Next Article in Journal
The Distinct Role of HIF-1α and HIF-2α in Hypoxia and Angiogenesis
Previous Article in Journal
Hydrogen Peroxide Modulates the Timely Activation of Jun and Erk in Schwann Cells at the Injury Site and Is Required for Motor Axon Regeneration
Previous Article in Special Issue
Natural Autophagy Activators to Fight Age-Related Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Iron Metabolism and Muscle Aging: Where Ferritinophagy Meets Mitochondrial Quality Control

by
Rosa Di Lorenzo
1,
Emanuele Marzetti
2,3,*,
Helio José Coelho-Junior
3,
Riccardo Calvani
2,3,
Vito Pesce
1,
Francesco Landi
2,3,
Christiaan Leeuwenburgh
4,* and
Anna Picca
3,5
1
Department of Biosciences, Biotechnologies, and Environment, Università degli Studi di Bari Aldo Moro, Via Edoardo Orabona 4, 70125 Bari, Italy
2
Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
3
Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
4
Department of Physiology and Aging, University of Florida, 2004 Mowry Road, Gainesville, FL 32611, USA
5
Department of Medicine and Surgery, LUM University, Str. Statale 100, 70010 Casamassima, Italy
*
Authors to whom correspondence should be addressed.
Cells 2025, 14(9), 672; https://doi.org/10.3390/cells14090672
Submission received: 4 April 2025 / Revised: 30 April 2025 / Accepted: 2 May 2025 / Published: 3 May 2025
(This article belongs to the Special Issue Autophagy Meets Aging 2025)

Abstract

:
In older adults with reduced physical performance, an increase in the labile iron pool within skeletal muscle is observed. This accumulation is associated with an altered expression of mitochondrial quality control (MQC) markers and increased mitochondrial DNA damage, supporting the hypothesis that impaired MQC contributes to muscle dysfunction during aging. The autophagy–lysosome system plays a critical role in MQC by tagging and engulfing proteins and organelles for degradation in lysosomes. The endolysosomal system is also instrumental in transferrin recycling, which, in turn, regulates cellular iron uptake. In the neuromuscular system, the autophagy–lysosome system supports the structural integrity of neuromuscular junctions, and its dysfunction contributes to muscle atrophy. While MQC was thought to protect against iron-induced cell death, the discovery of ferroptosis, a form of iron-dependent cell death, has highlighted a complex interplay between MQC and iron-inflicted damage. Ferritinophagy, the autophagic degradation of ferritin, if overactivated, can induce ferroptosis. Alternatively, aging may impair ferritinophagy, leading to ferritin accumulation and the release of toxic labile iron under stress, exacerbating oxidative damage and cellular senescence. Physical activity supports muscle health also by preserving mitochondrial quantity and quality and enhancing bioenergetics. However, therapeutic strategies for preventing or reversing physical function decline in aging are still lacking due to the insufficient understanding of the underlying mechanisms. Unveiling how disruptions in iron homeostasis impact muscle quality in older adults may allow for the development of therapeutic strategies targeting iron handling to alleviate age-associated muscle decline.

1. Introduction

Among the biological pathways contributing to muscle aging and the associated decline in physical performance, alterations in mitochondrial quality and function, alongside structural and functional muscle remodeling, play a critical role [1,2]. Recent studies have uncovered a link between defective iron handling in muscle, mitochondrial dysfunction, and systemic inflammation in older adults [3,4]. The labile iron pool has been found to be elevated in the muscles of older individuals with low physical performance, a phenomenon that has been associated with dysregulation in the expression of mitochondrial quality control (MQC) markers and an increase in mitochondrial DNA (mtDNA) damage [3,4]. These findings highlight the significance of inefficient MQC as a factor contributing to muscle dysfunction during aging [1].
The autophagy–lysosome system plays a central role in MQC. This system facilitates the tagging, engulfment, and degradation of proteins and organelles through autophagosomes, which subsequently deliver these materials to lysosomes for breakdown. Preclinical research has shown that inhibition of the autophagy–lysosome system results in dysfunctional neuromuscular junctions, muscle atrophy, and weakness [5]. In addition, the accumulation of lipofuscin, a nondegradable lysosomal aggregate composed of polymerized lipid and protein residues, has been documented in the muscles of both aged mice and humans. This accumulation may be an indicator of lysosomal dysfunction and compromised cellular quality control processes. Thus, alterations in autophagy and mitophagy are believed to contribute to muscle atrophy and the decline in physical function that characterizes aging [6].
MQC was initially believed to prevent cell death induced by iron overload. However, the discovery of ferroptosis, a form of iron-dependent cell death resulting from the lipid peroxidation of cellular membranes, has introduced a new perspective on the role of MQC in regulating iron-induced cell death [7]. An integral component of this process is ferritinophagy, the autophagic degradation and recycling of ferritin [8], which, if overactivated, can lead to ferroptosis [9].
Physical activity conveys several beneficial effects on muscle health, including the preservation of mitochondrial mass, promotion of mitochondrial dynamics and autophagy, and support of mitochondrial bioenergetics [10]. Nevertheless, a significant gap remains in our understanding of the underlying mechanisms driving the loss of physical function with aging, which hampers the development of effective therapeutic interventions. Given the rapidly growing population of older adults, identifying therapeutic targets to mitigate age-related muscle decline has become an urgent priority. In this context, understanding the mechanisms whereby iron dyshomeostasis contributes to the deterioration of muscle quality in older adults could have major implications for the development of therapeutic strategies targeting iron handling. Such interventions may offer new avenues to alleviate age-related muscle decline, thereby improving the health and quality of life of aging populations.

2. Iron, Mitochondria, and Aging

2.1. Iron and Mitochondria

Iron plays essential physiological roles, including its involvement in heme biosynthesis for oxygen transport by hemoglobin and muscle oxygenation by myoglobin, as well as its incorporation into iron–sulfur (Fe–S) clusters in cytochromes and cofactors of the mitochondrial electron transport chain complexes [11]. Iron homeostasis is tightly regulated at the molecular, cellular, and systemic levels. Indeed, low iron levels impair erythropoiesis, leading to anemia, while excess labile iron promotes reactive oxygen species (ROS) generation, which can damage cellular structures and organs [12].
Iron in the body exists in two distinct forms: heme iron (HI) and non-heme iron (NHI). HI is found at the center of a porphyrin ring in functional proteins such as hemoglobin, myoglobin, cytochromes, and Fe–S clusters [13,14] and is primarily derived from animal-based dietary sources such as meat and fish. In contrast, NHI, which constitutes approximately 30% of the bodily iron stores, is not associated with the porphyrin structure and is mainly found in plant-based foods [13,14]. Systemic iron uptake occurs in the duodenum, where enterocytes absorb dietary iron from the intestinal lumen. HI absorption is facilitated by the heme carrier protein 1 (HCP1), whereas NHI is taken up via the divalent metal transporter 1 (DMT1) [15] (Figure 1).
In addition, reticuloendothelial macrophages recycle iron from aged erythrocytes mostly in the spleen and release it into the bloodstream [16,17]. Here, iron is transported bound to transferrin (Tf), a glycoprotein synthesized by hepatocytes that possesses a high-affinity iron-binding site (Kd ≈ 10−20 M) [18,19,20,21]. The binding of iron to specific transporters is crucial for preventing oxidative damage to macromolecules, organelles, and cells.
Iron can exist in two states: the ferrous state (Fe2+), which is highly reactive and capable of reducing oxygen, and the ferric state (Fe3+). In conditions of iron overload, plasma Tf may become saturated, leading to an inability to fully bind free circulating iron [22]. In such circumstances, divalent metal transporters from the Zrt-Irt-like protein (ZIP) family intervene to absorb excess free iron [23]. The Fe3+–Tf complex enters cells through clathrin-mediated endocytosis upon Tf interaction with the transferrin receptor 1 (TfR1), a homodimeric glycoprotein. The binding of Fe3+–Tf to TfR1 is pH-dependent: at physiological pH (7.4), TfR1 binds Fe3+–Tf but not iron-free Tf, while the acidic environment of the endosome promotes the release of Fe3+, which is then reduced to Fe2+ by the ferrireductase six-transmembrane epithelial antigen of prostate 3 (STEAP3) and transferred to the cytoplasm via DMT1, coupled with H+ transport [24,25]. Cytoplasmic iron exists in a labile pool (~0.001 mM), typically bound to low-molecular weight molecules such as peptides, carboxylates, phosphates, or metallochaperone proteins [26,27]. The iron-free Tf–TfR1 complex is returned to the cell surface for recycling, enabling subsequent rounds of iron uptake (Figure 1).
The labile iron pool is regulated by two primary proteins: ferritin (Fer) and ferroportin (FPN). Ferritin, abundant in the bone marrow, spleen, and liver, is a multimeric protein composed of ferritin light (FTL) and heavy (FTH1) subunits that can sequester over 4000 Fe3+ ions in its core, utilizing the ferroxidase activity of its heavy chain to prevent oxidative damage [28,29]. The influx of labile iron into the cytoplasm induces the production of ferritin, highlighting the importance of storing and detoxifying excess iron to prevent cellular oxidative stress [30]. Iron is delivered to ferritin via poly (rC) binding protein 1 (PCBP1), which binds both RNA and iron at distinct sites [31] (Figure 1). Mice deficient in liver-specific PCBP1 exhibit oxidative stress, lipid peroxidation, and steatosis, highlighting the role of PCBP1 in mitigating cytoplasmic iron toxicity [32]. FPN, a secondary active transporter belonging to the major facilitator superfamily (MFS), is the only iron exporter responsible for transporting iron from cells into the circulation [33,34,35]. The levels of FPN are regulated by hepcidin, a defensin-like hormone that binds to FPN and induces its degradation [36,37]. Hepcidin expression is influenced by systemic iron levels and increases during iron overload while decreasing under conditions of anemia and hypoxia [38]. An increase in Tf also triggers the expression of hepcidin, which retains iron in macrophages and prevents iron absorption into enterocytes to avoid systemic iron overload [37,39] (Figure 1). Other molecular signals responsible for the transcriptional regulation of hepcidin are Tf saturation, erythropoietic activity, and inflammation [40]. Conversely, mutations in FPN are associated with hemochromatosis type 4, a disorder characterized by excessive iron accumulation [41]. Iron response element-binding proteins (IRP1 and IRP2) regulate the expression of ferritin and TfR1 at the transcriptional level by acting on the 3′ or 5′ untranslated region (UTR) of iron-responsive elements (IREs) [42,43]. IRP1 and IRP2 are homologous of the aconitase gene family and can sense cytosolic iron concentrations and respond by controlling the translation of Fer and FPN mRNA and the stability of TfR mRNA. Elevated iron levels reduce the affinity of IRP1 for IREs, leading to the conversion of IRP1 into its aconitase form. Conversely, when iron levels are low, IRP1 binds to the IRE with high affinity. IRP1 inhibits the translation of ferritin (Fer) mRNA when it binds to a single IRE located in the 5′ untranslated region (UTR) of the mRNA. On the other hand, when IRP1 binds to IREs in the 3′ UTR, it prevents mRNA cleavage and endonucleolytic degradation, thereby maintaining the transcription of TfR mRNA active [42]. Furthermore, a direct correlation has been observed in endothelial cells between the saturation of Tf-bound iron and FPN levels on the plasma membrane [44]. Once iron is transported outside the cell, Fe2+ is oxidized to Fe3+ by ceruloplasmin and/or hephaestin, mainly in enterocytes, and is subsequently bound to Tf for transport throughout the extracellular space [45].
Within cells, mitochondria are the main users and storage sites of iron. Herein, iron is incorporated into iron-containing proteins such as Fe–S cluster enzymes (e.g., electron transfer flavoproteins, NADH:ubiquinone oxidoreductase, succinate dehydrogenase subunits), heme-containing proteins (e.g., cytochrome bc1, cytochrome c, cytochrome c oxidase), and other iron-dependent enzymes [46] (Figure 1). While the abundance of iron-containing proteins in mitochondria highlights its critical role in mitochondrial function, the precise mechanisms by which labile iron reaches mitochondria from endosomes remain incompletely understood. One proposed model in reticulocytes suggests a “kiss-and-run” mechanism, where endosomes briefly interact with the outer mitochondrial membrane (OMM), facilitating direct iron transfer to mitochondria without the risk of generating reactive radicals in the cytosol [47]. In cardiac cells, iron is thought to enter mitochondria via fluid-phase endocytosis in a partially solvent-occluded manner [48]. In addition, studies in rat hepatocytes and yeast suggest that Fe2+, the primary form of iron in the cytosolic labile iron pool, can be imported into mitochondria driven by the mitochondrial membrane potential [49,50]. Iron must pass through both the OMM and the inner mitochondrial membrane (IMM) to reach the matrix, where most mitochondrial iron metabolism occurs. The import of Fe2+ into the intermembrane space may involve porins or a mitochondrial isoform of DMT1 (mDMT1) that has been identified in the OMM of HEK293 cells [51,52]. Subsequently, iron can be transported into the matrix through mitochondrial carrier family (MCF) proteins such as mitoferrin 1 (MFRN1), highly expressed in developing erythroid cells, and mitoferrin 2 (MFRN2), expressed ubiquitously, which are both located on the IMM [53,54]. Mitochondrial sideroflexins (SFXN), a family of transmembrane proteins [55], are also implicated in mitochondrial iron homeostasis [56], and their altered expression has been associated with various disorders, including mitochondriopathy or transient embryonic and neonatal sideroblastic anemia [46]. As heme synthesis occurs in mitochondria in close contact with the endoplasmic reticulum (ER), it is possible that heme is trafficked to the cytoplasm via the mitochondria–ER network, as shown in Saccharomyces cerevisiae [57]. In mammals, mitochondria–ER MCSs are referred to as mitochondria-associated membranes (MAMs). Mitochondria-derived vesicles (MDVs) may also play a role in heme trafficking [58]. MDVs can have either a single- or double-membrane structure, with a diameter ranging from 70 to 100 nm, and contain ligase proteins that are anchored to the mitochondria. These vesicles are specifically directed toward lysosomes and peroxisomes [59,60]. A key function of MDV-mediated heme “isolation” is to protect the cell from the harmful effects of free iron.
Iron storage within mitochondria is achieved through a specific mitochondrial ferritin (FtMt) (Figure 1), which is expressed in a tissue-specific manner in mammals (e.g., brain, in the medulla spinal cord, testes, heart, kidneys, and islets of Langerhans) [61,62]. Its main task is to sequester labile iron to prevent ROS formation [63]. Notably, FtMt overexpression compromises cellular iron homeostasis due to cytosolic iron depletion and mitochondrial iron overload [64].

2.2. Age-Related Mitochondrial and Iron Dysregulation

The role of iron in age-related oxidative damage and associated conditions has long been recognized, although the involvement of iron homeostasis in physiological aging warrants further exploration [65]. Both iron overload and deficiency have been observed in aging individuals and are associated with age-related disorders [66]. Iron accumulation in older adults has been observed particularly in liver diseases, kidney disorders, and Alzheimer’s disease [65,66]. Iron excess is particularly dangerous in senescent cells due to their inability to proliferate, which limits the redistribution and buffering of iron [67]. In these conditions, elevated levels of Fe3+–Fer and labile Fe2+ promote ROS-induced senescence [67]. On the other hand, age-associated iron deficiency has been linked to a reduced efficiency of dietary iron absorption and the sequestration of iron within macrophages, leading to decreased bioavailability for erythropoiesis [68].
In the skeletal muscle of aged rats, a high expression of Fer, increased iron transport via Tf, and a marked downregulation of TfR1 were observed, which were correlated with significant increases in NHI levels and oxidative damage to proteins and RNA [69,70,71]. NHI is a potent pro-oxidant metal that facilitates ROS generation, and its excess in muscle tissue has been associated with tissue atrophy [72,73]. NHI and total iron concentrations increase with age in skeletal muscle, plateauing in late middle age. While NHI continues to rise independent of Fer expression, a reduction in TfR1 has been linked to IRP2 downregulation [73].
Altered cellular iron transport and its accumulation within mitochondria have been associated with mitochondrial dysfunction [3,4,74,75]. Iron overload, mitochondrial dysfunction, and lipofuscin accumulation have been documented in liver cells [76]. The accumulation of lipofuscin with aging is observed in most postmitotic cells as non-disposable waste stuck in secondary lysosomes [77]. Lipofuscin has been identified as a store of redox-active iron, which can potentially lead to oxidative damage [77]. On the other hand, iron deficiency has been linked to reduced mitochondrial number and function in muscle cells.
Damaged mitochondria are cleared via mitophagy or proteasomal degradation; however, alternative mitochondrial recycling pathways via MDVs have been proposed [78,79,80]. During mitophagy, an autophagosomal membrane envelops the damaged mitochondrion, which is subsequently fused with a lysosome for degradation. Mitophagy can occur through two distinct pathways based on the dependence/independence on the PINK1/PARKIN axis [6]. The PINK1/PARKIN pathway is the best characterized route. Under basal conditions, PINK1 is cleaved and inactivated; however, mitochondrial stress or programmed mitophagy prevents PINK1 cleavage, leading to its accumulation on the mitochondrial surface, where it recruits PARKIN to ubiquitinate proteins for proteasomal degradation [6]. Other proteins, such as BCL2-interacting protein 3 (BNIP3), NIP-3-like protein X (NIX), and FUN14 domain-containing protein 1 (FUNDC1), facilitate PINK1/PARKIN-independent mitophagy, promoting mitophagosome formation. Various receptors, including BCL-2-like protein 13 (BCL2L13), FK506-binding protein 8 (FKPB8), Prohibitin 2 (PHB2), cardiolipins, autophagy and beclin 1 regulator 1 (AMBRA1), and neighbor of BRCA1 gene 1 protein (NBR1), also contribute to the execution of mitophagy [6]. In the context of iron deficiency in myotubes, mitochondria become surrounded by double-membrane structures, with mitochondrion–lysosomal co-localization and increased expression of mitophagy proteins [81].
Frataxin (FXN), a 17 kDa protein, plays a crucial role in mitochondrial iron homeostasis by acting as an iron chaperone during the synthesis of heme and Fe–S clusters. FXN also serves as an iron storage protein in the presence of excess metal, aids in the repair of oxidatively damaged Fe–S clusters in aconitase, and reduces ROS concentrations within mitochondria [82]. Indeed, the overexpression of FXN in Drosophila has been shown to counteract oxidative stress and extend lifespan [83]. Age-related iron imbalance impacts mitochondrial function, particularly in skeletal muscle, which serves as the body’s primary iron reservoir. Consistent with the findings in murine models, an increased expression of mitoferrin (MFRN) and reduced FXN levels have been observed in association with high labile Fe2+ concentrations in the skeletal muscle of older adults with low physical performance [4]. This suggests a potential dysregulation of mitochondrial iron handling, where MFRN upregulation may serve as a compensatory response to iron excess. However, FXN alone is insufficient to counteract iron overload, leaving free iron to compromise mitochondrial integrity via Fenton’s reaction [4]. Preclinical studies indicate that iron uptake by skeletal myocytes through TfR1 significantly decreases with aging, accompanied by the accumulation of intracellular labile iron [84]. Similar changes have been observed in the skeletal muscle of older adults, where TfR1 protein expression correlates with impaired physical function [3]. A potential mechanism underpinning these findings could involve TfR1 being diverted to a degradative pathway instead of being recycled back to the plasma membrane, leading to reduced receptor levels. Alternatively, TfR1 may be rerouted to the exosomal secretory pathway, a process that will be further discussed in the next section. Proper TfR1 recycling is critical for maintaining muscle physiology, as the inactivation of TfR1 impairs energy production in muscle cells, causing growth arrest and metabolic dysregulation, due to a failure to shift to fatty acid β-oxidation [85]. The link between muscle quality decline and disrupted TfR1 recycling warrants further investigation, and understanding the lysosomal degradation of TfR1 and its exosomal cargo could provide insight into iron mishandling and muscle function deterioration.

3. The Endolysosomal System and Ferritinophagy for the Regulation of Iron Metabolism: Converging Pathways?

Mitophagy plays a crucial role in maintaining cellular quality by selectively eliminating damaged mitochondria and regulating organelle numbers according to metabolic demands; however, excessive mitophagy can be detrimental to cells [86,87]. Excessive mitochondrial fission generates abundant mitochondrial debris, which activates mitophagy [88]. In contrast, when mitochondrial damage is mild, oxidized protein cargoes may be selectively incorporated into MDVs, which bud off from the mitochondria [59,60]. MDVs may thus serve as an alternative pathway to mitophagy and contribute to mitochondrial homeostasis [89]. These vesicles can follow one of two possible fates: (1) undergo endolysosomal degradation or (2) be secreted into the extracellular space as part of the many extracellular vesicles (EVs) [90,91,92,93]. The endosomal degradation pathway involves the internalization of molecules and fluids through membrane invaginations, which subsequently form vesicles or endosomes that fuse with lysosomes. Lysosomes contain hydrolytic enzymes that operate at acidic pH to degrade proteins, lipids, and carbohydrates. In contrast, EVs are lipid bilayer vesicles, typically ranging from 40 to 1000 nm in size, that can be released from any cell type. EVs play an essential role in cell-to-cell communication, facilitating the exchange of biomolecules such as proteins, nucleic acids, and lipids over long distances, and are involved in both physiological and pathological processes [78]. EVs can be categorized into exosomes (40–160 nm in diameter), which originate from the endosomal system, and microvesicles, which bud directly from the plasma membrane [94]. Exosomes are derived from intraluminal vesicles (ILVs) within multivesicular bodies (MVBs), which mature and either fuse with lysosomes for degradation or with the plasma membrane to release exosomes into the extracellular environment [95]. Recently, entire organelles or their fragments have been found to be transferred via MDVs, which may contribute to mitochondrial homeostasis [92,96,97,98,99].
MDVs are not dependent on the nuclear fission GTPase dynamin-related protein 1 (DRP1) and mainly originate from the OMM, although they can also arise from the IMM, thus partially including the mitochondrial matrix [91,100,101]. By delivering mildly depolarized mitochondria, MDVs can reduce cellular ROS levels, thereby contributing to preserving mitochondrial quality [102]. For example, stressed brown adipocytes release EVs containing oxidized and damaged mitochondrial fragments to maintain cellular homeostasis and support adaptive thermogenesis [103]. However, in conditions where mitophagy or the mitochondria–lysosome axis is impaired, cellular quality control is compromised, resulting in the accumulation of damaged mitochondria, misfolded proteins, and lipofuscin [104].
In older adults with physical frailty and sarcopenia (PF&S), the finding of elevated levels of EVs containing mitochondrial components suggests an attempt by the cells to eliminate dysfunctional organelles [98]. The involvement of MDVs in the intercellular transfer of mitochondria or mitochondrial components allows for an alternative explanation for such findings to be proposed. Accordingly, metabolically competent cells may release EVs containing functional organelles and/or mitochondrial constituents holding signaling roles to support the viability of cells with high levels of mitochondrial damage [100].
Cardiac myocytes begin eliminating MDVs even when mitochondria are only slightly oxidized, indicating that MDV formation may be independent of mitochondrial depolarization, autophagy signaling, and fission processes [105]. Cultured H9c2 myoblasts have been shown to continuously release MDVs [105]. In this system, damaged proteins resulting from local organelle dysfunction are removed under basal conditions without mitochondrial depolarization [60]. In contrast, mitochondrial stressors such as the mitochondrial inhibitor antimycin-A lead to an overproduction of MDVs [105]. Notably, the cargo of MDVs is determined by the target destination and the type of mitochondrial stress. A study by Todkar et al. [92] demonstrated that, depending on the stimulus, the same protein cargo can be directed either to EVs or to lysosomes. In cases of damaged mitochondria, most MDVs are directed to lysosomes, thereby blocking the alternative pathway of mitochondrial protein release into EVs. Conversely, the PINK1/PARKIN pathway promotes the inclusion of oxidized, damaged proteins into MDVs and inhibits the degradation pathway toward lysosomes. In the skeletal muscle, iron chelation in myotubes leads to reduced mitochondrial respiratory capacity, decreased mitochondrial protein levels, and a reduction in the mtDNA copy number, which supports the secretion of mitochondrial components through MDVs [81].
Recent studies have also suggested that the EV pathway may serve as an alternative mechanism to FPN for iron efflux, wherein MDVs facilitate the transfer of mitochondrial iron into EVs. This process allows cells with accumulated iron to clear excess metal and mitigate iron-induced damage. However, this occurs at the expense of recipient cells that absorb iron-rich EVs, potentially leading to oxidative damage and ferroptosis, a type of cell death caused by iron-dependent lipid peroxidation [106,107,108]. Ferroptosis is induced in satellite cells lacking TfR1, which impairs skeletal muscle regeneration [109]. Ferroptosis is closely associated with lysosomal activity, as lysosomes regulate iron metabolism by degrading ferritin and releasing free iron [110]. Lysosomal dysfunction or damage can result in the accumulation of lipid peroxidation, exacerbating ferroptosis [111]. EVs represent an alternative disposal system for damaged cellular components in the event of lysosomal dysfunction [112]. Indeed, the impairment of lysosomal activity leads to the accumulation of MVBs and increased EV release, with the subsequent overproduction of EVs when lysosomal activity is blocked [113]. Furthermore, iron-binding proteins such as Fer that leak from damaged lysosomes are loaded into EVs and released into the extracellular environment, thus augmenting the cellular iron pool [114]. Furthermore, the IRE–IRP system has been identified as a regulator of the EV marker protein cluster of differentiation (CD) 63. The IRE–IRP system is activated by iron accumulation and controls the transfer of Fer into CD63-positive EVs [115]. Other iron-related proteins, such as Tf and TfR1, have also been found in EVs, with their secretion being primarily induced by oxidative stress, which activates autophagy to dispose of intracellular iron [116,117]. EVs are also involved in iron redistribution [118]. Cytosolic iron export via FPN lowers intracellular iron levels by promoting the degradation of Fer through the proteasome [119]. However, Fer can also be degraded in lysosomes through ferritinophagy, a selective autophagic process that allows for the recycling of stored iron and the reduction in oxidative damage from iron overload [120]. When cellular iron availability is low, the corepressor of the nuclear receptor for the silencing transcription factor RE1 (REST) binds to the Fer gene promoter to induce its transcription [121]. Conversely, in the setting of iron excess, NCOA4 binds to Fer and triggers its degradation via the autophagy–lysosomal pathway. Ferritinophagy is considered a selective form of autophagy, in which excess intracellular Fer can induce iron-mediated apoptosis by activating NCOA4 [122]. Specifically, the Fer–NCOA4 complex localizes to the membrane of a proto-autophagosome and promotes the formation of a fully enclosed autophagosome, which fuses with the lysosome to degrade Fer and release iron into the cytoplasm for cellular use [122]. NCOA4 expression is upregulated during iron deficiency via hypoxia-inducible factor (HIF) and suppressed during iron excess through proteasomal degradation [123]. NCOA4-deficient mice accumulate Fer and iron in tissues at the expense of circulating iron [124]. Several factors modulate NCOA4 expression and ferritinophagy induction, including autophagy-associated genes (ATGs), which activate NCOA4 and promote intracellular iron transport and ferritinophagy, leading to ferroptosis [125,126]. Conversely, reducing NCOA4 or ATG expression mitigates iron overload and lipid peroxidation, thus protecting cells from ferroptosis [127]. NCOA4 expression can also be regulated through additional signaling pathways. The FTH1-NCOA4 complex binds to microtubule-associated protein 1A/1B-light chain 3- phosphatidylethanolamine conjugate (LC3II) in the lysosome, resulting in the release of Fe2+ and enhancement of the Fenton reaction that promotes ferritinophagy and ferroptosis [128]. The binding of NCOA4 to FTH1 can be inhibited by Yes-associated protein 1 (YAP1), a key regulator of the Hippo signaling pathway [129]. Finally, interleukin-6 (IL-6)/signal transducer and activator of transcription 3 (STAT3) and c-Jun N-terminal kinase (JNK)-transcription factor Jun (JUN) are additional regulators of NCOA4 expression [130,131].

4. Promoting Muscle Quality and Function and Iron Homeostasis: Research Gaps and Opportunities

Physical function plays a critical role in maintaining independence during late life. Older adults with functional limitations experience a more rapid decline in physical function compared to those with better physical performance [132,133,134]. However, the biological mechanisms underlying this accelerated decline in mobility-limited older adults are not well understood, and few strategies have proven effective in preventing mobility disability. Measures of physical performance, such as walking speed and chair-stand tests, are strong predictors of the incidence of mobility disability and all-cause mortality. In addition, muscle quality has emerged as a reliable indicator of functional capacity and mobility [1,135].
Iron metabolism in aging muscle has become an area of active research, as an improper handling of iron can lead to its accumulation in parenchymal tissues, negatively impacting muscle quality and function [4,72]. This accumulation may develop in the context of age-related chronic low-grade inflammation, wherein hepcidin signaling becomes upregulated, promoting cellular iron retention [40,136]. Elevated levels of muscle iron and plasma hepcidin have been observed in older individuals compared with younger controls and have been linked to reduced physical performance and increased levels of inflammatory markers such as C-reactive protein (CRP) and IL6 [1,3,4]. Within this pro-inflammatory environment, increased hepcidin expression may disrupt iron homeostasis by inhibiting the expression of the iron export protein FPN, leading to intracellular iron overload.
In addition, reduced mtDNA content and increased mtDNA damage have been observed in the muscle of older adults in association with dysregulated iron homeostasis and mitochondrial dysfunction [3]. Although multiple factors contribute to physical disability, substantial evidence suggests that mitochondrial dysfunction is a significant factor in functional decline [137,138,139]. Mitochondrial dysfunction is closely linked to the molecular pathways identified as hallmarks of aging [140,141]. Disruptions in cellular and mitochondrial iron transport have also been implicated in muscle aging, as they promote oxidative damage-mediated mutations and deletions, thereby reducing mitochondrial genome stability and impairing mitochondrial function. Moreover, ferroptosis has been proposed as a protective mechanism against age-related chronic damage [107].
A significant gap in knowledge is that most of the available evidence on the relationship among iron homeostasis, MQC, and ferroptosis originates from studies conducted in cell lines or tissues other than the muscle. Although these studies provide valuable mechanistic insights, their direct applicability to the skeletal muscle remains speculative. Indeed, the muscle possesses unique structural, metabolic, and regenerative properties, which may influence how these pathways are regulated or respond to stress and aging. However, preclinical studies indicate that age-related iron overload does affect functional parameters such as grip strength, muscle power, and muscle structural integrity [70,72,142]. Nevertheless, the trajectory of muscle quality in older adults and its associations with mitochondrial quality, lysosomal activity, iron metabolism, and measures of lower extremity tissue composition and physical performance remain insufficiently studied.
Preliminary evidence in humans suggests that elevated circulating hepcidin levels and dysregulated muscle iron in older adults with low physical performance are associated with reduced mitochondrial quality [1,3,4]. To advance this field, longitudinal studies examining the relationship between markers of iron regulation and mitochondrial dysfunction in older adults with varying levels of physical performance are needed. In such studies, low-functioning individuals should be compared with high-functioning peers to determine whether greater impairments in these markers are present in the former group. Based on the current literature, it is hypothesized that higher levels of hepcidin, along with increased muscle iron dysregulation and more pronounced mitochondrial dysfunction, may correlate with diminished muscle performance (e.g., slower walking speed). Under the best auspices, the longitudinal analysis of blood and skeletal muscle markers would indicate that an increased level of circulating hepcidin, greater muscle iron dysregulation, and elevated mitochondrial dysfunction in older adults with low physical performance may predict a faster decline in walking speed and physical function. If iron overload is determined to be causally linked to muscle dysfunction and declining physical performance, iron chelators could potentially be used to enhance myocyte viability. However, a major limitation of current chelation therapies is their lack of selectivity for specific organs or macromolecular structures. Once distributed within tissues, these agents indiscriminately chelate iron, restricting their use in older adults who are especially susceptible to iron-deficiency anemia [143]. Furthermore, in skeletal muscle, iron is essential for oxygen transport via myoglobin. Thus, there is an urgent need for targeted therapeutic strategies that selectively reduce labile iron while maintaining the bioavailable iron required for normal physiological function [84].
An overview of the major preclinical and human studies that explored the relationship between iron metabolism and markers of muscle homeostasis is provided in Table 1.

5. Conclusions

Impaired structural and functional muscle remodeling, along with altered mitochondrial quality and function, plays a central role in muscle aging. While the molecular mechanisms underlying these processes remain largely unclear, emerging evidence points to a link between altered muscle iron handling, mitochondrial dysfunction, and systemic inflammation. Understanding the mechanisms through which iron dyshomeostasis contributes to muscle quality decline in advanced age could have significant implications for developing therapeutic strategies that target iron handling to alleviate age-associated muscle decline.

Author Contributions

Conceptualization, R.D.L. and A.P.; writing—original draft preparation, R.D.L. and A.P.; writing—review and editing, R.C., H.J.C.-J., V.P. and E.M.; supervision, C.L. and F.L.; funding acquisition, C.L., F.L. and E.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Institutes of Health [1RO1 AG075136], Università Cattolica del Sacro Cuore [D1.2023 and D1.2024], Next Generation EU PRIN [2022YNENP3], and the nonprofit research foundation Centro Studi Achille e Linda Lorenzon [N/A]. This work was also partially funded by the Italian Ministry of Health [Ricerca Corrente 2025] and Next Generation EU in the context of the National Recovery and Resilience Plan, Investment PE8—Project Age-It: “Ageing Well in an Ageing Society”. This resource was co-financed by the Next Generation EU [DM 1557 11.10.2022]. The views and opinions expressed are only those of the authors and do not necessarily reflect those of the European Union or the European Commission. Neither the European Union nor the European Commission can be held responsible for them.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript
AMBRA1Autophagy and beclin 1 regulator 1
ATGAutophagy-associated gene
BCL2L13BCL-2-like protein 13
BNIP3BCL2-interacting protein 3
CRPC reactive protein
DMT1Divalent metal transporter 1
EREndoplasmic reticulum
EVExtracellular vesicle
Fe2+Ferrous iron
Fe3+Ferric iron
FerFerritin
Fe–S clusterIron–sulfur cluster
FKPB8FK506-binding protein 8
FPNFerroportin
FTH1Ferritin heavy chain
FTLFerritin light chain
FtMtMitochondrial ferritin
FUNDC1FUN14 domain-containing protein 1
FXNFrataxin
HIHeme iron
HIFHypoxia-inducible factor
IL-6Interleukin-6
STAT3Signal transducer and activator of transcription 3
ILVsIntraluminal vesicles
IMMInner mitochondrial membrane
IREsIron-responsive elements
IRP1/2Iron responsive element binding protein-1/2
JNKc-Jun N-terminal kinase
JUNTranscription factor Jun
LC3IIMicrotubule-associated protein 1A/1B-light chain 3
LFLow functioning
LIPLabile iron pool
MAMMitochondria-associated membrane
MCFMitochondrial carrier family
MCSMembrane contact sites
mDMT1Mitochondrial divalent metal transporter 1
MDVsMitochondria-derived vesicles
MFRNMitoferrin
MFSMajor facilitator superfamily
MQCMitochondrial quality control
mtDNAMitochondrial DNA
MVBMultivesicular body
NBR1Neighbor of BRCA1 gene 1 protein
NCOA4Nuclear receptor coactivator 4
NHINon-heme iron
NIXNIP-3-like protein X
OMMOuter mitochondrial membrane
PCBP1Poly(rC) binding protein 1
PF&SPhysical frailty and sarcopenia
PHB2Prohibitin 2
PINK1PTEN-induced kinase 1
RESTNuclear receptor of the silencing transcription factor RE1
ROSReactive oxygen species
SFXNSideroflexins
STEAP3Six-transmembrane epithelial antigen of prostate 3
TfTransferrin
TfR1Transferrin receptor 1
UTRUntranslated region
YAP1Yes-associated protein 1
ZIPZRT-IRT-like protein

References

  1. Picca, A.; Triolo, M.; Wohlgemuth, S.E.; Martenson, M.S.; Mankowski, R.T.; Anton, S.D.; Marzetti, E.; Leeuwenburgh, C.; Hood, D.A. Relationship between mitochondrial quality control markers, lower extremity tissue composition, and physical performance in physically inactive older adults. Cells 2023, 12, 183. [Google Scholar] [CrossRef] [PubMed]
  2. Marzetti, E.; Di Lorenzo, R.; Calvani, R.; Pesce, V.; Landi, F.; Coelho-Júnior, H.J.; Picca, A. From cell architecture to mitochondrial signaling: Role of intermediate filaments in health, aging, and disease. Int. J. Mol. Sci. 2025, 26, 1100. [Google Scholar] [CrossRef] [PubMed]
  3. Picca, A.; Mankowski, R.T.; Kamenov, G.; Anton, S.D.; Manini, T.M.; Buford, T.W.; Saini, S.K.; Calvani, R.; Landi, F.; Bernabei, R.; et al. Advanced age is associated with iron dyshomeostasis and mitochondrial DNA damage in human skeletal muscle. Cells 2019, 8, 1525. [Google Scholar] [CrossRef] [PubMed]
  4. Picca, A.; Saini, S.K.; Mankowski, R.T.; Kamenov, G.; Anton, S.D.; Manini, T.M.; Buford, T.W.; Wohlgemuth, S.E.; Xiao, R.; Calvani, R.; et al. Altered expression of mitoferrin and frataxin, larger labile iron pool and greater mitochondrial DNA damage in the skeletal muscle of older adults. Cells 2020, 9, 2579. [Google Scholar] [CrossRef]
  5. Triolo, M.; Hood, D.A. Manifestations of age on autophagy, mitophagy and lysosomes in skeletal muscle. Cells 2021, 10, 1054. [Google Scholar] [CrossRef]
  6. Picca, A.; Faitg, J.; Auwerx, J.; Ferrucci, L.; D’Amico, D. Mitophagy in human health, ageing and disease. Nat. Metab. 2023, 5, 2047–2061. [Google Scholar] [CrossRef]
  7. Zhou, Q.Y.; Ren, C.; Li, J.Y.; Wang, L.; Duan, Y.; Yao, R.Q.; Tian, Y.P.; Yao, Y.M. The crosstalk between mitochondrial quality control and metal-dependent cell death. Cell Death Dis. 2024, 15, 299. [Google Scholar] [CrossRef]
  8. Hoelzgen, F.; Nguyen, T.T.P.; Klukin, E.; Boumaiza, M.; Srivastava, A.K.; Kim, E.Y.; Zalk, R.; Shahar, A.; Cohen-Schwartz, S.; Meyron-Holtz, E.G.; et al. Structural basis for the intracellular regulation of ferritin degradation. Nat. Commun. 2024, 15, 3802. [Google Scholar] [CrossRef]
  9. Li, J.Y.; Feng, Y.H.; Li, Y.X.; He, P.Y.; Zhou, Q.Y.; Tian, Y.P.; Yao, R.Q.; Yao, Y.M. Ferritinophagy: A novel insight into the double-edged sword in ferritinophagy-ferroptosis axis and human diseases. Cell Prolif. 2024, 57, e13621. [Google Scholar] [CrossRef]
  10. Joseph, A.M.; Adhihetty, P.J.; Leeuwenburgh, C. Beneficial effects of exercise on age-related mitochondrial dysfunction and oxidative stress in skeletal muscle. J. Physiol. 2016, 594, 5105–5123. [Google Scholar] [CrossRef]
  11. Maio, N.; Rouault, T.A. Iron-sulfur cluster biogenesis in mammalian cells: New insights into the molecular mechanisms of cluster delivery. Biochim. Biophys. Acta 2015, 1853, 1493–1512. [Google Scholar] [CrossRef] [PubMed]
  12. Kohgo, Y.; Ikuta, K.; Ohtake, T.; Torimoto, Y.; Kato, J. Body iron metabolism and pathophysiology of iron overload. Int. J. Hematol. 2008, 88, 7–15. [Google Scholar] [CrossRef]
  13. Kafina, M.D.; Paw, B.H. intracellular iron and heme trafficking and metabolism in developing erythroblasts. Metallomics 2017, 9, 1193–1203. [Google Scholar] [CrossRef] [PubMed]
  14. Dutt, S.; Hamza, I.; Bartnikas, T.B. Molecular mechanisms of iron and heme metabolism. Annu. Rev. Nutr. 2022, 42, 311–335. [Google Scholar] [CrossRef] [PubMed]
  15. Latunde-Dada, G.O.; Takeuchi, K.; Simpson, R.J.; McKie, A.T. Haem carrier protein 1 (HCP1): Expression and functional studies in cultured cells. FEBS Lett. 2006, 580, 6865–6870. [Google Scholar] [CrossRef]
  16. Andrews, N.C.; Schmidt, P.J. Iron homeostasis. Annu. Rev. Physiol. 2007, 69, 69–85. [Google Scholar] [CrossRef]
  17. Fraser, S.T.; Midwinter, R.G.; Berger, B.S.; Stocker, R. Heme oxygenase-1: A critical link between iron metabolism, erythropoiesis, and development. Adv. Hematol. 2011, 2011, 473709. [Google Scholar] [CrossRef]
  18. Gulec, S.; Anderson, G.J.; Collins, J.F. Mechanistic and regulatory aspects of intestinal iron absorption. Am. J. Physiol. Gastrointest. Liver. Physiol. 2014, 307, G397–G409. [Google Scholar] [CrossRef]
  19. Knutson, M.; Wessling-Resnick, M. Iron metabolism in the reticuloendothelial system. Crit. Rev. Biochem. Mol. Biol. 2003, 38, 61–88. [Google Scholar] [CrossRef]
  20. Knutson, M.D.; Oukka, M.; Koss, L.M.; Aydemir, F.; Wessling-Resnick, M. Iron release from macrophages after erythrophagocytosis is up-regulated by ferroportin 1 overexpression and down-regulated by hepcidin. Proc. Natl. Acad. Sci. USA 2005, 102, 1324–1328. [Google Scholar] [CrossRef]
  21. Sharp, P.; Srai, S.K. Molecular mechanisms involved in intestinal iron absorption. World J. Gastroenterol. 2007, 13, 4716–4724. [Google Scholar] [CrossRef] [PubMed]
  22. Hentze, M.W.; Muckenthaler, M.U.; Andrews, N.C. Balancing acts: Molecular control of mammalian iron metabolism. Cell 2004, 117, 285–297. [Google Scholar] [CrossRef] [PubMed]
  23. Lichten, L.A.; Cousins, R.J. Mammalian zinc transporters: Nutritional and physiologic regulation. Annu. Rev. Nutr. 2009, 29, 153–176. [Google Scholar] [CrossRef] [PubMed]
  24. Fleming, M.D.; Romano, M.A.; Maureen, A.S.U.; Garrick, L.M.; Garrick, M.D.; Andrews, N.C. Nramp2 is mutated in the anemic belgrade (b) rat: Evidence of a role for Nramp2 in endosomal iron transport. Proc. Natl. Acad. Sci. USA 1998, 95, 1148–1153. [Google Scholar] [CrossRef]
  25. Ohgami, R.S.; Campagna, D.R.; Greer, E.L.; Antiochos, B.; McDonald, A.; Chen, J.; Sharp, J.J.; Fujiwara, Y.; Barker, J.E.; Fleming, M.D. Identification of a ferrireductase required for efficient transferrin-dependent iron uptake in erythroid cells. Nat. Genet. 2005, 37, 1264–1269. [Google Scholar] [CrossRef]
  26. Finney, L.A.; O’Halloran, T.V. Transition metal speciation in the cell: Insights from the chemistry of metal ion receptors. Science 2003, 300, 931–936. [Google Scholar] [CrossRef]
  27. Kakhlon, O.; Cabantchik, Z.I. The labile iron pool: Characterization, measurement, and participation in cellular processes. Free Radic. Biol. Med. 2002, 33, 1037–1046. [Google Scholar] [CrossRef]
  28. Dai, Y.; Zhu, C.; Xiao, W.; Chen, X.; Cai, Y. Mycobacterium tuberculosis induces host autophagic ferritin degradation for enhanced iron bioavailability and bacterial growth. Autophagy 2024, 20, 943–945. [Google Scholar] [CrossRef]
  29. Mleczko-Sanecka, K.; Silvestri, L. Cell-type-specific insights into iron regulatory processes. Am. J. Hematol. 2021, 96, 110–127. [Google Scholar] [CrossRef]
  30. Linder, M.C. Mobilization of stored iron in mammals: A review. Nutrients 2013, 5, 4022–4050. [Google Scholar] [CrossRef]
  31. Patel, S.J.; Protchenko, O.; Shakoury-Elizeh, M.; Baratz, E.; Jadhav, S.; Philpott, C.C. The iron chaperone and nucleic acid-binding activities of poly(rC)-binding protein 1 are separable and independently essential. Proc. Natl. Acad. Sci. USA 2021, 118, e2104666118. [Google Scholar] [CrossRef] [PubMed]
  32. Protchenko, O.; Baratz, E.; Jadhav, S.; Li, F.; Shakoury-Elizeh, M.; Gavrilova, O.; Ghosh, M.C.; Cox, J.E.; Maschek, J.A.; Tyurin, V.A.; et al. Iron chaperone poly RC binding protein 1 protects mouse liver from lipid peroxidation and steatosis. Hepatology 2021, 73, 1176–1193. [Google Scholar] [CrossRef] [PubMed]
  33. Drakesmith, H.; Nemeth, E.; Ganz, T. Ironing out ferroportin. Cell Metab. 2015, 22, 777–787. [Google Scholar] [CrossRef] [PubMed]
  34. Bonaccorsi Di Patti, M.C.; Polticelli, F.; Cece, G.; Cutone, A.; Felici, F.; Persichini, T.; Musci, G. A structural model of human ferroportin and of its iron binding site. FEBS J. 2014, 281, 2851–2860. [Google Scholar] [CrossRef]
  35. Le Gac, G.; Ka, C.; Joubrel, R.; Gourlaouen, I.; Lehn, P.; Mornon, J.P.; Férec, C.; Callebaut, I. Structure-function analysis of the human ferroportin iron exporter (SLC40A1): Effect of hemochromatosis type 4 disease mutations and identification of critical residues. Hum. Mutat. 2013, 34, 1371–1380. [Google Scholar] [CrossRef]
  36. Ganz, T.; Nemeth, E. Hepcidin and disorders of iron metabolism. Annu. Rev. Med. 2011, 62, 347–360. [Google Scholar] [CrossRef]
  37. Nemeth, E.; Tuttle, M.S.; Powelson, J.; Vaughn, M.B.; Donovan, A.; Ward, D.M.; Ganz, T.; Kaplan, J. Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science 2004, 306, 2090–2093. [Google Scholar] [CrossRef]
  38. den Elzen, W.P.J.; de Craen, A.J.M.; Wiegerinck, E.T.; Westendorp, R.G.J.; Swinkels, D.W.; Gussekloo, J. Plasma hepcidin levels and anemia in old age. The Leiden 85-Plus Study. Haematologica 2013, 98, 448–454. [Google Scholar] [CrossRef]
  39. Lin, L.; Valore, E.V.; Nemeth, E.; Goodnough, J.B.; Gabayan, V.; Ganz, T. Iron transferrin regulates hepcidin synthesis in primary hepatocyte culture through hemojuvelin and BMP2/4. Blood 2007, 110, 2182–2189. [Google Scholar] [CrossRef]
  40. Sukhbaatar, N.; Weichhart, T. Iron regulation: Macrophages in control. Pharmaceuticals 2018, 11, 137. [Google Scholar] [CrossRef]
  41. Majore, S.; Bonaccorsi di Patti, M.C.; Valiante, M.; Polticelli, F.; Cortese, A.; Di Bartolomeo, S.; De Bernardo, C.; De Muro, M.; Faienza, F.; Radio, F.C.; et al. Characterization of three novel pathogenic SLC40A1 mutations and genotype/phenotype correlations in 7 Italian families with type 4 hereditary hemochromatosis. Biochim. Biophys. Acta Mol. Basis Dis. 2018, 1864, 464–470. [Google Scholar] [CrossRef] [PubMed]
  42. Rouault, T.A. The role of iron regulatory proteins in mammalian iron homeostasis and disease. Nat. Chem. Biol. 2006, 2, 406–414. [Google Scholar] [CrossRef] [PubMed]
  43. Galy, B.; Ferring-Appel, D.; Kaden, S.; Gröne, H.J.; Hentze, M.W. Iron regulatory proteins are essential for intestinal function and control key iron absorption molecules in the duodenum. Cell Metab. 2008, 7, 79–85. [Google Scholar] [CrossRef] [PubMed]
  44. Baringer, S.L.; Palsa, K.; Spiegelman, V.S.; Simpson, I.A.; Connor, J.R. Apo- and holo-transferrin differentially interact with hephaestin and ferroportin in a novel mechanism of cellular iron release regulation. J. Biomed. Sci. 2023, 30, 36. [Google Scholar] [CrossRef]
  45. Papanikolaou, G.; Pantopoulos, K. Iron metabolism and toxicity. Toxicol. Appl. Pharmacol. 2005, 202, 199–211. [Google Scholar] [CrossRef]
  46. Paul, B.T.; Manz, D.H.; Torti, F.M.; Torti, S.V. Mitochondria and iron: Current questions. Expert Rev. Hematol. 2017, 10, 65–79. [Google Scholar] [CrossRef]
  47. Sheftel, A.D.; Zhang, A.S.; Brown, C.; Shirihai, O.S.; Ponka, P. Direct interorganellar transfer of iron from endosome to mitochondrion. Blood 2007, 110, 125–132. [Google Scholar] [CrossRef]
  48. Shvartsman, M.; Kikkeri, R.; Shanzer, A.; Cabantchik, Z.I. Non-transferrin-bound iron reaches mitochondria by a chelator-inaccessible mechanism: Biological and clinical implications. Am. J. Physiol. Cell Physiol. 2007, 293, C1383–C1394. [Google Scholar] [CrossRef]
  49. Romslo, I. Energy-dependent accumulation of iron by isolated rat liver mitochondria. IV. Relationship to the energy state of the mitochondria. Biochim. Biophys. Acta 1975, 387, 69–79. [Google Scholar] [CrossRef]
  50. Lange, H.; Kispal, G.; Lill, R. Mechanism of iron transport to the site of heme synthesis inside yeast mitochondria. J. Biol. Chem. 1999, 274, 18989–18996. [Google Scholar] [CrossRef]
  51. Wolff, N.A.; Ghio, A.J.; Garrick, L.M.; Garrick, M.D.; Zhao, L.; Fenton, R.A.; Thévenod, F. Evidence for mitochondrial localization of divalent metal transporter 1 (DMT1). FASEB J. 2014, 28, 2134–2145. [Google Scholar] [CrossRef] [PubMed]
  52. Lane, D.J.R.; Merlot, A.M.; Huang, M.L.H.; Bae, D.H.; Jansson, P.J.; Sahni, S.; Kalinowski, D.S.; Richardson, D.R. Cellular iron uptake, trafficking and metabolism: Key molecules and mechanisms and their roles in disease. Biochim. Biophys. Acta 2015, 1853, 1130–1144. [Google Scholar] [CrossRef] [PubMed]
  53. Ali, M.Y.; Oliva, C.R.; Flor, S.; Griguer, C.E. Mitoferrin, cellular and mitochondrial iron homeostasis. Cells 2022, 11, 3464. [Google Scholar] [CrossRef] [PubMed]
  54. Shaw, G.C.; Cope, J.J.; Li, L.; Corson, K.; Hersey, C.; Ackermann, G.E.; Gwynn, B.; Lambert, A.J.; Wingert, R.A.; Traver, D.; et al. Mitoferrin is essential for erythroid iron assimilation. Nature 2006, 440, 96–100. [Google Scholar] [CrossRef]
  55. Hildick-Smith, G.J.; Cooney, J.D.; Garone, C.; Kremer, L.S.; Haack, T.B.; Thon, J.N.; Miyata, N.; Lieber, D.S.; Calvo, S.E.; Akman, H.O.; et al. Macrocytic anemia and mitochondriopathy resulting from a defect in sideroflexin 4. Am. J. Hum. Genet. 2013, 93, 906–914. [Google Scholar] [CrossRef]
  56. Richardson, D.R.; Lane, D.J.R.; Becker, E.M.; Huang, M.L.H.; Whitnall, M.; Rahmanto, Y.S.; Sheftel, A.D.; Ponka, P. Mitochondrial iron trafficking and the integration of iron metabolism between the mitochondrion and cytosol. Proc. Natl. Acad. Sci. USA 2010, 107, 10775–10782. [Google Scholar] [CrossRef]
  57. Martinez-Guzman, O.; Willoughby, M.M.; Saini, A.; Dietz, J.V.; Bohovych, I.; Medlock, A.E.; Khalimonchuk, O.; Reddi, A.R. Mitochondrial-nuclear heme trafficking in budding yeast is regulated by GTPases that control mitochondrial dynamics and ER contact sites. J. Cell Sci. 2020, 133, jcs237917. [Google Scholar] [CrossRef]
  58. Chambers, I.G.; Willoughby, M.M.; Hamza, I.; Reddi, A.R. One Ring to bring them all and in the darkness bind them: The trafficking of heme without deliverers. Biochim. Biophys. Acta Mol. Cell Res. 2021, 1868, 118881. [Google Scholar] [CrossRef]
  59. Neuspiel, M.; Schauss, A.C.; Braschi, E.; Zunino, R.; Rippstein, P.; Rachubinski, R.A.; Andrade-Navarro, M.A.; McBride, H.M. Cargo-selected transport from the mitochondria to peroxisomes is mediated by vesicular carriers. Curr. Biol. 2008, 18, 102–108. [Google Scholar] [CrossRef]
  60. Soubannier, V.; McLelland, G.-L.; Zunino, R.; Braschi, E.; Rippstein, P.; Fon, E.A.; McBride, H.M. A vesicular transport pathway shuttles cargo from mitochondria to lysosomes. Curr. Biol. 2012, 22, 135–141. [Google Scholar] [CrossRef]
  61. Santambrogio, P.; Biasiotto, G.; Sanvito, F.; Olivieri, S.; Arosio, P.; Levi, S. Mitochondrial ferritin expression in adult mouse tissues. J. Histochem. Cytochem. 2007, 55, 1129–1137. [Google Scholar] [CrossRef] [PubMed]
  62. Focht, S.J.; Snyder, B.S.; Beard, J.L.; Van Gelder, W.; Williams, L.R.; Connor, J.R. Regional distribution of iron, transferrin, ferritin, and oxidatively- modified proteins in young and aged fischer 344 rat brains. Neuroscience 1997, 79, 255–261. [Google Scholar] [CrossRef] [PubMed]
  63. Campanella, A.; Rovelli, E.; Santambrogio, P.; Cozzi, A.; Taroni, F.; Levi, S. Mitochondrial ferritin limits oxidative damage regulating mitochondrial iron availability: Hypothesis for a protective role in Friedreich ataxia. Hum. Mol. Genet. 2009, 18, 1–11. [Google Scholar] [CrossRef] [PubMed]
  64. Nie, G.; Sheftel, A.D.; Kim, S.F.; Ponka, P. Overexpression of mitochondrial ferritin causes cytosolic iron depletion and changes cellular iron homeostasis. Blood 2005, 105, 2161–2167. [Google Scholar] [CrossRef]
  65. Zeidan, R.S.; Han, S.M.; Leeuwenburgh, C.; Xiao, R. Iron homeostasis and organismal aging. Ageing Res. Rev. 2021, 72, 101510. [Google Scholar] [CrossRef]
  66. Tian, Y.; Tian, Y.; Yuan, Z.; Zeng, Y.; Wang, S.; Fan, X.; Yang, D.; Yang, M. Iron metabolism in aging and age-related diseases. Int. J. Mol. Sci. 2022, 23, 3612. [Google Scholar] [CrossRef]
  67. Maus, M.; López-Polo, V.; Mateo, L.; Lafarga, M.; Aguilera, M.; De Lama, E.; Meyer, K.; Sola, A.; Lopez-Martinez, C.; López-Alonso, I.; et al. Iron accumulation drives fibrosis, senescence and the senescence-associated secretory phenotype. Nat. Metab. 2023, 5, 2111–2130. [Google Scholar] [CrossRef]
  68. Zeidan, R.S.; Martenson, M.; Tamargo, J.A.; McLaren, C.; Ezzati, A.; Lin, Y.; Yang, J.J.; Yoon, H.S.; McElroy, T.; Collins, J.F.; et al. Iron homeostasis in older adults: Balancing nutritional requirements and health risks. J. Nutr. Health Aging 2024, 28, 100212. [Google Scholar] [CrossRef]
  69. Hofer, T.; Marzetti, E.; Xu, J.; Seo, A.Y.; Gulec, S.; Knutson, M.D.; Leeuwenburgh, C.; Dupont-Versteegden, E.E. Increased iron content and RNA oxidative damage in skeletal muscle with aging and disuse atrophy. Exp. Gerontol. 2008, 43, 563–570. [Google Scholar] [CrossRef]
  70. Jung, S.H.; DeRuisseau, L.R.; Kavazis, A.N.; DeRuisseau, K.C. Plantaris muscle of aged rats demonstrates iron accumulation and altered expression of iron regulation proteins. Exp. Physiol. 2008, 93, 407–414. [Google Scholar] [CrossRef]
  71. Altun, M.; Edström, E.; Spooner, E.; Flores-Moralez, A.; Bergman, E.; Tollet-Egnell, P.; Norstedt, G.; Kessler, B.M.; Ulfhake, B. Iron load and redox stress in skeletal muscle of aged rats. Muscle Nerve 2007, 36, 223–233. [Google Scholar] [CrossRef] [PubMed]
  72. Xu, J.; Knutson, M.D.; Carter, C.S.; Leeuwenburgh, C. Iron accumulation with age, oxidative stress and functional decline. PLoS ONE 2008, 3, e2865. [Google Scholar] [CrossRef] [PubMed]
  73. DeRuisseau, K.C.; Park, Y.M.; DeRuisseau, L.R.; Cowley, P.M.; Fazen, C.H.; Doyle, R.P. Aging-related changes in the iron status of skeletal muscle. Exp. Gerontol. 2013, 48, 1294–1302. [Google Scholar] [CrossRef] [PubMed]
  74. Xu, J.; Marzetti, E.; Seo, A.Y.; Kim, J.-S.; Prolla, T.A.; Leeuwenburgh, C. The emerging role of iron dyshomeostasis in the mitochondrial decay of aging. Mech. Ageing Dev. 2010, 131, 487–493. [Google Scholar] [CrossRef]
  75. Seo, A.Y.; Xu, J.; Servais, S.; Hofer, T.; Marzetti, E.; Wohlgemuth, S.E.; Knutson, M.D.; Chung, H.Y.; Leeuwenburgh, C. Mitochondrial iron accumulation with age and functional consequences. Aging Cell 2008, 7, 706–716. [Google Scholar] [CrossRef]
  76. Song, C.C.; Pantopoulos, K.; Chen, G.H.; Zhong, C.C.; Zhao, T.; Zhang, D.G.; Luo, Z. Iron increases lipid deposition via oxidative stress-mediated mitochondrial dysfunction and the HIF1α-PPARγ pathway. Cell. Mol. Life Sci. 2022, 79, 394. [Google Scholar] [CrossRef]
  77. Terman, A.; Brunk, U.T. Aging as a catabolic malfunction. Int. J. Biochem. Cell Biol. 2004, 36, 2365–2375. [Google Scholar] [CrossRef]
  78. Ferrucci, L.; Guerra, F.; Bucci, C.; Marzetti, E.; Picca, A. Mitochondria break free: Mitochondria-derived vesicles in aging and associated conditions. Ageing Res. Rev. 2024, 102, 102549. [Google Scholar] [CrossRef]
  79. Falchi, A.M.; Sogos, V.; Saba, F.; Piras, M.; Congiu, T.; Piludu, M. Astrocytes shed large membrane vesicles that contain mitochondria, lipid droplets and ATP. Histochem. Cell Biol. 2013, 139, 221–231. [Google Scholar] [CrossRef]
  80. Davis, C.H.O.; Kim, K.Y.; Bushong, E.A.; Mills, E.A.; Boassa, D.; Shih, T.; Kinebuchi, M.; Phan, S.; Zhou, Y.; Bihlmeyer, N.A.; et al. Transcellular degradation of axonal mitochondria. Proc. Natl. Acad. Sci. USA 2014, 111, 9633–9638. [Google Scholar] [CrossRef]
  81. Leermakers, P.A.; Remels, A.H.V.; Zonneveld, M.I.; Rouschop, K.M.A.; Schols, A.M.W.J.; Gosker, H.R. Iron deficiency-induced loss of skeletal muscle mitochondrial proteins and respiratory capacity; the role of mitophagy and secretion of mitochondria-containing vesicles. FASEB J. 2020, 34, 6703–6717. [Google Scholar] [CrossRef]
  82. Bencze, K.Z.; Kondapalli, K.C.; Cook, J.D.; McMahon, S.; Millán-Pacheco, C.; Pastor, N.; Stemmler, T.L. The structure and function of frataxin. Crit. Rev. Biochem. Mol. Biol. 2006, 41, 269–291. [Google Scholar] [CrossRef] [PubMed]
  83. Runko, A.P.; Griswold, A.J.; Min, K.T. Overexpression of frataxin in the mitochondria increases resistance to oxidative stress and extends lifespan in Drosophila. FEBS Lett. 2008, 582, 715–719. [Google Scholar] [CrossRef] [PubMed]
  84. Xu, J.; Hwang, J.C.Y.; Lees, H.A.; Wohlgemuth, S.E.; Knutson, M.D.; Judge, A.R.; Dupont-Versteegden, E.E.; Marzetti, E.; Leeuwenburgh, C. Long-term perturbation of muscle iron homeostasis following hindlimb suspension in old rats is associated with high levels of oxidative stress and impaired recovery from atrophy. Exp. Gerontol. 2012, 47, 100–108. [Google Scholar] [CrossRef]
  85. Barrientos, T.; Laothamatas, I.; Koves, T.R.; Soderblom, E.J.; Bryan, M.; Moseley, M.A.; Muoio, D.M.; Andrews, N.C. Metabolic catastrophe in mice lacking transferrin receptor in muscle. EBioMedicine 2015, 2, 1705–1717. [Google Scholar] [CrossRef]
  86. Choi, G.E.; Lee, H.J.; Chae, C.W.; Cho, J.H.; Jung, Y.H.; Kim, J.S.; Kim, S.Y.; Lim, J.R.; Han, H.J. BNIP3L/NIX-Mediated mitophagy protects against glucocorticoid-induced synapse defects. Nat. Commun. 2021, 12, 487. [Google Scholar] [CrossRef] [PubMed]
  87. Liu, C.; Li, H.J.; Duan, W.X.; Duan, Y.; Yu, Q.; Zhang, T.; Sun, Y.P.; Li, Y.Y.; Liu, Y.S.; Xu, S.C. MCU upregulation overactivates mitophagy by promoting VDAC1 dimerization and ubiquitination in the hepatotoxicity of cadmium. Adv. Sci. (Weinh) 2023, 10, e2203869. [Google Scholar] [CrossRef]
  88. Chen, M.; Chen, Z.; Wang, Y.; Tan, Z.; Zhu, C.; Li, Y.; Han, Z.; Chen, L.; Gao, R.; Liu, L.; et al. Mitophagy receptor FUNDC1 regulates mitochondrial dynamics and mitophagy. Autophagy 2016, 12, 689–702. [Google Scholar] [CrossRef]
  89. Picca, A.; Guerra, F.; Calvani, R.; Romano, R.; Coelho-Junior, H.J.; Bucci, C.; Leeuwenburgh, C.; Marzetti, E. Mitochondrial-derived vesicles in skeletal muscle remodeling and adaptation. Semin. Cell Dev. Biol. 2023, 143, 37–45. [Google Scholar] [CrossRef]
  90. Soubannier, V.; Rippstein, P.; Kaufman, B.A.; Shoubridge, E.A.; McBride, H.M. Reconstitution of mitochondria derived vesicle formation demonstrates selective enrichment of oxidized cargo. PLoS ONE 2012, 7, e52830. [Google Scholar] [CrossRef]
  91. Sugiura, A.; McLelland, G.-L.; Fon, E.A.; McBride, H.M. A new pathway for mitochondrial quality control: Mitochondrial-derived vesicles. EMBO J. 2014, 33, 2142–2156. [Google Scholar] [CrossRef] [PubMed]
  92. Todkar, K.; Chikhi, L.; Desjardins, V.; El-Mortada, F.; Pépin, G.; Germain, M. Selective packaging of mitochondrial proteins into extracellular vesicles prevents the release of mitochondrial DAMPs. Nat. Commun. 2021, 12, 1971. [Google Scholar] [CrossRef] [PubMed]
  93. McLelland, G.-L.; Soubannier, V.; Chen, C.X.; McBride, H.M.; Fon, E.A. Parkin and PINK1 function in a vesicular trafficking pathway regulating mitochondrial quality control. EMBO J. 2014, 33, 282–295. [Google Scholar] [CrossRef]
  94. Welsh, J.A.; Goberdhan, D.C.I.; O’Driscoll, L.; Buzas, E.I.; Blenkiron, C.; Bussolati, B.; Cai, H.; Di Vizio, D.; Driedonks, T.A.P.; Erdbrügger, U.; et al. Minimal information for studies of extracellular vesicles (MISEV2023): From basic to advanced approaches. J. Extracell. Vesicles 2024, 13, e12404. [Google Scholar] [CrossRef] [PubMed]
  95. Colombo, M.; Raposo, G.; Théry, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef]
  96. Marzetti, E.; Coelho-Júnior, H.J.; Calvani, R.; Girolimetti, G.; Di Corato, R.; Ciciarello, F.; Galluzzo, V.; Di Mario, C.; Tolusso, B.; Santoro, L.; et al. Mitochondria-derived vesicles and inflammatory profiles of adults with long COVID supplemented with red beetroot juice: Secondary analysis of a randomized controlled trial. Int. J. Mol. Sci. 2025, 26, 1224. [Google Scholar] [CrossRef]
  97. Picca, A.; Guerra, F.; Calvani, R.; Marini, F.; Biancolillo, A.; Landi, G.; Beli, R.; Landi, F.; Bernabei, R.; Bentivoglio, A.R.; et al. Mitochondrial signatures in circulating extracellular vesicles of older adults with Parkinson’s disease: Results from the EXosomes in PArkiNson’s Disease (EXPAND) study. J. Clin. Med. 2020, 9, 504. [Google Scholar] [CrossRef]
  98. Picca, A.; Beli, R.; Calvani, R.; Coelho-Júnior, H.J.; Landi, F.; Bernabei, R.; Bucci, C.; Guerra, F.; Marzetti, E. Older adults with physical frailty and sarcopenia show increased levels of circulating small extracellular vesicles with a specific mitochondrial signature. Cells 2020, 9, 973. [Google Scholar] [CrossRef]
  99. Marzetti, E.; Guerra, F.; Calvani, R.; Marini, F.; Biancolillo, A.; Gervasoni, J.; Primiano, A.; Coelho-Júnior, H.J.; Landi, F.; Bernabei, R.; et al. Circulating mitochondrial-derived vesicles, inflammatory biomarkers and amino acids in older adults with physical frailty and sarcopenia: A preliminary BIOSPHERE multi-marker study using sequential and orthogonalized covariance selection—Linear discriminant analysis. Front. Cell Dev. Biol. 2020, 8, 564417. [Google Scholar] [CrossRef]
  100. Picca, A.; Guerra, F.; Calvani, R.; Coelho-Júnior, H.J.; Landi, F.; Bucci, C.; Marzetti, E. Mitochondrial-derived vesicles: The good, the bad, and the ugly. Int. J. Mol. Sci. 2023, 24, 3835. [Google Scholar] [CrossRef]
  101. König, T.; McBride, H.M. Mitochondrial-derived vesicles in metabolism, disease, and aging. Cell Metab. 2024, 36, 21–35. [Google Scholar] [CrossRef] [PubMed]
  102. Choong, C.J.; Okuno, T.; Ikenaka, K.; Baba, K.; Hayakawa, H.; Koike, M.; Yokota, M.; Doi, J.; Kakuda, K.; Takeuchi, T.; et al. Alternative mitochondrial quality control mediated by extracellular release. Autophagy 2020, 17, 2962–2974. [Google Scholar] [CrossRef] [PubMed]
  103. Rosina, M.; Ceci, V.; Turchi, R.; Chuan, L.; Borcherding, N.; Sciarretta, F.; Sánchez-Díaz, M.; Tortolici, F.; Karlinsey, K.; Chiurchiù, V.; et al. Ejection of damaged mitochondria and their removal by macrophages ensure efficient thermogenesis in brown adipose tissue. Cell Metab. 2022, 34, 533–548.e12. [Google Scholar] [CrossRef] [PubMed]
  104. Terman, A.; Kurz, T.; Navratil, M.; Arriaga, E.A.; Brunk, U.T. Mitochondrial turnover and aging of long-lived postmitotic cells: The mitochondrial-lysosomal axis theory of aging. Antioxid. Redox Signal. 2010, 12, 503–535. [Google Scholar] [CrossRef]
  105. Cadete, V.J.J.; Deschênes, S.; Cuillerier, A.; Brisebois, F.; Sugiura, A.; Vincent, A.; Turnbull, D.; Picard, M.; McBride, H.M.; Burelle, Y. Formation of mitochondrial-derived vesicles is an active and physiologically relevant mitochondrial quality control process in the cardiac system. J. Physiol. 2016, 594, 5343–5362. [Google Scholar] [CrossRef]
  106. Stockwell, B.R.; Friedmann Angeli, J.P.; Bayir, H.; Bush, A.I.; Conrad, M.; Dixon, S.J.; Fulda, S.; Gascón, S.; Hatzios, S.K.; Kagan, V.E.; et al. Ferroptosis: A regulated cell death nexus linking metabolism, redox biology, and disease. Cell 2017, 171, 273–285. [Google Scholar] [CrossRef]
  107. Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef]
  108. Zhou, B.; Liu, J.; Kang, R.; Klionsky, D.J.; Kroemer, G.; Tang, D. Ferroptosis is a type of autophagy-dependent cell death. Semin. Cancer Biol. 2020, 66, 89–100. [Google Scholar] [CrossRef]
  109. Ding, H.; Chen, S.; Pan, X.; Dai, X.; Pan, G.; Li, Z.; Mai, X.; Tian, Y.; Zhang, S.; Liu, B.; et al. Transferrin receptor 1 ablation in satellite cells impedes skeletal muscle regeneration through activation of ferroptosis. J. Cachexia Sarcopenia Muscle 2021, 12, 746–768. [Google Scholar] [CrossRef]
  110. Rizzollo, F.; More, S.; Vangheluwe, P.; Agostinis, P. The lysosome as a master regulator of iron metabolism. Trends Biochem. Sci. 2021, 46, 960–975. [Google Scholar] [CrossRef]
  111. Li, S.; Liao, Z.; Yin, H.; Liu, O.; Hua, W.; Wu, X.; Zhang, Y.; Gao, Y.; Yang, C. G3BP1 Coordinates lysophagy activity to protect against compression-induced cell ferroptosis during intervertebral disc degeneration. Cell Prolif. 2023, 56, e13368. [Google Scholar] [CrossRef] [PubMed]
  112. Eitan, E.; Suire, C.; Zhang, S.; Mattson, M.P. Impact of lysosome status on extracellular vesicle content and release. Ageing Res. Rev. 2016, 32, 65–74. [Google Scholar] [CrossRef]
  113. Adams, S.D.; Csere, J.; D’angelo, G.; Carter, E.P.; Romao, M.; Arnandis, T.; Dodel, M.; Kocher, H.M.; Grose, R.; Raposo, G.; et al. Centrosome amplification mediates small extracellular vesicle secretion via lysosome disruption. Curr. Biol. 2021, 31, 1403–1416.e7. [Google Scholar] [CrossRef] [PubMed]
  114. Atipimonpat, A.; Siwaponanan, P.; Khuhapinant, A.; Svasti, S.; Sukapirom, K.; Khowawisetsut, L.; Pattanapanyasat, K. Extracellular vesicles from thalassemia patients carry iron-containing ferritin and hemichrome that promote cardiac cell proliferation. Ann. Hematol. 2021, 100, 1929–1946. [Google Scholar] [CrossRef] [PubMed]
  115. Yanatori, I.; Richardson, D.R.; Dhekne, H.S.; Toyokuni, S.; Kishi, F. CD63 is regulated by iron via the IRE-IRP system and is important for ferritin secretion by extracellular vesicles. Blood 2021, 138, 1490–1503. [Google Scholar] [CrossRef] [PubMed]
  116. Gardner, J.O.; Leidal, A.M.; Nguyen, T.A.; Debnath, J. LC3-dependent EV loading and secretion (LDELS)promotes TFRC (transferrin receptor) secretion via extracellular vesicles. Autophagy 2023, 19, 1551–1561. [Google Scholar] [CrossRef]
  117. Carayon, K.; Chaoui, K.; Ronzier, E.; Lazar, I.; Bertrand-Michel, J.; Roques, V.; Balor, S.; Terce, F.; Lopez, A.; Salomé, L.; et al. Proteolipidic composition of exosomes changes during reticulocyte maturation. J. Biol. Chem. 2011, 286, 34426–34439. [Google Scholar] [CrossRef]
  118. Chen, H.; Tang, Y. Iron-loaded extracellular vesicles: Angel or demon? Free Radic. Res. 2023, 57, 61–68. [Google Scholar] [CrossRef]
  119. De Domenico, I.; Vaughn, M.B.; Li, L.; Bagley, D.; Musci, G.; Ward, D.M.; Kaplan, J. Ferroportin-mediated mobilization of ferritin iron precedes ferritin degradation by the proteasome. EMBO J. 2006, 25, 5396–5404. [Google Scholar] [CrossRef]
  120. Jin, X.; Jiang, C.; Zou, Z.; Huang, H.; Li, X.; Xu, S.; Tan, R. Ferritinophagy in the etiopathogenic mechanism of related diseases. J. Nutr. Biochem. 2023, 117, 109339. [Google Scholar] [CrossRef]
  121. Maksour, S.; Ooi, L.; Dottori, M. More than a corepressor: The role of CoREST proteins in neurodevelopment. eNeuro 2020, 7, ENEURO.0337-19.2020. [Google Scholar] [CrossRef] [PubMed]
  122. Santana-Codina, N.; Gikandi, A.; Mancias, J.D. The role of NCOA4-mediated ferritinophagy in ferroptosis. Adv. Exp. Med. Biol. 2021, 1301, 41–57. [Google Scholar] [CrossRef] [PubMed]
  123. Li, X.; Lozovatsky, L.; Sukumaran, A.; Gonzalez, L.; Jain, A.; Liu, D.; Ayala-Lopez, N.; Finberg, K.E. NCOA4 is regulated by HIF and mediates mobilization of murine hepatic iron stores after blood loss. Blood 2020, 136, 2691–2702. [Google Scholar] [CrossRef] [PubMed]
  124. Bellelli, R.; Federico, G.; Matte’, A.; Colecchia, D.; Iolascon, A.; Chiariello, M.; Santoro, M.; De Franceschi, L.; Carlomagno, F. NCOA4 deficiency impairs systemic iron homeostasis. Cell Rep. 2016, 14, 411–421. [Google Scholar] [CrossRef]
  125. Li, J.; Yuan, J.; Li, Y.; Wang, J.; Xie, Q.; Ma, R.; Wang, J.; Ren, M.; Lu, D.; Xu, Z. D-borneol enhances cisplatin sensitivity via autophagy dependent EMT signaling and NCOA4-mediated ferritinophagy. Phytomedicine 2022, 106, 154411. [Google Scholar] [CrossRef]
  126. Nai, A.; Lidonnici, M.R.; Federico, G.; Pettinato, M.; Olivari, V.; Carrillo, F.; Crich, S.G.; Ferrari, G.; Camaschella, C.; Silvestri, L.; et al. NCOA4-mediated ferritinophagy in macrophages is crucial to sustain erythropoiesis in mice. Haematologica 2021, 106, 795–805. [Google Scholar] [CrossRef]
  127. Hou, W.; Xie, Y.; Song, X.; Sun, X.; Lotze, M.T.; Zeh, H.J.; Kang, R.; Tang, D. Autophagy promotes ferroptosis by degradation of ferritin. Autophagy 2016, 12, 1425–1428. [Google Scholar] [CrossRef]
  128. Fang, Y.; Chen, X.; Tan, Q.; Zhou, H.; Xu, J.; Gu, Q. Inhibiting ferroptosis through disrupting the NCOA4-FTH1 interaction: A new mechanism of action. ACS Cent. Sci. 2021, 7, 980–989. [Google Scholar] [CrossRef]
  129. Wang, J.; Zhu, Q.; Li, R.; Zhang, J.; Ye, X.; Li, X. YAP1 protects against septic liver injury via ferroptosis resistance. Cell Biosci. 2022, 12, 163. [Google Scholar] [CrossRef]
  130. Sun, K.; Hou, L.; Guo, Z.; Wang, G.; Guo, J.; Xu, J.; Zhang, X.; Guo, F. JNK-JUN-NCOA4 axis contributes to chondrocyte ferroptosis and aggravates osteoarthritis via ferritinophagy. Free Radic. Biol. Med. 2023, 200, 87–101. [Google Scholar] [CrossRef]
  131. Zhu, M.; Peng, L.; Huo, S.; Peng, D.; Gou, J.; Shi, W.; Tao, J.; Jiang, T.; Jiang, Y.; Wang, Q.; et al. stat3 signaling promotes cardiac injury by upregulating NCOA4-mediated ferritinophagy and ferroptosis in high-fat-diet fed mice. Free Radic. Biol. Med. 2023, 201, 111–125. [Google Scholar] [CrossRef] [PubMed]
  132. Chang, M.; Cohen-Mansfield, J.; Ferrucci, L.; Leveille, S.; Volpato, S.; De Rekeneire, N.; Guralnik, J.M. Incidence of loss of ability to walk 400 meters in a functionally limited older population. J. Am. Geriatr. Soc. 2004, 52, 2094–2098. [Google Scholar] [CrossRef] [PubMed]
  133. Guralnik, J.M.; Ferrucci, L.; Simonsick, E.M.; Salive, M.E.; Wallace, R.B. Lower-extremity function in persons over the age of 70 years as a predictor of subsequent disability. N. Engl. J. Med. 1995, 332, 556–562. [Google Scholar] [CrossRef]
  134. Pavasini, R.; Guralnik, J.; Brown, J.C.; di Bari, M.; Cesari, M.; Landi, F.; Vaes, B.; Legrand, D.; Verghese, J.; Wang, C.; et al. Short physical performance battery and all-cause mortality: Systematic review and meta-analysis. BMC Med. 2016, 14, 215. [Google Scholar] [CrossRef] [PubMed]
  135. Coen, P.M.; Huo, Z.; Tranah, G.J.; Barnes, H.N.; Zhang, X.; Wolff, C.A.; Wu, K.; Cawthon, P.M.; Hepple, R.T.; Toledo, F.G.S.; et al. Autophagy gene expression in skeletal muscle of older individuals is associated with physical performance, muscle volume and mitochondrial function in the Study of Muscle, Mobility and Aging (SOMMA). Aging Cell 2024, 23, e14118. [Google Scholar] [CrossRef]
  136. Camaschella, C.; Nai, A.; Silvestri, L. Iron metabolism and iron disorders revisited in the hepcidin era. Haematologica 2020, 105, 260–272. [Google Scholar] [CrossRef]
  137. Mau, T.; Barnes, H.N.; Blackwell, T.L.; Kramer, P.A.; Bauer, S.R.; Marcinek, D.J.; Ramos, S.V.; Forman, D.E.; Toledo, F.G.S.; Hepple, R.T.; et al. Lower muscle mitochondrial energetics is associated with greater phenotypic frailty in older women and men: The Study of Muscle, Mobility and Aging. Geroscience 2024, 46, 2409–2424. [Google Scholar] [CrossRef]
  138. Cummings, S.R.; Lui, L.Y.; Glynn, N.W.; Mau, T.; Cawthon, P.M.; Kritchevsky, S.B.; Coen, P.M.; Goodpaster, B.; Marcinek, D.J.; Hepple, R.T.; et al. Energetics and clinical factors for the time required to walk 400 m: The Study of Muscle, Mobility and Aging (SOMMA). J. Am. Geriatr. Soc. 2024, 72, 1035–1047. [Google Scholar] [CrossRef]
  139. Gonzalez-Freire, M.; Scalzo, P.; D’Agostino, J.; Moore, Z.A.; Diaz-Ruiz, A.; Fabbri, E.; Zane, A.; Chen, B.; Becker, K.G.; Lehrmann, E.; et al. Skeletal muscle ex vivo mitochondrial respiration parallels decline in vivo oxidative capacity, cardiorespiratory fitness, and muscle strength: The Baltimore Longitudinal Study of Aging. Aging Cell 2018, 17, e12725. [Google Scholar] [CrossRef]
  140. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef]
  141. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. Hallmarks of aging: An expanding universe. Cell 2023, 186, 243–278. [Google Scholar] [CrossRef] [PubMed]
  142. Liang, C.; Mickey, M.C.; Receno, C.N.; Atalay, M.; DeRuisseau, K.C. Functional and biochemical responses of skeletal muscle following a moderate degree of systemic iron loading in mice. J. Appl. Physiol. (1985) 2019, 126, 799–809. [Google Scholar] [CrossRef] [PubMed]
  143. Stahl-Gugger, A.; de Godoi Rezende Costa Molino, C.; Wieczorek, M.; Chocano-Bedoya, P.O.; Abderhalden, L.A.; Schaer, D.J.; Spahn, D.R.; Orav, E.J.; Vellas, B.; da Silva, J.A.P.; et al. Prevalence and incidence of iron deficiency in European community-dwelling older adults: An observational analysis of the DO-HEALTH trial. Aging Clin. Exp. Res. 2022, 34, 2205–2215. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Mechanisms of iron transport and mitochondrial homeostasis. Dietary heme iron and ferrous iron (Fe2+) are absorbed by duodenal enterocytes and transferred into the bloodstream. Herein, iron binds Apo-transferrin (Apo-Tf), which allows for its mobilization. For cellular iron uptake, the ferric iron (Fe3+)–Tf complex binds to Tf receptor 1 (TfR1) at the plasma membrane and is internalized via a clathrin-coated vesicle. Iron released from Apo-Tf composes the cytoplasmic pool of labile iron that can be stored bound to ferritin (Fer) or can enter the mitochondria and form iron–sulfur (Fe–S) clusters and heme required for electron transport chain complexes. In response to low intracellular iron levels, nuclear receptor coactivator 4 (NCOA4) induces ferritinophagy, which can trigger Fe2+ overload and consequently reactive oxygen species (ROS) burst. The latter triggers ferroptosis, mitochondrial damage, and fission. To preserve mitochondrial homeostasis, damaged mitochondria, depending on damage severity, can undergo mitophagy and/or release mitochondria-derived vesicle (MDV) containing oxidized proteins and nucleic acids. In the cytosol, MDVs can form multivesicular bodies (MVBs) that can either pursue endolysosomal degradation or give rise to extracellular vesicles (EVs) released outside the cell. Red-dashed arrows highlight the pathways that are more relevant to iron dyshomeostasis and alterations in mitochondrial quality control. Abbreviations: BNIP3, BCL2-interacting protein 3; CP, ceruloplasmin; DcytB, duodenal cytochrome b; DMT1, divalent metal transporter 1; FPN, ferroportin; FtMt, mitochondrial ferritin; FUNDC1, FUN14 domain-containing protein 1; HCP1, heme carrier protein 1; HEPH, hephaestin; LIP, labile iron pool; MFRN, mitoferrin; NIX, NIP-3-like protein X; PINK1, PTEN-induced kinase 1; STEAP3, six-transmembrane epithelial antigen of prostate 3; Ub, ubiquitin. Created in https://BioRender.com (accessed on 24 April 2025).
Figure 1. Mechanisms of iron transport and mitochondrial homeostasis. Dietary heme iron and ferrous iron (Fe2+) are absorbed by duodenal enterocytes and transferred into the bloodstream. Herein, iron binds Apo-transferrin (Apo-Tf), which allows for its mobilization. For cellular iron uptake, the ferric iron (Fe3+)–Tf complex binds to Tf receptor 1 (TfR1) at the plasma membrane and is internalized via a clathrin-coated vesicle. Iron released from Apo-Tf composes the cytoplasmic pool of labile iron that can be stored bound to ferritin (Fer) or can enter the mitochondria and form iron–sulfur (Fe–S) clusters and heme required for electron transport chain complexes. In response to low intracellular iron levels, nuclear receptor coactivator 4 (NCOA4) induces ferritinophagy, which can trigger Fe2+ overload and consequently reactive oxygen species (ROS) burst. The latter triggers ferroptosis, mitochondrial damage, and fission. To preserve mitochondrial homeostasis, damaged mitochondria, depending on damage severity, can undergo mitophagy and/or release mitochondria-derived vesicle (MDV) containing oxidized proteins and nucleic acids. In the cytosol, MDVs can form multivesicular bodies (MVBs) that can either pursue endolysosomal degradation or give rise to extracellular vesicles (EVs) released outside the cell. Red-dashed arrows highlight the pathways that are more relevant to iron dyshomeostasis and alterations in mitochondrial quality control. Abbreviations: BNIP3, BCL2-interacting protein 3; CP, ceruloplasmin; DcytB, duodenal cytochrome b; DMT1, divalent metal transporter 1; FPN, ferroportin; FtMt, mitochondrial ferritin; FUNDC1, FUN14 domain-containing protein 1; HCP1, heme carrier protein 1; HEPH, hephaestin; LIP, labile iron pool; MFRN, mitoferrin; NIX, NIP-3-like protein X; PINK1, PTEN-induced kinase 1; STEAP3, six-transmembrane epithelial antigen of prostate 3; Ub, ubiquitin. Created in https://BioRender.com (accessed on 24 April 2025).
Cells 14 00672 g001
Table 1. List of the main studies investigating the relationship between iron metabolism and mitochondrial homeostasis in muscle.
Table 1. List of the main studies investigating the relationship between iron metabolism and mitochondrial homeostasis in muscle.
Type of StudyMain FindingsMain Biochemical MeasuresReferences
PreclinicalElevated total iron in muscles of old rats and altered expression of genes and proteins involved in redox homeostasis and iron handlingTotal iron quantification
Gene expression assays (cDNA array and RT-PCR)
2-D proteomic analysis and mass spectrometry of 87 protein spots
Altun et al., 2007 [71]
PreclinicalElevated non-heme iron content and RNA oxidative damage in muscles of old rats, especially following hindlimb suspension for two weeksNuclei acid oxidation
Non-heme iron quantification
Hofer et al., 2008 [69]
PreclinicalElevated non-heme iron content in muscles of old rats and altered expression of proteins involved in iron handlingNon-heme iron quantification
Protein expression of TfR1, DMT1, IRP1, and ferritin subunits
Jung et al., 2008 [70]
PreclinicalElevated non-heme iron content, mtRNA oxidation, and pro-apoptotic signaling in muscles of old ratsNon-heme iron quantification
Nuclei acid oxidation
Mitochondrial transition pore opening assay
Caspase-3 and caspase-9 enzymatic activity
Seo et al., 2008 [75]
PreclinicalElevated non-heme iron content and RNA oxidative damage in muscles of old rats, which were attenuated by lifelong moderate calorie restrictionNuclei acid oxidation
Non-heme iron quantification
Xu et al., 2008 [72]
PreclinicalElevated non-heme iron content and macromolecular oxidative damage, altered expression of proteins involved in iron handling in muscles of old ratsNon-heme iron quantification
Oxidative damage to nucleic acids, lipids, and proteins
Gene and protein expression of TfR1, DMT1, ZIP14, ferroportin, hemojuvelin
Xu et al., 2012 [84]
PreclinicalElevated non-heme iron content in muscles of old rats and altered expression of proteins involved in iron handling, with no effect of systemic administration of iron chelatorNon-heme iron quantification
Gene and protein expression of TfR1, IRP1, IRP2, and ferritin light chain, ferroportin
DeRuisseau et al., 2013 [73]
PreclinicalGrowth arrest and blunted energy production in muscles via fatty acid oxidation in mice with tissue-specific inactivation of TfR1Targeted metabolomic and proteomic analysis
Enzymatic activity of aconitase
Protein expression of OXPHOS complexes and TfR1
Barrientos et al., 2015 [85]
PreclinicalElevated non-heme iron content and macromolecular oxidative damage in muscles of rats injected with iron dextranNon-heme iron quantification
Protein expression of thioredoxin, TXNIP, catalase, SOD1, SOD2, GRX2, RyR1, Calstabin 1
Protein and lipid oxidation
Enzymatic activity of GPx and SOD
Liang et al., 2019 [142]
HumanElevated total iron content and mtDNA damage, altered expression of proteins involved in redox homeostasis and iron handling in muscles of physically inactive older adultsTotal iron content
Protein expression of OGG1, ZIP8, and ZIP14
Plasma quantification of ferritin, hepcidin, IL6, and CRP
Oxidative damage to mtDNA
Picca et al., 2019 [3]
HumanLarger labile iron pool, altered expression of proteins involved in mitophagy and iron handling, increased mtDNA damage in muscles of physically inactive older adultsQuantification of labile iron pool
Protein expression of LC3BII/I and p62
Quantification of mtDNA4977
Picca et al., 2020 [4]
PreclinicalExposure to iron chelator increased mitochondria-derived vesicle secretion by myotubes knocked down for mitophagy-related proteinsmtDNA copy number
Protein expression of BNIP3, BNIP3L, DNM1L, FUNDC1, LC3B, HSP70, Flotillin-1, OXPHOS complexes
Mitochondria-derived vesicle isolation
Enzymatic activity of citrate synthase and HADH
Mitochondrial respirometry
Leermakers et al., 2020 [81]
PreclinicalActivation of ferroptosis and impaired skeletal muscle regeneration in satellite cell-specific TfR1 deletionNon-heme and total iron quantification
Gene and protein expression of ferritin, MyoD, NRF2, Pax7, PGC-1α, and transferrin
Ding et al., 2021 [109]
Abbreviations: BNIP3, Bcl-2 and adenovirus E1B 19-kDa-interacting protein 3; BNIP3L, Bcl-2/E1B 19 kDa-interacting protein 3-like protein; cDNA, complementary DNA; CRP, C-reactive protein; DMT1, divalent metal transporter 1; DNM1L, dynamin-1-like protein; FUNDC1, FUN14 domain-containing protein 1; GPx, glutathione peroxidase; GRX2, glutaredoxin 2; HADH, β-hydroxyacyl-CoA dehydrogenase; HSP70, heat shock protein 70; IL6, interleukin 6; IRP, iron regulatory protein; LC3B, microtubule-associated proteins 1A/1B light chain 3B; mtDNA, mitochondrial DNA; mtRNA, mitochondrial RNA; MyoD, myogenic differentiation 1; NRF2, nuclear respiratory factor 2; OGG1, 8-oxoguanine DNA glycosylase; OXPHOS, oxidative phosphorylation; Pax7, paired box domain 7; PGC-1α, peroxisome proliferator-activated receptor-gamma coactivator 1 alpha; RT-PCR, real-time polymerase chain reaction; RyR1, ryanodine receptor 1; SOD, superoxide dismutase; TfR1, transferrin receptor 1; TXNIP, thioredoxin-interacting protein; ZIP, Zrt/Irt-like protein.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Di Lorenzo, R.; Marzetti, E.; Coelho-Junior, H.J.; Calvani, R.; Pesce, V.; Landi, F.; Leeuwenburgh, C.; Picca, A. Iron Metabolism and Muscle Aging: Where Ferritinophagy Meets Mitochondrial Quality Control. Cells 2025, 14, 672. https://doi.org/10.3390/cells14090672

AMA Style

Di Lorenzo R, Marzetti E, Coelho-Junior HJ, Calvani R, Pesce V, Landi F, Leeuwenburgh C, Picca A. Iron Metabolism and Muscle Aging: Where Ferritinophagy Meets Mitochondrial Quality Control. Cells. 2025; 14(9):672. https://doi.org/10.3390/cells14090672

Chicago/Turabian Style

Di Lorenzo, Rosa, Emanuele Marzetti, Helio José Coelho-Junior, Riccardo Calvani, Vito Pesce, Francesco Landi, Christiaan Leeuwenburgh, and Anna Picca. 2025. "Iron Metabolism and Muscle Aging: Where Ferritinophagy Meets Mitochondrial Quality Control" Cells 14, no. 9: 672. https://doi.org/10.3390/cells14090672

APA Style

Di Lorenzo, R., Marzetti, E., Coelho-Junior, H. J., Calvani, R., Pesce, V., Landi, F., Leeuwenburgh, C., & Picca, A. (2025). Iron Metabolism and Muscle Aging: Where Ferritinophagy Meets Mitochondrial Quality Control. Cells, 14(9), 672. https://doi.org/10.3390/cells14090672

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

Article Metrics

Back to TopTop