Exploring the potential regulation of DUOX in thyroid hormone‑autophagy signaling via IGF‑1 in the skeletal muscle (Review)
- Authors:
- Published online on: December 24, 2024 https://doi.org/10.3892/br.2024.1917
- Article Number: 39
-
Copyright: © Then et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 4.0].
Abstract
1. Introduction
Skeletal muscle is an important body tissue with the largest mass in the human body, accounting for ~40% of total body weight and is the main source of protein reserves in the body (1,2). Skeletal muscle is the most flexible and plastic tissue in the human body and is responsible for carrying out its functions in daily physical activities, including movement, gestures and life activities (2,3). Skeletal muscles also serve as the primary tissue involved in energy metabolism, taking in, using and storing substrates, including glucose, lipids, and amino acids (2,3).
In its development into the tissue with the largest mass in the human body, the development of skeletal muscle is influenced by a number of factors, such as nutritional status, physical activity, exercise, injury or disease, autophagy processes and hormones, one of which is thyroid hormone (4,5). Thus, thyroid hormone deficiency has an effect on skeletal muscle and can cause muscle atrophy if it remains at low levels (6,7). The interaction of thyroid hormone with insulin-like growth factor 1 (IGF-1) and autophagy can also affect muscle development (8-11). One form of thyroid hormone deficiency can be caused by interference with thyroid hormone synthesis, such as a lack of protein enzyme dual oxidases (DUOX) in the formation process (12).
Consequently, DUOX and muscle development are related via autophagy and IGF-1. To the best of the authors' knowledge, no review or study has addressed this mechanism. The regulatory relationships that will subsequently be connected to muscle growth in relation to DUOX, thyroid hormone, IGF-1 and autophagy are covered in the present review.
2. DUOX
Dual oxidase is an enzyme that belongs to the Nox family and performs a role in the oxidation of nicotinamide adenine dinucleotide phosphate (NADPH) (13,14). The DUOX enzyme has 2 types, DUOX1 and DUOX2, whose function is mainly to produce reactive oxygen species (ROS) in various tissues such as thyroid, colon, kidney, testis, salivary glands, respiratory and lymphoid (13-15) DUOX has a major contribution in the synthesis of H2O2, a substance that has an important role in the host defense system, fertilization, embryogenesis, signal transduction, cell differentiation, cell death programs and hormone synthesis, especially thyroid hormone (16-18). During the maturation phase, additional proteins known as dual oxidase maturation factors (DUOXA1 and DUOXA2) are needed to support the production of H2O2 (19,20).
The DUOX and DUOXA genes are located next to each other in an operon-like unit and are paired with each other on the long arm of chromosome 15 (Fig. 1) (21,22). In addition to thyroid cells, the DUOXA1 and DUOXA2 genes are also expressed in human respiratory epithelial cells (DUOXA1) and salivary glands (DUOXA2), although the highest expression occurs in thyroid cells (23,24). Research on mice and zebrafish demonstrates that DUOX expression emerges only when the follicle structure is functioning optimally, specifically at the final stage of cell differentiation during thyroid embryogenesis (25,26).
DUOX then travels toward the apex of the thyroid cell and the N-linked glycosylation process occurs in the Golgi apparatus, where it changes into an active form (27,28). In the absence of DUOXA as a maturation factor for DUOX, the oxidase process is arrested in the endoplasmic reticulum and only a small amount of superoxide is detected (29). The activation of DUOX1 and DUOX2 isoform occurs through Ca2+ binding to the EF-hand motif found in the N-terminal cytoplasmic segment (30,31). Based on its sequence homology with NADPH oxidase 2 (NOX2), DUOX should produce only superoxide. However, DUOX1 and DUOX2 co-expressed with DUOXA1 and DUOXA2 produced more H2O2 (32). This difference is caused by the presence of the seventh transmembrane domain and the N-terminal peroxidase ectodomain, which showed 40% homology to thyroid peroxidase (TPO), so that the superoxide produced was directly converted into H2O2 (33-35).
In the process of forming H2O2, DUOX1 requires a second intracellular loop and the COOH terminal tail of DUOXA1, while DUOX2 requires the integrity of the NH2 terminal end of DUOXA2(36) DUOX2 will produce superoxide if paired with DUOXA1 or DUOXA2, which has changes at the NH2 terminal end (36-38). The expression level of DUOX2 is fivefold higher than that of DUOX1, although both are produced at the same site (34,35,39).
Thyroid stimulating hormone (TSH), via cAMP transmission, significantly regulates DUOX2 mRNA transcription in dog and pig thyrocytes (14,40). A study conducted in mice showed an autoregulatory mechanism by thyroglobulin (Tg) that suppresses DUOX2 and DUOXA2 mRNA to control thyroid hormone synthesis (41). In addition to TSH, increasing intracellular H2O2 concentration may affect DUOX function (27,33,42). Excess amounts of iodide (I-) will inhibit the production of H2O2, which causes a decrease in TPO activity and reduced incorporation of I- into Tg. This effect is called ‘Wolf-Chaikof effect’ (42-45).
3. Modulation of DUOX protein expression in thyroid hormone formation
The formation of thyroid hormone (Fig. 2) requires I- as a basic ingredient. I- is taken from the blood vessel circulation by thyrocytes through the Na/I transporter, which is located in the basolateral plasma membrane of the cell (46). This intracellular Na+ gradient is maintained by Na+/K+-ATPase (17) I- is then transferred into the lumen of the follicle via the chlorine channel CIC5 and pendrin, which are transporters that do not depend on sodium (17). The I- is then catalyzed by the TPO enzyme using H2O2 as an oxidizer produced by the DUOX/DUOXA complex. This process is called ‘iodide organification’ and is directly sent into the colloid cells (17,47). Studies examining mutations in the DUOX2 gene show a defect in I- organization in two unrelated families, causing congenital goiter hypothyroidism (48,49). This indicates that the DUOX protein regulates thyroid hormones through its production as an oxidizer during the formation of thyroid hormones.
Tyrosine in Tg produced in follicular cells by the Golgi complex is transported into colloids through the process of exocytosis and binds to the resulting iodotyrosine residue, which ultimately forms iodinated Tg (TgI). TgI then forms a complex with proteolytic cleavage to produce monoiodotyrosine, diiodotyrosine, 3,5,3'-triiodothyronine (T3); and 3,5,3',5'-tetraiodothyronine (T4/thyroxin), all of which will be stored in colloids and released when needed (17,47). When the stimulus to release hormones is received, thyrosite will engulf some of the colloid, form an endocytosis and proteolysis process assisted by lysosomes and then separate Tg from T3 and T4, which will then be transported by monocarboxylate transporters and diffuse into the blood (17,47,50). It has been reported that mice with a double knockout of the DUOXA1/DUOXA2 gene show that the loss of DUOX cells resulted in hypothyroidism and a decrease in H2O2 levels due to disruption of T4 production in the thyroid follicles (51,52).
4. Modulation of thyroid hormone levels with IGF-1
Thyroid hormones, particularly T3, have a direct effect on the pituitary gland and regulate the secretion of growth hormone (GH). This direct influence operates through thyroid hormone receptors found in the somatotroph cells of the anterior pituitary gland. Once T3 binds to these receptors, it can adjust the transcription of the GH gene, thereby affecting the synthesis and release of GH. Maintaining appropriate thyroid hormone levels is crucial for normal GH production, as evidenced by decreased GH mRNA levels in the pituitary gland and reduced GH secretion in hypothyroidism cases. Conversely, administering thyroid hormone under hypothyroid conditions can restore GH secretion to normal levels, underscoring the necessity of adequate thyroid hormone levels for regular GH synthesis and release (53,54).
Thyroid hormones also indirectly affect GH secretion by affecting the hypothalamus. The hypothalamus synthesizes two key hormones, growth hormone-releasing hormone (GHRH), which prompts GH secretion and somatostatin, which inhibits GH secretion. Thyroid hormones can regulate the release of hypothalamic hormones, thus indirectly influencing GH secretion. For instance, thyroid hormones can increase the production of GHRH in the hypothalamus, resulting in increased GH release from the pituitary gland (54,55). The amount of thyroid hormone circulating in the blood is related to an increase in IGF-1, a polypeptide that shares structural similarities with human pro-insulin and is an essential hormone for the growth and development of the body. The primary source of GH production is the liver and is triggered by GH secreted by the anterior pituitary gland (56). The bioavailability and physiological effects of IGF-1 are controlled by a set of proteins called IGF-binding proteins (IGFBP) that are secreted. These proteins have a strong affinity for IGF-1 and serve as transporters of circulating IGF-1(57).
Thyroid hormone can modulate GH and subsequently affect the production of IGF-1. Some studies have mentioned that hypothyroidism leads to decreased GH levels and consequently lower IGF-1 levels, whereas replacement therapy with thyroid hormone can elevate IGF-1 levels (58,59). Another study found that patients with hyperthyroidism have higher serum IGF-1 levels than those with euthyroidism (60). On the other hand, previous studies have shown that not all effects of thyroid hormones on the IGF-1 pathway are mediated through GH. Thyroid hormones directly affect the transcription of the IGF-1 gene. Specifically, triiodothyronine can attach to thyroid hormone receptors, which function as transcription factors. These thyroid hormone receptors can subsequently bind to thyroid hormone response elements situated in the promoter region of IGF-1, resulting in altered IGF-1 mRNA. This direct regulation of transcription occurs in different tissues, such as the liver and bone, where IGF-1 plays a crucial role in growth and development (60-62).
Thyroid hormones can also adjust intracellular signaling pathways of IGF-1 intracellularly. They can regulate the expression of IGF-1 receptors (IGF-1R) on target cells, thus influencing the sensitivity and responsiveness of these cells to IGF-1. Furthermore, thyroid hormones can affect the production of IGFBP, which controls the availability and function of IGF-1 by binding to it in the bloodstream. Variations in IGFBP levels can change the quantity of unbound or free IGF-1 that can be attached to its receptor to produce its effects (58,60,61). One study showed that T4 replacement therapy increased serum IGFBP1 levels. In hypothyroid animals, serum IGFBP3 and IGFBP4 levels are reduced, and thyroid hormone replacement can correct these changes. Patients who undergo thyroidectomy and have their thyroid hormone replacement discontinued experience a decrease in the levels of circulating IGFBP1. Treatment with thyroxine raises these levels (58). These findings highlight the complex relationship between thyroid hormone levels and the IGF-1 pathway, suggesting that thyroid hormones may influence the activity of IGF-1 and related pathways in multiple ways.
5. Modulation of IGF-1 expression in autophagy
Research has revealed that IGF-1 signaling controls autophagy in a bidirectional manner (63). Autophagy is a widely occurring recycling process in which cellular material, including organelles, is taken up by membrane-bound vacuoles referred to as autophagosomes and transported to lysosomes and are degraded by the lysosomal compartment's store of proteolytic enzymes. This process is essential for maintaining cell, tissue, and organism homeostasis (64,65).
As an antagonist of autophagy (Fig. 3), IGF-1 binds tightly to the α subunit of IGF-1R, a type 2 tyrosine kinase membrane receptor, thereby exerting cell proliferation, differentiation and survival (66,67). This binding initiates the phosphorylation of tyrosine residues and subsequent activation of intrinsic tyrosine kinase activity of the β subunit. This further causes adaptor proteins, including insulin receptor substrate 1 and 2 (IRS1/2) and Src homology 2 domain containing E (SHE), to become phosphorylated (68). Consequently, they initiate a cascade of reactions involving various intracellular signaling pathways. These reactions ultimately modulate the expression of genes associated with cell proliferation, autophagy, or apoptosis, primarily through the PI3k/AKT/mTOR and Ras-Raf-ERK1/2 (MAPK) pathway (67,69,70). An adaptor protein, Src homology and Collagen (Shc), binds with son-of-sevenless and growth factor receptor-bound protein 2 to activate RAS, whereas IRS1/2 interacts with the p85 regulatory subunit to activate PI3K (66-68).
The PI3K/AKT/mTOR activation pathway involves the phosphorylation of phosphatidylinositol 4,5-bisphosphate (PIP2), a lipid protein, to phosphatidylinositol 3,4,5-trisphosphate (PIP3) by the PI3K kinase subunits p85 and p110. PIP3 signaling proteins, such as phosphoinositide-dependent kinase-1 (PDK1), then activate AKT, which in turn suppresses serine and threonine residues on its targets, including glycogen synthase kinase-3 beta (GSK-3β) and Tuberous sclerosis complex 1/Tuberous sclerosis complex 2 (TSC1/TSC2) (67,71). Inactive GSK-3β and TSC1/TSC2 prevent their inhibition, activating the small G protein Ras homolog enriched in the brain (Rheb) that binds with GTP. GTP-bound Rheb activates mTOR-complex 1 (mTORC1) at the lysosomal surface by binding to specific domains, including N-heat, M-heat, and the focal adhesion targeting domain. This binding allosterically modulates ATP binding at the active site, facilitating subsequent phosphorylation events (67,72).
Through the MAPK pathway, Shc/Ras/Raf/MEK modulates ERK1/2 and phosphorylates and inhibits the TSC complex, thus activating mTORC1. Subsequently, this activation affects downstream effectors such as ribosomal S6 kinase (S6K), Eukaryotic translation initiation factor 4E-binding protein 1 (4EBP1) and Unc-51-like kinase (ULK) 1/2, which in turn control processes such as autophagy inhibition, delayed apoptosis, protein synthesis, cell survival and proliferation (73). Several studies have found that changes in IGF-1 levels alter autophagy (74,75) Renna et al (76) found that IGF-1R knockdown reduced LC3-II levels in HeLa cells grown in normal media. In addition, IGF-1R knockdown reduced autophagosome formation in mouse embryonic fibroblasts derived from hemizygous IGF-1R mice. In another study, it was found that an increase in IGF-1 was accompanied by an increase in Beclin1, ULK1, and autophagy-related 5 (Atg5), which are markers of autophagy (77).
Conversely, IGF-1 promotes mitophagy (Fig. 4). Following IGF-1 signaling stimulation, there is a notable increase in the expression of peroxisome proliferator-activated receptor-gamma coactivator (PGC)-1alpha. This results in increased levels of cytochrome c oxidase subunit 7A1 (COX7A1), transcription factor B1, mitochondrial (TFB1M), and mtDNA, which improve mitochondrial function (78). In addition, IGF-1 also plays a critical role in mitochondrial biogenesis and turnover through the upregulation of factors such as PGC-1β and PGC-1-related coactivator, as well as the expression of the mitophagy receptor Bcl2/adenovirus E1B 19 kDa protein-interacting protein 3 (BNIP3) (79,80) BNIP3 contains LC3-interacting regions domains that enable interaction with LC3II in autophagosomes. Through AKT-mediated inhibitory phosphorylation of serine 9 of GSK-3β, IGF-1 stimulates the expression of the mitophagy receptor BNIP3, which in turn activates nuclear factor erythroid 2-related factor 2 (Nrf2) and the downstream transcriptional regulators Nrf1 and hypoxia-inducible factor 1-alpha (HIF-1α), implying that BNIP3 is a secondary target gene of Nrf2. Therefore, IGF-1 signaling ensures mitochondrial homeostasis and promotes tumor growth by linking mitochondrial biogenesis to basal levels of mitochondrial turnover through Nrf2 and BNIP3 (67,79).
The bidirectional effects of IGF-1 are context-dependent and are influenced by various factors, including the cellular context and situations. Under normal conditions, where cellular growth and proliferation are prioritized, IGF-1 signaling inhibits autophagy (63,67,69,81). Nonetheless, it stimulates autophagy and mitophagy in pathological situations where cellular recycling processes are required, such as energy deficiencies, starvation, hypoxia and cancer (78-80,82).
6. Modulation of thyroid hormone in autophagy
Muscle development can be directly influenced by thyroid hormones through several pathways and one of them is the autophagy pathway (83) thyroid hormones require autophagy to regulate lipid homeostasis and mitochondrial quality control in the liver (84,85). To date, research data are insufficient to explain the role of thyroid hormones in autophagy.
Lesmana et al (86) revealed that thyroid hormone induces autophagy through 5' AMP-activated protein kinase (AMPK) activation by increasing its phosphorylation, inhibiting mTOR signaling as demonstrated by a decrease in mTOR phosphorylation and increasing mRNA and protein expression in LC3, p62 and Ulk1 found in skeletal muscle, which is the key autophagy initiator (86). On the other hand, studies by Kurashige et al (87) discovered that T4 suppressed autophagy by decreasing LC3 and increasing p62. Conversely, the authors discovered that TSH increased the process of autophagy through the cAMP-PKA-cAMP response element binding protein/ERK and PKC signaling pathways (87). These disparities in the data suggest that more investigation is required to fully understand the effects of thyroid hormones on autophagy.
7. Modulation of thyroid hormone levels in skeletal muscle growth
Thyroid hormone has a major role in the growth, regeneration and differentiation of skeletal muscle through the induction of autophagy, which involves the stimulation of ROS of AMPK and mTOR-ULK1 signaling (86). Thyroid hormone can also trigger changes in the muscle fiber profile, such as the loss of embryonic and neonatal myosin and increase in fast or slow myosin genes in certain muscles (88). In addition, rats with hypothyroidism exhibit delayed transition to adult myosin in their fast muscles but not in their slow muscles (89-91).
Weight-bearing exercise and electrical stimulation are essential for the postnatal growth of slow fibers, whereas T3 signaling is critical for the development of fast fibers, particularly for the conversion of neonatal fiber to fiber IIb (89,91-93). The typical pattern of fiber dispersion in every muscle is determined in part by the physiological levels of thyroid hormone (92,94). Thyroid hormone, especially T3, induces muscle contractions to become faster in rats by increasing the expression of myosin heavy chain (MYH)2, MYH 1, MYH 4, fibers IIa, IIx, and IIb; and suppressing the expression of MYH7 and myosin from fiber type I. Furthermore, T3 promotes the conversion of muscle fiber types from slow to fast by causing changes from MYH7 to MYH2, MYH2 to MYH1, and MYH1 to MYH4 (95,96). Triiodothyronine induces miR-133a expression in fast-twitch muscles and also induces slow-to-fast muscle fiber transition (97). Furthermore, mice with miR-133a deletion exhibit a fast-to-slow muscle transition (98).
Muscle growth can be directly regulated by T3 hormone, which stimulates signals to myoblast determination protein (MYOD)1, a protein that regulates the transcription process during myogenesis (5). Moreover, MYOD1 stimulates muscle satellite cells to differentiate into myoblasts and myotubes. Furthermore, myogenin in immature myotubes and myosin heavy chain (MYH) in mature myotubes are two additional mechanisms that directly affect muscle development and function caused by T3 (5,99).
8. Modulation of autophagy in skeletal muscle growth
Autophagy provides the fundamental components for metabolism and cellular renewal (11,100,101). Additionally, autophagy controls intracellular quality control, which aids in the breakdown of defective proteins and basal protein turnover (102). Inhibition of autophagy causes the aggregation of ubiquitin proteins and inclusion bodies in various types of cells, and the abnormalities can also occur in mitochondria, peroxisomes, the endoplasmic reticulum, and Golgi bodies (103-105). Studies using Atg7 knockout mice and focused on muscles that presented abnormal concentric membranous formations, reticulum distension, disordered sarcomere and aberrant mitochondria. The Atg7 knockout mice displayed muscular phenotypes such myopathy's morphological characteristics, muscle atrophy and degeneration under catabolic environments, this showed how autophagy provides benefits for preserving the integrity of myofiber and muscle mass (104,105). Additionally, an Atg16L hypomorph mouse model showed reduced autophagy flux but still present and impaired muscle fiber development and generation (105,106).
Degeneration and atrophy of the muscles are caused by impaired autophagy (105,107). However, excessive autophagy can also cause atrophy, which is associated with mutations in the laminin α2 chain, which cause muscle fibrosis, atrophy and apoptotic phenotypes as well as an increase in the expression of genes related to autophagy (108-110).
Autophagy plays an important role in skeletal muscle regeneration due to its ability to regenerate muscle stem cells/satellite cells by maintaining a state of quiescence and preventing aging (111,112). In senescent muscle stem cells, the phosphorylation of AMPK and its downstream target P27Kip1 is reduced and the accompanying stress of inhibited autophagy renders muscle stem cells more susceptible to apoptosis (113). Autophagy helps prevent aging by clearing the autophagosome and providing an energy source for activation (111,112). Failure of autophagy in satellite cells will cause aging, oxidative stress and mitochondrial dysfunction, as well as accumulation of organelles and proteins, but satellite cells are not the cause of muscle fiber hypertrophy because satellite cells are only needed for the de novo formation of new fibers (114-118). These results show that the decrease in the number of satellite cells may not be due to atrophy.
Previous studies have reported that the autophagy process occurs throughout the entire myoblast differentiation cycle, so it can be concluded that there is a two-way process between autophagy and muscle cell differentiation (112,119-121). There is a relationship with thyroid hormone, where T3 is needed for the process of differentiation and fusion of myoblasts, which will later trigger upregulation of autophagy (122). Disruption of the autophagy process, such as knockdown of Atg5 and Atg7, affects myogenesis, which is followed by mitochondrial dysfunction (121).
The differentiation of primitive myoblasts into mature myotubes necessitates a metabolic change to meet the increasing energy demand, which involves mitochondrial renewal, which has been proven to be an essential step (123,124). In this situation, autophagy plays an important role, as evidenced by an increase in mitophagy prior to a rise in the amount of mitochondrial proteins at the beginning of the mitochondrial renewal process during myogenic differentiation (124).
Autophagy is required in the myoblast differentiation process due to its relationship with signaling preventing the apoptosis process (125,126). Inhibition of Atg7 enhanced transient caspase 3 activation, DNA fragmentation and the proportion of apoptotic nuclei (125). In addition, mitophagy also has the function of removing damaged mitochondria prior to apoptotic signaling, reducing cell stress and death (126). Notably, the increased ROS levels induced by cellular stress are also required for skeletal muscle development and inhibition of mitochondrial ROS production leads to the failure of myoblast differentiation (127,128). Furthermore, DUOX, which is a member of the NADPH family, may produce ROS, leading to the hypothesis that there is a link between DUOX gene and skeletal muscle growth.
9. Modulation of DUOX, thyroid hormone, IGF-1, and autophagy in skeletal muscle growth
From the various interactions, Fig. 5 summarizes of the relationships among DUOX, thyroid hormone, IGF-1 and autophagy in skeletal muscle. The DUOX gene generates H2O2, which is an oxidant that aids in the production of thyroid hormone (17,18). Thyroid hormones can promote autophagy by inhibiting mTOR and increasing the production of AMPK (86). Nevertheless, another study claims that thyroid hormones, particularly T4, have an effect and that they directly inhibit the autophagy process (87). In turn, autophagy recycles damaged organelles and macromolecules and the byproducts of this degradation are used by muscle cells for their development, differentiation and regeneration (105,107,121,125,129). Moreover, thyroid hormone also affects autophagy and the development of muscle cells in another pathway by increasing GH and IGFBP through IGF-1 (58-60). Furthermore, IGF-1 raises At, which then cascades down into two pathways: Upregulating mTOR, which inhibits autophagy and downregulating GSK-3β, which increases autophagy, especially mitophagy (67,73). Despite the opposing effects on autophagy, both of these processes directly enhance muscle development. Thyroid hormone, particularly T3, can directly affect muscle growth and differentiation by raising MYOD1, myogenin, and MYH (5,99). Although several pathways still require further research, such as the relationship between DUOX, autophagy and muscle growth, it si hypothesized that there is a relationship between these components that can be explained through available research.
To the best of the authors' knowledge, DUOX research on autophagy and cell or tissue growth is currently limited to smooth muscle organs such as the respiratory system. A study using mice with inflammation in their lungs showed that autophagy regulates the increase in superoxide levels by directing DUOX1 to the apical surface of the airway epithelium (130). Another similar study in Drosophila showed the activation of the DUOX gene downstream of autophagy by activating the ATG1-dependent lipophagy pathway, which is required for tumor necrosis factor receptor-associated factor 3 (TRAF3)-AMPK/Warts gene (WTS)-pathway-induced DUOX activation (131,132). Additionally, NOX, which is in the same family as DUOX, produces ROS, which affects autophagy. The exact methods by which ROS trigger autophagy remain unclear. For example, by directly oxidizing parts of the autophagic machinery, this activation serves as a compensatory and survival mechanism to reduce cellular death caused by excess ROS (133,134). Another study reported that ROS produced from NOX activates autophagy by stimulating the protein kinase RNA-like endoplasmic reticulum kinase signaling pathway, which in turn enhances the activation of autophagy and survival in cardiomyocytes in response to food restriction and ischaemia (135).
Hypothetically, ROS produced by the DUOX gene has a similar effect to NOX in directly regulating autophagy, where autophagy is triggered when the cell is under stress (136-142) or, in the other words, DUOX and autophagy have a reciprocal relationship (143). Furthermore, ROS has an effect on muscle tissue development and it is hypothesized that DUOX has a direct effect on muscle development via ROS production. Skeletal muscle formation depends on elevated ROS levels brought on by cellular stress, and inhibition of mitochondrial ROS synthesis impairs myoblast differentiation (127,128). It is also hypothesized that IGF-1 promotes muscle growth by upregulating DUOX. However, no studies have specifically investigated this relationship, and the direct interactions are largely unknown.
10. Conclusion
In conclusion, the DUOX gene has several benefits in various life processes, including muscular development. By understanding the function of DUOX, clinicians will be able to diagnose and administer appropriate treatment if a disorder occurs in this gene. DUOX is an enzyme that can produce H2O2, which is needed for thyroid hormone production. In addition, thyroid hormone can trigger muscle growth directly and indirectly through the IGF-1 signaling pathway. IGF-1 will later have a bidirectional effect on the autophagy process. Autophagy itself is a process necessary for muscle development. In this pathway, the effect of DUOX on skeletal muscle growth is unclear. It was hypothesized that there is a direct relationship between DUOX and IGF-1, autophagy and muscle development. Therefore, further studies are required to provide new insights into the influence of DUOX on skeletal muscle growth through IGF-1 signaling.
Acknowledgements
The authors are grateful to Dr Aurelia Angelica Erawan, Dr Ivana Iman Santosa, and Dr Gladys Danielle Novianto (Universitas Padjadjaran, Bandung, Indonesia) for the insightful suggestions and constructive feedback on the manuscript and to Dr Audrey Averina Santoso, Dr Cici Pratama Dea Mantong, and Dr Jody Garcia Hartanto Sitorus (Universitas Padjadjaran, Bandung, Indonesia) who provided assistance in the editing of images of the current paper.
Funding
Funding: The present study was supported by the Ministry of Education, Culture, Research and Technology of Indonesia through the Fundamental Research-Regular grant no. 3018/UN6.3.1/PT.00/2023 to RL.
Availability of data and materials
Not applicable.
Authors' contributions
AAT completed the first draft of the manuscript. HG and RL proposed ideas. HG, RL and AC reviewed and edited the manuscript. NS and JWG analyzed the data and revised the manuscript. All authors have read and approved the final manuscript. Data authentication is not applicable.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Liang JL, Xie JF, Wang CY and Chen N: Regulatory roles of microRNAs in sarcopenia and exercise intervention. Sheng Li Xue Bao. 72:667–676. 2020.PubMed/NCBI(In Chinese). | |
Evans WJ: Skeletal muscle loss: Cachexia, sarcopenia, and inactivity. Am J Clin Nutr. 91:1123S–1127S. 2010.PubMed/NCBI View Article : Google Scholar | |
Frontera WR and Ochala J: Skeletal muscle: A brief review of structure and function. Calcif Tissue Int. 96:183–195. 2015.PubMed/NCBI View Article : Google Scholar | |
Xia Q, Huang X, Huang J, Zheng Y, March ME, Li J and Wei Y: The role of autophagy in skeletal muscle diseases. Front Physiol. 12(638983)2021.PubMed/NCBI View Article : Google Scholar | |
Muscat GE, Mynett-johnson L, Dowhan D, Downes M and Griggs R: Activation of myoD gene transcription by 3,5,3'-triiodo-L-thyronine: A direct role for the thyroid hormone and retinoid X receptors. Nucleic Acids Res. 22:583–591. 1994.PubMed/NCBI View Article : Google Scholar | |
Brent GA: Mechanisms of thyroid hormone action. J Clin Invest. 122:3035–3043. 2012.PubMed/NCBI View Article : Google Scholar | |
Yu F, Göthe S, Wikström L, Forrest D, Vennström B and Larsson L: Effects of thyroid hormone receptor gene disruption on myosin isoform expression in mouse skeletal muscles. Am J Physiol Regul Integr Comp Physiol. 278:R1545–R1554. 2000.PubMed/NCBI View Article : Google Scholar | |
Grosvenor CE and Turner CW: Effect of growth hormone upon thyroid secretion rate in the rat. Proc Soc Exp Biol Med. 100:70–72. 1959.PubMed/NCBI View Article : Google Scholar | |
Saji M, Tsushima T, Isozaki O, Murakami H, Ohba Y, Sato K, Arai M, Mariko A and Shizume K: Interaction of insulin-like growth factor I with porcine thyroid cells cultured in monolayer. Endocrinology. 121:749–756. 1987.PubMed/NCBI View Article : Google Scholar | |
Malaguarnera R, Frasca F, Garozzo A, Gianì F, Pandini G, Vella V, Vigneri R and Belfiore A: Insulin receptor isoforms and insulin-like growth factor receptor in human follicular cell precursors from papillary thyroid cancer and normal thyroid. J Clin Endocrinol Metab. 96:766–774. 2011.PubMed/NCBI View Article : Google Scholar | |
Kang C, You NJ and Avery L: Dual roles of autophagy in the survival of Caenorhabditis elegans during starvation. Genes Dev. 21:2161–2171. 2007.PubMed/NCBI View Article : Google Scholar | |
Carvalho DP and Dupuy C: Role of the NADPH oxidases DUOX and NOX4 in thyroid oxidative stress. Eur Thyroid J. 2:160–167. 2013.PubMed/NCBI View Article : Google Scholar | |
Donkó Á, Péterfi Z, Sum A, Leto T and Geiszt M: Dual oxidases. Philos Trans R Soc Lond B Biol Sci. 360:2301–2308. 2005.PubMed/NCBI View Article : Google Scholar | |
Dupuy C, Ohayon R, Valent A, Noël-Hudson MS, Dème D and Virion A: Purification of a novel flavoprotein involved in the thyroid NADPH oxidase. Cloning of the porcine and human cdnas. J Biol Chem. 274:37265–37269. 1999.PubMed/NCBI View Article : Google Scholar | |
Geiszt M and Leto TL: The Nox family of NAD(P)H oxidases: Host defense and beyond. J Biol Chem. 279:51715–51718. 2004.PubMed/NCBI View Article : Google Scholar | |
Conner GE: Regulation of dual oxidase hydrogen peroxide synthesis results in an epithelial respiratory burst. Redox Biol. 41(101931)2021.PubMed/NCBI View Article : Google Scholar | |
Szanto I, Pusztaszeri M and Mavromati M: H2O2 metabolism in normal thyroid cells and in thyroid tumorigenesis: Focus on NADPH oxidases. Antioxidants (Basel). 8(126)2019.PubMed/NCBI View Article : Google Scholar | |
Korzeniowska A, Donkó ÁP, Morand S and Leto TL: Functional characterization of DUOX enzymes in reconstituted cell models. Methods Mol Biol. 1982:173–190. 2019.PubMed/NCBI View Article : Google Scholar | |
Grasberger H and Refetoff S: Identification of the maturation factor for dual oxidase. Evolution of an eukaryotic operon equivalent. J Biol Chem. 281:18269–18272. 2006.PubMed/NCBI View Article : Google Scholar | |
Hulur I, Hermanns P, Nestoris C, Heger S, Refetoff S, Pohlenz J and Grasberger H: A single copy of the recently identified dual oxidase maturation factor (DUOXA) 1 gene produces only mild transient hypothyroidism in a patient with a novel biallelic DUOXA2 mutation and monoallelic DUOXA1 deletion. J Clin Endocrinol Metab. 96:E841–E851. 2011.PubMed/NCBI View Article : Google Scholar | |
Xu C, Linderholm A, Grasberger H and Harper RW: Dual oxidase 2 bidirectional promoter polymorphisms confer differential immune responses in airway epithelia. Am J Respir Cell Mol Biol. 47:484–490. 2012.PubMed/NCBI View Article : Google Scholar | |
Christophe-Hobertus C and Christophe D: Delimitation and functional characterization of the bidirectional THOX-DUOXA promoter regions in thyrocytes. Mol Cell Endocrinol. 317:161–167. 2010.PubMed/NCBI View Article : Google Scholar | |
Luxen S, Belinsky SA and Knaus UG: Silencing of DUOX NADPH oxidases by promoter hypermethylation in lung cancer. Cancer Res. 68:1037–1045. 2008.PubMed/NCBI View Article : Google Scholar | |
Grasberger H, De Deken X, Miot F, Pohlenz J and Refetoff S: Missense mutations of dual oxidase 2 (DUOX2) implicated in congenital hypothyroidism have impaired trafficking in cells reconstituted with DUOX2 maturation factor. Mol Endocrinol. 21:1408–1421. 2007.PubMed/NCBI View Article : Google Scholar | |
Milenkovic M, De Deken X, Jin L, De Felice M, Di Lauro R, Dumont JE, Corvilain B and Miot F: Duox expression and related H2O2 measurement in mouse thyroid: Onset in embryonic development and regulation by TSH in adult. J Endocrinol. 192:615–626. 2007.PubMed/NCBI View Article : Google Scholar | |
Opitz R, Maquet E, Zoenen M, Dadhich R and Costagliola S: TSH receptor function is required for normal thyroid differentiation in zebrafish. Mol Endocrinol. 25:1579–1599. 2011.PubMed/NCBI View Article : Google Scholar | |
De Deken X, Wang D, Dumont JE and Miot F: Characterization of ThOX proteins as components of the thyroid H(2)O(2)-generating system. Exp Cell Res. 273:187–196. 2002.PubMed/NCBI View Article : Google Scholar | |
Raad H, Eskalli Z, Corvilain B, Miot F and De Deken X: Thyroid hydrogen peroxide production is enhanced by the Th2 cytokines, IL-4 and IL-13, through increased expression of the dual oxidase 2 and its maturation factor DUOXA2. Free Radic Biol Med. 56:216–225. 2013.PubMed/NCBI View Article : Google Scholar | |
El Hassani RA, Benfares N, Caillou B, Talbot M, Sabourin JC, Belotte V, Morand S, Gnidehou S, Agnandji D, Ohayon R, et al: Dual oxidase2 is expressed all along the digestive tract. Am J Physiol Gastrointest Liver Physiol. 288:G933–G942. 2005.PubMed/NCBI View Article : Google Scholar | |
Rigutto S, Hoste C, Grasberger H, Milenkovic M, Communi D, Dumont JE, Corvilain B, Miot F and De Deken X: Activation of dual oxidases Duox1 and Duox2: differential regulation mediated by camp-dependent protein kinase and protein kinase C-dependent phosphorylation. J Biol Chem. 284:6725–6734. 2009.PubMed/NCBI View Article : Google Scholar | |
Ameziane-El-Hassani R, Schlumberger M and Dupuy C: NADPH oxidases: New actors in thyroid cancer? Nat Rev Endocrinol. 12:485–494. 2016.PubMed/NCBI View Article : Google Scholar | |
Lambeth JD: Nox enzymes, ROS, and chronic disease: An example of antagonistic pleiotropy. Free Radic Biol Med. 43:332–347. 2007.PubMed/NCBI View Article : Google Scholar | |
Song Y, Ruf J, Lothaire P, Dequanter D, Andry G, Willemse E, Dumont JE, Van Sande J and De Deken X: Association of duoxes with thyroid peroxidase and its regulation in thyrocytes. J Clin Endocrinol Metab. 95:375–382. 2010.PubMed/NCBI View Article : Google Scholar | |
Ameziane-El-Hassani R, Morand S, Boucher JL, Frapart YM, Apostolou D, Agnandji D, Gnidehou S, Ohayon R, Noël-Hudson MS, Francon J, et al: Dual oxidase-2 has an intrinsic Ca2+-dependent H2O2-generating activity. J Biol Chem. 280:30046–30054. 2005.PubMed/NCBI View Article : Google Scholar | |
Caillou B, Dupuy C, Lacroix L, Nocera M, Talbot M, Ohayon R, Dème D, Bidart JM, Schlumberger M and Virion A: Expression of reduced nicotinamide adenine dinucleotide phosphate oxidase (ThoX, LNOX, Duox) genes and proteins in human thyroid tissues. J Clin Endocrinol Metab. 86:3351–3358. 2001.PubMed/NCBI View Article : Google Scholar | |
Hoste C, Dumont JE, Miot F and De Deken X: The type of DUOX-dependent ROS production is dictated by defined sequences in DUOXA. Exp Cell Res. 318:2353–2364. 2012.PubMed/NCBI View Article : Google Scholar | |
Zamproni I, Grasberger H, Cortinovis F, Vigone MC, Chiumello G, Mora S, Onigata K, Fugazzola L, Refetoff S, Persani L and Weber G: Biallelic inactivation of the dual oxidase maturation factor 2 (DUOXA2) gene as a novel cause of congenital hypothyroidism. J Clin Endocrinol Metab. 93:605–610. 2008.PubMed/NCBI View Article : Google Scholar | |
Morand S, Ueyama T, Tsujibe S, Saito N, Korzeniowska A and Leto TL: Duox maturation factors form cell surface complexes with Duox affecting the specificity of reactive oxygen species generation. FASEB J. 23:1205–1218. 2009.PubMed/NCBI View Article : Google Scholar | |
Pachucki J, Wang D, Christophe D and Miot F: Structural and functional characterization of the two human ThOX/Duox genes and their 5'-flanking regions. Mol Cell Endocrinol. 214:53–62. 2004.PubMed/NCBI View Article : Google Scholar | |
De Deken X, Wang D, Many MC, Costagliola S, Libert F, Vassart G, Dumont JE and Miot F: Cloning of two human thyroid cDNAs encoding new members of the NADPH oxidase family. J Biol Chem. 275:23227–23233. 2000.PubMed/NCBI View Article : Google Scholar | |
Yoshihara A, Hara T, Kawashima A, Akama T, Tanigawa K, Wu H, Sue M, Ishido Y, Hiroi N, Ishii N, et al: Regulation of dual oxidase expression and H2O2 production by thyroglobulin. Thyroid. 22:1054–1062. 2012.PubMed/NCBI View Article : Google Scholar | |
Cardoso LC, Martins DC, Figueiredo MD, Rosenthal D, Vaisman M, Violante AH and Carvalho DP: Ca(2+)/nicotinamide adenine dinucleotide phosphate-dependent H(2)O(2) generation is inhibited by iodide in human thyroids. J Clin Endocrinol Metab. 86:4339–4343. 2001.PubMed/NCBI View Article : Google Scholar | |
Wolff J and Chaikoff IL: Plasma inorganic iodide, a chemical regulator of normal thyroid function. Endocrinology. 42:468–471. 1948.PubMed/NCBI View Article : Google Scholar | |
Carvalho DP, Dupuy C, Gorin Y, Legue O, Pommier J, Haye B and Virion HA: The Ca2+- and reduced nicotinamide adenine dinucleotide phosphate-dependent hydrogen peroxide generating system is induced by thyrotropin in porcine thyroid cells. Endocrinology. 137:1007–1012. 1996.PubMed/NCBI View Article : Google Scholar | |
Corvilain B, Van Sande J and Dumont JE: Inhibition by iodide of iodide binding to proteins: The ‘Wolff-Chaikoff’ effect is caused by inhibition of H2O2 generation. Biochem Biophys Res Commun. 154:1287–1292. 1988.PubMed/NCBI View Article : Google Scholar | |
Pochin EE: Investigation of thyroid function and disease with radioactive iodine. Lancet. 2:84–91. 1950.PubMed/NCBI View Article : Google Scholar | |
Godlewska M, Góra M, Buckle AM, Porebski BT, Kemp EH, Sutton BJ, Czarnocka B and Banga JP: A redundant role of human thyroid peroxidase propeptide for cellular, enzymatic, and immunological activity. Thyroid. 24:371–382. 2014.PubMed/NCBI View Article : Google Scholar | |
Varela V, Rivolta CM, Esperante SA, Gruñeiro-Papendieck L, Chiesa A and Targovnik HM: Three mutations (p.Q36H, p.G418fsX482, and g.IVS19-2A>C) in the dual oxidase 2 gene responsible for congenital goiter and iodide organification defect. Clin Chem. 52:182–191. 2006.PubMed/NCBI View Article : Google Scholar | |
Di Candia S, Zamproni I, Cortinovis F, Passoni A, Vigone MC, Fugazzola L, Persani L and Weber G: Congenital hypothyroidism and partial iodide organification defects: Two mutations in DUOX2 gene. Horm Res. 65(38)2006. | |
Thomas J, Sairoz Jose A, Poojari VG, Shetty S, K SP, Prabhu R V K and Rao M: Role and clinical significance of monocarboxylate transporter 8 (MCT8) during pregnancy. Reprod Sci. 30:1758–1769. 2023.PubMed/NCBI View Article : Google Scholar | |
Grasberger H, De Deken X, Mayo OB, Raad H, Weiss M, Liao XH and Refetoff S: Mice deficient in dual oxidase maturation factors are severely hypothyroid. Mol Endocrinol. 26:481–492. 2012.PubMed/NCBI View Article : Google Scholar | |
Eskalli Z, Achouri Y, Hahn S, Many MC, Craps J, Refetoff S, Liao XH, Dumont JE, Van Sande J, Corvilain B, et al: Overexpression of interleukin-4 in the thyroid of transgenic mice upregulates the expression of Duox1 and the anion transporter pendrin. Thyroid. 26:1499–1512. 2016.PubMed/NCBI View Article : Google Scholar | |
Pappa T and Refetoff S: Resistance to thyroid hormone beta: A focused review. Front Endocrinol (Lausanne). 12(656551)2021.PubMed/NCBI View Article : Google Scholar | |
Giustina A and Wehrenberg WB: Influence of thyroid hormones on the regulation of growth hormone secretion. Eur J Endocrinol. 133:646–653. 1995.PubMed/NCBI View Article : Google Scholar | |
Kamegai J, Tamura H, Ishii S, Sugihara H and Wakabayashi I: Thyroid hormones regulate pituitary growth hormone secretagogue receptor gene expression. J Neuroendocrinol. 13:275–278. 2001.PubMed/NCBI View Article : Google Scholar | |
Al-Samerria S and Radovick S: The role of insulin-like growth factor-1 (IGF-1) in the control of neuroendocrine regulation of growth. Cells. 10(2664)2021.PubMed/NCBI View Article : Google Scholar | |
Yakar S and Adamo ML: Insulin-like growth factor-1 physiology: Lessons from mouse models. Endocrinol Metab Clin North Am. 41:231–247, v. 2012.PubMed/NCBI View Article : Google Scholar | |
Smith TJ: Insulin-like growth factor pathway and the thyroid. Front Endocrinol (Lausanne). 12(653627)2021.PubMed/NCBI View Article : Google Scholar | |
Chang YJ, Hwu CM, Yeh CC, Wang PS and Wang SW: Effects of subacute hypothyroidism on metabolism and growth-related molecules. Molecules. 19:11178–11195. 2014.PubMed/NCBI View Article : Google Scholar | |
Tseng FY, Chen YT, Chi YC, Chen PL and Yang WS: Serum levels of insulin-like growth factor 1 are negatively associated with log transformation of thyroid-stimulating hormone in Graves' disease patients with hyperthyroidism or subjects with euthyroidism: A prospective observational study. Medicine (Baltimore). 98(e14862)2019.PubMed/NCBI View Article : Google Scholar | |
Smith TJ and Janssen JAMJL: Insulin-like growth factor-i receptor and thyroid-associated ophthalmopathy. Endocr Rev. 40:236–267. 2019.PubMed/NCBI View Article : Google Scholar | |
Robson H, Siebler T, Shalet SM and Williams GR: Interactions between GH, IGF-I, glucocorticoids, and thyroid hormones during skeletal growth. Pediatr Res. 52:137–147. 2002.PubMed/NCBI View Article : Google Scholar | |
Sipos F, Székely H, Kis ID, Tulassay Z and Műzes G: Relation of the IGF/IGF1R system to autophagy in colitis and colorectal cancer. World J Gastroenterol. 23:8109–8119. 2017.PubMed/NCBI View Article : Google Scholar | |
Gómez-Virgilio L, Silva-Lucero MDC, Flores-Morelos DS, Gallardo-Nieto J, Lopez-Toledo G, Abarca-Fernandez AM, Zacapala-Gómez AE, Luna-Muñoz J, Montiel-Sosa F, Soto-Rojas LO, et al: Autophagy: A key regulator of homeostasis and disease: An overview of molecular mechanisms and modulators. Cells. 11(2262)2022.PubMed/NCBI View Article : Google Scholar | |
Levine B and Kroemer G: SnapShot: Macroautophagy. Cell. 132:162.e1–162.e3. 2008.PubMed/NCBI View Article : Google Scholar | |
Liu Q, Guan JZ, Sun Y, Le Z, Zhang P, Yu D and Liu Y: Insulin-like growth factor 1 receptor-mediated cell survival in hypoxia depends on the promotion of autophagy via suppression of the PI3K/Akt/mTOR signaling pathway. Mol Med Rep. 15:2136–2142. 2017.PubMed/NCBI View Article : Google Scholar | |
Kasprzak A: Autophagy and the insulin-like growth factor (IGF) system in colonic cells: Implications for colorectal neoplasia. Int J Mol Sci. 24(3665)2023.PubMed/NCBI View Article : Google Scholar | |
Wang Z, Li W, Guo Q, Wang Y, Ma L and Zhang X: Insulin-like growth factor-1 signaling in lung development and inflammatory lung diseases. Biomed Res Int. 2018(6057589)2018.PubMed/NCBI View Article : Google Scholar | |
Gonçalves DA, Silveira WA, Manfredi LH, Graça FA, Armani A, Bertaggia E, O Neill BT, Lautherbach N, Machado J, Nogara L, et al: Insulin/IGF1 signalling mediates the effects of β2-adrenergic agonist on muscle proteostasis and growth. J Cachexia Sarcopenia Muscle. 10:455–475. 2019.PubMed/NCBI View Article : Google Scholar | |
Zhang B, Li H, Wang Y, Li Y, Zhou Z, Hou X, Zhang X and Liu T: Mechanism of autophagy mediated by IGF-1 signaling pathway in the neurotoxicity of lead in pubertal rats. Ecotoxicol Environ Saf. 251(114557)2023.PubMed/NCBI View Article : Google Scholar | |
Mercurio L, Albanesi C and Madonna S: Recent updates on the involvement of PI3K/AKT/mTOR molecular cascade in the pathogenesis of hyperproliferative skin disorders. Front Med (Lausanne). 8(665647)2021.PubMed/NCBI View Article : Google Scholar | |
Shams R, Ito Y and Miyatake H: Evaluation of the binding kinetics of RHEB with mTORC1 by in-cell and in vitro assays. Int J Mol Sci. 22(8766)2021.PubMed/NCBI View Article : Google Scholar | |
Germano CA, Clemente G, Storniolo A, Romeo MA, Ferretti E, Cirone M and Di Renzo L: mTORC1/ERK1/2 interplay regulates protein synthesis and survival in acute myeloid leukemia cell lines. Biology (Basel). 12(676)2023.PubMed/NCBI View Article : Google Scholar | |
Jia G, Cheng G, Gangahar DM and Agrawal DK: Insulin-like growth factor-1 and TNF-alpha regulate autophagy through c-jun N-terminal kinase and Akt pathways in human atherosclerotic vascular smooth cells. Immunol Cell Biol. 84:448–454. 2006.PubMed/NCBI View Article : Google Scholar | |
Ravikumar B, Vacher C, Berger Z, Davies JE, Luo S, Oroz LG, Scaravilli F, Easton DF, Duden R, O'Kane CJ and Rubinsztein DC: Inhibition of mTOR induces autophagy and reduces toxicity of polyglutamine expansions in fly and mouse models of Huntington disease. Nat Genet. 36:585–595. 2004.PubMed/NCBI View Article : Google Scholar | |
Renna M, Bento CF, Fleming A, Menzies FM, Siddiqi FH, Ravikumar B, Puri C, Garcia-Arencibia M, Sadiq O, Corrochano S, et al: IGF-1 receptor antagonism inhibits autophagy. Hum Mol Genet. 22:4528–4544. 2013.PubMed/NCBI View Article : Google Scholar | |
Yu Q, Zhao B, He Q, Zhang Y and Peng XB: microRNA-206 is required for osteoarthritis development through its effect on apoptosis and autophagy of articular chondrocytes via modulating the phosphoinositide 3-kinase/protein kinase B-mTOR pathway by targeting insulin-like growth factor-1. J Cell Biochem. 120:5287–5303. 2019.PubMed/NCBI View Article : Google Scholar | |
Guan X, Yan Q, Wang D, Du G and Zhou J: IGF-1 signaling regulates mitochondrial remodeling during myogenic differentiation. Nutrients. 14(1249)2022.PubMed/NCBI View Article : Google Scholar | |
Riis S, Murray JB and O'Connor R: IGF-1 signalling regulates mitochondria dynamics and turnover through a conserved GSK-3β-Nrf2-BNIP3 pathway. Cells. 9(147)2020.PubMed/NCBI View Article : Google Scholar | |
Lyons A, Coleman M, Riis S, Favre C, O'Flanagan CH, Zhdanov AV, Papkovsky DB, Hursting SD and O'Connor R: Insulin-like growth factor 1 signaling is essential for mitochondrial biogenesis and mitophagy in cancer cells. J Biol Chem. 292:16983–16998. 2017.PubMed/NCBI View Article : Google Scholar | |
Zecchini S, Giovarelli M, Perrotta C, Morisi F, Touvier T, Di Renzo I, Moscheni C, Bassi MT, Cervia D, Sandri M, et al: Autophagy controls neonatal myogenesis by regulating the GH-IGF1 system through a NFE2L2- and DDIT3-mediated mechanism. Autophagy. 15:58–77. 2019.PubMed/NCBI View Article : Google Scholar | |
Shan Y, Lu C, Wang J, Li M, Ye S, Wu S, Huang J, Bu S and Wang F: IGF-1 contributes to liver cancer development in diabetes patients by promoting autophagy. Ann Hepatol. 27(100697)2022.PubMed/NCBI View Article : Google Scholar | |
Dentice M, Marsili A, Ambrosio R, Guardiola O, Sibilio A, Paik JH, Minchiotti G, DePinho RA, Fenzi G, Larsen PR and Salvatore D: The FoxO3/type 2 deiodinase pathway is required for normal mouse myogenesis and muscle regeneration. J Clin Invest. 120:4021–4030. 2010.PubMed/NCBI View Article : Google Scholar | |
Sinha RA, Singh BK, Zhou J, Wu Y, Farah BL, Ohba K, Lesmana R, Gooding J, Bay BH and Yen PM: Thyroid hormone induction of mitochondrial activity is coupled to mitophagy via ROS-AMPK-ULK1 signaling. Autophagy. 11:1341–1357. 2015.PubMed/NCBI View Article : Google Scholar | |
Sinha RA, You SH, Zhou J, Siddique MM, Bay BH, Zhu X, Privalsky ML, Cheng SY, Stevens RD, Summers SA, et al: Thyroid hormone stimulates hepatic lipid catabolism via activation of autophagy. J Clin Invest. 122:2428–2438. 2012.PubMed/NCBI View Article : Google Scholar | |
Lesmana R, Sinha RA, Singh BK, Zhou J, Ohba K, Wu Y, Yau WW, Bay BH and Yen PM: Thyroid hormone stimulation of autophagy is essential for mitochondrial biogenesis and activity in skeletal muscle. Endocrinology. 157:23–38. 2016.PubMed/NCBI View Article : Google Scholar | |
Kurashige T, Nakajima Y, Shimamura M, Yamada M and Nagayama Y: Hormonal regulation of autophagy in thyroid PCCL3 cells and the thyroids of male mice. J Endocr Soc. 4(bvaa054)2020.PubMed/NCBI View Article : Google Scholar | |
Schiaffino S, Rossi AC, Smerdu V, Leinwand LA and Reggiani C: Developmental myosins: Expression patterns and functional significance. Skelet Muscle. 5(22)2015.PubMed/NCBI View Article : Google Scholar | |
Gambke B, Lyons GE, Haselgrove J, Kelly AM and Rubinstein NA: Thyroidal and neural control of myosin transitions during development of rat fast and slow muscles. FEBS Lett. 156:335–339. 1983.PubMed/NCBI View Article : Google Scholar | |
Butler-Browne GS, Herlicoviez D and Whalen RG: Effects of hypothyroidism on myosin isozyme transitions in developing rat muscle. FEBS Lett. 166:71–75. 1984.PubMed/NCBI View Article : Google Scholar | |
Di Maso NA, Caiozzo VJ and Baldwin KM: Single-fiber myosin heavy chain polymorphism during postnatal development: Modulation by hypothyroidism. Am J Physiol Regul Integr Comp Physiol. 278:R1099–R1106. 2000.PubMed/NCBI View Article : Google Scholar | |
Baldwin KM and Haddad F: Effects of different activity and inactivity paradigms on myosin heavy chain gene expression in striated muscle. J Appl Physiol (1985). 90:345–357. 2001.PubMed/NCBI View Article : Google Scholar | |
Adams GR, Haddad F and Baldwin KM: The interaction of space flight and thyroid state on somatic and skeletal muscle growth and myosin heavy chain expression on neonatal rodents. J Gravit Physiol. 7:P15–P18. 2000.PubMed/NCBI | |
Mahdavi V, Izumo S and Nadal-Ginard B: Developmental and hormonal regulation of sarcomeric myosin heavy chain gene family. Circ Res. 60:804–814. 1987.PubMed/NCBI View Article : Google Scholar | |
Simonides WS and Van Hardeveld C: Thyroid hormone as a determinant of metabolic and contractile phenotype of skeletal muscle. Thyroid. 18:205–216. 2008.PubMed/NCBI View Article : Google Scholar | |
Larsson L, Li X, Teresi A and Salviati G: Effects of thyroid hormone on fast- and slow-twitch skeletal muscles in young and old rats. J Physiol. 481:149–161. 1994.PubMed/NCBI View Article : Google Scholar | |
Zhang D, Wang X, Li Y, Zhao L, Lu M, Yao X, Xia H, Wang YC, Liu MF, Jiang J, et al: Thyroid hormone regulates muscle fiber type conversion via miR-133a1. J Cell Biol. 207:753–766. 2014.PubMed/NCBI View Article : Google Scholar | |
Liu N, Bezprozvannaya S, Shelton JM, Frisard MI, Hulver MW, McMillan RP, Wu Y, Voelker KA, Grange RW, Richardson JA, et al: Mice lacking microRNA 133a develop dynamin 2-dependent centronuclear myopathy. J Clin Invest. 121:3258–3268. 2011.PubMed/NCBI View Article : Google Scholar | |
Downes M, Griggs R, Atkins A, Olson EN and Muscat GE: Identification of a thyroid hormone response element in the mouse myogenin gene: Characterization of the thyroid hormone and retinoid X receptor heterodimeric binding site. Cell Growth Differ. 4:901–910. 1993.PubMed/NCBI | |
Ito K and Suda T: Metabolic requirements for the maintenance of self-renewing stem cells. Nat Rev Mol Cell Biol. 15:243–256. 2014.PubMed/NCBI View Article : Google Scholar | |
Scott RC, Schuldiner O and Neufeld TP: Role and regulation of starvation-induced autophagy in the Drosophila fat body. Dev Cell. 7:167–178. 2004.PubMed/NCBI View Article : Google Scholar | |
Mizushima N: The pleiotropic role of autophagy: From protein metabolism to bactericide. Cell Death Differ. 12 (Suppl 2):S1535–S1541. 2005.PubMed/NCBI View Article : Google Scholar | |
Jung HS, Chung KW, Won Kim J, Kim J, Komatsu M, Tanaka K, Nguyen YH, Kang TM, Yoon KH, Kim JW, et al: Loss of autophagy diminishes pancreatic beta cell mass and function with resultant hyperglycemia. Cell Metab. 8:318–224. 2008.PubMed/NCBI View Article : Google Scholar | |
Komatsu M, Waguri S, Ueno T, Iwata J, Murata S, Tanida I, Ezaki J, Mizushima N, Ohsumi Y, Uchiyama Y, et al: Impairment of starvation-induced and constitutive autophagy in Atg7-deficient mice. J Cell Biol. 169:425–434. 2005.PubMed/NCBI View Article : Google Scholar | |
Masiero E, Agatea L, Mammucari C, Blaauw B, Loro E, Komatsu M, Metzger D, Reggiani C, Schiaffino S and Sandri M: Autophagy is required to maintain muscle mass. Cell Metab. 10:507–515. 2009.PubMed/NCBI View Article : Google Scholar | |
Paolini A, Omairi S, Mitchell R, Vaughan D, Matsakas A, Vaiyapuri S, Ricketts T, Rubinsztein DC and Patel K: Attenuation of autophagy impacts on muscle fibre development, starvation induced stress and fibre regeneration following acute injury. Sci Rep. 8(9062)2018.PubMed/NCBI View Article : Google Scholar | |
Carnio S, LoVerso F, Baraibar MA, Longa E, Khan MM, Maffei M, Reischl M, Canepari M, Loefler S, Kern H, et al: Autophagy impairment in muscle induces neuromuscular junction degeneration and precocious aging. Cell Rep. 8:1509–1521. 2014.PubMed/NCBI View Article : Google Scholar | |
Vainshtein A, Grumati P, Sandri M and Bonaldo P: Skeletal muscle, autophagy, and physical activity: The ménage à trois of metabolic regulation in health and disease. J Mol Med (Berl). 92:127–137. 2014.PubMed/NCBI View Article : Google Scholar | |
Carmignac V, Svensson M, Körner Z, Elowsson L, Matsumura C, Gawlik KI, Allamand V and Durbeej M: Autophagy is increased in laminin α2 chain-deficient muscle and its inhibition improves muscle morphology in a mouse model of MDC1A. Hum Mol Genet. 20:4891–4902. 2011.PubMed/NCBI View Article : Google Scholar | |
Grumati P, Coletto L, Sabatelli P, Cescon M, Angelin A, Bertaggia E, Blaauw B, Urciuolo A, Tiepolo T, Merlini L, et al: Autophagy is defective in collagen VI muscular dystrophies, and its reactivation rescues myofiber degeneration. Nat Med. 16:1313–1320. 2010.PubMed/NCBI View Article : Google Scholar | |
Rayagiri SS, Ranaldi D, Raven A, Mohamad Azhar NIF, Lefebvre O, Zammit PS and Borycki AG: Basal lamina remodeling at the skeletal muscle stem cell niche mediates stem cell self-renewal. Nat Commun. 9(1075)2018.PubMed/NCBI View Article : Google Scholar | |
Tang AH and Rando TA: Induction of autophagy supports the bioenergetic demands of quiescent muscle stem cell activation. EMBO J. 33:2782–2797. 2014.PubMed/NCBI View Article : Google Scholar | |
White JP, Billin AN, Campbell ME, Russell AJ, Huffman KM and Kraus WE: The AMPK/p27Kip1 axis regulates autophagy/apoptosis decisions in aged skeletal muscle stem cells. Stem Cell Reports. 11:425–439. 2018.PubMed/NCBI View Article : Google Scholar | |
Fukada SI: The roles of muscle stem cells in muscle injury, atrophy and hypertrophy. J Biochem. 163:353–358. 2018.PubMed/NCBI View Article : Google Scholar | |
Mccarthy JJ, Mula J, Miyazaki M, Erfani R, Garrison K, Farooqui AB, Srikuea R, Lawson BA, Grimes B, Keller C, et al: Effective fiber hypertrophy in satellite cell-depleted skeletal muscle. Development. 138:3657–3666. 2011.PubMed/NCBI View Article : Google Scholar | |
Sousa-Victor P, Gutarra S, García-Prat L, Rodriguez-Ubreva J, Ortet L, Ruiz-Bonilla V, Jardí M, Ballestar E, González S, Serrano AL, et al: Geriatric muscle stem cells switch reversible quiescence into senescence. Nature. 506:316–321. 2014.PubMed/NCBI View Article : Google Scholar | |
Cosgrove BD, Gilbert PM, Porpiglia E, Mourkioti F, Lee SP, Corbel SY, Llewellyn ME, Delp SL and Blau HM: Rejuvenation of the muscle stem cell population restores strength to injured aged muscles. Nat Med. 20:255–264. 2014.PubMed/NCBI View Article : Google Scholar | |
García-Prat L, Martínez-Vicente M, Perdiguero E, Ortet L, Rodríguez-Ubreva J, Rebollo E, Ruiz-Bonilla V, Gutarra S, Ballestar E, Serrano AL, et al: Autophagy maintains stemness by preventing senescence. Nature. 529:37–42. 2016.PubMed/NCBI View Article : Google Scholar | |
Call JA, Wilson RJ, Laker RC, Zhang M, Kundu M and Yan Z: Ulk1-mediated autophagy plays an essential role in mitochondrial remodeling and functional regeneration of skeletal muscle. Am J Physiol Cell Physiol. 312:C724–C732. 2017.PubMed/NCBI View Article : Google Scholar | |
Fortini P, Ferretti C, Iorio E, Cagnin M, Garribba L, Pietraforte D, Falchi M, Pascucci B, Baccarini S, Morani F, et al: The fine tuning of metabolism, autophagy and differentiation during in vitro myogenesis. Cell Death Dis. 7(e2168)2016.PubMed/NCBI View Article : Google Scholar | |
Sin J, Andres AM, Taylor DJR, Weston T, Hiraumi Y, Stotland A, Kim BJ, Huang C, Doran KS and Gottlieb RA: Mitophagy is required for mitochondrial biogenesis and myogenic differentiation of C2C12 myoblasts. Autophagy. 12:369–80. 2016.PubMed/NCBI View Article : Google Scholar | |
Chargé SBP and Rudnicki MA: Cellular and molecular regulation of muscle regeneration. Physiol Rev. 84:209–238. 2004.PubMed/NCBI View Article : Google Scholar | |
Horie T, Kawamata T, Matsunami M and Ohsumi Y: Recycling of iron via autophagy is critical for the transition from glycolytic to respiratory growth. J Biol Chem. 292:8533–8543. 2017.PubMed/NCBI View Article : Google Scholar | |
Duguez S, Féasson L, Denis C and Freyssenet D: Mitochondrial biogenesis during skeletal muscle regeneration. Am J Physiol Endocrinol Metab. 282:E802–E809. 2002.PubMed/NCBI View Article : Google Scholar | |
McMillan EM and Quadrilatero J: Autophagy is required and protects against apoptosis during myoblast differentiation. Biochem J. 462:267–277. 2014.PubMed/NCBI View Article : Google Scholar | |
Hoshino A, Matoba S, Iwai-Kanai E, Nakamura H, Kimata M, Nakaoka M, Katamura M, Okawa Y, Ariyoshi M, Mita Y, et al: p53-TIGAR axis attenuates mitophagy to exacerbate cardiac damage after ischemia. J Mol Cell Cardiol. 52:175–184. 2012.PubMed/NCBI View Article : Google Scholar | |
Le Moal E, Pialoux V, Juban G, Groussard C, Zouhal H, Chazaud B and Mounier R: Redox control of skeletal muscle regeneration. Antioxid Redox Signal. 27:276–310. 2017.PubMed/NCBI View Article : Google Scholar | |
Yin W, Yang L, Kong D, Nie Y, Liang Y and Teng CB: Guanine-rich RNA binding protein GRSF1 inhibits myoblast differentiation through repressing mitochondrial ROS production. Exp Cell Res. 381:139–149. 2019.PubMed/NCBI View Article : Google Scholar | |
Ornatowski W, Lu Q, Yegambaram M, Garcia AE, Zemskov EA, Maltepe E, Fineman JR, Wang T and Black SM: Complex interplay between autophagy and oxidative stress in the development of pulmonary disease. Redox Biol. 36(101679)2020.PubMed/NCBI View Article : Google Scholar | |
Dickinson JD, Sweeter JM, Warren KJ, Ahmad IM, De Deken X, Zimmerman MC and Brody SL: Autophagy regulates DUOX1 localization and superoxide production in airway epithelial cells during chronic IL-13 stimulation. Redox Biol. 14:272–284. 2018.PubMed/NCBI View Article : Google Scholar | |
Lee KA, Kim B, Bhin J, Kim DH, You H, Kim EK, Kim SH, Ryu JH, Hwang D and Lee WJ: Bacterial uracil modulates Drosophila DUOX-dependent gut immunity via Hedgehog-induced signaling endosomes. Cell Host Microbe. 17:191–204. 2015.PubMed/NCBI View Article : Google Scholar | |
Lee KA, Cho KC, Kim B, Jang IH, Nam K, Kwon YE, Kim M, Hyeon DY, Hwang D, Seol JH and Lee WJ: Inflammation-modulated metabolic reprogramming is required for DUOX-dependent gut immunity in Drosophila. Cell Host Microbe. 23:338–352.e5. 2018.PubMed/NCBI View Article : Google Scholar | |
Tian Y, Kuo CF, Sir D, Wang L, Govindarajan S, Petrovic LM and Ou JHJ: Autophagy inhibits oxidative stress and tumor suppressors to exert its dual effect on hepatocarcinogenesis. Cell Death Differ. 22:1025–1034. 2015.PubMed/NCBI View Article : Google Scholar | |
Peng YF, Shi YH, Shen YH, Ding Bin Z, Ke AW, Zhou J, Qiu SJ and Fan J: Promoting colonization in metastatic HCC cells by modulation of autophagy. PLoS One. 8(e74407)2013.PubMed/NCBI View Article : Google Scholar | |
Sciarretta S, Zhai P, Shao D, Zablocki D, Nagarajan N, Terada LS, Volpe M and Sadoshima J: Activation of NADPH oxidase 4 in the endoplasmic reticulum promotes cardiomyocyte autophagy and survival during energy stress through the protein kinase RNA-activated-like endoplasmic reticulum kinase/eukaryotic initiation factor 2α/activating transcription factor 4 pathway. Circ Res. 113:1253–1264. 2013.PubMed/NCBI View Article : Google Scholar | |
Chen Y, Azad MB and Gibson SB: Superoxide is the major reactive oxygen species regulating autophagy. Cell Death Differ. 16:1040–1052. 2009.PubMed/NCBI View Article : Google Scholar | |
Lee SJ, Ryter SW, Xu JF, Nakahira K, Kim HP, Choi AMK and Kim YS: Carbon monoxide activates autophagy via mitochondrial reactive oxygen species formation. Am J Respir Cell Mol Biol. 45:867–873. 2011.PubMed/NCBI View Article : Google Scholar | |
Cho IH, Choi YJ, Gong JH, Shin D, Kang MK and Kang YH: Astragalin inhibits autophagy-associated airway epithelial fibrosis. Respir Res. 16(51)2015.PubMed/NCBI View Article : Google Scholar | |
Filomeni G, De Zio D and Cecconi F: Oxidative stress and autophagy: The clash between damage and metabolic needs. Cell Death Differ. 22:377–388. 2015.PubMed/NCBI View Article : Google Scholar | |
Scherz-Shouval R and Elazar Z: Regulation of autophagy by ROS: Physiology and pathology. Trends Biochem Sci. 36:30–38. 2011.PubMed/NCBI View Article : Google Scholar | |
Scherz-Shouval R and Elazar Z: ROS, mitochondria and the regulation of autophagy. Trends Cell Biol. 17:422–427. 2007.PubMed/NCBI View Article : Google Scholar | |
Scherz-Shouval R, Shvets E, Fass E, Shorer H, Gil L and Elazar Z: Reactive oxygen species are essential for autophagy and specifically regulate the activity of Atg4. EMBO J. 26:1749–1760. 2007.PubMed/NCBI View Article : Google Scholar | |
Recuero M, Munive VA, Sastre I, Aldudo J, Valdivieso F and Bullido MJ: A free radical-generating system regulates AβPP metabolism/processing: involvement of the ubiquitin/proteasome and autophagy/lysosome pathways. J Alzheimers Dis. 34:637–647. 2013.PubMed/NCBI View Article : Google Scholar | |
De Deken X and Miot F: DUOX defects and their roles in congenital hypothyroidism. Methods Mol Biol. 1982:667–693. 2019.PubMed/NCBI View Article : Google Scholar |