocase of the inner membrane (TIM23) in the IBM [1217]. Ub: ubiquitin; SUMO: small ubiquitin-like modifier; tBID: proapoptotic truncated BID; VDAC: voltage-dependent anion channel; SOD2: superoxide dismutase 2; GPX: glutathione peroxidase; GSSG: glutathione disulfide; O2•−: superoxide; OH: hydroxyl radical

    Note. Adapted from “Electron Transport Chain”, “Mitochondrial Membrane (Phospholipid Bilayers)”, and “Protein Import into the Mitochondria”, by BioRender.com (2024). Retrieved from https://app.biorender.com/biorender-templates

    Mitochondrial membranes exhibit predominantly phospholipids, such as phosphatidylcholine (PC), phosphatidylethanolamine (PE), and phosphatidylserine (PS), and low levels of sphingolipids and sterols [1821]. A unique mitochondrial characteristic consists in the presence of cardiolipin (CL), a negatively charged phospholipid located in the IMM, which plays an essential role in the maintenance of membrane integrity and cristae morphology [22]. Its particular conical shape allows for hexagonal structure formation that generates strongly curved areas within the IMM. Moreover, CL is involved in the generation and preservation of protein-protein and protein-membrane interactions [23, 24], conservation of the mitochondrial respiratory chain complexes [25, 26], organization of supercomplexes [27], and assembly of F1F0-ATP synthase dimers. Therefore, compromised CL biosynthesis is linked to damaged cristae morphology and consequently modifies mitochondria shape and dynamics [2830], respiration, and capability to cope with energy demands through oxidative phosphorylation (OXPHOS) [31, 32]. CL can also flip to the OMM, serving as a signaling molecule to activate mitophagy and apoptotic signaling pathways [33, 34] under stress conditions.

    Another feature of mitochondria is the high content of proteins, the majority of which are implicated in OXPHOS and organized in respiratory chain complexes, including the F1F0-ATP synthase and the presence of solute carriers in IMM [3537]. Most mitochondrial proteins are generated as precursors on cytosolic ribosomes and then transferred and organized into distinct organelle compartments by protein translocases [1216]. Moreover, mitochondria continuously experience fusion and fission, which implicate transitory disruption of the classical membrane bilayer and mixture of phospholipids. This event promotes the interchange of mitochondrial content, including mtDNA, which is critical for the generation of ATP in the OXPHOS [10, 11]. Besides, the IMM splits into inner borders and cristae to define specific invaginations where the respiratory chain machinery is located, which can undergo extensive transformation to release Cyt C during apoptotic signaling [9]. The mitochondrial electron transport chain (ETC) is one of the principal sources of reactive oxygen species (ROS) [8], principally due to complex I (CI, NADH coenzyme Q reductase) and complex III (CI, ubiquinol Cyt C reductase) activities [38, 39]. During electron trafficking to molecular oxygen (O2) for ATP production in the ETC, electron leakage at these two complexes is accepted by O2, generating superoxide anion (O2•−) as a side product of OXPHOS, that subsequently dismutates to hydrogen peroxide (H2O2) [40, 41].

    The endoplasmic reticulum (ER) is responsible for lipid biosynthesis in cells, arising as the main source of essential components of membrane bilayers such as phospholipids and cholesterol, which are distributed to different intracellular compartments, including mitochondria. Unlike other lipid species, CL is synthesized within mitochondria where it plays essential roles in mitochondrial function (see below), Thus, the communication between mitochondria and the ER via mitochondria—ER membrane contact sites is crucial to mitochondrial lipid biosynthesis and calcium exchange [42].

    In this review, we will provide evidence for the contribution that alterations in mitochondrial membrane lipid composition have on the development and progression of metabolic liver diseases, including metabolic dysfunction-associated steatotic liver disease (MASLD) and alcohol-related liver disease (ARLD).

    Mitochondrial lipid composition

    Lipid composition influences the biophysical properties of membrane bilayers. In turn, membrane fluidity controls diverse membrane-associated processes, including membrane—protein contacts and function, signal transduction, and membrane fusion and fission events that ultimately regulate organelle biogenesis and growth, dynamics, and vesicular trafficking [43, 44]. In addition, lipids can act as signaling intermediates that trigger specific signaling pathways, as best illustrated by the hydrolysis of sphingomyelin by sphingomyelinases.

    Focusing on the molecular organization of membrane lipids, they can generally be classified as phospholipids, sphingolipids, and sterols [45]. Phospholipids, the most abundant lipids in bilayers, are formed by a glycerol backbone with one (or two, in CL) phosphate group(s) together with fatty acid (FA) side chains [46]. The main phospholipids of mitochondrial membranes are PC and PE, accounting for 40–45% and 25–30% respectively of the total lipid composition, with phosphatidylinositol (PI) and CL representing 10–15%, while PS is the less abundant phospholipid (3–5%) [1821]. The amount of these lipids differs between the IMM and OMM. For example, the IMM is enriched in PE and CL [21, 24].

    In contrast, sphingolipids are characterized by a sphingoid backbone formed by the condensation between palmitoyl-CoA with serine [47]. Sphingosine is the singular sphingoid precursor, consisting of the palmitoyl-CoA-serine backbone. Fatty acyl chains of varying length acylate the sphingosine backbone to form sphingolipids of which ceramides are the most intensively studied. Ceramides are a heterogeneous family of sphingolipids that exhibit a large degree of FA of different lengths determined by 6 ceramide synthases (CerS1–CerS6), which exhibit different affinity towards short or long-chain FA. Ceramides, like most sphingolipids, are synthesized in the ER and then undergo exchange and transport to different membrane bilayers in the cell, including mitochondria through the ER-mitochondria contact sites [48, 49].

    Not only does the presence of diverse fatty acyl chains with different lengths and saturation regulates the flexibility and stiffness of the bilayer, but also the existence of cholesterol is key in membrane fluidity regulation [50]. For instance, although the amount of cholesterol in mitochondrial membranes is low compared to its abundance in plasma membranes, the OMM is relatively enriched in this sterol compared to IMM.

    Fatty acyl chains in the backbone of lipids can be oxidized by lipid peroxidation, which results in membrane bilayer damage thereby affecting cell integrity and organelle performance [5154]. Oxidative damage to the mitochondrial lipidome induces mitochondrial dysfunction [55]. As mentioned above, CL is crucial for OXPHOS [23, 27, 56, 57], mitochondrial dynamics [58], ATP production, and apoptosis [59, 60]. Due to this key role in mitochondrial physiology, decreased CL levels caused either by low biosynthesis of CL, as in the rare genetic disorder of Barth syndrome, or CL loss by lipid peroxidation due to its susceptibility to ROS attach, compromise mitochondrial function with low OXPHOS activity and ATP generation [61].

    Mitochondrial dynamics are controlled by lipid composition via diacylglyceride (DAG), PE, PS, and cholesterol, promoting negative membrane bends essential for mitochondrial fusion [62]. In addition, the import of proteins is pivotal for mitochondrial synthesis and activity. Many mitochondrial proteins are produced as precursors on cytosolic ribosomes and then are transferred to mitochondria. Phospholipids possess important functions in protein trafficking through and into mitochondrial membranes. Concretely, CL, PE, and PC distinctly influence protein translocases of both OMM and IMM, such as the translocase of the OMM (TOM) complex activity, which depends on OMM phospholipids [17], and the dynamic translocase of the inner membrane 23 (TIM23) and S-adenosylmethionine carrier (SAM) complexes. The association of phospholipids-proteins in specific domains displays a central task in the biogenesis and function of mitochondria [63, 64]. Modifications in the content of lipids within the organelle induce expanded, fragmented, and dysfunctional mitochondria, which is associated with diverse diseases [65].

    Mitochondria in steatotic liver diseases

    The liver is a central hub for lipid metabolism and the disruption of hepatic lipid homeostasis is a key step in metabolic liver disease development. As liver mitochondria oxidize FA to obtain energy by ETC, the loss of mitochondrial function due to changes in mitochondrial membrane lipid composition can contribute to the accumulation of lipids, resulting in steatosis. This first phase of MASLD can progress to advanced stages of the disease, characterized by mitochondrial dysfunction, generation of oxidative stress, hepatocellular damage, inflammation, and fibrosis. In Table 1, we have summarized recent results on how changes in different lipids (from mitochondrial membranes or as a result of mitochondria-lipid interaction), influence the development of liver diseases.

    Changes in mitochondrial lipid composition in different disease models of MASLD, ARLD, and HCC

    LipidsMitochondrial changesDisease outcomesDisease model
    Fatty acids↑ mitochondrial fatty acid β-oxidation at early stagesARLD [66]
    MASLD [67]
    Mice
    ↓ mitochondrial fatty acid β-oxidation at advanced stagesARLD [6870]
    MASH [67, 68, 71]
    Mice
    ↓ nicotinamide adenine dinucleotide (NAD+/NADH) levels in mitochondriaARLD [69]
    MASLD [68]
    ↑ carnitine palmitoyltransferase-1 in mitochondrial membraneMASLD [67]Mice
    ↑ lipid peroxidation in mitochondriaARLD [72]
    MASLD [71]
    MASH [71]
    Rat
    HepG2
    ↓ ETC coupling (CI, CIV)MASLD [71]
    MASH [67, 71]
    Mice
    ↓ mitophagy mediated by NLRP3 activation and AMPK inhibitionMASLD [73, 74]Mice
    Cells
    ↑ mitochondrial attachment to lipid droplets because of diacylglycerol-O-acyltransferase-2 increased activityMASLD [67]
    ↑ lipid peroxidation in mitochondriaARLD [72]
    MASLD [71]
    MASH [71]
    Rat
    HepG2
    ↓ mesh due to altered mitochondrial membrane compositionARLD [75, 76]
    MASLD [68]
    Rat
    GlycerideDiacylglycerides↑ pyroptosis via NLRP3 activationMASH [77]Mice
    Human
    Triglycerides↑ mitochondrial oxidative fluxMASLD [68]
    ↓ membrane fluidity if the cholesterol/triglycerides ratio is alteredARLD [76]Rat
    ↑ tumor anabolismHCC [78]
    PhospholipidCardiolipin↑ NLRP3 and apoptosis by CL peroxidation and redistribution from IMM to OMMARLD [69, 75, 76]
    MASLD [68]
    Rat
    ↓ ETC complex activity (CI, CIII, CIV, and ADP/ATP carrier)ARLD [69]
    MASLD [67, 71, 79]
    Rat
    ↑ mPTP opening and cytochrome c release by Bcl-2 family proteins interaction (Bax)ARLD [80]
    MASLD [67, 71]
    MASH [80]
    Rat
    Phosphatidylcholine↓ mitochondrial ROS production by CYP2E1 inhibitionARLD [72]
    ↑apoptosis due to changes in mitochondrial phosphatidylcholine redox state and through JNK activationARLD [76]
    MASLD [68, 79]
    MASH [68, 77]
    Mice
    Rat
    Human
    Phosphatidylethanolamine↓ membrane fluidityARLD [76]
    SphingolipidCeramide↑mitochondrial ROS generation and apoptosis by TNFα/Fas signalingARLD [70, 75, 81]
    MASLD [68, 73]
    MASH [81]
    PMH
    ↓ ETC (CIII)ARLD [70]
    MASH [67]
    Mice
    ↓ mitochondrial fatty acid β-oxidationMASLD [68]
    MASH [67]
    Mice
    ↓ mitophagy through NLRP3 activation    MASLD [73]
    ↓ mitochondrial membrane permeabilizationHCC [82]Cell line
    ↑ mitochondrial depolarizationMASLD [82]
    Ganglioside↑ ETC (CIII)MASH [67]
    SterolCholesterol↑ mitochondrial ROS productionARLD [75, 83]
    MASH [83]
    Cells
    Human
    ↓ ETC (CI)MASLD [84]
    ARLD [69]
    HCC [83]
    ↑survival by a defective assembly of the apoptosomeHCC [80, 83]Rat
    ↓ mitochondrial membrane permeabilizationARLD [75, 76, 83]
    MASLD [84]
    MASH [67, 83, 85]
    HCC [80, 83]
    HepG2
    Mice
    Rats
    Monkeys
    Human
    ↓ mitochondrial protein transport (SLC25A11) by TNFα and Fas-induced apoptosisARLD [69, 75, 80, 83, 86]
    MASH [80, 83, 85, 87]
    PMH
    Mice
    Human
    ↑ mitochondrial fusion (megamitochondria)ARLD [69]Mice
    ↑ mPTP by JNK-dependent proinflammatory pathwayARLD [75]
    MASH [68]
    PMH
    ↑ alternative (acidic) bile synthesis pathwayMASLD [84]
    MASH [88, 89]
    HCC [88]
    PRH
    Mice
    Lipid droplets↓ motility and fusion rates of peridroplets mitochondriaMASLD [67]
    ↑ megamitochondria through fusion-fission rates alterationARLD [66]Mice
    ↑ function of cytosolic mitochondriaMASLD [90]
    HCC [90]
    Display full size

    MASLD: metabolic dysfunction-associated steatotic liver disease; ARLD: alcohol-related liver disease; HCC: hepatocellular carcinoma; MASH: metabolic-associated steatohepatitis; ETC: electron transport chain; NLRP3: NLR family pyrin domain containing 3; IMM: inner mitochondrial membrane; OMM: outer mitochondrial membrane; mPTP: mitochondrial permeability transition pore; ROS: reactive oxygen species; JNK: c-Jun N-terminal kinase; TNFα: tumor necrosis factor-alpha

    Mitochondria in MASLD and progression to advanced stages

    MASLD is a multifaceted spectrum of liver alterations linked to genetic, epigenetic, and environmental risk factors that affect lipid homeostasis, altering the generation, oxidation, and release of free fatty acids (FFA), cholesterol, triglycerides (TG), and bile acids (BAs). First considered as a consequence of “two-hits” in a simplistic initial hypothesis, the pathogenesis of this complex liver disease was later recognized in the “multiple hits” hypothesis in which the accumulation of lipids in hepatocytes in the onset of MASLD sensitizes the liver to the action of several hits [9193]. Therefore, steatosis can progress to advanced forms, such as metabolic-associated steatohepatitis (MASH), where lipid deposition coexists with inflammation, liver injury, and fibrosis. Then, it can progress to cirrhosis and culminate in hepatocellular carcinoma (HCC) [94, 95]. MASLD is recognized as the most prevalent chronic liver disease worldwide (25% occurrence of normal population) and more than 50% of type 2 diabetic and overweight patients suffer it [96]. Hence, the current definition of MASLD includes steatotic, overweight, type 2 diabetes, or metabolic dysregulated patients [97, 98].

    During steatosis, impaired lipid and glucose metabolism arises principally from high-fat diet (HFD) consumption, resulting in diverse lipid accumulation (FFAs, DAG, TG, ceramides, and cholesterol). The complex process associated with mitochondrial proteome alteration and mitochondrial dysfunction is considered a major player in the transition from MASLD to MASH [99]. Mitochondria play a central role in the regulation of lipid metabolism and hence disruption of mitochondrial function contributes to hepatic steatosis [100].

    Mitochondrial lipid variation has been described in patients at different stages of the MASLD continuum, with increased CL and ubiquinone in early MASLD but increased acylcarnitine in MASH [101]. The impact of these changes on mitochondrial function during MASLD is not well established. Patients with MASH display reduced activities of the respiratory chain complexes, which correlated with serum tumor necrotic factor-alpha (TNFα), and insulin resistance (IR) [101]. There are findings either indicating impaired respiration or increased mitochondrial mass but lower maximal respiration in obese subjects with MASH, which contrasts with reports of increased hepatic mitochondrial function in the same category of patients [102]. In line with the impaired mitochondrial performance, increased hepatic oxidative stress and oxidative DNA damage have been reported in parallel with reduced antioxidant defense [74, 103].

    MASH patients also accumulate microtubule-associated protein 1A/1B-light chain 3 (LC3-II) and sequestosome-1 (p62), a marker of Mallory-Denk bodies, which are associated with disease complications [104]. As mitochondrial quality control is regulated by mitophagy, intervention in this process modulates the implication of mitochondria in MASLD progression. In this regard, knocking down the cell survival regulator macrophage stimulating 1 (MST1), promoted PTEN-induced kinase 1 (PINK1)/Parkin-related mitophagy and reduced HFD-associated liver damage [105]. Besides, optic atrophy type 1 protein (OPA1) inhibition prevented mitophagy intermediates overload and mitigated methionine-choline-deficient (MCD) diet-promoted liver damage [106].

    Mitochondrial morphology changes during MASLD in experimental models, with the appearance of fragmented morphology in parallel with Ca2+ increment, reduced OXPHOS activity, and increased ROS generation. Mitochondrial ROS generation has detrimental effects on the development of MASLD [107]. For instance, elevated ROS levels induce the production of cytokines and c-Jun N-terminal kinase (JNK) activation, which plays a feed-forward loop in mitochondrial dysfunction. This scenario leads to impaired β-oxidation, promoting FA accumulation and ATP depletion in hepatocytes while worsening insulin signaling. Damaged mitochondria accumulation within hepatocytes drives necrotic cell death and the leakage of DNA-enhanced mitochondria-induced danger-associated molecular patterns (DAMPs), such as mtDNA, N-formyl peptides, and ATP, which promote the inflammasomes NLR family pyrin domain containing 3 (NLRP3) and absent in melanoma 2 (AIM2) through toll-like receptors (TLRs) recognition [108110]. It is also important to mention that the translocation of CL to the OMM acts as a docking site to recruit and bind inflammasome components like NLRP3. Furthermore, these signals promote TLR9 activation on Kupffer cells (KCs) and hepatic stellate cells (HSCs), and formyl peptide receptor 1 (FPR1) stimulation, which induces interferon regulatory factor (IRF) and nuclear factor kappa β (NFκβ) action [111]. In turn, the generation of inflammatory cytokines and activation of fibrogenic players establish a chronic inflammatory environment that participates in the progression of MASLD towards MASH and fibrosis (Figure 2). DAMPs have been detected in the plasma of MASH patients, linking mitochondrial dysfunction and inflammation during MASH [112]. Mitochondrial injury also promotes the depletion of nicotinamide adenine dinucleotide (NAD+/NADH) levels, which regulate an adaptive reaction to increased FFA hepatic levels [113] together with an augmented synthesis of mitochondrial free cholesterol and the JNK-dependent proinflammatory routes [114, 115]. The last promotes leakage of mitochondrial components via the mitochondrial permeability transition pore (mPTP) induction and hepatocyte death [116].

    Mitochondrial dysfunction in metabolic-associated steatohepatitis-hepatocellular carcinoma (MASH-HCC) progression. In the cytoplasm, fatty acids bound to CoA (FA-CoA) are transported through the outer (OMM) and inner mitochondrial membranes (IMM), for β-oxidation. The resulting acetyl-CoA is metabolized in the tricarboxylic acid cycle (TCA) or used in the mevalonate pathway to synthesize cholesterol, which can also come from diet and be transported to the mitochondria via steroidogenic acute regulatory protein (STARD1) transporter [87, 88]. STARD1 is not only overexpressed in hepatic cells but also stellate hepatic cells. Changes in the fluidity of the mitochondrial membrane due to cholesterol deposition lead to reduced activity of mitochondrial proteins such as the 2-oxoglutarate carrier (2-OGC). Alterations in OXPHOS, reactive oxygen species (ROS) production [91], the low levels of mitochondrial glutathione (mGSH), and tumor necrosis factor-alpha (TNFα) signaling [101, 117] stimulate c-Jun N-terminals kinase (JNK) action, leading to altered glucose metabolism, apoptosis via mitochondrial permeability transition pore (mPTP) formation [87] and the apoptosome formation by cytochrome c release. In the transition between MASH and HCC, mitophagy is compromised: as the mitochondrial membrane potential shrinks, PTEN-induced kinase 1 (PINK1) recruits Parkin, which ubiquitinates the voltage-dependent anion channel (VDAC) [108110]. The overexpression of macrophage stimulating 1 (MST1) enhances repressed Parkin, leading to mitochondrial fission activation and mitophagy inhibition [104, 105]. A high-fat diet can lead to intestinal microbiota dysbiosis. By entering the portal circulation, pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) bind to toll-like receptors (TLR) from Kupffer cells and activate nuclear factor kappa B (NFκβ) [108110, 118]. This initiates the gene transcription of inflammasome components, such as NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3). Active caspase 1 promotes pro-interleukin-1β (pro-IL-1β) and gasdermin D (GSMD) cleavage into their mature forms. The amino terminal fragments of N-terminal of gasdermin D (GSDMD) form pores at the cell membrane by oligomerization for pyropoptosis. In turn, IL-1β release promotes attraction and activation of further immune cells to the liver as well as stimulates HSC [77, 119, 120]. PAMPs and DAMPs can bind also to formyl peptide receptor 1 (FPR1), stimulating fibrogenic gene transcription. Foam hepatocytes release vesicles containing mitochondrial DNA (mtDNA) and intact mitochondria. Once DAMPs are internalized, TLR9 recognizes mtDNA at the endosomes of Kupffer cells and hepatic stellate cells, inducing proinflammatory cytokines secretion [112, 121, 122], such as TNFα [101] and IL-1β, which can further enhance hepatic damage. LD: lipid droplets; TAG: triacylglycerides; LC3-II: microtubule-associated protein 1A/1B-light chain 3; IRF1: interferon regulatory factor 1; OPA1: optic atrophy type 1 protein; Cyt C: cytochrome c; ROS: reactive oxygen species; ETC: electron transport chain

    Note. Adapted from “Suppression of Inflammasome by IRF4 and IRF8 is critical for T cell Priming”, by BioRender.com (2024). Retrieved from https://app.biorender.com/biorender-templates

    Concerning the transition from steatosis to MASH, pioneering observations pointed at free cholesterol increment. Its putative trafficking to mitochondria has emerged as an important player as shown in a cohort of patients with established MASH, which exhibited increased free cholesterol content and high expression of the steroidogenic acute regulatory protein (STARD1) compared to patients with simple steatosis [87]. These findings have been identified in the progression of MASLD towards advanced stages like HCC [122124]. Thus, the cholesterol deposition at IMM by STARD1 has important negative consequences for the mitochondrial status, including the depletion of a crucial antioxidant defense like GSH, which reflects the defect in the activity of the 2-oxoglutarate carrier (2-OGC; SLC25A11) due to its sensitivity to cholesterol-promoted modifications in membrane fluidity [125, 126], and the impairment in the assembly of respiratory supercomplexes and thus OXPHOS [117, 127]. Hence, STARD1 activation reflecting increased mitochondrial cholesterol (mChol) levels [128], account for the mitochondrial GSH (mGSH) depletion found in MASH patients [129, 130], who also presented mitochondria ultrastructural abnormalities [131] and mitochondrial dysfunction [132]. In addition, the accumulation of cholesterol in KCs and HSCs accounts for stimulated inflammation and fibrosis, characteristic of the advanced stages of MASLD [87, 121, 122, 133]. Overall, the evidences in patients and experimental models indicate that the type rather than the amount of fat is a key player in MASLD progression, with the increase in cholesterol and in particular in mitochondrial membrane emerging as a putative new target for intervention to prevent progression of MASLD.

    Mitochondria function in HCC

    HCC is the main type of liver cancer and the second principal cause of cancer-related demise in the world due to the delay in diagnosis and modest therapeutic strategies, becoming a global health concern [134, 135]. HCC is the final stage of chronic liver diseases caused by several etiologies, including viral hepatitis, ARLD, or MASH [136, 137]. The pathogenesis of HCC is complex and multifactorial and, besides genetic factors and DNA damage, a plethora of players, including lipotoxicity, mitochondrial dysfunction, oxidative stress, inflammation, ER stress, and the disruption in Ca2+ homeostasis induce the perfect milieu for tumor development [136] (Figure 3).

    Mitochondrial changes in hepatocellular carcinoma (HCC) development. Lipid accumulation in the liver results in an alteration of Ca2+ homeostasis [138]. The loss of the cation in the endoplasmic reticulum (ER) and consequent increase in the cytoplasm and mitochondrial matrix of the hepatocyte leads to the creation of reactive oxygen species (ROS) that causes mutations in nuclear and mitochondrial DNA (mtDNA), thus activating proliferative genes and inhibiting oncosuppressor genes [138]. Among them, nuclear factor erythroid 2-related factor 2 (Nrf2) activation defends from oxidative stress, reduces cytokeratin 19 (CK-19) expression, and promotes the ER stress response mediated by the fibroblast growth factor 19 (FGF19) pathway [139, 140]. Also, the overexpression of proliferator-activated receptor-gamma coactivator-1 beta (PGC-1β) induces fatty acid oxidation gene expression and promotes tumor growth and anabolic metabolism. The main lipids in HCC are cholesterol and cardiolipin (CL) [78]. Increased cholesterol in the inner mitochondrial membrane (IMM) due to overexpression of the STARD1 transporter, alters membrane fluidity, endowing tumor cells with reduced permeability and thus increased resistance to chemotherapy [84, 141143]. Furthermore, the acidic pathway of bile acid (BA) synthesis is overactive. Although membrane stiffness affects the activity of the glutathione transporter 2-oxoglutarate (2-OGC), stabilization of hypoxia-inducible factor 1 (HIF-1) promotes its overexpression, thus maintaining glutathione (GSH) levels in the mitochondrial matrix [125, 126]. The high antioxidant capacity of tumor cells prevents oxidation of CL, keeping the electron transport chain stable and preventing the release of cytochrome c [28, 80]. In turn, cholesterol accumulation increases the synthesis of lipotoxic BAs resulting from the acid pathway of BA synthesis in mitochondria [88]. In HCC, mitochondrial dynamics are altered by overexpression of dynamin-related protein 1 (DRP1) and mitochondrial fission 1 protein (Fis1) and decreased expression of mitofusin 1/2 (MFN1/2), leading to increased mitochondrial fission [144146]. DAG: diacylglyceride; PKC: protein kinase C; MAMs: mitochondria-associated membranes; ATF4: activating transcription factor 4; HO-1: heme oxygenase 1; GSK-3β: glycogen synthase kinase-3 beta; FGFR4: fibroblast growth factor receptor 4; OPA1: optic atrophy type 1 protein; Ub: ubiquitin. Created with BioRender.com

    Metabolic variations are abundant in cancer pathogenesis. HCC cells subsist in a potently abundant fat milieu [78, 147]. Peroxisome proliferator-activated receptor gamma co-activator 1β (PGC-1β) controls hepatic oxidative metabolism, mitochondria biogenesis, and antioxidant defense mechanisms. It also plays a pivotal function in cancer progression, supporting metabolic modifications by lipogenic enzyme activation, driving tumor growth and anabolism, and activating gene expression in FA and TG production [78]. Elevated PGC-1β levels activate the expression of ROS scavengers, such as superoxide dismutase (SOD), catalase, glutathione peroxidase (GPX), glutathione reductase (GR), thioredoxin (Trx) and peroxiredoxins (Prx), reducing ROS overload and subsequent apoptosis. In contrast, PGC-1β knockdown protects mice from cancer development [78].

    Linked to the progression of MASLD, unphysiological overload of mChol has been found in HCC and has been associated with tumor growth and malignancy [141, 142, 148]. As previously described, mChol promotes a reduction of the mitochondria membrane fluidity [84, 149], increasing the mitochondrial membrane order of HCC cancer cells [143, 150152]. Paradoxically, this event does not decrease mGSH in cancer cells because of the adaptive overexpression of 2-oxoglutarate transporter (2-OGC or SLC25A11) through hypoxia-inducible factor-1 (HIF-1) stabilization [125, 126, 152]. Consequently, ATP production is supported via OXPHOS and glycolysis [152, 153], which is an intriguing vis-à-vis the reported negative effect of mChol in the assembly of mitochondrial respiratory supercomplexes to support OXPHOS [84]. Therefore, this scenario favors tumor growth by the synergism between protection against mitochondrial OMM permeabilization and defense against oxidative stress [117, 154]. In line with these effects of mChol in cancer cell survival, recent findings indicated that mChol overload promotes sorafenib resistance of HCC cells [155], validating the critical function of mChol in HCC progression.

    As alluded to above, CL is vital for mitochondrial ATP production in the respiratory chain and for preserving IMM organization [28, 80]. Oxidative modifications of CL, due to ROS attack to double bonds of its FA constituents, not only influence CI, CIII, and complex IV (CIV) function [156158] but also regulate programmed cell death by controlling Cyt C leakage and by attaching to the B-cell lymphoma 2 (Bcl-2) family protein Bid to promote Bax and Bak oligomerization and subsequent OMM permeabilization [34, 71, 79, 159, 160]. Thus, the outcome of mChol acting as a proapoptotic versus an antiapoptotic factor in HCC cells depends on the oxidized status of CL, which in turn depends on the mGSH levels. In stages pre-HCC like early MASLD, mChol accumulation causes mGSH depletion and CL peroxidation, which overall contribute to hepatocellular cell death and mitochondrial dysfunction. However, in established HCC, CL is intact due to increased expression of SLC25A11, contributing to apoptosis resistance and tumor growth promotion.

    The transport and metabolism of cholesterol in IMM acts as an alternative pathway for the generation of BAs generation in the so-called mitochondrial acidic pathway [161163]. BAs are synthesized in hepatocytes predominantly by the classic pathway, which is controlled by 7α-hydroxylase (CYP7A1), and to a lesser extent by the mitochondrial alternative pathway, which is determined by the availability of cholesterol in the IMM for its metabolism by sterol 27-hydroxylase (CYP27A1). As the transport of cholesterol to IMM is determined by STARD1, the overexpression of this carrier in MASLD dictates a unique role for STARD1 in stimulating BA synthesis in mitochondria. BAs are not only essential for fat digestion but they also regulate gene expression and promote inflammation. Thus, the expression of STARD1 favors the switch in the synthesis of BAs from the classic to the alternative pathway, and the generation of mitochondrial-derived chenodeoxycholic acid and its taurine forms have been described as a crucial step in the MASLD-driven HCC development [88].

    Mitochondria in ARLD

    ARLD encompasses a spectrum of hepatic alterations as the result of the ingestion of elevated doses of alcohol that comprises from steatosis, the first stage, to alcoholic hepatitis, cirrhosis, and HCC. Alcohol oxidative metabolism induces several events that are involved in ARLD progression, including the disruption in the balance between fat synthesis and degradation that underlies the onset of steatosis and the alterations in mitochondrial function and morphology.

    Hepatic ethanol metabolism initiates with the enzyme alcohol dehydrogenase (ADH) transforming alcohol to acetaldehyde, which is then metabolized to acetate by the acetaldehyde dehydrogenase (ALDH). In addition, alcohol is also metabolized via the microsomal system cytochrome P450, CYP2E1, to acetaldehyde. Although the ADH has a higher affinity for alcohol than CYP2E1, the chronic consumption of alcohol induces the expression of CYP2E1 and becomes the preferential metabolic pathway for the metabolism of alcohol. In turn, acetaldehyde and the derived malonaldehyde can form protein adduct due to their reactivity, which can be enclosed by KCs, endothelial, and HSCs. This activates an inflammatory reaction that contributes to ARLD progression [164]. The molecular mechanisms that promote the deleterious effects of alcohol metabolism to cause ARLD are intricate and multifactorial and include disruption of lipid and methionine metabolism, alterations in mitochondrial dynamics, respiration, membrane structure, mtDNA oxidation, and ROS production [69], whose final impact in ARLD onset is determined by genetic and environmental factors.

    Mitochondrial function in ARLD

    Mitochondrial membrane structure and cholesterol homeostasis in ARLD

    Lipid organization influences membrane conformation and modifies the function of proteins embedded in the bilayer. Two major modifications have been documented in the lipid configuration of hepatic mitochondria from rodents fed with ethanol: elevated cholesterol content and depleted CL level. Changes in lipid composition directly affect the physical properties of the bilayer, as exemplified by the relative ratio of PC to PE and particularly by the cholesterol/phospholipid molar ratio, which is a pivotal factor of membrane fluidity. The length and saturation of the fatty acyl chains of the PC/PE molecular species is another factor that regulates physical membrane properties, but especially the increase in cholesterol in a particular bilayer restricts the rotation of acyl chains and determines the fluidity of the bilayer, which in turn can affect the activity of mitochondrial membrane proteins [75]. In this regard, alcohol consumption stimulates cholesterol content in hepatocytes and STARD1 expression, resulting in the trafficking and accumulation of cholesterol in mitochondrial membranes. Interestingly, the increase in the expression of STARD1 occurs in a zonal-dependent fashion with the predominant expression in the perivenous zone of the liver, coinciding with the predominant expression of CYP2E1 and the site of injury as a consequence of alcohol consumption [165]. The zonal-dependent increase in mChol by STARD1 causes mGSH depletion through impairment of the SLC25A11 function and the onset of oxidative stress, thus contributing to the pericentral damage caused by alcohol intake [125, 166]. As mentioned above, although mChol is the precursor for the synthesis of BAs in mitochondria and cholestasis is an accompanying complication of patients with alcoholic hepatitis, it remains to be established whether or not the increase in mChol by alcohol feeding in the pericentral zone contributes to the cholestatic manifestations in ARLD.

    Mitochondrial dynamics in ARLD

    Mitochondria are dynamic organelles that relocate in the cytoskeleton and control their morphology and function by a fine-tuned and highly regulated fusion/fission process, which is pivotal for the preservation of mitochondrial tasks and the management of metabolism and cellular signaling [167]. The equilibrium between fission and fusion impacts mitochondrial morphology and adapts their function to diverse stresses. It is also associated with cellular division, apoptosis, and autophagy. Mitochondrial dynamics are regulated by mitochondria-shaping proteins (MSP), of which mitofusin 1 and 2 (MFN1/2) and OPA1 are involved in mitochondrial fusion, while the cytosolic dynamin-related protein 1 (Drp-1) has a crucial role in mitochondrial fission [168170].

    ARLD patients exhibit alterations in the morphology of hepatic mitochondria, with the pioneering description of the presence of megamitochondria (large and elongated mitochondria) as a result of an elevated mitochondrial fusion activity, which triggers mitochondrial elongation and is related to the induction of OXPHOS activity and mtDNA relocation in the mild stage of the ARLD spectrum [171]. In addition, patients with alcoholic hepatitis displayed an elevated expression of Drp-1 in severe stages of the disease, an event that correlates with the data described in human precision-cut hepatic slices incubated with different ethanol concentrations [169]. Thus, chronic alcohol ingestion induces mitochondrial hyperfragmentation through Drp-1 overexpression, promoting more serious hepatic damage [169], pointing at Drp-1 inhibition as a potential therapy to regenerate the balance between mitochondrial dynamics. In line with this possibility, Drp-1 genetic knockdown protected against alcohol-induced hepatotoxicity in VL-17A cells and abolished the growth impairment induced by ethanol exposure. Besides, mice with Drp-1 liver-specific deletion fed the acute-on-chronic alcohol model exhibited less liver injury and the appearance of megamitochondria, with similar findings observed in MASLD following Drp-1 inhibition, which caused decreased steatosis and oxidative stress [170, 172]. Thus, based on this existing evidence it is clear that the presence of megamitochondria is a positive adaptive reaction to alcohol intake and their presence in liver biopsies is a clinical and histological factor linked to a better prognosis in AH patients [173176]. However, the elimination of megamitochondria via mitophagy is not an efficient process due to the size of this type of mitochondria, raising the question of whether the presence of megamitochondria is a transient process during the early adaptation to alcohol intake or not, which in case of persistence may contribute to mitochondrial maladaptation and disruption in the innate immune reaction that increases liver injury in the late phase of chronic ARLD [177179]. The influence that the increase in cholesterol accumulation in mitochondria and subsequent change in membrane fluidity has on mitochondrial dynamics in ARLD remains to be fully established.

    mtDNA in ARLD

    In comparison with nuclear DNA, mtDNA is highly susceptible to free radical attack since is not protected by histones, and it is located near the IMM, the main cellular ROS source. Mitochondria display several mtDNA copies, and while some degree of mtDNA oxidation can occur, this may not necessarily compromise mitochondrial function, providing that intact copies are still available [180]. Cells mainly remove damaged mtDNA and replicate intact copies to preserve the global mtDNA pool integrity. Thus, a remarkable transient decline of murine hepatic mtDNA was reported within hours after acute ethanol exposure, which was further prominent in older animals. This is consistent with the appearance of mtDNA fragments in the serum of patients with ARLD [181].

    Alcohol intake and its oxidative metabolism induce oxidative stress, reflecting the unbalance between ROS generation and its scavenging by compromised antioxidant defense strategies. This disrupts mitochondrial organization and function, and subsequently drives to ROS formation in a vicious cycle of injury, which is more evident with aging. The “mitochondrial theory of aging” proposes that alterations in mtDNA overload compromise cellular energy metabolism, affecting cellular life span [182]. Besides O2•− and H2O2 generated by changes in cholesterol homeostasis in mitochondria by ethanol intake, these species can also generate hydroxyl radical (OH) by the Fenton reaction, which can attack mtDNA, leading to the release of purine and pyrimidine bases from mtDNA and strand breakdown [183]. Moreover, OH directly modifies purine and pyrimidine bases, generating 8-oxoguanine (8-oxoG), which is regularly used as an indicator of free radical damage to DNA. It has been demonstrated that 8-oxoG and mtDNA strand break accumulation were remarkably increased in the livers of long-term ethanol-fed rats [184]. Since mtDNA encodes for 13 constituents of the respiratory chain machinery, ethanol-driven mtDNA harm via ROS production can injure the mitochondrial respiratory complexes [185]. Moreover, alcohol impairs mitochondrial ribosomes leading to a reduction in mitochondrial protein synthesis, and ROS can oxidize intramitochondrial proteins [186], contributing to alcohol-induced liver injury.

    Damaged mitochondria can activate the cascade of apoptosis and induce an innate and sterile inflammatory reaction by releasing apoptotic factors and/or mtDNA. In turn, mtDNA triggers cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS), which elevates secondary messenger cyclic 2’3’-cGAMP generation that initiates stimulator of interferon genes (STING), leading to IRF3 and IRF7 activation [187, 188]. This cGAS-IRF3 route is activated in experimental ARLD models and is positively associated with disorder severity in ARLD patients [189].

    Mitochondrial respiration in ARLD

    Alcohol-induced structural changes in mitochondria reflect alterations in mitochondrial OXPHOS. Although primary findings in alcohol-fed rat hepatic mitochondria have revealed depleted respiration via the downregulation of CIII and V synthesis [186, 190], recent findings described an opposing effect in mice, in which alcohol intake elevated state III respiration in hepatic mitochondria [66]. Remarkably, this increment in state III was more dramatic in the intragastric ethanol-fed model, which is associated with increased hepatic damage and advanced stage of ARLD, compared to the milder effect observed in oral ethanol consumption models. The impact of alcohol ingestion on mitochondrial performance is linked to the regeneration of NAD+ from NADH oxidation to stimulate oxidative alcohol metabolism. The transfer of electrons from NADH to the respiratory machinery to oxidize it to NAD+ in murine mitochondria stimulates the generation of ROS from the increased consumption of O2 in the ETC. Interestingly, the elevated respiration in mice vs rats correlates with the species-dependent susceptibility to ethanol-induced liver damage. That fact matches the mitochondrial regeneration of NAD+ from NADH, indicating that augmented respiration links ethanol consumption with elevated metabolism and ROS production. The importance of putative variations of mitochondrial respiration to ARLD patients is not well defined. Indirect evidence indicates a reduced mitochondrial function in ARLD patients, who exhibited a decreased peak exhalation of 13CO2 from 2-keto[1-13C]isocaproic acid whereas aminopyrine breath assay and galactose removal capability were not modified [191]. Besides, in patients with high alcohol consumption, ARLD severity was linked to mtDNA fragment expression in hepatic tissue [192, 193]. Hence, these data suggest that ethanol ingestion is related to mitochondrial respiration disturbances, correlating with the severity of ARLD progression.

    As alluded to above, a consequence of NAD+ regeneration from NADH oxidation, which is required for endured ethanol metabolism, is the increase in the leakage of electrons transported directly to O2 to produce O2•− [72]. In addition to this event, the consumption of ethanol further increased ROS generation via its metabolism through CYP2E1, as besides ER, it is also found in mitochondria and ethanol consumption induces its expression [194, 195]. In mitochondria, the principal defense against O2•− production is SOD2, which generates H2O2, a powerful oxidant that forms reactive radicals via the Fenton reaction. Mitochondrial H2O2 detoxification can occur via the GSH redox cycle and the Prx-III systems. Reduced GSH level is critical for this GSH redox cycle which requires the participation of GPX-4, while the mitochondrial oxidized form of Prx-III, generated after the reduction of H2O2, is regenerated by Trx2 [196]. Mitochondria do not generate GSH de novo, therefore, they depend on cytosolic GSH to provide mGSH to the mitochondrial matrix. mGSH acts as a cofactor for GPX4 to detoxify H2O2 and other FAs-derived peroxides [190]. As mentioned above, the transport of cytosolic GSH through SLC25A11 is susceptible to changes in cholesterol-dependent modifications in membrane fluidity [190, 196, 197]. Although the relative importance between mGSH/GPX and the Prx-III/Trx2 systems in detoxifying H2O2 remains to be established, both antioxidant systems are interconnected, and mGSH depletion determines the efficiency of the Prx-III/Trx2 pair. Thus, alcohol-induced mChol trafficking may result in the perturbation of membrane physical properties, which can alter efficient antioxidant defense mechanisms, leading to alterations in the mechanisms of mitochondrial respiration.

    Mitochondria and ER stress crosstalk in ARLD

    The ER regulates the transport and maturation of membrane and secretory proteins, post-translational protein processing, and Ca2+ homeostasis. ER stress leads to lipid overload, inflammation, and cell death [198], and has been also described to play a key role in hepatic steatosis via sterol regulatory element-binding proteins (SREBPs) activation (Figure 4). Disturbances in protein folding or modifications in lipid homeostasis lead to the initiation of the unfolded protein response (UPR) that induces the expression of the transcription of chaperones (GRP78/BiP) and the ER-associated degradation (ERAD) mechanisms [199201] to reestablish homeostasis. Alcohol ingestion promotes ER stress via diverse mechanisms, such as ceramide production via the acid sphingomyelinase (ASMase) activation, the production of acetaldehyde as a result of protein adducts formation in the ER, and also via oxidative stress [202]. A crucial mechanism of ethanol-promoted ER stress is the disruption of methionine metabolism, followed by a rise in homocysteine levels. In this regard, it has been demonstrated that feeding mice with betaine reduced ethanol-induced ER stress, liver steatosis, and hepatic damage [203]. In addition, ER stress can control mitochondrial function. Thus, Ca2+ homeostasis disturbances in the ER affect mitochondria due to the transport of Ca2+, which induces mitochondrial mPTP and cellular damage [70]. ER and mitochondria display physical interaction via mitochondrial-associated membranes (MAMs) defining the boundary of ER and mitochondria, which operate as a channel for the transfer of ions and lipids. In addition, ER stress can regulate cholesterol overload in mitochondrial membranes via STARD1 upregulation [86], a pivotal participant in metabolic hepatic disorders such as MASH and ARLD as mentioned above [87, 88]. Moreover, another connection between methionine metabolism and mitochondrial crosstalk has been revealed in ARLD. Methionine adenosyltransferase 1A (MAT1A) is normally located in the cytosol and nucleus, although new data described MAT1A also present in mitochondria, where it protects mitochondrial proteome and regulates mitochondrial performance [204]. Hepatic tissue from ARLD patients and mice fed alcohol displayed a remarkable reduction in MAT1A localization in mitochondria, facilitated by the isomerase peptidyl-prolyl cis/trans isomerase (PIN1) and the casein kinase (CK2). Prevention of PIN1-MAT1A interaction increases MAT1A levels in mitochondria and protects against alcohol-promoted mitochondrial malfunction and fat storage. However, whether the advantageous outcomes of targeting mitochondrial MAT1A results from SAM local generation to induce stronger methylation and elevated mitochondrial proteins remains to be established vis-à-vis the direct transport of cytosolic SAM to mitochondrial fraction by a transport system that is insensitive to changes in mitochondrial membrane fluidity [205].

    Alterations in mitochondrial function during alcohol-related liver disease (ARLD) development. Alcohol is metabolized by alcohol dehydrogenase (ADH), leading to acetaldehyde and subsequently to acetate by acetaldehyde dehydrogenase (ALDH). CYP2E1 can also metabolize alcohol in the endoplasmic reticulum. This process, together with the adducts formed by acetaldehyde and its by-products, causes high endoplasmic reticulum stress [69, 164]. Also contributing to this phenomenon is the overexpression of acid sphingomyelinase (ASMase) and the consequent increase in ceramides, oxidative stress, and alterations in methionine metabolism [75, 86]. ER stress promotes the activation of the three branches of the unfolded protein response (UPR) and the overexpression of the cholesterol transporter steroidogenic acute regulatory protein (STARD1) [199, 200, 202]. Thus, in alcoholic steatohepatitis, there is an increased accumulation of cholesterol in the inner mitochondrial membrane (IMM). This cholesterol can come from both the diet and the mevalonate pathway. Its deposition in the IMM together with the decrease in cardiolipin causes increased mitochondrial membrane stiffness, reducing the activity of the 2-oxoglutarate carrier (2-OGC) transporter and, consequently, mitochondrial glutathione (mGSH) levels [75, 206]. In turn, cholesterol induces increased bile acid synthesis through the alternative route, further affecting mitochondrial antioxidant levels. The reduction of mGSH levels together with the high production of reactive species due to the alteration of the mitochondrial electron transport chain leads to an alteration of the glutathione peroxidase (GPX)-peroxiredoxin-3 (Prx-III)-thioredoxin (Trx-2) system [190, 196, 197]. In ARLD, the action of peptidyl-prolyl cis/trans isomerase NIMA-interacting 1 (PIN1) and casein kinase 2 (CK2) are reduced and, consequently, methionine adenosyltransferase 1A (MAT1α) is able to metabolize methionine to S-adenosylmethionine (SAM), which is transported to the mitochondria [204206]. Calcium (Ca2+) homeostasis is altered in this environment and, consequently, mitochondrial permeability transition pore (mPTP) is stimulated. This whole scenario triggers inflammasome activation, IL-1β and IL-18 releasing to recruit immune cells, and leads to a translocation of cardiolipin from the IMM to the outer mitochondrial membrane (OMM) in the mitochondria-associated membranes (MAMs) [120, 207209]. LDL: low-density lipoprotein; LDLR: LDL receptor; PCSK9: proprotein convertase subtilisin/kexin type 9; SOD2: superoxid dismutase 2; O2•−: superoxide; OH: hydroxyl radical; CYP7A1: cholesterol 7α-hydroxylase; CYP27A1: cytochrome P450 family 27 subfamily A member 1; VDAC: voltage-dependent anion channel; MCU: mitochondrial calcium uniporter; IP3R: inositol 1,4,5-trisphosphate receptor; NLRP3: NLR family pyrin domain containing 3; SAH: S-adenosylhomocysteine; IL: interleukin; GSDMD: N-terminal of gasdermin D; TLR: toll-like receptor; PAMPs: pathogen-associated molecular pattern molecules; DAMPs: damage-associated molecular patterns; XBP1: X-box binding protein 1; ERAD: endoplasmic-reticulum-associated protein degradation; ATF4: activating transcription factor 4; ATF6: activating transcription factor 4; CHOP: C/EBP homologous protein; Cyt c: cytochrome c

    Note. Adapted from “Electron Transport Chain”, “PCSK9 Inhibitors”, and “Suppression of Inflammasome by IRF4 and IRF8 is Critical for T cell Priming”, by BioRender.com (2024). Retrieved from https://app.biorender.com/biorender-templates

    Mitochondria and inflammasome crosstalk in ARLD

    Inflammasome is known to be activated in ARLD [207] and it consists of a set of intracellular multiprotein oligomers [NLRP3, caspase-1, interleukin (IL)-1β] found in the cytosol. Inflammasome senses the pathogen-associated molecular patterns (PAMPs) and DAMPs, RNA viruses, pore-forming toxins, cholesterol crystals and uric acid [120, 208], and triggers the generation and release of pro-inflammatory cytokines, as IL-1β and IL-18 [119, 209]. Diverse processes can activate NLRP3 inflammasome at MAMs, such as ROS generation by dysfunctional mitochondria [210], impaired mitophagy [118], mtDNA oxidation [120], and disturbances of the intestinal barrier that trigger the translocation of DAMPs and PAMPs. CL, located in MAMs, flips from the IMM to the OMM triggering NLRP3 initiation [211]. Furthermore, in MAMs, the trafficking of lipids and Ca2+ between ER and mitochondria takes place via voltage-dependent anion channel (VDAC) [212], which enables the NLRP3 inflammasome assembly [73]. Then, the association between ethanol metabolism and NLRP3 initiation is orchestrated via mitochondrial disruption, since mitochondria participate in the oxidative metabolism of ethanol, the initiation of ROS production, and oxidative stress. Thus, the crosstalk between mitochondria and inflammasome promotes ARLD progression and arises as a possible target for therapeutic intervention.

    Conclusions and perspectives

    Lipids are intricate biomolecules that determine the structural and physical properties of membranes, which in turn can regulate multiple signaling pathways. The multifaceted mitochondrial double membrane participates in ATP generation, oxidative stress, and cell death regulation. The lipid composition of the mitochondrial membrane, particularly the IMM, is distinctive in performing its numerous functions. Oxidative stress and apoptosis are well-recognized outcomes from the alterations of CL levels and redox status. Hence, genetic disorders disrupting enzymes involved in the synthesis and maturation of CL result in the disruption of mitochondrial organization with defective ATP generation and early cell death. Furthermore, modified fat metabolism and membrane content are critical characteristics of cancer cells. Alterations in mitochondrial membrane lipid configuration and fluidity are hallmarks of solid tumors, and CL with highly saturated acyl chains has been described in cell death-resistant cancer cell lines. In this regard, the mChol pool regulates metabolism and redox biology and contributes to the development of hepatic disorders, such as MASLD, MASH, ARLD, or HCC. Upregulation of STARD1 expression is responsible for the cholesterol transport and accumulation in mitochondria and its expression is induced by ER stress, thus establishing a link between ER stress and mitochondrial dysfunction in metabolic liver diseases, MASLD and ARLD. In the liver, cholesterol accumulation alters mitochondria membrane fluidity and prevents the trafficking of GSH from the cytosol to the mitochondrial matrix promoting oxidative damage through excess ROS formation. mChol also raises BAs synthesis in the mitochondrial alternative pathway, which influences liver tumorigenesis. Thus, although changes in lipid composition can have a significant impact in membrane physical properties, which in turn affect mitochondrial function, the cumulative evidence points to a crucial role for cholesterol accumulation in orchestrating these deleterious effects in mitochondria via impaired antioxidant defense and subsequent ROS generation, affecting essential unique lipid components like CL. In addition to promoting ROS generation, increased cholesterol in mitochondria may fuel the synthesis of oxysterols and BAs, which can contribute to the progression of MASLD to HCC development. In addition, alterations in hepatic mitochondrial lipid composition may also contribute to liver metastatic colonization by invasive cancer cells [213]. Hence, future developments aimed to prevent the increase in mChol to maintain appropriate mitochondrial function may be promising for the treatments/preventions of these prevalent chronic liver diseases.

    Abbreviations

    ARLD:

    alcohol-related liver disease

    BAs:

    bile acids

    CL:

    cardiolipin

    Cyt C:

    cytochrome c

    DAMPs:

    danger-associated molecular patterns

    Drp-1:

    dynamin-related protein 1

    ER:

    endoplasmic reticulum

    ETC:

    electron transport chain

    FA:

    fatty acid

    FFA:

    free fatty acids

    GPX:

    glutathione peroxidase

    GSH:

    glutathione

    H2O2:

    hydrogen peroxide

    HCC:

    hepatocellular carcinoma

    HSCs:

    hepatic stellate cells

    IL:

    interleukin

    IMM:

    inner mitochondrial membrane

    IRF:

    interferon regulatory factor

    KCs:

    Kupffer cells

    MAMs:

    mitochondrial-associated membranes

    MASH:

    metabolic-associated steatohepatitis

    MASLD:

    metabolic dysfunction-associated steatotic liver disease

    MAT1A:

    methionine adenosyltransferase 1A

    mChol:

    mitochondrial cholesterol

    mGSH:

    mitochondrial glutathione

    mtDNA:

    mitochondrial DNA

    NLRP3:

    NLR family pyrin domain containing 3

    O2:

    molecular oxygen

    O2•−:

    superoxide anion

    OMM:

    outer mitochondrial membrane

    OXPHOS:

    oxidative phosphorylation

    PC:

    phosphatidylcholine

    PE:

    phosphatidylethanolamine

    PGC-1β:

    peroxisome proliferator-activated receptor gamma co-activator 1β

    Prx:

    peroxiredoxins

    PS:

    phosphatidylserine

    ROS:

    reactive oxygen species

    SAM:

    S-adenosylmethionine carrier

    STARD1:

    steroidogenic acute regulatory protein

    TG:

    triglycerides

    Trx:

    thioredoxin

    Declarations

    Acknowledgments

    Cartoons in Figures were created with BioRender.com. Carmen Garcia-Ruiz and José C. Fernández-Checa acknowledge the support of the Spanish National Research Council’s Cancer Hub.

    Author contributions

    LF and LCdlR: Conceptualization, Writing—original draft, Writing—review & editing. JCFC and CGR: Conceptualization, Writing—original draft, Writing—review & editing, Funding acquisition, Supervision. All authors have read and agreed to the published version of the manuscript.

    Conflicts of interest

    Prof. José C. Fernández-Checa is the Editor-in-Chief of Exploration of Digestive Diseases, and Prof. Carmen Garcia-Ruiz is a member of the Editorial Board and a Guest Editor of Exploration of Digestive Diseases. However, neither was involved in the decision-making or review process for this manuscript.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    We are grateful for the support from grants [PID2020-115055RB-I00], [PID2022-1429560B-I00] and [2023AEP068] from Plan Nacional de I+D funded by the Agencia Estatal de Investigación (AEI) and the Fondo Europeo de Desarrollo Regional (FEDER) and from the CIBEREHD; as well as support from AGAUR of the Generalitat de Catalunya [SGR-2021-00491], European Cooperation in Science & Technology (COST) [ACTION CA17112], Prospective European Drug-Induced Liver Injury Network, the 2018-102799-T “Enfermedades Metabólicas y Cancer” from the Red Nacional of the Spanish Health Ministry and the COVID grant from the Spanish Association for the Study of the Liver (AEEH). In addition, this project has received funding from the European Horizon’s research and innovation program HORIZON-HLTH-2022-STAYHLTH-02 under agreement No [101095679]. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

    Copyright

    © The Author(s) 2024.

    References

    Zimorski V, Ku C, Martin WF, Gould SB. Endosymbiotic theory for organelle origins. Curr Opin Microbiol. 2014;22:3848. [DOI] [PubMed]
    Archibald JM. Endosymbiosis and Eukaryotic Cell Evolution. Curr Biol. 2015;25:R91121. [DOI] [PubMed]
    Yang Z, Wang L, Yang C, Pu S, Guo Z, Wu Q, et al. Mitochondrial Membrane Remodeling. Front Bioeng Biotechnol. 2022;9:786806. [DOI] [PubMed] [PMC]
    Iovine JC, Claypool SM, Alder NN. Mitochondrial compartmentalization: emerging themes in structure and function. Trends Biochem Sci. 2021;46:90217. [DOI] [PubMed] [PMC]
    Backes S, Herrmann JM. Protein Translocation into the Intermembrane Space and Matrix of Mitochondria: Mechanisms and Driving Forces. Front Mol Biosci. 2017;4:83. [DOI] [PubMed] [PMC]
    Muñoz-Gómez SA, Slamovits CH, Dacks JB, Wideman JG. The evolution of MICOS: Ancestral and derived functions and interactions. Commun Integr Biol. 2015;8:e1094593. [DOI] [PubMed] [PMC]
    van der Laan M, Horvath SE, Pfanner N. Mitochondrial contact site and cristae organizing system. Curr Opin Cell Biol. 2016;41:3342. [DOI] [PubMed]
    Murphy MP. How mitochondria produce reactive oxygen species. Biochem J. 2009;417:113. [DOI] [PubMed] [PMC]
    Cogliati S, Enriquez JA, Scorrano L. Mitochondrial Cristae: Where Beauty Meets Functionality. Trends Biochem Sci. 2016;41:261273. [DOI] [PubMed]
    Westermann B. Mitochondrial fusion and fission in cell life and death. Nat Rev Mol Cell Biol. 2010;11:87284. [DOI] [PubMed]
    Friedman JR, Nunnari J. Mitochondrial form and function. Nature. 2014;505:33543. [DOI] [PubMed] [PMC]
    Neupert W, Herrmann JM. Translocation of proteins into mitochondria. Annu Rev Biochem. 2007;76:72349. [DOI] [PubMed]
    Chacinska A, Koehler CM, Milenkovic D, Lithgow T, Pfanner N. Importing mitochondrial proteins: machineries and mechanisms. Cell. 2009;138:62844. [DOI] [PubMed] [PMC]
    Endo T, Yamano K, Kawano S. Structural insight into the mitochondrial protein import system. Biochim Biophys Acta. 2011;1808:95570. [DOI] [PubMed]
    Becker T, Böttinger L, Pfanner N. Mitochondrial protein import: from transport pathways to an integrated network. Trends Biochem Sci. 2012;37:8591. [DOI] [PubMed]
    Hewitt V, Alcock F, Lithgow T. Minor modifications and major adaptations: the evolution of molecular machines driving mitochondrial protein import. Biochim Biophys Acta. 2011;1808:94754. [DOI] [PubMed]
    Stan T, Ahting U, Dembowski M, Künkele KP, Nussberger S, Neupert W, et al. Recognition of preproteins by the isolated TOM complex of mitochondria. EMBO J. 2000;19:4895902. [DOI] [PubMed] [PMC]
    Osman C, Voelker DR, Langer T. Making heads or tails of phospholipids in mitochondria. J Cell Biol. 2011;192:716. [DOI] [PubMed] [PMC]
    Horvath SE, Daum G. Lipids of mitochondria. Prog Lipid Res. 2013;52:590614. [DOI] [PubMed]
    van Meer G, Voelker DR, Feigenson GW. Membrane lipids: where they are and how they behave. Nat Rev Mol Cell Biol. 2008;9:11224. [DOI] [PubMed] [PMC]
    Vance JE. Phospholipid synthesis and transport in mammalian cells. Traffic. 2015;16:118. [DOI] [PubMed]
    Schlame M, Greenberg ML. Biosynthesis, remodeling and turnover of mitochondrial cardiolipin. Biochim Biophys Acta Mol Cell Biol Lipids. 2017;1862:37. [DOI] [PubMed] [PMC]
    Claypool SM, Oktay Y, Boontheung P, Loo JA, Koehler CM. Cardiolipin defines the interactome of the major ADP/ATP carrier protein of the mitochondrial inner membrane. J Cell Biol. 2008;182:93750. [DOI] [PubMed] [PMC]
    Gebert N, Joshi AS, Kutik S, Becker T, McKenzie M, Guan XL, et al. Mitochondrial cardiolipin involved in outer-membrane protein biogenesis: implications for Barth syndrome. Curr Biol. 2009;19:21339. [DOI] [PubMed] [PMC]
    Fiedorczuk K, Letts JA, Degliesposti G, Kaszuba K, Skehel M, Sazanov LA. Atomic structure of the entire mammalian mitochondrial complex I. Nature. 2016;538:40610. [DOI] [PubMed] [PMC]
    Gomez B Jr, Robinson NC. Phospholipase digestion of bound cardiolipin reversibly inactivates bovine cytochrome bc1. Biochemistry. 1999;38:90318. [DOI] [PubMed]
    Zhang M, Mileykovskaya E, Dowhan W. Gluing the respiratory chain together. Cardiolipin is required for supercomplex formation in the inner mitochondrial membrane. J Biol Chem. 2002;277:435536. [DOI] [PubMed]
    Paradies G, Paradies V, Benedictis VD, Ruggiero FM, Petrosillo G. Functional role of cardiolipin in mitochondrial bioenergetics. Biochim Biophys Acta. 2014;1837:40817. [DOI] [PubMed]
    Ren M, Phoon CKL, Schlame M. Metabolism and function of mitochondrial cardiolipin. Prog Lipid Res. 2014;55:116. [DOI] [PubMed]
    Shen Z, Ye C, McCain K, Greenberg ML. The Role of Cardiolipin in Cardiovascular Health. Biomed Res Int. 2015;2015:891707. [DOI] [PubMed] [PMC]
    Zhang J, Guan Z, Murphy AN, Wiley SE, Perkins GA, Worby CA, et al. Mitochondrial phosphatase PTPMT1 is essential for cardiolipin biosynthesis. Cell Metab. 2011;13:690700. [DOI] [PubMed] [PMC]
    Sustarsic EG, Ma T, Lynes MD, Larsen M, Karavaeva I, Havelund JF, et al. Cardiolipin Synthesis in Brown and Beige Fat Mitochondria Is Essential for Systemic Energy Homeostasis. Cell Metab. 2018;28:15974.e11. [DOI] [PubMed] [PMC]
    Kagan VE, Tyurin VA, Jiang J, Tyurina YY, Ritov VB, Amoscato AA, et al. Cytochrome c acts as a cardiolipin oxygenase required for release of proapoptotic factors. Nat Chem Biol. 2005;1:22332. [DOI] [PubMed]
    Lutter M, Fang M, Luo X, Nishijima M, Xie X, Wang X. Cardiolipin provides specificity for targeting of tBid to mitochondria. Nat Cell Biol. 2000;2:75461. [DOI] [PubMed]
    Nunnari J, Suomalainen A. Mitochondria: in sickness and in health. Cell. 2012;148:114559. [DOI] [PubMed] [PMC]
    Wallace DC. Mitochondria and cancer. Nat Rev Cancer. 2012;12:68598. [DOI] [PubMed] [PMC]
    Palmieri F, Monné M. Discoveries, metabolic roles and diseases of mitochondrial carriers: A review. Biochim Biophys Acta. 2016;1863:236278. [DOI] [PubMed]
    Starkov AA. The role of mitochondria in reactive oxygen species metabolism and signaling. Ann N Y Acad Sci. 2008;1147:3752. [DOI] [PubMed] [PMC]
    Cadenas E, Boveris A, Ragan CI, Stoppani AO. Production of superoxide radicals and hydrogen peroxide by NADH-ubiquinone reductase and ubiquinol-cytochrome c reductase from beef-heart mitochondria. Arch Biochem Biophys. 1977;180:24857. [DOI] [PubMed]
    Brand MD, Affourtit C, Esteves TC, Green K, Lambert AJ, Miwa S, et al. Mitochondrial superoxide: production, biological effects, and activation of uncoupling proteins. Free Radic Biol Med. 2004;37:75567. [DOI] [PubMed]
    Muller FL, Liu Y, Remmen HV. Complex III releases superoxide to both sides of the inner mitochondrial membrane. J Biol Chem. 2004;279:4906473. [DOI] [PubMed]
    Falabella M, Vernon HJ, Hanna MG, Claypool SM, Pitceathly RDS. Cardiolipin, Mitochondria, and Neurological Disease. Trends Endocrinol Metab. 2021;32:22437. [DOI] [PubMed] [PMC]
    Harayama T, Riezman H. Understanding the diversity of membrane lipid composition. Nat Rev Mol Cell Biol. 2018;19:28196. [DOI] [PubMed]
    Klose C, Surma MA, Simons K. Organellar lipidomics--background and perspectives. Curr Opin Cell Biol. 2013;25:40613. [DOI] [PubMed]
    Dowhan W. Molecular basis for membrane phospholipid diversity: why are there so many lipids? Annu Rev Biochem. 1997;66:199232. [DOI] [PubMed]
    Hoch FL. Cardiolipins and biomembrane function. Biochim Biophys Acta. 1992;1113:71133. [DOI] [PubMed]
    Gault CR, Obeid LM, Hannun YA. An overview of sphingolipid metabolism: from synthesis to breakdown. Adv Exp Med Biol. 2010;688:123. [DOI] [PubMed] [PMC]
    Breslow DK, Weissman JS. Membranes in balance: mechanisms of sphingolipid homeostasis. Mol Cell. 2010;40:26779. [DOI] [PubMed] [PMC]
    Scharwey M, Tatsuta T, Langer T. Mitochondrial lipid transport at a glance. J Cell Sci. 2013;126:531723. [DOI] [PubMed]
    Antonny B, Vanni S, Shindou H, Ferreira T. From zero to six double bonds: phospholipid unsaturation and organelle function. Trends Cell Biol. 2015;25:42736. [DOI] [PubMed]
    Hajeyah AA, Griffiths WJ, Wang Y, Finch AJ, O’Donnell VB. The Biosynthesis of Enzymatically Oxidized Lipids. Front Endocrinol (Lausanne). 2020;11:591819. [DOI] [PubMed] [PMC]
    Bochkov V, Gesslbauer B, Mauerhofer C, Philippova M, Erne P, Oskolkova OV. Pleiotropic effects of oxidized phospholipids. Free Radic Biol Med. 2017;111:624. [DOI] [PubMed]
    Dixon SJ, Stockwell BR. The hallmarks of ferroptosis. Annu Rev Cancer Biol. 2019;3:3554. [DOI]
    Veglia F, Tyurin VA, Mohammadyani D, Blasi M, Duperret EK, Donthireddy L, et al. Lipid bodies containing oxidatively truncated lipids block antigen cross-presentation by dendritic cells in cancer. Nat Commun. 2017;8:2122. [DOI] [PubMed] [PMC]
    Vamecq J, Dessein AF, Fontaine M, Briand G, Porchet N, Latruffe N, et al. Mitochondrial dysfunction and lipid homeostasis. Curr Drug Metab. 2012;13:1388400. [DOI] [PubMed]
    Pfeiffer K, Gohil V, Stuart RA, Hunte C, Brandt U, Greenberg ML, et al. Cardiolipin stabilizes respiratory chain supercomplexes. J Biol Chem. 2003;278:5287380. [DOI] [PubMed]
    Böttinger L, Horvath SE, Kleinschroth T, Hunte C, Daum G, Pfanner N, et al. Phosphatidylethanolamine and cardiolipin differentially affect the stability of mitochondrial respiratory chain supercomplexes. J Mol Biol. 2012;423:67786. [DOI] [PubMed] [PMC]
    DeVay RM, Dominguez-Ramirez L, Lackner LL, Hoppins S, Stahlberg H, Nunnari J. Coassembly of Mgm1 isoforms requires cardiolipin and mediates mitochondrial inner membrane fusion. J Cell Biol. 2009;186:793803. [DOI] [PubMed] [PMC]
    Schug ZT, Gottlieb E. Cardiolipin acts as a mitochondrial signalling platform to launch apoptosis. Biochim Biophys Acta. 2009;1788:202231. [DOI] [PubMed]
    Chu CT, Ji J, Dagda RK, Jiang JF, Tyurina YY, Kapralov AA, et al. Cardiolipin externalization to the outer mitochondrial membrane acts as an elimination signal for mitophagy in neuronal cells. Nat Cell Biol. 2013;15:1197205. [DOI] [PubMed] [PMC]
    Ames BN, Liu J. Delaying the mitochondrial decay of aging with acetylcarnitine. Ann N Y Acad Sci. 2004;1033:10816. [DOI] [PubMed]
    Furt F, Moreau P. Importance of lipid metabolism for intracellular and mitochondrial membrane fusion/fission processes. Int J Biochem Cell Biol. 2009;41:182836. [DOI] [PubMed]
    Ardail D, Gasnier F, Lermé F, Simonot C, Louisot P, Gateau-Roesch O. Involvement of mitochondrial contact sites in the subcellular compartmentalization of phospholipid biosynthetic enzymes. J Biol Chem. 1993;268:2598592. [DOI] [PubMed]
    Vogel F, Bornhövd C, Neupert W, Reichert AS. Dynamic subcompartmentalization of the mitochondrial inner membrane. J Cell Biol. 2006;175:23747. [DOI] [PubMed] [PMC]
    Balboa D, Weltner J, Novik Y, Eurola S, Wartiovaara K, Otonkoski T. Generation of an OCT4 reporter human induced pluripotent stem cell line using CRISPR/SpCas9. Stem Cell Res. 2017;23:1058. [DOI] [PubMed]
    Han D, Ybanez MD, Johnson HS, McDonald JN, Mesropyan L, Sancheti H, et al. Dynamic adaptation of liver mitochondria to chronic alcohol feeding in mice: biogenesis, remodeling, and functional alterations. J Biol Chem. 2012;287:4216579. [DOI] [PubMed] [PMC]
    Amorim R, Magalhães CC, Borges F, Oliveira PJ, Teixeira J. From Non-Alcoholic Fatty Liver to Hepatocellular Carcinoma: A Story of (Mal)Adapted Mitochondria. Biology (Basel). 2023;12:595. [DOI] [PubMed] [PMC]
    Di Ciaula A, Passarella S, Shanmugam H, Noviello M, Bonfrate L, Wang DQ, et al. Nonalcoholic Fatty Liver Disease (NAFLD). Mitochondria as Players and Targets of Therapies? Int J Mol Sci. 2021;22:5375. [DOI] [PubMed] [PMC]
    Torres S, Segalés P, Conde de la Rosa L, Garcia-Ruiz C, Fernandez-checa JC. Mitochondria and Alcohol. In: Mueller S, Heilig M, editors. Alcohol and Alcohol-related Diseases. 1st ed. Cham: Springer; 2023. pp. 1043–74. [DOI]
    Hoek JB, Cahill A, Pastorino JG. Alcohol and mitochondria: a dysfunctional relationship. Gastroenterology. 2002;122:204963. [DOI] [PubMed] [PMC]
    Paradies G, Paradies V, Ruggiero FM, Petrosillo G. Oxidative stress, cardiolipin and mitochondrial dysfunction in nonalcoholic fatty liver disease. World J Gastroenterol. 2014;20:1420518. [DOI] [PubMed] [PMC]
    Cederbaum AI, Lu Y, Wu D. Role of oxidative stress in alcohol-induced liver injury. Arch Toxicol. 2009;83:51948. [DOI] [PubMed]
    Yu JW, Lee MS. Mitochondria and the NLRP3 inflammasome: physiological and pathological relevance. Arch Pharm Res. 2016;39:150318. [DOI] [PubMed]
    Ma X, McKeen T, Zhang J, Ding W. Role and Mechanisms of Mitophagy in Liver Diseases. Cells. 2020;9:837. [DOI] [PubMed] [PMC]
    Marí M, Morales A, Colell A, García-Ruiz C, Fernández-Checa JC. Mitochondrial cholesterol accumulation in alcoholic liver disease: Role of ASMase and endoplasmic reticulum stress. Redox Biol. 2014;3:1008. [DOI] [PubMed] [PMC]
    Colell A, García-Ruiz C, Morales A, Ballesta A, Ookhtens M, Rodés J, et al. Transport of reduced glutathione in hepatic mitochondria and mitoplasts from ethanol-treated rats: effect of membrane physical properties and S-adenosyl-L-methionine. Hepatology. 1997;26:699708. [DOI] [PubMed]
    Koh EH, Yoon JE, Ko MS, Leem J, Yun JY, Hong CH, et al. Sphingomyelin synthase 1 mediates hepatocyte pyroptosis to trigger non-alcoholic steatohepatitis. Gut. 2021;70:195464. [DOI] [PubMed] [PMC]
    Piccinin E, Peres C, Bellafante E, Ducheix S, Pinto C, Villani G, et al. Hepatic peroxisome proliferator-activated receptor γ coactivator 1β drives mitochondrial and anabolic signatures that contribute to hepatocellular carcinoma progression in mice. Hepatology. 2018;67:88498. [DOI] [PubMed]
    Petrosillo G, Portincasa P, Grattagliano I, Casanova G, Matera M, Ruggiero FM, et al. Mitochondrial dysfunction in rat with nonalcoholic fatty liver Involvement of complex I, reactive oxygen species and cardiolipin. Biochim Biophys Acta. 2007;1767:12607. [DOI] [PubMed]
    Montero J, Mari M, Colell A, Morales A, Basañez G, Garcia-Ruiz C, et al. Cholesterol and peroxidized cardiolipin in mitochondrial membrane properties, permeabilization and cell death. Biochim Biophys Acta. 2010;1797:121724. [DOI] [PubMed] [PMC]
    Marí M, Fernández-Checa JC. Sphingolipid signalling and liver diseases. Liver Int. 2007;27:44050. [DOI] [PubMed]
    Jamil M, Cowart LA. Sphingolipids in mitochondria-from function to disease. Front Cell Dev Biol. 2023;11:1302472. [DOI] [PubMed] [PMC]
    Garcia-Ruiz C, Conde de la Rosa L, Ribas V, Fernandez-Checa JC. Mitochondrial cholesterol and cancer. Semin Cancer Biol. 2021;73:7685. [DOI] [PubMed] [PMC]
    Solsona-Vilarrasa E, Fucho R, Torres S, Nuñez S, Nuño-Lámbarri N, Enrich C, et al. Cholesterol enrichment in liver mitochondria impairs oxidative phosphorylation and disrupts the assembly of respiratory supercomplexes. Redox Biol. 2019;24:101214. [DOI] [PubMed] [PMC]
    Garcia-Ruiz C, Mari M, Colell A, Morales A, Caballero F, Montero J, et al. Mitochondrial cholesterol in health and disease. Histol Histopathol. 2009;24:11732. [DOI] [PubMed]
    Fernandez A, Matias N, Fucho R, Ribas V, Von Montfort C, Nuño N, et al. ASMase is required for chronic alcohol induced hepatic endoplasmic reticulum stress and mitochondrial cholesterol loading. J Hepatol. 2013;59:80513. [DOI] [PubMed] [PMC]
    Caballero F, Fernández A, De Lacy AM, Fernández-Checa JC, Caballería J, García-Ruiz C. Enhanced free cholesterol, SREBP-2 and StAR expression in human NASH. J Hepatol. 2009;50:78996. [DOI] [PubMed]
    Conde de la Rosa L, Garcia-Ruiz C, Vallejo C, Baulies A, Nuñez S, Monte MJ, et al. STARD1 promotes NASH-driven HCC by sustaining the generation of bile acids through the alternative mitochondrial pathway. J Hepatol. 2021;74:142941. [DOI] [PubMed] [PMC]
    Pandak WM, Ren S, Marques D, Hall E, Redford K, Mallonee D, et al. Transport of cholesterol into mitochondria is rate-limiting for bile acid synthesis via the alternative pathway in primary rat hepatocytes. J Biol Chem. 2002;277:4815864. [DOI] [PubMed]
    Benador IY, Veliova M, Liesa M, Shirihai OS. Mitochondria Bound to Lipid Droplets: Where Mitochondrial Dynamics Regulate Lipid Storage and Utilization. Cell Metab. 2019;29:82735. [DOI] [PubMed] [PMC]
    Rolo AP, Teodoro JS, Palmeira CM. Role of oxidative stress in the pathogenesis of nonalcoholic steatohepatitis. Free Radic Biol Med. 2012;52:5969. [DOI] [PubMed]
    Serviddio G, Bellanti F, Vendemiale G. Free radical biology for medicine: learning from nonalcoholic fatty liver disease. Free Radic Biol Med. 2013;65:95268. [DOI] [PubMed]
    Parthasarathy G, Revelo X, Malhi H. Pathogenesis of Nonalcoholic Steatohepatitis: An Overview. Hepatol Commun. 2020;4:47892. [DOI] [PubMed] [PMC]
    Chalasani N, Younossi Z, Lavine JE, Charlton M, Cusi K, Rinella M, et al. The diagnosis and management of nonalcoholic fatty liver disease: Practice guidance from the American Association for the Study of Liver Diseases. Hepatology. 2018;67:32857. [DOI] [PubMed]
    Tilg H, Moschen AR. Evolution of inflammation in nonalcoholic fatty liver disease: the multiple parallel hits hypothesis. Hepatology. 2010;52:183646. [DOI] [PubMed]
    Younossi ZM, Golabi P, de Avila L, Paik JM, Srishord M, Fukui N, et al. The global epidemiology of NAFLD and NASH in patients with type 2 diabetes: A systematic review and meta-analysis. J Hepatol. 2019;71:793801. [DOI] [PubMed]
    Younossi ZM, Rinella ME, Sanyal AJ, Harrison SA, Brunt EM, Goodman Z, et al. From NAFLD to MAFLD: Implications of a Premature Change in Terminology. Hepatology. 2021;73:11948. [DOI] [PubMed]
    Eslam M, Newsome PN, Sarin SK, Anstee QM, Targher G, Romero-Gomez M, et al. A new definition for metabolic dysfunction-associated fatty liver disease: An international expert consensus statement. J Hepatol. 2020;73:2029. [DOI] [PubMed]
    Eccleston HB, Andringa KK, Betancourt AM, King AL, Mantena SK, Swain TM, et al. 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;15:44759. [DOI] [PubMed] [PMC]
    Win S, Than TA, Kaplowitz N, Wong N, Arya A, Win ZT, et al. The central role of mitochondrial metabolism in hepatic steatosis. Explor Dig Dis. 2024;3:4268.
    Peng KY, Watt MJ, Rensen S, Greve JW, Huynh K, Jayawardana KS, et al. Mitochondrial dysfunction-related lipid changes occur in nonalcoholic fatty liver disease progression. J Lipid Res. 2018;59:197786. [DOI] [PubMed] [PMC]
    Koliaki C, Szendroedi J, Kaul K, Jelenik T, Nowotny P, Jankowiak F, et al. Adaptation of hepatic mitochondrial function in humans with non-alcoholic fatty liver is lost in steatohepatitis. Cell Metab. 2015;21:73946. [DOI] [PubMed]
    Li J, Zou B, Yeo YH, Feng Y, Xie X, Lee DH, et al. Prevalence, incidence, and outcome of non-alcoholic fatty liver disease in Asia, 1999-2019: a systematic review and meta-analysis. Lancet Gastroenterol Hepatol. 2019;4:38998. [DOI] [PubMed]
    González-Rodríguez A, Mayoral R, Agra N, Valdecantos MP, Pardo V, Miquilena-Colina ME, et al. Impaired autophagic flux is associated with increased endoplasmic reticulum stress during the development of NAFLD. Cell Death Dis. 2014;5:e1179. [DOI] [PubMed] [PMC]
    Zhou T, Chang L, Luo Y, Zhou Y, Zhang J. Mst1 inhibition attenuates non-alcoholic fatty liver disease via reversing Parkin-related mitophagy. Redox Biol. 2019;21:101120. [DOI] [PubMed] [PMC]
    Yamada T, Murata D, Kleiner DE, Anders R, Rosenberg AZ, Kaplan J, et al. Prevention and regression of megamitochondria and steatosis by blocking mitochondrial fusion in the liver. iScience. 2022;25:103996. [DOI] [PubMed] [PMC]
    Chen Z, Tian R, She Z, Cai J, Li H. Role of oxidative stress in the pathogenesis of nonalcoholic fatty liver disease. Free Radic Biol Med. 2020;152:11641. [DOI] [PubMed]
    Elsheikh A, Lavergne SN, Castrejon JL, Farrell J, Wang H, Sathish J, et al. Drug antigenicity, immunogenicity, and costimulatory signaling: evidence for formation of a functional antigen through immune cell metabolism. J Immunol. 2010;185:644860. [DOI] [PubMed] [PMC]
    Marques PE, Oliveira AG, Pereira RV, David BA, Gomides LF, Saraiva AM, et al. Hepatic DNA deposition drives drug-induced liver injury and inflammation in mice. Hepatology. 2015;61:34860. [DOI] [PubMed]
    Lee J, Park JS, Roh YS. Molecular insights into the role of mitochondria in non-alcoholic fatty liver disease. Arch Pharm Res. 2019;42:93546. [DOI] [PubMed]
    Garcia-Martinez I, Santoro N, Chen Y, Hoque R, Ouyang X, Caprio S, et al. Hepatocyte mitochondrial DNA drives nonalcoholic steatohepatitis by activation of TLR9. J Clin Invest. 2016;126:85964. [DOI] [PubMed] [PMC]
    Inzaugarat ME, Wree A, Feldstein AE. Hepatocyte mitochondrial DNA released in microparticles and toll-like receptor 9 activation: A link between lipotoxicity and inflammation during nonalcoholic steatohepatitis. Hepatology. 2016;64:66971. [DOI] [PubMed] [PMC]
    Gariani K, Menzies KJ, Ryu D, Wegner CJ, Wang X, Ropelle ER, et al. Eliciting the mitochondrial unfolded protein response by nicotinamide adenine dinucleotide repletion reverses fatty liver disease in mice. Hepatology. 2016;63:1190204. [DOI] [PubMed] [PMC]
    Bellanti F, Mitarotonda D, Tamborra R, Blonda M, Iannelli G, Petrella A, et al. Oxysterols induce mitochondrial impairment and hepatocellular toxicity in non-alcoholic fatty liver disease. Free Radic Biol Med. 2014;75:S167. [DOI] [PubMed]
    Win S, Than TA, Le BH, García-Ruiz C, Fernandez-Checa JC, Kaplowitz N. Sab (Sh3bp5) dependence of JNK mediated inhibition of mitochondrial respiration in palmitic acid induced hepatocyte lipotoxicity. J Hepatol. 2015;62:136774. [DOI] [PubMed] [PMC]
    Chavin KD, Yang S, Lin HZ, Chatham J, Chacko VP, Hoek JB, et al. Obesity induces expression of uncoupling protein-2 in hepatocytes and promotes liver ATP depletion. J Biol Chem. 1999;274:5692700. [DOI] [PubMed]
    Marí M, Caballero F, Colell A, Morales A, Caballeria J, Fernandez A, et al. Mitochondrial free cholesterol loading sensitizes to TNF- and Fas-mediated steatohepatitis. Cell Metab. 2006;4:18598. [DOI] [PubMed]
    Zhong Z, Umemura A, Sanchez-Lopez E, Liang S, Shalapour S, Wong J, et al. NF-κB Restricts Inflammasome Activation via Elimination of Damaged Mitochondria. Cell. 2016;164:896910. [DOI] [PubMed] [PMC]
    Guo H, Callaway JB, Ting JP. Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat Med. 2015;21:67787. [DOI] [PubMed] [PMC]
    Kelley N, Jeltema D, Duan Y, He Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int J Mol Sci. 2019;20:3328. [DOI] [PubMed] [PMC]
    Teratani T, Tomita K, Suzuki T, Oshikawa T, Yokoyama H, Shimamura K, et al. A high-cholesterol diet exacerbates liver fibrosis in mice via accumulation of free cholesterol in hepatic stellate cells. Gastroenterology. 2012;142:15264.e10. [DOI] [PubMed]
    Tomita K, Teratani T, Suzuki T, Shimizu M, Sato H, Narimatsu K, et al. Free cholesterol accumulation in hepatic stellate cells: mechanism of liver fibrosis aggravation in nonalcoholic steatohepatitis in mice. Hepatology. 2014;59:15469. [DOI] [PubMed]
    Ribas V, de la Rosa LC, Robles D, Núñez S, Segalés P, Insausti-Urkia N, et al. Dietary and Genetic Cholesterol Loading Rather Than Steatosis Promotes Liver Tumorigenesis and NASH-Driven HCC. Cancers (Basel). 2021;13:4091. [DOI] [PubMed] [PMC]
    Ioannou GN. The Role of Cholesterol in the Pathogenesis of NASH. Trends Endocrinol Metab. 2016;27:8495. [DOI] [PubMed]
    Coll O, Colell A, García-Ruiz C, Kaplowitz N, Fernández-Checa JC. Sensitivity of the 2-oxoglutarate carrier to alcohol intake contributes to mitochondrial glutathione depletion. Hepatology. 2003;38:692702. [DOI] [PubMed]
    Ribas V, García-Ruiz C, Fernández-Checa JC. Glutathione and mitochondria. Front Pharmacol. 2014;5:151. [DOI] [PubMed] [PMC]
    Caballero F, Fernández A, Matías N, Martínez L, Fucho R, Elena M, et al. Specific contribution of methionine and choline in nutritional nonalcoholic steatohepatitis: impact on mitochondrial S-adenosyl-L-methionine and glutathione. J Biol Chem. 2010;285:1852836. [DOI] [PubMed] [PMC]
    Horn CL, Morales AL, Savard C, Farrell GC, Ioannou GN. Role of Cholesterol-Associated Steatohepatitis in the Development of NASH. Hepatol Commun. 2022;6:1235. [DOI] [PubMed] [PMC]
    Serviddio G, Bellanti F, Tamborra R, Rollo T, Capitanio N, Romano AD, et al. Uncoupling protein-2 (UCP2) induces mitochondrial proton leak and increases susceptibility of non-alcoholic steatohepatitis (NASH) liver to ischaemia-reperfusion injury. Gut. 2008;57:95765. [DOI] [PubMed]
    Gan LT, Van Rooyen DM, Koina ME, McCuskey RS, Teoh NC, Farrell GC. Hepatocyte free cholesterol lipotoxicity results from JNK1-mediated mitochondrial injury and is HMGB1 and TLR4-dependent. J Hepatol. 2014;61:137684. [DOI] [PubMed]
    Sanyal AJ, Campbell-Sargent C, Mirshahi F, Rizzo WB, Contos MJ, Sterling RK, et al. Nonalcoholic steatohepatitis: association of insulin resistance and mitochondrial abnormalities. Gastroenterology. 2001;120:118392. [DOI] [PubMed]
    von Montfort C, Matias N, Fernandez A, Fucho R, Conde de la Rosa L, Martinez-Chantar ML, et al. Mitochondrial GSH determines the toxic or therapeutic potential of superoxide scavenging in steatohepatitis. J Hepatol. 2012;57:8529. [DOI] [PubMed] [PMC]
    Vallejo C, Robles D, Baulies A, Fucho R, Fernandez-Checa JC, Garcia-Ruiz C. Ganglioside GD3 acetylation contributes to hepatic stellate cell activation. Hepatology. 2016;2:840A.
    Bruix J, Chan SL, Galle PR, Rimassa L, Sangro B. Systemic treatment of hepatocellular carcinoma: An EASL position paper. J Hepatol. 2021;75:96074. [DOI] [PubMed]
    Singal AG, Kudo M, Bruix J. Breakthroughs in Hepatocellular Carcinoma Therapies. Clin Gastroenterol Hepatol. 2023;21:213549. [DOI] [PubMed] [PMC]
    Ande SR, Nguyen KH, Grégoire Nyomba BL, Mishra S. Prohibitin-induced, obesity-associated insulin resistance and accompanying low-grade inflammation causes NASH and HCC. Sci Rep. 2016;6:23608. [DOI] [PubMed] [PMC]
    Shalapour S, Karin M. Fatty Acid-Induced T Cell Loss Greases Liver Carcinogenesis. Cell Metab. 2016;23:75961. [DOI] [PubMed]
    Ali ES, Rychkov GY, Barritt GJ. Deranged hepatocyte intracellular Ca2+ homeostasis and the progression of non-alcoholic fatty liver disease to hepatocellular carcinoma. Cell Calcium. 2019;82:102057. [DOI] [PubMed]
    Xu D, Xu M, Jeong S, Qian Y, Wu H, Xia Q, et al. The Role of Nrf2 in Liver Disease: Novel Molecular Mechanisms and Therapeutic Approaches. Front Pharmacol. 2019;9:1428. [DOI] [PubMed] [PMC]
    Teng Y, Zhao H, Gao L, Zhang W, Shull AY, Shay C. FGF19 Protects Hepatocellular Carcinoma Cells against Endoplasmic Reticulum Stress via Activation of FGFR4-GSK3β-Nrf2 Signaling. Cancer Res. 2017;77:621525. [DOI] [PubMed]
    Feo F, Canuto RA, Garcea R, Gabriel L. Effect of cholesterol content on some physical and functional properties of mitochondria isolated from adult rat liver, fetal liver, cholesterol-enriched liver and hepatomas AH-130, 3924A and 5123. Biochim Biophys Acta. 1975;413:11634. [DOI] [PubMed]
    Crain RC, Clark RW, Harvey BE. Role of lipid transfer proteins in the abnormal lipid content of Morris hepatoma mitochondria and microsomes. Cancer Res. 1983;43:3197202. [PubMed]
    Montero J, Morales A, Llacuna L, Lluis JM, Terrones O, Basañez G, et al. Mitochondrial cholesterol contributes to chemotherapy resistance in hepatocellular carcinoma. Cancer Res. 2008;68:524656. [DOI] [PubMed]
    Simula L, Nazio F, Campello S. The mitochondrial dynamics in cancer and immune-surveillance. Semin Cancer Biol. 2017;47:2942. [DOI] [PubMed]
    Sun X, Cao H, Zhan L, Yin C, Wang G, Liang P, et al. Mitochondrial fission promotes cell migration by Ca2+/CaMKII/ERK/FAK pathway in hepatocellular carcinoma. Liver Int. 2018;38:126372. [DOI] [PubMed]
    Zhang Z, Li TE, Chen M, Xu D, Zhu Y, Hu BY, et al. MFN1-dependent alteration of mitochondrial dynamics drives hepatocellular carcinoma metastasis by glucose metabolic reprogramming. Br J Cancer. 2020;122:20920. [DOI] [PubMed] [PMC]
    Calvisi DF, Wang C, Ho C, Ladu S, Lee SA, Mattu S, et al. Increased lipogenesis, induced by AKT-mTORC1-RPS6 signaling, promotes development of human hepatocellular carcinoma. Gastroenterology. 2011;140:107183. [DOI] [PubMed] [PMC]
    Ribas V. Role of cholesterol homeostasis in MASH-driven hepatocellular carcinoma: not just a neutral fat. Explor Dig Dis. 2024;3:20325. [DOI]
    Torres S, Solsona-Vilarrasa E, Nuñez S, Matías N, Insausti-Urkia N, Castro F, et al. Acid ceramidase improves mitochondrial function and oxidative stress in Niemann-Pick type C disease by repressing STARD1 expression and mitochondrial cholesterol accumulation. Redox Biol. 2021;45:102052. [DOI] [PubMed] [PMC]
    Colell A, García-Ruiz C, Lluis JM, Coll O, Mari M, Fernández-Checa JC. Cholesterol impairs the adenine nucleotide translocator-mediated mitochondrial permeability transition through altered membrane fluidity. J Biol Chem. 2003;278:3392835. [DOI] [PubMed]
    Bosch M, Marí M, Herms A, Fernández A, Fajardo A, Kassan A, et al. Caveolin-1 deficiency causes cholesterol-dependent mitochondrial dysfunction and apoptotic susceptibility. Curr Biol. 2011;21:6816. [DOI] [PubMed] [PMC]
    Baulies A, Montero J, Matías N, Insausti N, Terrones O, Basañez G, et al. The 2-oxoglutarate carrier promotes liver cancer by sustaining mitochondrial GSH despite cholesterol loading. Redox Biol. 2018;14:16477. [DOI] [PubMed] [PMC]
    Lee JS, Lee H, Lee S, Kang JH, Lee SH, Kim SG, et al. Loss of SLC25A11 causes suppression of NSCLC and melanoma tumor formation. EBioMedicine. 2019;40:18497. [DOI] [PubMed] [PMC]
    Colell A, García-Ruiz C, Miranda M, Ardite E, Marí M, Morales A, et al. Selective glutathione depletion of mitochondria by ethanol sensitizes hepatocytes to tumor necrosis factor. Gastroenterology. 1998;115:154151. [DOI] [PubMed]
    Yue X, Kong Y, Zhang Y, Sun M, Liu S, Wu Z, et al. SREBF2-STARD4 axis confers sorafenib resistance in hepatocellular carcinoma by regulating mitochondrial cholesterol homeostasis. Cancer Sci. 2023;114:47789. [DOI] [PubMed] [PMC]
    Paradies G, Petrosillo G, Pistolese M, Ruggiero FM. The effect of reactive oxygen species generated from the mitochondrial electron transport chain on the cytochrome c oxidase activity and on the cardiolipin content in bovine heart submitochondrial particles. FEBS Lett. 2000;466:3236. [DOI] [PubMed]
    Paradies G, Petrosillo G, Pistolese M, Ruggiero FM. Reactive oxygen species affect mitochondrial electron transport complex I activity through oxidative cardiolipin damage. Gene. 2002;286:13541. [DOI] [PubMed]
    Paradies G, Petrosillo G, Pistolese M, Ruggiero FM. Reactive oxygen species generated by the mitochondrial respiratory chain affect the complex III activity via cardiolipin peroxidation in beef-heart submitochondrial particles. Mitochondrion. 2001;1:1519. [DOI] [PubMed]
    Li J, Romestaing C, Han X, Li Y, Hao X, Wu Y, et al. Cardiolipin remodeling by ALCAT1 links oxidative stress and mitochondrial dysfunction to obesity. Cell Metab. 2010;12:15465. [DOI] [PubMed] [PMC]
    Marí M, Colell A, Morales A, Caballero F, Moles A, Fernández A, et al. Mechanism of mitochondrial glutathione-dependent hepatocellular susceptibility to TNF despite NF-kappaB activation. Gastroenterology. 2008;134:150720. [DOI] [PubMed]
    Liu D, Wong CC, Fu L, Chen H, Zhao L, Li C, et al. Squalene epoxidase drives NAFLD-induced hepatocellular carcinoma and is a pharmaceutical target. Sci Transl Med. 2018;10:eaap9840. [DOI] [PubMed]
    Bakiri L, Hamacher R, Graña O, Guío-Carrión A, Campos-Olivas R, Martinez L, et al. Liver carcinogenesis by FOS-dependent inflammation and cholesterol dysregulation. J Exp Med. 2017;214:1387409. [DOI] [PubMed] [PMC]
    Liang JQ, Teoh N, Xu L, Pok S, Li X, Chu ESH, et al. Dietary cholesterol promotes steatohepatitis related hepatocellular carcinoma through dysregulated metabolism and calcium signaling. Nat Commun. 2018;9:4490. [DOI] [PubMed] [PMC]
    Ambade A, Mandrekar P. Oxidative stress and inflammation: essential partners in alcoholic liver disease. Int J Hepatol. 2012;2012:853175. [DOI] [PubMed] [PMC]
    Fucho R, Solsona-Vilarrasa E, Torres S, Nuñez S, Insausti-Urkia N, Edo A, et al. Zonal expression of StARD1 and oxidative stress in alcoholic-related liver disease. J Lipid Res. 2023;64:100413. [DOI] [PubMed] [PMC]
    Fernandez-Checa JC, Kaplowitz N. Hepatic mitochondrial glutathione: transport and role in disease and toxicity. Toxicol Appl Pharmacol. 2005;204:26373. [DOI] [PubMed]
    Giacomello M, Pyakurel A, Glytsou C, Scorrano L. The cell biology of mitochondrial membrane dynamics. Nat Rev Mol Cell Biol. 2020;21:20424. [DOI] [PubMed]
    Gao S, Hu J. Mitochondrial Fusion: The Machineries In and Out. Trends Cell Biol. 2021;31:6274. [DOI] [PubMed]
    Palma E, Riva A, Moreno C, Odena G, Mudan S, Manyakin N, et al. Perturbations in Mitochondrial Dynamics Are Closely Involved in the Progression of Alcoholic Liver Disease. Alcohol Clin Exp Res. 2020;44:85665. [DOI] [PubMed] [PMC]
    Palma E, Ma X, Riva A, Iansante V, Dhawan A, Wang S, et al. Dynamin-1-Like Protein Inhibition Drives Megamitochondria Formation as an Adaptive Response in Alcohol-Induced Hepatotoxicity. Am J Pathol. 2019;189:5809. [DOI] [PubMed] [PMC]
    Bruguera M, Rodes J, Bordas JM. Histological diagnosis of alcoholic hepatitis. Clinical meaning and prognosis of Mallory’s hyaline bodies. Rev Clin Esp. 1975;136:1317. Spanish. [PubMed]
    Galloway CA, Lee H, Brookes PS, Yoon Y. Decreasing mitochondrial fission alleviates hepatic steatosis in a murine model of nonalcoholic fatty liver disease. Am J Physiol Gastrointest Liver Physiol. 2014;307:G63241. [DOI] [PubMed] [PMC]
    Altamirano J, Miquel R, Katoonizadeh A, Abraldes JG, Duarte-Rojo A, Louvet A, et al. A histologic scoring system for prognosis of patients with alcoholic hepatitis. Gastroenterology. 2014;146:12319.e6. [DOI] [PubMed] [PMC]
    Andrade P, Silva M, Rodrigues S, Lopes J, Lopes S, Macedo G. Alcoholic hepatitis histological score has high accuracy to predict 90-day mortality and response to steroids. Dig Liver Dis. 2016;48:65660. [DOI] [PubMed]
    Kim W, Choi YI, Joo SK, Jung YJ. Alcoholic hepatitis histological scores predict short-term survival in Asian patients with biopsy-proven alcoholic hepatitis. J Hepatol. 2017;66:S118. [DOI]
    Chen H, Chan DC. Physiological functions of mitochondrial fusion. Ann N Y Acad Sci. 2010;1201:215. [DOI] [PubMed]
    Kageyama Y, Hoshijima M, Seo K, Bedja D, Sysa-Shah P, Andrabi SA, et al. Parkin-independent mitophagy requires Drp1 and maintains the integrity of mammalian heart and brain. EMBO J. 2014;33:2798813. [DOI] [PubMed] [PMC]
    Yamada T, Murata D, Adachi Y, Itoh K, Kameoka S, Igarashi A, et al. Mitochondrial Stasis Reveals p62-Mediated Ubiquitination in Parkin-Independent Mitophagy and Mitigates Nonalcoholic Fatty Liver Disease. Cell Metab. 2018;28:588604.e5. [DOI] [PubMed] [PMC]
    Ma X, Chen A, Melo L, Clemente-Sanchez A, Chao X, Ahmadi AR, et al. Loss of hepatic DRP1 exacerbates alcoholic hepatitis by inducing megamitochondria and mitochondrial maladaptation. Hepatology. 2023;77:15975. [DOI] [PubMed] [PMC]
    DiMauro S, Schon EA. Mitochondrial DNA mutations in human disease. Am J Med Genet. 2001;106:1826. [DOI] [PubMed]
    Mansouri A, Gaou I, De Kerguenec C, Amsellem S, Haouzi D, Berson A, et al. An alcoholic binge causes massive degradation of hepatic mitochondrial DNA in mice. Gastroenterology. 1999;117:18190. [DOI] [PubMed]
    Raha S, Robinson BH. Mitochondria, oxygen free radicals, disease and ageing. Trends Biochem Sci. 2000;25:5028. [DOI] [PubMed]
    Halliwell B, Aruoma OI. DNA damage by oxygen-derived species. Its mechanism and measurement in mammalian systems. FEBS Lett. 1991;281:919. [DOI] [PubMed]
    Cahill A, Stabley GJ, Wang X, Hoek JB. Chronic ethanol consumption causes alterations in the structural integrity of mitochondrial DNA in aged rats. Hepatology. 1999;30:8818. [DOI] [PubMed] [PMC]
    Sadikot RT, Bedi B, Li J, Yeligar SM. Alcohol-induced mitochondrial DNA damage promotes injurious crosstalk between alveolar epithelial cells and alveolar macrophages. Alcohol. 2019;80:6572. [DOI] [PubMed] [PMC]
    Venkatraman A, Landar A, Davis AJ, Chamlee L, Sanderson T, Kim H, et al. Modification of the mitochondrial proteome in response to the stress of ethanol-dependent hepatotoxicity. J Biol Chem. 2004;279:22092101. [DOI] [PubMed]
    Motwani M, Pesiridis S, Fitzgerald KA. DNA sensing by the cGAS-STING pathway in health and disease. Nat Rev Genet. 2019;20:65774. [DOI] [PubMed]
    Ablasser A, Goldeck M, Cavlar T, Deimling T, Witte G, Röhl I, et al. cGAS produces a 2’-5’-linked cyclic dinucleotide second messenger that activates STING. Nature. 2013;498:3804. [DOI] [PubMed] [PMC]
    Luther J, Khan S, Gala MK, Kedrin D, Sridharan G, Goodman RP, et al. Hepatic gap junctions amplify alcohol liver injury by propagating cGAS-mediated IRF3 activation. Proc Natl Acad Sci U S A. 2020;117:1166773. [DOI] [PubMed] [PMC]
    García-Ruiz C, Kaplowitz N, Fernandez-Checa JC. Role of Mitochondria in Alcoholic Liver Disease. Curr Pathobiol Rep. 2013;1:15968. [DOI] [PubMed] [PMC]
    Witschi A, Mossi S, Meyer B, Junker E, Lauterburg BH. Mitochondrial function reflected by the decarboxylation of [13C]ketoisocaproate is impaired in alcoholics. Alcohol Clin Exp Res. 1994;18:9515. [DOI] [PubMed]
    Caldwell SH, Swerdlow RH, Khan EM, Iezzoni JC, Hespenheide EE, Parks JK, et al. Mitochondrial abnormalities in non-alcoholic steatohepatitis. J Hepatol. 1999;31:4304. [DOI] [PubMed]
    Fromenty B, Grimbert S, Mansouri A, Beaugrand M, Erlinger S, Rötig A, et al. Hepatic mitochondrial DNA deletion in alcoholics: association with microvesicular steatosis. Gastroenterology. 1995;108:193200. [DOI] [PubMed]
    Knockaert L, Fromenty B, Robin M. Mechanisms of mitochondrial targeting of cytochrome P450 2E1: physiopathological role in liver injury and obesity. FEBS J. 2011;278:425260. [DOI] [PubMed]
    Bai J, Cederbaum AI. Overexpression of CYP2E1 in mitochondria sensitizes HepG2 cells to the toxicity caused by depletion of glutathione. J Biol Chem. 2006;281:512836. [DOI] [PubMed]
    Marí M, Morales A, Colell A, García-Ruiz C, Kaplowitz N, Fernández-Checa JC. Mitochondrial glutathione: features, regulation and role in disease. Biochim Biophys Acta. 2013;1830:331728. [DOI] [PubMed] [PMC]
    Zhang H, Go YM, Jones DP. Mitochondrial thioredoxin-2/peroxiredoxin-3 system functions in parallel with mitochondrial GSH system in protection against oxidative stress. Arch Biochem Biophys. 2007;465:11926. [DOI] [PubMed]
    Yang F, Luo J. Endoplasmic Reticulum Stress and Ethanol Neurotoxicity. Biomolecules. 2015;5:253853. [DOI] [PubMed] [PMC]
    Cao SS, Kaufman RJ. Targeting endoplasmic reticulum stress in metabolic disease. Expert Opin Ther Targets. 2013;17:43748. [DOI] [PubMed]
    Yoshida H. ER stress and diseases. FEBS J. 2007;274:63058. [DOI] [PubMed]
    Wolff S, Weissman JS, Dillin A. Differential scales of protein quality control. Cell. 2014;157:5264. [DOI] [PubMed]
    Galligan JJ, Smathers RL, Shearn CT, Fritz KS, Backos DS, Jiang H, et al. Oxidative Stress and the ER Stress Response in a Murine Model for Early-Stage Alcoholic Liver Disease. J Toxicol. 2012;2012:207594. [DOI] [PubMed] [PMC]
    Ji C, Kaplowitz N. Betaine decreases hyperhomocysteinemia, endoplasmic reticulum stress, and liver injury in alcohol-fed mice. Gastroenterology. 2003;124:148899. [DOI] [PubMed]
    Barbier-Torres L, Murray B, Yang JW, Wang J, Matsuda M, Robinson A, et al. Depletion of mitochondrial methionine adenosyltransferase α1 triggers mitochondrial dysfunction in alcohol-associated liver disease. Nat Commun. 2022;13:557. [DOI] [PubMed] [PMC]
    Fernández A, Colell A, Caballero F, Matías N, García-Ruiz C, Fernández-Checa JC. Mitochondrial S-adenosyl-L-methionine transport is insensitive to alcohol-mediated changes in membrane dynamics. Alcohol Clin Exp Res. 2009;33:116980. [DOI] [PubMed]
    Maxfield FR, Tabas I. Role of cholesterol and lipid organization in disease. Nature. 2005;438:61221. [DOI] [PubMed]
    Knorr J, Wree A, Tacke F, Feldstein AE. The NLRP3 Inflammasome in Alcoholic and Nonalcoholic Steatohepatitis. Semin Liver Dis. 2020;40:298306. [DOI] [PubMed]
    Zhang Y, Dong Z, Song W. NLRP3 inflammasome as a novel therapeutic target for Alzheimer’s disease. Signal Transduct Target Ther. 2020;5:37. [DOI] [PubMed] [PMC]
    Schroder K, Tschopp J. The inflammasomes. Cell. 2010;140:82132. [DOI] [PubMed]
    Zhou R, Yazdi AS, Menu P, Tschopp J. A role for mitochondria in NLRP3 inflammasome activation. Nature. 2011;469:2215. [DOI] [PubMed]
    Iyer SS, He Q, Janczy JR, Elliott EI, Zhong Z, Olivier AK, et al. Mitochondrial cardiolipin is required for Nlrp3 inflammasome activation. Immunity. 2013;39:31123. [DOI] [PubMed] [PMC]
    Hayashi T, Rizzuto R, Hajnoczky G, Su TP. MAM: more than just a housekeeper. Trends Cell Biol. 2009;19:818. [DOI] [PubMed] [PMC]
    Pouliquen DL. Liver metastatic colonization by invasive cancer cells: a review of potential biomarkers with mitochondrial involvement. Explor Dig Dis. 2024;3:6985. [DOI]